Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL MODIFIED MACROCYCLIC COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2017/060167
Kind Code:
A1
Abstract:
The present invention relates to novel modified macrocyclic compounds of general formula (I) as described and defined herein, and methods for their preparation, their use for the treatment and/or prophylaxis of disorders, in particular of hyper-proliferative disorders and/or virally induced infectious diseases and/or of cardiovascular diseases. The invention further relates to intermediate compounds useful in the preparation of said compounds of general formula (I).

Inventors:
LÜCKING ULRICH (DE)
HOG DANIEL (DE)
GEISLER JENS (DE)
SCHOLZ ARNE (DE)
PETERSEN KIRSTIN (DE)
LIENAU PHILIP (DE)
STEGMANN CHRISTIAN (DE)
ANDRES DOROTHEE (DE)
SIEMEISTER GERHARD (DE)
WERBECK NICOLAS (DE)
Application Number:
PCT/EP2016/073399
Publication Date:
April 13, 2017
Filing Date:
September 30, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAYER PHARMA AG (DE)
International Classes:
C07D498/08; A61K31/529; A61P35/00; C07D498/18; C07D515/08
Domestic Patent References:
WO2013037894A12013-03-21
WO2014060375A22014-04-24
WO2014060376A12014-04-24
WO2015155197A12015-10-15
WO2008129070A12008-10-30
WO2008129071A12008-10-30
WO2008129080A12008-10-30
WO2005026129A12005-03-24
WO2009118567A22009-10-01
WO2011116951A12011-09-29
WO2012117048A12012-09-07
WO2012117059A12012-09-07
WO2012143399A12012-10-26
WO2001025220A12001-04-12
WO2008079933A22008-07-03
WO2011012661A12011-02-03
WO2011026917A12011-03-10
WO2012066065A12012-05-24
WO2012066070A12012-05-24
WO2012101062A12012-08-02
WO2012101063A12012-08-02
WO2012101064A12012-08-02
WO2012101065A22012-08-02
WO2012101066A12012-08-02
WO2011077171A12011-06-30
WO2014031937A12014-02-27
WO2013037896A12013-03-21
WO2013037894A12013-03-21
WO2014060376A12014-04-24
WO2014060375A22014-04-24
WO2014060493A22014-04-24
WO2014076028A12014-05-22
WO2014076091A12014-05-22
WO2014076111A12014-05-22
WO2015001021A12015-01-08
WO2015136028A12015-09-17
WO2004009562A12004-01-29
WO2004072063A12004-08-26
WO2010009155A22010-01-21
WO2003037346A12003-05-08
WO2005037800A12005-04-28
WO2008025556A12008-03-06
WO2002066481A12002-08-29
WO2008109943A12008-09-18
WO2009032861A12009-03-12
WO2011046970A12011-04-21
WO2012142329A12012-10-18
WO2012139499A12012-10-18
WO2014106762A12014-07-10
WO2007147574A12007-12-27
WO2007147575A22007-12-27
WO2006066957A22006-06-29
WO2006066956A22006-06-29
WO2004026881A12004-04-01
WO2007079982A12007-07-19
WO2006106895A12006-10-12
WO2012009309A12012-01-19
WO2009132202A22009-10-29
WO2007071455A12007-06-28
WO2005037800A12005-04-28
WO2011029537A12011-03-17
WO2015155197A12015-10-15
WO2015155197A12015-10-15
Foreign References:
US20040209895A12004-10-21
EP1218360B12008-05-28
US20040116388A12004-06-17
US7074789B22006-07-11
US7618968B22009-11-17
US7291616B22007-11-06
US20080064700A12008-03-13
US20030153570A12003-08-14
EP1674470A12006-06-28
EP1674469A12006-06-28
DE10239042A12004-03-04
EP1803723A12007-07-04
EP1710246A12006-10-11
US20070232632A12007-10-04
US20070191393A12007-08-16
US20070191393A12007-08-16
DE2129678A11971-12-30
Other References:
WILLIAM ET AL.: "Discovery of Kinase Spectrum Selective Macrocycle (16 E )-14-Methyl-20-oxa-5,7,14,26-tetraazatetracyclo[19.3.1.1(2,6).1(8,12)]heptacosa-1(25),2(26),3,5,8(27),9,11,16,21,23-decaene (SB1317/TG02), a Potent Inhibitor of Cyclin Dependent Kinases (CDKs), Janus Kinase 2 (JAK2), and Fms-like Tyrosine Kinas", J. MED. CHEM., vol. 55, no. 1, 12 January 2012 (2012-01-12), pages 169 - 196, XP055071583, DOI: 10.1021/jm201112g
CHO ET AL., CELL CYCLE, vol. 9, 2010, pages 1697
HE ET AL., MOL CELL, vol. 29, 2008, pages 588
YANG ET AL., MOL CELL, vol. 19, 2005, pages 535
ZHOU; YIK, MICROBIOL MOL BIOL REV, vol. 70, 2006, pages 646
WANG; FISCHER, TRENDS PHARMACOL SCI, vol. 29, 2008, pages 302
BARK-JONES ET AL., ONCOGENE, vol. 25, 2006, pages 1775
ZHOU ET AL., J VIROL., vol. 80, 2006, pages 4781
DEY ET AL., CELL CYCLE, vol. 6, 2007, pages 1856
WANG ET AL., CHEMISTRY & BIOLOGY, vol. 17, 2010, pages 1111 - 1121
CHEMMEDCHEM, vol. 2, no. 1, 2007, pages 63 - 77
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
R. COPELAND ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 5, 2006, pages 730 - 739
TUMMINO, P.J.; R.A. COPELAND: "Residence time of receptor- ligand complexes and its effect on biological function", BIOCHEMISTRY, vol. 47, no. 20, 2008, pages 5481 - 5492
COPELAND, R.A.; D.L. POMPLIANO; T.D. MEEK: "Drug-target residence time and its implications for lead optimization", NATURE REVIEWS DRUG DISCOVERY, vol. 5, no. 9, 2006, pages 730 - 739, XP002570141, DOI: doi:10.1038/nrd2082
LU, H.; P.J. TONGE: "Drug-target residence time: critical information for lead optimization", CURR OPIN CHEM BIOL, vol. 14, no. 4, 2010, pages 467 - 474
VAUQUELIN, G.; S.J. CHARLTON: "Long-lasting target binding and rebinding as mechanisms to prolong in vivo drug action", BR J PHARMACOL, vol. 161, no. 3, 2010, pages 488 - 508
PRICE, D.; A. SHARMA; F. CERASOLI, BIOCHEMICAL PROPERTIES, PHARMACOKINETICS AND PHARMACOLOGICAL RESPONSE OF TIOTROPIUM IN CHRONIC OBSTRUCTIVE PULMONARY DISEASE PATIENTS, 2009
DOWLING, M., BR. J. PHARMACOL., vol. 148, 2006, pages 927 - 937
WOOD ET AL., CANCER RES., vol. 64, 2004, pages 6652 - 6659
LACKEY, CURRENT TOPICS IN MEDICINAL CHEMISTRY, vol. 6, no. 5, 2006
E.H. KERNS; L. DI: "Drug-like Properties: Concepts, Structure Design and Methods", 2008, ACADEMIC PRESS, pages: 276 - 286
EDWARD H. KERNS; LI DI: "Solubility Methods in: Drug-like Properties: Concepts, Structure Design and Methods", 2008, ACADEMIC PRESS, pages: 276 - 286
JONSSON U ET AL., ANN BIOL CLIN., vol. 51, no. 1, 1993, pages 19 - 26
JOHNSSON B ET AL., ANAL BIOCHEM., vol. 198, no. 2, 1991, pages 268 - 277
DAY Y ET AL., PROTEIN SCIENCE, vol. 11, 2002, pages 1017 - 1025
MYSKZA DG, ANAL BIOCHEM., vol. 329, 2004, pages 316 - 323
TUMMINO; COPELAND, BIOCHEMISTRY, vol. 47, no. 20, 2008, pages 5481 - 5492
JOHNSSON B ET AL., ANAL BIOCHEM., vol. 198, no. 2, 1 November 1991 (1991-11-01), pages 268 - 277
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 2006, WILEY
L. JIANG; S.L. BUCHWALD: "Metal-Catalyzed Cross-Coupling Reactions", 2004, WILEY-VCH
P. G. M. WUTS; T. W. GREEN: "Protective Groups in Organic Synthesis", 2006, JOHN WILEY & SONS
C.R. JOHNSON, J. ORG. CHEM., vol. 58, 1993, pages 1922
C. BOLM ET AL., SYNTHESIS, vol. 10, 2009, pages 1601
C. BOLM ET AL., CHEM. EUROP. J., vol. 10, 2004, pages 2942
C. BOLM ET AL., SYNTHESIS, vol. 7, 2002, pages 879
C. BOLM ET AL., CHEM. EUR. J., vol. 7, 2001, pages 1118
C. BOLM ET AL., TET. LETT., vol. 39, 1998, pages 5731
C. BOLM ET AL., J. ORG. CHEM., vol. 65, 2000, pages 169
C. BOLM ET AL., SYNTHESIS, vol. 7, 2000, pages 911
C. BOLM ET AL., J. ORG. CHEM., vol. 70, 2005, pages 2346
V.J. BAUER ET AL., J. ORG. CHEM., vol. 31, 1966, pages 3440
C. R. JOHNSON ET AL., J. AM. CHEM. SOC., vol. 92, 1970, pages 6594
S. ALLENMARK ET AL., ACTA CHEM. SCAND. SER. B, 1983, pages 325
D.J. CRAM ET AL., J. AM. CHEM. SOC., vol. 92, 1970, pages 7369
C.R. JOHNSON ET AL., J. ORG. CHEM., vol. 43, 1978, pages 4136
A.C. BARNES, J. MED. CHEM., vol. 22, 1979, pages 418
D. CRAIG ET AL., TET, vol. 51, 1995, pages 6071
D.J. CRAM ET AL., J. AM. CHEM. SOC., vol. 96, 1974, pages 2183
P. STOSS ET AL., CHEM. BER., vol. 111, 1978, pages 1453
D.T. SAUER ET AL., INORGANIC CHEMISTRY, vol. 11, 1972, pages 238
C. BOLM ET AL., ORG. LETT., vol. 9, 2007, pages 2951
M.H. ALI ET AL., SYNTHESIS, 1997, pages 764
M.C. CARRENO, CHEM. REV., vol. 95, 1995, pages 1717
I. PATEL ET AL., ORG. PROC. RES. DEV., vol. 6, 2002, pages 225
N. KHIAR ET AL., CHEM. REV., vol. 103, 2003, pages 3651
BOLM ET AL., ORG. LETT., vol. 6, 2004, pages 1305
BULL ET AL., J. ORG. CHEM., vol. 80, 2015, pages 6391
R. CHINCHILLA; C. NAJERA, CHEM. REV., vol. 107, 2007, pages 874 - 922
S. NISHIMURA: "Handbook of Heterogeneous Catalytic Hydrogenation for Organic Synthesis", 2001, WILEY-VCH
H. R. BENTLEY ET AL., J. CHEM. SOC., 1952, pages 1572
SATZINGER ET AL., ANGEW. CHEM., vol. 83, 1971, pages 83
SAMMOND ET AL., BIOORG. MED. CHEM. LETT., vol. 15, 2005, pages 3519
D.G. HALL: "Boronic Acids", 2005, WILEY-VCH VERLAG GMBH & CO. KGAA
K.C.K. SWAMY ET AL., CHEM. REV., vol. 109, 2009, pages 2551
R.C. LAROCK: "Comprehensive Organic Transformations", 1989, VCH, pages: 411 - 415
J.A. BULL, J. ORG. CHEM., vol. 80, 2015, pages 6391
Attorney, Agent or Firm:
BIP PATENTS (DE)
Download PDF:
Claims:
Patent claims

1. A compound of general formula (I)

0)

wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-,

-S(=0)(=NR5)-;

G, E represent, independently from each other, a bivalent moiety selected from the group consisting of -O-, -N(RA)-, -CH2-, -CH(Ci-C6-alkyl)-, -C(Ci-C6-alkyl)2-, -S-, -S(=0)-, -S(=0)2-, with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-C8-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, -NR6R7, C2-C3-alkenyl-, C2-C3-alkynyl-, C3-C4- cycloalkyl-, hydroxy-Ci-C3-alkyl, -(CH2)NR6R7, and/or

(ii) one or two or three or four substituents, identically or differently, selected from halogen and Ci-C3-alkyl-,

or wherein

one carbon atom of said C2-C8-alkylene moiety forms a three- or four-membered ring together with a bivalent moiety to which it is attached, wherein said bivalent moiety is selected from - CH2CH2-, -CH2CH2CH2-, -CH2OCH2-;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a group selected from Ci-C6-alkyl-, C3-C6-alkenyl-, C3-C6-alkynyl-,

C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-, phenyl-Ci-C3-alkyl- and heteroaryl-Ci- C3-alkyl-, wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C6-alkyl-, halo-Ci- C3-alkyl-, Ci-C6-alkoxy-, Ci-C3-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, -OP(=0)(OH)¾ -C(=0)OH, -C(=0)NH2;

R2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R3, R4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R5 represents a group selected from a hydrogen atom, cyano,

-C(=0)NR6R7, Ci-Ce-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-,

wherein said Ci-C6-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R6, R7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said Ci-Ce-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -ΝΗ2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-, or

R6 and R7, together with the nitrogen atom they are attached to, form a cyclic amine;

R8 represents a group selected from Ci-C6-alkyl-, halo-Ci-C3-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-,

RA represents a hydrogen atom or a Ci-C6-alkyl- group, or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

2. The compound of general formula (I) according to claim 1 , wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-,

-S(=0)(=NR5)-;

G, E each represent, independently from each other, a bivalent moiety selected from the group consisting of-O-, -N(RA)-, -CH2-, -CH(Ci-C3-alkyl)-, -S-, -S(=0)2-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-Cs-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, C3-C4-cycloalkyl-, hydroxy-Ci-C3-alkyl-,

-(CH2)NR6R7, and/or

(ii) one or two or three additional substituents, identically or differently, selected from a fluorine atom and a Ci-C3-alkyl- group;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a group selected from Ci-C6-alkyl- and C3-Cs-cycloalkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C3-alkyl-, fluoro- Ci-C2-alkyl-, Ci-C3-alkoxy-, Ci-C2-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, -OP(=0)(OH)¾ -C(=0)OH, -C(=0)NH2;

R2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, C1-C2- alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C2-alkyl-;

R3, R4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano Ci-C2-alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-;

R5 represents a group selected from a hydrogen atom, cyano,

-C(=0)NR6R7, Ci-Ce-alkyl-, C3-C5-cycloalkyl-, phenyl-,

wherein said Ci-C6-alkyl-, C3-C5-cycloalkyl- or phenyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-;

R6, R7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C3-C5-cycloalkyl-, phenyl- and benzyl-, wherein said Ci-C6-alkyl-, Cs-Cs-cycloalkyl-, phenyl- or benzyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-, or

R6 and R7, together with the nitrogen atom they are attached to, form a cyclic amine;

R8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl-, Cs-Cs-cycloalkyl-, phenyl- and benzyl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-;

RA represents a hydrogen atom or a Ci-C3-alkyl- group, or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

3. The compound of general formula (I) according to any one of claims 1 or 2, wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-, -S(=0)(=NR5)-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-, -CH2-,

-CH(Ci-C3-alkyl)-, -S-; G represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-, -CH2-,

-CH(Ci-C3-alkyl)-, -S-, -S(=0)2-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-Cs-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from C3-C4-cycloalkyl- and hydroxymethyl-, and/or

(ii) one or two additional substituents, identically or differently, selected from Ci-C2-alkyl-,

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a group selected from Ci-C i-alkyl- and C3-Cs-cycloalkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, a fluorine atom, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2, -C(=0)OH; represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-;

R represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-;

R4 represents a hydrogen atom or a fluorine atom;

R5 represents a group selected from a hydrogen atom, cyano, -C(=0)R8,

-C(=0)OR8, -S(=0)2R8, -C(=0)NR6R7, Ci-C4-alkyl-,

wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of a fluorine atom, hydroxy, cyano, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines;

R6, R7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C i-alkyl- and C3-C5-cycloalkyl-,

wherein said Ci-C i-alkyl- or Cs-Cs-cycloalkyl- group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, or

R6 and R7, together with the nitrogen atom they are attached to, form a cyclic amine;

R8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl-, Cs-Cs-cycloalkyl- and benzyl-, wherein said group is optionally substituted with one substituent selected from the group consisting of halogen, hydroxy, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2;

RA represents a hydrogen atom or a Ci-C3-alkyl- group, or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

4. The compound of general formula (I) according to any one of claim 1, 2 or 3, wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR5)-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-, -CH2-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-, -CH2-, -S-; with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-Cs-alkylene moiety; X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a Ci-C i-alkyl- group,

wherein said group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkoxy-, -Ν¾ -C(=0)OH;

R2 represents a hydrogen atom or a fluorine atom;

R3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R4 represents a hydrogen atom;

R5 represents a group selected from a hydrogen atom, cyano, -C(=0)R8, -C(=0)OR8, Ci-C4-alkyl-,

wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of hydroxy, cyano, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-;

R8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl- and benzyl-;

RA represents a hydrogen atom, a methyl- or an ethyl- group,

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

5. The compound of general formula (I) according to any one of claim 1 to 4, wherein L represents a C3-C4-alkylene moiety,

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof. 6. The compound of general formula (I) according to any one of claim 1 to 5, wherein

A represents a bivalent moiety -S(=0)(=NR5)-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(H) -;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

R5 represents a hydrogen atom;

RA represents a hydrogen atom or a methyl- group, or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

7. The compound of general formula (I) according to claim 1 , wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR5)-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, -N(H)-, -S-; with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-Cs-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a Ci-C3-alkyl- group;

R2 represents a hydrogen atom or a fluorine atom;

R3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R4 represents a hydrogen atom;

R5 represents a group selected from a hydrogen atom, cyano, -C(=0)R8,

-C(=0)OR8, Ci-C3-alkyl-;

R8 represents a group selected from Ci-C i-alkyl-, trifluoromethyl- and benzyl-;

RA represents a hydrogen atom or a methyl- group,

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

8. The compound of general formula (I) according to claim 1, wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR5)-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-C i-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a methyl- group;

R2 represents a hydrogen atom;

R3 represents a hydrogen atom or a fluorine atom;

R4 represents a hydrogen atom;

R5 represents a group selected from a hydrogen atom, -C(=0)OR8;

R8 represents a group selected from tert-butyl- and benzyl-;

RA represents a hydrogen atom or a methyl- group,

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

9. The compound of general formula (I) according to any one of claim 1 to 8, wherein

R represents a fluorine atom, and

R4 represents a hydrogen atom,

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

10. The compound of general formula (I) according to claim 1, wherein

A represents a bivalent moiety -S(=0)(=NR5)-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(RA)-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(H)-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-C4-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R1 represents a methyl- group;

R2 represents a hydrogen atom;

R3 represents a hydrogen atom or a fluorine atom;

R4 represents a hydrogen atom;

R5 represents a hydrogen atom;

RA represents a hydrogen atom or a methyl- group,

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

11. The compound according to claim 1 , which is selected from

15,19-difluoro-8- [(methylsulfanyl)methyl]-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 12,16-(metheno)- 1,5,11, 13-benzoxatriazacyclooctadecine;

- (rac)-benzyl [{[16,20-difluoro-3,4,5,6-tetrahydro-2H,12H-l l,7-(azeno)-13,17-(metheno)-l,6,12,14- benzoxatriazacyclononadecin-9-yl]methyl}(methyl)oxido-λ6-sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-3,4,5,6-tetrahydro-2H, 12H- 11 ,7-(azeno)- 13,17-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine;

2, 18-difluoro-9-[(methylsulfanyl)methyl]-l 3, 14, 15,16-tetrahydro-6H-7, 11 -(azeno)-5, 1 -(metheno)- 12,4,6-benzoxadiazacyclooctadecine;

- (rac)-2, 18-difluoro-9- [(methylsulfmyl)methyl]- 13, 14,15,16-tetrahydro-6H-7, 11 -(azeno)-5, 1 - (metheno)- 12,4,6-benzoxadiazacyclooctadecine; - (rac)-tert-butyl [{[16,20-difluoro-6-methyl-3,4,5,6-tetrahydro-2H,12H-13,17-(azeno)-l 1,7-

(metheno)- 1 ,6,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl) oxido-λ6- sulfanylidene]carbamate;

(rac)-tert-butyl [{[15,19-difluoro-2,3,4,5-tetrahydro-l lH-10,6-(azeno)-16,12-(metheno)-l,5,l 1,13- benzoxatriazacyclooctadecin-8-yl]methyl}(methyl)oxido-λ6-sulfanylidene]carbamate;

(rac)- 15,19-difluoro-8- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 16,12-(metheno)- 1,5,11, 13-benzoxatriazacyclooctadecine;

(rac)-tert-butyl [{[17,21-difluoro-2,3,4,5,6,7-hexahydro-13H-12,8-(azeno)-14,18-(metheno)- 1,7,13,15-benzoxatriazacycloicosin- 10-yl]methyl} (methyl)oxido-λ6-sulfanylidene]carbamate;

- (rac)-17,21-difluoro-10-[(S-methylsulfonimidoyl)methyl]-2,3,4,5,6,7-hexahydro-13H-12,8-(azeno)- 14,18-(metheno)- 1,7,13,15-benzoxatriazacycloicosine

(rac)-16,20-difluoro-6-methyl-9-[(S-methylsulfonimidoyl)methyl]-3,4,5,6-tetrahydro-2H-7,l l- (azeno)- 17, 13-(metheno)- 1 ,6,12,14-benzoxatriazacyclononadecine

8,16,20-trifluoro-6-methyl-9-[(methylsulfanyl)methyl]-3,4,5,6-tetrahydro-2H,12H-l l,7-(azeno)- 13,17-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine

(rac)-fert-butyl [{[15,19-difluoro- 1 ,2,3,4-tetrahydro- 16,12-(azeno)-6, 10-(metheno)-5, 1,11,13- benzoxatriazacyclooctadecin-8(l lH)-yl]methyl}(methyl)oxido-λ6-sulfanylidene]carbamate;

(rac)- 15,19-difluoro-8- [(S-methylsulfonimidoyl)methyl]- 1 ,2,3,4-tetrahydro- 16,12-(azeno)-6, 10- (metheno)-5, 1,11, 13-benzoxatriazacyclooctadecine;

- (rac)-tert-butyl [{[16,20-difluoro-2,3,4,5-tetrahydro-lH,12H-13,17-(azeno)-l l,7-(metheno)-

6,1,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl)oxido^6-sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 1 H- 17, 13-(azeno)-7, 11 - (metheno)-6, 1,12,14-benzoxatriazacyclononadecine;

(rac)- 16,20-difluoro-6-methyl-9-[(S-methylsulfonimidoyl)methyl] - 1 ,2,3,4,5, 6-hexahydro- 12H- 17, 13 -(azeno)- 1 l,7-(metheno)-l,6,12,14-benzotetraazacyclononadecine; salt with formic acid;

2, 19-difluoro-9-[(methylsulfanyl)methyl]- 14,15,16,17-tetrahydro-6H, 13H-7, 11 -(azeno)-5, 1 - (metheno)- 12,4,6-benzoxadiazacyclononadecine;

(rac)-2, 19-difluoro-9- [(methylsulfmyl)methyl]- 14,15,16,17-tetrahydro-6H, 13H-7, 11 -(azeno)-5, 1 - (metheno)- 12,4,6-benzoxadiazacyclononadecine;

- (rac)-2,19-difluoro-9-[(S-methylsulfonimidoyl)methyl]-14,15,16,17-tetrahydro-6H,13H-7,l 1- (azeno)-5, 1 -(metheno)- 12,4,6-benzoxadiazacyclononadecine; (rac)-tert-butyl [ { [ 16,20-difluoro-2,3 ,4,5-tetrahydro- 12H- 17, 13-(azeno)- 11 ,7-(metheno)-6, 1,12,14- benzoxathiadiazacyclononadecin-9-yl]methyl}(methyl)oxido-λ6-sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 12H- 17, 13-(azeno)- 11 ,7-(metheno)-6, 1,12,14-benzoxathiadiazacyclononadecine;

- (rac)-16,20-difluoro-6-methyl-9-[(S-methylsulfo^

13,17-(azeno)- 11 ,7-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine; [{[16,20-difluoro-3,4,5,6-tetrahydro-2H,12H-13,17-(azeno)-l l,7-(metheno)- 1,6,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl)oxido^6-sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-3,4,5,6-tetrahydro-2H, 12H- 13,17-(azeno)- 11 ,7-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine;

and an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

12. A compound of general formula (I) according to any one of claims 1 to 11 for the use as a

medicament.

13. A compound of general formula (I) according to any one of claims 1 to 11 for the use of treating and/or prophylaxis of hyper-pro liferative disorders, virally induced infectious diseases and/or of cardiovascular diseases. 14. A compound of general formula (I) according to any one of claims 1 to 11 for the use of treating and/or prophylaxis of lung carcinomas, prostate carcinomas, cervical carcinomas, colorectal carcinomas, melanomas or ovarian carcinomas.

15. Use of a compound of general formula (I) according to any one of claims 1 to 11 in the manufacture of a medicament for the treatment and/or prophylaxis of hyper-pro liferative disorders, virally induced infectious diseases and/or of cardiovascular diseases.

Use of a compound of general formula (I) according to any one of claims 1 to 11 in the manufacture of a medicament for the treatment and/or prophylaxis of lung carcinomas, prostate carcinomas, cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias.

Use of a compound of general formula (I) according to any one of claims 1 to 11 in the manufacture of a medicament for the treatment and/or prophylaxis of non-small cell lung carcinomas, hormone- independent human prostate carcinomas, multidrug-resistant human cervical carcinomas or human acute myeloid leukemias.

18. A pharmaceutical combination comprising a compound according to any one of claims 1 to 11 in combination with at least one or more further active ingredients.

19. The pharmaceutical combination according to claim 18 for use of the treatment and/or prophylaxis of hyper-proliferative disorders, virally induced infectious diseases and/or of cardiovascular diseases.

20. The pharmaceutical combination according to claim 18 for use of the treatment and/or prophylaxis of lung carcinomas, prostate carcinomas, cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias.

21. A pharmaceutical composition comprising a compound according to any one of claims 1 to 11 in combination with an inert, nontoxic, pharmaceutically suitable adjuvant.

22. The pharmaceutical composition according to claim 21 for use of the treatment and/or prophylaxis of hyper-proliferative disorders, virally induced infectious diseases and/or of cardiovascular diseases.

23. The pharmaceutical composition according to claim 21 for use of the treatment and/or prophylaxis of lung carcinomas, prostate carcinomas, cervical carcinomas, colorectal carcinomas, melanomas ovarian carcinomas or leukemias.

24. A compound of general formula (44)

wherein R1, R2, R3, R4 and L are as defined according to any one of claims 1 to 10 for the compound of general formula (I),

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

25. A compound of general formula (34)

wherein R1, R2, R3, R4, RA and L are as defined according to any one of claims 1 to 10 for the compound of general formula (I),

or an enantiomer, diastereomer, salt, solvate or salt of solvate thereof.

26. A process for the preparation of a compound of formula (45), in which process a compound of the formula (44), in which R1, R2, R3, R4 and L are as defined for the compound of formula (I) according to any one of the claims 1 to 10,

is reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

45

to give a compound of the formula (45),

and in which process the resulting compound is optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

27. A process for the preparation of a compound of formula (35), in which process a compound of the formula (34), in which R1, R2, R3, R4, RA and L are as defined for the compound of formula (I) according to any one of the claims 1 to 10,

is reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give a compound of the formula (35),

and in which process the resulting compound is optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

Description:
Novel modified macrocyclic compounds

The present invention relates to novel modified macrocyclic compounds of general formula (I) as described and defined herein, and methods for their preparation, their use for the treatment and/or prophylaxis of disorders, in particular of hyper-pro liferative disorders and/or virally induced infectious diseases and/or of cardiovascular diseases. The invention further relates to intermediate compounds useful in the preparation of said compounds of general formula (I).

The family of cyclin-dependent kinase (CDK) proteins consists of members that are key regulators of the cell division cycle (cell cycle CDK's), that are involved in regulation of gene transcription (transcriptional CDK's), and of members with other functions. CDKs require for activation the association with a regulatory cyclin subunit. The cell cycle CDKs CDKl/cyclin B, CDK2/cyclin A, CDK2/cyclinE, CDK4/cyclinD, and CDK6/cyclinD get activated in a sequential order to drive a cell into and through the cell division cycle. The transcriptional CDKs CDK9/cyclin T and CDK7/cyclin H regulate the activity of RNApolymerase II via phosphorylation of the carboxy-terminal domain (CTD). Positive transcription factor b (PTEFb) is a heterodimer of CDK9 and one of four cyclin partners, cyclin Tl, cyclin K, cyclin T2a or T2b.

Whereas CDK9 (NCBI GenBank Gene ID 1025) is exclusively involved in transcriptional regulation, CDK7 in addition participates in cell cycle regulation as CDK-activating kinase (CAK).

Transcription of genes by RNA polymerase II is initiated by assembly of the pre-initiation complex at the promoter region and phosphorylation of Ser 5 and Ser 7 of the CTD by CDK7/cyclin H. For a major fraction of genes RNA polymerase II stops mRNA transcription after it moved 20-40 nucleotides along the DNA template. This promoter-proximal pausing of RNA polymerase II is mediated by negative elongation factors and is recognized as a major control mechanism to regulate expression of rapidly induced genes in response to a variety of stimuli (Cho et al., Cell Cycle 9, 1697, 2010). PTEFb is crucially involved in overcoming promoter-proximal pausing of RNA polymerase II and transition into a productive elongation state by phosphorylation of Ser 2 of the CTD as well as by phosphorylation and inactivation of negative elongation factors.

Activity of PTEFb itself is regulated by several mechanisms. About half of cellular PTEFb exists in an inactive complex with 7SK small nuclear RNA (7SK snRNA), La-related protein 7 (LARP7/PIP7S) and hexamethylene bis-acetamide inducible proteins 1/2 (HEXIM1/2, He et al., Mol Cell 29, 588, 2008). The remaining half of PTEFb exists in an active complex containing the bromodomain protein Brd4 (Yang et al., Mol Cell 19, 535, 2005). Brd4 recruits PTEFb through interaction with acetylated histones to chromatin areas primed for gene transcription. Through alternately interacting with its positive and negative regulators, PTEFb is maintained in a functional equilibrium: PTEFb bound to the 7SK snRNA complex represents a reservoir from which active PTEFb can be released on demand of cellular transcription and cell proliferation (Zhou & Yik, Microbiol Mol Biol Rev 70, 646, 2006). Furthermore, the activity of PTEFb is regulated by posttranslational modifications including phosphorylation/de- phosphorylation, ubiquitination, and acetylation (reviewed in Cho et al., Cell Cycle 9, 1697, 2010).

Deregulated activity of CDK9 kinase activity of the PTEFb heterodimer is associated with a variety of human pathological settings such as hyper-proliferative diseases (e.g. cancer), virally induced infectious diseases or cardiovascular diseases: Cancer is regarded as a hyper-proliferative disorder mediated by a disbalance of proliferation and cell death (apoptosis). High levels of anti-apoptotic Bcl-2-family proteins are found in various human tumors and account for prolonged survival of tumor cells and therapy resistance. Inhibition of PTEFb kinase activity was shown to reduce transcriptional activity of RNA polymerase II leading to a decline of shortlived anti-apoptotic proteins, especially Mcl-1 and XIAP, reinstalling the ability of tumor cells to undergo apoptosis. A number of other proteins associated with the transformed tumor phenotype (such as Myc, NF-kB responsive gene transcripts, mitotic kinases) are either short-lived proteins or are encoded by short-lived transcripts which are sensitive to reduced RNA polymerase II activity mediated by PTEFb inhibition (reviewed in Wang & Fischer, Trends Pharmacol Sci 29, 302, 2008). Many viruses rely on the transcriptional machinery of the host cell for the transcription of their own genome. In case of HIV-1, RNA polymerase II gets recruited to the promoter region within the viral LTR's. The viral transcription activator (Tat) protein binds to nascent viral transcripts and overcomes promoter-proximal RNA polymerase II pausing by recruitment of PTEFb which in turn promotes transcriptional elongation. Furthermore, the Tat protein increases the fraction of active PTEFb by replacement of the PTEFb inhibitory proteins HEXIM1/2 within the 7SK snRNA complex. Recent data have shown that inhibition of the kinase activity of PTEFb is sufficient to block HIV-1 repliction at kinase inhibitor concentrations that are not cytotoxic to the host cells (reviewed in Wang & Fischer, Trends Pharmacol Sci 29, 302, 2008). Similarly, recruitment of PTEFb by viral proteins has been reported for other viruses such as B-cell cancer-associated Epstein-Barr virus, where the nuclear antigen EBNA2 protein interacts with PTEFb (Bark- Jones et al., Oncogene, 25, 1775, 2006), and the human T- lymphotropic virus type 1 (HTLV-1), where the transcriptional activator Tax recruits PTEFb (Zhou et al., J Virol. 80, 4781, 2006).

Cardiac hypertrophy, the heart's adaptive response to mechanical overload and pressure (hemodynamic stress e.g. hypertension, myocardial infarction), can lead, on a long term, to heart failure and death. Cardiac hypertrophy was shown to be associated with increased transcriptional activity and RNA polymerase II CTD phosphorylation in cardiac muscle cells. PTEFb was found to be activated by dissociation from the inactive 7SK snRNA/HEXIMl/2 complex. These findings suggest pharmacological inhibition of PTEFb kinase activity as a therapeutic approach to treat cardiac hypertrophy (reviewed in Dey et al., Cell Cycle 6, 1856, 2007). In summary, multiple lines of evidence suggest that selective inhibition of the CDK9 kinase activity of the PTEFb heterodimer (= CDK9 and one of four cyclin partners, cyclin Tl, cyclin K, cyclin T2a or T2b) represents an innovative approach for the treatment of diseases such as cancer, viral diseases, and/or diseases of the heart. CDK9 belongs to a family of at least 13 closely related kinases of which the subgroup of the cell cycle CDK's fulfills multiple roles in regulation of cell proliferation. Thus, co- inhibition of cell cycle CDKs (e.g. CDKl/cyclin B, CDK2/cyclin A, CDK2/cyclinE, CDK4/cyclinD, CDK6/cyclinD) and of CDK9, is expected to impact normal proliferating tissues such as intestinal mucosa, lymphatic and hematopoietic organs, and reproductive organs. To maximize the therapeutic value of CDK9 kinase inhibitors, molecules with improved duration of action and/or high potency and efficacy and/or selectivity towards CDK9 are required.

CDK inhibitors in general as well as CDK9 inhibitors are described in a number of different publications: WO2008129070 and WO2008129071 both describe 2,4 disubstituted aminopyrimidines as CDK inhibitors in general. It is also asserted that some of these compounds may act as selective CDK9 inhibitors (WO2008129070) and as CDK5 inhibitors (WO2008129071), respectively, but no specific CDK9 IC 50 (WO2008129070) or CDK5 IC 50 (WO2008129071) data is presented. These compounds do not contain a fluorine atom in 5-position of the pyrimidine core.

WO2008129080 discloses 4,6 disubstituted aminopyrimidines and demonstrates that these compounds show an inhibitory effect on the protein kinase activity of various protein kinases, such as CDK1 , CDK2, CDK4, CDK5, CDK6 and CDK9, with a preference for CDK9 inhibition (example 80).

WO2005026129 discloses 4,6 disubstituted aminopyrimidines and demonstrates that these compounds show an inhibitory effect on the protein kinase activity of various protein kinases, in particular CDK2, CDK4, and CDK9.

WO 2009118567 discloses pyrimidine and [l,3,5]triazine derivatives as protein kinase inhibitors, in particular CDK2, CDK7 and CDK9.

WO2011116951 discloses substituted triazine derivatives as selective CDK9 inhibitors.

WO2012117048 discloses disubstituted triazine derivatives as selective CDK9 inhibitors.

WO2012117059 discloses disubstituted pyridine derivatives as selective CDK9 inhibitors.

WO2012143399 discloses substituted 4-aryl-N-phenyl-l,3,5-triazin-2-amines as selective CDK9 inhibitors. EP1218360 Bl, which corresponds to US2004116388A1, US7074789B2 and WO2001025220A1, describes triazine derivatives as kinase inhibitors, but does not disclose potent or selective CDK9 inhibitors. WO2008079933 discloses aminopyridine and aminopyrimidine derivatives and their use as CDK1, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8 or CDK9 inhibitors.

WO2011012661 describes aminopyridine derivatives useful as CDK inhibitors. WO2011026917 discloses carboxamides derived from substituted 4-phenylpyridine-2-amines as inhibitors ofCDK9.

WO2012066065 discloses phenyl-heterorayl amines as inhibitors of CDK9. A selectivity towards CDK9 over other CDK isoforms is preferred, however disclosure of CDK-inhibition data is confined to CDK 9. No bicyclic ring systems are disclosed attached to the C4 position of the pyrimidine core. Within the group attached to C4 of the pyrimidine core, alkoxy phenyls can be regarded as encompassed, but there is no suggestion for a specific substitution pattern characterised by a fluorine atom attached to C5 of the pyrimidine ring, and an aniline at C2 of the pyrimidine, featuring a substituted sulfonyl-methylene group in meta position. Compounds shown in the examples typically feature a substituted cycloalkyl group as R 1 but no phenyl.

WO2012066070 discloses 3-(aminoaryl)-pyridine compounds as inhibitors of CDK9. The biaryl core mandatorily consists of two heteroaromatic rings.

WO2012101062 discloses substituted bi-heteroaryl compounds featuring a 2-aminopyridine core as inhibitors of CDK9. The biaryl core mandatorily consists of two heteroaromatic rings.

WO2012101063 discloses carboxamides derived from substituted 4-(heteroaryl)-pyridine-2-amines as inhibitors of CDK9. WO 2012101064 discloses N-acyl pyrimidine biaryl compounds as inhibitors of CDK9.

WO 2012101065 discloses pyrimidine biaryl compounds as inhibitors of CDK9. The biaryl core mandatorily consists of two heteroaromatic rings.

WO 2012101066 discloses pyrimidine biaryl compounds as inhibitors of CDK9. Substitution R 1 of the amino group attached to the heteroaromatic core is confined to non-aromatic groups but does not cover substituted phenyls. Furthermore, the biaryl core mandatorily consists of two heteroaromatic rings. WO 2011077171 discloses 4,6-disubstituted aminopyrimidine derivatives as inhibitors of CDK9.

WO 2014031937 discloses 4,6-disubstituted aminopyrimidine derivatives as inhibitors of CDK9. WO 2013037896 discloses disubstituted 5-fluoropyrimidines as selective inhibitors of CDK9.

WO 2013037894 discloses disubstituted 5-fluoropyrimidine derivatives containing a sulfoximine group as selective inhibitors of CDK9. Wang et al. (Chemistry & Biology 17, 1111-1121, 2010) describe 2-anilino-4-(thiazol-5-yl)pyrimidine transcriptional CDK inhibitors, which show anticancer activity in animal models.

WO 2014060376 discloses substituted 4-(ortho)-fluorophenyl-5-fluoropyrimidin-2-yl amine derivatives containing a sulfone group as selective inhibitors of CDK9.

WO 2014060375 discloses substituted 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group as selective inhibitors of CDK9.

WO 2014060493 discloses substituted N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfone group as selective inhibitors of CDK9.

WO 2014076028 discloses substituted 4-(ortho)-fluorophenyl-5-fluoropyrimidin-2-yl amine derivatives containing a sulfoximine group as selective inhibitors of CDK9. WO 2014076091 discloses substituted 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group as selective inhibitors of CDK9.

WO 2014076111 discloses substituted N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfoximine group as selective inhibitors of CDK9.

WO 2015001021 discloses 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group as selective inhibitors of CDK9.

WO 2015136028 discloses 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group as selective inhibitors of CDK9. WO2004009562 discloses substituted triazine kinase inhibitors. For selected compounds CDK1 and CDK4 test data, but no CDK9 data is presented.

WO2004072063 describes heteroaryl (pyrimidine, triazine) substituted pyrroles as inhibitors of protein kinases such as ERK2, GSK3, PKA or CDK2.

WO2010009155 discloses triazine and pyrimidine derivatives as inhibitors of histone deacetylase and/or cyclin dependent kinases (CDKs). For selected compounds CDK2 test data is described. WO2003037346 (corresponding to US7618968B2, US7291616B2, US2008064700A1, US2003153570A1) relates to aryl triazines and uses thereof, including to inhibit lysophosphatidic acid acyltransferase beta (LPAAT-beta) activity and/or proliferation of cells such as tumor cells.

WO2005037800 discloses sulfoximine substituted anilino-pyrimidines as inhibitors of VEGFR and CDK kinases, in particular VEGFR2, CDK1 and CDK2, having no aromatic ring directly bonded to the pyrimidine ring and having the sulfoximine group directly bonded to the aniline group. No CDK9 data are disclosed.

WO2008025556 describes carbamoyl sulfoximides having a pyrimidine core, which are useful as kinase inhibitors. No CDK9 data is presented. No molecules are exemplified, which possess a fluoropyrimidine core.

WO2002066481 describes pyrimidine derivatives as cyclin dependent kinase inhibitors. CDK9 is not mentioned and no CDK9 data is presented.

WO2008109943 concerns phenyl aminopyri(mi)dine compounds and their use as kinase inhibitors, in particular as JAK2 kinase inhibitors. The specific examples mainly focus on compounds having a pyrimidine core. WO2009032861 describes substituted pyrimidinyl amines as JNK kinase inhibitors. The specific examples mainly focus on compounds having a pyrimidine core.

WO2011046970 concerns amino-pyrimidine compounds as inhibitors of TBK1 and/or IKK epsilon. The specific examples mainly focus on compounds having a pyrimidine core.

WO2012142329 concerns amino-pyrimidine compounds as inhibitors of TBK1 and/or IKK epsilon.

WO2012139499 discloses urea substituted anilino-pyrimidines as inhibitors of various protein kinases. WO2014106762 discloses 4-pyrimidinylamino-benzenesulfonamide derivatives as inhibitors of polo-like kinase- 1.

Macrocyclic compounds have been described as therapeutically useful substances, in particular of various protein kinases including cyclin dependent kinases. However, the documents listed below do not disclose specific compounds as inhibitors of CDK9.

WO 2007147574 discloses sulfonamido-macrocycles as inhibitors of Tie2 showing selectivity over CDK2 and Aurora kinase C, inter alia for the treatment of diseases accompanied with dysregulated vascular growth.

WO 2007147575 discloses further sulfonamido-macrocycles as inhibitors of Tie2 and KDR showing selectivity over CDK2 and Plkl, inter alia for the treatment of diseases accompanied with dysregulated vascular growth.

WO 2006066957 / EP 1674470 discloses further sulfonamido-macrocycles as inhibitors of Tie2 showing low cytotoxicity, inter alia for the treatment of diseases accompanied with dysregulated vascular growth.

WO 2006066956 / EP 1674469 discloses further sulfonamido-macrocycles as inhibitors of Tie2 showing low cytotoxicity, inter alia for the treatment of diseases accompanied with dysregulated vascular growth.

WO 2004026881 / DE 10239042 discloses macrocyclic pyrimidine derivatives as inhibitors of cyclin dependent kinases, in particular CDKl and CDK2, as well as VEGF-R, inter alia for the treatment of cancer. The compounds of the present invention differ from those disclosed in WO 2004026881 in featuring a mandatory biaromatic portion within the macrocyclic ring system. Furthermore, none of the example compounds disclosed in WO 2004026881 features a group -CFh-A-R 1 , in which A and R 1 are as defined for the compounds of the formula (I) of the present invention, attached to one of the two aromatic portions of the macrocyclic ring system. WO 2007079982 / EP 1803723 discloses macrocyclic benzenacyclononaphanes as inhibtors of multiple protein kinases, e.g. Aurora kinases A and C, CDKl, CDK2 and c-Kit, inter alia for the treatment of cancer. The compounds of the present invention differ from those disclosed in WO 2007079982 in featuring a mandatory biaromatic portion within the macrocyclic ring system. Furthermore, the compounds of the present invention do not feature a group -S(=0)(N=R 2 )R 1 directly attached to the phenylene portion of the macrocyclic ring system as disclosed in WO 2007079982. WO 2006106895 / EP 1710246 discloses sulfoximine-macrocycle compounds as inhibitors of Tie2 showing low cytotoxicity, inter alia for the treatment of diseases accompanied with dysregulated vascular growth. WO 2012009309 discloses macrocyclic compounds fused to benzene and pyridine rings for the reduction of beta-amyloid production.

WO 2009132202 discloses macrocyclic compounds as inhibitors of JAK 1, 2 and 3, TYK2 and ALK and their use in the treatment of JAK/ALK-associated diseases, including inflammatory and autoimmune disease as well as cancer.

ChemMedChem 2007, 2(1), 63-77 describes macrocyclic aminopyrimidines as multitarget CDK and VEGF-R inhibitors with potent antiproliferative activity. The compounds of the present invention differ from those disclosed in said journal publication in featuring a mandatory biaromatic portion within the macrocyclic ring system. Furthermore, none of the compounds disclosed in ChemMedChem 2007, 2(1), 63-77 features a group -CH2-A-R 1 in which A and R 1 are as defined for the compounds of the formula (I) or the present invention, attached to one of the two aromatic portions of the macrocyclic ring system.

Despite the fact that various inhibitors of CDKs are known, there remains a need for selective CDK9 inhibitors, especially CDK9 inhibitors which are selective at high ATP concentrations, to be used for the treatment of diseases such as hyper-proliferative diseases, viral diseases, and/or diseases of the heart, which offer one or more advantages over the compounds known from prior art, such as :

• improved activity and / or efficacy, allowing e.g. a dose reduction

• improved side effect profile, such as fewer undesired side effects, lower intensity of side effects, or reduced (cyto)toxicity

• improved duration of action, e.g. by improved pharmacokinetics and / or improved target residence time

A particular object of the invention is to provide selective CDK9 kinase inhibitors, which show a high anti-proliferative activity in tumor cell lines, such as HeLa, HeLa-MaTu-ADR, NCI-H460, DU145, Caco-2, B16F10, A2780 or MOLM-13, compared to compounds known from prior art.

Another object of the invention is to provide selective CDK9 kinase inhibitors which show an increased potency to inhibit CDK9 activity (demonstrated by a lower IC50 value for CDK9/Cyclin Tl) compared to the compounds known from prior art. Another particular object of the invention is to provide selective CDK9 kinase inhibitors which show an increased potency to inhibit CDK9 activity at high ATP concentrations compared to compounds known from prior art. Another particular object of the invention is to provide selective CDK9 kinase inhibitors which show an increased target residence time compared to compounds known from prior art.

Another particular object of the invention is to provide selective CDK9 kinase inhibitors which show an improved duration of action, e.g. by improved pharmacokinetics and / or improved target residence time.

Further, it is an object of the present invention to provide selective CDK9 kinase inhibitors, which, compared to the compounds known from prior art, show a high anti-proliferative activity in tumor cell lines, such as HeLa, HeLa-MaTu-ADR, NCI-H460, DU145, Caco-2, B16F10, A2780 or MOLM-13, and/or which show an increased potency to inhibit CDK9 activity (demonstrated by a lower IC50 value for CDK9/Cyclin Tl), especially an increased potency to inhibit CDK9 activity at high ATP concentrations, and/or which show an increased target residence time compared to the compounds known from prior art.

The CDK9 kinase inhibitors according to the invention shall have simultaneously selectivity for CDK9/Cyclin Tl over CDK2/Cyclin E, especially at high ATP concentrations.

