Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
REVERSIBLE MACROCYCLIC KINASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2023/110936
Kind Code:
A1
Abstract:
The present invention relates to macrocyclic compounds and compositions containing said compounds acting as kinase inhibitors. Moreover, the present invention provides processes for the preparation of the disclosed compounds, as well as methods of using them, for instance as a medicament, in particular for the treatment specific kinase mediated disorders, such as cancer.

Inventors:
DE MAN ADRIANUS PETRUS ANTONIUS (NL)
BUIJSMAN ROGIER CHRISTIAN (NL)
STERRENBURG JAN GERARD (NL)
DE WIT JOERI JOHANNES PETRUS (NL)
VAN CAUTER FREEK (NL)
PRINSEN MARTINE BERENDINA WILHEMINA (NL)
MULLER MICHELLE (NL)
VAN MIL YVONNE GERTRUDA THEODORA HENDRIKA (NL)
Application Number:
PCT/EP2022/085713
Publication Date:
June 22, 2023
Filing Date:
December 13, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NETHERLANDS TRANSLATIONAL RES CENTER HOLDING B V (NL)
International Classes:
C07D471/04; A61K31/4353; A61K31/4985; A61P35/00; C07D487/04
Domestic Patent References:
WO2016210165A12016-12-29
WO2013010380A12013-01-24
WO2016106627A12016-07-07
WO2013010380A12013-01-24
WO2016210165A12016-12-29
WO2015058084A12015-04-23
WO2015095099A12015-06-25
WO2015095102A12015-06-25
WO2013081016A12013-06-06
WO2017106429A22017-06-22
WO2019091441A12019-05-16
WO2014082598A12014-06-05
WO2014025976A12014-02-13
WO2019236631A12019-12-12
WO2010011538A12010-01-28
WO2013010380A12013-01-24
Other References:
ATTWOOD M.M ET AL.: "Trends in kinase drug discovery targets, indications and inhibitor design", NAT REV DRUG DISCOV, vol. 20, 2021, pages 839 - 861, XP037602806, DOI: 10.1038/s41573-021-00252-y
COHEN ET AL.: "Kinase drug discovery 20 years after imatinib: progress and future directions", NATURE REVIEWS DRUG DISCOVERY, vol. 20, 2021, pages 551 - 569, XP037497182, DOI: 10.1038/s41573-021-00195-4
WILLEMSEN-SEEGERS N. ET AL., J MOL BIOL, vol. 429, no. 2017, 2016, pages 574 - 586
COPELAND R.A ET AL., NAT REV DRUG DISCOV, vol. 5, 2006, pages 730
BARF TKAPTEIN A, J MED CHEM, vol. 55, no. 14, 2012, pages 6243 - 62
DRIGGERS E.M., NAT REV DRUG DISCOV, 2008
MALLINSON J.M., COLLINS I., FUTURE MED CHEM, 2012
BOGA S B ET AL., BIOORG MED CHEM LETT, vol. 27, 2017, pages 3939 - 3943
LIU J ET AL., ACS MED CHEM LETT, vol. 6, 2016, pages 198 - 203
YIFAN FENGWEIMING DUANXIAOCHUAN CUCHENGYUAN LIANGMINHANG XIN: "Bruton's tyrosine kinase (BTK) inhibitors in treating cancer: a patent review", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 29, no. 4, 2010, pages 217 - 241
"Pharmaceutical Sciences", 2000, MEADE PUBLISHING CO
STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
"Cancer Principles and Practice of Oncology", 15 February 2001
BIOORG. MED. CHEM., vol. 23, no. 4, 2015, pages 564 - 578
J. MED. CHEM., vol. 60, no. 7, 2017, pages 2944 - 2962
ACS MED. CHEM. LETTERS, vol. 12, 2021, pages 1912 - 1919
ACS MED. CHEM. LETTERS, vol. 4, no. 7, 2013, pages 627 - 631
ORG. LETT., vol. 22, 2020, pages 2991 - 2994
XIANHUI WANG ET AL.: "Bruton's Tyrosine Kinase and Its Isoforms in Cancer", FRONT. CELL DEV. BIOL, vol. 9, 2021, pages 668996
KOKABEE L ET AL.: "Bruton's Tyrosine kinase is a potential therapeutic target in prostate cancer cells", CANCER BIOLOGY & THERAPY, vol. 16, no. 11, 2015, pages 1604 - 1615