The present invention relates to compounds of general formula (I)

R

(I)

wherein A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-, -S(=0)(=NR 5 )-;

G, E represent, independently from each other, a bivalent moiety selected from the group consisting of-O-, -N(R A )-, -CH2-, -CH(Ci-C 6 -alkyl)-, -C(Ci-C 6 -alkyl) 2 -, -S-, -S(=0)-, -S(=0) 2 -, with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-C8-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, -NR 6 R 7 , C2-C3-alkenyl-, C2-C3-alkynyl-, C3-C4- cycloalkyl-, hydroxy-Ci-C 3 -alkyl, -(CH 2 )NR 6 R 7 , and/or

(ii) one or two or three or four substituents, identically or differently, selected from halogen and

Ci-C 3 -alkyl-,

or wherein

one carbon atom of said C2-C8-alkylene moiety forms a three- or four-membered ring together with a bivalent moiety to which it is attached, wherein said bivalent moiety is selected from - CH2CH2-, -CH2CH2CH2-, -CH2OCH2-;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a group selected from Ci-C6-alkyl-, C3-C6-alkenyl-, C3-C6-alkynyl-,

C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-, phenyl-Ci-C3-alkyl- and heteroaryl-Ci- C 3 -alkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C6-alkyl-, halo-Ci- C3-alkyl-, Ci-C6-alkoxy-, Ci-C3-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, -OP(=0)(OH) 2 , -C(=0)OH, -C(=0)NH 2 ; R 2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R 3 , R 4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R 5 represents a group selected from a hydrogen atom, cyano,

-C(=0)NR 6 R 7 , Ci-Ce-alkyl-, C 3 -C 7 -cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-, wherein said Ci-C6-alkyl-, C3-C 7 -cycloalkyl-, heterocyclyl-, phenyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R 6 , R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said Ci-C6-alkyl-, C3-C 7 -cycloalkyl-, heterocyclyl-, phenyl-, benzyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine;

R 8 represents a group selected from Ci-C6-alkyl-, halo-Ci-C3-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -ΝΗ2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-,

R A represents a hydrogen atom or a Ci-C6-alkyl- group, or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

Compounds according to the invention are the compounds of the formula (I) and the salts, solvates and solvates of the salts thereof, the compounds of the hereinafter recited formula which are encompassed by formula (I) and the salts, solvates and solvates of the salts thereof, and the compounds which are encompassed by formula (I) and are mentioned hereinafter as exemplary embodiments and the salts, solvates and solvates of the salts thereof, where the compounds which are encompassed by formula (I) and are mentioned hereinafter are not already salts, solvates and solvates of the salts.

The compounds according to the invention may, depending on their structure, exist in stereoisomeric forms (enantiomers, diastereomers). The invention therefore relates to the enantiomers or diastereomers and respective mixtures thereof. The stereoisomerically pure constituents can be isolated in a known manner from such mixtures of enantiomers and/or diastereomers. If the compounds according to the invention can be in tautomeric forms, the present invention encompasses all tautomeric forms.

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

Salts which are preferred for the purposes of the present invention are physiologically acceptable salts of the compounds according to the invention. However, salts which are not suitable for pharmaceutical applications per se, but which, for example, can be used for the isolation or purification of the compounds according to the invention, are also comprised.

The term "physiologically acceptable salt" refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention, for example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm Sci. 1977, 66, 1-19.

Physiologically acceptable salts of the compounds according to the invention encompass acid addition salts of mineral acids, carboxylic acids and sulfonic acids, for example salts of hydrochloric acid, hydrobromic acid, hydroiodic, sulfuric acid, bisulfuric acid, phosphoric acid, nitric acid or with an organic acid, such as formic, acetic, acetoacetic, pyruvic, trifluoroacetic, propionic, butyric, hexanoic, heptanoic, undecanoic, lauric, benzoic, salicylic, 2-(4-hydroxybenzoyl)-benzoic, camphoric, cinnamic, cyclopentanepropionic, digluconic, 3 -hydroxy-2 -naphthoic, nicotinic, pamoic, pectinic, persulfuric, 3-phenylpropionic, picric, pivalic, 2-hydroxyethanesulfonate, itaconic, sulfamic, trifluoromethanesulfonic, dodecylsulfuric, ethansulfonic, benzenesulfonic, para-toluenesulfonic, methansulfonic, 2-naphthalenesulfonic, naphthalinedisulfonic, camphorsulfonic acid, citric, tartaric, stearic, lactic, oxalic, malonic, succinic, malic, adipic, alginic, maleic, fumaric, D-gluconic, mandelic, ascorbic, glucoheptanoic, glycerophosphoric, aspartic, sulfosalicylic, hemisulfuric, or thiocyanic acid, for example. Physiologically acceptable salts of the compounds according to the invention also comprise salts of conventional bases, such as, by way of example and by preference, alkali metal salts (for example sodium and potassium salts), alkaline earth metal salts (for example calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines with 1 to 16 C atoms, such as, by way of example and by preference, ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, arginine, lysine, ethylenediamine, N-methylpiperidine, N-methylglucamine, dimethylglucamine, ethylglucamine, 1,6-hexadiamine, glucosamine, sarcosine, serinol, tris(hydroxymethyl)aminomethane, aminopropanediol, Sovak base, and l-amino-2,3,4- butanetriol. Additionally, the compounds according to the invention may form salts with a quartemary ammonium ion obtainable e.g. by quarternisation of a basic nitrogen containing group with agents such as lower alkylhalides such as methyl-, ethyl-, propyl-, and butylchlorides, -bromides and -iodides ; dialkylsulfates such as dimethyl-, diethyl-, dibutyl- and diamylsulfates, long chain halides such as decyl-, lauryl-, myristyl- and stearylchlorides, -bromides and -iodides, aralkylhalides such as benzyl- and phenethylbromides and others. Examples of suitable quartemary ammonium ions are tetramethylammonium, tetraethylammonium, tetra(«-propyl)ammonium, tetra (n-butyl)ammonium, or N-b enzy 1-N N N-trimethy lammonium.

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

Solvates is the term used for the purposes of the invention for those forms of the compounds according to the invention which form a complex with solvent molecules by coordination in the solid or liquid state. Hydrates are a special form of solvates in which the coordination takes place with water. Hydrates are preferred as solvates within the scope of the present invention.

The invention also includes all suitable isotopic variations of a compound of the invention. An isotopic variation of a compound of the invention is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually or predominantly found in nature. Examples of isotopes that can be incorporated into a compound of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine and iodine, such as 2 H (deuterium), 3 H (tritium), 13 C, 14 C, 15 N, 17 0, 18 0, 32 P, 33 P, 33 S, 34 S, 35 S, 36 S, 18 F, 36 C1, 82 Br, 123 I, 124 I, 129 I and 131 I, respectively. Certain isotopic variations of a compound of the invention, for example, those in which one or more radioactive isotopes such as 3 H or 14 C are incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. Isotopic variations of a compound of the invention can generally be prepared by conventional procedures known by a person skilled in the art such as by the illustrative methods or by the preparations described in the examples hereafter using appropriate isotopic variations of suitable reagents.

In addition, the present invention also encompasses prodrugs of the compounds according to the invention. The term "prodrugs" encompasses compounds which themselves may be biologically active or inactive, but are converted (for example by metabolism or hydrolysis) to compounds according to the invention during their residence time in the body.

Furthermore, the present invention includes all possible crystalline forms, or polymorphs, of the compounds of the present invention, either as single polymorphs, or as a mixture of more than one polymorphs, in any ratio.

Accordingly, the present invention includes all possible salts, polymorphs, metabolites, hydrates, solvates, prodrugs (e.g. : esters) thereof, and diastereoisomeric forms of the the compounds of the present invention as single salt, polymorph, metabolite, hydrate, solvate, prodrug (e.g. : esters) thereof, or diastereoisomeric form, or as mixture of more than one salt, polymorph, metabolite, hydrate, solvate, prodrug (e.g. : esters) thereof, or diastereoisomeric form in any ratio.

For the purposes of the present invention, the substituents have the following meaning, unless otherwise specified:

The term "halogen", "halogen atom" or "halo" represents fluorine, chlorine, bromine and iodine, particularly bromine, chlorine or fluorine, preferably chlorine or fluorine, more preferably fluorine. The term "alkyl-" represents a linear or branched alkyl- group having the number of carbon atoms specifically indicated, e.g. Ci-Cio one, two, three, four, five, six, seven, eight, nine or ten carbon atoms, e.g. methyl-, ethyl-, « -propyl-, isopropyl-, « -butyl-, isobutyl-, sec-butyl-, teri-butyl-, pentyl-, isopentyl-, hexyl-, heptyl-, octyl-, nonyl-, decyl-, 2-methylbutyl-, 1 -methylbutyl-, 1 -ethylpropyl-, 1 ,2-dimethylpropyl-, neo-pentyl-, 1 , 1 -dimethylpropyl-, 4-methylpentyl-, 3-methylpentyl-, 2-methylpentyl-, 1 -methylpentyl-, 2-ethylbutyl-, 1 -ethylbutyl-, 3,3-dimethylbutyl-, 2,2-dimethylbutyl-,

1.1 - dimethylbutyl-, 2,3-dimethylbutyl-, 1 ,3-dimethylbutyl-, or 1 ,2-dimethylbutyl-. If the number of carbon atoms is not specifically indicated, the term "alkyl-" represents a linear or branched alkyl- group having, as a rule, 1 to 9, particularly 1 to 6, preferably 1 to 4 carbon atoms. Particularly, the alkyl- group has 1 , 2, 3, 4, 5 or 6 carbon atoms ("Ci-C6-alkyl-"), e.g. methyl-, ethyl-, ^-propyl-, isopropyl-, n-butyl-, teri-butyl-, pentyl-, isopentyl-, hexyl-, 2-methylbutyl-, 1 -methylbutyl-, 1 -ethylpropyl-,

1.2- dimethylpropyl-, neo-pentyl-, 1 , 1 -dimethylpropyl-, 4-methylpentyl-, 3-methylpentyl-, 2-methylpentyl-, 1 -methylpentyl-, 2-ethylbutyl-, 1 -ethylbutyl-, 3,3-dimethylbutyl-, 2,2-dimethylbutyl-, 1 , 1 -dimethylbutyl-, 2,3-dimethylbutyl-, 1 ,3-dimethylbutyl-, or 1 ,2-dimethylbutyl-. Preferably, the alkyl- group has 1 , 2 or 3 carbon atoms ("Ci-C3-alkyl"), methyl-, ethyl-, ^-propyl- or isopropyl-.

The term "C2-C8-alkylene" is to be understood as preferably meaning a linear, bivalent and saturated hydrocarbon moiety having 2 to 8, particularly 2, 3, 4 or 5 carbon atoms, as in "C2-C5-alkylene", more particularly 2, 3 or 4 carbon atoms, as in "C2-C i-alkylene" e.g. ethylene, ^-propylene, n-butylene, n-pentylene, or n-hexylene, preferably ^-propylene or n-butylene.

The term "C 2 -C6-alkenyl-" is to be understood as preferably meaning a linear or branched, monovalent hydrocarbon group, which contains one double bond, and which has 2, 3, 4, 5 or 6 carbon atoms ("C2-C6- alkenyl-"). Particularly, said alkenyl group is a C 2 -C3-alkenyl-, C3-C6-alkenyl- or C3-C i-alkenyl- group. Said alkenyl- group is, for example, a vinyl-, allyl-, (£')-2-methylvinyl-, (Z)-2-methylvinyl- or isopropenyl- group. The term "C 2 -C6-alkynyl-" is to be understood as preferably meaning a linear or branched, monovalent hydrocarbon group which contains one triple bond, and which contains 2, 3, 4, 5 or 6 carbon atoms. Particularly, said alkynyl- group is a C 2 -C3-alkynyl-, C3-C6-alkynyl- or C3-C i-alkynyl- group. Said C 2 -C3-alkynyl- group is, for example, an ethynyl-, prop-l-ynyl- or prop-2-ynyl- group. The term "C3-C7-cycloalkyl-" is to be understood as preferably meaning a saturated or partially unsaturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4, 5, 6 or 7 carbon atoms. Said C3-C7-cycloalkyl- group is for example, a monocyclic hydrocarbon ring, e.g. a cyclopropyl-, cyclobutyl-, cyclopentyl-, cyclohexyl- or cycloheptyl- group. Said cycloalkyl- ring is non-aromatic but can optionally contain one or more double bonds e.g. cycloalkenyl-, such as a cyclopropenyl-, cyclobutenyl-, cyclopentenyl-, cyclohexenyl- or cycloheptenyl- group, wherein the bond between said ring with the rest of the molecule may be to any carbon atom of said ring, be it saturated or unsaturated. Particularly, said cycloalkyl- group is a C i-C6-cycloalkyl-, a Cs-Ce-cycloalkyl- or a cyclohexyl- group.

The term "C3-C5-cycloalkyl-" is to be understood as preferably meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3, 4 or 5 carbon atoms. In particular said C3-C5-cycloalkyl- group is a monocyclic hydrocarbon ring such as a cyclopropyl-, cyclobutyl- or cyclopentyl- group. Preferably said "C3-C5-cycloalkyl-" group is a cyclopropyl- group.

The term "C3-C4-cycloalkyl-" is to be understood as preferably meaning a saturated, monovalent, monocyclic hydrocarbon ring which contains 3 or 4 carbon atoms. In particular, said C3-C4-cycloalkyl- group is a monocyclic hydrocarbon ring such as a cyclopropyl- or cyclobutyl- group.

The term "heterocyclyl-" is to be understood as meaning a saturated or partially unsaturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8 or 9 carbon atoms and further containing 1, 2 or 3 heteroatom-containing groups selected from oxygen, sulfur, nitrogen. Particularly, the term "heterocyclyl-" is to be understood as meaning a "4- to 10-membered heterocyclic ring". The term "a 4- to 10-membered heterocyclic ring" is to be understood as meaning a saturated or partially unsaturated, monovalent, mono- or bicyclic hydrocarbon ring which contains 3, 4, 5, 6, 7, 8 or 9 carbon atoms, and further containing 1, 2 or 3 heteroatom-containing groups selected from oxygen, sulfur, nitrogen.

A C3-C9-heterocyclyl- is to be understood as meaning a heterocyclyl- which contains at least 3, 4, 5, 6, 7, 8 or 9 carbon atoms and additionally at least one heteroatom as ring atoms. Accordingly in case of one heteroatom the ring is 4- to 10-membered, in case of two heteroatoms the ring is 5- to 11 -membered and in case of three heteroatoms the ring is 6- to 12-membered. Said heterocyclic ring is for example, a monocyclic heterocyclic ring such as an oxetanyl-, azetidinyl-, tetrahydrofuranyl-, pyrrolidinyl-, 1,3-dioxolanyl-, imidazolidinyl-, pyrazolidinyl-, oxazolidinyl-, isoxazolidinyl-, 1 ,4-dioxanyl-, pyrrolinyl-, tetrahydropyranyl-, piperidinyl-, morpholinyl-, 1,3-dithianyl-, thiomorpholinyl-, piperazinyl-, or chinuclidinyl- group. Optionally, said heterocyclic ring can contain one or more double bonds, e.g. 4H-pyranyl-, 2H-pyranyl-, 2,5-dihydro-lH-pyrrolyl-, 1,3-dioxolyl-, 4H- 1,3,4-thiadiazinyl-, 2,5-dihydrofuranyl-, 2,3-dihydrofuranyl-, 2,5-dihydrothienyl-, 2,3-dihydrothienyl-, 4,5-dihydrooxazolyl-, 4,5-dihydroisoxazolyl-, or 4H-l,4-thiazinyl- group, or, it may be benzo fused.

Particularly, a C3-C7-heterocyclyl- is to be understood as meaning a heterocyclyl- which contains at least 3, 4, 5, 6, or 7 carbon atoms and additionally at least one heteroatom as ring atoms. Accordingly in case of one heteroatom the ring is 4- to 8-membered, in case of two heteroatoms the ring is 5- to 9-membered and in case of three heteroatoms the ring is 6- to 10-membered.

Particularly, a C3-C6-heterocyclyl- is to be understood as meaning a heterocyclyl- which contains at least 3, 4, 5 or 6 carbon atoms and additionally at least one heteroatom as ring atoms. Accordingly in case of one heteroatom the ring is 4- to 7-membered, in case of two heteroatoms the ring is 5- to 8-membered and in case of three heteroatoms the ring is 6- to 9-membered.

Particularly, the term "heterocyclyl-" is to be understood as being a heterocyclic ring which contains 3, 4 or 5 carbon atoms, and 1, 2 or 3 of the above-mentioned heteroatom-containing groups (a "4- to 8- membered heterocyclic ring"), more particularly said ring can contain 4 or 5 carbon atoms, and 1, 2 or 3 of the above-mentioned heteroatom-containing groups (a "5- to 8-membered heterocyclic ring"), more particularly said heterocyclic ring is a "6-membered heterocyclic ring", which is to be understood as containing 4 carbon atoms and 2 of the above-mentioned heteroatom-containing groups or 5 carbon atoms and one of the above-mentioned heteroatom-containing groups, preferably 4 carbon atoms and 2 of the above-mentioned heteroatom-containing groups. The term "Ci-C6-alkoxy-" is to be understood as preferably meaning a linear or branched, saturated, monovalent, hydrocarbon group of formula -O-alkyl-, in which the term "alkyl-" is defined supra, e.g. a methoxy-, ethoxy-, n-propoxy-, z ' o-propoxy-, n-butoxy-, z ' o-butoxy-, teri-butoxy-, eobutoxy-, pentyloxy-, z ' o-pentyloxy-, n-hexyloxy- group, or an isomer thereof. Particularly, the "Ci-C6-alkoxy-" group is a "Ci-C i-alkoxy-", a "Ci-C3-alkoxy-", a methoxy-, ethoxy-, or propoxy- group, preferably a methoxy-, ethoxy- or propoxy- group. Further preferred is a "Ci-C2-alkoxy-" group, particularly a methoxy- or ethoxy- group.

The term „Ci-C3-fluoroalkoxy-" is to be understood as preferably meaning a linear or branched, saturated, monovalent, Ci-C3-alkoxy- group, as defined supra, in which one or more of the hydrogen atoms is replaced, identically or differently, by one or more fluorine atoms. Said Ci-C3-fluoroalkoxy- group is, for example a 1 , 1 -difluoromethoxy-, a 1 , 1 , 1 -trifluoromethoxy-, a 2-fluoroethoxy-, a 3-fluoropropoxy-, a 2,2,2-trifluoroethoxy-, a 3,3,3-trifluoropropoxy-, particularly a "Ci-C2-fluoroalkoxy-" group.

The term„alkylamino-" is to be understood as preferably meaning an alkylamino group with one linear or branched alkyl- group as defined supra. (Ci-C3)-alkylamino- for example means a monoalkylamino- group with 1, 2 oder 3 carbon atoms, (Ci-C6)-alkylamino- with 1, 2, 3, 4, 5 or 6 carbon atoms. The term "alkylamino-" comprises for example methylamino-, ethylamino-, n-propylamino-, z o-propylamino-, tert-butylamino-, n-pentylamino- or n-hexylamino-.

The term„dialkylamino-" is to be understood as preferably meaning an alkylamino- group having two linear or branched alkyl- groups as defined supra, which are independent from each other. (Ci-C3)-dialkylamino- for example represents a dialkylamino- group with two alkyl- groups each of them having 1 to 3 carbon atoms per alkyl- group. The term "dialkylamino-" comprises for example: N,N-dimethylamino-, N,N-diethylamino-, N-ethyl-N-methylamino-, N-methyl-N-n-propylamino-, N-z o-propyl-N-«-propylamino-, N-tert-butyl-N-methylamino-, N-ethyl-N-n-pentylamino- and N-n-hexyl-N-methylamino-.

The term "cyclic amine" is to be understood as preferably meaning a cyclic amine group. Preferably, a cyclic amine means a saturated, monocyclic group with 4 to 10, preferably 4 to 7 ring atoms of which at least one ring atom is a nitrogen atom. Suitable cyclic amines are especially azetidine, pyrrolidine, piperidine, piperazine, 1 -methylpiperazine, morpholine, thiomorpholine, which could be optionally substituted by one or two methyl groups. The term "halo-Ci-C3-alkyl-", or, used synonymously, "Ci-C3-haloalkyl-", is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C3-alkyl-" is defined supra, and in which one or more hydrogen atoms is replaced by a halogen atom, identically or differently, i.e. one halogen atom being independent from another. Preferably, a halo-Ci-C3-alkyl- group is a fluoro-Ci-C3-alkyl- or a fluoro-Ci-C2-alkyl- group, such as for example -CF3, -CHF2, -CH2F, -CF2CF3, or -CH2CF3, more preferably it is -CF3. The term "hydroxy-Ci-C3-alkyl-", is to be understood as preferably meaning a linear or branched, saturated, monovalent hydrocarbon group in which the term "Ci-C3-alkyl-" is defined supra, and in which one or more hydrogen atoms is replaced by hydroxy group, preferably not more than one hydrogen atom per carbon atom being replaced by a hydroxy group. Particularly, a hydroxy-Ci-C3-alkyl- group is, for example, -CH 2 OH, -CH 2 -CH 2 OH, -C(H)OH-CH 2 OH, -CH 2 -CH 2 -CH 2 OH.

The term "phenyl-Ci-C3-alkyl-" is to be understood as preferably meaning a phenyl group, in which one of the hydrogen atoms is replaced by a Ci-C3-alkyl group, as defined supra, which links the phenyl-Ci-C3- alkyl- group to the rest of the molecule. Particularly, the "phenyl-Ci-C3-alkyl-" is a phenyl-Ci-C 2 -alkyl-, preferably it is a benzyl- group.

The term "heteroaryl-" is to be understood as preferably meaning a monovalent, aromatic ring system having 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms (a "5- to 14-membered heteroaryl-" group), particularly 5 (a "5-membered heteroaryl-") or 6 (a "6-membered heteroaryl-") or 9 (a"9-membered heteroaryl-") or 10 ring atoms (a "10-membered heteroaryl-"), and which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen or sulfur, and can be monocyclic, bicyclic, or tricyclic, and in addition in each case can be benzo- condensed. Particularly, heteroaryl- is selected from thienyl-, furanyl-, pyrrolyl-, oxazolyl-, thiazolyl-, imidazolyl-, pyrazolyl-, isoxazolyl-, isothiazolyl-, oxadiazolyl-, triazolyl-, thiadiazolyl-, tetrazolyl- etc., and benzo derivatives thereof, such as, for example, benzofuranyl-, benzothienyl-, benzoxazolyl-, benzisoxazolyl-, benzimidazolyl-, benzotriazolyl-, indazolyl-, indolyl-, isoindolyl-, etc.; or pyridyl-, pyridazinyl-, pyrimidinyl-, pyrazinyl-, triazinyl-, etc., and benzo derivatives thereof, such as, for example, quinolinyl-, quinazolinyl-, isoquinolinyl-, etc.; or azocinyl-, indolizinyl-, purinyl-, etc., and benzo derivatives thereof; or cinnolinyl-, phthalazinyl-, quinazolinyl-, quinoxalinyl-, naphthyridinyl-, pteridinyl-, carbazolyl-, acridinyl-, phenazinyl-, phenothiazinyl-, phenoxazinyl-, xanthenyl-, or oxepinyl-, etc. Preferably, heteroaryl- is selected from monocyclic heteroaryl-, 5-membered heteroaryl- or 6-membered heteroaryl-.

The term "5-membered heteroaryl-" is understood as preferably meaning a monovalent, aromatic ring system having 5 ring atoms and which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen or sulfur. Particularly, "5-membered heteroaryl-" is selected from thienyl-, furanyl-, pyrrolyl-, oxazolyl-, thiazolyl-, imidazolyl-, pyrazolyl-, isoxazolyl-, isothiazolyl-, oxadiazolyl-, triazolyl-, thiadiazolyl-, tetrazolyl-. The term "6-membered heteroaryl-" is understood as preferably meaning a monovalent, aromatic ring system having 6 ring atoms and which contains at least one heteroatom which may be identical or different, said heteroatom being such as oxygen, nitrogen or sulfur. Particularly, "6-membered heteroaryl-" is selected from pyridyl-, pyridazinyl-, pyrimidinyl-, pyrazinyl-, triazinyl-.

The term "heteroaryl-Ci-C3-alkyl-" is to be understood as preferably meaning a heteroaryl-, a 5-membered heteroaryl- or a 6-membered heteroaryl- group, each as defined supra, in which one of the hydrogen atoms is replaced by a Ci-C3-alkyl- group, as defined supra, which links the heteroaryl-Ci-C3- alkyl- group to the rest of the molecule. Particularly, the "heteroaryl-Ci-C3-alkyl-" is a heteroaryl-Ci-C2- alkyl-, a pyridinyl-Ci-C3-alkyl-, a pyridinylmethyl-, a pyridinylethyl-, a pyridinylpropyl-, a pyrimidinyl- Ci-C3-alkyl-, a pyrimidinylmethyl-, a pyrimidinylethyl-, a pyrimidinylpropyl-, preferably a pyridinylmethyl- or a pyridinylethyl- or a pyrimidinylethyl- or a pyrimidinylpropyl- group.

As used herein, the term "leaving group" refers to an atom or a group of atoms that is displaced in a chemical reaction as stable species taking with it the bonding electrons. Preferably, a leaving group is selected from the group comprising: halo, in particular chloro, bromo or iodo, methanesulfonyloxy-, p-toluenesulfonyloxy-, trifluoromethanesulfonyloxy-, nonafluorobutanesulfonyloxy-, (4-bromo- benzene)sulfonyloxy-, (4-nitro-benzene)sulfonyloxy-, (2-nitro-benzene)-sulfonyloxy-, (4-isopropyl- benzene)sulfonyloxy-, (2,4,6-tri-isopropyl-benzene)-sulfonyloxy-, (2,4,6-trimethyl-benzene)sulfonyloxy- , (4-fert-butyl-benzene)sulfonyloxy-, benzenesulfonyloxy-, and (4-methoxy-benzene)sulfonyloxy-.

As used herein, the term "Ci-C3-alkylbenzene" refers to a partially aromatic hydrocarbon consisting of a benzene ring which is substituted by one or two Ci-C3-alkyl- groups, as defined supra. Particularly, "Ci-C3-alkylbenzene" is toluene, ethylbenzene, cumene, n-propylbenzene, ori/zo-xylene, meia-xylene or para-xylene. Preferably, "Ci-C3-alkylbenzene" is toluene.

As used herein, the term "carboxamide based solvent" refers to lower aliphatic carboxamides of the formula Ci-C2-alkyl-C(=0)-N(Ci-C2-alkyl)2, lic aliphatic carboxamides of the formula

C r C 2 -alkyl in which G represents -CH2-, -CH2-CH2- or -CH2-CH2-CH2-. Particularly, "carboxamide based solvent" is N,N-dimethylformamide, NN-dimethylacetamide or N-methylpyrrolidin-2-one. Preferably, "carboxamide based solvent" is N,N-dimethylformamide or N-methyl-pyrrolidin-2-one. The term "Ci-Cio", as used throughout this text, e.g. in the context of the definition of "Ci-Cio-alkyl" is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 10, i.e. 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. It is to be understood further that said term "Ci-Cio" is to be interpreted as any sub-range comprised therein, e.g. Ci-Cio, C1-C9, Ci-Cs , C1-C7 , Ci-Ce C1-C5, C1-C4, Ci- C3, C1-C2, C2-C10, C2-C9, C2-C8, C2-C7, C2-C6, C2-C5, C2-C4, C2-C3, C3-C10, C3-C9, C3-C8, C3-C7, C3-C6, C3-C5, C3-C4, C4-C10, C4-C9, C4-C8, C4-C7, C4-C6, C4-C5, C5-C10, C5-C9, C5-C8, C5-C7, C5-C6, Ce-Cio, C6-C9,

C6-C8, C6-C7, C7-C10, C7-C9, C7-C8, C8-C10, C8-C9, C9-C10.

Similarly, as used herein, the term "Ci-Ce", as used throughout this text, e.g. in the context of the definition of "Ci-Ce-alkyl", "Ci-C6-alkoxy" is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 6, i.e. 1, 2, 3, 4, 5 or 6 carbon atoms. It is to be understood further that said term "Ci-Ce" is to be interpreted as any sub-range comprised therein, e.g. Ci-Ce C1-C5, C1-C4, C1-C3, C1-C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, C5-C6. Similarly, as used herein, the term "C1-C4", as used throughout this text, e.g. in the context of the definition of "Ci-C4-alkyl", "Ci-C4-alkoxy" is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 4, i.e. 1, 2, 3 or 4 carbon atoms. It is to be understood further that said term "C1-C4" is to be interpreted as any sub-range comprised therein, e.g. C1-C4, C1-C3, C1-C2, C2-

Similarly, as used herein, the term "C1-C3", as used throughout this text, e.g. in the context of the definition of "Ci-C3-alkyl", "Ci-C3-alkoxy" or "Ci-C3-fluoroalkoxy" is to be understood as meaning an alkyl group having a finite number of carbon atoms of 1 to 3, i.e. 1, 2 or 3 carbon atoms. It is to be understood further that said term "C1-C3" is to be interpreted as any sub-range comprised therein, e.g.

Further, as used herein, the term "C3-C6", as used throughout this text, e.g. in the context of the definition of "C3-C6-cycloalkyl", is to be understood as meaning a cycloalkyl group having a finite number of carbon atoms of 3 to 6, i.e. 3, 4, 5 or 6 carbon atoms. It is to be understood further that said term "C3-C6" is to be interpreted as any sub-range comprised therein, e.g. C3-C6 , C3-C5 , C3-C4 , C4-C6 , C4-C5 , C5-C6. Further, as used herein, the term "C3-C7", as used throughout this text, e.g. in the context of the definition of "C3-C7-cycloalkyl", is to be understood as meaning a cycloalkyl group having a finite number of carbon atoms of 3 to 7, i.e. 3, 4, 5, 6 or 7 carbon atoms, particularly 3, 4, 5 or 6 carbon atoms. It is to be understood further that said term "C3-C7" is to be interpreted as any sub-range comprised therein, e.g. C3- C7 , C3-C6 , C3-C5 , C3-C4 , C4-C7 , C4-C6 , C4-C5, C5-C7 , C5-C6, C6-C7.

A symbol ^ at a bond denotes the linkage site in the molecule. As used herein, the term "one or more times", e.g. in the definition of the substituents of the compounds of the general formulae of the present invention, is understood as meaning one, two, three, four or five times, particularly one, two, three or four times, more particularly one, two or three times, even more particularly one or two times.

Where the plural form of the word compounds, salts, hydrates, solvates and the like, is used herein, this is taken to mean also a single compound, salt, isomer, hydrate, solvate or the like.

In another embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-,

-S(=0)(=NR 5 )-;

G, E represent, independently from each other, a bivalent moiety selected from the group consisting of -O-, -N(R A )-, -CH2-, -CH(Ci-C 6 -alkyl)-, -C(Ci-C 6 -alkyl) 2 -, -S-, -S(=0)-, -S(=0) 2 -, with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-C8-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, -NR 6 R 7 , C2-C3-alkenyl-, C2-C3-alkynyl-, C3-C4- cycloalkyl-, hydroxy-Ci-C 3 -alkyl, -(CH 2 )NR 6 R 7 , and/or

(ii) one or two or three or four substituents, identically or differently, selected from halogen and Ci-Cs-alkyl-,

or wherein

one carbon atom of said C2-C6-alkylene moiety forms a three- or four-membered ring together with a bivalent moiety to which it is attached, wherein said bivalent moiety is selected from - CH2CH2-, -CH2CH2CH2-, -CH2OCH2-;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a group selected from Ci-C6-alkyl-, C3-C6-alkenyl-, C3-C6-alkynyl-,

C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-, phenyl-Ci-C3-alkyl- and heteroaryl-Ci- C 3 -alkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C6-alkyl-, halo-Ci- C3-alkyl-, Ci-C6-alkoxy-, Ci-C3-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, -OP(=0)(OH)¾ -C(=0)OH, -C(=0)NH 2 ;

R represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R 3 , R 4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R 5 represents a group selected from a hydrogen atom, cyano,

-C(=0)NR 6 R 7 , Ci-Ce-alkyl-, C 3 -C 7 -cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-,

wherein said Ci-C6-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-;

R 6 , R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said Ci-Ce-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -ΝΗ2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine;

R 8 represents a group selected from Ci-C6-alkyl-, halo-Ci-C3-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH 2 , alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-,

R A represents a hydrogen atom or a Ci-C6-alkyl- group, or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. In another embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-,

-S(=0)(=NR 5 )-;

G, E each represent, independently from each other, a bivalent moiety selected from the group consisting of-O-, -N(R A )-, -CH 2 -, -CH(Ci-C 3 -alkyl)-, -S-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-Cs-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, C3-C4-cycloalkyl-, hydroxy-Ci-C3-alkyl-, -(CH 2 )NR 6 R 7 , and/or

(ii) one or two or three additional substituents, identically or differently, selected from a fluorine atom and a Ci-C3-alkyl- group;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a group selected from Ci-C6-alkyl- and C3-Cs-cycloalkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C3-alkyl-, fluoro- Ci-C2-alkyl-, Ci-C3-alkoxy-, Ci-C2-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, -OP(=0)(OH) ¾ -C(=0)OH, -C(=0)NH 2 ;

R 2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, C1-C2- alkyl-, Ci-C 2 -alkoxy-, fluoro-Ci-C 2 -alkyl-;

R 3 , R 4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, Ci-C2-alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-;

R 5 represents a group selected from a hydrogen atom, cyano,

-C(=0)NR 6 R 7 , Ci-Ce-alkyl-, C 3 -C 5 -cycloalkyl-, phenyl-,

wherein said Ci-C6-alkyl-, C3-C5-cycloalkyl- or phenyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-;

R 6 , R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C3-C5-cycloalkyl-, phenyl- and benzyl-, wherein said Ci-C6-alkyl-, Cs-Cs-cycloalkyl-, phenyl- or benzyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C 2 -alkyl-, Ci-C 2 -fluoroalkoxy-, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine;

R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl-, Cs-Cs-cycloalkyl-, phenyl- and benzyl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-;

R A represents a hydrogen atom or a Ci-C3-alkyl- group, or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In a preferred embodiment, the present invention concerns compounds of general formula (I), wherein A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-,

-CH(Ci-C 3 -alkyl)-, -S-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-,

-CH(Ci-C 3 -alkyl)-, -S-, -S(=0) 2 -;

with the proviso that at least one of said bivalent moieties G and E is different from -0-; L represents a C2-Cs-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from C3-C4-cycloalkyl- and hydroxymethyl-, and/or

(ii) one or two additional substituents, identically or differently, selected from Ci-C 2 -alkyl-; X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a group selected from Ci-C i-alkyl- and C3-Cs-cycloalkyl-, wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, a fluorine atom, G-C 2 -alkyl-, Ci-C 2 -alkoxy-, -NH 2 , -C(=0)OH; represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-; represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-; represents a hydrogen atom or a fluorine atom; represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , -S(=0) 2 R 8 , -C(=0)NR 6 R 7 , Ci-C 4 -alkyl-,

wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of a fluorine atom, hydroxy, cyano, Ci-C3-alkoxy-, -NH 2 , alkylamino-, dialkylamino-, cyclic amines;

R 6 , R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C i-alkyl- and C3-C5-cycloalkyl-,

wherein said Ci-C i-alkyl- or Cs-Cs-cycloalkyl- group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C 2 -alkyl-, Ci-C 2 -alkoxy-, -NH 2 , alkylamino-, dialkylamino-, cyclic amines, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine;

R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl-, Cs-Cs-cycloalkyl- and benzyl-, wherein said group is optionally substituted with one substituent selected from the group consisting of halogen, hydroxy, Ci-C 2 -alkyl-, Ci-C 2 -alkoxy-, -NH 2 ; R A represents a hydrogen atom or a Ci-C3-alkyl- group, or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. In another preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -S-; with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-Cs-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a Ci-C i-alkyl- group,

wherein said group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkoxy-, -NH2, -C(=0)OH;

R 2 represents a hydrogen atom or a fluorine atom;

R 3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 ,

-C(=0)OR 8 , Ci-C 4 -alkyl-,

wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of hydroxy, cyano, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-;

R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl- and benzyl-;

R A represents a hydrogen atom, a methyl- or an ethyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In another preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C2-C i-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N; R 1 represents a Ci-C i-alkyl- group,

wherein said group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkoxy-, -NH2, -C(=0)OH;

R 2 represents a hydrogen atom;

R 3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 ,

-C(=0)OR 8 , Ci-C 4 -alkyl-,

wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of hydroxy, cyano, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-;

R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl- and benzyl-;

R A represents a hydrogen atom, a methyl- or an ethyl- group, or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In another preferred embodiment, the present invention concerns compounds of general formula (I), wherein A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, -N(H)-, -S-; with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-Cs-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a Ci-C3-alkyl- group;

R 2 represents a hydrogen atom or a fluorine atom;

R 3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 ,

-C(=0)OR 8 , Ci-C 3 -alkyl-;

R 8 represents a group selected from Ci-C4-alkyl-, trifluoromethyl- and benzyl-;

R A represents a hydrogen atom or a methyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. In another preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety -S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(H) -;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-C i-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a Ci-C3-alkyl- group;

R 2 represents a hydrogen atom;

R 3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R 4 represents a hydrogen atom;

R 5 represents a hydrogen atom;

R A represents a hydrogen atom or a methyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In another preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-C i-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a Ci-C3-alkyl- group;

R 2 represents a hydrogen atom;

R 3 represents a group selected from a hydrogen atom, a fluorine atom and a methoxy- group;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 ,

-C(=0)OR 8 , Ci-C 3 -alkyl-;

R 8 represents a group selected from Ci-C i-alkyl-, trifluoromethyl- and benzyl-;

R A represents a hydrogen atom or a methyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. In a particularly preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, -N(H)-, -S-; with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-Cs-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a methyl- group;

R 2 represents a hydrogen atom or a fluorine atom;

R 3 represents a hydrogen atom or a fluorine atom;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, -C(=0)OR 8 ;

R 8 represents a group selected from tert-butyl- and benzyl-;

R A represents a hydrogen atom or a methyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In a particularly preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety -S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(H)-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-C i-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a methyl- group;

R 2 represents a hydrogen atom;

R 3 represents a hydrogen atom or a fluorine atom;

R 4 represents a hydrogen atom;

R 5 represents a hydrogen atom;

R A represents a hydrogen atom or a methyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. In another particularly preferred embodiment, the present invention concerns compounds of general formula

(I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a C3-C i-alkylene moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a methyl- group;

R 2 represents a hydrogen atom;

R 3 represents a hydrogen atom or a fluorine atom;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, -C(=0)OR 8 ;

R 8 represents a group selected from tert-butyl- and benzyl-;

R A represents a hydrogen atom or a methyl- group,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In another particularly preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(H)-, -N(CH3)-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, -N(H)-, -S-; with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a -CH 2 CH 2 CH 2 -, -CH2CH2CH2CH2- or a -CH2CH2CH2CH2CH2- moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a methyl- group;

R 2 represents a hydrogen atom or a fluorine atom;

R 3 represents a fluorine atom, wherein R 3 is attached in /jara-position to the ring directly bonded to the phenyl- ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N;

R 4 represents a hydrogen atom; R 5 represents a group selected from a hydrogen atom, -C(=0)OC(CH3)3,

-C(=0)OCH 2 Ph,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. In another particularly preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety -S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(H)-, -Ν(0¾)-;

G represents a bivalent moiety selected from the group consisting of -0-, -N(H)-;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a -CH 2 CH 2 CH 2 - or a -CH 2 CH 2 CH 2 CH 2 - moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a methyl- group;

R 2 represents a hydrogen atom;

R 3 represents a fluorine atom, wherein R 3 is attached in /jara-position to the ring directly bonded to the phenyl- ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N;

R 4 represents a hydrogen atom;

R 5 represents a hydrogen atom,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In another particularly preferred embodiment, the present invention concerns compounds of general formula (I), wherein

A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-,

-S(=0)(=NR 5 )-;

E represents a bivalent moiety selected from the group consisting of -0-, -N(H)-, -N(CH3)-;

G represents a bivalent moiety selected from the group consisting of -0-, -CH 2 -;

with the proviso that at least one of said bivalent moieties G and E is different from -0-;

L represents a -CH 2 CH 2 CH 2 - or a -CH 2 CH 2 CH 2 CH 2 - moiety;

X, Y represent CH or N with the proviso that one of X and Y represents CH and one of X and Y represents N;

R 1 represents a methyl- group;

R 2 represents a hydrogen atom; R represents a fluorine atom, wherein R is attached in /jara-position to the ring directly bonded to the phenyl- ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N;

R 4 represents a hydrogen atom;

R 5 represents a group selected from a hydrogen atom, -C(=0)OC(CH3)3,

-C(=0)OCH 2 Ph,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

In another embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)2-, -S(=0)(=NR 5 )-.

In another embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety selected from the group consisting of -S(=0)2-, -S(=0)(=NR 5 )-.

In another embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)2-.

In a preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety selected from the group consisting of -S-, -S(=0)-, -S(=0)(=NR 5 )-.

In another preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S-. In another preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)-.

In another preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)(=NR 5 )-.

In another embodiment the invention relates to compounds of formula (I), in which G and E, independently from each other, represent a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -CH(Ci-C 6 -alkyl)-, -C(Ci-C 6 -alkyl) 2 -, -S-, -S(=0)-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-. In another embodiment the invention relates to compounds of formula (I), in which G and E, independently from each other, represent a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -CH(Ci-C 3 -alkyl)-, -S-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(R A )-, -CH2-, -CH(Ci-C3-alkyl)-, -S-, and in which G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -CH(Ci-C 3 -alkyl)-, -S-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In a preferred embodiment the invention relates to compounds of formula (I), in which G and E, independently from each other, represent a bivalent moiety selected from the group consisting of -0-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another preferred embodiment the invention relates to compounds of formula (I), in which G and E, independently from each other, represent a bivalent moiety selected from the group consisting of -0-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(R A )-,

and in which G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, -N(H)-, -S-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(R A )-,

and in which G represents a bivalent moiety selected from the group consisting of-O-, -CH2-, with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(H)-, -Ν(0¾)-,

and in which G represents a bivalent moiety selected from the group consisting of -O-, -CH2-, -N(H)-, -S-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of -0-, -N(H)-, -Ν(0¾)-, and in which G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, with the proviso that at least one of said bivalent moieties G and E is different from -0-.

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

E represents a bivalent moiety -N(H)-,

and in which G represents a bivalent moiety -0-. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -Ν(0¾)-,

and in which G represents a bivalent moiety -0-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -0-,

and in which G represents a bivalent moiety -N(H)-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -Ν(0¾)-,

and in which G represents a bivalent moiety -N(H)-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)(=NR 5 )-,

in which E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-,

in which G represents a bivalent moiety selected from the group consisting of -0-, -N(H)-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-, in which R A represents a hydrogen atom or a methyl- group,

and in which R 5 represents a hydrogen atom. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)(=NR 5 )-,

in which E represents a bivalent moiety -N(H)-,

in which G represents a bivalent moiety -0-,

and in which R 5 represents a hydrogen atom. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)(=NR 5 )-,

in which E represents a bivalent moiety -Ν(0¾)-,

in which G represents a bivalent moiety -0-,

and in which R 5 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -0-,

in which G represents a bivalent moiety -N(H)-,

and in which R 5 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which A represents a bivalent moiety -S(=0)(=NR 5 )-,

in which E represents a bivalent moiety -Ν(0¾)-,

in which G represents a bivalent moiety -N(H)-,

and in which R 5 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -0-,

and in which G represents a bivalent moiety -CH2 -.

In another embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -CH(Ci-C6-alkyl)-, -C(Ci-C 6 -alkyl) 2 -, -S-, -S(=0)-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -CH(Ci-C6-alkyl)-, -C(Ci-C 6 -alkyl) 2 -, -S-, -S(=0)-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH 2 -, -CH(Ci-C3-alkyl)-, -S-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-. In another embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(R A )-, -CH2-, -CH(Ci-C3-alkyl)-, -S-, with the proviso that at least one of said bivalent moieties G and E is different from -0-. In a preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(R A )-, -CH2-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(R A )-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety selected from the group consisting of-O-, -N(H)-, -Ν(0¾)-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -N(H)-. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -Ν(0¾)-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which E represents a bivalent moiety -0-,

with the proviso that the bivalent moiety G is different from -0-.

In another embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH 2 -, -CH(Ci-C6-alkyl)-, -C(Ci-C 6 -alkyl) 2 -, -S-, -S(=0)-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -CH(Ci-C3-alkyl)-, -S-, -S(=0) 2 -,

with the proviso that at least one of said bivalent moieties G and E is different from -0-. In a preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-, -S-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-. In another preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety selected from the group consisting of -0-, -N(R A )-, -CH2-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety selected from the group consisting of -0-, -CH2-, -N(H)-, -S-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety selected from the group consisting of -0-, -CH2-,

with the proviso that at least one of said bivalent moieties G and E is different from -0-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety -0-,

with the proviso that the bivalent moiety E is different from -0-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety-CH2-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety-N(H)-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which G represents a bivalent moiety-S-. In another embodiment the invention relates to compounds of formula (I), in which L represents a C2-C8-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, -NR 6 R 7 , C 2 -C3-alkenyl-, C 2 -C3-alkynyl-, C3-C4-cycloalkyl-, hydroxy-Ci-Cs-alkyl-, -(CH 2 )NR 6 R 7 , and/or

(ii) one or two or three or four substituents, identically or differently, selected from halogen and Ci-C 3 -alkyl-,

or wherein one carbon atom of said C2-C8-alkylene moiety forms a three- or four-membered ring together with a bivalent moiety to which it is attached, wherein said bivalent moiety is selected from -CH2CH2-, -CH2CH2CH2-, -CH2OCH2-. In another embodiment the invention relates to compounds of formula (I), in which L represents a C2-C5-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from hydroxy, C3-C4-cycloalkyl-, hydroxy-Ci-C3-alkyl-, -(CH 2 )NR 6 R 7 , and/or

(ii) one or two or three additional substituents, identically or differently, selected from a fluorine atom and a Ci-C3-alkyl- group.

In a preferred embodiment the invention relates to compounds of formula (I), in which L represents a C2-C5-alkylene moiety,

wherein said moiety is optionally substituted with

(i) one substituent selected from C3-C4-cycloalkyl- and hydroxymethyl-, and/or

(ii) one or two additional substituents, identically or differently, selected from Ci-C 2 -alkyl-.

In another preferred embodiment the invention relates to compounds of formula (I), in which L represents a C2-Cs-alkylene moiety, wherein said moiety is optionally substituted with one or two methyl- groups.

In another preferred embodiment the invention relates to compounds of formula (I), in which L represents a C2-C i-alkylene moiety, wherein said moiety is optionally substituted with one or two methyl- groups.

In a another preferred embodiment the invention relates to compounds of formula (I), in which L represents a C2-Cs-alkylene moiety. In a another preferred embodiment the invention relates to compounds of formula (I), in which L represents a C2-C i-alkylene moiety.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which L represents a C3-Cs-alkylene moiety.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which L represents a C3-C i-alkylene moiety. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which L represents a moiety -CH 2 CH 2 CH 2 -, -CH2CH2CH2CH2- or -CH2CH2CH2CH2CH2-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which L represents a moiety -CH2CH2CH2-.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which L represents a moiety -CH2CH2CH2CH2-. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which L represents a moiety -CH2CH2CH2CH2CH2-.

In another embodiment the invention relates to compounds of formula (I), in which X represents N, and in which Y represents CH.

In another embodiment the invention relates to compounds of formula (I), in which X represents CH, and in which Y represents N.

In another embodiment the invention relates to compounds of formula (I), in which R 1 represents a group selected from Ci-C6-alkyl-, C3-C6-alkenyl-, C3-C6-alkynyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-, phenyl-Ci-C3-alkyl- and heteroaryl-Ci-C3-alkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C6-alkyl-, halo-Ci-C3-alkyl-, Ci-Ce- alkoxy-, Ci-C3-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, -OP(=0)(OH) ¾ -C(=0)OH, -C(=0)NH 2 .

In another embodiment the invention relates to compounds of formula (I), in which R 1 represents a group selected from Ci-C6-alkyl- and C3-C5-cycloalkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, halogen, Ci-C3-alkyl-, fluoro-Ci-C2-alkyl-, C1-C3- alkoxy-, Ci-C2-fluoroalkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, -OP(=0)(OH)2, -C(=0)OH, -C(=0)NH 2 .

In a preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a group selected from Ci-C i-alkyl- and C3-C5-cycloalkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, a fluorine atom, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2. In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a group selected from Ci-C i-alkyl- and C3-Cs-cycloalkyl-,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, a fluorine atom, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2, -C(=0)OH.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C i-alkyl- group,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, cyano, a fluorine atom, Ci-C2-alkoxy-, -NH2, -C(=0)OH.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C i-alkyl- group,

wherein said group is optionally substituted with one or two or three substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkoxy-, -NH2, -C(=0)OH.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C i-alkyl- group. In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C3-alkyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C2-alkyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- or an ethyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents an ethyl- group.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group. In a preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C i-alkyl- group, and R 2 represents a hydrogen atom or a fluorine atom. In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a Ci-C i-alkyl- group, and R 2 represents a hydrogen atom.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group, and R 2 represents a hydrogen atom or a fluorine atom.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group, and R 2 represents a hydrogen atom. In another embodiment the invention relates to compounds of formula (I), in which R 2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-.

In another embodiment the invention relates to compounds of formula (I), in which R 2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, Ci-C2-alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C 2 -alkyl-.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 2 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 2 represents a hydrogen atom or a fluorine atom. In another preferred embodiment the invention relates to compounds of formula (I), in which R 2 represents a fluorine atom.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 2 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 2 represents a hydrogen atom, R 3 represents a fluorine atom, and R 4 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group, R 2 represents a hydrogen atom, R 3 represents a fluorine atom, and R 4 represents a hydrogen atom. In another embodiment the invention relates to compounds of formula (I), in which R and R represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-. In another embodiment the invention relates to compounds of formula (I), in which R 3 and R 4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C2-alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-.

In another embodiment the invention relates to compounds of formula (I), in which R 3 and R 4 represent, independently from each other, a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-.

In another embodiment the invention relates to compounds of formula (I), in which R 3 and R 4 represent, independently from each other, a hydrogen atom, a fluorine atom or a methoxy- group.

In another embodiment the invention relates to compounds of formula (I), in which R 3 and R 4 represent, independently from each other, a hydrogen atom or a fluorine atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-, and in which R 4 represents a hydrogen atom or a fluorine atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano Ci-C 2 -alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-, and in which R 4 represents a hydrogen atom or a fluorine atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, a bromine atom, cyano Ci-C 2 -alkyl-, Ci-C2-alkoxy-, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-, and in which R 4 represents a hydrogen atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-, and in which R 4 represents a hydrogen atom. In another embodiment the invention relates to compounds of formula (I), in which R represents a hydrogen atom, a fluorine atom or a methoxy- group, and in which R 4 represents a hydrogen atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a hydrogen atom or a fluorine atom, and in which R 4 represents a hydrogen atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a methoxy- group, and in which R 4 represents a hydrogen atom. In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-, and in which R 4 represents a hydrogen atom,

wherein R 3 is attached in para-position to the ring directly bonded to the phenyl-ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a hydrogen atom or a fluorine atom, and in which R 4 represents a hydrogen atom,

wherein R 3 is attached in /jara-position to the ring directly bonded to the phenyl-ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a fluorine atom, and in which R 4 represents a hydrogen atom,

wherein R 3 is attached in /jara-position to the ring directly bonded to the phenyl-ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a fluorine atom, and in which R 4 represents a hydrogen atom.

In another embodiment the invention relates to compounds of formula (I), in which R 3 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a hydrogen atom or a fluorine atom.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a hydrogen atom, a fluorine atom or a methoxy- group. In another preferred embodiment the invention relates to compounds of formula (I), in which R represents a hydrogen atom.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a fluorine atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a fluorine atom,

wherein R 3 is attached in /jara-position to the ring directly bonded to the phenyl-ring to which R 3 is attached, which is a pyridine ring if Y represents CH and a pyrimidine ring if Y represents N.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 4 represents a group selected from a hydrogen atom, a fluorine atom, a chlorine atom, cyano, methyl-, methoxy-, trifluoromethyl-, trifluoromethoxy-.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 4 represents a hydrogen atom or a fluorine atom.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 4 represents a fluorine atom.

In aparticularly preferred embodiment the invention relates to compounds of formula (I), in which R 4 represents a hydrogen atom. In another embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , -S(=0) 2 R 8 , -C(=0)NR 6 R 7 , Ci-Ce-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, heteroaryl-,

wherein said Ci-C6-alkyl, C3-C7-cycloalkyl-, heterocyclyl-, phenyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, Ci-C3-fluoroalkoxy-.

In another embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , -S(=0) 2 R 8 , -C(=0)NR 6 R 7 , G-Ce-alkyl-, C3-C5-cycloalkyl-, phenyl-,

wherein said Ci-C6-alkyl-, C3-C5-cycloalkyl- or phenyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, cyano, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, C1-C2- fluoroalkoxy-.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , -S(=0) 2 R 8 , -C(=0)NR 6 R 7 , Ci-C 4 -alkyl-, wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of a fluorine atom, hydroxy, cyano, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines. In another preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , Ci-C i-alkyl-,

wherein said Ci-C i-alkyl- group is optionally substituted with one substituent selected from the group consisting of hydroxy, cyano, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-. In another preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)NR 6 R 7 , -C(=0)R 8 , -C(=0)OR 8 , -S(=0) 2 R 8 , Ci-C 4 -alkyl-.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , Ci-C 4 -alkyl-.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, cyano, -C(=0)R 8 , -C(=0)OR 8 , Ci-C 3 -alkyl-.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, -C(=0)OR 8 . In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a -C(=0)OR 8 group.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from a hydrogen atom, -C(=0)OC(CH3)3, -C(=0)OCH2Ph.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a hydrogen atom. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group selected from -C(=0)OC(CH 3 ) 3 , -C(=0)OCH 2 Ph.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group -C(=0)OC(CH 3 ) 3 .

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a group -C(=0)OCH 2 Ph. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 5 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 3 represents a fluorine atom, R 4 represents a hydrogen atom, and R 5 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group, R 3 represents a fluorine atom, R 4 represents a hydrogen atom, and R 5 represents a hydrogen atom. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group, R 2 represents a hydrogen atom, R 3 represents a fluorine atom, R 4 represents a hydrogen atom, and R 5 represents a hydrogen atom.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R 1 represents a methyl- group and R 5 represents a hydrogen atom.

In another embodiment the invention relates to compounds of formula (I), in which R 6 and R 7

represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C 3 -C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said Ci-C6-alkyl-, C 3 -C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- or heteroaryl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C 3 -alkyl-, Ci-C 3 -alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C 3 -alkyl-, Ci-C 3 -fluoroalkoxy-, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine. In another embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C6-alkyl-, C3-C5-cycloalkyl-, phenyl- and benzyl-,

wherein said Ci-C6-alkyl-, Cs-Cs-cycloalkyl-, phenyl- or benzyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, C1-C2- fluoroalkoxy-, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine. In another embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom, Ci-C6-alkyl-, Cs-Cs-cycloalkyl-, phenyl- and benzyl-,

wherein said Ci-C6-alkyl-, Cs-Cs-cycloalkyl-, phenyl- or benzyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, C1-C2- fluoroalkoxy-, and in which R 7 represents a hydrogen atom or a C1-C3 alkyl- group, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine.

In another embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom, Ci-C6-alkyl- and phenyl-,

wherein said Ci-Ce-alkyl- or phenyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, C1-C3- alkoxy-, dialkylamino-, and in which R 7 represents a hydrogen atom or a C1-C3 alkyl- group, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine. In another embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom, Ci-C6-alkyl- and phenyl-,

wherein said Ci-Ce-alkyl- or phenyl- group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, C1-C3- alkoxy-, dialkylamino-, and in which R 7 represents a hydrogen atom or a C1-C3 alkyl- group.

In another embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C i-alkyl- and C3- C5-cycloalkyl-, wherein said Ci-C i-alkyl- or Cs-Cs-cycloalkyl- group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom, Ci-C i-alkyl- and Cs-Cs-cycloalkyl-,

wherein said Ci-C4-alkyl- or Cs-Cs-cycloalkyl- group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines,

and in which R 7 represents a hydrogen atom or a C1-C3 alkyl- group, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom, Ci-C4-alkyl- and Cs-Cs-cycloalkyl-,

wherein said Ci-C4-alkyl- or Cs-Cs-cycloalkyl- group is optionally substituted with one or two substituents, identically or differently, selected from the group consisting of hydroxy, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines,

and in which R 7 represents a hydrogen atom or a C1-C3 alkyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 represent, independently from each other, a group selected from a hydrogen atom and Ci-C4-alkyl-, wherein said Ci-C4-alkyl- group is optionally substituted with one substituent selected from the group consisting of hydroxy, Ci-C2-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom and Ci-C4-alkyl-,

wherein said Ci-C4-alkyl- group is optionally substituted with one substituent selected from the group consisting of hydroxy, Ci-C2-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines,

and in which R 7 represents a hydrogen atom or a C1-C3 alkyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C4-alkyl- and C3- C5-cycloalkyl-, or

R 6 and R 7 , together with the nitrogen atom they are attached to, form a cyclic amine In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 represent, independently from each other, a group selected from a hydrogen atom, Ci-C i-alkyl- and C3- Cs-cycloalkyl-. In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 6 and R 7 represent, independently from each other, a group selected from a hydrogen atom and Ci-C 2 -alkyl-.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 6 represents a group selected from a hydrogen atom and Ci-C2-alkyl-, and in which R 7 represents a hydrogen atom.

In another embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from Ci-C6-alkyl-, halo-Ci-C3-alkyl-, C3-C7-cycloalkyl-, heterocyclyl-, phenyl-, benzyl- and heteroaryl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, acetylamino-, N-methyl-N-acetylamino-, cyclic amines, halo-Ci-C3-alkyl-, C1-C3- fluoroalkoxy-.

In another embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl-, C3-C5-cycloalkyl-, phenyl- and benzyl-,

wherein said group is optionally substituted with one, two or three substituents, identically or differently, selected from the group consisting of halogen, hydroxy, Ci-C3-alkyl-, Ci-C3-alkoxy-, -NH2, alkylamino-, dialkylamino-, cyclic amines, fluoro-Ci-C2-alkyl-, Ci-C2-fluoroalkoxy-.

In a preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl-, C3-C5-cycloalkyl- and benzyl-,

wherein said group is optionally substituted with one substituent selected from the group consisting of halogen, hydroxy, Ci-C2-alkyl-, Ci-C2-alkoxy-, -NH2.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from Ci-C6-alkyl-, fluoro-Ci-C3-alkyl- and benzyl-. In another preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from Ci-C i-alkyl-, trifluoromethyl- and benzyl-. In another preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from teri-butyl-, trifluoromethyl- and benzyl-.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a Ci-C i-alkyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a trifluoromethyl- group. In another preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a benzyl- group.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a group selected from teri-butyl- and benzyl-.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a teri-butyl- group.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R 8 represents a benzyl- group.

In another embodiment the invention relates to compounds of formula (I), in which R A represents a hydrogen atom or a Ci-C6-alkyl- group. In another embodiment the invention relates to compounds of formula (I), in which R A represents a hydrogen atom or a Ci-C i-alkyl- group.

In a preferred embodiment the invention relates to compounds of formula (I), in which R A represents a hydrogen atom or a Ci-C3-alkyl- group.

In another preferred embodiment the invention relates to compounds of formula (I), in which R A represents a hydrogen atom, a methyl- or an ethyl- group.

In a particularly preferred embodiment the invention relates to compounds of formula (I), in which R A represents a hydrogen atom or a methyl- group.

In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R A represents a hydrogen atom. In another particularly preferred embodiment the invention relates to compounds of formula (I), in which R A represents a methyl- group.

It is to be understood that the present invention relates to any sub-combination within any embodiment of the present invention of compounds of formula (I), supra.

More particularly still, the present invention covers compounds of formula (I) which are disclosed in the Example section of this text, infra. Very specially preferred are combinations of two or more of the abovementioned preferred embodiments.

In particular, a preferred subject of the present invention is a compound selected from:

15,19-difluoro-8- [(methylsulfanyl)methyl]-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 12,16-(metheno)- 1,5,11,13-benzoxatriazacyclooctadecine;

- (rac)-benzyl [{[16,20-difluoro-3,4,5,6-tetrahydro-2H,12H-l l,7-(azeno)-13,17-(metheno)-l,6,12,14- benzoxatriazacyclononadecin-9-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate;

{rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-3,4,5,6-tetrahydro-2H, 12H- 11 ,7-(azeno)-

13.17- (metheno)- 1,6,12,14-benzoxatriazacyclononadecine;

- 2, 18-difluoro-9-[(methylsulfanyl)methyl]-l 3, 14, 15, 16-tetrahydro-6H-7, 11 -(azeno)-5, 1 -(metheno)- 12,4,6-benzoxadiazacyclooctadecine;

- (rac)-2, 18-difluoro-9- [(methylsulfmyl)methyl]- 13, 14,15,16-tetrahydro-6H-7, 11 -(azeno)-5, 1 - (metheno)- 12,4,6-benzoxadiazacyclooctadecine;

- (rac)-tert-butyl [{[16,20-difluoro-6-methyl-3,4,5,6-tetrahydro-2H,12H-13,17-( azeno)-l 1,7- (metheno)- 1 ,6,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl) oxido-λ 6 - sulfanylidene]carbamate;

(rac)-tert-butyl [{[15,19-difluoro-2,3,4,5-tetrahydro-l lH-10,6-(azeno)-16,12-(metheno)-l,5,l 1,13- benzoxatriazacyclooctadecin-8-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate;

{rac)- 15,19-difluoro-8- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 16,12-(metheno)-l,5,l 1,13-benzoxatriazacyclooctadecine;

(rac)-tert-butyl [{[17,21-difluoro-2,3,4,5,6,7-hexahydro-13H-12,8-(azeno)-14, 18-(metheno)- 1,7,13,15-benzoxatriazacycloicosin- 10-yl]methyl} (methyl)oxido^ 6 -sulfanylidene]carbamate;

(rac)-17,21-difluoro-10-[(S-methylsulfonimidoyl)methyl]-2,3, 4,5,6,7-hexahydro-13H-12,8-(azeno)-

14.18- (metheno)- 1,7,13,15-benzoxatriazacycloicosine (rac)-16,20-difluoro-6-methyl-9-[(S-methylsulfo^

(azeno)- 17, 13-(metheno)- 1 ,6,12,14-benzoxatriazacyclononadecine

8,16,20-trifluoro-6-methyl-9-[(methylsulfanyl)methyl]-3,4,5, 6-tetra^

13,17-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine

- (rac)-fert-butyl [{[15,19-difluoro-l,2,3,4-tetrahydro-16,12-(azeno)-6,10-(met heno)-5,l,l 1,13- benzoxatriazacyclooctadecin-8(l lH)-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate;

(rac)- 15,19-difluoro-8- [(S-methylsulfonimidoyl)methyl]- 1 ,2,3,4-tetrahydro- 16,12-(azeno)-6, 10- (metheno)-5, 1,11, 13-benzoxatriazacyclooctadecine;

(rac)-tert-butyl [{[16,20-difluoro-2,3,4,5-tetrahydro-lH,12H-13,17-(azeno)-l l,7-(metheno)- 6,1,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl)oxido^ 6 -sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 1 H- 17, 13-(azeno)-7, 11 - (metheno)-6, 1,12,14-benzoxatriazacyclononadecine;

(rac)- 16,20-difluoro-6-methyl-9-[(S-methylsulfonimidoyl)methyl] - 1 ,2,3,4,5, 6-hexahydro- 12H- 17, 13 -(azeno)- 1 l,7-(metheno)-l,6,12,14-benzotetraazacyclononadecine; salt with formic acid; - 2,19-difluoro-9-[(methylsulfanyl)methyl]-14,15,16,17-tetrahy dro-6H,13H-7,l l-(azeno)-5,l- (metheno)- 12,4,6-benzoxadiazacyclononadecine;

(rac)-2, 19-difluoro-9- [(methylsulfmyl)methyl]- 14,15,16,17-tetrahydro-6H, 13H-7, 11 -(azeno)-5, 1 - (metheno)- 12,4,6-benzoxadiazacyclononadecine;

(rac)-2, 19-difluoro-9- [(S-methylsulfonimidoyl)methyl] -14,15,16,17-tetrahydro-6H, 13H-7, 11 - (azeno)-5, 1 -(metheno)- 12,4,6-benzoxadiazacyclononadecine;

(rac)-tert-butyl [ { [ 16,20-difluoro-2,3 ,4,5-tetrahydro- 12H- 17, 13 -(azeno)- 11 ,7-(metheno)-6, 1,12,14- benzoxathiadiazacyclononadecin-9-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 12H- 17, 13-(azeno)- 11 ,7-(metheno)-6, 1,12,14-benzoxathiadiazacyclononadecine;

- (rac)-16,20-difluoro-6-methyl-9-[(S-methylsulfonimidoyl)meth yl]-3,4,5,6-tetrahydro-2H,12H- 13,17-(azeno)- 11 ,7-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine;

(rac)-fert-butyl [{[16,20-difluoro-3,4,5,6-tetrahydro-2H,12H-13,17-(azeno)-l l,7-(metheno)-

1,6,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl)oxido-λ 6 -sulfanylidene]carbamate;

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-3,4,5,6-tetrahydro-2H, 12H- 13,17-(azeno)- 1 l,7-(metheno)- 1,6, 12,14-benzoxatriazacyclononadecine;

and the enantiomers, diastereomers, salts, solvates or salts of solvates thereof. The above mentioned definitions of groups and radicals which have been detailed in general terms or in preferred ranges also apply to the end products of the formula (I) and, analogously, to the starting materials or intermediates required in each case for the preparation.

The present invention further relates to a process for the preparation of the compounds of formula (11), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (10)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (11),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (15), in which R 1 , R 2 , R 3 , R 4 , R 5 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (14)

in which R 1 , R 2 , R 3 , R 4 , R 5 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (15),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (25), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (24)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (25),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (35), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (34)

in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catal zed C-N cross-coupling reaction,

to give compounds of the formula (35),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (35a), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, said process comprising the steps of

a. allowing compounds of formula (34)

in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, to react in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (35),

followed by

b. allowing said compounds of formula (35), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, to react with an acid,

to give compounds of the formula (35a),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

The present invention further relates to a process for the preparation of the compounds of formula (35c), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula 34a)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (35c),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

The present invention further relates to a process for the preparation of the compounds of formula (35d), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, said process comprising the steps of

a. allowing compounds of formula 34a)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

35c

to give compounds of the formula (35c),

followed by

b. allowing said compounds of formula (35c), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react with an acid,

35d to give compounds of the formula (35d),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (45), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (44)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

45

to give compounds of the formula (45),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (45a), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, said process comprising the steps of

a. allowing compounds of formula (44)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (45),

followed by b. allowing said compounds of formula (45), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react with an acid,

45a

to give compounds of the formula (45a),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

The present invention further relates to a process for the preparation of the compounds of formula (45), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula 58)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular nucleophilic displacement reaction,

to give compounds of the formula (45),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

The present invention further relates to a process for the preparation of the compounds of formula (45a), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, said process comprising the steps of

a. allowing compounds of formula 58)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react in an intramolecular nucleophilic displacement reaction,

45

to give compounds of the formula (45),

followed by

b. allowing said compounds of formula (45), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react with an acid,

to give compounds of the formula (45a),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (66), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (65)

in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular nucleophilic displacement reaction,

to give compounds of the formula (66),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The present invention further relates to a process for the preparation of the compounds of formula (66a), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, said process comprising the steps of

a. allowing compounds of formula (65)

in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, to react in an intramolecular nucleophilic displacement reaction,

to give compounds of the formula (66),

followed by b. allowing said compounds of formula (66), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention, to react with an acid,

to give compounds of the formula (66a),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

The present invention further relates to a process for the preparation of the compounds of formula (66), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, in which process compounds of formula (71)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, are reacted in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (72),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof.

The present invention further relates to a process for the preparation of the compounds of formula (66a), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, said process comprising the steps of

a. allowing compounds of formula (71)

in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react in an intramolecular Palladium-catalyzed C-N cross-coupling reaction,

to give compounds of the formula (72),

followed by

b. allowing said compounds of formula (72), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention, to react with an acid,

to give compounds of the formula (72a),

and in which process the resulting compounds are optionally, if appropriate, converted with the corresponding (i) solvents and/or (ii) bases or acids to the solvates, salts and/or solvates of the salts thereof. The invention further relates to compounds of the formula (10), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (10), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (14), in which R 1 , R 2 , R 3 , R 4 , R 5 and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (14), in which R 1 , R 2 , R 3 , R 4 , R 5 and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (24), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (24), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (34), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (34), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (34a), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (34a), in which R 1 , R 2 , R 3 , R and L are as defined for the com ound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (44), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (44), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (58), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (58), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (65), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (34), in which R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The invention further relates to compounds of the formula (71), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

or the enantiomers, diastereomers, salts, solvates or salts of solvates thereof.

The invention further relates to the use of the compounds of the formula (71), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of formula (I) according to the invention,

for the preparation of compounds of the formula (I). The compounds according to the invention show a valuable pharmacological and pharmacokinetic spectrum of action which could not have been predicted.

They are therefore suitable for use as medicaments for the treatment and/or prophylaxis of disorders in humans and animals.

The pharmaceutical activity of the compounds according to the invention can be explained by their action as selective inhibitors of CDK9, and, more significantly, as selective inhibitors of CDK9 at high ATP concentrations.

Thus, the compounds according to the general formula (I) as well as the enantiomers, diastereomers, salts, solvates and salts of solvates thereof are used as selective inhibitors for CDK9.

Furthermore, the compounds according to the invention show a particularly high potency (demonstrated by a low IC 5 0 value in the CDK9/CycTl assay) for selectively inhibiting CDK9 activity, in particular at high ATP concentrations.

In context of the present invention, the IC50 value with respect to CDK9 can be determined by the methods described in the method section below.

As compared to many CDK9 inhibitors described in the prior art, compounds of the present invention according to general formula (I) show a surprisingly high potency for inhibiting CDK9 activity, especially at high ATP concentrations, which is demonstrated by their low IC50 value in the CDK9/CycTl high ATP kinase assay. Thus, these compounds have a lower probability to be competed out of the ATP-binding pocket of CDK9/CycTl kinase due to the high intracellular ATP concentration (R. Copeland et al., Nature Reviews Drug Discovery 2006, 5, 730-739). According to this property the compounds of the present invention are particularly able to inhibit CDK9/CycTl within cells for a longer period of time as compared to classical ATP competitive kinase inhibitors. This increases the anti-tumor cell efficacy at pharmacokinetic clearance-mediated declining serum concentrations of the inhibitor after dosing of a patient or an animal.

As compared to CDK9 inhibitors in the prior art, compounds in the present invention show a surprisingly long target residence time. It has been suggested earlier that the target residence time is an appropriate predictor for drug efficacy on the basis that equilibrium-based in vitro assays inadequately reflect in vivo situations where drug concentrations fluctuate due to adsorption, distribution and elimination processes and the target protein concentration may be dynamically regulated (Tummino, P.J. and R.A. Copeland, Residence time of receptor- ligand complexes and its effect on biological function. Biochemistry, 2008. 47(20): p. 5481-5492; Copeland, R.A., D.L. Pompliano, and T.D. Meek, Drug-target residence time and its implications for lead optimization. Nature Reviews Drug Discovery, 2006. 5(9): p. 730-739).

Therefore, the equilibrium binding parameter, KD, or the functional representative, IC50, may not fully reflect requirements for in vivo efficacy. Assuming that a drug molecule can only act as long as it remains bound to its target, the "lifetime" (residence time), of the drug-target complex may serve as a more reliable predictor for drug efficacy in a non- equilibrium in vivo system. Several publications appreciated and discussed its implications for in vivo efficacy (Lu, H. and P.J. Tonge, Drug-target residence time: critical information for lead optimization. Curr Opin Chem Biol, 2010. 14(4): p. 467-74; Vauquelin, G. and S.J. Charlton, Long-lasting target binding and rebinding as mechanisms to prolong in vivo drug action. Br J Pharmacol, 2010. 161(3): p. 488-508).

One example for the impact of target residence time is given by the drug tiotropium that is used in COPD treatment. Tiotropium binds to the Ml , M2 and M3 subtype of the muscarinic receptors with comparable affinities, but is kinetically selective as it has the desired long residence times only for the M3 receptor. Its drug-target residence time is sufficiently long that after washout from human trachea in vitro, tiotropium maintains inhibition of cholinergic activity with a half-life of 9 hours. This translates to protection against bronchospasms for more than 6 hours in vivo (Price, D., A. Sharma, and F. Cerasoli, Biochemical properties, pharmacokinetics and pharmacological response of tiotropium in chronic obstructive pulmonary disease patients. 2009; Dowling, M. (2006) Br. J. Pharmacol. 148, 927-937).

Another example is Lapatinib (Tykerb). It was found was that the long target residence time found for lapatinib in the purified intracellular domain enzyme reaction correlated with the observed, prolonged signal inhibition in tumor cells based on receptor tyrosine phosphorylation measurements. It was subsequently concluded that the slow binding kinetics may offer increased signal inhibition in the tumor, thus leading to greater potential to affect the tumor growth rates or effectiveness of co-dosing with other chemotherapeutic agents. (Wood et al (2004) Cancer Res. 64: 6652-6659; Lackey (2006) Current Topics in Medicinal Chemistry, 2006, Vol. 6, No. 5) In context of the present invention, the IC50 value with respect to CDK9 at high ATP concentrations can be determined by the methods described in the method section below. Preferably, it is determined according to Method lb ("CDK9/CycTl high ATP kinase assay") as described in the Materials and Method section below.

If desired, the IC50 value with respect to CDK9 at low ATP concentration can e.g. be determined by the methods described in the method section below, according to Method la. ("CDK9/CycTl kinase assay") described in the Materials and Method section below. In context of the present invention, the target resident time of selective CDK9 inhibitors according to the present invention can be determined by the methods described in the method section below. Preferably, it is determined according to Method 8 ("Surface Plasmon Resonance PTEFb") as described in the Materials and Method section below.

Further, compounds of the present invention according to formula (I) surprisingly show a surprisingly high anti-proliferative activity in tumor cell lines, such as HeLa, HeLa-MaTu-ADR, NCI-H460, DU145, Caco-2, B16F10, A2780 or MOLM-13, compared to CDK9 inhibitors described in the prior art. In context of the present invention, the anti-proliferative activity in tumor cell lines such as HeLa, HeLa- MaTu-ADR, NCI-H460, DU145, Caco-2, B16F10, A2780 or MOLM-13 is preferably determined according to Method 3. ("Proliferation Assay") as described in the Materials and Method section below.

In context of the present invention, the aqueous solubility is preferably determined according to Method 4. ("Equilibrium Shake Flask Solubility Assay") described in the Materials and Method section below.

In context of the present invention, the metabolic stability in rat hepatocytes is preferably determined according to Method 6. ("Investigation of in vitro metabolic stability in rat hepatocytes") described in the Materials and Method section below.

In context of the present invention, the half-life in rats upon administration in vivo is preferably determined according to Method 7. ("In vivo pharmacokinetics in rats") described in the Materials and Method section below. In context of the present invention, the apparent Caco-2 permeability values from the basal to apical compartment (P app A-B) or the efflux ratio (defined as the ratio ((P apP B-A) / (P app A-B)) are preferably determined according to Method 5. ("Caco-2 Permeation Assay") described in the Materials and Method section below. A further subject matter of the present invention is the use of the compounds of general formula (I) according to the invention for the treatment and/or prophylaxis of disorders, preferably of disorders relating to or mediated by CDK9 activity, in particular of hyper-proliferative disorders, virally induced infectious diseases and/or of cardiovascular diseases, more preferably of hyper-proliferative disorders. The compounds of the present invention may be used to inhibit selectively the activity or expression of CDK9. Therefore, the compounds of formula (I) are expected to be valuable as therapeutic agents. Accordingly, in another embodiment, the present invention provides a method of treating disorders relating to or mediated by CDK9 activity in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of formula (I) as defined above. In certain embodiments, the disorders relating to CDK9 activity are hyper-proliferative disorders, virally induced infectious diseases and/or of cardiovascular diseases, more preferably hyper-proliferative disorders, particularly cancer.

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

The term "subject" or "patient" includes organisms which are capable of suffering from a cell proliferative disorder or a disorder associated with reduced or insufficient programmed cell death (apoptosis) or who could otherwise benefit from the administration of a compound of the invention, such as human and non-human animals. Preferred humans include human patients suffering from or prone to suffering from a cell proliferative disorder or associated state, as described herein. The term "non-human animals" includes vertebrates, e.g., mammals, such as non-human primates, sheep, cow, dog, cat and rodents, e.g., mice, and non-mammals, such as chickens, amphibians, reptiles, etc. The term "disorders relating to or mediated by CDK9" shall include diseases associated with or implicating CDK9 activity, for example the hyperactivity of CDK9, and conditions that accompany with these diseases. Examples of "disorders relating to or mediated by CDK9" include disorders resulting from increased CDK9 activity due to mutations in genes regulating CDK9 activity auch as LARP7, HEXIM1/2 or 7sk snRNA, or disorders resulting from increased CDK9 activity due to activation of the CDK9/cyclinT/RNApolymerase II complex by viral proteins such as HIV-TAT or HTLV-TAX or disorders resulting from increased CDK9 activity due to activation of mitogenic signaling pathways. The term "hyperactivity of CDK9" refers to increased enzymatic activity of CDK9 as compared to normal non-diseased cells, or it refers to increased CDK9 activity leading to unwanted cell proliferation, or to reduced or insufficient programmed cell death (apoptosis), or mutations leading to constitutive activation of CDK9.

The term "hyper-proliferative disorder" includes disorders involving the undesired or uncontrolled proliferation of a cell and it includes disorders involving reduced or insufficient programmed cell death (apoptosis). The compounds of the present invention can be utilized to prevent, inhibit, block, reduce, decrease, control, etc., cell proliferation and/or cell division, and/or produce apoptosis. This method comprises administering to a subject in need thereof, including a mammal, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt, hydrate or solvate thereof which is effective to treat or prevent the disorder.

Hyper-pro liferative disorders in the context of this invention include, but are not limited to, e.g., psoriasis, keloids and other hyperplasias affecting the skin, endometriosis, skeletal disorders, angiogenic or blood vessel proliferative disorders, pulmonary hypertension, fibrotic disorders, mesangial cell proliferative disorders, colonic polyps, polycystic kidney disease, benign prostate hyperplasia (BPH), and solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid, and their distant metastases. Those disorders also include lymphomas, sarcomas and leukemias.

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

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

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

Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal and vulvar cancer, as well as sarcoma of the uterus.

Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, salivary gland cancers, anal gland adenocarcinomas, and mast cell tumors.

Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral, and hereditary and sporadic papillary renal cancers.

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

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

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

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

Lymphomas include, but are not limited to AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin's disease, and lymphoma of the central nervous system. Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, rhabdomyosarcoma, malignant histiocytosis, fibrosarcoma, hemangiosarcoma, hemangiopericytoma, and leiomyosarcoma.

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

Fibrotic proliferative disorders, i.e. the abnormal formation of extracellular matrices, that may be treated with the compounds and methods of the present invention include lung fibrosis, atherosclerosis, restenosis, hepatic cirrhosis, and mesangial cell proliferative disorders, including renal diseases such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic microangiopathy syn- dromes, transplant rejection, and glomerulopathies.

Other conditions in humans or other mammals that may be treated by administering a compound of the present invention include tumor growth, retinopathy, including diabetic retinopathy, ischemic retinal- vein occlusion, retinopathy of prematurity and age-related macular degeneration, rheumatoid arthritis, psoriasis, and bullous disorders associated with subepidermal blister formation, including bullous pemphigoid, erythema multiforme and dermatitis herpetiformis.

The compounds of the present invention may also be used to prevent and treat diseases of the airways and the lung, diseases of the gastrointestinal tract as well as diseases of the bladder and bile duct.

The disorders mentioned above have been well characterized in humans, but also exist with a similar etiology in other animals, including mammals, and can be treated by administering pharmaceutical compositions of the present invention. In a further aspect of the present invention, the compounds according to the invention are used in a method for preventing and/or treating infectious diseases, in particular virally induced infectious diseases. The virally induced infectious diseases, including opportunistic diseases, are caused by retroviruses, hepadnaviruses, herpesviruses, flaviviridae, and/or adenoviruses. In a further preferred embodiment of this method, the retroviruses are selected from lentiviruses or oncoretroviruses, wherein the lentivirus is selected from the group comprising: HIV-1, HIV-2, FIV, BIV, SIVs, SHIV, CAEV, VMV or EIAV, preferably HIV-1 or HIV-2 and wherein the oncoretro virus is selected from the group of: HTLV-I, HTLV-II or BLV. In a further preferred embodiment of this method, the hepadnavirus is selected from HBV, GSHV or WHV, preferably HBV, the herpesvirus is selected from the group comprising: HSV I, HSV II, EBV, VZV, HCMV or HHV 8, preferably HCMV and the flaviviridae is selected from HCV, West nile or Yellow Fever. The compounds according to general formula (I) are also useful for prophylaxis and/or treatment of cardiovascular diseases such as cardiac hypertrophy, adult congenital heart disease, aneurysm, stable angina, unstable angina, angina pectoris, angioneurotic edema, aortic valve stenosis, aortic aneurysm, arrhythmia, arrhythmogenic right ventricular dysplasia, arteriosclerosis, arteriovenous malformations, atrial fibrillation, Behcet syndrome, bradycardia, cardiac tamponade, cardiomegaly, congestive cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, cardiovascular disease prevention, carotid stenosis, cerebral hemorrhage, Churg-Strauss syndrome, diabetes, Ebstein's Anomaly, Eisenmenger complex, cholesterol embolism, bacterial endocarditis, fibromuscular dysplasia, congenital heart defects, heart diseases, congestive heart failure, heart valve diseases, heart attack, epidural hematoma, hematoma, subdural, Hippel-Lindau disease, hyperemia, hypertension, pulmonary hypertension, hypertrophic growth, left ventricular hypertrophy, right ventricular hypertrophy, hypoplastic left heart syndrome, hypotension, intermittent claudication, ischemic heart disease, Klippel- Trenaunay- Weber syndrome, lateral medullary syndrome, long QT syndrome mitral valve prolapse, moyamoya disease, mucocutaneous lymph node syndrome, myocardial infarction, myocardial ischemia, myocarditis, pericarditis, peripheral vascular diseases, phlebitis, polyarteritis nodosa, pulmonary atresia, Raynaud disease, restenosis, Sneddon syndrome, stenosis, superior vena cava syndrome, syndrome X, tachycardia, Takayasu's arteritis, hereditary hemorrhagic telangiectasia, telangiectasis, temporal arteritis, tetralogy of fallot, thromboangiitis obliterans, thrombosis, thromboembolism, tricuspid atresia, varicose veins, vascular diseases, vasculitis, vasospasm, ventricular fibrillation, Williams syndrome, peripheral vascular disease, varicose veins and leg ulcers, deep vein thrombosis, Wolff-Parkinson- White syndrome.

Preferred are cardiac hypertrophy, adult congenital heart disease, aneurysms, angina, angina pectoris, arrhythmias, cardiovascular disease prevention, cardiomyopathies, congestive heart failure, myocardial infarction, pulmonary hypertension, hypertrophic growth, restenosis, stenosis, thrombosis and arteriosclerosis.

A further subject matter of the present invention is the use of the compounds of general formula (I) according to the invention as a medicament. A further subject matter of the present invention is the use of the compounds of general formula (I) according to the invention for the treatment and/or prophylaxis of disorders, in particular of the disorders mentioned above.

A further subject matter of the present invention is the use of the compounds of general formula (I) according to the invention for the treatment and/or prophylaxis of hyper-pro liferative disorders, virally induced infectious diseases and/or of cardiovascular diseases. A preferred subject matter of the present invention is the use of the compounds of general formula (I) according to the invention for the treatment and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone-independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias.

A further subject matter of the present invention are the compounds of general formula (I) according to the invention for the use as a medicament. A further subject matter of the present invention are the compounds of general formula (I) according to the invention for the use of treating and/or prophylaxis of the disorders mentioned above.

A further subject matter of the present invention are the compounds of general formula (I) according to the invention for the use of treating and/or prophylaxis of hyper-pro liferative disorders, virally induced infectious diseases and/or of cardiovascular diseases.

A preferred subject matter of the present invention are the compounds of general formula (I) according to the invention for the use of treating and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone-independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias.

A further subject matter of the present invention are the compounds of general formula (I) according to the invention for the use in a method for the treatment and/or prophylaxis of the disorders mentioned above.

A further subject matter of the present invention are the compounds of general formula (I) according to the invention for the use in a method for the treatment and/or prophylaxis of hyper-pro liferative disorders, virally induced infectious diseases and/or of cardiovascular diseases.

A preferred subject matter of the present invention are the compounds of general formula (I) according to the invention for the use in a method of treatment and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone-independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias. A further subject matter of the present invention is the use of the compounds of general formula (I) according to the invention in the manufacture of a medicament for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above. A further subject matter of the present invention is the use of the compounds of general formula (I) according to the invention in the manufacture of a medicament for the treatment and/or prophylaxis of hyper-proliferative disorders, virally induced infectious diseases and/or of cardiovascular diseases.

A preferred subject matter of the present invention is the use of the compounds of general formula (I) according to the invention in the manufacture of a medicament for the treatment and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone- independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias.

A further subject matter of the present invention is a method for the treatment and/or prophylaxis of disorders, in particular the disorders mentioned above, using an effective amount of the compounds of general formula (I) according to the invention. A further subject matter of the present invention is a method for the treatment and/or prophylaxis of hyper-proliferative disorders, virally induced infectious diseases and/or of cardiovascular diseases, using an effective amount of the compounds of general formula (I) according to the invention.

A preferred subject matter of the present invention is a method for the treatment and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone- independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias using an effective amount of the compounds of general formula (I) according to the invention.

Another aspect of the present invention relates to pharmaceutical combinations comprising a compound of general formula (I) according to the invention in combination with at least one or more further active ingredients. As used herein the term "pharmaceutical combination" refers to a combination of at least one compound of general formula (I) according to the invention as active ingredient together with at least one other active ingredient with or without further ingredients, carrier, diluents and/or solvents. Another aspect of the present invention relates to pharmaceutical compositions comprising a compound of general formula (I) according to the invention in combination with an inert, nontoxic, pharmaceutically suitable adjuvant. As used herein the term "pharmaceutical composition" refers to a galenic formulation of at least one pharmaceutically active agent together with at least one further ingredient, carrier, diluent and/or solvent.

Another aspect of the present invention relates to the use of the pharmaceutical combinations and/or the pharmaceutical compositions according to the invention for the treatment and/or prophylaxis of disorders, in particular of the disorders mentioned above.

Another aspect of the present invention relates to the use of the pharmaceutical combinations and/or the pharmaceutical compositions according to the invention for the treatment and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone- independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias.

Another aspect of the present invention relates to pharmaceutical combinations and/or the pharmaceutical compositions according to the invention for use of the treatment and/or prophylaxis of disorders, in particular of the disorders mentioned above.

Another aspect of the present invention relates to pharmaceutical combinations and/or the pharmaceutical compositions according to the invention for use of the treatment and/or prophylaxis of lung carcinomas, especially non-small cell lung carcinomas, prostate carcinomas, especially hormone- independent human prostate carcinomas, cervical carcinomas, including multidrug-resistant human cervical carcinomas, colorectal carcinomas, melanomas, ovarian carcinomas or leukemias, especially acute myeloid leukemias. Compounds of formula (I) may be administered as the sole pharmaceutical agent or in combination with one or more additional therapeutic agents where the combination causes no unacceptable adverse effects. This pharmaceutical combination includes administration of a single pharmaceutical dosage formulation which contains a compound of formula (I) and one or more additional therapeutic agents, as well as administration of the compound of formula (I) and each additional therapeutic agent in its own separate pharmaceutical dosage formulation. For example, a compound of formula (I) and a therapeutic agent may be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent may be administered in separate dosage formulations. Where separate dosage formulations are used, the compound of formula (I) and one or more additional therapeutic agents may be administered at essentially the same time (e.g., concurrently) or at separately staggered times (e.g., sequentially).

In particular, the compounds of the present invention may be used in fixed or separate combination with other anti-tumor agents such as alkylating agents, anti-metabolites, plant-derived anti-tumor agents, hormonal therapy agents, topoisomerase inhibitors, camptothecin derivatives, kinase inhibitors, targeted drugs, antibodies, interferons and/or biological response modifiers, anti-angiogenic compounds, and other anti-tumor drugs. In this regard, the following is a non-limiting list of examples of secondary agents that may be used in combination with the compounds of the present invention:

• Alkylating agents include, but are not limited to, nitrogen mustard N-oxide, cyclophosphamide, ifosfamide, thiotepa, ranimustine, nimustine, temozolomide, altretamine, apaziquone, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, mafosfamide, bendamustin, and mitolactol; platinum-coordinated alkylating compounds include, but are not limited to, cisplatin, carboplatin, eptaplatin, lobaplatin, nedaplatin, oxaliplatin, and satraplatin;

• Anti-metabolites include, but are not limited to, methotrexate, 6-mercaptopurine riboside, mercaptopurine, 5-fluorouracil alone or in combination with leucovorin, tegafur, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, gemcitabine, fludarabin, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethynylcytidine, cytosine arabinoside, hydroxyurea, melphalan, nelarabine, nolatrexed, ocfosfite, disodium premetrexed, pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincristine, and vinorelbine;

• Hormonal therapy agents include, but are not limited to, exemestane, Lupron, anastrozole, doxercalciferol, fadrozole, formestane, 1 1 -beta hydroxysteroid dehydrogenase 1 inhibitors, 17-alpha hydroxylase/17,20 lyase inhibitors such as abiraterone acetate, 5-alpha reductase inhibitors such as finasteride and epristeride, anti- estrogens such as tamoxifen citrate and fulvestrant, Trelstar,toremifene, raloxifene, lasofoxifene, letrozole, anti-androgens such as bicalutamide, flutamide, mifepristone, nilutamide, Casodex, and anti-progesterones and combinations thereof;

• Plant-derived anti-tumor substances include, e.g., those selected from mitotic inhibitors, for example epothilones such as sagopilone, ixabepilone and epothilone B, vinblastine, vinflunine, docetaxel, and paclitaxel;

• Cytotoxic topoisomerase inhibiting agents include, but are not limited to, aclarubicin, doxorubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan, topotecan, edotecarin, epimbicin, etoposide, exatecan, gimatecan, lurtotecan, mitoxantrone, pirambicin, pixantrone, rubitecan, sobuzoxane, tafluposide, and combinations thereof; • Immunologicals include interferons such as interferon alpha, interferon alpha-2a, interferon alpha- 2b, interferon beta, interferon gamma- la and interferon gamma-nl, and other immune enhancing agents such as L19-IL2 and other IL2 derivatives, filgrastim, lentinan, sizofilan, TheraCys, ubenimex, aldesleukin, alemtuzumab, BAM-002, dacarbazine, daclizumab, denileukin, gemtuzumab, ozogamicin, ibritumomab, imiquimod, lenograstim, lentinan, melanoma vaccine (Corixa), molgramostim, sargramostim, tasonermin, tecleukin, thymalasin, tositumomab, Vimlizin, epratuzumab, mitumomab, oregovomab, pemtumomab, and Provenge; Merial melanoma vaccine

• Biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses such as survival, growth or differentiation of tissue cells to direct them to have anti-tumor activity; such agents include, e.g., krestin, lentinan, sizofiran, picibanil, ProMune, and ubenimex;

• Anti-angiogenic compounds include, but are not limited to, acitretin, aflibercept, angiostatin, aplidine, asentar, axitinib, recentin, bevacizumab, brivanib alaninat, cilengtide, combretastatin, DAST, endostatin, fenretinide, halofuginone, pazopanib, ranibizumab, rebimastat, removab, revlimid, sorafenib, vatalanib, squalamine, sunitinib, telatinib, thalidomide, ukrain, and vitaxin;

• Antibodies include, but are not limited to, trastuzumab, cetuximab, bevacizumab, rituximab, ticilimumab, ipilimumab, lumiliximab, catumaxomab, atacicept, oregovomab, and alemtuzumab;

• VEGF inhibitors such as, e.g., sorafenib, DAST, bevacizumab, sunitinib, recentin, axitinib, aflibercept, telatinib, brivanib alaninate, vatalanib, pazopanib, and ranibizumab; Palladia

• EGFR (HER1) inhibitors such as, e.g., cetuximab, panitumumab, vectibix, gefitinib, erlotinib, and Zactima;

• HER2 inhibitors such as, e.g., lapatinib, tratuzumab, and pertuzumab;

• mTOR inhibitors such as, e.g., temsirolimus, sirolimus/Rapamycin, and everolimus;

• c-Met inhibitors;

• PI3K and AKT inhibitors;

• CDK inhibitors such as roscovitine and flavopiridol;

• Spindle assembly checkpoints inhibitors and targeted anti-mitotic agents such as PLK inhibitors, Aurora inhibitors (e.g. Hesperadin), checkpoint kinase inhibitors, and KSP inhibitors;

• HDAC inhibitors such as, e.g., panobinostat, vorinostat, MS275, belinostat, and LBH589;

• HSP90 and HSP70 inhibitors;

• Proteasome inhibitors such as bortezomib and carfilzomib;

• Serine/threonine kinase inhibitors including MEK inhibitors (such as e.g. RDEA 119) and Raf inhibitors such as sorafenib;

• Farnesyl transferase inhibitors such as, e.g., tipifarnib;

• Tyrosine kinase inhibitors including, e.g., dasatinib, nilotibib, DAST, bosutinib, sorafenib, bevacizumab, sunitinib, AZD2171, axitinib, aflibercept, telatinib, imatinib mesylate, brivanib alaninate, pazopanib, ranibizumab, vatalanib, cetuximab, panitumumab, vectibix, gefitinib, erlotinib, lapatinib, tratuzumab, pertuzumab, and c-Kit inhibitors; Palladia, masitinib

• Vitamin D receptor agonists;

• Bcl-2 protein inhibitors such as obatoclax, oblimersen sodium, and gossypol;

· Cluster of differentiation 20 receptor antagonists such as, e.g., rituximab;

• Ribonucleotide reductase inhibitors such as, e.g., gemcitabine;

• Tumor necrosis apoptosis inducing ligand receptor 1 agonists such as, e.g., mapatumumab;

• 5-Hydroxytryptamine receptor antagonists such as, e.g., rEV598, xaliprode, palonosetron hydrochloride, granisetron, Zindol, and AB- 1001 ;

· Integrin inhibitors including alpha5-betal integrin inhibitors such as, e.g., E7820, JSM 6425, volociximab, and endostatin;

• Androgen receptor antagonists including, e.g., nandrolone decanoate, fluoxymesterone, Android, Prost-aid, andromustine, bicalutamide, flutamide, apo-cyproterone, apo-flutamide, chlormadinone acetate, Androcur, Tabi, cyproterone acetate, and nilutamide;

· Aromatase inhibitors such as, e.g., anastrozole, letrozole, testolactone, exemestane, amino- glutethimide, and formestane;

• Matrix metalloproteinase inhibitors;

• Other anti-cancer agents including, e.g., alitretinoin, ampligen, atrasentan bexarotene, bortezomib, bosentan, calcitriol, exisulind, fotemustine, ibandronic acid, miltefosine, mitoxantrone, I- asparaginase, procarbazine, dacarbazine, hydroxycarbamide, pegaspargase, pentostatin, tazaroten, velcade, gallium nitrate, canfosfamide, darinaparsin, and tretinoin.