WANG ET AL.: "Bruton's Tyrosine Kinase inhibitors prevent therapeutic escape in breast cancer cells", MOL CANCER THER, vol. 15, no. 9, 2016, pages 2198 - 2208, XP055345697, DOI: 10.1158/1535-7163.MCT-15-0813
WEI ET AL., ONCOTARGET, vol. 7, no. 43, 2016, pages 69961 - 69975
WANG ET AL., J EXP CLIN CANCER RES, vol. 36, 2017, pages 96
STEFAN F.H. NEYS ET AL.: "Targeting Bruton's Tyrosine Kinase in inflammatory and autoimmune pathalogies", FRONT CELL DEV BIOL, vol. 9, 2021, pages 668131
RULA ZAIN: "Structure-Function relationships of covalent and non-covalent BTK inhibitors", FRONT IMMUNOL, vol. 12, 2021, pages 694853
WANG ET AL.: "Noncovalent inhibitors reveal BTK gatekeeper and auto-inhibitory residues that control its transforming activity", JCI INSIGHT, vol. 4, no. 12, 2019, pages e127566
DRIGGERS, EM ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 7, no. 7, 2008, pages 608 - 624
MALLINSON J ET AL.: "Macrocycles in new drug discovery", FUTURE MED. CHEM, vol. 4, no. 11, 2012, pages 1409 - 1438, XP055071666, DOI: 10.4155/fmc.12.93
P.J. TUMMINO, R.A: "Copeland, Residence time of receptor-ligand complexes and its effect on biological function", BIOCHEMISTRY, vol. 47, 2008, pages 5481 - 5492, XP055375021, DOI: 10.1021/bi8002023
Attorney, Agent or Firm:
NEDERLANDSCH OCTROOIBUREAU (NL)
Download PDF:
Claims:
CLAIMS 1. A compound of Formula (I-a) to (I-h) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is selected from the group consisting of: wherein R1 is wherein : W is a direct bond or an aryl group having 6-10 carbon or a heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; V is selected from the group consisting of: a direct bond, O, -OCH2- , -CH(R1v)-, -C(O)-, -C(O)-N(R2v)-, -N(R2v)-C(O)-,-NH-C(O)-NH-, -NH-C(O)-C(R3v)(R4v)-C(O)-NH-, -NH-SO2-, -NH-C(O)-O-, -CH(R1v)-NH-C(O)-, -CH(R1v)-C(O)-NH-, -C≡C-, and -CH2O- ; R1v is hydrogen or (1-2C)alkyl; R2v is hydrogen or (1-2C)alkyl; R3v is hydrogen or (1-2C)alkyl; R4v is hydrogen or (1-2C)alkyl; or R3v and R4v form together with the carbon atom they are attached to a (3-6C)cycloalkyl; U is an aryl group having 6-10 carbon, a heteroaryl group having 1-9 carbon or a cycloalkyl group having 3-6 carbon; wherein any of said aryl group, heteroaryl group and cycloalkyl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1-6C)alkoxy, (3-6C)cycloalkyl or (3- 6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; Wherein R2 is of Formula (IIa) to (IIf) selected from the group consisting of: , ; wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X1, X2 and X3 are independently selected from CH2, -CH2CH2-, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with one or more substituents selected from halogen, hydroxy, (1-3C)alkyl, (1-3C)alkoxy, (1- 4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R3 and R4 together represent a linker having Formula (III-1 to III-40) selected from the group consisting of:

whereby the marks the position of R3 in any one of Formula I-a to I-h, and whereby the marks the position of R4 in any one of Formula II-a to II-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CD3, (1-4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl, (3-6C)cycloalkoxy or (1-6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein R5 is hydrogen, NH2 or Methyl.

2. Compound according to claim 1, wherein the linker represented by R3 and R4 is selected from the group consisting of: whereby the marks the position of R3 in any one of Formula I-a to I-h, and whereby the marks the position of R4 in any one of Formula II-a to II-f; wherein any of said linker is optionally and independently substituted with one or more groups selected from deuterium, halogen, oxo, hydroxy, CD3, (1-4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl, (3-6C)cycloalkoxy or (1-6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 3. Compound according to any one of the preceding claims, wherein the linker represented by R3 and R4 is selected from the group consisting of:

whereby the marks the position of R3 in any one of Formula I-a to I-h, and whereby the marks the position of R4 in any one of Formula II-a to II-f; wherein any of said linker is optionally and independently substituted with one or more substituents selected from deuterium, hydroxy, CD3, (1-4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3- 6C)cycloalkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 4. Compound according to any one of the preceding claims, wherein the linker represented by R3 and R4 is selected from the group consisting of:

whereby the marks the position of R3 in any one of Formula I-a to I-h, and whereby the marks the position of R4 in any one of Formula II-a to II-f; wherein any of said linker is optionally and independently substituted with one or more substituents selected from deuterium, hydroxy, CD3, (1-2C)alkyl, (1-2C)alkoxy, (3-6C)cycloalkyl or (3- 6C)cycloalkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 5. Compound according to any one of the preceding claims, wherein R2 is selected from the group consisting of: wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X1, X2 and X3 are independently selected from CH2, -CH2CH2-, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with halogen, hydroxy, (1-3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 6. Compound according to any one of the preceding claims, wherein R2 is selected from the group consisting of:

wherein Q is a monocyclic ring selected from a (3-7)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X1, X2 and X3 are independently selected from CH2, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with halogen, hydroxy, (1-3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 7. Compound according to any one of the preceding claims, wherein R2 is selected from the group consisting of: wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with hydroxy, methyl or methoxy. 8. Compound according to any one of the preceding claims, wherein the compound comprises a bicyclic scaffold selected from:

wherein R5 is hydrogen, NH2 or Methyl. 9. Compound according to any one of the preceding claims, wherein the compound comprises a bicyclic scaffold selected from: . 10. Compound according to any one of the preceding claims 1 - 9, wherein R1 is: W is an aryl group having 6-10 carbon or a heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and V is a direct bond. 11. Compound according to any one of the preceding claims 1 - 10, wherein R1 is: the phenyl group is optionally and independently substituted with one or more substituents selected from halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and U is an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1- 6C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 12. Compound according to any one of the preceding claims 1 – 9 and 11, wherein V is any one of: - OCH2- , -C(O)-N(R2v)-, -N(R2v)-C(O)-,-NH-C(O)-NH-, -NH-C(O)-C(R3v)(R4v)-C(O)-NH-, -NH-SO2-, -NH-C(O)-O-, -CH(R1v)-NH-C(O)-, -CH(R1v)-C(O)-NH- ; R1v is hydrogen or (1-2C)alkyl; R2v is hydrogen or (1-2C)alkyl; R3v is hydrogen or (1-2C)alkyl; R4v is hydrogen or (1-2C)alkyl; or R3v and R4v form together with the carbon atom they are attached to a (3-6C)cycloalkyl. 13. Compound according to any one of the preceding claims 1 – 9 and 11 - 12, wherein R1 is any one of: wherein: the phenyl group is optionally and independently substituted with one or more substituents selected from halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and U is an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon, wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1- 6C)alkoxy, (3-6C)cycloalkyl and (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 14. Compound according to any one of the preceding claims 1 - 9, wherein R1 is: , wherein: R1w is selected from: hydrogen, halogen, (1-2C)alkyl, (1-2C)alkoxy), (3-6C)cycloalkyl, (6-10C)aryl, and (1-5C)heteroaryl; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1-2C)alkyl, (1- 2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and V is selected from the group consisting of: a direct bond, -CH(R1v)-, -CH(R1v)-NH-C(O)-, -CH2O- ; R1v is hydrogen or (1-2C)alkyl. 15. Compound according to any one of the preceding claims 1 – 9 and 14, wherein R1 is: , wherein : wherein R1w is selected from hydrogen, halogen, (1-2C)alkyl, (1-2C)alkoxy, (3-6C)cycloalkyl, (6- 10C)aryl, and (1-5C)heteroaryl; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1-2C)alkyl, (1- 2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and V is a direct bond. 16. Compound according to any one of the preceding claims 1 - 9, wherein R1 is: , wherein: W is a direct bond; and U is hydrogen or an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1- 6C)alkoxy, (3-6C)cycloalkyl and (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 17. Compound according to any one of the preceding claims 1 - 9, wherein R1 is: , wherein : W is a direct bond, and U is hydrogen or an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1- 6C)alkoxy, (3-6C)cycloalkyl and (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 18. Compound according to any one of the preceding claims 1 - 9, wherein R1 is selected from the group consisting of: ; wherein: R1w and R2w are independently selected from hydrogen, halogen, (1-2C)alkyl, and (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R1v)-NH-C(O)-, -CH(R1v)- ; R1v is hydrogen or (1-2C)alkyl; and U is an aryl group having 6-10 carbon or an heteroaryl group having 1-5 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (1-4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl and (3- 6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. 19. Compound according to any one of the preceding claims 1 – 9 and 18, wherein R1 is selected from the group consisting of: wherein: R1w and R2w are independently selected from hydrogen, halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R1v)-NH-C(O)-, -CH(R1v)- ; R1v is hydrogen or (1-2C)alkyl; wherein R1u and R2u are independently selected from hydrogen, halogen, cyano, (1-4C)alkyl, (1- 5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein Xu is selected from CH and N. 20. Compound according to any one of the preceding claims 1 – 9 and 18 - 19, wherein V is any one of O, -C(O)-NH-, -CH(R1v)-NH-C(O)-, and -CH(R1v)-; wherein R1v is hydrogen or (1-2C)alkyl. 21. Compound according to any one of the preceding claims 1 – 9 and 18 - 20, wherein R1 is selected from the group consisting of: wherein R1w and R2w are independently selected from hydrogen, halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R1u and R2u are independently selected from hydrogen, halogen, cyano, (1-4C)alkyl, (1- 5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein Xu is selected from CH and N. 22. Compound according to any one of the preceding claims 1 – 9 and 18 - 21, wherein R1 is selected from the group consisting of: ; wherein R2w is selected from hydrogen, halogen, (1-2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and wherein R3u is selected from hydrogen, halogen, cyano, (1-4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro. 23. Compound according to any one of the preceding claims 1 – 9 and 18 - 22, wherein R1 is: wherein R2w is selected from hydrogen, fluoro, methyl or methoxy; wherein R3u is selected from hydrogen, halogen, cyano, (1-4C)alkyl, (1-2C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro.

24. Compound according to any one of the preceding claims, wherein the compound is selected from the group consisting of:

25. Compound according to any one of the preceding claims, wherein said compound is preferable suitable for inhibiting Bruton’s Tyrosine Kinase (BTK), wherein said compound is selected from the group consisting of:

26. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Bruton’s Tyrosine Kinase C481S mutant (BTK C481S), wherein said compound is selected from the group consisting of: 27. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Aurora B Kinase (AurB), wherein said compound is selected from the group consisting of: .

28. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Epidermal Growth Factor Receptor (EGFR), wherein said compound is selected from the group consisting of: 29. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Fibroblast Growth Factor Receptor 1 (FGFR1), wherein said compound is selected from the group consisting of:

30. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting FMS related tyrosine kinase 3 (FLT-3), wherein said compound is selected from the group consisting of: 31. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Colony Stimulating Factor 1 Receptor (FMS), wherein said compound is selected from the group consisting of:

32. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting IL2 inducible T cell kinase (ITK), wherein said compound is selected from the group consisting of:

. 33. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Lymphocyte-Specific Protein Tyrosine Kinase (LCK), wherein said compound is selected from the group consisting of:

34. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting LCK/YES Novel Tyrosine Kinase (LYN), wherein said compound is selected from the group consisting of: 35. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Platelet Derived Growth Factor Receptor Beta (PDGFR-β), wherein said compound is selected from the group consisting of:

36. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting TEC Protein Tyrosine Kinase (TEC), wherein said compound is selected from the group consisting of: 37. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Vascular Endothelial Growth Factor Receptor (VEGFR), wherein said compound is selected from the group consisting of: 38. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Mitogen-Activated Protein Kinase Kinase 1 (MEK1), wherein said compound is selected from the group consisting of:

39. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Abelson Tyrosine Kinase (ABL), wherein said compound is selected from the group consisting of: 40. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Axl Receptor Tyrosine Kinase (AXL), wherein said compound is selected from the group consisting of: 41. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting or hepatocyte growth factor receptor or tyrosine-protein kinase Met (c-MET), wherein said compound is selected from the group consisting of: . 42. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Fibroblast Growth Factor Receptor 3 (FGFR3), wherein said compound is selected from the group consisting of:

43. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting Insuline Like Growth Factor Receptor 1 (IGFR1), wherein said compound is selected from the group consisting of: . 44. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting "REarranged during Transfection" receptor tyrosine kinase (RET), wherein said compound is selected from the group consisting of:

45. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting non-receptor tyrosine kinase Src (SRC), wherein said compound is selected from the group consisting of: 46. Compound according to any one of the claims 1 to 24, wherein said compound is preferable suitable for inhibiting non-receptor tyrosine kinase Yes (YES), wherein said compound is selected from the group consisting of:

47. Compound according to any of the preceding claims or a pharmaceutically acceptable salt thereof, for use as a medicament. 48. Compound according to any of the claims 1 - 46 or a pharmaceutically acceptable salt thereof, for use in therapy. 49. Compound according to any of the claims 1 - 46 or a pharmaceutically acceptable salt thereof, for use in the treatment of Bruton’s Tyrosine Kinase (Btk) mediated disorders. 