The compounds of the present invention may also be employed in cancer treatment in conjunction with radiation therapy and/or surgical intervention.

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

(1) yield better efficacy in reducing the growth of a tumor or even eliminate the tumor as

compared to administration of either agent alone,

(2) provide for the administration of lesser amounts of the administered chemotherapeutic agents, (3) provide for a chemotherapeutic treatment that is well tolerated in the patient with fewer

deleterious pharmacological complications than observed with single agent chemotherapies and certain other combined therapies,

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

humans,

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

provide a longer time for tumor progression, and/or

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

Furthermore, the compounds of formula (I) may be utilized, as such or in compositions, in research and diagnostics, or as analytical reference standards, and the like, which are well known in the art. The compounds according to the invention can act systemically and/or locally. For this purpose, they can be administered in a suitable way, such as, for example, by the oral, parenteral, pulmonal, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival or otic route, or as an implant or stent.

For these administration routes, it is possible to administer the compounds according to the invention in suitable application forms.

Suitable for oral administration are administration forms which work as described in the prior art and deliver the compounds according to the invention rapidly and/or in modified form, which comprise the compounds according to the invention in crystalline and/or amorphous and/or dissolved form, such as, for example, tablets (coated or uncoated, for example tablets provided with enteric coatings or coatings whose dissolution is delayed or which are insoluble and which control the release of the compound according to the invention), tablets which rapidly decompose in the oral cavity, or films/wafers, films/lyophilizates, capsules (for example hard or soft gelatin capsules), sugar-coated tablets, granules, pellets, powders, emulsions, suspensions, aerosols or solutions.

Parenteral administration can take place with avoidance of an absorption step (for example intravenously, intraarterially, intracardially, intraspinally or intralumbally) or with inclusion of absorption (for example intramuscularly, subcutaneously, intracutaneously, percutaneously or intraperitoneally). Administration forms suitable for parenteral administration are, inter alia, preparations for injection and infusion in the form of solutions, suspensions, emulsions, lyophilizates or sterile powders.

Examples suitable for the other administration routes are pharmaceutical forms for inhalation (inter alia powder inhalers, nebulizers), nasal drops/solutions/sprays; tablets to be administered lingually, sub lingually or buccally, films/wafers or capsules, suppositories, preparations for the eyes or ears, vaginal capsules, aqueous suspensions (lotions, shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems (such as plasters, for example), milk, pastes, foams, dusting powders, implants or stents.

The compounds according to the invention can be converted into the stated administration forms. This can take place in a manner known per se by mixing with inert, nontoxic, pharmaceutically suitable adjuvants. These adjuvants include, inter alia, carriers (for example microcrystalline cellulose, lactose, mannitol), solvents (for example liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (for example sodium dodecyl sulphate, polyoxysorbitan oleate), binders (for example polyvinylpyrrolidone), synthetic and natural polymers (for example albumin), stabilizers (for example antioxidants, such as, for example, ascorbic acid), colorants (for example inorganic pigments, such as, for example, iron oxides) and flavour- and/or odour-masking agents.

The present invention furthermore provides medicaments comprising at least one compound according to the invention, usually together with one or more inert, nontoxic, pharmaceutically suitable adjuvants, and their use for the purposes mentioned above.

When the compounds of the present invention are administered as pharmaceuticals, to humans or animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1% to 99,5% (more preferably 0.5% to 90%>) of active ingredient in combination with one or more inert, nontoxic, pharmaceutically suitable adjuvants.

Regardless of the route of administration selected, the compounds according to the invention of general formula (I) and/or the pharmaceutical composition of the present invention are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Actual dosage levels and time course of administration of the active ingredients in the pharmaceutical compositions of the invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient without being toxic to the patient.

Materials and Methods:

The percentage data in the following tests and examples are percentages by weight unless otherwise indicated; parts are parts by weight. Solvent ratios, dilution ratios and concentration data of liquid/liquid solutions are in each case based on volume.

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

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

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

Examples were synthesized one or more times. When synthesized more than once, data from biological and/or physicochemical assays represent average values or median values calculated utilizing data sets obtained from testing of one or more synthetic batch. The in vitro pharmacological, pharmacokinetic and physicochemical properties of the compounds can be determined according to the following assays and methods.

Noteworthily, in the CDK9 assays described below the resolution power is limited by the enzyme concentrations, the lower limit for IC50S is about 1-2 nM in the CDK9 high ATP assay and 2-4 nM in the CDK low ATP assays. For compounds exhibiting IC50S in this range the true affinity to CDK9 and thus the selectivity for CDK9 over CDK2 might be even higher, i.e. for these compounds the selectivity factors calculated in columns 4 and 7 of Table 2, infra, are minimal values, they could be also higher.

la. CDK9/CycTl kinase assay:

CDK9/CycTl -inhibitory activity of compounds of the present invention was quantified employing the CDK9/CycTl TR-FRET assay as described in the following paragraphs:

Recombinant full-length His-tagged human CDK9 and CycTl, expressed in insect cells and purified by Ni-NTA affinity chromatography, were purchased from Invitrogen (Cat. No PV4131). As substrate for the kinase reaction biotinylated peptide biotin-Ttds-YISPLKSPYKISEG (C-terminus in amid form) was used which can be purchased e.g. from the company JERTNI Peptide Technologies (Berlin, Germany).

For the assay 50 nl of a lOOfold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΐ of a solution of CDK9/CycTl in aqueous assay buffer [50 mM Tris/HCl pH 8.0, lO mM MgCl 2 , 1.0 mM dithiothreitol, 0.1 mM sodium ortho-vanadate, 0.01% (v/v) Nonidet-P40 (Sigma)] were added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΐ of a solution of adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΐ assay volume is 10 μΜ) and substrate (1.67 μΜ => final cone, in the 5 μΐ assay volume is 1 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 25 min at 22°C. The concentration of CDK9/CycTl was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 1 μg/mL. The reaction was stopped by the addition of 5 μΐ of a solution of TR-FRET detection reagents (0.2 μΜ streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-RB(pSer807/pSer81 l)-antibody from BD Pharmingen [# 558389] and 1.2 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no. AD0077]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 100 mM HEPES pH 7.5).

The resulting mixture was incubated 1 h at 22°C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu- chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm was measured in a HTRF reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 11 different concentrations in the range of 20 μΜ to 0.1 nM (20 μΜ, 5.9 μΜ, 1.7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 nM, 13 nM, 3.8 nM, 1.1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the lOOfold concentrated solutions in DMSO by serial 1 :3.4 dilutions) in duplicate values for each concentration and IC50 values were calculated by a 4 parameter fit using an inhouse software.

lb. CDK9/CycTl high ATP kinase assay

CDK9/CycTl -inhibitory activity of compounds of the present invention at a high ATP concentration after preincubation of enzyme and test compounds was quantified employing the CDK9/CycTl TR- FRET assay as described in the following paragraphs.

Recombinant full-length His-tagged human CDK9 and CycTl, expressed in insect cells and purified by Ni-NTA affinity chromatography, were purchase from Invitrogen (Cat. No PV4131). As substrate for the kinase reaction biotinylated peptide biotin-Ttds-YISPLKSPYKISEG (C-terminus in amid form) was used which can be purchased e.g. from the company JERINI peptide technologies (Berlin, Germany). For the assay 50 nl of a lOOfold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΐ of a solution of CDK9/CycTl in aqueous assay buffer [50 mM Tris/HCl pH 8.0, 10 mM MgCl 2 , 1.0 mM dithiothreitol, 0.1 mM sodium ortho-vanadate, 0.01% (v/v) Nonidet-P40 (Sigma)] were added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΐ of a solution of adenosine-tri-phosphate (ATP, 3.3 mM => final cone, in the 5 μΐ assay volume is 2 mM) and substrate (1.67 μΜ => final cone, in the 5 μΐ assay volume is 1 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 25 min at 22°C. The concentration of CDK9/CycTl was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 0.5 μg/mL. The reaction was stopped by the addition of 5 μΐ of a solution of TR-FRET detection reagents (0.2 μΜ streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-RB(pSer807/pSer81 l)-antibody from BD Pharmingen [# 558389] and 1.2 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no. AD0077]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 100 mM HEPES pH 7.5).

The resulting mixture was incubated 1 h at 22°C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu- chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm was measured in a HTRF reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 11 different concentrations in the range of 20 μΜ to 0.1 nM (20 μΜ, 5.9 μΜ, 1.7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 nM, 13 nM, 3.8 nM, 1.1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the lOOfold concentrated solutions in DMSO by serial 1 :3.4 dilutions) in duplicate values for each concentration and IC50 values were calculated by a 4 parameter fit using an inhouse software.

2a. CDK2/CycE kinase assay:

CDK2/CycE -inhibitory activity of compounds of the present invention was quantified employing the CDK2/CycE TR-FRET assay as described in the following paragraphs:

Recombinant fusion proteins of GST and human CDK2 and of GST and human CycE, expressed in insect cells (Sf9) and purified by Glutathion-Sepharose affinity chromatography, were purchased from ProQinase GmbH (Freiburg, Germany). As substrate for the kinase reaction biotinylated peptide biotin- Ttds-YISPLKSPYKISEG (C-terminus in amid form) was used which can be purchased e.g. from the company JERINI Peptide Technologies (Berlin, Germany).

For the assay 50 nl of a lOOfold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΐ of a solution of CDK2/CycE in aqueous assay buffer [50 mM Tris/HCl pH 8.0, lO mM MgCl 2 , 1.0 mM dithiothreitol, 0.1 mM sodium ortho-vanadate, 0.01% (v/v) Nonidet-P40 (Sigma)] were added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΐ of a solution of adenosine-tri-phosphate (ATP, 16.7 μΜ => final cone, in the 5 μΐ assay volume is 10 μΜ) and substrate (1.25 μΜ => final cone, in the 5 μΐ assay volume is 0.75 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 25 min at 22°C. The concentration of CDK2/CycE was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 130 ng/mL. The reaction was stopped by the addition of 5 μΐ of a solution of TR-FRET detection reagents (0.2 μΜ streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-RB(pSer807/pSer81 l)-antibody from BD Pharmingen [# 558389] and 1.2 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no. AD0077]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 100 mM HEPES pH 7.5).

The resulting mixture was incubated 1 h at 22°C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu- chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm was measured in a TR-FRET reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 11 different concentrations in the range of 20 μΜ to 0.1 nM (20 μΜ, 5.9 μΜ, 1.7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 nM, 13 nM, 3.8 nM, 1.1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the lOOfold concentrated solutions in DMSO by serial 1 :3.4 dilutions) in duplicate values for each concentration and IC50 values were calculated by a 4 parameter fit using an inhouse software.

2b. CDK2/CycE high ATP kinase assay:

CDK2/CycE -inhibitory activity of compounds of the present invention at 2 mM adenosine-tri-phosphate (ATP) was quantified employing the CDK2/CycE TR-FRET (TR-FRET = Time Resolved Fluorescence Resonance Energy Transfer) assay as described in the following paragraphs. Recombinant fusion proteins of GST and human CDK2 and of GST and human CycE, expressed in insect cells (Sf9) and purified by Glutathion-Sepharose affinity chromatography, were purchase from ProQinase GmbH (Freiburg, Germany). As substrate for the kinase reaction biotinylated peptide biotin- Ttds-YISPLKSPYKISEG (C-terminus in amid form) was used which can be purchased e.g. from the company JERINI peptide technologies (Berlin, Germany).

For the assay 50 nl of a lOOfold concentrated solution of the test compound in DMSO was pipetted into a black low volume 384well microtiter plate (Greiner Bio-One, Frickenhausen, Germany), 2 μΐ of a solution of CDK2/CycE in aqueous assay buffer [50 mM Tris/HCl pH 8.0, 10 mM MgCl 2 , 1.0 mM dithiothreitol, 0.1 mM sodium ortho-vanadate, 0.01% (v/v) Nonidet-P40 (Sigma)] were added and the mixture was incubated for 15 min at 22°C to allow pre-binding of the test compounds to the enzyme before the start of the kinase reaction. Then the kinase reaction was started by the addition of 3 μΐ of a solution ATP (3.33 mM => final cone, in the 5 μΐ assay volume is 2 mM) and substrate (1.25 μΜ => final cone, in the 5 μΐ assay volume is 0.75 μΜ) in assay buffer and the resulting mixture was incubated for a reaction time of 25 min at 22°C. The concentration of CDK2/CycE was adjusted depending of the activity of the enzyme lot and was chosen appropriate to have the assay in the linear range, typical concentrations were in the range of 15 ng/ml. The reaction was stopped by the addition of 5 μΐ of a solution of TR-FRET detection reagents (0.2 μΜ streptavidine-XL665 [Cisbio Bioassays, Codolet, France] and 1 nM anti-RB(pSer807/pSer81 l)-antibody from BD Pharmingen [# 558389] and 1.2 nM LANCE EU-W1024 labeled anti-mouse IgG antibody [Perkin-Elmer, product no. AD0077, as an alternative a Terbium-cryptate-labeled anti-mouse IgG antibody from Cisbio Bioassays can be used]) in an aqueous EDTA-solution (100 mM EDTA, 0.2 % (w/v) bovine serum albumin in 100 mM HEPES pH 7.5).

The resulting mixture was incubated 1 h at 22°C to allow the formation of complex between the phosphorylated biotinylated peptide and the detection reagents. Subsequently the amount of phosphorylated substrate was evaluated by measurement of the resonance energy transfer from the Eu- chelate to the streptavidine-XL. Therefore, the fluorescence emissions at 620 nm and 665 nm after excitation at 350 nm wer measured in a TR-FRET reader, e.g. a Rubystar (BMG Labtechnologies, Offenburg, Germany) or a Viewlux (Perkin-Elmer). The ratio of the emissions at 665 nm and at 622 nm was taken as the measure for the amount of phosphorylated substrate. The data were normalised (enzyme reaction without inhibitor = 0 % inhibition, all other assay components but no enzyme = 100 % inhibition). Usually the test compounds were tested on the same microtiterplate in 11 different concentrations in the range of 20 μΜ to 0.1 nM (20 μΜ, 5.9 μΜ, 1.7 μΜ, 0.51 μΜ, 0.15 μΜ, 44 nM, 13 nM, 3.8 nM, 1.1 nM, 0.33 nM and 0.1 nM, the dilution series prepared separately before the assay on the level of the lOOfold concentrated solutions in DMSO by serial 1 :3.4 dilutions) in duplicate values for each concentration and IC50 values were calculated by a 4 parameter fit using an inhouse software. 3. Proliferation Assay:

Cultivated tumour cells (HeLa, human cervical tumour cells, ATCC CCL-2; NCI-H460, human non- small cell lung carcinoma cells, ATCC HTB-177; DU 145, hormone- independent human prostate carcinoma cells, ATCC HTB-81 ; HeLa-MaTu-ADR, multidrug-resistant human cervical carcinoma cells, EPO-GmbH Berlin; Caco-2, human colorectal carcinoma cells, ATCC HTB-37; B16F10, mouse melanoma cells, ATCC CRL-6475) were plated at a density of 5,000 cells/well (DU145, HeLa-MaTu- ADR), 3,000 cells/well (NCI-H460, HeLa), 1,500 cells/well (Caco-2), or 1,000 cells/well (B16F10) in a 96-well multititer plate in 200 μΐ ^ of their respective growth medium supplemented 10% fetal calf serum. After 24 hours, the cells of one plate (zero-point plate) were stained with crystal violet (see below), while the medium of the other plates was supplemented with the test substances in various concentrations (0 μΜ, as well as in the range of 0.0001-10 μΜ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.1%) using a Hewlett-Packard HP D300 Digital Dispenser. The cells were incubated for 4 days in the presence of test substances. Cell proliferation was determined by staining the cells with crystal violet: the cells were fixed by adding 20 μΐ/measuring point of an 11% glutaric aldehyde solution for 15 minutes at room temperature. After three washing cycles of the fixed cells with water, the plates were dried at room temperature. The cells were stained by adding 100 μΐ/measuring point of a 0.1 % crystal violet solution (pH 3.0). After three washing cycles of the stained cells with water, the plates were dried at room temperature. The dye was dissolved by adding 100 μΐ/measuring point of a 10% acetic acid solution. The extinction was determined by photometry at a wavelength of 595 nm. The change of cell number, in percent, was calculated by normalization of the measured values to the extinction values of the zero-point plate (=0%) and the extinction of the untreated (0 μιη) cells (=100%). The IC50 values (inhibitory concentration at 50% of maximal effect) were determined by means of a 4 parameter fit. A2780 human ovarian carcinoma cells (ECACC # 93112519) and non-adherent MOLM-13 human acute myeloid leukemia cells (DSMZ ACC 554) were seeded at a density of 3,000 cell/well (A2780) or 5,000 cells/well (MOLM-13) in a 96-well multititer plate in 150 μΕ of growth medium supplemented 10% fetal calf serum. After 24 hours, cell viability of one plate (zero-point plate) was determined with the Cell Titre-Glo Luminescent Cell Viability Assay (Promega), while the medium of the other plates was supplemented with the test substances in various concentrations (0 μΜ, as well as in the range of 0.0001- 10 μΜ; the final concentration of the solvent dimethyl sulfoxide was adjusted to 0.1%) using a Hewlett- Packard HP D300 Digital Dispenser. Cell viability was assessed after 72-hour exposure with the Cell Titre-Glo Luminescent Cell Viability Assay (Promega). IC 5 0 values (inhibitory concentration at 50% of maximal effect) were determined by means of a 4 parameter fit on measurement data which were normalized to vehicle (DMSO) treated cells (=100%) and measurement readings taken immediately before compound exposure (=0%). 4. Equilibrium Shake Flask Solubility Assay:

4a) High Throughput determination of aqueous drug solubility (100 mmolar in DMSO)

The high throughput screening method to determine aqueous drug solubility is based on:

Thomas Onofrey and Greg Kazan, Performance and correlation of a 96-well high throughput screening method to determine aqueous drug solubility,

http://ww.millipore.com/publications.nsf/a73664f9f981af8c 852569b9005b4eee/e565516fb76e7435852 56da30052db77/$FILE/AN1731EN00.pdf

The assay was run in a 96-well plate format. Each well was filled with an individual compound.

All pipetting steps were performed using a robot platform.

100 μΐ of a 10 mmolar solution of drug in DMSO were concentrated by vacuum centrifugation and resolved in 10 μΐ DMSO. 990 μΐ phosphate buffer pH 6.5 were added. The content of DMSO amounts to 1%. The multititer plate was put on a shaker and mixed for 24 hrs at room temperature.150 μΐ of the suspension were transferred to a filtration plate. After filtration using a vacuum manifold the filtrate was diluted 1 :400 and 1 :8000. A second microtiter plate with 20 μΐ of a 10 mM solution of drug in DMSO served for calibration. Two concentrations (0.005 μΜ and 0.0025 μΜ) were prepared by dilution in DMSO / water 1 :1 and used for calibration. Filtrate and calibration plates were quantified by HPLC- MS/MS.

Chemicals:

Preparation of 0.1 m phosphate buffer pH 6.5:

61.86 g NaCl and 39.54 mg KH2PO4 were solved in water and filled up to 1 1. The mixture was diluted 1 : 10 with water and the pH adjusted to 6.5 by NaOH.

Materials:

Millipore MultiScreen HT s-HV Plate 0.45 μιη

Chromatographic conditions were as follows:

HPLC column: Ascentis Express CI 8 2.7 μιη 4.6 x 30 mm

Injection volume: 1 μΐ

Flow: 1.5 ml/min

Mobile phase: acidic gradient

A: Water / 0.05% HCOOH

B: Acetonitrile / 0.05% HCOOH

0 min → 95%A 5%B

0.75 min→ 5%A 95%B

2.75 min→ 5%A 95%B

2.76 min→ 95%A 5%B

3 min → 95%A 5%B The areas of sample- and calibration injections were determined by using mass spectromety software (AB SCIEX: Discovery Quant 2.1.3. and Analyst 1.6.1). The calculation of the solubility value (in mg/1) was executed by an inhouse developed Excel macro. 4b) Thermodynamic solubility in water from powder

The thermodynamic solubility of compounds in water was determined by an equilibrium shake flask method (see for example: E.H. Kerns, L. Di: Drug-like Properties: Concepts, Structure Design and Methods, 276-286, Burlington, MA, Academic Press, 2008). A saturated solution of the drug was prepared and the solution was mixed for 24 h to ensure that equilibrium was reached. The solution was centrifuged to remove the insoluble fraction and the concentration of the compound in solution was determined using a standard calibration curve. To prepare the sample, 2 mg solid compound was weighed in a 4 mL glass vial. 1 mL phosphate buffer pH 6.5 was added. The suspension was stirred for 24 hrs at room temperature. The solution was centrifuged afterwards. To prepare the sample for the standard calibration, 2 mg solid sample was dissolved in 30 mL acetonitrile. After sonification the solution was diluted with water to 50 mL. Sample and standards were quantified by HPLC with UV- detection. For each sample two injection volumes (5 and 50 μΐ) in triplicates were made. Three injection volumes (5 μΐ, 10 μΐ and 20 μΐ) were made for the standard.

Chromatographic conditions:

HPLC column: Xterra MS CI 8 2.5 μιη 4.6 x 30 mm

Injection volume: Sample: 3χ5μ1 and 3χ50μ1

Standard: 5μ1, ΙΟμΙ, 20μ1

Flow: 1.5mL/min

Mobile phase: acidic gradient:

A: Water / 0.01% TFA

B: Acetonitrile / 0.01% TFA

0 min → 95%A 5%B

0-3 min→ 35%>A 65%>B, linear gradient

3-5 min→ 35%>A 65%>B, isocratic

5-6 min→ 95%A 5%B, isocratic

UV detector: wavelength near the absorption maximum (between 200 and 400nm)

The areas of sample- and standard injections as well as the calculation of the solubility value (in mg/1) were determined by using HPLC software (Waters Empower 2 FR).

4c) Thermodynamic solubility in Citrate buffer pH 4

Thermodynamic solubility was determined by an equilibrium shake flask method [Literature: Edward H. Kerns and Li Di (2008) Solubility Methods in: Drug-like Properties: Concepts, Structure Design and Methods, p276-286. Burlington, MA: Academic Press]. A saturated solution of the drug was prepared and the solution was mixed for 24 h to ensure that equilibrium has been reached. The solution was centrifuged to remove the insoluble fraction and the concentration of the compound in solution was determined using a standard calibration curve.

To prepare the sample, 1.5 mg solid compound was weighed in a 4 ml glass vial. 1 ml Citrate buffer pH 4 was added. The suspension was put on a stirrer and mixed for 24 hrs at room temperature. The solution was centrifuged afterwards. To prepare the sample for the standard calibration, 0.6 mg solid sample was dissolved in 19 ml acetonitrile/water 1 : 1. After sonification the solution was filled up with acetonitrile/water 1 :1 to 20 ml.

Sample and standards were quantified by HPLC with UV-detection. For each sample two injection volumes (5 and 50 μΐ) in triplicates were made. Three injection volumes (5 μΐ, 10 μΐ and 20 μΐ) were made for the standard.

Chemicals:

Citrate buffer pH 4 (MERCK Art. 109435; 1 L buffer consisting of 11,768 g citric acid,

4,480 g sodium hydroxide, 1,604 g hydrogen chloride)

Chromatographic conditions were as follows:

HPLC column: Xterra MS CI 8 2.5 μιη 4.6 x 30 mm

Injection volume: Sample: 3χ5μ1 and 3χ50μ1

Standard: 5μ1, ΙΟμΙ, 20μ1

Flow: 1.5ml/min

Mobile phase: acidic gradient:

A: Water / 0.01% TFA

B: Acetonitrile / 0.01% TFA

O min: 95%A 5%B

0-3 min: 35%A 65%B, linear gradient

3-5 min: 35%A 65%B, isocratic

5-6 min: 95%A 5%B, isocratic

UV detector: wavelength near the absorption maximum (between 200 and 400nm)

The areas of sample- and standard injections as well as the calculation of the solubility value (in mg/1) were determined by using HPLC software (Waters Empower 2 FR).

The areas of sample- and standard injections as well as the calculation of the solubility value (in mg/1) were determined by using HPLC software (Waters Empower 2 FR). 5. Caco-2 Permeation Assay:

Caco-2 cells (purchased from DSMZ Braunschweig, Germany) were seeded at a density of 4.5 x 10 4 cells per well on 24 well insert plates, 0.4 μιη pore size, and grown for 15 days in DMEM medium supplemented with 10% fetal bovine serum, 1% GlutaMAX (lOOx, GIBCO), 100 U/mL penicillin, 100μg/mL streptomycin (GIBCO) and 1% non essential amino acids (100 x). Cells were maintained at 37°C in a humified 5% CO2 atmosphere. Medium was changed every 2-3 day. Before running the permeation assay, the culture medium was replaced by a FCS-free hepes-carbonate transport buffer (pH 7.2). For assessment of monolayer integrity the transepithelial electrical resistance (TEER) was measured. Test compounds were predissolved in DMSO and added either to the apical or basolateral compartment in final concentration of 2 μΜ in transport buffer. Before and after 2h incubation at 37°C samples were taken from both compartments. Analysis of compound content was done after precipitation with methanol by LC/MS/MS analysis. Permeability (Papp) was calculated in the apical to basolateral (A → B) and basolateral to apical (B→ A) directions. The apparent permeability was calculated using following equation:

Papp = (Vr/Po)(l/S)(P2/t)

Where Vr is the volume of medium in the receiver chamber, Po is the measured peak area or height of the test drug in the donor chamber at t=o, S the surface area of the monolayer, P2 is the measured peak area of the test drug in the acceptor chamber after 2h of incubation, and t is the incubation time. The efflux ratio basolateral (B) to apical (A) was calculated by dividing the Papp B-A by the Papp A-B. In addition the compound recovery was calculated.

6. Investigation of in vitro metabolic stability in rat hepatocytes

Hepatocytes from Han Wistar rats were isolated via a 2-step perfusion method. After perfusion, the liver was carefully removed from the rat: the liver capsule was opened and the hepatocytes were gently shaken out into a Petri dish with ice-cold Williams medium E (purchased from Sigma Aldrich Life Science, St Louis, MO). The resulting cell suspension was filtered through sterile gaze in 50 ml falcon tubes and centrifuged at 50 x g for 3 min at room temperature. The cell pellet was resuspended in 30 ml WME and centrifuged through a Percoll® gradient for 2 times at 100 x g. The hepatocytes were washed again with Williams' medium E (WME) and resuspended in medium containing 5% Fetal calf serum (FCS, purchased from Invitrogen, Auckland, NZ). Cell viability was determined by trypan blue exclusion. For the metabolic stability assay liver cells were distributed in WME containing 5% FCS to glass vials at a density of 1.0 x 10 6 vital cells/ml. The test compound was added to a final concentration of 1 μΜ. During incubation, the hepatocyte suspensions were continuously shaken and aliquots were taken at 2, 8, 16, 30, 45 and 90 min, to which equal volumes of cold acetonitrile were immediately added. Samples were frozen at -20° C over night, after subsequently centrifuged for 15 minutes at 3000 rpm and the supernatant was analyzed with an Agilent 1200 HPLC-system with LCMS/MS detection. The half- life of a test compound was determined from the concentration-time plot. From the half- life the intrinsic clearances were calculated. Together with the additional parameters liver blood flow, amount of liver cells in vivo and in vitro, the maximal oral bioavailability (Fmax) was calculated using the following scaling parameters: Liver blood flow (rat) - 4.2 L/h/kg; specific liver weight - 32 g/kg rat body weight; liver cells in vivo- 1.1 x 10 s cells/g liver, liver cells in vitro - 0.5 x 10 6 /ml.

7. In vivo pharmacokinetics in rats

For in vivo pharmacokinetic experiments test compounds were administered to male Wistar rats intravenously at doses of 0.3 to 1 mg/kg formulated as solutions using either rat plasma or solubilizers such as PEG400 in well-tolerated amounts.

For pharmacokinetics after intravenous administration test compounds were given as i.v. bolus and blood samples were taken at 2 min, 8 min, 15 min, 30 min, 45 min, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h after dosing. Depending on the expected half- life additional samples were taken at later time points (e.g. 48 h, 72 h). Blood was collected into Lithium-Heparin tubes (Monovetten® , Sarstedt) and centrifuged for 15 min at 3000 rpm. An aliquot of 100 \L from the supernatant (plasma) was taken and precipitated by addition of 400 L ice cold acetonitrile and frozen at -20 °C over night. Samples were subsequently thawed and centrifuged at 3000 rpm, 4 °C for 20 minutes. Aliquots of the supernatants were taken for analytical testing using an Agilent 1200 HPLC-system with LCMS/MS detection. PK parameters were calculated by non-compartmental analysis using a PK calculation software.

PK parameters derived from concentration-time profiles after i.v.: CLplasma: Total plasma clearance of test compound (in L/kg/h); CLblood: Total blood clearance of test compound: CLplasma*Cp/Cb (in L/kg/h) with Cp/Cb being the ratio of concentrations in plasma and blood, AUCnorm: Area under the concentration-time curve from t=0h to infinity (extrapolated) divided by the administered dose (in kg*h/L); ti/2: terminal half-life (in h).

8. Surface Plasmon Resonance PTEFb

Defintions

The term "surface plasmon resonance", as used herein, refers to an optical phenomenon that allows for the analysis of the reversible associations of biological molecules in real time within a biosensor matrix, for example using the Biacore® system (GE Healthcare Biosciences, Uppsala, Sweden). Biacore® uses the optical properties of surface plasmon resonance (SPR) to detect alterations in the refractive index of a buffer, which changes as molecules in solution interact with the target immobilized on the surface. In brief, proteins are covalently bound to the dextran matrix at a known concentration and a ligand for the protein is injected through the dextran matrix. Near infrared light, directed onto the opposite side of the sensor chip surface is reflected and also induces an evanescent wave in the gold film, which in turn, causes an intensity dip in the reflected light at a particular angle known as the resonance angle. If the refractive index of the sensor chip surface is altered (e.g. by compound binding to the bound protein) a shift occurs in the resonance angle. This angle shift can be measured. These changes are displayed with respect to time along the y-axis of a sensorgram, which depicts the association and dissociation of any biological reaction.

The term "KD", as used herein, is intended to refer to the equilibrium dissociation constant of a particular compound / target protein complex.

The term "koff", as used herein, is intended to refer to the off-rate, i.e. the dissociation rate constant of a particular compound / target protein complex.

The term "target residence time", as used herein, is intended to refer to the inverse of the rate of dissociation rate constant ( 1 / koff ) of a particular compound / target protein complex.

For further descriptions see:

Jonsson U et al al., 1993 Ann Biol Clin.;51(l): 19-26.

Johnsson B et al, Anal Biochem. 1991 ;198(2):268-77.

Day Y et al, Protein Science, 2002; 11, 1017-1025

Myskza DG, Anal Biochem., 2004; 329, 316-323

Tummino and Copeland, Biochemistry, 2008;47(20):5481-5492.

Biological activity

The biological activity (e.g. as inhibitors of PTEFb) of the compounds according to the invention can be measured using the SPR assay described.

The level of activity exhibited by a given compound in the SPR assay can be defined in terms of the KD value, and preferred compounds of the present invention are compounds having a KD value of less than 1 micromolar, more preferably less than 0.1 micromolar.

Furthermore, the time in residence at its target of a given compound can be defined in terms of the target residence time (TRT), and preferred compounds of the present invention are compounds having a TRT value of more than 10 minutes, more preferably more than 1 hour. The ability of the compounds according to the invention to bind human PTEFb may be determined using surface plasmon resonance (SPR). KD values and k 0 ff values may be measured using a Biacore® T200 instrument (GE Healthcare, Uppsala, Sweden). For SPR measurements, recombinant human PTEFb (CDK9/Cyclin Tl recombinant human active protein kinase purchased from ProQinase, Freiburg, Germany) is immobilized using standard amine coupling (Johnsson B et al, Anal Biochem. 1991 Nov l;198(2):268-77). Briefly, carboxymethylated dextran biosensor chips (CM7, GE Healthcare) are activated with N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Human PTEFb is diluted in lx HBS-EP+ (GE Healthcare) and injected on the activated chip surface. Subsequently, a 1 :1 solution of 1 M ethanolamine-HCl (GE Healthcare) and lx HBS-EP is injected to block unreacted groups, resulting in approximately 4000 response units (RU) of immobilized protein. A reference surface is generated by treatment with NHS-EDC and ethanolamine-HCl. Compounds are dissolved in 100% dimethylsulfoxide (DMSO, Sigma- Aldrich, Germany) to a concentration of 10 mM and subsequently diluted in running buffer (lx HBS-EP+ pH 7.4 [generated from HBS-EP+ Buffer lOx (GE Healthcare): 0.1 M HEPES, 1.5 M NaCl, 30 mM EDTA and 0.5% v/v Surfactant P20], 1% v/v DMSO). For kinetic measurements, serial dilutions of compound (0.82 nM up to 2 μΜ) are injected over immobilized protein. Binding kinetics is measured at 25 °C with a flow rate of 50 μΐ/min in running buffer. Compound concentrations are injected for 60 s followed by a dissociation time of 1800 s. Slight variations of these parameters are indicated in Table 4a and 4b. SPR measurements performed at 37°C are summarized in Table 4b. The resulting sensorgrams are double-referenced against the reference surface as well as against blank injections.

The double-referenced sensorgrams are fit to a simple reversible Langmuir 1 : 1 reaction mechanism as implemented in the Biacore® T200 evaluation software 2.0 (GE Healthcare). In cases were full compound dissociation has not occurred at the end of the dissociation phase, the Rmax parameter (response at saturation) is fit as local variable. In all other cases, Rmax is fit as global variable.

Preparative Examples

Syntheses of compounds

The syntheses of the macrocyclic compounds of formula (I) according to the present invention are preferably carried out according to the general synthetic sequences as shown in Schemes la, lb, lc, Id, 2, 3a, 3b, 3c, 3d, 3e, 4a, 4b, 4c, 5, 6a, 6b, 6c, 6d, 6e, 6f, 6g, 7a, 7b, 7c, 8a, 8b, 8c, 9a, 9b, 9c, 9d, 9e and 9f.

In addition to said routes described below, also other routes may be used to synthesise the target compounds, in accordance with common general knowledge of a person skilled in the art of organic synthesis. The order of transformations exemplified in the following Schemes is therefore not intended to be limiting, and suitable synthesis steps from various schemes can be combined to form additional synthesis sequences. In addition, modification of any of the substituents R 1 , R 2 , R 3 , R 4 and/or R 5 can be achieved before and/or after the exemplified transformations. These modifications can be such as the introduction of protective groups, cleavage of protective groups, reduction or oxidation of functional groups, halogenation, metallation, metal catalysed coupling reactions, substitution or other reactions known to a person skilled in the art. These transformations include those which introduce a functionality allowing for further interconversion of substituents. Appropriate protective groups and their introduction and cleavage are well-known to a person skilled in the art (see for example T.W. Greene and P.G.M. Wuts in Protective Groups in Organic Synthesis, 4 th edition, Wiley 2006). Specific examples are described in the subsequent paragraphs. Further, it is possible that two or more successive steps may be performed without work-up being performed between said steps, e.g. a "one-pot" reaction, as it is well- known to a person skilled in the art.

The geometry of the sulfinyl and sulfoximine moiety renders some of the compounds of the general formula (I) chiral. Separation of racemic sulfoxides and sulfoximines into their enantiomers can be achieved by methods known to the person skilled in the art, preferably by means of preparative HPLC on chiral stationary phase.

The syntheses of the pyridine derivatives of formulae (11), (15), (15a) and (15b), all of them constituting subsets of the general formula (I) according to the present invention, are preferably carried out according to the general synthetic sequences as shown in Schemes la, lb, lc and Id.

Schemes la, lb and lc, wherein R 1 , R 2 , R 3 , R 4 and R 5 are as defined for the compound of general formula (I) according to the present invention, outline the preparation of pyridine-based macrocyclic compounds of of formulae (11) and (15), from 2-chloro-5-fluoro-4-iodopyridine (1; CAS# 884494-49-9). As outlined in Scheme la, said starting material (1) can be reacted with a boronic acid derivative of formula (2), in which R 3 and R 4 are as defined for the compound of general formula (I), to give a compound of formula (3). The boronic acid derivative (2) may be a boronic acid (R = -H) or an ester of the boronic acid, e.g. its isopropyl ester (R = -0Η(Ο¾)2), or an ester derived from pinacol in which the boronic acid intermediate forms a 2-aryl-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (R-R = -C(C¾)2- C(CH 3 ) 2 -).

Said coupling reaction can be catalyzed by palladium catalysts, e.g. by Pd(0) catalysts such as tetrakis(triphenylphosphine)palladium(0) [Pd(PPh3) i] , tris(dibenzylideneacetone)di-palladium(0) [Pd2(dba)3], or by Pd(II) catalysts such as dichlorobis(triphenylphosphine)-palladium(II) [Pd(PPh3)2Cb], palladium(II) acetate and triphenylphosphine or by [l,l'-bis(diphenylphosphino)ferrocene]palladium dichloride.

The reaction is preferably carried out in a mixture of a solvent such as 1 ,2-dimethoxyethane, dioxane, DMF, THF, or isopropanol with water and in the presence of a base such as potassium carbonate, sodium bicarbonate or potassium phosphate.

(review: D.G. Hall, Boronic Acids, 2005 WILEY- VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527-30991-8 and references cited therein).

The reaction is performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. The reaction is preferably completed after 1 to 36 hours of reaction time. In the second step, a compound of formula (3) can be converted to a compound of formula (4). This reaction can be carried out by a Palladium-catalyzed C-N cross-coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross- Coupling Reactions', 2 nd ed. : A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004).

Preferred is the herein described use of lithium bis(trimethylsilyl)amide, tris(dibenzylideneacetone)dipalladium(0) and 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl in THF. The reactions are preferably run under an atmosphere of argon for 3-24 hours at a temperature of 40°C to 80°C in an oil bath. In the third step, a compound of formula (4) can be converted to a compound of formula (5), by means of cleaving the methyl ether present in compounds of formula (4).

Preferred is the herein described use of boron tribromide in DCM. The reactions are preferably run for 1 to 24 hours at 0°C to room temperature. In the fourth step, a compound of formula (5) can be coupled with a compound of formula (6), in which L is as defined for the compound of general formula (I), and in which LG 1 is a leaving group such as chloro, bromo, iodo, Ci-C4-alkyl-S(=0)20-, trifluoromethanesulfonyloxy-, benzenesulfonyloxy-, or /jara-toluenesulfonyloxy-, preferably bromo, to give a compound of formula (7). This reaction is carried out preferentially in the presence of an inorganic base such as potassium carbonate in MeCN at 80 °C in a sealed vessel and reaction times of 3 to 24 hours. Compounds of the formula (6) are well known to the person skilled in the art and widely commercially available.

Scheme la In the fifth step, and as shown in Scheme lb, a compound of formula (7) can be coupled with a compound of formula (8), in which R 1 and R 2 are as defined for the compound of general formula (I), to give a compound of formula (9). This reaction can be carried out by a Palladium-catalyzed C-N cross- coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions', 2 nd ed.: A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004).

Preferred is the herein described use of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, - biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

Compounds of the formula (8) can be prepared as outlined in Scheme 2, infra.

In the sixth step, a compound of formula (9) can be converted to a compound of formula (10). This deprotection is preferably run using an aqueous solution of hydrazine in an aliphatic alcohol of the formula Ci-C i-alkyl-OH, preferably MeOH or EtOH, as a solvent, at temperatures ranging from room temperature {i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. Optionally, a cyclic ether such as tetrahydrofuran, tetrahydropyran or 1,4 dioxane can be added as co- solvent in order to support complete dissolution of the starting material. The reaction is preferably completed after 1 to 36 hours of reaction time. (P. G. M. Wuts, T. W. Green, Protective Groups in Organic Synthesis, 4 th Edition, John Wiley & Sons, Hoboken, USA, 2006).

In the seventh step, a compound of formula (10) can be converted to a macrocyclic compound of formula (11). This cyclization reaction can be carried out by an intramolecular Palladium-catalyzed C-N cross- coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions', 2 nd ed.: A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004).

Preferred is the herein described use of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, - biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

As outlined in Scheme lc, a compound of formula (7), in which R , R and L are as defined for the compound of general formula (I), can also be converted to a compound of formula (12). This deprotection is preferably run using an aqueous solution of hydrazine in an aliphatic alcohol of the formula Ci-C i-alkyl-OH, preferably MeOH or EtOH, as a solvent, at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. Optionally, a cyclic ether such as tetrahydrofuran, tetrahydropyran or 1,4 dioxane can be added as co- solvent in order to support complete dissolution of the starting material. The reaction is preferably completed after 1 to 36 hours of reaction time. (P. G. M. Wuts, T. W. Green, Protective Groups in Organic Synthesis, 4 th Edition, John Wiley & Sons, Hoboken, USA, 2006).

In the next step, a compound of formula (12) can be coupled with a compound of formula (13), in which R 1 , R 2 and R 5 are as defined for the compound of general formula (I), to give a compound of formula

(14) . This reaction is preferentially carried out in the presence of an organic or inorganic base, such as pyridine which is optionally substituted on, two or three times, identically or differently, with C1-C3- alkyl-, such as lutidine, or an aliphatic amine of the formula (Ci-C3-alkyl-)3N or an alkali carbonate, preferentially NN-diisopropylethylamine in a dipolar aprotic solvent such as a carboxamide based solvent or dimethyl sulfoxide, preferably NMP. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 160°C in a microwave oven or in an oil bath.

Compounds of the formula (13) can be prepared as outlined in Scheme 2, infra.

Subsequently, a compound of formula (14) can be converted to a macrocyclic compound of formula

(15) . This cyclization reaction can be carried out by an intramolecular Palladium-catalyzed C-N cross- coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L.

Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions', 2 nd ed.: A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004).

Preferred is the herein described use of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, - biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath. In case the R 5 group present in the compound formula (15) is a protective group, such as trifluoroacetyl- (-C(=0)CF3), tert-butoxycarbonyl- (-C(=0)OC(CH3)3; also referred to herein as "Boc"), or benzyloxycarbonyl- (-C(=0)OCH2Ph), R 5 can be readily converted into a hydrogen atom, using methods well known to the person skilled in the art.

114 As outlined in Scheme Id, N-unprotected sulfoximines of formula (15a) (R 5 = H) may be further converted into N-functionalized derivatives of formula (15b). There are multiple methods for the preparation of N-functionalized sulfoximines by functionalization of the nitrogen of the sulfoximine group:

- Alkylation: see for example: a) U. Liicking et al, US 2007/0232632; b) C.R. Johnson, J. Org. Chem. 1993, 58, 1922; c) C. Bolm et al, Synthesis 2009, 10, 1601.

- Acylation: see for example: a) C. Bolm et al, Chem. Europ. J. 2004, 10, 2942; b) C. Bolm et al, Synthesis 2002, 7, 879; c) C. Bolm et al, Chem. Eur. J. 2001, 7, 1118.

- Arylation: see for example: a) C. Bolm et al, Tet. Lett. 1998, 39, 5731 ; b) C. Bolm et al., J. Org. Chem. 2000, 65, 169; c) C. Bolm et al, Synthesis 2000, 7, 911 ; d) C. Bolm et al, J. Org. Chem. 2005, 70, 2346; e) U. Lucking et al, WO2007/71455.

- Reaction with isocyanates: see for example: a) V.J. Bauer et al, J. Org. Chem. 1966, 31, 3440; b) C. R. Johnson et al, J. Am. Chem. Soc. 1970, 92, 6594; c) S. Allenmark et al, Acta Chem. Scand. Ser. B 1983, 325; d) U. Lucking et al, US2007/0191393.

- Reaction with sulfonylchlorides: see for example: a) D.J. Cram et al, J. Am. Chem. Soc. 1970, 92, 7369; b) C.R. Johnson et al, J. Org. Chem. 1978, 43, 4136; c) A.C. Barnes, J. Med. Chem. 1979, 22, 418; d) D. Craig et al, Tet. 1995, 51, 6071 ; e) U. Lucking et al, US2007/191393.

- Reaction with chloroformiates: see for example: a) P.B. Kirby et al, DE2129678; b) D.J. Cram et al, J. Am. Chem. Soc. 1974, 96, 2183; c) P. Stoss et al, Chem. Ber. 1978, 111, 1453; d) U. Lucking et al,

WO2005/37800.

- Reaction with bromocyane: see for example: a) D.T. Sauer et al, Inorganic Chemistry 1972, 1 1, 238; b) C. Bolm et al, Org. Lett. 2007, 9, 2951 ; c) U. Lucking et al, WO 2011/29537.

Compounds of the formula (8), in which R 1 and R 2 are as defined for the compound of general formula (I) and compounds of the formula (13), in which R 1 , R 2 and R 5 are as defined for the compound of general formula (I), can be prepared according to Scheme 2, starting e.g. from a 2,6-dichloroisonicotinic acid derivative of formula (16), in which R 2 is as defined for the compound of general formula (I), which can be reduced to the corresponding pyridinemethanol of formula (17), by means of reduction. Preferred is the herein described use of sulfanediyldimethane - borane (1 : 1 complex) in tetrahydrofuran.

Derivatives of isonicotinic acid of formula (16), and esters thereof, are well known to the person skilled in the art, and are often commercially available. In a second step, a pyridinemethanol of formula (17) can be reacted to give a compound of formula (18), in which LG 2 represents a leaving group such as chloro, bromo, iodo, Ci-C4-alkyl-S(=0)20-, trifluoromethanesulfonyloxy-, benzenesulfonyloxy-, or /jara-toluenesulfonyloxy-. Such conversions are well known to the person skilled in the art; preferred is the herein described use of methanesulfonyl chloride in the presence of triethylamine as a base, in dichloromethane as a solvent, to give a compound of formula (18) in which LG 2 represents methanesulfonyloxy-, or the herein described use of thionyl chloride in the presence of pyridine as a base, in toluene as a solvent, to give a compound of formula (18) in which LG 2 represents chloro.

In a third step, a compound of formula (18) can be reacted with a thiol (or a salt of said thiol) of the formula R'-SH, in which R 1 is as defined for the compound of general formula (I), to give a thioether derivative of formula (8). Thiols of the formula R'SH, and salts thereof, are well known to the person skilled in the art and are commercially available in considerable variety.

Oxidation of a thioether of formula (8) can be used to obtain the corresponding sulfoxide of formula (19). The oxidation can be performed analogously to known processes (see for example: (a) M.H. Ali et al, Synthesis 1997, 764; (b) M.C. Carreno, Chem. Rev. 1995, 95, 1717; (c) I. Patel et al, Org. Proc. Res. Dev. 2002, 6, 225; (d) N. Khiar et al, Chem. Rev. 2003, 103, 3651).

Preferred is the herein described use of periodic acid und iron(III)chloride. Rhodium-catalyzed imination of a sulfoxide of formula (19) can be used to prepare the corresponding sulfoximines of formula (13) (see for example: a) Bolm et al, Org. Lett. 2004, 6, 1305; b) Bull et al, J. Org. Chem. 2015, 80, 6391). In this type of reaction, R 5 preferably represents a -C(=0)R 8 or C(=0)OR 8 group, in which R 8 is as defined for the compound of general formula (I); more preferably, R 5 represents a group selected from trifluoroacetyl- (-C(=0)CF3), teri-butoxycarbonyl- (-C(=0)OC(CH3)3), and, as described herein, benzyloxycarbonyl- (-C(=0)OCH2Ph).

13

Scheme 2

The syntheses of pyridine derivatives of formulae (25), (26), (27), (27a) and (28), all of them constituting further subsets of the general formula (I) according to the present invention, are preferably carried out according to the general synthetic sequences as shown in Schemes 3a, 3b, 3c, 3d and 3e.