50. Compound according to any of the claims 1 - 46 or a pharmaceutically acceptable salt thereof, for use in the treatment of Bruton’s Tyrosine Kinase (Btk) mediated disorders, wherein the Btk mediated disorder is selected from the group consisting of an allergic disease, an immunological disease, an autoimmune disease, an inflammatory disease, heteroimmune condition, fibrosis, a thromboembolic disease, a bone-related disease, and cancer. 51. Compound according to any of the claims 1 - 46 or claim 49 – 50 or a pharmaceutically acceptable salt thereof, for use in the treatment of Bruton’s Tyrosine Kinase (Btk) mediated disorders, wherein the Btk mediated disorder is selected from the group consisting of arthritis, rheumatoid arthritis, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, deforming arthritis, osteoarthritis, traumatic arthritis, gouty arthritis, Reiter’s syndrome, polychondritis, acute synovitis and spondylitis, glomerulonephritis (with or without nephrotic syndrome), autoimmune hematologic disorders, hemolytic anemia, aplasic anemia, idiopathic thrombocytopenia, and neutropenia, autoimmune gastritis, and autoimmune inflammatory bowel diseases, ulcerative colitis, Crohn’s disease, host versus graft disease, allograft rejection, chronic thyroiditis, Graves’ disease, schleroderma, diabetes (type I and type II), active hepatitis (acute and chronic), pancreatitis, primary billiary cirrhosis, myasthenia gravis, multiple sclerosis, systemic lupus erythematosis, psoriasis, atopic dermatitis, contact dermatitis, eczema, skin sunburns, vasculitis (e.g. Behcet’s disease) chronic renal insufficiency, Stevens-Johnson syndrome, inflammatory pain, idiopathic sprue, cachexia, sarcoidosis, Guillain-Barré syndrome, uveitis, conjunctivitis, kerato conjunctivitis, otitis media, periodontal disease, pulmonary interstitial fibrosis, asthma, bronchitis, rhinitis, sinusitis, pneumoconiosis, pulmonary insufficiency syndrome, pulmonary emphysema, pulmonary fibrosis, silicosis, chronic inflammatory pulmonary disease, and chronic obstructive pulmonary disease. 52. Compound according to any of the claims 1 - 46 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, lymphoma or leukemia. 53. Compound according to any of the claims 1 - 46 or claim 52 or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from B-cell malignancy, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocyte leukemia, non-Hodgkin lymphoma for example ABC-DLBCL, mantle cell lymphoma, follicular lymphoma, hairy cell leukemia B-cell non-Hodgkin lymphoma, Waldenstrom’s macroglobulinemia, Richter transformation, multiple myeloma, bone cancer, bone metastasis, chronic lymphocytic lymphomas, B-cell prolymphocyte leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell lymphoma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, and lymphomatoid granulomatosis. 54. Use of the compound according to any of the claims 1 - 46 or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament. 55. A pharmaceutical composition which comprises the compound according to any of the claims 1 – 46 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients. 56. The pharmaceutical composition of claim 55, which further comprises at least one additional therapeutically active agent. 57. A method for treating of cancer in a subject in need thereof comprising administering to the subject the compound according to any of the claims 1 – 46 or a pharmaceutically acceptable salt thereof in an amount effective to treat cancer. 58. A method for treating a subject suffering with a Bruton's Tyrosine Kinase (Btk) mediated disorder comprising administering to the subject the compound of any of the claims 1 – 46 or a pharmaceutically acceptable salt thereof in an amount effective to treat the Btk mediated disorder. 59. Compound according to any of the claims 1 – 46, in particular claim 27, or a pharmaceutically acceptable salt thereof, for use in the treatment of Aurora B Kinase mediated disorders.