As outlined in Scheme 3a, a compound of formula (5, prepared as described above; see Scheme la), in which R 3 and R 4 are as defined for the compound of general formula (I), can be converted into a triflate compound of formula (20), in which OTf represents -OS(=0)2CF3, in the first step. This reaction is carried out preferentially using N-phenyl trifluoromethanesulfonimide and a base, preferentially trimethylamine, in solvents such as DCM at temperatures from room temperature to reflux and reaction times from 1 to 24 hours. In a second step, a compound of formula (20) can be coupled with an alkyne of formula (21), in which L' represents a Ci-Cvalkylene group featuring one carbon atom less as compared to the corresponding group L in formula (23), to give a compound of formula (22). Said coupling reaction, also known as Sonogashira coupling, is catalyzed by palladium catalysts, e.g. by Pd(0) catalysts such as tetrakis(triphenylphosphine)palladium(0) [Pd(PPh3) i], tris(dibenzylideneacetone)di-palladium(0) [Pd2(dba)3], or by Pd(II) catalysts such as dichlorobis(triphenylphosphine)-palladium(II) [Pd(PPh3)2Cb], also being referred herein to as bis(triphenylphosphino)palladium(II) chloride, or by palladium(II) acetate and triphenylphosphine or by [l, -bis(diphenylphosphino)ferrocene]palladium dichloride. The reaction is promoted by a copper(I) salt such as copper(I) iodide.

The reaction is preferably carried out in a solvent such as 1 ,2-dimethoxyethane, dioxane, DMF, THF and in the presence of a base such as triethylamine, DIPEA or diisopropylamine. The reaction is performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. The reaction is preferably completed after 1 to 36 hours of reaction time. (For a review on the Sonogashira reaction, see: R. Chinchilla, C. Najera, Chem. Rev. 2007, 107, 874- 922)

Compounds of formula (21) are well known to the person skilled in the art and are widely commercially available.

In a third step, a compound of formula (22) can be hydrogenated to give a compound of formula (23). This reaction is preferentially run in the presence of a hydrogenation catalyst such as palladium on activated charcoal under positive pressure of hydrogen (1 to 50 bar) and solvents such as MeOH, EtOH or THF. (For an overview on heterogeneous catalytic hydrogenation, see: S. Nishimura, 'Handbook of Heterogeneous Catalytic Hydrogenation for Organic Synthesis', Wiley- VCH, Weinheim, 2001).

In a fourth step and as shown in Scheme 3b, a compound of formula (23) can be coupled with a compound of formula (8, see Scheme 2), in which R 1 and R 2 are as defined for the compound of general formula (I), to provide a compound of formula (24). This coupling is preferentially carried out in the presence of a base such as alkali carbonates, alkali phosphates, alkali hydrides or organic bases such as alkali tert-butoxides or amines. The reaction is preferentially run in solvents such as diethylether, methyl- teri-butyl ether, cyclopentylmethyl ether, THF, DMF, DME, NMP or DMSO at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent.

Scheme 3b

In a fifth step and as shown in Scheme 3c, a compound of formula (24) is converted to a macrocyclic compound of formula (25). This cyclization reaction can be carried out by an intramolecular Palladium- catalyzed C-N cross-coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions ', 2 nd ed.: A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004).

Preferred is the herein described use of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, - biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

Scheme 3c As outlined in Scheme 3d, oxidation of a thioether of formula (25) can be used to obtain the corresponding sulfoxide of formula (26). The oxidation can be performed analogously to known processes (see for example: (a) M.H. Ali et al, Synthesis 1997, 764; (b) M.C. Carreno, Chem. Rev. 1995, 95, 1717; (c) I. Patel et al, Org. Proc. Res. Dev. 2002, 6, 225; (d) N. Khiar et al, Chem. Rev. 2003, 103, 3651).

Preferred is the herein described use of periodic acid und iron(III) chloride.

Imination of a sulfoxide of formula (26) gives the corresponding unsubstituted sulfoximine of formula (27). Preferred is the well-known use of sodium azide and sulfuric acid in trichloromethane or DCM at 45°C (see for example: a) H. R. Bentley et al, J. Chem. Soc. 1952, 1572; b) C. R. Johnson et al, J. Am. Chem. Soc. 1970, 92, 6594; c) Satzinger et al, Angew. Chem. 1971, 83, 83).

Furthermore, thioethers of formula (25) can also be oxidized to the corresponding sulfones of formula (28). The oxidation can be prepared analogously to known processes (see for example: Sammond et al; Bioor . Med. Chem. Lett. 2005, 15, 3519).

Scheme 3d N-unprotected sulfoximines of formula (27) (R 5 = H) may be further converted into N-functionalized derivatives of formula (27a), as shown in Scheme 3e. There are multiple methods for the preparation of N-functionalized sulfoximines by functionalization of the nitrogen of the sulfoximine group; for details see the references listed in context of the conversion of N-unprotected sulfoximines of formula (15a) (R 5 = H) into N-functionalized derivatives of formula 15b) / Scheme lc.

Scheme 3e The syntheses of the pyrimidine derivatives of formula (35), (35a) and (35b), constituting further subsets of the general formula (I) according to the present invention, are preferably carried out according to the general synthetic sequences as shown in Schemes 4a, 4b and 4c.

Schemes 4a, 4b and 4c, wherein R 1 , R 2 , R 3 , R 4 and R A and L are as defined for the compound of general formula (I) according to the present invention, outline the preparation of pyrimidine compounds of the general formula (I) from 2,4-dichloro-5-fluoropyrimidine (CAS# 2927-71-1, 29).

As shown in Scheme 4a, said starting material (29) can be reacted with a boronic acid derivative of formula (2) to give a compound of formula (30). The boronic acid derivative (2) may be a boronic acid (R = -H) or an ester of the boronic acid, e.g. its isopropyl ester (R = -CH(Ci¾)2), or an ester derived from pinacol in which the boronic acid intermediate forms a 2-aryl-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (R-R = -C(Ci¾)2-C(CH3)2-). Boronic acids and their esters are commercially available and well-known to the person skilled in the art; see e.g. D.G. Hall, Boronic Acids, 2005 WILEY- VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527-30991-8 and references cited therein. The coupling reaction can be catalyzed by Pd catalysts, e.g. by Pd(0) catalysts such as tetrakis(triphenylphosphine)palladium(0) [Pd(PPh3) i] , tris(dibenzylideneacetone)di-palladium(0) [Pd2(dba)3], or by Pd(II) catalysts such as dichlorobis(triphenylphosphine)-palladium(II) [Pd(PPh3)2Cb], palladium(II) acetate and triphenylphosphine or by [l,l'-bis(diphenylphosphino)ferrocene]palladium dichloride [Pd(dppf)Cl 2 ]. The reaction is preferably carried out in a mixture of a solvent such as 1 ,2-dimethoxyethane, dioxane, DMF, THF, or isopropanol with water and in the presence of a base such as aqueous potassium carbonate, aqueous sodium bicarbonate or potassium phosphate. The reaction is performed at temperatures ranging from room temperature (=20°C) to the boiling point of the solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven, (review: D.G. Hall, Boronic Acids, 2005 WILEY- VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527-30991-8 and references cited therein). The reaction is preferably completed after 1 to 36 hours of reaction time.

In the second step, a compound of formula (30) is converted to a compound of formula (31), by means of cleaving the methyl ether present in the compound of formula (30). Preferred is the herein described use of boron tribromide in DCM. The reactions are preferably run for 1 to 24 hours at 0°C to room temperature.

In the third step, a compound of formula (31) is coupled with a compound of formula (32) to give a compound of formula (33). This reaction can be carried out by a Mitsunobu reaction (see for example: a) K.C.K. Swamy et al, Chem. Rev. 2009, 109, 2551). Compounds of the formula (32) can be prepared as outlined in Scheme 5 infra

Scheme 4a As outlined in Scheme 4b, a compound of formula (33), wherein R 1 , R 2 , R 3 , R 4 , L and R A are as defined for the compound of general formula (I) according to the present invention, can be reduced to give an aniline of formula (34). The reduction can be prepared analogously to known processes (see for example: (a) Sammond et al; Bioorg. Med. Chem. Lett. 2005, 15, 3519; (b) R.C. Larock, Comprehensive Organic Transformations, VCH, New York, 1989, 411-415). Preferred is the herein described use of hydrogen gas in the presence of commercial catalysts containing platinum and vanadium on carbon, preferably on activated carbon, in a suitable solvent such as an aliphatic alcohol of the formula Ci-C3-alkyl-OH, optionally containing a cyclic ether such as tetrahydroiuran or 1,4-dioxane as co-solvent, preferably in methanol or a mixture of methanol and tetrahydroiuran. Alternatively, titanium(III)chloride in a mixture of aqueous hydrochloric acid and tetrahydroiuran can be used.

The resulting compounds of formula (34) can be converted to a macrocyclic compound of formula (35). This cyclization reaction can be carried out by an intramolecular Palladium-catalyzed C-N cross- coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions', 2 nd ed.: A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004). Preferred is the herein described use of chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath. Finally, the tert-butoxycarbonyl- group attached to the sulfoximine nitrogen can be cleaved under acidic conditions to give unprotected sulfoximines of formula (35a) (see for example: J.A. Bull, J. Org. Chem. 2015, 80, 6391). Preferred is the herein described use of an acid, preferably trifluoroacetic acid in dichloromethane as a solvent.

Scheme 4b

Scheme 4c, wherein R 1 , R 2 , R 3 , R 4 , R A and L are as defined for the compound of general formula (I) according to the present invention, outlines the preparation of an N-substituted sulfoximine compound of the formula (35b) from an N-unsubstituted sulfoximine compound of the formula (35a). An N-unprotected sulfoximine of formula (35a) (R 5 = H) may be reacted to to give an N-functionalized derivative of formula (35b). Formulae (35a) and (35b) both constitute further subsets of the general formula (I). There are multiple methods known for the preparation of N-functionalized sulfoximines by functionalization of the nitrogen of the sulfoximine group; for details see the references listed in context of the conversion of N-unprotected sulfoximines of formula (15a) (R 5 = H) into N-functionalized derivatives of formula (15b) / Scheme lc.

Compounds of the formula (32), in which R 1 and R 2 are as defined for the compound of general formula (I) according to the present invention, can be prepared according to Scheme 5, starting e.g. from a sulfoxide derivative of the formula (36), in which R 1 and R 2 are as defined for the compound of general formula (I) according to the present invention, which are reacted to give a Boc-protected sulfoximine compound of formula (37) (see for example: J.A. Bull, J. Org. Chem. 2015, 80, 6391).

In a second step, a compound of formula (37) is reacted with an amine of the formula HO-L-NHR A , in which L and R A are as defined for the compound of general formula (I) according to the present invention to give compounds of the formula (32). Sulfoxide derivatives of formula (36) and methods for their preparation are known to the person skilled in the art (see e.g. WO 2013037894).

Scheme 5

The syntheses of pyrimidine derivatives of formulae (45). (45a) and (45b), all of them constituting further subsets of the general formula (I) according to the present invention, can be preferably carried out according to the general synthetic sequences as shown in Schemes 6a, 6b, 6c, 6d and 6e.

Scheme 6a In a first step, pyrimidine of formula (29) can be reacted with a boronic acid derivative of formula (38), in which R 3 and R 4 are as defined for the compound of general formula (I), to give a compound of formula (39). The boronic acid derivative (38) may be a boronic acid (R = -H) or an ester of the boronic acid, e.g. its isopropyl ester (R = -CH(Ci¾)2), or an ester derived from pinacol in which the boronic acid intermediate forms a 2-aryl-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (R-R = -0(Ο¾)2-0(Ο¾)2-).

Said coupling reaction can be catalyzed by palladium catalysts, e.g. by Pd(0) catalysts such as tetrakis(triphenylphosphine)palladium(0) [Pd(PPh3) i] , tris(dibenzylideneacetone)di-palladium(0) [Pd2(dba)3], or by Pd(II) catalysts such as dichlorobis(triphenylphosphine)-palladium(II) [Pd(PPh3)2Cb], palladium(II) acetate and triphenylphosphine or by [l,l'-bis(diphenylphosphino)ferrocene]palladium dichloride.

The reaction can be preferably carried out in a mixture of a solvent such as 1 ,2-dimethoxyethane, dioxane, DMF, THF, or isopropanol with water and in the presence of a base such as potassium carbonate, sodium bicarbonate or potassium phosphate, (review: D.G. Hall, Boronic Acids, 2005 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527-30991-8 and references cited therein).

The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. The reaction is preferably completed after 1 to 36 hours of reaction time.

In a second step, the compounds of formula (39) can be reacted with 2-nitrobenzenesulfonyl chloride (NsCl) to give compounds of the formula (40). This reaction can be carried out in the presence of an organic base, preferably pyridine, and catalytic amounts of 4-dimethylaminopyridine in solvents such as dichloromethane. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

In a third step, compounds of the formula (40), in which Ns represents a 2-nitrobenzenesulfonyl group, can be reacted with with an alcohol of formula (41), in which R 1 , R 2 and L are as defined for the compound of the general formula (I) and which can be prepared according to Scheme 6e, in the presence of a tertiary phosphine, such as triphenylphosphine, and a dialkyl diazodicarboxylate (known as Mitsunobu reaction, see for example: K.C.K. Swamy et al, Chem. Rev. 2009, 109, 2551), to yield compounds of formula (42). Preferred is the use of diisopropyl azodicarboxylate and triphenylphosphine as coupling reagent in a solvent such as dichloromethane or THF. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Scheme 6b

As outlined in Scheme 6b, compounds of the formula (42), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of the general formula (I), and in which Ns represents a 2-nitrobenzenesulfonyl group, can be reacted with thiophenol to yield compounds of the formula (43). This reaction can be carried out in the presence of an organic or an inorganic base, such as cesium carbonate and preferably in a carboxamide based solvent, such as DMF or NMP. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Subsequently, the resulting compounds of formula (43) can be reduced to give aniline derivatives of formula (44). The reduction can be performed analogously to known processes (see for example: (a) Sammond et al; Bioorg. Med. Chem. Lett. 2005, 15, 3519; (b) R.C. Larock, Comprehensive Organic Transformations, VCH, New York, 1989, 411-415). Preferred is the herein described use of platinum and vanadium on activated carbon under an atmosphere of hydrogen gas in a solvent mixture of methanol and THF. (For an overview on heterogeneous catalytic hydrogenation, see: S. Nishimura, 'Handbook of Heterogeneous Catalytic Hydrogenation for Organic Synthesis', Wiley- VCH, Weinheim, 2001).

Scheme 6c

The resulting compounds of formula (44) can be converted to macrocyclic compounds of formula (45) (Scheme 6c). This cyclization reaction can be carried out by an intramolecular Palladium-catalyzed C-N cross-coupling reaction (see above, e.g. Scheme lb).

Preferred is the herein described use of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, - biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-teri-butylether adduct, 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions are preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

Finally, the teri-butoxycarbonyl- group attached to the sulfoximine nitrogen can be cleaved under acidic conditions to give unprotected sulfoximines of formula (45a) (see for example: J.A. Bull, J. Org. Chem. 2015, 80, 6391). Preferred is the herein described use of an acid, preferably trifluoroacetic acid in dichloromethane as a solvent.

Scheme 6d, wherein R 1 , R 2 , R 3 , R 4 , R 5 and L are as defined for the compound of general formula (I) according to the present invention, outlines the preparation of N-substituted sulfoximine compounds of the formula (45b) from N-unsubstituted sulfoximine compounds of the formula (45a).

Scheme 6d N-Unprotected sulfoximines of formula (45a) (R 5 = H) may be reacted to give N-functionalized derivatives of formula (45b). Formulae (45a) and (45b) both constitute further subsets of the general formula (I). There are multiple methods known for the preparation of N-functionalized sulfoximines by functionalization of the nitrogen of the sulfoximine group; for details see the references listed in context of the conversion of N-unprotected sulfoximines of formula (15a) (R 5 = H) into N-functionalized derivatives of formula (15b) / Scheme lc.

Compounds of the formula (41) can be obtained as outlined in Scheme 6e. In a first step, a thioether derivative of formula (46), in which R 1 and R 2 are as defined for the compound of general formula (I), can be reacted with a carboxylic ester of formula (47), in which R E represents a Ci-C i-alkyl group, and LG 3 represents a leaving group such as chloro, bromo, iodo, Ci-C4-alkyl-S(=0)20-, trifluoromethanesulfonyloxy-, benzenesulfonyloxy-, or /jara-toluenesulfonyloxy-, and in which L' represents a Ci-Cs-alkylene group featuring one carbon atom less as compared to the corresponding group L in formula (41), L in turn being as defined for the the compound of general formula (I), in the presence of a base, such as an alkali carbonate, preferably potassium carbonate, in N,N- dimethylformamide (DMF) as a solvent, to give a compound of formula (48). Compounds of the formula (46) are known to the person skilled in the art and described in literature (see e.g. WO 2015/155197).

Scheme 6e In a second step, oxidation of a thioether of formula (48) can be used to obtain the corresponding sulfoxide of formula (49). The oxidation can be performed analogously to known processes as outlined above (e.g. as discussed in context of Scheme 3d, conversion of compounds of formula (25) into compounds of formula (26)). Preferred is the herein described use of periodic acid und iron(III)chloride. In a third step, Rhodium-catalyzed imination of a sulfoxide of formula (49) can be used to prepare the corresponding sulfoximines of formula (50) (see for example: a) Bolm et al, Org. Lett. 2004, 6, 1305; b) Bull et al, J. Org. Chem. 2015, 80, 6391). This type of reaction can be also performed with R 5 groups different from teri-butoxycarbonyl- (-C(=0)OC(CH3)3) as shown here, R 5 optionally representing a -C(=0)R 8 or C(=0)OR 8 group, in which R 8 is as defined for the compound of general formula (I); more specifically, R 5 may represent a group such as trifluoroacetyl- (-C(=0)CF3), or benzyloxycarbonyl- (-C(=0)OCH 2 Ph).

In a fourth step, an ester of the formula (50) can be reduced using a reducing agent such as lithium aluminium hydride or di-z o-butylaluminiumhydride (DIBAL), in an ether, preferably tetrahydrofuran, as a solvent, to give compound of the formula (41) which can be further elaborated e.g. as shown in Scheme 6a.

Scheme 6f Alternatively, macrocycles of the formula (45) can be obtained as outlined in Schemes 6f and 6g.

In a first step, phenol derivatives of the formula (51), in which R 1 and R 2 are as defined for the compound of general formula (I), can be reacted with a compound of formula (6), in which L is as defined for the compound of general formula (I), and in which LG 1 is a leaving group such as chloro, bromo, iodo, Ci-C4-alkyl-S(=0)20-, trifluoromethanesulfonyloxy-, benzenesulfonyloxy-, or para- toluenesulfonyloxy-, preferably bromo, to give compounds of formula (52). This reaction can be carried out preferentially in the presence of an inorganic base such as potassium carbonate in a carboxamide solvent such as DMF or NMP. The reaction can be preferably carried out at temperatures ranging from room temperature (= 20 °C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Phenols of the formula (51) are well known to the person skilled in the art and literature known. Compounds of the formula (6) are well known to the person skilled in the art and widely commercially available.

In a next step, oxidation of thioethers of formula (52) can be used to obtain the corresponding sulfoxides of formula (53). The oxidation can be performed analogously to known processes as outlined above {e.g. as discussed in context of Scheme 3d, conversion of compounds of formula (25) into compounds of formula (26)). Preferred is the herein described use of periodic acid und iron(III)chloride.

Rhodium-catalyzed imination of sulfoxides of formula (53) can be used to prepare the corresponding sulfoximines of formula (54) (see for example: a) Bolm et al, Org. Lett. 2004, 6, 1305; b) Bull et al, J. Org. Chem. 2015, 80, 6391). This type of reaction can be also performed with R 5 groups different from teri-butoxycarbonyl- (-C(=0)OC(CH3)3) as shown here, R 5 optionally representing a -C(=0)R 8 or C(=0)OR 8 group, in which R 8 is as defined for the compound of general formula (I); more specifically, R 5 may represent a group such as trifluoroacetyl- (-C(=0)CF3), or benzyloxycarbonyl- (-C(=0)OCH 2 Ph).

Subsequently, compounds of formula (54) can be reduced to give aniline derivatives of formula (55). This conversion can be accomplished according to known methods, e.g. as discussed in context of Scheme 6b, conversion of compounds of formula (43) into compounds of formula (44). Preferred is the herein described use of platinum and vanadium on activated carbon under an atmosphere of hydrogen gas in a solvent mixture of methanol and THF.

Scheme 6g

In a next step, anilines of the formula (55) can be reacted with 2-chloro pyrimidines of the formula (56), in which R 3 and R 4 are as defined for the compound of general formula (I), to give compounds of formula (57). Said 2-chloro pyrimidines of formula (56) are well known to the person skilled in the art and are often commercially available, or can be prepared according to known methods, e.g. as discussed in context of Scheme 1 a. This reaction can be carried out by an intermolecular Palladium-catalyzed C-N cross-coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal- Catalyzed Cross-Coupling Reactions', 2 nd ed. : A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004). Preferred is the herein described use of chloro(2-dicyclohexylphosphino- 2',4',6'-tri-z ' ,yo-propyl- 1 , 1 '-biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a C1-C3- alkylbenzene and a carboxamide based solvent or an ethereal solvent such as 1,4-dioxane, preferably a mixture of toluene and NMP, as a solvent. The reactions can be preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

Said compounds of formula (57) can be further converted to free amino compounds of formula (58). This deprotection reaction can be preferably run using an aqueous solution of hydrazine in an aliphatic alcohol of the formula Ci-C i-alkyl-OH, preferably MeOH or EtOH, as a solvent, at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. Optionally, a cyclic ether such as tetrahydrofuran, tetrahydropyran or 1 ,4 dioxane can be added as co-solvent in order to support complete dissolution of the starting material. The reaction is preferably completed after 1 to 36 hours of reaction time.

Finally, the macrocyclic compounds of formula (45) can be obtained by an intramolecular nucleophilic discplacement. The reaction can be carried out by treating amine compounds of the formula (58) in the presence of an organic base such as triethylamine, ethyldiisopropylamine or 2,6-lutidine in polar solvents such as DMSO. The reaction can be preferably carried out at temperatures ranging from 120 °C to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Schemes 7a, b and c outline the synthesis of compounds of the formulae (66), (66a) and (66b), all of them constituting further subsets of the general formula (I) according to the present invention.

In a first step, compounds of the formula (59), in which L and R A are as defined for the compound of general formula (I), and which are known to the person skilled in the art and are typically commercially available, can be reacted with phthalimide in the presence of a tertiary phosphine, such as triphenylphosphine, and a dialkyl diazodicarboxylate (known as Mitsunobu reaction, see for example: K.C.K. Swamy et al, Chem. Rev. 2009, 109, 2551), to yield compounds of formula (60). Preferred is the use of diisopropyl azodicarboxylate and triphenylphosphine as coupling reagent in a solvent such as dichloromethane or THF. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time. In a second step, the feri-butoxycarbonyl group in compounds of formula (60) can be cleaved off to yield compounds of formula (61). This reaction can be carried out in the presence of a Bronstedt acid, preferably trifluoroacetic acid or hydrochloric acid, in dichloromethane. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Scheme 7a

In a third step, compounds of the structure (37), in which R 1 and R 2 are as defined for the compound of general formula (I) and the synthesis of which is outlined above in Scheme 5, can be reacted with amines of formula (61) to yield compounds of formula (62). This conversion can be preferably carried out in the presence of an organic base such as triethylamine and in DMSO. The reaction can be preferably carried out at temperatures ranging from 120 °C to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

In a fourth step, compounds of formula (62) can be reduced to give aniline derivatives of formula (63). This conversion can be accomplished according to known methods, e.g. as discussed in context of Scheme 6b, conversion of compounds of formula (43) into compounds of formula (44). Preferred is the herein described use of platinum and vanadium on activated carbon under an atmosphere of hydrogen as in a solvent mixture of methanol and THF.

Scheme 7b In a next step, anilines of the formula (63) can be reacted with a 2-chloro pyrimidine of the formula (56) (see also Scheme 6g), to give compounds of formula (64). This coupling reaction can be carried out by an intermolecular Palladium-catalyzed C-N cross-coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions', 2 nd ed. : A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004). Preferred is the herein described use of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2- aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2-(dicyclohexylphosphino)-2',4',6'- triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent or an ethereal solvent such as 1,4-dioxane, as a solvent. The reactions can be preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

The resulting compounds of formula (64) can be further converted to compounds of formula (65). This deprotection reaction can be preferably run using an aqueous solution of hydrazine in an aliphatic alcohol of the formula Ci-C i-alkyl-OH, preferably MeOH or EtOH, as a solvent, at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. Optionally, a cyclic ether such as tetrahydrofuran, tetrahydropyran or 1 ,4 dioxane can be added as co-solvent in order to support complete dissolution of the starting material. The reaction is preferably completed after 1 to 36 hours of reaction time.

Subsequently, macrocyclic compounds of formula (66) can be obtained by an intramolecular nucleophilic displacement. The reaction is carried out by treating amine compounds of the formula (65) in the presence of an organic base such as triethylamine, ethyldiisopropylamine or 2,6-lutidine in polar solvents such as DMSO. The reaction can be preferably carried out at temperatures ranging from 120 °C to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Finally, the tert-butoxycarbonyl- group attached to the sulfoximine nitrogen in compounds of formula (66) can be cleaved under acidic conditions to give unprotected sulfoximines of formula (66a) (see for example: J.A. Bull, J. Org. Chem. 2015, 80, 6391). Preferred is the herein described use of an acid, preferably trifluoroacetic acid in dichloromethane as a solvent. Optionally, said unprotected sulfoximines of formula (66a) can be converted into N-substituted sulfoximine compounds of the formula (66b). Formulae (66a) and (66b) both constitute further subsets of the general formula (I). There are multiple methods known for the preparation of N-functionalized sulfoximines by functionalization of the nitrogen of the sulfoximine group; for details see the references listed in context of the conversion of N- unprotected sulfoximines of formula (15a) (R 5 = H) into N-functionalized derivatives of formula (15b) / Scheme lc.

Scheme 7c

Schemes 8a, b and c outline the synthesis of thioether compounds of the formulae (72), (72a) and (72b), all of them constituting further subsets of the general formula (I) according to the present invention.

As outlined in Scheme 8a, thiol derivatives of formula (67), in which R 3 and R 4 are as defined for the compound of the general formula (I), can be reacted with an alcohol of formula (41) (see also Scheme

6e), in which R 1 , R 2 and L are as defined for the compound of the general formula (I), in the presence of a tertiary phosphine, such as triphenylphosphine, and a dialkyl diazodicarboxylate (known as Mitsunobu reaction, see for example: K.C.K. Swamy et al, Chem. Rev. 2009, 109, 2551), to yield compounds of formula (68). Thiol derivatives of formula (67) are known to the person skilled in the art and are commercially available in certain cases. Preferred is the use of diisopropyl azodicarboxylate and triphenylphosphine as coupling reagent in as solvent such as dichloromethane or THF. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

141 In a second step, compounds of formula (68) can be reacted with 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi- 1,3,2-dioxaborolane in the presence of a palladium catalyst, preferentially dichlorobis(tricyclohexylphosphine)palladium(II), and an organic or an inorganic base, preferentially potassium acetate, to access compounds of formula (69), in which the two groups R together form a group -C(CH3)2-C(CH3)2-). The reaction can be preferably carried out in an ethereal solvent such as THF or 1,4-dioxane at temperatures from temperatures ranging from room temperature (=20°C) to the boiling point of the solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. The reaction is preferably completed after 1 to 36 hours of reaction time.

In a third step, said boronic acid derivatives of formula (69) can be reacted with 2-chloro-5-fluoro-4- iodopyrimidine (CAS# 884494-49-9) to give compounds of the formula (70).

Said reaction can be catalyzed by Pd catalysts, e.g. by Pd(0) catalysts such as tetrakis(triphenylphosphine)palladium(0) [Pd(PPh3) i] , tris(dibenzylideneacetone)di-palladium(0) [Pd2(dba)3], or by Pd(II) catalysts such as dichlorobis(triphenylphosphine)-palladium(II) [Pd(PPh3)2Cb], palladium(II) acetate and triphenylphosphine or by [l,l'-bis(diphenylphosphino)ferrocene]palladium dichloride [Pd(dppf)Cl 2 ].

The reaction can be preferably carried out in a mixture of a solvent such as 1 ,2-dimethoxyethane, dioxane, DMF, THF, or isopropanol with water and in the presence of a base such as aqueous potassium carbonate, aqueous sodium bicarbonate or potassium phosphate.

The reaction can be performed at temperatures ranging from room temperature (=20°C) to the boiling point of the solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. The reaction is preferably completed after 1 to 36 hours of reaction time, (review: D.G. Hall, Boronic Acids, 2005 WILEY- VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527-30991-8 and references cited therein).

As outlined in Scheme 8b, compounds of formula (70) can be reduced to give aniline derivatives of formula (71) analogously to known processes. This conversion can be accomplished according to known methods, e.g. as discussed in context of Scheme 6b, conversion of compounds of formula (43) into compounds of formula (44). Preferred is the herein described use of platinum and vanadium on activated carbon under an atmosphere of hydrogen gas in a solvent mixture of methanol and THF.

ı43 The resulting compounds of formula (71) can be converted to macrocyclic compounds of formula (72). This cyclization reaction can be carried out by an intramolecular Palladium-catalyzed C-N cross- coupling reaction (for a review on C-N cross coupling reactions see for example: a) L. Jiang, S.L. Buchwald in 'Metal-Catalyzed Cross-Coupling Reactions', 2 nd ed.: A. de Meijere, F. Diederich, Eds.: Wiley- VCH: Weinheim, Germany, 2004). Preferred is the herein described use of chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct, 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl as catalyst and ligand, an alkali carbonate or an alkali phosphate, preferably potassium phosphate, as a base, in a mixture of a Ci-C3-alkylbenzene and a carboxamide based solvent, preferably a mixture of toluene and NMP, as a solvent. The reactions can be preferably run under an atmosphere of argon for 2 to 24 hours at 100 to 130°C in a microwave oven or in an oil bath.

Finally, the tert-butoxycarbonyl- group attached to the sulfoximine nitrogen present in compounds of formula (72) can be cleaved under acidic conditions to give unprotected sulfoximines of formula (72a) (see for example: J.A. Bull, J. Org. Chem. 2015, 80, 6391). Preferred is the herein described use of an acid, preferably trifluoroacetic acid in dichloromethane as a solvent. Optionally, said unprotected sulfoximines of formula (66a) can be converted into N-substituted sulfoximine compounds of the formula (72b).

Scheme 8c

Formulae (72a) and (72b) both constitute further subsets of the general formula (I). There are multiple methods known for the preparation of N-functionalized sulfoximines by functionalization of the nitrogen of the sulfoximine group; for details see the references listed in context of the conversion of N- unprotected sulfoximines of formula (15a) (R 5 = H) into N-functionalized derivatives of formula (15b) / Scheme lc. Schemes 9a, 9b, 9c, 9d and 9e outline an additional synthesis route to compounds of the formula (34a), constituting a subset of formula (34) (in that -NR A - as of the formula (34) is -NH- in formula (34a)), described in context of Scheme 4b.

In a first step, benzylic alcohols of formula (73), in which R 2 is as defined as for the compound of general formula (I), can be converted into the corresponding thioethers of formula (74) by reaction with a halogenating agent, preferably thionyl chloride, in a solvent such as dichloromethane, followed by reaction with a thiol of formula R'-SH or a salt therof, in which R 1 is as defined for the compound of general formula (I), in a solvent such as acetone. Benzylic alcohols of formula (73) as well as thiols of formula R'-SH, and their salts, are known to the person skilled in the art and are commercially available in many cases.

In a second step, said thioethers of formula (74) can be reacted with 2-nitrobenzenesulfonyl chloride (NsCl) to give compounds of the formula (75). This reaction can be carried out in the presence of an organic base, preferably pyridine, and, optionally, catalytic amounts of 4-dimethylaminopyridine, in solvents such as dichloromethane. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Scheme 9a

In a third step, the resulting compounds of formula (75), in which R 1 and R 2 are as defined for the compound of general formula (I), and in which Ns represents a 2-nitrobenzenesulfonyl group, can be reacted with compounds of formula (76), in which R 3 , R 4 and L are as defined as for the compound of general formula (I), in the presence of a tertiary phosphine, such as triphenylphosphine, and a dialkyl diazodicarboxylate (known as Mitsunobu reaction, see for example: K.C.K. Swamy et al, Chem. Rev. 2009, 109, 2551), to yield compounds of formula (77). Preferred is the use of diisopropyl azodicarboxylate and triphenylphosphine as coupling reagent in a solvent such as dichloromethane or THF. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time. Compounds of formula (76) can be prepared according to Scheme 9f.

Scheme 9b In a fourth step, oxidation of thioethers of formula (77) can be used to obtain the corresponding sulfoxides of formula (78). The oxidation can be performed analogously to known processes as outlined above (e.g. as discussed in context of Scheme 3d, conversion of compounds of formula (25) into compounds of formula (26)). Preferred is the herein described use of periodic acid und iron(III)chloride. Rhodium-catalyzed imination of sulfoxides of formula (78) can be used to prepare the corresponding sulfoximines of formula (79) (see for example: a) Bolm et al, Org. Lett. 2004, 6, 1305; b) Bull et al, J. Org. Chem. 2015, 80, 6391). This type of reaction can be also performed with R 5 groups different from teri-butoxycarbonyl- (-C(=0)OC(CH3)3) as shown here, R 5 optionally representing a -C(=0)R 8 or C(=0)OR 8 group, in which R 8 is as defined for the compound of general formula (I); more specifically, R 5 may represent a group such as trifluoroacetyl- (-C(=0)CF3), or benzyloxycarbonyl- (-C(=0)OCH 2 Ph).

Scheme 9c In a fifth step, and as outlined in Scheme 6d, compounds of the formula (79), in which R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of the general formula (I), and in which Ns represents a 2- nitrobenzenesulfonyl group, can be reacted with thiophenol to yield compounds of the formula (80). This reaction can be carried out in the presence of an organic or an inorganic base, such as cesium carbonate and preferably in a carboxamide based solvent, such as DMF or NMP. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time.

Subsequently, compounds of formula (80) can be reduced to give aniline derivatives of formula (34a). which constitutes a subset of formula (34). This conversion can be accomplished according to known methods, e.g. as discussed in context of Scheme 6b, conversion of compounds of formula (43) into compounds of formula (44). Preferred is the herein described use of platinum and vanadium on activated carbon under an atmosphere of hydrogen gas in methanol as a solvent.

Scheme 9d

Scheme 9e, wherein R 1 , R 2 , R 3 , R 4 and L are as defined for the compound of general formula (I) according to the present invention, outlines preparation of macrocyclic sulfoximines of formulae (35c), (35d) and (35e), representing subsets of formulae (35), (35a) and (35b), respectively, in analogy to the corresponding conversions discussed in the context of Schemes 4b and 4c.

Scheme 9e According to Scheme 9f, compounds of formula (31), in which R 3 and R 4 are as defined for the compound of general formula (I) and which are described in context of Scheme 4a, can be reacted with compounds of formula (81), in L is as defined for the compound of general formula (I), and in which PG 1 represents a protective group for a hydroxy group, preferably teri-butyldimethylsilyl-, in the presence of a tertiary phosphine, such as triphenylphosphine, and a dialkyl diazodicarboxylate (known as Mitsunobu reaction, see for example: K.C.K. Swamy et al, Chem. Rev. 2009, 109, 2551), to yield compounds of formula (82). Preferred is the use of diisopropyl azodicarboxylate and triphenylphosphine as coupling reagent in a solvent such as dichloromethane or THF. The reaction can be performed at temperatures ranging from room temperature (i.e. approx. 20°C) to the boiling point of the respective solvent. The reaction is preferably completed after 1 to 36 hours of reaction time

The protective group PG 1 can subsequently be removed using methods known to the person skilled in the art, such as reaction with aqueous hydrochloric acid or tetrabutylammonium fluoride in case PG 1 represents a teri-butyldimethylsilyl- group. Protective groups for a hydroxy group, and methods for their introduction as well for their removal, are well known to the person skilled in the art, see e.g. P. G. M. Wuts, T. W. Green, Protective Groups in Organic Synthesis, 4 th Edition, John Wiley & Sons, Hoboken, USA, 2006.

Scheme 9f

Abbreviations used in the description of the chemistry and in the Examples that follow are:

br. (broad, ¾ NMR signal); CDCI3 (deuterated chloroform); cHex (cyclohexane); DCE (dichloroethane); d (doublet, ¾ NMR signal); DCM (dichloromethane); DIPEA (di-wo-propylethylamine); DMAP (4- NN-dimethylaminopyridine), DME (1,2-dimethoxyethane), DMF (NN-dimethylformamide); DMSO (dimethyl sulfoxide); ES (electrospray); EtOAc (ethyl acetate); EtOH (ethanol); h (hour(s)); ¾ NMR (proton nuclear magnetic resonance spectroscopy); HPLC (High Performance Liquid Chromatography), iPrOH (z o-propanol); m (multiplet, ¾ NMR signal); mCPBA (meia-chloroperoxybenzoic acid), MeCN (acetonitrile), MeOH (methanol); min (minute(s)); MS (mass spectrometry); MTBE (methyl teri-butyl ether); NMP (N-Methylpyrrolidin-2-one); NMR (nuclear magnetic resonance); Pd(dppf)Cl2 ([1,1 '- bis(diphenylphosphino)ferrocene]dichloro palladium(II) complex with dichloromethane); q (quartet, ¾ NMR signal); quin (quintet, ¾ NMR signal); rac (racemic); RT (room temperature); s (singlet, ¾ NMR signal); sat. aq. (saturated aqueous); S1O2 (silica gel); t (triplet, ¾ NMR signal); TFA (trifluoroacetic acid); TFAA (trifluoroacetic anhydride), THF (tetrahydrofuran); UPLC (Ultra-High Performance Liquid Chromatography), UV (ultraviolet), wt-% (percent by weight). 'H-NMR spectra

'H-NMR signals are specified with their multiplicity / combined multiplicities as apparent from the spectrum; possible higher-order effects are not considered. Chemical shifts of the signals (δ) are specified as ppm (parts per million).

Chemical naming:

Chemical names were generated using the ACD/Name software from ACD/Labs. In some cases generally accepted names of commercially available reagents were used in place of ACD/Name generated names.

Salt stoichiometry:

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

Unless specified otherwise, suffixes to chemical names or structural formulae such as "hydrochloride", "trifluoroacetate", "sodium salt", or "x HC1", "x CF3COOH", "x Na + ", for example, are to be understood as not a stoichiometric specification, but solely as a salt form.

This applies analogously to cases in which synthesis intermediates or example compounds or salts thereof have been obtained, by the preparation and/or purification processes described, as solvates, such as hydrates with (if defined) unknown stoichiometric composition.

Preparative HPLC methods:

Autopurifier: acidic conditions General methods for LC-MS analysis

Method a:

Instrament: Waters Acquity UPLCMS SingleQuad; Column: Acquity UPLC BEH CI 8 1.7 μηι, 50x2. lmm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-1.6 min 1- 99% B, 1.6-2.0 min 99% B; flow 0.8 ml/min; temperature: 60 °C; DAD scan: 210-400 nm.

Method b:

Instrament: Waters Acquity UPLCMS SingleQuad; Column: Acquity UPLC BEH CI 8 1.7 μηι, 50x2. lmm; eluent A: water + 0.2 vol % aqueous ammonia (32%>), eluent B: acetonitrile; gradient: 0-1.6 min 1-99% B, 1.6-2.0 min 99% B; flow 0.8 ml/min; temperature: 60 °C; DAD scan: 210-400 nm.

Example 1:

15,19-Difluoro-8-[(methylsulfanyl)methyl]-2,3,4,5-tetrahydro -llH-10,6-(azeno)-12,16-(metheno)- 1,5,11, 13-benzoxatriazacyclooctadecine

Preparation of Intermediate 1.1:

2-Chloro-5-fluoro-4-(4-fluoro-2-methoxyphenyl)pyridine

A batch with 2-chloro-5-fluoro-4-iodopyridine (1000 mg, 3.88 mmol, APAC Pharmaceutical, LLC), (4- fluoro-2-methoxyphenyl)boronic acid (660 mg, 3.88 mmol, Aldrich Chemical Company Inc.) and tetrakis(triphenylphosphin)palladium(0) (449 mg, 0.38 mmol) in 1 ,2-dimethoxyethane (10.0 mL) and an aqueous 2 M solution of potassium carbonate (5.8 mL) was degassed using argon. The batch was stirred under an atmosphere of argon for 4 h at 100 °C. After cooling, the batch was diluted with ethyl acetate and THF and washed with a saturated aqueous solution of sodium chloride. The organic layer was filtered using a Whatman filter and concentrated. The residue was purified by column chromatography (hexane to hexane / ethyl acetate 50%) to give the desired title compound (947 mg, 3.70 mmol).

¾ NMR (400MHz, CDC1 3 , 295 K) δ/ppm = 8.27 (m, 1H), 7.33 (m, 1H), 7.24 (m, 1H), 6.75 (m, 2H), 3.83 (s, 3H).

Preparation of Intermediate 1.2:

5-Fluoro-4-(4-fluoro-2-methoxyphenyl)pyridin-2-amine

A solution of lithium bis(trimethylsilyl)amide in THF (1M, 20.5 mL, 20.5 mmol, Aldrich Chemical Company Inc.) was added to a mixture of 2-chloro-5-fluoro-4-(4-fluoro-2-methoxyphenyl)pyridine (2.50 g, 9.78 mmol, see Intermediate 1.1), tris(dibenzylideneacetone)dipalladium (0) (0.18 g, 0.20 mmol, Aldrich Chemical Company Inc.) and 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (0.19 g, 0.39 mmol, Aldrich Chemical Company Inc.) in THF (16.3 mL) under an atmosphere of argon at room temperature. The mixture was stirred at 60 °C for 6 h. The mixture was cooled to -40 °C and water (10 ml) was added. The mixture was slowly warmed to room temperature under stirring, solid sodium chloride was added and the mixture was extracted twice with ethyl acetate. The combined organic layers were filtered using a Whatman filter and concentrated. The residue was purified by column chromatography on silica gel (hexane to hexane / ethyl acetate 60%>) to give the desired title compound (2.04 g, 8.64 mmol).

¾ NMR (400MHz, CDCI3, 295 K) δ/ppm = 7.95 (1H), 7.20 (1H), 6.72 (2H), 6.46 (1H), 4.33 (2H), 3.61 (3H).

Preparation of Intermediate 1.3:

2- 2-Amino-5-fluoropyridin-4-yl)-5-fluorophenol

A solution of boron tribromide in DCM (1M, 47.1 mL, 47.1 mmol, Aldrich Chemical Company Inc.) was added dropwise to a stirred solution of 5-fluoro-4-(4-fluoro-2-methoxyphenyl)pyridin-2-amine (2.00 g, 8.47 mmol) in DCM (205 mL) at 0 °C. The mixture was slowly warmed to room temperature while stirring overnight. The mixture was cautiously diluted with an aqueous solution of sodium bicarbonate under stirring at 0°C and stirred at room temperature for 1 h. A saturated aqueous solution of sodium chloride was added and the mixture was extracted with ethyl acetate. The combined organic layers were filtered using a Whatman filter and concentrated to give the crude title compound (1.92 g) that was used without further purification.

¾ NMR (400MHz, DMSO-i 6 , 295 K) 8/ppm = 10.21 (1H), 7.84 (1H), 7.19 (1H), 6.71 (2H), 6.39 (1H), 5.80 (2H).

Prepration of Intermediate 1.4:

2-{3-[2-(2-Amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]propy l}-lH-isoindol-l,3(2H)-dione

A suspension of 2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenol (700 mg, 3.15 mmol), 2-(3- bromopropyl)-lH-isoindole-l,3(2H)-dione (1.01 g, 3.78 mmol) and potassium carbonate (870 mg, 6.30 mmol) in MeCN (14 mL) in a sealed tube was heated to 80 °C and stirred for 16 h. The mixture was cooled to room temperature and the reaction was stopped by the addition of water (20 mL). The mixture was extracted with ethyl acetate (3 x 20 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (20 mL), dried over sodium sulfate, filtered and concentrated to obtain the title compound (1.3 g, 90% pure) which was used without further purification in the next step.

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.94 - 2.00 (2H), 3.63 - 3.70 (2H), 4.04 - 4.11 (2H), 5.81 (2H), 6.39 (1H), 6.83 - 6.92 (1H), 6.96 - 7.04 (1H), 7.28 (1H), 7.77 - 7.92 (5H).

Preparation of Intermediate 1.5:

(2,6-Dichloropyridin-4-yl)methanol

To a stirred solution 2,6-dichloroisonicotinic acid (10.0 g, 52.1 mmol) in THF (300 mL) at 0 °C was added a solution of sulfanediyldimethane - borane (1 :1) (16.0 g, 210.5 mmol) in THF. The mixture was allowed to react at room temperature overnight. Then, MeOH (22 mL) was cautiously added to the stirred mixture while cooling with an ice bath. The reaction mixture was diluted with ethyl acetate (300 mL), washed with an aqueous sodium hydroxide solution (IN, 100 mL) and saturated aqueous sodium chloride solution. The organic layer was concentrated and the residue was purified by column chromatography on silica gel (hexane / ethyl acetate = 7: 1 to 3 :1) to give the desired title compound (8.3 g, 46.6 mmol).

¾ NMR (300MHz, CDC1 3 , 295 K) δ/ppm = 7.25 (2H), 4.72 (2H), 2.24 (1H).

Preparation of Intermediate 1.6:

(2,6-Dichloropyridin-4-yl)methyl methanesulfonate

(2,6-Dichloropyridin-4-yl)methanol (1.0 g, 5.62 mmol) was dissolved in DCM (20 mL) and triethyl amine (1.0 g, 9.88 mmol) was added. The resulting mixture was cooled to 0 °C and methanesulfonyl chloride (0.9 g, 7.89 mmol) was added. The mixture was stirred at room temperature for 1 h. By adding an aqueous hydrogen choride solution (IN), the pH value of the mixture was adjusted to 3, before it was extracted three times with ethyl acetate. The combined organic layers were concentrated to give the crude title compound (1.4 g) that was used without further purification.

Preparation of Intermediate 1.7:

2,6-Dichloro-4-[(methylsulfanyl)methyl]pyridine

(2,6-Dichloropyridin-4-yl)methyl methanesulfonate (1.40 g, 5.47 mmol) was dissolved in THF (20 mL) and a mixture of sodium thiomethoxide and sodium hydroxide (wt 1/1, 0.70 g, 5 mmol, supplied by Shanghai DEMO Medical Tech Co., Ltd) was added. The resulting mixture was stirred overnight at room temperature. The reaction mixture was diluted with water (10 mL) and extracted three times with ethyl acetate. The combined organic layers were concentrated and the residue was purified by column chromatography on silica gel (hexane / ethyl acetate = 6:1 to 3: 1) to give the desired product (0.54 g, 2.60 mmol).

¾ NMR (300MHz, CDC1 3 , 295 K) δ/ppm = 7.18 (2H), 3.55 (2H), 1.98 (3H).

Prepration of Intermediate 1.8:

2-(3-{2-[2-({6-Chloro-4-[(methylsulfanyl)methyl]pyridin-2-yl }amino)-5-fluoropyridin-4-yl]-5- fluorophenoxy}propyl)-lH-isoindol-l 3(2H)-dione

To a degassed suspension of 2- {3-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]propyl} -lH- isoindol-l,3(2H)-dione (435 mg, 90% pure, 956 μιηοΐ; see Intermediate 1.4), 2,6-dichloro-4- [(methylsulfanyl)methyl]pyridine (199 mg, 956 μιηοΐ), potassium phosphate (1.01 g, 4.78 mmol) and 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (91.2 mg, 191 μιηοΐ) in toluene (8.7 mL) and NMP (0.87 mL) was added chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2- aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (158 mg, 191 μιηοΐ) and the mixture was heated to 130 °C overnight. Additional 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (46 mg, 95 μιηοΐ) and chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (79 mg, 95 μιηοΐ) was added and the mixture was stirred for an additional 2 h at 130 °C. The mixture was cooled to room temperature and stopped by the addition of water (50 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, filtered (Whatman- filter) and concentrated. The residue was redissolved in diethylether and the solution was washed with brine, dried (Whatman filter) and concentrated. The organic layer was dried (Whatman filter) and concentrated. The crude product was combined with a second reaction batch carried out analogously as described above. The two reaction batches were combined and purified by flash column chromatography (hexanes/ ethyl acetate to ethyl acetate /MeOH) to yield the title compound (750 mg, 80% pure), which was contaminated by catalyst impurities and used without further purification in the next step. Prepration of Intermediate 1.9:

4-[2-(3-Aminopropoxy)-4-fluorophenyl]-N-{6-chlor-4-[(methyls ulfanyl)methyl]pyridin-2-yl}-5- fluoropyridin-2-amine

To a suspension of 2-(3- {2-[2-({6-chloro-4-[(methylsulfanyl)methyl]pyridin-2-yl}amin o)-5- fluoropyridin-4-yl]-5-fluorophenoxy}propyl)-lH-isoindol-l,3( 2H)-dione (400 mg, 80% pure, 0.54 mmol) in EtOH (20 mL) was added an aqueous solution of hydrazine (35 wt-%, 290 μΐ, 3.2 mmol) and the mixture was heated to reflux for 4 h. The mixture was cooled to 5°C, the precipitate was filtered off and the filtrate was concentrated. Purification by flash column chromatography on silica gel (DCM/MeOH) yielded the title compound (116 mg, 0.23 mmol).

'H NMR (400 MHz, DMSO-ifc, 295 K) 8/ppm = 1.65 - 1.80 (2H), 2.01 (3H), 2.58 (2H), 3.24 - 3.53 (2H), 3.68 (2H), 4.06 - 4.15 (2H), 6.89 - 6.98 (2H), 7.05 - 7.16 (1H), 7.35 (1H), 7.54 (1H), 7.72 - 7.90 (2H), 8.03 -8.13 (1H), 8.27 (1H), 10.16 (1H).

Example 1 - Preparation of the End Product:

To a degassed suspension of 4-[2-(3-aminopropoxy)-4-fluorophenyl]-N- {6-chloro-4- [(methylsulfanyl)methyl]pyridin-2-yl}-5-fluoropyridin-2-amin e (60 mg, 0.133 mmol), potassium phosphate (141 mg, 0.665 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (12.7 mg, 27 μιηοΐ) in toluene (9.9 mL) and NMP (1.2 mL) was added chloro(2-dicyclohexylphosphino-2',4',6'-tri- wo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (22 mg, 27 μιηοΐ) and the mixture was heated in a sealed tube to 130 °C overnight. Additional 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (12.7 mg, 27 μιηοΐ) and chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (22 mg, 27 μιηοΐ) was added and the mixture was heated to 130 °C over night. The mixture was diluted with saturated aqueous sodium chloride solution (60 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried (Whatman filter) and concentrated. The crude product was combined with a second reaction batch carried out analogously as described above, but with 50 mg (0.111 mmol) of 4-[2-(3-aminopropoxy)-4-fluorophenyl]-N- {6-chloro-4- [(methylsulfanyl)methyl]pyridin-2-yl}-5-fluoropyridin-2-amin e and reaction at 110 °C. Sequential purification by flash column chromatography on silica gel (hexanes/ ethyl acetate) and preparative HPLC (autopurifier: acidic conditions) yielded the title compound (5.5 mg, 0.01 mmol).

1H NMR (400 MHz, DMSO-i 6 , 295 K) δ/ppm = 1.81-1.91 (2H), 2.00 (3H), 3.43 (4H), 4.04-4.13 (2H), 5.83-5.88 (1H), 6.05-6.10 (1H), 6.73-6.81 (1H), 6.83-6.92 (1H), 7.05 (1H), 7.54 (1H), 8.24 (1H), 8.91 (1H), 9.24 (1H).

Example 2:

(r c)-Benzyl [{[16,20-difluoro-3,4,5,6-tetrahydro-2H,12H-ll,7-(azeno)-13, 17-(metheno)-l,6,12,14- benzoxatriazac clononadecin-9- l]meth l}(meth l)oxido-λ 6 -sulfan lidene]carbamate

Preparation of Intermediate 2.1:

(r c)-2,6-Dichloro-4-[(methylsulfinyl)methyl]pyridine

Iron(III)chloride (0.11 g, 0.7 mmol) was added to a mixture of 2,6-dichloro-4- [(methylsulfanyl)methyl]pyridine (5.00 g, 24.0 mmol; see Intermediate 1.7) in acetonitrile (57 mL) and the batch was stirred at room temperature for 10 minutes. The batch was cooled to 0°C and periodic acid (5.86 g, 25.7 mmol) was added under stirring in one portion. After 90 min at 0°C, the mixture was added to a stirred solution of sodium thiosulfate pentahydrate (33.4 g, 134.5 mmol) in ice water (306 mL). The batch was saturated with solid sodium chloride and extracted twice with THF. The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by column chromatography on silica gel (ethyl acetate) to give the desired title compound (4.83 g, 21.6 mmol). ¾ NMR (400MHz, DMSO-i/ 6 , 295 K) δ/ppm = 4.01 (d, 1H), 4.25 (d, 1H), 7.51 (s, 2H). Preparation of Intermediate 2.2:

(r c)-Benzyl {[(2,6-dichloro ridin-4- l)meth l](meth l)oxido-λ 6 -sulfan lidene}carbamate

To a suspension of (rac)-2,6-dichloro-4-[(methylsulfinyl)methyl]pyridine (1000 mg, 4.46 mmol), benzyl carbamate (1349 mg, 8.92 mmol), magnesium oxide (719 mg, 17.85 mmol) and rhodium(II) acetate dimer (49 mg, 0.11 mmol) in DCM (44 mL) was added iodobenzene diacetate (2.16 g, 6.69 mmol) at room temperature. The batch was stirred for 18 h at room temperature. The volume of the reaction mixture was reduced to about 10 ml and the mixture was purified by column chromatography on silica gel (hexane/ethyl acetate 0 to 75%) to give the desired title compound (960 mg, 2.6 mmol).

¾ NMR (400MHz, DMSO-i/ 6 , 295 K) δ/ppm = 3.32 (s, 3H), 5.02 (m, 4H), 7.22 - 7.49 (m, 5H), 7.59 (s,

2H).

Prepration of Intermediate 2.3:

2-{4-[2-(2-Amino-5-fluoro indole-l,3(2H)-dione

A suspension of 2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenol (500 mg, 2.25 mmol; see Intermediate 1.3), 2-(4-bromobutyl)-lH-isoindole-l,3(2H)-dione (762 mg, 2.70 mmol) and potassium carbonate (622 mg, 4.50 mmol) in MeCN (14 mL) in a sealed tube was heated to 80 °C and stirred for 16 h. The mixture was cooled to room temperature and the reaction was stopped by the addition of water (20 mL). The mixture was extracted with ethyl acetate (3 x 20 mL). The combined organic layers were washed with saturated aqueous solution of sodium chloride, dried over sodium sulfate, filtered and concentrated to obtain the crude product (1095 mg) which was used without further purification.

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ = 1.59 - 1.67 (4H), 3.51 - 3.67 (2H), 4.03 (2H), 5.81 (2H), 6.34 (1H), 6.85 (1H), 7.02 (1H), 7.26 (1H), 7.78 - 7.89 (m, 5H). Prepration of Intermediate 2.4:

4- [2-(4-Aminobutoxy)-4-fluorophenyl] -5-fluoropyridin-2-amine

To a suspension of 2- {4-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]butyl}- lH-isoindole- l,3(2H)-dione (2190 mg, 85% pure, 4.40 mmol) in EtOH (93 mL) was added a solution of hydrazine in water (35 wt-%, 2.4 mL, 26.4 mmol) and the mixture was heated to reflux for 150 min. The mixture was cooled to 5°C, the precipitate was filtered off and the filtrate was concentrated. Purification by flash column chromatography on silica gel (hexane / ethyl acetate 1 :4 to 0:1 followed by ethyl acetate / methanol 4:1) yielded the title compound (1070 mg, 3.57 mmol).

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.30 - 1.45 (4H), 1.58 - 1.67 (2H), 3.96 - 4.05 (2H), 5.85 (2H), 6.36 (1H), 6.85 (1H), 7.02 (1H), 7.04 (1H), 7.26 (1H), 7.86 (1H).

Prepration of Intermediate 2.5:

(r c)-Benzyl [{[2-({4-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]b utyl}amino)-6- chlorop ridin-4- l]meth l}(meth l)oxido-λ 6 -sulfan lidene]carbamate

N,N-Diisopropylethylamine (0.36 mL, 2.05 mmol) was added to a suspension of 4-[2-(4-aminobutoxy)- 4-fluorophenyl]-5-fluoropyridin-2-amine (200 mg, 0.68 mmol) and benzyl {[(2,6-dichloropyridin-4- yl)methyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (509 mg; see Intermediate 2.2) in NMP (26.8 mL) and the mixture was stirred at 130°C for 90 min. After cooling, the mixture was diluted with water and extracted with ethyl acetate (3 x 50 mL). The combined organic phases were washed with aqueous saturated sodium chloride solution, dried (Whatman filter) and concentrated. The residue was purified by column chromatography on silica gel (hexane to ethyl acetate) to give the desired title compound (92 mg, 0.11 mmol).

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) 8/ppm = 1.51 - 1.62 (2H), 1.63 - 1.75 (2H), 3.17 (2H), 3.25 (3H), 4.02 (2H), 4.75 (2H), 5.03 (2H), 5.83 (2H), 6.36 (1H), 6.42 - 6.49 (1H), 6.49 - 6.57 (1H), 6.85 (1H), 7.03 (1H), 7.16 - 7.23 (1H), 7.19 - 7.38 (6H), 7.75 - 7.95 (1H). Example 2 - Preparation of the End Product:

To a degassed suspension of (rac)-benzyl [{[2-({4-[2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenoxy]butyl}amino)-6-chloropyridin-4-yl]methyl}(meth yl)oxido-λ 6 -sulfanylidene]carbamate (270 mg, 0.43 mmol), potassium phosphate (455 mg, 2.14 mmol) and 2-dicyclohexylphosphino-2',4',6'- triisopropylbiphenyl (41 mg, 0.09 mmol) in toluene (44 mL) and NMP (6 mL) was added chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (71 mg, 0.09 mmol) and the mixture was heated in a sealed tube to 130 °C for 6 h. The mixture was diluted with water (80 mL) and extracted with ethyl acetate (3 x 60 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried (Whatman filter) and concentrated. The residue was purified by column chromatography on silica gel (hexane/ ethyl acetate 1 :4 to 0: 1) to give the desired product (26 mg, 0.04 mmol).

1H NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.69 - 1.90 (4H), 3.13 - 3.30 (5H), 4.08 - 4.22 (2H), 4.50 - 4.76 (2H), 4.90 - 5.17 (2H), 5.89 - 6.01 (1H), 6.01 - 6.20 (1H), 6.78 - 6.95 (2H), 7.01 - 7.14 (1H), 7.27 - 7.48 (6H), 8.17 - 8.43 (2H), 9.30 - 9.60 (1H).

Example 3:

(r c)-16,20-Difluoro-9-[(S-methylsulfonimidoyl)methyl]-3,4,5,6- tetrahydro-2H,12H-ll,7-(azeno)- 13,17-(metheno)-l,6,12,14-benzoxatriazacyclononadecine

To a degassed suspension of (rac)-benzyl [{[2-({4-[2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenoxy]butyl}amino)-6-chloropyridin-4-yl]methyl}(meth yl)oxido-λ 6 -sulfanylidene]carbamate (Intermediate 2.5, 90 mg, 0.14 mmol), potassium phosphate (152 mg, 0.714 mmol) and 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (13.6 mg, 0.029 mmol) in toluene (14.8 mL) and NMP (1.9 mL) was added chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2- aminoethyl)phenyl] palladium(II) methyl-feri-butylether adduct (23 mg, 0.029 mmol) and the mixture was heated in a sealed tube to 130 °C overnight. The mixture was diluted with water (80 mL) and extracted with ethyl acetate (3 x 60 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried (Whatman filter) and concentrated. The residue was sequentially purified by column chromatography on silica gel (hexane/ ethyl acetate, then ethyl acetate /MeOH), followed by preparative HPLC to give the desired title compound (1.2 mg, 2.6 μιηοΐ). HPLC Purification:

Instrument: Waters Autopurificationsystem: Pump 2545, Sample Manager 2767, CFO,

DAD 2996, ELSD 2424, SQD; Column: YMC Triart CI 8 5μιη 100x30mm;

Eluent A: water + 0.1 vol- % formic acid (99%); Eluent B: acetonitrile;

Gradient: 0.00-0.50 min 40% B (25->70 mL/min), 0.51-5.50 min 40-60% B (70 mL/min);

Detection: DAD scan, 210-400 nm.

'H NMR (400 MHz, DMSO-ifc, 295K) 8/ppm = 1.72 - 1.88 (4H), 2.87 (3H), 3.15 - 3.26 (2H), 3.59 (1H), 4.03 - 4.20 (4H), 5.91 (1H), 6.07 (1H), 6.76 (1H), 6.82 - 6.90 (1H), 7.09 (1H), 7.31 (1H), 8.26 (1H), 8.33 (1H), 9.33 (1H).

Example 4:

2,18-Difluoro-9-[(methylsulfanyl)methyl]-13,14,15,16-tetrahy dro-6H-7,ll-(azeno)-5,l-(metheno)- 12,4,6-benzoxadiazacyclooctadecine

Preparation of Intermediate 4.1

2-(2-Amino-5-fluoropyridin-4-yl)-5-fluorophenyl trifluormethansulfonate

To a solution of 2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenol (500 mg, 2.25 mmol; see Intermediate 1.3) in DCM (22 mL) was sequentially added triethylamine (380 μΐ, 2.7 mmol) and N-phenyl trifluoromethanesulfonimide (1.21 g, 3.38 mmol) and the mixture was heated to reflux for 2 h. The mixture was cooled to room temperature and concentrated. The residue was purified by flash column chromatography (silica, hexanes/ethyl acetate) to yield the title compound. Such material was contaminated by N-phenyl trifluoromethanesulfonimide byproducts and was used without further purification.

¾ NMR (400 MHz, DMSO-i/ 6 , 295K) δ/ppm = 6.10 (2H), 6.46 (1H), 7.56 (1H), 7.67 - 7.78 (2H), 8.01 (1H). Preparation of Intermediate 4.2

4-[2-(2-Amino-5-fluoropyridin-4-yl)-5-fluorophenyl]but-3-yn- l-ol

To a mixture of crude 2-(2-Amino-5-fluoropyridin-4-yl)-5-fluorophenyl-trifluormeth ansulfonate (1.24 g), but-3-yn-l-ol (294 mg, 4.20 mmol), triethylamine (980 μΐ, 7.0 mmol) and copper(I) iodide (133 mg, 700 μιηοΐ) in DMF was added bis(triphenylphosphino)palladium(II) chloride (246 mg, 350 μιηοΐ) and the mixture was heated to 110 °C overnight. The mixture was cooled to room temperature, filtered over a pad of Celite ® and concentrated. The residue was dissolved in ethyl acetate (250 mL) and water (100 mL) and the layers were separated. The organic layer was sequentially washed with saturated aqueous sodium bicarbonate solution and saturated aqueous sodium chloride solution, dried (Whatman filter) and concentrated. The crude product was purified by column chromatography on silica gel, hexanes/ethyl acetate to ethyl acetate/MeOH) to yield the title compound (473 mg, 1.55 mmol).

¾ NMR (400 MHz, DMSO-i/ 6 , 295K) δ/ppm = 2.42 (2H), 3.42 (2H), 4.80 - 4.87 (1H), 5.92 (2H), 6.44 (1H), 7.28 - 7.33 (1H), 7.35 - 7.44 (2H), 7.92 (1H). Preparation of Intermediate 4.3

4-[2-(2-Amino-5-fluoropyridin-4-yl)-5-fluorophenyl]butan-l-o l

To a solution of 4-[2-(2-Amino-5-fluoropyridin-4-yl)-5-fluorophenyl]but-3-yn- l-ol (473 mg, 1.72 mmol) in MeOH (15 mL) in an autoclave was added palladium on charcoal (91.7 mg, 10 wt-% Pd, 86.2 μιηοΐ) and the mixture was reacted with an atmosphere of hydrogen (20 bar ¾) for 3 h. The hydrogen atmosphere was replaced by argon and reaction control indicated incomplete conversion. Additional palladium on charcoal (120 mg, 10 wt-% Pd, 113 μιηοΐ) was added and the mixture was hydrogenated (20 bar ¾) for an additional 2.5 h. The hydrogen atmosphere was replaced by argon, the mixture was filtered and the filter cake was washed with MeOH. The filtrate was concentrated to yield the title compound (432 mg, 1.40 mmol).

'H NMR (400 MHz, DMSO-i 6 , 295K) 8/ppm = 1.24 - 1.34 (3H), 1.45 (2H), 3.24 - 3.31 (2H), 4.33 (1H), 5.94 (2H), 6.29 (1H), 7.09 - 7.16 (1H), 7.18 - 7.28 (2H), 7.92 (1H). Two protons are overlayed by residual DMSO.

Preparation of Intermediate 4.4

4-{2-[4-({6-Chloro-4-[(methylsulfanyl)methyl]pyridin-2-yl}ox y)butyl]-4-fluorophenyl}-5- fluoropyridin-2-amine

To a solution of 4-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenyl]butan-l-o l (382 mg, 1.37 mmol) in THF (10 mL) at 0 °C was added sodium hydride (82.3 mg, 60 wt-% in mineral oil, 2.06 mmol). The mixture was allowed to warm to room temperature and stirred for 30 min. Then, 2,6-dichloro-4- [(methylsulfanyl)methyl]pyridine (428 mg, 2.06 mmol, see Intermediate 1.7) was added and the mixture was heated to 90 °C for 2 h and subsequently cooled to room temperature. The mixture was carefully concentrated and the residue was dissolved in ethyl acetate (50 mL) and water (20 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate (3 x 50 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried by filtering over a Whatman filter and concentrated. The crude product was purified by column chromatography on silica gel (hexanes/ ethyl acetate) to yield the desired title compound (304 mg, 0.64 mmol).

'H NMR (400 MHz, DMSO-ifc, 295K) 8/ppm = 1.52 - 1.62 (4H), 1.93 - 1.93 (1H), 1.95 (3H), 3.65 (2H), 4.08 - 4.16 (2H), 5.93 (2H), 6.31 (1H), 6.68 (1H), 7.02 (1H), 7.09 - 7.15 (1H), 7.20 - 7.28 (2H), 7.89 (1H). Two protons are overlayed by residual DMSO.

Example 4 - Preparation of the End Product:

To a solution of 4- {2-[4-({6-chloro-4-[(methylsulfanyl)methyl]pyridin-2-yl}oxy) butyl]-4-fluorophenyl}- 5-fluoropyridin-2-amine (254 mg, 565 μιηοΐ) in toluene (57 mL) and NMP (6.9 mL) was sequentially added potassium phosphate (599 mg, 2.82 mmol), 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (26.9 mg, 56.5 μιηοΐ) and chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l,r- biphenyl)[2-(2- aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (46.7 mg, 56.5 μιηοΐ). The suspension was degassed and heated to 130 °C overnight. The mixture was allowed to cool to room temperature and filtered. The filtrate was partitioned between ethyl acetate (170 mL) and saturated aqueous sodium chloride solution (100 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 170 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried (Whatman filter) and concentrated. The residue was combined with a reaction batch carried out analogously as described above but with 50 mg of 4- {2-[4-({6-chloro-4- [(methylsulfanyl)methyl]pyridin-2-yl}oxy)butyl]-4-fluorophen yl}-5-fluoropyridin-2-amine (111 μιηοΐ). The residue was partitioned between diethyl ether and saturated aqueous sodium chloride solution, the layers were separated and the aqueous layer was extracted with diethyl ether. The combined organic layers were dried and concentrated. The residue was dissolved in DMSO (8 mL) and water (1 mL). The resulting suspension was filtered and the filtrate was subjected to preparative HPLC to yield the title compound (82 mg, 0.19 mmol). HPLC Purification:

Instrument: Pump: LABOMATIC HD-5000; Manual injection valve: Rheodyne 3725Ϊ038; Detector: Knauer AZURA UVD 2.15; Collector: LABOMATIC LABOCOL Vario-4000; Column: Chromatorex RP C-18 10 μιη, 125x30mm;

Eluent A: water + 0.1 vol- % formic acid (99%); Eluent B: acetonitrile;

Gradient: 0.00-0.50 min 65% B (60 mL/min), 0.50-10.00 min 65-100% B (60 mL/min), 10.00-10.10 min 65-100% B (60 mL/min), 10.10-12.00 min 100% B (60 mL/min);

Detection: UV 269 nm.

1 H NMR (400 MHz, DMSO-i 6 , 295 K) 8/ppm = 1.81 (4H), 1.98 (3H), 2.61 - 2.86 (2H), 3.56 (2H), 3.91 4.89 (2H), 6.16 (1H), 6.41 (1H), 7.12 (1H), 7.29 - 7.43 (2H), 7.54 (1H), 8.32 (1H), 9.70 (1H).

Example 5:

(r c)-2,18-Difluoro-9-[(methylsulfmyl)methyl]-13,14,15,16-tetra hydro-6H-7,ll-(azeno)-5,l- (metheno)-12,4,6-benzoxadiazacyclooctadecine

To a solution of 2,18-difluoro-9-[(methylsulfanyl)methyl]-13, 14,15, 16-tetrahydro-6H-7,l l-(azeno)-5,l- (metheno)-12,4,6-benzoxadiazacyclooctadecine (60.0 mg, 145 μιηοΐ, see Example 4) in MeCN (4.4 mL) at 0 °C was added iron(III) chloride (2.35 mg, 14.5 μιηοΐ) and the mixture was stirred for 10 min. Then, periodic acid (99.2 mg, 435 μιηοΐ) was added amd the mixture was stirred for 2 h. Additional iron(III) chloride (2.35 mg, 14.5 μιηοΐ) and periodic acid (99.2 mg, 435 μιηοΐ) was added and the mixture was stirred for 1 h. The reaction was stopped by pouring onto ice and saturated aqueous sodium thiosulfate (15 mL) was added. The mixture was stirred for 10 min and the product was extracted with ethyl acetate (2 x 35 mL). The combined organic layers were washed with a saturated solution of aqueous sodium chloride, dried (Whatman filter) and concentrated. The residue was combined with the crude material of a second reaction batch carried out analogously as described above, but with 20 mg of 2,18-difluoro-9- [(methylsulfanyl)methyl]- 13, 14,15,16-tetrahydro-6H-7, 1 1 -(azeno)-5, 1 -(metheno)- 12,4,6- benzoxadiazacyclooctadecine (48 μιηοΐ). The crude material was purified by preparative HPLC to yield the title compound (17.2 mg, 0.04 mmol). HPLC Purification:

Instrument: Pump: LABOMATIC HD-5000; Manual injextion valve: Rheodyne 3725Ϊ038; Detector: Knauer AZURA UVD 2.15; Collector: LABOMATIC LABOCOL Vario-4000; Column: Chromatorex RP C-18 10 μιη, 125x30mm;

Eluent A: water + 0.1 vol- % formic acid (99%); Eluent B: acetonitrile;

Gradient: 0.00-1.00 min 30% B (60 mL/min), 1.00-10.00 min 35-70% B (60 mL/min), 10.00-10.10 min 70-100% B (60 mL/min), 10.10-12.53 min 100% B (60 mL/min);

Detection: UV 269 nm. ¾ NMR (400 MHz, DMSO-i 6 , 295 K) δ/ppm = 1.82 (4H), 2.55 (3H), 2.65 - 2.81 (2H), 3.84 (1H), 4.03 (1H), 4.09 - 5.05 (2H), 6.17 (1H), 6.39 (1H), 7.13 (1H), 7.31 - 7.45 (2H), 7.56 (1H), 8.34 (1H), 9.80 (1H)

Example 6:

(rac)-tert-but \ [{[16,20-difluoro-6-methyl-3,4,5,6-tetrahydro-2H,12H-13,17-( azeno)-ll,7- (metheno)-l,6,12,14-benzoxatriazacyclononadecin-9-yl]methyl} (methyl)oxido^ 6 - sulfanylidene] carbamate

Preparation of Intermediate 6.1:

2-Chloro-5-fluoro-4-(4-fluoro-2-methoxyphenyl)pyrimidine

A batch with 2,4-dichloro-5-fluoropyrimidine (200 mg; 1.20 mmol; Aldrich Chemical Company Inc.), (4-fluoro-2-methoxyphenyl)boronic acid (224 mg; 1.31 mmol; Aldrich Chemical Company Inc.) and tetrakis(triphenylphosphin)palladium(0) (138 mg; 0.12 mmol) in 1,2-dimethoxyethane (3.6 ml) and an aqueous 2M solution of potassium carbonate (1.8 ml) was degassed using argon. The batch was stirred under an atmosphere of argon for 16 hours at 90°C. After cooling the batch was diluted with ethyl acetate and washed with saturated aqueous sodium chloride solution. The organic layer was filtered using a Whatman filter and concentrated. The residue was purified by column chromatography (hexane / ethyl acetate 1 :1) to give the desired title compound (106 mg; 0.41 mmol).

¾ NMR (400MHz, CDC1 3 , 295 K) δ/ppm = 8.47 (1H), 7.51 (1H), 6.82 (1H), 6.73 (1H), 3.85 (3H).

Preparation of Intermediate 6.2:

2-(2-Chloro-5-fluoropyrimidin-4-yl)-5-fluorophenol

A solution of boron tribromide in DCM (1M; 43.3 mL; 43.3 mmol; Aldrich Chemical Company Inc.) was added dropwise to a stirred solution of 2-chloro-5-fluoro-4-(4-fluoro-2-methoxyphenyl)pyrimidine (2.00 g; 7.79 mmol) in DCM (189 mL) at 0°C. The mixture was slowly warmed to room temperature while stirring overnight. The mixture was cautiously diluted with an aqueous solution of sodium bicarbonate under stirring at 0°C and stirred at room temperature for 1 hour. Solid sodium chloride was added and the mixture filtered using a Whatman filter. The organic layer was concentrated to give the crude title compound (1.85 g) that was used without further purification.

¾ NMR (400MHz, DMSO- d 6 , 295 K) δ/ppm = 10.80 (1H), 8.90 (1H), 7.50 (1H), 6.83 (1H), 6.78 (1H)

Preparation of Intermediate 6.3:

l-Fluoro-3-[(methylsulfanyl)methyl]-

Intermediate 6.3 was prepared as described in WO 2013/037894 (Intermediate 9.1, page 94)

¾ NMR (400MHz, CDCI3, 295 K) δ/ppm = 8.00 (m, 1H), 7.82 (m, 1H), 7.44 (m, 1H), 3.74 (s, 2H), 2.03 (s, 3H). Preparation of Intermediate 6.4:

(r c)-l-Fluoro-3-[(methylsulfinyl)methyl]-5-nitrobenzene

Intermediate 6.4 was prepared as described in WO 2013/037894 (Intermediate 9.2, page 94)

¾ NMR (400MHz, DMSO- d 6 , 295 K) δ/ppm = 8.06 (m, 2H), 7.63 (m, 1H), 4.32 (d, 1H), 4.08 (d, 1H),

2.45 (s, 3H).

Preparation of Intermediate 6.5:

(rac)-tert-but \ [(3-fluoro-5-nitrobenzyl)(methyl)oxido^ 6 -sulfanylidene]carbamate

To a suspension of (rac)-l-fluoro-3-[(methylsulfinyl)methyl]-5-nitrobenzene (834 mg; 3.84 mmol), tert- butyl carbamate (675 mg; 5.76 mmol), magnesium oxide (619 mg; 15.36 mmol) and rhodium(II)-acetate dimer (85 mg; 0.19 mmol) in DCM (38 mL) was added iodobenzene diacetate (1855 mg; 5.76 mmol) at room temperature. The batch was stirred for 16 hours at room temperature and additional 2 hours at 40°C. After cooling, the batch was filtered and concentrated. The residue was purified by chromatography (hexane to hexane / ethyl acetate 45%) to give the desired title compound (998 mg; 3.00 mmol).

¾ NMR (400MHz, DMSO- d 6 , 295 K) δ/ppm = 1.29 - 1.42 (9H), 3.17 - 3.25 (3H), 5.04 - 5.13 (2H), 7.75 (1H), 8.18 - 8.23 (2H).

Preparation of Intermediate 6.6:

(rac)-tert-but \ [{3-[(4-hydroxybutyl)(methyl)amino]-5-nitrobenzyl}(methyl)ox ido-λ 6 - sulfanylidene] carbamate

A mixture of (rac)-tert-butyl [(3-fluoro-5-nitrobenzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (200 mg; 0.60 mmol) and 4-(methylamino)butan-l-ol (186 mg; 1.81 mmol) in NMP (2.0 mL) was stirred at 100°C overnight. The mixture was stirred for additional 2 h at 110°C. After cooling, the mixture was diluted with ethyl acetate and washed twice with water and once with an aqueous solution of sodium chloride. The organic phase was filtered using a Whatman filter and concentrated. The residue was purified by column chromatography on silica gel (hexane to hexane / ethyl acetate 0:1) followed by preparative HPLC to give the title compound (95 mg; 0.23 mmol).

HPLC Purification:

Instrument: Pump: LABOMATIC HD-5000; Manual injection valve: Rheodyne 3725Ϊ038; Detector: Knauer AZURA UVD 2.15; Collector: LABOMATIC LABOCOL Vario-4000; Column: Chromatorex RP C-18 10 μιη, 125x30mm;

Eluent A: water + 0.1 vol- % formic acid (99%); Eluent B: acetonitrile;

Gradient: 0.00-0.50 min 15% B (150 mL/min), 0.50-6.00 min 15-55% B (150 mL/min), 6.00-6.10 min 55-100% B (150 mL/min), 6.10-8.00 min 100% B (150 mL/min).

Detection: UV 254 nm.

¾ NMR (400MHz, DMSO- d 6 , 295 K) 8/ppm = 1.37 - 1.59 (13H), 2.98 (3H), 3.11 - 3.18 (3H), 3.36 - 3.48 (4H), 4.44 (1H), 4.87 - 4.98 (2H), 7.17 (1H), 7.39 (1H), 7.47 (1H).

Preparation of Intermediate 6.7:

(rac)-tert-but \ N-[[3-[4-[2-(2-chloro-5-fluoro-pyrimidin-4-yl)-5-fluoro-phen oxy]butyl-methyl- amino] -5-nitro-phenyl] methyl-methyl-oxo^ 6 -sulfanylidene] carbamate

A solution of diisopropyl azodicarboxylate (16 mg; 79 μιηοΐ) in DCM (0.2 mL) was added dropwise to a mixture of (rac)-tert-butyl [{3-[(4-hydroxybutyl)(methyl)amino]-5-nitrobenzyl}(methyl)ox ido-λ 6 - sulfanylidene]carbamate (30 mg; 72 μιηοΐ), 2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenol (19 mg; 79 μιηοΐ) and triphenylphosphine (21 mg; 79 μιηοΐ) in DCM (0.3 mL) at 0°C and the batch was stirred at room temperature overnight. The mixture was concentrated and the residue was purified by preparative HPLC to give the title compound (26 mg; 40 μιηοΐ).

HPLC Purification:

Instrument: Pump: LABOMATIC HD-5000; Manual injection valve: Rheodyne 3725Ϊ038; Detector: Knauer AZURA UVD 2.15; Collector: LABOMATIC LABOCOL Vario-4000; Column: Chromatorex RP C-18 10 μιη, 125x30mm;

Eluent A: water + 0.1 vol- % formic acid (99%); Eluent B: acetonitrile;

Gradient: 0.00-0.50 min 65% B (150 mL/min), 0.50-6.00 min 65-100% B (150 mL/min), 6.00-8.00 min 100% B (150 mL/min);

Detection: UV 254 nm.

¾ NMR (400MHz, DMSO- d 6 , 295 K) δ/ppm = 1.37 (9H), 1.51 - 1.71 (4H), 2.95 (3H), 3.12 (3H), 3.36 - 3.45 (2H), 4.1 1 (2H), 4.85 - 4.96 (2H), 6.97 (1H), 7.12 - 7.18 (2H), 7.36 (1H), 7.49 (1H), 7.50 - 7.55 (1H), 8.80 (1H).

Preparation of Intermediate 6.8:

(r c)-teri-butyl[{3-amino-5-[{4-[2-(2-chloro-5-fluoropyrimidin- 4-yl)-5- fluoro henox ]but l}(meth l)amino]benz l}(meth l)oxido-λ 6 -sulfan lidene]carbamate

Platinum 1%> and vanadium 2%>, on activated carbon (50-70%) wetted powder, 10 mg) was added to a solution of (rac)-tert-butyl N-[[3-[4-[2-(2-chloro-5-fluoro-pyrimidin-4-yl)-5-fluoro- phenoxy]butyl-methyl-amino]-5-nitro-phenyl]methyl-methyl-oxo -λ 6 -sulfanylidene]carbamate (25 mg; 39 μιηοΐ) in methanol (5 mL) and the mixture was stirred for 10 min at room temperature under a hydrogen atmosphere. The mixture was filtered and the filtrate was concentrated to give the crude title compound (17 mg) that was used without further purification.

¾ NMR (400MHz, DMSO- da, 295 K) δ/ppm = 1.31 - 1.40 (9H), 1.41 - 1.67 (4H), 2.65 - 2.83 (3H), 3.04 (3H), 3.14 - 3.28 (2H), 4.08 (2H), 4.46 - 4.62 (2H), 4.95 (2H), 5.91 - 5.99 (3H), 6.97 (1H), 7.15 (1H), 7.53 (1H), 8.84 (1H). Example 6 - Preparation of the End Product:

A mixture of crude (rflc)-tert-butyl[{3-amino-5-[{4-[2-(2-chloro-5-fluoropyrimi din-4-yl)-5- fluorophenoxy]butyl} (methyl)amino]benzyl} (methyl)oxido^ 6 -sulfanylidene]carbamate (17.0 mg), chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl- 1 , 1 '-biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-tert-butylether adduct (4.6 mg; 5.6 μιηοΐ; ABCR GmbH & CO. KG) and 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (2.7 mg; 5.6 μmol; Aldrich Chemical Company Inc.) and potassium phosphate (29.6 mg; 139 μιηοΐ) in toluene (2.0 ml) and NMP (0.2 mL) was stirred under an atmosphere of argon at 110°C in a closed vessel for 4 hours. After cooling, the batch was diluted with aqueous sodium chloride solution and extracted with ethyl acetate / THF. The organic layer was filtered using a Whatman filter and concentrated. The residue was purified by preparative HPLC to give the title compound (11.0 mg; 20 μιηοΐ).

HPLC Purification:

Instrument: Waters Autopurificationsystem; Column: Waters XBrigde CI 8 5μ 100x30mm;

Eluent A: H 2 0 + 0.1 vol% formic acid (99%), Eluent B: MeCN;

Gradient: 0.00-0.50 min 25% B (25-> 70 mL/min), 0.51-5.50 min 50-70% B (70 mL/min),

DAD scan: 210-400 nm

¾ NMR (400MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.38 (9H), 1.67 (2H), 1.82 (2H), 2.89 (3H), 3.10 (3H), 3.22 - 3.31 (m, 2H), 4.19 - 4.27 (2H), 4.61 - 4.69 (2H), 6.25 (1H), 6.42 (1H), 6.86 (1H), 7.13 (1H), 7.33 - 7.39 (1H), 7.71 (1H), 8.62 (1H), 9.56 (1H).

Example 7;

(rac)-tert-butyl [{[15,19-difluoro-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 16, 12-(metheno)- 1,5,11,13- benzoxatriazacyclooctadecin-8-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

Preparation of Intermediate 7.1:

4-[2-(3-aminopropoxy)-4-fluorophenyl]-5-fluoropyridin-2-amin e 2

To a solution of 2- {3-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]propyl} -lH-isoindole-l,3(2H)- dione (see Intermediate 1.4; 2.01 g, 90 % purity) in ethanol (100 mL) was added aqueous hydrazine (2.4 mL, 35 % purity) and the mixture was heated to reflux for 3 h. The mixture was cooled to 5 °C, filtered and concentrated. The crude product was purified by flash column chromatography (silica gel, ethyl acetate/methanol) to yield the title compound (1.14 g, 95% purity).

LC-MS (Method a): R t = 0.53 min; MS (ESIpos): m/z = 280 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.62 - 1.72 (m, 2H), 2.54 - 2.59 (m, 2H), 3.13 - 3.19 (m, 2H), 4.02 - 4.08 (m, 2H), 5.78 - 5.89 (m, 2H), 6.33 - 6.39 (m, 1H), 6.81 - 6.87 (m, 1H), 7.01 - 7.08 (m, 1 H), 7.20 - 7.28 (m, 1 H), 7.82 - 7.88 (m, 1 H).

Preparation of Intermediate 7.2:

(rac)-tert-butyl {[(2,6-dichloropyridin-4-yl)methyl](methyl)oxido-λ 6 -sulfanylidene}carbamate

To a suspension of (rac)-2,6-dichloro-4-[(methylsulfinyl)methyl]pyridine (see Intermediate 2.1 ; 6.00 g), tert-butyl carbamate (4.70 g), magnesium oxide (4.32 g) and rhodium(II)acetate dimer (296 mg) in dichloromethane (270 ml) was added portionwise iodobenzene diacetate (12.9 g) and the mixture was stirred at room temperature for 15 h and subsequently at 40 °C for 3 h. The mixture was allowed to cool to room temperature, filtered over a pad of Celite and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (7.33 g, 97%> purity).

LC-MS (Method a): R t = 1.10 min; MS (ESIpos): m/z = 339 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.38 - 1.42 (m, 9H), 3.20 - 3.26 (m, 3H), 4.96 - 5.04 (m, 2H), 7.55 - 7.61 (m, 2H). Preparation of Intermediate 7.3:

(rac)-fert-butyl [{[2-({3-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]p ropyl}

chloropyridin-4-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

A solution of 4-[2-(3-aminopropoxy)-4-fluorophenyl]-5-fluoropyridin-2-amin e (580 mg, 2.08 mmol), (rac)-fert-butyl {[(2,6-dichloropyridin-4-yl)methyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (587 mg) and 2,6-lutidine (600 μΐ.) in DMSO (19 mL) was heated to 130 °C for 3 h and subsequently to 150 °C for 1.5 h. The mixture was allowed to cool to room temperature, diluted with water (100 mL) and extracted three times with ethyl acetate (60 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (233 mg, 90% purity).

LC-MS (Method a): R t = 1.17 min; MS (ESIpos): m/z = 582 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 22°C) δ/ppm = 1.38 (s, 9H), 1.81 - 1.93 (m, 2H), 3.14 - 3.18 (m, 3H), 3.20 - 3.29 (m, 2H), 4.05 - 4.13 (m, 2H), 4.66 - 4.78 (m, 2H), 5.78 - 5.90 (m, 2H), 6.36 - 6.39 (m, 1H), 6.40 - 6.44 (m, 1H), 6.50 - 6.53 (m, 1H), 6.79 - 6.92 (m, 1H), 7.01 - 7.10 (m, 1H), 7.19 - 7.25 (m, 1H), 7.25 - 7.31 (m, 1H), 7.84 - 7.90 (m, 1H).

Example 7 - Preparation of the End Product:

A degassed suspension of rac-tert-butyl [{[2-({3-[2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenoxy]propyl}amino)-6-chloropyridin-4-yl]methyl}(met hyl)oxido-λ 6 -sulfanylidene]carbamate

(220 mg), chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l,r- biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (31.3 mg), 2-(dicyclohexylphosphino)-2',4',6'-tri-z ' sO-propyl- l,l'-biphenyl (18.0 mg) and potassium phosphate (401 mg) in toluene (36 mL) and N-methylpyrrolidone

(4.6 mL) was heated at 130 °C for 5.5 h and subsequently at 110 °C for 12 h. The mixture was allowed to cool to room temperature, diluted with water (100 mL) and extracted three times with ethyl acetate (50 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate -> ethyl acetate/methanol yielded the title compound (145 mg, 84% purity). A pure sample was obtained by preparative HPLC.

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.31 min; MS (ESIpos): m/z = 546 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.38 (s, 9H), 1.82 - 1.94 (m, 2H), 3.21 (s, 3H), 3.41 (br s, 2H), 4.03 - 4.13 (m, 2H), 4.47 - 4.65 (m, 2H), 5.93 (d, 1H), 6.10 - 6.18 (m, 1H), 6.83 - 6.96 (m, 2H), 7.05 (dd, 1H), 7.54 (dd, 1H), 8.26 (d, 1H), 8.83 - 8.92 (m, 1H), 9.35 - 9.43 (m, 1H).

Example 8:

(rac)- 15,19-difluoro-8- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 16,12- (metheno)- 1 ,5,11, 13-benzoxatriazacyclooctadecine

To a solution of (rac)-tert-butyl [{[15,19-difluoro-2,3,4,5-tetrahydro-l lH-10,6-(azeno)-16,12- (metheno)- 1 ,5,11, 13-benzoxatriazacyclooctadecin-8-yl]methyl} (methyl) oxido-λ 6 - sulfanylidene] carbamate (90.0 mg) in dichloromethane (900 μΐ.) was added trifluoroacetic acid (410 μΐ.) and the mixture was stirred for 2 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate solution (5 mL). The mixture was combined with a second reaction batch carried out as described above but with 20 mg of (rac)-tert-butyl [{[15,19- difluoro-2,3,4,5-tetrahydro- 11 H- 10,6-(azeno)- 16,12-(metheno)- 1 ,5,11, 13-benzoxatriazacyclooctadecin- 8-yl]methyl}(methyl)oxido^ 6 -sulfanylidene]carbamateThe mixture was extracted three times with dichloromethane (25 mL each), the combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by preparative HPLC to yield the title compound (38 mg, 99%> purity). HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100% B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.07 min; MS (ESIpos): m/z = 446 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm 1.83 - 1.96 (m, 2H), 2.88 (s, 3H), 3.37 - 3.47 (m, 2H), 3.55 - 3.62 (m, 1H), 4.05 - 4.20 (m, 4H), 5.86 - 5.95 (m, 1H), 6.06 - 6.15 (m, 1H), 6.83 - 6.92 (m, 2H), 7.02 - 7.09 (m, 1H), 7.48 - 7.60 (m, 1H), 8.23 - 8.29 (m, 1H), 8.88 - 8.91 (m, 1H), 9.31 (s, 1H).

Example 9:

(rac)-tert-butyl [{[17,21-difluoro-2,3,4,5,6,7-hexahydro-13H-12,8-(azeno)-14, 18-(metheno)-l,7,13,15- benzoxatriazacycloicosin-10-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

Preparation of Intermediate 9.1:

2- {5-[2-(2-amino-5-fluoropyridin-4- l)-5-fluorophenoxy]pentyl}-lH-isoindole-l,3(2H)-dione

To a suspension of 2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenol (see Intermediate 1.3; 2.00 g) and 2- (5-bromopentyl)-lH-isoindole-l,3(2H)-dione (3.20 g) in acetonitrile (57 ml) was added potassium carbonate (2.49 g) and the mixture was heated to 80 °C for 22 h. The mixture was allowed to cool to room temperature, diluted with water (150 mL) and extracted three times with ethyl acetate (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated to yield the title compound (4.58 g, 82%> purity) that was used without further purification. LC-MS (Method a): R t = 1.11 min; MS (ESIpos): m/z = 438 [M+H] + ¾ NMR (400 MHz, DMSO-i 6 , 295 K) δ/ρρηι = 1.27 - 1.35 (m, 2H), 1.55 - 1.67 (m, 4H), 3.52 - 3.61 (m, 2H), 3.93 - 4.01 (m, 2H), 5.74 - 5.92 (m, 2H), 6.28 - 6.37 (m, 1H), 6.80 - 6.91 (m, 1H), 6.97 - 7.04 (m, 1H), 7.20 - 7.28 (m, 1H), 7.62 - 7.69 (m, 1H), 7.80 - 7.90 (m, 4H). Preparation of Intermediate 9.2:

4- {2-[(5-aminopentyl)oxy]-4-fluorophenyl}-5-fluoropyridin-2-am ine

To a suspension of 2- {5-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]pentyl} -lH-isoindole- l,3(2H)-dione (4.58 g, 82% purity) in ethanol (220 niL) was added aqueous hydrazine (4.7 mL, 35 % purity) and the mixture was heated to reflux for 3.5 h. The mixture was cooled to 5 °C, filtered and concentrated. The residue was treated with dichloromethane and the resulting suspension was filtered. The filtrate was treated with ethanol, cooled to 0 °C and filtered. The combined filter cakes were purified by flash column chromatography (silica gel, ethyl acetate/methanol) to yield the title compound (1.53 g, 95% purity).

LC-MS (Method a): R t = 0.65 min; MS (ESIpos): m/z = 308 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.27 - 1.37 (m, 4H), 1.56 - 1.64 (m, 2H), 3.14 - 3.19 (m, 2H), 3.96 - 4.02 (m, 2H), 5.79 - 5.93 (m, 2H), 6.34 - 6.41 (m, 1H), 6.79 - 6.88 (m, 1H), 6.98 - 7.07 (m, 1H), 7.20 - 7.30 (m, 1H), 7.82 - 7.87 (m, 1H) (one methylene group obscured). Preparation of Intermediate 9.3:

(rac)-tert-buty\ [{[2-({5-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]p entyl}amino)-6- chloropyridin-4-yl]methyl}(methyl)oxido-

A solution of 4- {2-[(5-aminopentyl)oxy]-4-fluorophenyl}-5-fluoropyridin-2-am ine (1.60 g, 70 % purity), (rac)-fert-butyl { [(2,6-dichloropyridin-4-yl)methyl] s(methyl)oxido^ 6 -sulfanylidene} carbamate (see Intermediate 7.2; 1.03 g) and 2,6-lutidine (1.1 mL) in DMSO (37 mL) was heated to 130 °C for 2.5h. The mixture was allowed to cool to room temperature, diluted with water (150 mL) and extracted three times with ethyl acetate (70 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (623 mg, 95% purity). LC-MS (Method a): R t = 1.23 min; MS (ESIpos): m/z = 610 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.31 - 1.41 (m, 11H), 1.44 - 1.54 (m, 2H), 1.58 - 1.69 (m, 2H), 3.10 - 3.19 (m, 5H), 3.97 - 4.02 (m, 2H), 4.68 - 4.79 (m, 2H), 5.79 - 5.86 (m, 2H), 6.32 - 6.39 (m, 1H), 6.41 - 6.46 (m, 1H), 6.47 - 6.53 (m, 1H), 6.82 - 6.91 (m, 1H), 6.97 - 7.07 (m, 1H), 7.13 - 7.20 (m, 1H), 7.24 - 7.29 (m, 1H), 7.83 - 7.87 (m, 1H).

Example 9 - Preparation of the End Product

A degassed suspension of (rac)-fert-butyl [{[2-({5-[2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenoxy]pentyl}amino)-6-chloropyridin-4-yl]methyl}(met hyl)oxido-λ 6 -sulfanylidene]carbamate (310 mg), chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl- 1 , 1 '-biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (84.0 mg), 2-(dicyclohexylphosphino)-2',4',6'- triisopropylbiphenyl (84.0 mg) and potassium phosphate (539 mg) in toluene (31 mL) and N- methylpyrrolidone (3.1 mL) was heated at 130 °C for 18 h. Additional portions of chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (84.0 mg) and 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (84.0 mg) were added, and the mixture was stirred for an additional 7 h at 130 °C. The mixture was allowed to cool to room temperature, diluted with water (100 mL) and extracted three times with ethyl acetate (50 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was sequentially purified by flash column chromatography (silica gel, dichloromethane/methanol) followed by preparative HPLC to yield the title compound (78 mg, 99% purity).

HPLC Purification:

Instrument: pump: Labomatic HD-5000, head HDK 280, lowpressure gradient module ND-B1000; manual injection valve: Rheodyne 3725Ϊ038; detector: Knauer Azura UVD 2.15; collector: Labomatic Labocol Vario-4000; column: Chromatorex RP C-18 10 μηι, 125x30mm; solvent A: water + 0.1 vol-%> formic acid, solvent B: acetonitrile; gradient: 0.00-0.50 min 40% B (150 ml/min), 0.50-6.00 min 40-80% B (150 ml/min), 6.00-6.10 min 80-100% B (150 ml/min), 6.10-8.00 min 100% B (150 ml/min); detection: UV 279 nm.

LC-MS (Method a): R t = 1.36 min; MS (ESIpos): m/z = 574 [M+H] + ¾ NMR (400 MHz, DMSO-d 6 , 295 K) δ/ρρηι = 1.35 - 1.41 (m, 11H), 1.43 - 1.49 (m, 2H), 1.59 - 1.70 (m, 2H), 3.10 - 3.17 (m, 2H), 3.17 - 3.23 (m, 3H), 4.13 - 4.20 (m, 2H), 4.50 - 4.62 (m, 2H), 5.89 - 5.95 (m, 1H), 6.10 - 6.16 (m, 1H), 6.67 - 6.74 (m, 1H), 6.84 - 6.93 (m, 1H), 7.12 - 7.18 (m, 1H), 7.24 - 7.33 (m, 1H), 8.17 - 8.28 (m, 2H), 9.38 (s, 1H).

Example 10:

(rac)- 17,21 -difluoro- 10-[(S-methylsulfonm^

14,18-(metheno)- 1,7,13,15-benzoxatriazacycloicosine

To a solution of (rac)-tert-butyl [{[17,21-difluoro-2,3,4,5,6,7-hexahydro-13H-12,8-(azeno)-14, 18- (metheno)- 1 ,7,13,15-benzoxatriazacycloicosin- 10-yl]methyl} (methyl)oxido^ 6 -sulfanylidene]carbamate (75.0 mg) in dichloromethane (25 mL) was added trifluoroacetic acid (250 μΐ.) and the mixture was stirred for 4 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate solution, and the mixture was extracted three times with dichloromethane (20 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by preparative HPLC to yield the title compound (32 mg, 99% purity).

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.11 min; MS (ESIpos): m/z = 474 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.32 - 1.41 (m, 2H), 1.41 - 1.52 (m, 2H), 1.61 - 1.69 (m, 2H), 2.82 - 2.91 (m, 3H), 3.06 - 3.20 (m, 2H), 3.56 - 3.62 (m, 1H), 4.03 - 4.12 (m, 1H), 4.12 - 4.20 (m, 3H), 5.79 - 5.97 (m, 1H), 6.10 - 6.17 (m, 1H), 6.55 - 6.66 (m, 1H), 6.81 - 6.92 (m, 1H), 7.11 - 7.19 (m, 1H), 7.25 - 7.34 (m, 1H), 8.18 - 8.26 (m, 2H), 9.25 - 9.34 (m, 1H). Example 11:

(rac)-16,20-difluoro-6-methyl-9-[(S-methylsulfo^

17,13 -(metheno)- 1,6,12,14-benzoxatriazacyclononadecine

Preparation of Intermediate 11.1:

(rac)-feri-butyl [({2-chloro-6-[(4-hydroxybutyl)(methyl)amino] yridin-4-yl}methyl)(methyl)oxido-λ 6 - sulfanylidene]carbamate

To a solution of (rac)-fert-butyl {[(2,6-dichloropyridin-4-yl)methyl](methyl)oxido^ 6 - sulfanylidene} carbamate (see Intermediate 7.2; 1.00 g) and 4-(methylamino)butan-l-ol (365 mg) in DMSO (35 mL) was added 2,6-lutidine (1.0 mL) and the mixture was heated at 130 °C for 4.5 h. The mixture was allowed to cool to room temperature, diluted with water (150 mL) and extracted three times with ethyl acetate (70 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (539 mg, 99% purity).

LC-MS (Method a): R t = 1.07 min; MS (ESIneg): m/z = 404 [M-H] "

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.35 - 1.45 (m, 11H), 1.50 - 1.59 (m, 2H), 2.93 - 3.01 (m, 3H), 3.12 - 3.17 (m, 3H), 3.37 - 3.43 (m, 2H), 3.44 - 3.49 (m, 2H), 4.39 - 4.44 (m, 1H), 4.72 - 4.87 (m, 2H), 6.54 - 6.60 (m, 1H), 6.60 - 6.65 (m, 1H). Preparation of Intermediate 11.2

(rac)-fert-butyl [( {2-[ {4-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]butyl} (methyl)

chloropyridin-4-yl}methyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate

To a suspension of (rac)-fert-butyl [( {2-chloro-6-[(4-hydroxybutyl)(methyl)amino]pyridin-4- yl}methyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (350 mg), 2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenol (see Intermediate 1.3; 160 mg) and triphenylphospine (283 mg) in dichloromethane (5 mL) was added a solution of diisoproyl azodicarboxylate (210 μΐ) in dichloromethane (1 mL), and the mixture was stirred for 18 h at room temperature. Additional portions of 2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenol (80 mg), triphenylphosphine (141 mg) and diisoproyl azodicarboxylate (105 μΐ) were added, and the mixture was stirred for an additional 4 h. The reaction mixture was concentrated and the crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate). Further purification by flash column chromatography (silica gel hexanes/ethyl acetate) yielded the title compound (180 mg, 95% purity).

LC-MS (Method a): R t = 1.26 min; MS (ESIpos): m/z = 610 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.38 (s, 9H), 1.50 - 1.66 (m, 4H), 2.89 - 2.96 (m, 3H), 3.11 - 3.18 (m, 3H), 3.41 - 3.49 (m, 2H), 3.99 - 4.06 (m, 2H), 4.71 - 4.84 (m, 2H), 5.79 - 5.88 (m, 2H), 6.32 - 6.38 (m, 1H), 6.56 - 6.59 (m, 1H), 6.59 - 6.61 (m, 1H), 6.83 - 6.92 (m, 1H), 6.96 - 7.06 (m, 1H), 7.23 - 7.29 (m, 1H), 7.80 - 7.86 (m, 1H).

Example 11 - Preparation of the End Product

A degassed suspension of (rac)-fert-butyl [({2-[{4-[2-(2-amino-5-fluoropyridin-4-yl)-5- fluorophenoxy]butyl}(methyl)amino]-6-chloropyridin-4-yl}meth yl)(methyl)oxido-λ 6 - sulfanylidene]carbamate (120 mg), chloro(2-dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, - biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (32.5 mg), 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (18.8 mg) and potassium phosphate (209 mg) in toluene (19 mL) and N-methylpyrrolidone (2.4 mL) was heated at 130 °C for 18 h. Additional portions of chloro(2-dicyclohexylphosphino-2',4',6'-tri-wo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (16.3 mg) and 2-(dicyclohexylphosphino)-2',4',6'- triisopropylbiphenyl (9.4 mg) were added, and the mixture was stirred for an additional 4 h at 130 °C. The mixture was allowed to cool to room temperature, diluted with water (50 mL) and extracted three times with ethyl acetate (30 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to afford crude (rac)-fert-butyl [{[16,20-difluoro-6- methyl-3,4,5,6-tetrahydro-2H-7, 11 -(azeno)- 17, 13-(metheno)- 1 ,6,12,14-benzoxatriazacyclononadecin- 9(12H)-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate (71 mg, 92% purity,) that was contaminated by some impurities and used without further purification.

To a solution of crude (rac)-fert-butyl [{[16,20-difluoro-6-methyl-3,4,5,6-tetrahydro-2H-7,l l-(azeno)- 17, 13-(metheno)- 1,6,12,14-benzoxatriazacyclononadecin-9(l 2H)-yl]methyl} (methyl) oxido-λ 6 - sulfanylidene]carbamate (70.0 mg) in dichloromethane (1.5 mL) was added trifluoroacetic acid (240 μΐ) and the mixture was stirred for 4.5 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate solution, and the mixture was extracted three times with dichloromethane (15 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated. The crude product was purified by preparative HPLC. Further purification by preparative HPLC yielded the title compound (8 mg, 99% purity).

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Colum: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.2 vol %> aqueous ammonia (32%>), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.17 min; MS (ESIpos): m/z = 474 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.68 - 1.90 (m, 4 H), 2.85 - 2.89 (m, 3 H), 2.93 (s, 3 H), 3.48 - 3.60 (m, 2 H), 3.62 - 3.73 (m, 1 H), 4.10 - 4.26 (m, 4 H), 5.96 - 6.08 (m, 1 H), 6.17 - 6.28 (m, 1 H), 6.80 - 6.97 (m, 1 H), 7.02 - 7.17 (m, 1 H), 7.29 - 7.37 (m, 1 H), 8.23 - 8.34 (m, 2 H), 9.31 - 9.46 (m, 1 H).

Example 12:

8,16,20-trifluoro-6-methyl-9-[(methylsulfanyl)methyl]-3,4,5, 6-tetrahydro-2H, 12H- 11 ,7-(azeno)- 13, 17- (metheno)- 1 ,6,12,14-benzoxatriazacyclononadecine

Preparation of Intermediate 12.1:

(2,6-dichloro-3-fluoropyridin-4-yl)methanol

To a solution of 2,6-dichloro-3-fluoropyridine-4-carboxylic acid (4.90 g, purchased from FCH group) in THF (86 mL) at 0 °C was slowly added borane THF complex solution (93 mL, 1.0 M). The mixture was allowed to warm to room temperature and stirred for 16 h. The reaction was carefully stopped by the addition of MeOH (20 mL). The mixture was partitioned between ethyl acetate (100 mL) and saturated aqueous sodium chloride solution (100 mL), and the aqueous layer was extracted three times with ethyl acetate (150 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield (2,6-dichloro-3-fluoropyridin-4-yl)methanol (4.41 g, 99% purity). LC-MS (Method a): R t = 0.90 min; MS (ESIpos): m/z = 196 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 4.60 - 4.69 (m, 2H), 5.75 - 5.83 (m, 1H), 7.51 - 7.60 (m, 1H). Preparation of Intermediate 12.2:

2,6-dichloro-4-(chloromethyl)-3-fluoropyridine

To a solution of (2,6-dichloro-3-fluoropyridin-4-yl)methanol (4.30 g) and pyridine (2.0 mL) in toluene (17 mL) at 0 °C was added thionyl chloride (2.1 mL). The mixture was stirred for an additional 30 min at 0 °C and for 24 h at room temperature. The reaction was stopped by the addition of water (200 mL) and the mixture was extracted three times with ethyl acetate (150 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated to yield the title compound (4.67 g, 80% purity) that was used without further purification.

¾ NMR (400 MHz, DMSO-i 6 , 295 K) δ/ppm = 4.81 - 4.88 (m, 2H), 7.80 - 7.86 (m, 1H).

Preparation of Intermediate 12.3:

2,6-dichloro-3-fluoro-4-[(methylsulfanyl)methyl]pyridine

To a solution of crude 2,6-dichloro-4-(chloromethyl)-3-fluoropyridine (4.67 g) in ethanol (41 mL) at 0 °C was added sodium thiomethoxide (1.10 g) portionwise. The mixture was allowed to warm to room temperature and stirred for 3 h. The reaction was stopped by the addition of half-saturated aqueous sodium chloride solution (200 mL) and the mixture was extracted three times with ethyl acetate (150 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was combined with a second reaction batch carried out as described above but with 100 mg of 2,6-dichloro-4-(chloromethyl)-3-fluoropyridine. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate) yielded the title compound (2.22 g, 95% purity).

LC-MS (Method a): R t = 1.26 min; MS (ESIpos): m/z = 226 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 2.03 (s, 3H), 3.75 - 3.82 (m, 2H), 7.63 - 7.69 (m, 1H). Preparation of Intermediate 12.4:

4-[{6-chloro-3-fluoro-4-[(methylsulfanyl)methyl]pyridin-2-yl }(methyl)amino]butan-l-ol

A solution of 2,6-dichloro-3-fluoro-4-[(methylsulfanyl)methyl]pyridine (2.00 g), 4-(methylamino)butan- l-ol (1.10 g) and 2,6-lutidine (3.1 mL) in DMSO (1 10 mL) was heated at 130 °C for 3.5 h. The mixture was allowed to cool to room temperature, diluted with water (300 mL) and extracted three times with ethyl acetate (150 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate). Further purification by flash column chromatography (silica gel, hexanes/ethyl acetate) yielded the title compound (935 mg, 95% purity).

LC-MS (Method a): R t = 1.24 min; MS (ESIpos): m/z = 293 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.34 - 1.45 (m, 2H), 1.48 - 1.63 (m, 2H), 2.02 (s, 3H), 3.01 (s, 3H), 3.36 - 3.46 (m, 4H), 3.60 - 3.66 (m, 2H), 4.38 - 4.44 (m, 1H), 6.67 - 6.70 (m, 1H).

Preparation of Intermediate 12.5:

N- {4-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]butyl} -6-chloro-3-fluoro-N-methyl-4- [(methylsulfanyl)methyl]pyridin-2-amine

To a suspension of 4-[{6-chloro-3-fluoro-4-[(methylsulfanyl)methyl]pyridin-2-yl }(methyl)amino]butan- l-ol (930 mg), 2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenol (see Intermediate 1.3; 847 mg) and triphenylphosphine (916 mg) in dichloromethane (12 mL) was added diisopropyl azodicarboxylate (690 μΐ) in dichloromethane (3 mL), and the mixture was stirred at room temperature for 7.5 h. The mixture was concentrated and the crude product was purified by flash column chromatography to yield the title compound (2.05 g, 80% purity) that was used without further purification.

LC-MS (Method a): R t = 1.44 min; MS (ESIpos): m/z = 497 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.54 - 1.66 (m, 4H), 1.97 - 2.00 (m, 2H), 2.92 - 2.97 (m, 3H), 3.37 - 3.44 (m, 2H), 3.60 - 3.64 (m, 2H), 4.00 - 4.04 (m, 2H), 5.80 - 5.86 (m, 2H), 6.32 - 6.38 (m, 1H), 6.67 - 6.72 (m, 1H), 6.80 - 6.90 (m, 1H), 6.98 - 7.05 (m, 1H), 7.23 - 7.33 (m, 1H), 7.76 - 7.83 (m, 1H).

Example 12 - Preparation of the End Product:

A degassed suspension of crude N- {4-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenoxy]butyl}- 6- chloro-3-fluoro-N-methyl-4-[(methylsulfanyl)methyl]pyridin-2 -amine (1.90 g), chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (506 mg), 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (292 mg) and potassium phosphate (1.95 g) in toluene (150 mL) and N-methylpyrrolidone (15 mL) was heated at 130 °C for 18 h. Additional portions of chloro(2-dicyclohexylphosphino-2',4',6'-tri-z ' o-propyl- l,l'-biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (506 mg) and 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (292 mgk) were added, and the mixture was stirred for an additional 7 h at 130 °C. The mixture was allowed to cool to room temperature, diluted with water (150 mL) and extracted three times with ethyl acetate (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate). Further purification by flash column chromatography (silica gel, hexanes/ethyl acetate) and preparative HPLC yielded the title compound (221 mg, 95% purity).

HPLC Purification:

Instrument: pump: Labomatic HD-5000, head HDK 280, low pressure gradient module ND-B1000; manual injection valve: Rheodyne 3725Ϊ038; detector: Knauer Azura UVD 2.15; collector: Labomatic Labocol Vario-4000; column: Chromatorex RP C-18 10 μηι, 125x30mm; solvent A: water + 0.2 vol-%> ammonia (32%), solvent B: acetonitrile; gradient: 0.00-0.50 min 65% B (150 ml/min), 0.50-6.00 min 65- 100% B (150 ml/min), 6.00-8.00 min 100% B (150 ml/min); detection: UV 277 nm.

LC-MS (Method b): R t = 1.59 min; MS (ESIpos): m/z = 461 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) 8/ppm = 1.75 - 1.91 (m, 4H), 2.01 - 2.08 (m, 3H), 3.01 - 3.10 (m, 3H), 3.33 - 3.41 (m, 2H), 3.51 - 3.59 (m, 2H), 4.03 - 4.15 (m, 2H), 6.19 - 6.27 (m, 1H), 6.81 - 6.93 (m, 1H), 7.06 - 7.13 (m, 1H), 7.25 - 7.34 (m, 1H), 8.08 - 8.15 (m, 1H), 8.22 - 8.29 (m, 1H), 8.22 - 8.29 (m, 1H), 9.35 - 9.45 (m, 1H). Example 13:

(rac)-fert-butyl [{[15,19-difluoro- 1 ,2,3,4-tetrahydro- 16,12-(azeno)-6, 10-(metheno)-5, 1,11,13- benzoxatriazacyclooctadecin-8(l lH)-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

Preparation of Intermediate 13.1

2-(3- {3-[(methylsulfanyl)methyl]-5-nitrophenoxy}propyl)-lH-isoind ole-l,3(2H)-dione

To a solution of 3-[(methylsulfanyl)methyl]-5-nitrophenol (2.21 g, prepared according to WO2015/155197 Al, 2015, Intermediate 1.2) in DMF (22 mL) was sequentially added potassium carbonate (2.30 g) and 2-(3-bromopropyl)-lH-isoindole-l,3(2H)-dione (3.27 g) and the mixture was stirred at room temperature for 22 h. The reaction mixture was diluted with EtOAc (50 mL), and the reaction was stopped by the addition of saturated aqueous sodium bicarbonate solution (50 mL). The layers were separated and the aqueous layer was extracted twice with EtOAc (50 mL each). The combined organic layers were sequentially washed with water and saturated aqueous sodium chloride, dried and concentrated to yield the title compound (4.25 g, 95% purity) that was used without further purification.

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.93 (s, 3H), 2.06 - 2.13 (m, 2H), 3.73 - 3.81 (m, 4H), 4.11 - 4.17 (m, 2H), 7.15 - 7.20 (m, 1H), 7.37 - 7.44 (m, 1H), 7.72 - 7.78 (m, 1H), 7.81 - 7.89 (m, 4H). Preparation of Intermediate 13.2:

rac-2-[3-(3- {[(S)-methylsulfinyl]methy^

O

To a solution of 2-(3- {3-[(methylsulfanyl)methyl]-5-nitrophenoxy}propyl)-lH-isoind ole-l,3(2H)-dione (4.23 g, 95 % purity) in acetonitrile (320 ml) at 0 °C was added iron trichloride (169 mg) and the mixture was stirred for 10 min. Then, periodic acid (7.11 g) was added and the mixture was stirred for an additional 2 h at 0 °C. The reaction was stopped by the addition of saturated aqueous sodium thiosulfate solution (100 mL). The mixture was stirred for 10 min and extracted twice with ethyl acetate (250 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated to yield crude the title compound (4.55 g, 90% purity) that was used without further purification.

LC-MS (Method a): R t = 0.99 min; MS (ESIpos): m/z = 403 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 2.06 - 2.15 (m, 2H), 2.48 (s, 3H), 3.75 - 3.83 (m, 2H), 3.99 - 4.08 (m, 1H), 4.13 - 4.20 (m, 2H), 4.22 - 4.29 (m, 1H), 7.20 - 7.26 (m, 1H), 7.50 - 7.55 (m, 1H), 7.75 - 7.80 (m, 1H), 7.82 - 7.89 (m, 4H).

Preparation of Intermediate 13.3:

(rac)-fert-butyl [ {3-[3-(l ,3-dioxo-l ,3-dihydro-2H-isoindol-2-yl)propoxy]-5-nitrobenzyl} (methyl) oxido-

To a suspension of crude rac-2-[3-(3- {[(S)-methylsulfinyl]methyl}-5-nitrophenoxy)propyl]-lH- isoindole-l,3(2H)-dione (4.55 g), tert-butyl carbamate (1.98 g), magnesium oxide (1.82 g) and rhodium(II)acetate dimer (125 mg) in dichloromethane (120 mL) was added portionwise iodobenzene diacetate (5.46 g), and the mixture was heated to 40 °C for 4 h. The mixture was kept at - 20 °C for 72 h and subsequently stirred at 40 °C for an additional 3 h. The mixture was allowed to cool to room temperature, filtered over a pad of Celite and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (5.83 g, 80% purity) that was used without further purification.

LC-MS (Method a): R t = 1.20 min; MS (ESIpos): m/z = 518 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.36 (s, 9H), 1.93 - 1.93 (m, 1H), 2.06 - 2.15 (m, 2H), 3.13 (s, 3H), 3.74 - 3.80 (m, 2H), 4.13 - 4.20 (m, 2H), 4.93 - 5.04 (m, 2H), 7.35 - 7.41 (m, 1H), 7.59 - 7.64 (m, 1H), 7.81 - 7.91 (m, 5H).

Preparation of Intermediate 13.4

(rac)-tert-buty\ [{3-amino-5-[3-(l,3-dioxo-l,3-dihydro-2H-isoindol-2- yl)propoxy]benzyl}(methyl)oxido^ 6 -sulfanylidene]carbamate

To a solution of crude (rac)-tert-butyl [{3-[3-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)propoxy]-5- nitrobenzyl}(methyl)oxido^ 6 -sulfanylidene]carbamate (2.90 g) in methanol (120 mL) was added platinum 1% and vanadium 2% on activated carbon (344 mg). The mixture was purged with hydrogen gas (1 atm) and stirred for 0.5 h. Additional platinum 1% and vanadium 2% on activated carbon (200 mg) was added and the mixture was stirred under an atmosphere of hydrogen for 2 h. Additional platinum 1% and vanadium 2% on activated carbon (150 mg) was added and the mixture was stirred under an atmosphere of hydrogen for 1.5 h. The mixture was stirred under nitrogen for 15 h. Additional platinum 1% and vanadium 2% on activated carbon (200 mg) was added and the mixture was stirred under an atmosphere of hydrogen for 1 h. The mixture was diluted with THF (75 mL) and additional platinum 1% and vanadium 2% on activated carbon (200 mg) was added and the mixture was stirred under an atmosphere of hydrogen for 1 h. The mixture was filtered and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (740 mg, 95% purity). ¾ NMR (400 MHz, DMSO-i 6 , 295 K) δ/ρρηι = 1.38 (s, 9H), 1.98 - 2.06 (m, 2H), 3.04 (s, 3H), 3.71 - 3.76 (m, 2H), 3.85 - 3.93 (m, 2H), 4.48 - 4.62 (m, 2H), 5.16 - 5.23 (m, 2H), 6.03 - 6.12 (m, 2H), 6.13 - 6.19 (m, 1H), 7.81 - 7.89 (m, 4H).

Preparation of Intermediate 13.5:

(rac)-tert-buty\ [(3- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2-yl]amino}-5-[3- (l,3-dioxo-l,3- dihydro-2H-isoindol-2-yl)propoxy]benzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate

A suspension of (rac)-tert-butyl [{3-amino-5-[3-(l,3-dioxo-l,3-dihydro-2H-isoindol-2- yl)propoxy]benzyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate (250 mg), 2-chloro-4-(2,4- difluorophenyl)-5-fluoropyrimidine (251 mg, purchased from Apollo Scientific), chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (42.4 mg), 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (24.4 mg) and potassium phosphate (544 mg) in toluene (51 mL) and N-methylpyrrolidone (6.2 mL) was stirred for 18 h at 130 °C. Additional portions of 2-chloro-4-(2,4-difluorophenyl)-5-fluoropyrimidine (125 mg), chloro(2-dicyclohexylphosphino-2',4',6' ri-wo-propyl-l,r-biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (21.2 mg) and 2-(dicyclohexylphosphino)-2',4',6'- triisopropylbiphenyl (12.2 mg) were added, and the mixture was stirred for 4 h at 130 °C. Additional portions of 2-chloro-4-(2,4-difluorophenyl)-5-fluoropyrimidine (63 mg), chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l,r-biphenyl) [2-(2-aminoethyl)phenyl] palladium(II) methyl-tert-butylether adduct (10.6 mg) and 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (6.1 mg) were added, and the mixture was stirred for 16 h at 1 10 C. The mixture was allowed to cool to room temperature, filtered, diluted with water (100 mL) and extracted three times with ethyl acetate (150 mL each). The combined organic layers were washes with saturated aqueous sodium chloride solution, dried and concentrated. The residue was treated with saturated aqueous sodium chloride solution (100 mL) and extracted three times with diethyl ether (120 mL each). The combined organic layers were purified and concentrated. The crude product was combined with a second reaction batch carried out similarly as described above but with 200 mg of (rac)-fert-butyl [ {3-amino-5-[3-(l,3-dioxo-l,3-dihydro-2H-isoindol- 2-yl) ro oxy]benzyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (337 mg, 80% purity) that was contaminated by impurities and used without further purification.

LC-MS (Method a): R t = 1.45 min; MS (ESIpos): m/z = 696 [M+H] + Preparation of Intermediate 13.6:

(rac)-fert-butyl {[3-(3-aminopropoxy)-5- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2- yl] amino} benzyl] (methyl)oxido^ 6 -sulfanylidene } carbamate

To a suspension of crude (rac)-tert-butyl [(3- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2-yl]amino}-5- [3-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)propoxy]benzyl)(m ethyl)oxido-λ 6 -sulfanylidene]carbamate (337 mg, 80% purity) in ethanol (14 mL) and THF (3.0 mL) was added aqueous hydrazine (210 L, 35% purity) and the mixture was heated to reflux for 4 h. The mixture was cooled to 5 °C, filtered and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl actetate/methanol) to yield the title compound (165 mg, 95% purity).

LC-MS (Method a): R t = 1.03 min; MS (ESIpos): m/z = 566 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.36 (s, 9H), 1.75 - 1.78 (m, 2H), 2.65 - 2.70 (m, 2 H), 3.07 - 3.12 (m, 2H), 3.13 - 3.19 (m, 2H), 3.96 - 4.00 (m, 2H), 4.69 - 4.83 (m, 2H), 6.54 - 6.64 (m, 1H), 7.30 - 7.41 (m, 2H), 7.42 - 7.54 (m, 2H), 7.79 - 7.91 (m, 1H), 8.65 - 8.74 (m, 1H), 9.95 - 10.02 (m, 1H).

Example 13 - Preparation of the End Product:

To a solution of (rac)-fert-butyl {[3-(3-aminopropoxy)-5- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2- yl]amino}benzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (165 mg) in DMSO (11 mL) was added triethylamine (49 \L) and the mixture was heated to 150 °C for 5 h. The mixture was allowed to cool to room temperature, diluted with water (25 mL) and extracted twice with with ethyl acetate (100 mL each). The combined organic layers were sequentially washed with saturated aqueous sodium bicarbonate solution and saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (255 mg, 88% purity) that was used without further purification. A pure sample was obtained by preparative HPLC.

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.37 min; MS (ESIpos): m/z = 546 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.38 (s, 9H), 1.94 (br s, 2H), 3.11 - 3.21 (m, 5H), 4.26 (br t, 2H), 4.72 (s, 2H), 6.38 - 6.50 (m, 1H), 6.55 (s, 1H), 6.59 - 6.64 (m, 1H), 6.79 (s, 1H), 7.58 - 7.66 (m, 1H), 7.67 - 7.75 (m, 1H), 8.29 (s, 1H), 8.52 - 8.57 (m, 1H), 8.65 (d, 1H), 9.94 (s, 1H).

Example 14:

rac- 15,19-difluoro-8-[(S-methylsulfonimidoyl)methyl] - 1 ,2,3,4-tetrahydro- 16,12-(azeno)-6, 10- (metheno)-5, 1,11, 13-benzoxatriazacyclooctadecine

To a solution of (rac)-tert-butyl [{[15,19-difluoro-l,2,3,4-tetrahydro-16,12-(azeno)-6,10-(met heno)- 5,1,11, 13-benzoxatriazacyclooctadecin-8(l lH)-yl]methyl} (methyl)oxido^ 6 -sulfanylidene]carbamate (215 mg, 88%o purity) in dichloromethane (2.7 mL) was added trifluoroacetic acid (870 μΐ) and the mixture was stirred for 2 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate (30 mL), and the mixture was extracted three times with dichloromethane (60 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated. The crude product was purified by preparative HPLC to yield the title compound (14.6 mg, 94% purity, 9%). HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.14 min; MS (ESIpos): m/z = 446 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.86 - 2.02 (m, 2H), 2.81 (s, 3H), 3.08 - 3.22 (m, 2H), 3.57 (s, 1H), 4.16 - 4.30 (m, 4H), 6.39 - 6.51 (m, 1H), 6.51 - 6.66 (m, 2H), 6.67 - 6.85 (m, 1H), 7.60 - 7.71 (m, 1H), 7.75 - 7.85 (m, 1H), 8.11 - 8.28 (m, 1H), 8.59 - 8.69 (m, 1H), 9.81 - 9.95 (m, 1H).

Example 15:

(rac)-tert-butyl [{[16,20-difluoro-2,3,4,5-tetrahydro-lH,12H-13,17-(azeno)-l l,7-(metheno)-6,l, 12,14- benzoxatriazacyclononadecin-9-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

Prepration of Intermediate 15.1:

ethyl 4- {3-[(methylsulfanyl)methyl]-5-nitrophenoxy}butanoate

To a suspension of 3-[(methylsulfanyl)methyl]-5-nitrophenol (6.00 g) and potassium carbonate (4.99 g) in DMF (58 ml) at 0°C was added dropwise ethyl 4-bromobutanoate (4.7 mL). The mixture was allowed to warm to room temperature and stirred for 24 h. The reaction was diluted with water (300 mL) and the mixture was extracted three times with ethyl acetate (200 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated to yield the title compound (11.69 g, 90% purity) that was contaminated by DMF and excess ethyl 4-bromobutanoate and which was used without further purification.

LC-MS (Method a): R t = 1.35 min; MS (ESIpos): m/z = 314 [M+H] +

TI NMR ^OO MHz, DMSO-i/ 6 , 295 K) 8/ppm = 1.15 - 1.21 (m, 3H), 1.94 - 2.03 (m, 5H), 3.74 - 3.81 (m, 2H), 4.02 - 4.14 (m, 4H), 7.33 - 7.36 (m, 1H), 7.57 - 7.61 (m, 1H), 7.75 - 7.80 (m, 1H) (one methylene group is overlayed by residual DMSO).

Prepration of Intermediate 15.2:

(rac)-ethyl 4-(3 - { [ S-methylsulfinyl]methyl} -5-nitrophenoxy)butanoate

To a solution of crude ethyl 4- {3-[(methylsulfanyl)methyl]-5-nitrophenoxy}butanoate (11.7 g) in acetonitrile (410 mL) at 0 °C was added iron trichloride (605 mg) and the mixture was stirred for 15 min. Then, periodic acid (25.5 g) was added and the reaction was stirred for 1.5 h at 0 °C. The reaction was stopped by the addition of saturated aqueous sodium thiosulfate solution, and the mixture was extracted three times with ethyl acetate (300 mL each). The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated to yield the title compound (10.5 g, 99%> purity) that was used without further purification.

LC-MS (Method a): R t = 0.96 min; MS (ESIpos): m/z = 330 [M+H] +

TI NMR ^OO MHz, DMSO-i/ 6 , 295 K) 8/ppm = 1.15 - 1.21 (m, 3H), 1.97 - 2.06 (m, 2H), 4.04 - 4.15 (m, 5H), 4.24 - 4.31 (m, 1H), 7.28 - 7.36 (m, 1H), 7.65 - 7.69 (m, 1H), 7.76 - 7.82 (m, 1H) (the methyl group and one methylene group is overlayed by residual DMSO). Prepration of Intermediate 15.3:

(rac)-ethyl 4-(3- {[N-(tert-butoxycarbonyl)-S-methylsulfonimidoyl]methyl}-5-ni rrophenoxy)butanoate

To a suspension of (rac)-ethyl 4-(3- {[ S-methylsulfinyl]methyl}-5-nitrophenoxy)butanoate (10.5 g), tert- butyl carbamate (5.60 g), magnesium oxide (5.14 g), and rhodium(II)acetate dimer (352 mg) in dichloromethane (530 mL) was added iodobenzene diacetate (15.4 g), and the mixture was stirred for 4.5 h at 45 °C. Additional portions of teri-butyl carbamate (1.87 g), rhodium(II)acetate dimer (117 mg) and iodobenzene diacetate (5.1 g) were added, and the mixture was stirred for further 12 h at 45 °C. The mixture was allowed to cool to room temperature, filtered over a pad of Celite and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (12.8 g, 97% purity).

LC-MS (Method a): R t = 1.22 min; MS (ESIpos): m/z = 445 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.15 - 1.24 (m, 3H), 1.39 (s, 9H), 1.98 - 2.06 (m, 2H), 2.44 - 2.44 (m, 1H), 3.09 - 3.19 (m, 3H), 4.04 - 4.16 (m, 4H), 4.95 - 5.10 (m, 2H), 7.41 - 7.47 (m, 1H), 7.73 - 7.80 (m, 1H), 7.88 - 7.94 (m, 1H) (two protons overlayed by residual DMSO).

Prepration of Intermediate 15.4:

(rac)-tert-butyl {[3-(4-hydroxybutoxy)-5-nitrobenzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate

To a solution of (rac)-ethyl 4-(3- {[N-(teri-butoxycarbonyl)-S-methylsulfonimidoyl]methyl}-5- nitrophenoxy)butanoate (12.8 g) in THF (210 mL) at -20 °C was added dropwise diisobutylaluminum hydride (120 mL, 1.0 M in THF). The mixture was allowed to warm to room temperature and stirred for 2.5 h. The reaction was stopped by the addition of saturated aqueous sodium potassium tartrate solution. The mixture was vigorously stirred for 2 h and subsequently extracted three times with ethyl acetate (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate -> ethyl acetate/methanol) to yield the title compound (8.01 g, 97% purity).

LC-MS (Method a): R t = 1.02 min; MS (ESIpos): m/z = 403 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.39 (s, 9 H), 1.51 - 1.65 (m, 2 H), 1.73 - 1.83 (m, 2 H), 3.14 (s, 3 H), 3.43 - 3.51 (m, 2 H), 4.09 - 4.16 (m, 2 H), 4.46 - 4.51 (m, 1 H), 4.93 - 5.07 (m, 2 H), 7.37 - 7.50 (m, 1 H), 7.73 - 7.79 (m, 1 H), 7.86 - 7.92 (m, 1 H).

Prepration of Intermediate 15.5:

2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluoroaniline

To a solution of 2,4-dichloro-5-fluoropyrimidine (2.46 g), 5-fluoro-2-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)aniline (3.50 g, purchased from Milestone Pharmtech USA Inc.) and tetrakis(triphenylphosphino)palladium(0) (1.71 g) in 1 ,2-dimethoxy ethane (120 mL) was added aqueous potassium carbonate (30 mL, 1.5 M) and the mixture was heated to 90 °C for 16 h. The mixture was allowed to cool to room temperature and diluted with ethyl acetate. The mixture was washed with saturated aqueous sodium chloride, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl actate) to yield the title compound (2.33 g, 95%> purity).

LC-MS (Method a): R t = 1.11 min; MS (ESIpos): m/z = 242 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 6.29 - 6.40 (m, 2H), 6.42 - 6.51 (m, 1H), 6.54 - 6.61 (m, 1H), 7.41 - 7.48 (m, 1H), 8.84 - 8.89 (m, 1H).

Prepration of Intermediate 15.6:

N-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenyl]-2-nit robenzenesulfonamide

To a suspension of 2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluoroaniline (2.33 g), 2-nitrobenzenesulfonyl chloride (2.56 g) and 4-dimethylaminopyridine (58.9 mg) in dichloromethane (12 mL) was added pyridine (940 μΐ.) and the mixture was stirred for 16 h. Additional portions of 2-nitrobenzenesulfonyl chloride (2.56 g) (0.4 eq.), 4-dimethylaminopyridine (58.9 mg) and pyridine (940 μΐ.) were added, and the mixture was stirred for an additional 4 h. The reaction was stopped by the addition of aqueous hydrochloric acid (IN, 100 mL) and the mixture was extracted three times with dichloromethane (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (3.46 g, 95% purity).

LC-MS (Method a): R t = 1.22 min; MS (ESIpos): m/z = 427 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 7.11 - 7.22 (m, 1H), 7.27 - 7.38 (m, 1H), 7.54 - 7.65 (m, 1H), 7.78 - 7.93 (m, 3H), 7.94 - 8.02 (m, 1H), 8.84 - 8.94 (m, 1H), 10.50 - 10.69 (m, 1H). Prepration of Intermediate 15.7

(rac)-tert-buty\ {[3-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenyl] [(2- nitrophenyl)sulfonyl] amino } butoxy)-5 -nitrobenzyl] (methyl)oxido^ 6 -sulfanylidene } carbamate

To a solution of N-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenyl]-2-nit robenzenesulfonamide (3.00 g), (rac)-fert-butyl {[3-(4-hydroxybutoxy)-5-nitrobenzyl](methyl)oxido^ 6 - sulfanylidene} carbamate (2.83 g) and triphenylphopshine (3.69 g) in dichloromethane (140 mL) at 0 °C was added diisopropyl azodicarboxylate (2.8 mL). The mixture was allowed to warm to room temperature, stirred for 4 h and concentrated. The residue was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (4.58 g, 70% purity) that was used without further purification.

LC-MS (Method a): R t = 1.39 min; MS (ESIpos): m/z = 812 [M+H] + Prepration of Intermediate 15.8:

(rac)-fert-butyl {[3-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenyl]amino}b utoxy)-5- nitrobenzyl](methyl)oxido^ 6 -sulfanylidene}carbamate

To a solution of crude (rac)-tert-butyl {[3-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenyl] [(2- nitrophenyl)sulfonyl] amino } butoxy)-5 -nitrobenzyl] (methyl)oxido^ 6 -sulfanylidene} carbamate (4.58 g) in DMF (75 ml) was added cesium carbonate (2.58 g) and the mixture was stirred for 2 min. Then, thiophenol (490 μΐ) was added and the mixture was stirred for 18 h. The mixture was diluted with water (150 mL) and extracted three times with ethyl acetate (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by preparative HPLC to yield the title compound (1.09 g, 99 % purity). HPLC Purification:

Instrument: Labomatic HD3000, AS-3000, Labcol Vario 4000 Plus, Knauer DAD 2600; column: YMC 10μ; eluent A: water + 0.1 Vol-% formic acid (99%), eluent B: acetonitrile; gradient: 0.00-1.00 min 60% B (50->200mL/min), 1.00-10.00 min 60-80% B (200mL/min), DAD scan: 254 nm

LC-MS (Method a): R t = 1.50 min; MS (ESIpos): m/z = 626 [M+H] +

Ή NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.37 (s, 9H), 1.68 - 1.81 (m, 2H), 1.81 - 1.92 (m, 2H), 3.12 - 3.17 (m, 3H), 3.18 - 3.24 (m, 2H), 4.15 - 4.21 (m, 2H), 4.93 - 5.06 (m, 2H), 6.44 - 6.55 (m, 1H), 6.56 - 6.65 (m, 1H), 6.91 - 6.99 (m, 1H), 7.40 - 7.46 (m, 1H), 7.46 - 7.52 (m, 1H), 7.73 - 7.81 (m, 1H), 7.87 - 7.93 (m, 1H), 8.84 - 8.89 (m, 1H)

Prepration of Intermediate 15.9:

(rac)-tert-buty\ {[3-amino-5-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenyl] amino } butoxy)benzyl] (methyl)oxido^ 6 -sulfanylidene } carbamate

To a solution of (rac)-tert-butyl {[3-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenyl] amino }butoxy)-5-nitrobenzyl](methyl)oxido^ 6 -sulfanylidene} carbamate (1.09 g) in methanol (28 mL) and THF (8.5 mL) was added platinum 1%> and vanadium 2% on activated carbon (170 mg). The mixture was purged with hydrogen gas (1 atm) and stirred for 2.5 h. The mixture was filtered and concentrated to yield the title compound (1.02 g, 99% purity) that was used without further purification.

LC-MS (Method a): R t = 1.39 min; MS (ESIpos): m/z = 597 [M+H] + 'H NMR (400 MHz, DMSO-de, 295 K) δ ppm 1.35 - 1.39 (m, 9H), 1.63 - 1.74 (m, 2H), 1.74 - 1.85 (m, 2H), 3.00 - 3.09 (m, 3H), 3.12 - 3.22 (m, 2H), 3.85 - 3.92 (m, 2H), 4.52 - 4.66 (m, 2H), 5.15 - 5.24 (m, 2H), 6.1 1 - 6.22 (m, 3H), 6.46 - 6.54 (m, 1H), 6.56 - 6.64 (m, 1H), 6.91 - 6.99 (m, 1H), 7.42 - 7.53 (m, 1H), 8.85 - 8.90 (m, 1H).

Example 15 - Prepration of the End Product:

A degassed suspension of (rac)-tert-butyl {[3-amino-5-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenyl]amino}butoxy)benzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (210 mg), chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (58.3 mg), 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (84.0 mg) and potassium phosphate (374 mg) in toluene (21 mL) and N-methylpyrrolidone (2.1 mL) was heated at 130 °C for 7 h. The mixture was allowed to cool to room temperature, diluted with water (60 mL) and extracted three times with ethyl acetate (30 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate) yielded the title compound (85 mg, 98% purity).

LC-MS (Method a): R t = 1.37 min; MS (ESIpos): m/z = 560 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.38 (s, 9H), 1.57 - 1.68 (m, 4H), 3.1 1 (s, 3H), 3.24 - 3.50 (m, 4H), 4.56 - 4.73 (m, 2H), 5.93 - 6.08 (m, 1H), 6.44 - 6.49 (m, 1H), 6.62 - 6.67 (m, 1H), 6.70 - 6.75 (m, 1H), 6.76 - 6.84 (m, 1H), 7.37 - 7.49 (m, 1H), 8.00 - 8.08 (m, 1H), 8.57 - 8.63 (m, 1H), 9.86 - 9.95 (m, 1H).

Example 16:

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 1 H- 17, 13-(azeno)-7, 1 1 - (metheno)-6, 1 ,12, 14-benzoxatriazacyclononadecine

To a solution of (rac)-tert-butyl [{[16,20-difluoro-2,3,4,5-tetrahydro-lH-17,13-(azeno)-7,l l-(metheno)- 6,1,12,14-benzoxatriazacyclononadecin-9(l 2H)-yl]methyl} (methyl)oxido-λ 6 -sulfanylidene]carbamate (80.0 mg) in dichloromethane (1.2 mL) was added trifluoroacetic acid (280 μΐ.) and the mixture was stirred for 1.5 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate solution (5 mL), and the mixture was extracted with dichloromethane. The combined organic layers were washed with saturated aqueous sodium chloride, dried and concentrated to yield the title compound (66 mg, 98% purity).

LC-MS (Method a): R t = 1.15 min; MS (ESIpos): m/z = 460 [M+H] +

¾ NMR (400 MHz, DMSO-de, 295 K) δ/ppm = 1.61 - 1.84 (m, 4H), 2.79 (s, 3H), 3.15 - 3.28 (m, 2H), 3.54 (s, 1H), 4.08 - 4.15 (m, 2H), 4.21 (d, 2H), 6.34 - 6.46 (m, 1H), 6.45 - 6.55 (m, 2H), 6.60 - 6.76 (m, 2H), 7.13 - 7.24 (m, 1H), 7.90 - 8.00 (m, 1H), 8.65 (d, 1 H), 9.75 (s, 1H).

Example 17:

(rac)- 16,20-difluoro-6-methyl-9-[(S-methylsulfonimidoyl)methyl] - 1 ,2,3,4,5, 6-hexahydro- 12H- 17,13- (azeno)-l l,7-(metheno)- 1,6, 12,14-benzotetraazacyclononadecine; salt with formic acid

Preparation of Intermediate 17.1:

teri-butyl [4-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)butyl]methylcarba mate

To a solution of tert-butyl (4-hydroxybutyl)methylcarbamate (1.25 g), lH-isoindole-l,3(2H)-dione (1.36 g) and triphenylphosphine (1.77 g) in dichloromethane (15 ml) at 0 °C was added diisopropyl azodicarboxylate (1.3 mL). The mixture was allowed to warm to room temperature and stirred for an additional 18 h. The reaction was stopped by the addition of water (60 mL) and the mixture was extracted twice with dichloromethane (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (3.33 g, 57% purity) that was used without further purification.

LC-MS (Method a): R t = 1.26 min; MS (ESIpos): m/z = 333 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.27 - 1.39 (br s, 9H), 1.41 - 1.59 (m, 4H), 2.68 - 2.79 (m, 3H), 3.06 - 3.23 (m, 2H), 3.51 - 3.70 (m, 2H), 7.76 - 7.91 (m, 4H).

Preparation of Intermediate 17.2:

2- [4-(methylamino)butyl] - 1 H-isoindole- 1 ,3 (2H)-dione

H 3 C

To a solution of tert-butyl [4-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)butyl]methylcarba mate (2.83 g) in dichloromethane (66 mL) was added trifluoroacetic acid (21 mL) and the mixture was stirred for 2 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate solution (40 mL) and the mixture was extracted three times with dichloromethane (40 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (870 mg, 99% purity).

LC-MS (Method a): R t = 0.58 min; MS (ESIpos): m/z = 233 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.32 - 1.46 (m, 2H), 1.55 - 1.65 (m, 2H), 2.18 - 2.24 (m, 3H), 2.39 - 2.46 (m, 2H), 3.52 - 3.61 (m, 2H), 7.81 - 7.89 (m, 4H) (NH obscured). Preparation of Intermediate 17.3:

(rac)-fert-butyl [(3- {[4-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)butyl](methyl)

nitrobenzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate

To a solution of 2-[4-(methylamino)butyl]-lH-isoindole-l,3(2H)-dione (795 mg) and (rac)-fert-butyl [(3-fluoro-5-nitrobenzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (see Intermediate 6.5; 1.14 g) in DMSO (34 mL) was added triethylamine (570 μΐ.) and the mixture was heated at 100 °C for 5 h, at 115 °C for 16 h and at 150 °C for 4 h. The mixture was allowed to cool to room temperature, diluted with water (50 mL) and extracted three times with ethyl acetate (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concenctrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (430 mg, 95% purity).

LC-MS (Method a): R t = 1.31 min; MS (ESIpos): m/z = 545 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm 1.38 (s, 9H), 1.50 - 1.65 (m, 4H), 2.95 - 3.00 (m, 3H), 3.05 - 3.12 (m, 3H), 3.39 - 3.48 (m, 2H), 3.57 - 3.62 (m, 2H), 4.83 - 4.98 (m, 2H), 7.15 - 7.19 (m, 1H), 7.34 - 7.38 (m, 1H), 7.43 - 7.49 (m, 1H), 7.80 - 7.89 (m, 4H).

Preparation of Intermediate 17.4:

(rac)-fert-butyl [(3-amino-5- {[4-(l,3-dioxo-l,3-dihydro-2H-isoindol-2- yl)butyl] (methyl)amino } benzyl)(methyl)oxido^ 6 -sulfanylidene] carbamate

To a solution of (rac)-tert-butyl[(3- {[4-(l,3-dioxo-l,3-dihydro-2H-isoindol-2-yl)butyl](methyl)am ino}- 5-nitrobenzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (430 mg) in methanol (19 mL) and THF (1.9 mL) was added platinum 1% and vanadium 2% on activated carbon (77 mg). The mixture was purged with hydrogen gas (1 atm) and stirred for 0.75 h. Additional platinum 1% and vanadium 2% on activated carbon (100 mg) was added .The mixture was purged with hydrogen gas (1 atm) and stirred for 2.5 h. The mixture was filtered and concentrated to yield the title compound (403 mg, 95% purity) that was used without further purification.

LC-MS (Method a): R t = 1.04 min; MS (ESIpos): m/z = 516 [M+H] +

'H NMR (400 MHz, DMSO-ifc, 295 K) 8/ppm = 1.36 - 1.40 (m, 9H), 1.44 - 1.55 (m, 2H), 1.55 - 1.63 (m, 2H), 2.76 - 2.81 (m, 3 H), 2.97 - 3.02 (m, 3H), 3.20 - 3.27 (m, 2H), 3.55 - 3.61 (m, 2H), 4.45 - 4.58 (m, 2H), 4.86 - 4.97 (m, 2H), 5.87 - 5.94 (m, 2H), 5.96 - 6.01 (m, 1H), 7.80 - 7.89 (m, 4H)

Preparation of Intermediate 17.5:

(rac)-fert-butyl [(3- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2-yl]amino}-5- {[4-(l,3-dioxo-l,3- dihydro-2H-isoindol-2-yl)butyl] (methyl)amino } benzyl)(methyl)oxido^ 6 -sulfanylidene] carbamate

A suspension of (rac)-fert-butyl [(3-amino-5- {[4-(l,3-dioxo-l,3-dihydro-2H-isoindol-2- yl)butyl](methyl)amino}benzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (378 mg), 2-chloro-4-(2,4- difluorophenyl)-5-fluoropyrimidine (359 mg, purchased from Apollo Scientific), chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (121 mg), 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (70.0 mg) and potassium phosphate (1.20 g) in 1,4-dioxane (23 mL) was heated to 115 °C for 3 h. The mixture was allowed to cool to room temperature, filtered, diluted with saturated aqueous sodium chloride solution (50 mL) and extracted three times with ethyl acetate (100 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (541 mg, 90% purity) that was used without further purification.

LC-MS (Method a): R t = 1.47 min; MS (ESIpos): m/z = 723 [M+H] +

Ti NMR ^OO MHz, DMSO-i/ 6 , 295 K) 8/ppm = 1.31 - 1.38 (m, 9H), 1.48 - 1.59 (m, 4H), 2.81 - 2.89 (m, 3H), 3.02 - 3.07 (m, 3H), 3.25 - 3.31 (m, 2H), 3.53 - 3.62 (m, 2H), 4.62 - 4.72 (m, 2H), 6.33 - 6.43 (m, 1H), 7.05 - 7.08 (m, 1H), 7.15 - 7.22 (m, 1H), 7.22 - 7.34 (m, 2H), 7.41 - 7.51 (m, 1H), 7.77 - 7.88 (m, 5H), 8.59 - 8.65 (m, 1H), 9.68 - 9.77 (m, 1H) Preparation of Intermediate 17.6:

(rac)-fert-butyl [(3-[(4-aminobutyl)(methyl)amino]-5- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2- yl]amino}benzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate

To a suspension of (rac)-fert-butyl [(3- {[4-(2,4-difluorophenyl)-5-fluoropyrimidin-2-yl]amino}-5- {[4- ( 1 ,3 -dioxo- 1 ,3 -dihydro-2H-isoindol-2-yl)butyl] (methyl)amino } benzyl)(methyl)oxido^ 6 - sulfanylidene]carbamate (441 mg) in ethanol (23 mL) and THF (4.7 mL) was added aqueous hydrazine (330 μL, 35 % purity) and the mixture was heated to reflux for 4 h. The mixture was cooled to 5 °C, filtered and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to yield the title compound (221 mg, 85% purity).

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.35 (s, 9H), 1.43 - 1.59 (m, 4H), 2.83 - 2.89 (m, 4H), 3.05 - 3.11 (m, 4H), 3.21 - 3.31 (m, 2H), 4.63 - 4.76 (m, 2H), 6.28 - 6.41 (m, 1H), 7.03 - 7.11 (m, 1H), 7.18 - 7.26 (m, 1H), 7.27 - 7.36 (m, 1H), 7.46 - 7.55 (m, 1H), 7.79 - 7.88 (m, 1H), 8.62 - 8.69 (m, 1H), 9.72 - 9.80 (m, 1H).

Example 17 - Preparation of the End Product

To a solution (rac)-fert-butyl [(3-[(4-aminobutyl)(methyl)amino]-5- {[4-(2,4-difluorophenyl)-5- fluoropyrimidin-2-yl]amino}benzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (211 mg) in DMSO (14 mL) was added triethylamine (60 μL) and the mixture was stirred at 120 °C for 4 h. The mixture was allowed to cool to room temperature, diluted with water (30 mL) and extracted twice with ethyl acetate (80 mL each). The combined organic layers were sequentially washed with saturated aqueous bicarbonate solution and saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate ethyl acetate/methanol) to crude (rac)-fert-butyl [{[16,20-difluoro-6-methyl-l,2,3,4,5,6-hexahydro-12H-17,13- (azeno)- 11 ,7-(metheno)- 1 ,6,12,14-benzotetraazacyclononadecin-9-yl]methyl} (methyl)oxido^ 6 - sulfanylidene]carbamate (92 mg) that was contaminated by impurities and which was used without further purification. To a solution of crude (rac)-tert-butyl [{[16,20-difluoro-6-methyl-l,2,3,4,5,6-hexahydro-12H-17,13- (azeno)- 11 ,7-(metheno)- 1 ,6,12,14-benzotetraazacyclononadecin-9-yl]methyl} (methyl)oxido^ 6 - sulfanylidene]carbamate (130 mg) in dichloromethane (1.9 mL) was added trifluoroacetic acid (570 μΐ) and the mixture was stirred for 1.5 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate (5 mL) and the mixture was extracted with dichloromethane. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by preparative HPLC to yield the title compound (16 mg, 96% purity).

HPLC Purification:

Instrument: pump: Labomatic HD-5000, head HDK 280, low pressure gradient module ND-B1000; manual injection valve: Rheodyne 3725Ϊ038; detector: Knauer Azura UVD 2.15; collector: Labomatic Labocol Vario-4000; column: Chromatorex RP C-18 10 μιη, 125x30mm; solvent A: water + 0.1 vol-% formic acid, solvent B: acetonitrile; gradient: 0.00-0.50 min 30% B (150 ml/min), 0.50-6.00 min 30-70% B (150 ml/min), 6.00-6.10 min 70-100% B (150 ml/min), 6.10-8.00 min 100% B (150 ml/min); detection: UV 254 nm.

LC-MS (Method a): R t = 1.17 min; MS (ESIneg): m/z = 471 [M-H] "

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.53 - 1.69 (m, 4H), 2.78 - 2.84 (m, 3H), 2.85 - 2.88 (m, 3H), 3.19 - 3.27 (m, 2H), 3.43 - 3.54 (m, 1H), 4.10 - 4.27 (m, 2H), 6.21 - 6.31 (m, 1H), 6.33 - 6.51 (m, 3H), 6.60 - 6.71 (m, 1H), 7.12 - 7.24 (m, 1H), 7.67 - 7.77 (m, 1H), 8.11 - 8.21 (m, 1H), 8.52 - 8.65 (m, 1H), 9.46 - 9.54 (m, 1H).

Example 18:

2, 19-difluoro-9-[(methylsulfanyl)methyl]- 14,15,16,17-tetrahydro-6H, 13H-7, 11 -(azeno)-5, 1 -(metheno)- 12,4,6-benzoxadiazacyclononadecine

Preparation of Intermediate 18.1:

5-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenyl]pent-4-yn -l-ol

To a solution of 2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenyl trifluoromethanesulfonate (see Intermediate 4.1 ; 3.00 g) in DMF (56 mL) was sequentially added pent-4-yn-l-ol (855 mg), copper(I)iodide (323 mg), triethylamine (2.4 mL) and bis(triphenylphosphino)palladium(II) chloride (594 mg), and the mixture was stirred at 80 °C for 16 h. The reaction was allowed to cool to room temperature, filtered over a pad of Celite and concentrated. The residue was partioned between ethyl acetate (150 mL) and water (50 mL). The organic layer was consecutively washed with saturated aqueous sodium bicarbonate solution and saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate to ethyl acetate/methanol) to yield the title compound (841 mg, 96% purity).

LC-MS (Method a): R t = 0.84 min; MS (ESIpos): m/z = 289 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.46 - 1.57 (m, 2H), 2.28 - 2.38 (m, 2H), 3.33 - 3.37 (m, 2H), 4.39 - 4.49 (m, 1H), 5.86 - 5.93 (m, 2H), 6.36 - 6.45 (m, 1H), 7.24 - 7.33 (m, 1H), 7.35 - 7.44 (m, 2H), 7.88 - 7.93 (m, 1H) (propargylic protons are overlayed by residual DMSO).

Preparation of Intermediate 18.2:

5-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenyl]pentan-l- ol

To a solution of 5-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenyl]pent-4-yn -l-ol (575 mg) in THF (19 mL) were sequentially added acetic acid (few drops) and palladium 10% on activated carbon (106 mg). The reactor was purged with hydrogen gas (48.9 bar) and the mixture was stirred at 50 °C for 18 h. The mixture was cooled to room temperature, filtered (washing with THF) and concentrated to give the title compound (652 mg, 96% purity) that was used without further purification.

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm 1.12 - 1.22 (m, 2 H), 1.25 - 1.33 (m, 2 H), 1.37 - 1.48 (m, 2 H), 1.88 - 1.93 (m, 3 H), 2.14 - 2.22 (m, 1 H), 2.42 - 2.47 (m, 2 H), 3.26 - 3.31 (m, 2 H), 4.26 - 4.33 (m, 1 H), 5.89 - 6.00 (m, 2 H), 6.23 - 6.34 (m, 1 H), 7.07 - 7.14 (m, 1 H), 7.17 - 7.25 (m, 2 H), 7.89 - 7.93 (m, 1 H).

Preparation of Intermediate 18.3:

4- {2-[5-( {6-chloro-4-[(methylsulfanyl)methyl]pyridin-2-yl} oxy)pentyl]-4-fluorophenyl} -5- fluoropyridin-2-amine

To a solution of 5-[2-(2-amino-5-fluoropyridin-4-yl)-5-fluorophenyl]pentan-l- ol (424 mg) in THF (11 mL) at 0 °C was added sodium hydride (87.0 mg, 60% purity). The mixture was allowed to warm to room temperature and stirred for 30 min. Then, 2,6-dichloro-4-[(methylsulfanyl)methyl]pyridine (see Intermediate 1.7; 453 mg) was added and the mixture was heated to 90 °C for 2 h. An additional portion of 2,6-dichloro-4-[(methylsulfanyl)methyl]pyridine (227 mg) was added and the mixture was stirred at 90 °C for an additional 2.5 h. The mixture was allowed to cool to room temperature and concentrated. The residue was diluted with water (20 mL) and ethyl acetate (50 mL). The layers were separated and the aqueous layer was extracted three times with ethyl acetate (50 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude product was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (560 mg, 95%> purity).

LC-MS (Method a): R t = 1.48 min; MS (ESIpos): m/z = 464 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.23 - 1.34 (m, 2H), 1.44 - 1.53 (m, 2H), 1.56 - 1.65 (m, 2H), 1.94 (s, 3H), 3.63 (s, 2H), 4.10 - 4.18 (m, 2H), 5.88 - 5.97 (m, 2H), 6.27 - 6.35 (m, 1H), 6.70 - 6.74 (m, 1H), 6.97 - 7.02 (m, 1H), 7.06 - 7.14 (m, 1H), 7.18 - 7.27 (m, 2H), 7.85 - 7.96 (m, 1H) (one methylene group overlayed by residual DMSO). Example 18 - Preparation of the End Product:

A degassed suspension of 4- {2-[5-( {6-chloro-4-[(methylsulfanyl)methyl]pyridin-2-yl} oxy)pentyl]-4- fluorophenyl} -5-fluoropyridin-2-amine (510 mg), chloro(2-dicyclohexylphosphino-2',4',6'-tri-z ' o-propyl- l,r-biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-feri-butylether adduct (182 mg), 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (105 mg) and potassium phosphate (1.17 g) in toluene (100 mL) and N-methylpyrrolidone (10 mL) was heated at 130 °C for 18 h. The mixture was allowed to cool to room temperature, diluted with water (100 mL) and extracted three times with ethyl acetate (150 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The residue was treated with saturated aqueous sodium chloride solution (100 mL) and extracted three times with diethyl ether (150 mL each). The combined organic layers were concentrated and the residue was purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to give the crude title compound (310 mg, 80% purity) that was used without further purification. A pure sample was obtained by purification by preparative HPLC.

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.59 min; MS (ESIpos): m/z = 428 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm 1.08 - 1.27 (m, 2H), 1.72 - 1.83 (m, 4H), 1.98 (s, 3H), 2.57 - 2.68 (m, 2H), 3.54 (s, 2H), 3.86 - 4.1 1 (m, 2H), 6.05 - 6.19 (m, 1H), 6.47 - 6.58 (m, 1H), 7.08 - 7.19 (m, 1H), 7.21 - 7.33 (m, 2H), 7.85 - 8.10 (m, 1H), 8.25 - 8.43 (m, 1H), 9.65 - 9.81 (m, 1H).

Example 19:

(rac)-2, 19-difluoro-9- [(methylsulfinyl)methyl]- 14, 15, 16, 17-tetrahydro-6H, 13H-7, 1 1 -(azeno)-5, 1 - (metheno)- 12,4,6-benzoxadiazacyclononadecine

To a solution of 2,19-difluoro-9-[(methylsulfanyl)methyl]-14,15,16,17-tetrahy dro-6H,13H-7,l l-(azeno)- 5,l-(metheno)-12,4,6-benzoxadiazacyclononadecine (260 mg) in acetonitrile (19 ml) at 0 °C was added iron trichloride (9.9 mg) and the mixture was stirred for 10 min. Then, periodic acid (415 mg) was added and the mixture was stirred for 3 h at 0 °C. The reaction was stopped by the addition of saturated aqueous sodium thiosulfate solution (50 mL) and the mixture was stirred for 10 min before being extracted twice with ethyl acetate (75 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated to give the crude title compound (239 mg, 80% purity) that was used without further purification. A pure sample was obtained by further purification by preparative HPLC.

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol % formic acid (99%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 mL/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.28 min; MS (ESIpos): m/z = 444 [M+H] +

¾ NMR (400 MHz, DMSO-i/ 6 , 295 K) δ/ppm = 1.15 - 1.26 (m, 2H), 1.68 - 1.89 (m, 4H), 2.54 (s,3 H), 2.58 - 2.67 (m, 2H), 3.77 - 3.88 (m, 1H), 3.94 - 4.08 (m, 3H), 6.07 - 6.17 (m, 1H), 6.44 - 6.53 (m, 1H), 7.10 - 7.19 (m, 1H), 7.21 - 7.34 (m, 2H), 7.84 - 7.96 (m, 1H), 8.29 - 8.35 (m, 1H), 9.66 - 9.86 (m, 1H).

Example 20:

(rac)-2, 19-difluoro-9- [(S-methylsulfonimidoyl)methyl] -14,15,16,17-tetrahydro-6H, 13H-7, 11 -(azeno)- 5, 1 -(metheno)- 12,4,6-benzoxadiazacyclononadecine

To a vigorously stirred solution of crude (rac)-2,19-difluoro-9-[(methylsulfinyl)methyl]-14,15, 16,17- tetrahydro-6H,13H-7,l l-(azeno)-5,l -(metheno)- 12,4,6-benzoxadiazacyclononadecine (50.0 mg) in chloroform (770 μΐ,) at 0 °C was sequentially added sodium azide (58.6 mg) and concentrated aqueous sulfuric acid (120 μΐ) (Caution: the reaction was carefully set up using a blast shield). The mixture was stirred at room temperature for 18 h. The mixture was carefully diluted with a mixture of saturated aqueous sodium chloride solution and saturated aqueous sodium bicarbonate solution (1 : 1, 35 mL), and then extracted three times with dichloromethane/methanol (9: 1, 20 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. The crude material was combined with two additional reaction batches carried out as described above. Purification by preparative HPLC yielded the title compound (11.1 mg, 97% purity).

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Colum: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.2 vol % aqueous ammonia (32%), eluent B: acetonitrile; gradient: 0-5.5 min 5-100%) B; flow 70 ml/min; temperature: 25 °C; DAD scan: 210-400 nm.

LC-MS (Method b): R t = 1.24 min; MS (ESIpos): m/z = 459 [M+H] +

¾ NMR (400 MHz, DMSO-de, 295 K) δ/ppm = 1.05 - 1.31 (m, 2 H), 1.55 - 2.00 (m, 4 H), 2.57 - 2.68 (m, 2 H), 2.86 (s, 3 H), 3.66 - 3.79 (m, 1 H), 3.87 - 4.10 (m, 2 H), 4.18 - 4.33 (m, 2 H), 6.16 - 6.30 (m, 1 H), 6.52 - 6.64 (m, 1 H), 7.07 - 7.19 (m, 1 H), 7.22 - 7.34 (m, 2 H), 7.86 - 7.97 (m, 1 H), 8.33 (s, 1 H), 9.82 (s, 1 H).

Example 21:

(rac)-tert-butyl [ { [ 16,20-difluoro-2,3,4,5-tetrahydro- 12H- 17, 13-(azeno)- 11 ,7-(metheno)-6, 1,12,14- benzoxathiadiazacyclononadecin-9-yl]methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

Preparation of Intermediate 21.1:

(rac)-fert-butyl [(3- {4-[(2-bromo-5-fluoro henyl)sulfanyl]butoxy}-5-nitrobenzyl)(methyl)oxido-λ 6 - sulfanylidene]carbamate

To a solution of (rac)-fert-butyl {[3-(4-hydroxybutoxy)-5-nitrobenzyl](methyl)oxido^ 6 - sulfanylidene} carbamate (see Intermediate 15.4; 1.00 g), 2-bromo-5-fluorobenzenethiol (514 mg, purchased from Oakwood Chemicals) and triphenylphosphine (717 mg) in dichloromethane (10 mL) was added diisopropyl azodicarboxylate (540 μΐ.) and the reaction was stirred for 3 h at room temperature. The mixture was concentrated and purified by flash column chromatography (silica gel, hexanes/ethyl acetate) to yield the title compound (1.03 g, 90% purity).

LC-MS (Method a): R t = 1.51 min; MS (ESIpos): m/z = 591 [M+H] +

¾ NMR (400 MHz, DMSO-ifc, 22°C) δ/ppm = 1.38 (s, 9H), 1.75 - 1.85 (m, 2 H), 1.88 - 1.96 (m, 2H), 3.07 - 3.19 (m, 5H), 4.14 - 4.22 (m, 2H), 4.94 - 5.08 (m, 2H), 6.90 - 7.02 (m, 1H), 7.19 - 7.27 (m, 1H), 7.40 - 7.49 (m, 1H), 7.59 - 7.66 (m, 1H), 7.74 - 7.80 (m, 1H), 7.88 - 7.93 (m, 1H).

Preparation of Intermediate 21.2:

(rac)-fert-butyl {[3-(4- {[5-fluoro-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl] sulfanyl} butoxy)-5 -nitrobenzyl] (methyl)oxido^ 6 -sulfanylidene } carbamate

A suspension of (rac)-tert-butyl [(3- {4-[(2-bromo-5-fluorophenyl)sulfanyl]butoxy}-5- nitrobenzyl)(methyl)oxido-λ 6 -sulfanylidene]carbamate (1.90 g), 4,4,4',4', 5,5,5', 5'-octamethyl-2,2'-bi- 1,3,2-dioxaborolane (979 mg), potassium acetate (1.58 g) and dichlorobis(tricyclohexylphosphine)palladium(II) (119 mg) in 1,4-dioxane (66 mL) was heated at 110 °C for 7 h. The mixture was cooled to room temperature, filtered (the filter cake was washed with ethyl acetate) and concentrated. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate) yielded the title compound (820 mg) as a mixture with the corresponding boronic acid and some further impurities. Such material was used without further purification in the next step.

LC-MS (Method a): R t = 1.53 min; MS (ESIpos): m/z = 639 [M+H] +

Preparation of Intermediate 21.3:

(rac)-fert-butyl {[3-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenyl]sulfany l}butoxy)-5- nitrobenzyl](methyl)oxido^ 6 -sulfanylidene}carbamate

To a suspension of crude (rac)-tert-butyl {[3-(4- {[5-fluoro-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]sulfanyl}butoxy)-5-nitrobenzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (820 mg), 2,4- dichloro-5-fluoropyrimidine (295 mg, purchased from Apollo Scientific) and [1,1 '- bis(diphenylphosphino)ferrocene]dichloro palladium(II) complex with dichloromethane (120 mg) in 1,2- dimethoxyethane (3.8 mL) was added aqueous potassium carbonate solution (2.2 mL, 2.0 M) and the mixture was stirred at 90 °C for 3.5 h. The mixture was cooled to room temperature, diluted with water (100 mL) and extracted three times with ethyl acetate (50 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate) yielded the crude title compound (300 mg) that was used in the next step without further purification.

LC-MS (Method a): R t = 1.53 min; MS (ESIpos): m/z = 643 [M+H] + Preparation of Intermediate 21.4:

(rac)-fert-butyl {[3-amino-5-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenyl]sulfanyl}butoxy)benzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate

To a solution of crude (rac)-fert-butyl {[3-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenyl]sulfanyl}butoxy)-5-nitrobenzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (300 mg) in methanol (11 mL) and THF (1.1 mL) was added platinum 1% and vanadium 2% on activated carbon (45.5 mg). The mixture was purged with hydrogen gas (1 atm) and stirred for 1.5 h at room temperature. The mixture was filtered and concentrated to yield the crude title compound (347 mg) that was used without further purification.

LC-MS (Method a): R t = 1.39 min; MS (ESIpos): m/z = 614 [M+H] Example 21 - Preparation of the End Product:

A degassed suspension of crude (rac)-fert-butyl {[3-amino-5-(4- {[2-(2-chloro-5-fluoropyrimidin-4-yl)- 5-fluorophenyl]sulfanyl}butoxy)benzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (170 mg), chloro(2- dicyclohexylphosphino-2',4',6'-tri-z,yo-propyl-l, -biphenyl)[2-(2-arninoethyl)phenyl] palladium(II) methyl-fert-butylether adduct (45.9 mg), 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (26.4 mg) and potassium phosphate (294 mg) in toluene (17 mL) and N-methylpyrrolidone (1.7 rriL) was heated at 130 °C for 18 h. The mixture was allowed to cool to room temperature, diluted with water (50 mL) and extracted three times with ethyl acetate (20 mL each). The combined organic layers were washed with saturated aqueous sodium chloride solution, dried and concentrated. Purification by preparative HPLC yielded the title compound (9.2 mg, 97% purity).

HPLC Purification

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.2 Vol-% aqueous ammonia (32%), eluent B: acetonitrile; gradient: 0.00-0.50 min 44% B (25->70mL/min), 0.51-7.50 min 44-64% B (70mL/min), DAD scan: 210-400 nm

LC-MS (Method b): R t = 1.35 min; MS (ESIpos): m/z = 577 [M+H] +

'H NMR (400 MHz, DMSO-de, 295 K) 8/ppm = 1.35 - 1.42 (m, 9H), 1.74 - 1.90 (m, 4H), 3.08 - 3.13 (m, 3H), 3.16 - 3.26 (m, 2H), 3.98 - 4.09 (m, 2H), 4.66 - 4.73 (m, 2H), 6.46 - 6.57 (m, 1H), 6.71 - 6.77 (m, 1H), 7.06 - 7.15 (m, 1H), 7.35 - 7.47 (m, 2H), 8.14 - 8.23 (m, 1H), 8.69 - 8.78 (m, 1H), 9.93 - 9.97 (m, 1H).

Example 22:

(rac)- 16,20-difluoro-9- [(S-methylsulfonimidoyl)methyl]-2,3,4,5-tetrahydro- 12H- 17, 13-(azeno)- 1 1 ,7- (metheno)-6, 1 ,12, 14-benzoxathiadiazacyclononadecine

A solution of (rac)-fert-butyl [ {[16,20-difluoro-2,3,4,5-tetrahydro-12H-17, 13-(azeno)-l l ,7-(metheno)- 6, 1 ,12, 14-benzoxathiadiazacyclononadecin-9-yl]methyl} (methyl)oxido^ 6 -sulfanylidene]carbamate (8.00 mg) in dichloromethane (2.0 mL) was treated with trifluoroacetic acid (27 μ ) and the mixture was stirred at room temperature for 3.5 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of saturated aqueous sodium bicarbonate solution. The mixture was then extracted with dichloromethane. The combined organic layers were dried and concentrated. Purification by HPLC yielded the title compound (3.9 mg, 99% purity).

HPLC Purification:

Instrument: Waters Autopurification MS SingleQuad; Column: Waters XBrigde CI 8 5μ 100x30mm; eluent A: water + 0.1 vol-% formic acid (99%), eluent B: acetonitrile; gradient: 0.00-0.50 min 17% B (25->70mL/min), 0.51-5.50 min 33-53% B (70mL/min), DAD scan: 210-400 nm.

LC-MS (Method a): R t = 1.10 min; MS (ESIpos): m/z = 477 [M+H] +

'H NMR (400 MHz, DMSO-ifc, 295 K) 8/ppm = 1.74 - 1.94 (m, 4H), 2.72 - 2.86 (m, 3H), 3.12 - 3.26 (m, 2H), 3.50 - 3.59 (m, 1H), 3.98 - 4.15 (m, 2H), 4.15 - 4.28 (m, 2H), 6.41 - 6.58 (m, 1H), 6.62 - 6.85 (m, 1H), 7.02 - 7.20 (m, 1H), 7.34 - 7.50 (m, 2H), 8.07 - 8.25 (m, 1H), 8.61 - 8.77 (m, 1H), 9.78 - 9.95 (m, 1H). Example 23:

(rac)- 16,20-difluoro-6-methyl-9-[(S-methylsulfonimidoyl)methyl] -3,4,5, 6-tetrahydro-2H, 12H- 13,17- (azeno)- 11 ,7-(metheno)- 1 ,6,12,14-benzoxatriazac clononadecine

Trifluoroacetic acid (0.056 mL) was added to a stirred solution of (rac)-tert-butyl [{[16,20-difluoro-6- methyl-3,4,5,6-tetrahydro-2H,12H-13,17-(azeno)-l l,7-(metheno)-l,6,12,14- benzoxatriazacyclononadecin-9-yl]methyl} (methyl)oxido^ 6 -sulfanylidene]carbamate (see Example 6; 13 mg) in DCM (0.1 mL) and the mixture was stirred for 2 h at room temperature. The pH value of the reaction mixture was adjusted to pH9 by the addition of an aqueous solution of potassium carbonate (2M) and extracted three times with dichloromethane. The combined organic phase was filtered using a Whatman filter and concentrated to give the title compound (7 mg).

¾-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.61 - 1.74 (m, 2H), 1.81 (br d, 2H), 2.65-2.88 (m, 6H), 3.14 - 3.31 (m, 2H), 4.03 (d, 1H), 4.14 - 4.35 (m, 4H), 6.27 (s, 1H), 6.40 (s, 1H), 6.86 (td, 1H), 7.13 (dd, 1H), 7.36 (ddd, 1H), 7.68 (s, 1H), 8.61 (d, 1H), 9.52 (s, 1H). Example 24:

(rac)-tert-butyl [{[16,20-difluoro-3,4,5,6-tetrahydro-2H,12H-13,17-(azeno)-l l,7-(metheno)-l,6,12,14- benzoxatriazacyclononadecin-9-yl methyl}(methyl)oxido-λ 6 -sulfanylidene]carbamate

Preparation of Intermediate 24.1:

3-[(methylsulfanyl)methyl]-5-nitroaniline

Thionyl chloride (12.9 mL) was added dropwise to a stirred suspension of (3-amino-5- nitrophenyl)methanol (4.95 g) in DCM (96 mL) at room temperature and the mixture was stirred under under reflux for 18 h. After cooling the mixture was concentrated and the residue was dissolved in acetone (440 mL). An aqueous solution of sodium thiomethoxide (21%, 39 mL) was added under stirring at 0°C and the mixture was stirred at room temperature for 72 h. The mixture was diluted with ethyl acetate and washed with an aqueous solution of sodium chloride. The organic phase was filtered using a Whatman filter and concentrated. The residue was purified by flash chromatography on silica gel (hexane to hexane/ethyl acetate 40%) to give the title compound (4.85 g).

'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.96 (s, 3H), 3.66 (s, 2H), 5.83 (s, 2H), 6.89 (t, 1H), 7.26 (s, 2H). Preparation of Intermediate 24.2:

N- {3-[(methylsulfanyl)methyl]-5-nitrophenyl} -2-nitrobenzenesulfonamide

Pyridine (0.49 mL) was added to a stirred suspension of 3-[(methylsulfanyl)methyl]-5-nitroaniline (1.00 g) and 2-nitrobenzenesulfonyl chloride (1.34 g) in dichloromethane (6.1 mL) at room temperature. The mixture was stirred overnight. An aqueous solution of hydrogen chloride (IN, 8 mL) was added before the mixture was extracted three times with ethyl acetate. The combined organic phase was washed with an aqueous solution of sodium chloride, filtered using a Whatman filter and concentrated. The residue was purified by column chromatography on silica gel (hexane to hexane/ ethyl acetate 50%) to give the title compound (1.80 g,).

'H-NMR (400 MHz, DMSO-ifc, 295 K) 8/ppm = 1.82 (s, 3H), 3.77 (s, 2H), 7.50 (t, 1H), 7.79 - 7.91 (m, 4H), 7.95 - 8.09 (m, 2H), 11.35 (br s, 1H).

Preparation of Intermediate 24.3:

4-[2-(4- {[tert-butyl(dimethyl)silyl]oxy}butoxy)-4-fluorophenyl]-2-ch loro-5-fluoropyrimidine

Diisopropyl azodicarboxylate (9.84 mL) was added dropwise to a stirred mixture of 2-(2-chloro-5- fluoropyrimidin-4-yl)-5-fluorophenol (6.00 g), 4- {[teri-butyl(dimethyl)silyl]oxy}butan-l -ol (10.11 g) and triphenylphosphine (13.49 g) in THF at 0 °C and the mixture was stirred overnight at room temperature. The mixture was concentrated and the residue was purified by column chromatography on silica gel (hexane to hexane / ethyl acetate 20%) to give the desired title compound (11.1 g; 25.9 mmol), still containing some impurities.

'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = -0.02 (s, 6H), 0.77 - 0.92 (m, 9H), 1.39 - 1.57 (m, 2H), 1.57 - 1.76 (m, 2H), 3.55 (s, 2H), 4.08 (s, 2H), 6.82 - 7.06 (m, 1H), 7.06 - 7.21 (m, 1H), 7.46 - 7.63 (m, 1H), 8.91 (d, 1H).

Preparation of Intermediate 24.4:

4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy]butan -l-ol

A mixture of 4-[2-(4- {[teri-butyl(dimethyl)silyl]oxy}butoxy)-4-fluorophenyl]-2-ch loro-5- fluoropyrimidine (11.1 g) in THF (666 mL) and an aqueous solution of hydrogen chloride (2M, 327 mL) was stirred at room temperature for 3 h. The pH value of the reaction mixture was adjusted to pH>7 by the addition of solid sodium bicarbonate. Solid sodium chloride was added and the mixture was extracted three times with ethyl acetate. The combined organic phase were dried and concentrated to give the crude product, that was used without further purification (9.86 g).

'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.37 - 1.52 (m, 2H), 1.59 - 1.75 (m, 2H), 3.35 - 3.45 (m, 2H), 3.95 - 4.16 (m, 2H), 4.32 - 4.51 (m, 1H), 6.87 - 7.02 (m, 1H), 7.09 - 7.21 (m, 1H), 7.54 (dd, 1H), 8.93 (d, 1H). Preparation of Intermediate 24.5:

N- {4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy]buty l} -N- {3-[(methylsulfanyl)methyl]-5- nitrophenyl} -2-nitrobenzenesulfonamide

Diisopropyl azodicarboxylate (0.74 mL) was added dropwise to a stirred mixture of 4-[2-(2-chloro-5- fluoropyrimidin-4-yl)-5-fluorophenoxy]butan-l-ol (591 mg; see Intermediate 1.3), N- {3- [(methylsulfanyl)methyl]-5-nitrophenyl}-2-nitrobenzenesulfon amide (720 mg) and triphenylphosphine (985 mg) in DCM (29 mL) at 0°C and the mixture was stirred for 6 h at room temperature. The mixture was concentrated and the residue was purified by preparative HPLC (basic conditions) to give the title compound (418 mg).

'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.39 - 1.53 (m, 2H), 1.62 - 1.71 (m, 2H), 1.81 (s, 3H), 3.76 - 3.85 (m, 4H), 4.05 (t, 2H), 6.96 (td, IH), 7.10 (dd, IH), 7.51 (dd, IH), 7.59 (t, IH), 7.77 - 7.81 (m, 2H), 7.86 - 7.97 (m, 3H), 8.21 (t, IH), 8.76 (d, IH).

Preparation of Intermediate 24.6:

(rac)-N- {4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy]buty l} -N- {3- [(methylsulfinyl)methyl]-5-nitrophenyl} -2-nitrobenzenesulfonamide

To a stirred solution of N- {4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy]buty l}-N- {3- [(methylsulfanyl)methyl]-5-nitrophenyl}-2-nitrobenzenesulfon amide (418 mg) in MeCN (16.4 mL) at 0 °C was added iron(III) chloride (2.9 mg) and the mixture was stirred for 10 minutes at room temperature. Then, periodic acid (150 mg) was added and the mixture was stirred for 2 h while cooling with an ice bath. The reaction was stopped by pouring onto ice and saturated aqueous sodium thiosulfate (30 mL) was added. The mixture was stirred for 10 min and the product was extracted twice with ethyl acetate / THF 1 : 1; (50 mL each). The combined organic layers were filtered using a Whatman filter and concentrated. The residue was purified by column chromatography (DCM to DCM/EtOH 50%) to yield the title compound (220 mg).

'H-NMR (400MHZ, DMSO-i 6 , 295 K) 8/ppm = 1.39 - 1.51 (m, 2H), 1.62 - 1.71 (m, 2H), 2.34 (s, 3H), 3.82 (br t, 2H), 4.01 - 4.11 (m, 3H), 4.30 (d, 1H), 6.96 (td, 1H), 7.10 (dd, 1H), 7.12 (d, 1H), 7.51 (dd, 1H), 7.66 (t, 1H), 7.75 - 7.82 (m, 2H), 7.89 - 8.00 (m, 3H), 8.20 (d, 1H), 8.20 (s, 1H), 8.78 (d, 1H).

Preparation of Intermediate 24.7:

(rac)-tert-butyl {[3-({4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy ]butyl}amino)-5- nitrobenzyl](methyl)oxido^ 6 -sulfanylidene}carbamate

To a suspension of (rac)-N- {4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy]buty l}-N- {3- [(methylsulfinyl)methyl]-5-nitrophenyl} -2-nitrobenzenesulfonamide (420 mg), tert-butyl carbamate (106 mg), magnesium oxide (97 mg) and rhodium(II) acetate dimer (7 mg) in DCM (21 mL) was added iodobenzene diacetate (292 mg) at room temperature under an atmosphere of argon. The mixture was stirred for 18 h at 45°C. The reaction mixture was concentrated to a volume of about 3 mL and the mixture was then purified by column chromatography on silica gel (hexane/ethyl acetate 20% to 70%>) to give the desired title compound (396 mg).

¾-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.37 (s, 9H), 1.40 - 1.53 (m, 2H), 1.61 - 1.71 (m, 2H), 3.03 (s, 3H), 3.76 - 3.88 (m, 2H), 4.00 - 4.07 (m, 2H), 4.98 - 5.09 (m, 2H), 6.96 (td, 1H), 7.10 (dd, 1H), 7.51 (dd, 1H), 7.76 - 7.84 (m, 3H), 7.89 - 7.98 (m, 2H), 8.06 (t, 1H), 8.31 (t, 1H), 8.77 (d, 1H). Preparation of Intermediate 24.8:

(rac)-fert-butyl {[3-({4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5-fluorophenoxy ]butyl}

nitrobenzyl](methyl)oxido-λ 6 -sulfan lidene}carbamate

A suspension of (rac)-tert-butyl {[3-({4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenoxy]butyl}amino)-5-nitrobenzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (200 mg) and cesium carbonate (161 mg) in DMF (4.7 mL) was stirred at room temperature for 2 minutes before thiophenol (33 mg) was added. The mixture was stirred at room temperature for 4 h. Then it was diluted with water and extracted three times with ethyl acetate. The combined organic phase was washed with aqueous sodium chloride solution, dried and concentrated. The residue was purified by preperative HPLC (Autopurifier; acidic conditions) to give the title compound (90 mg), still containing slight impurities.

Preparation of Intermediate 24.9:

(rac)-fert-butyl {[3-amino-5-({4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenoxy]butyl}amino)benzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate

Platinum 1% and vanadium 2%, on activated carbon (50-70% wetted powder, 21 mg) was added to a solution of (rac)-fert-butyl {[3-({4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenoxy] butyl} amino)-5-nitrobenzyl](methyl)oxido^ 6 -sulfanylidene} carbamate(80 mg) in methanol (21 mL) and the mixture was stirred for 1 h at room temperature under a hydrogen atmosphere. The mixture was filtered and the filtrate was concentrated. The residue was purified by preparative HPLC (Autopurifier, acidic conditions) to give the title compound (14 mg).

'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.39 (m, 9H), 1.50 - 1.63 (m, 2H), 1.67 - 1.77 (m, 2H), 2.97 (br t, 2H), 3.11 (s, 3H), 4.10 (t, 2H), 4.58 - 4.67 (m, 2H), 6.14 - 6.34 (m, 3H), 6.98 (td, 1H), 7.09 - 7.23 (m, 1H), 7.54 (dd, 1H), 8.87 (d, 1H).

Example 24 - Preparation of end product:

A mixture of (rac)-tert-butyl {[3-amino-5-({4-[2-(2-chloro-5-fluoropyrimidin-4-yl)-5- fluorophenoxy]butyl}amino)benzyl](methyl)oxido-λ 6 -sulfanylidene}carbamate (14.0 mg), chloro(2- dicyclohexylphosphino-2',4',6'-tri-wo-propyl-l, -biphenyl)[2-(2-aminoethyl)phenyl] palladium(II) methyl-tert-butylether adduct (1.9 mg; purchased from ABCR GmbH & CO. KG) and 2- (dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl (1.1 mg; purchased from Aldrich Chemical Company Inc.) and potassium phosphate (24.9 mg) in toluene (1.8 mL) and NMP (0.2 mL) was stirred under an atmosphere of argon at 110°C for 3 h. After cooling, the batch was diluted with with aqueous sodium chloride solution and extracted twice with ethyl acetate. The combined organic phase was dried and concentrated. The residue was purified by preparative HPLC (Autopurifier; basic conditions) to give the desired title compound (6 mg). 'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.37 (s, 9H), 1.66 (br d, 2H), 1.80 (br s, 2H), 3.02 - 3.14 (m, 5H), 4.25 (br t, 2H), 4.52 - 4.59 (m, 2H), 5.96 (t, 1H), 6.19 (s, 1H), 6.29 (s, 1H), 6.86 (td, 1H), 7.12 (dd, 1H), 7.32 - 7.38 (m, 1H), 7.48 (s, 1H), 8.61 (d, 1H), 9.50 (s, 1H).

Example 25:

(rac) 6,20-difluoro-9-[(S-methylsulfonimidoyl)methyl]-3,4,5,6 etrahydro-2H,12H-13,17-(azeno)-l l,7- (metheno)- 1 ,6,12,14-benzoxatriazacyclononadecine

Trifluoroacetic acid (0.03 mL) was added to a stirred solution of (rac)-tert-butyl [ {[16,20-difluoro- 3,4,5, 6-tetrahydro-2H, 12H- 13, 17-(azeno)- 1 1 ,7-(metheno)- 1 ,6, 12, 14-benzoxatriazacyclononadecin-9- yl]methyl} (methyl)oxido-λ 6 -sulfanylidene]carbamate (6 mg) in DCM (0.1 mL) and the mixture was stirred for 2 h at room temperature. The mixture was diluted with aqueous sodium bicarbonate solution and extracted three times with ethyl acetate/THF (1 : 1). The combined organic phase was dried and concentrated. The residue was purified by preparative HPLC (Autopurifier; basic conditions) to give the desired product (4 mg).

'H-NMR (400 MHz, DMSO-ifc, 295 K) δ/ppm = 1.67 (br s, 2H), 1.77 - 1.84 (m, 2H), 2.81 (s, 3H), 3.01 - 3.08 (m, 2H), 3.42 (s, 1H), 4.08 (s, 1H), 4.13 (s, 1H), 4.25 (br t, 2H), 5.91 (t, 1H), 6.18 (s, 1H), 6.27 (s, 1H), 6.86 (td, 1H), 7.12 (dd, 1H), 7.35 (t, 1H), 7.44 (s, 1H), 8.60 (d, 1H), 9.45 (s, 1H).

The following Table 1 provides an overview on the compounds described in the example section:

Table 1

Example No. Structure Name of compound

{rac)- 16,20-difluoro-9- [(S- methylsulfonimidoyl)methyl]-3,4,5,6- tetrahydro-2H, 12H- 1 1 ,7-(azeno)- 13, 17- (metheno)-l ,6,12, 14- benzoxatriazacyclononadecine

2, 18-difluoro-9-[(methylsulfanyl)methyl]- 13, 14,15, 16-tetrahydro-6H-7, 1 1 -(azeno)-5, 1 - (metheno)- 12,4,6- benzoxadiazacyclooctadecine

(rac)-2, 18-difluoro-9- [(methylsulfmyl)methyl] -13, 14, 15, 16- tetrahydro-6H-7, 1 1 -(azeno)-5, 1 -(metheno)- 12,4,6-benzoxadiazacyclooctadecine Example No. Structure Name of compound

8,16,20-trifluoro-6-methyl-9- [(methylsulfanyl)methyl]-3,4,5,6-tetrahydro-

12 2H, 12H- 11 ,7-(azeno)- 13, 17-(metheno)- 1,6,12,14-benzoxatriazacyclononadecine

(rac)-tert-butyl [{[15,19-difluoro- 1 ,2,3,4- tetrahydro- 16,12-(azeno)-6, 10-(metheno)-

13 5,1,11, 13-benzoxatriazacyclooctadecin- 8(l lH)-yl]methyl}(methyl)oxido^ 6 - sulfanylidene]carbamate

(rac)-15,19-difluoro-8-[(S- methylsulfonimidoyl)methyl] -1,2,3,4-

14 tetrahydro- 16,12-(azeno)-6, 1 O-(metheno)- 5,1,11, 13-benzoxatriazacyclooctadecine

Results:

Table 2: Inhibition for CDK9 and CDK2 of compounds according to the present invention

The IC50 (inhibitory concentration at 50% of maximal effect) values are indicated in nM, "n.t." means that the compounds have not been tested in the respective assay.

Φ : Example Number

©: CDK9: CDK9/CycTl kinase assay as described under Method la. of Materials and Methods

© : CDK2: CDK2/CycE kinase assay as described under Method 2. of Materials and Methods ©: Selectivity CDK9 over CDK2: IC50 (CDK2) / IC50 (CDK9) according to Methods la. and 2a. of Materials and Methods

©: high ATP CDK9: CDK9/CycTl kinase assay as described under Method lb. of Materials and Methods

©: high ATP CDK2: CDK2/CycE kinase assay as described under Method 2b. of Materials and Methods

®: Selectivity high ATP CDK9 over high ATP CDK2: IC50 (high ATP CDK2) / IC50 (high ATP CDK9) according to Methods lb. and 2b. of Materials and Methods Noteworthily, in the CDK9 assays, as described supra in the Methods la. and lb. of Materials and Methods, resolution power is limited by the enzyme concentrations, the lower limit for IC50S is about 1 -2 nM in the CDK9 high ATP assay and 2-4 nM in the CDK low ATP assays. For compounds exhibiting IC50S in this range the true affinity to CDK9 and thus the selectivity for CDK9 over CDK2 might be even higher, i.e. for these compounds the selectivity factors calculated in columns 4 and 7 of Table 2, infra, are minimal values, they could be also higher.

Table 2



240

Tables 3a and 3b: Inhibition of proliferation of HeLa, HeLa-MaTu-ADR, NCI-H460, DU145, Caco-2,

B16F10, A2780 and MOLM-13 cells by compounds according to the present invention, determined as described under Method 3. of Materials and Methods. All IC50 (inhibitory concentration at 50% of maximal effect) values are indicated in nM, "n.t." means that the compounds have not been tested in the respective assay.

Φ : Example Number

© : Inhibition of HeLa cell proliferation

© : Inhibition of HeLa-MaTu-ADR cell proliferation

© : Inhibition of NCI-H460 cell proliferation

© : Inhibition of DU 145 cell proliferation

© : Inhibition of Caco-2 cell proliferation

© : Inhibition of B 16F 10 cell proliferation

®: Inhibition of A2780 cell proliferation

® : Inhibition of MOLM- 13 cell proliferation Table 3a: Indications represented by cell lines

Table 3b: Inhibition of proliferation

 Table 4a: Equilibrium dissociation constants KD [1/s], dissociation rate constants kofr [1/s], and target resident times [min] as determined by Method 8. at 25°C as described in Materials and Methods. Slight variations of experimental parameters are indicated by letters (A-G): Parameters A: Described in Materials and Methods section 8.

Parameters B: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1200 s, Serial dilutions of compound (3.13 nM up to 100 nM)

Parameters C: Flow rate: 50 μΐ/min, Injection time: 60 s, Dissociation time: 1200 s, Serial dilutions of compound (0.82 nM up to 200 nM)

Parameters D: Flow rate: 100 μΐ/min, Injection time: 80 s, Dissociation time: 1200 s, Serial dilutions of compound (3.13 nM up to 100 nM)

Parameters E: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1100 s, Serial dilutions of compound (0.78 nM up to 25 nM) and measured at 37°C

Parameters F: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1100 s, Serial dilutions of compound (1.56 nM up to 50 nM)

Parameters G: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1100 s, Serial dilutions of compound (3.13 nM up to 100 nM)

Dissociation rate constants below of what is resolvable with the respective assay are reported using the "<"-symbol (e.g. < 2.5 E-5 s 1 )

Values labeled with "*" represent arithmetic means of more than one value.

Φ : Example Number

© : Equilibrium dissociation constant K D [ 1 /s]

© : Dissociation rate constant k of r [1/s]

© : Target resident time [min]

© : Experimental parameters as specified above [A-G]

Table 4a:

 Table 4b: Equilibrium dissociation constants KD [1/s], dissociation rate constants k 0 ff [1/s], and target resident times [min] as determined by Method 8. at 37°C as described in Materials and Methods. Slight variations of experimental parameters are indicated by letters (A-G):

Parameters A: Described in Materials and Methods section 8.

Parameters B: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1200 s, Serial dilutions of compound (3.13 nM up to 100 nM)

Parameters C: Flow rate: 50 μΐ/min, Injection time: 60 s, Dissociation time: 1200 s, Serial dilutions of compound (0.82 nM up to 200 nM)

Parameters D: Flow rate: 100 μΐ/min, Injection time: 80 s, Dissociation time: 1200 s, Serial dilutions of compound (3.13 nM up to 100 nM)

Parameters E: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1100 s, Serial dilutions of compound (0.78 nM up to 25 nM) and measured at 37°C

Parameters F: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1100 s, Serial dilutions of compound (1.56 nM up to 50 nM)

Parameters G: Flow rate: 100 μΐ/min, Injection time: 70 s, Dissociation time: 1100 s, Serial dilutions of compound (3.13 nM up to 100 nM)

Dissociation rate constants below of what is resolvable with the respective assay are reported using the "<"-symbol (e.g. < 8.0 E-5 s 1 ).

Values labeled with "*" represent arithmetic means of more than one value.

© : Example Number

© : Equilibrium dissociation constant K D [ 1 /s]

© : Dissociation rate constant koff [1/s]

© : Target resident time [min]

© : Experimental parameters as specified above [A-G]

It is expected that that the prolonged residence time of macrocyclic CDK9 inhibitors according to the invention will result in a sustained inhibitory effect on CDK9 signaling, ultimately contributing to sustained target engagement and anti-tumor efficacy. Table 4b:

Structure © © ©

1.10E-9* 9.54E-4* 17.5* E

5.84 E-10* 1.13 E-3* 14.7* E