60. Compound according to any of the claims 1 – 46, in particular claim 27 or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer. 61. Compound according to any of the claims 1 – 46, in particular claim 28, or a pharmaceutically acceptable salt thereof, for use in the treatment of Epidermal Growth Factor Receptor (EGFR) mediated disorders. 62. Compound according to any of the claims 1 – 46, in particular claim 28, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein preferably the cancer is selected from a lung cancer, non-small cell lung cancer, a pancreatic cancer, a colon cancer, a breast cancer, colorectal cancer, a prostate cancer, a head and neck cancer, an ovarian cancer, a brain cancer, a kidney carcinoma, pancreatic cancer, ovarian cancer, gastric cancer, glioma or prostate cancer. 63. Compound according to any of the claims 1 – 46, in particular claim 29, or a pharmaceutically acceptable salt thereof, for use in the treatment of Fibroblast Growth Factor Receptor 1 (FGFR1) mediated disorders. 64. Compound according to any of the claims 1 – 46, in particular claim 29, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein preferably the cancer is selected from brain cancer, head and neck cancer, gastric cancer or ovarian cancer. 65. Compound according to any of the claims 1 – 46, in particular claim 30, or a pharmaceutically acceptable salt thereof, for use in the treatment of FMS related tyrosine kinase 3 (FLT-3) mediated disorders. 66. Compound according to any of the claims 1 – 46, in particular claim 30, or a pharmaceutically acceptable salt thereof, for use in the treatment of a proliferative disease, such as cancer, preferably a proliferative disease selected from the group consisting of: presence or progression of solid tumor, sarcoma, lymphoma, B-cell lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Burkitt lymphoma, leukemias, chronic lymphocytic leukemia, B-cell prolymphocytic leukemia, plasma cell myeloma, plasmacytoma, lymphomatoid granulomatosis, melanoma, B-cell proliferative disease, brain cancer, kidney cancer, liver cancer, adrenal gland cancer, bladder cancer, breast cancer, breast ductal carcinoma, lobular carcinoma, stomach neoplasm, stomach cancer, esophagus cancer, ovarian cancer, colorectal cancer, prostate cancer, pancreas cancer, lung cancer, vagina cancer, membranous adenocarcinoma, thyroid cancer, neck cancer, CNS cancer, malignant glioma, myeloproliferative disease, glioblastoma, multiple myeloma, gastrointestinal cancer, colorectal carcinoma, head and neck neoplasms, brain tumor, epidermal hyperplasia, psoriasis, prostate hyperplasia, neoplasia, neoplasia of epithelial character, and a combination thereof. 67. Compound according to any of the claims 1 – 46, in particular claim 30, or a pharmaceutically acceptable salt thereof, for use in the treatment of a hematological malignancy selected from the group consisting of: myeloma, acute lymphocytic leukemia, acute myeloid leukemia, acute promyelocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, chronic neutrophilic leukemia, acute undifferentiated leukemia, anaplastic large-cell lymphoma, adult T-cell acute myelocytic leukemia, acute myelocytic leukemia with trilineage myelodysplasia, mixed lineage leukemia, myelodysplasia syndromes, myeloproliferative disorders, multiple myeloma, myeloid sarcoma and a combination thereof. 68. Compound according to any of the claims 1 – 46, in particular claim 31, or a pharmaceutically acceptable salt thereof, for use in the treatment of Colony Stimulating Factor 1 Receptor (FMS) mediated disorders. 69. Compound according to any of the claims 1 – 46, in particular claim 31, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from cancer, cardiovascular diseases, inflammatory diseases, and autoimmune diseases, including chronic graft vs host disease. 70. Compound according to any of the claims 1 – 46, in particular claim 31, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from osteoporosis, Paget's disease, rheumatoid arthritis and other forms of inflammatory arthritis, osteoarthritis, prosthesis failure, osteolytic sarcoma, myeloma, and tumor metastasis to bone. 71. Compound according to any of the claims 1 – 46, in particular claim 31, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from glomerulonephritis, inflammatory bowel disease, prosthesis failure, sarcoidosis, congestive obstructive pulmonary disease, idiopathic pulmonary fibrosis, asthma, pancreatitis, HIV infection, psoriasis, diabetes, tumor related angiogenesis, age-related macular degeneration, diabetic retinopathy, restenosis, schizophrenia and Alzheimer's dementia. 72. Compound according to any of the claims 1 – 46, in particular claim 31, or a pharmaceutically acceptable salt thereof, for use in the treatment of pain, including skeletal pain caused by tumor metastasis or osteoarthritis, or visceral, inflammatory, or neurogenic pain in a mammal. 73. Compound according to any of the claims 1 – 46, in particular claim 31, or a pharmaceutically acceptable salt thereof, for use in the treatment of an autoimmune disease selected from the group consisting of systemic lupus erythematosus, rheumatoid arthritis and other forms of inflammatory arthritis, psoriasis, Sjogren's syndrome, multiple sclerosis, or uveitis. 74. Compound according to any of the claims 1 – 46, in particular claim 32, or a pharmaceutically acceptable salt thereof, for use in the treatment of IL2 inducible T cell kinase (ITK) mediated disorders. 75. Compound according to any of the claims 1 – 46, in particular claim 32, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection. 76. Compound according to any of the claims 1 – 46, in particular claim 33, or a pharmaceutically acceptable salt thereof, for use in the treatment of Lymphocyte-Specific Protein Tyrosine Kinase (LCK) mediated disorders. 77. Compound according to any of the claims 1 – 46, in particular claim 33, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease or disorder mediated by immune cells selected from T lymphocytes, NK cells, B lymphocytes, e.g. acute or chronic rejection of organ or tissue allo- or xenografts, atherosclerosis, vascular occlusion due to vascular injury such as angioplasty, restenosis, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, hypertension, heart failure, chronic obstructive pulmonary disease, CNS disease such as Alzheimer disease or amyotrophic lateral sclerosis, cancer, cholangiocarcinoma, cytokine release syndrome, lymphodepletion in combination with immunotherapy, such as immunotherapy using NK cells, infectious disease such as AIDS, septic shock or adult respiratory distress syndrome, ischemia/reperfusion injury e.g. myocardial infarction, stroke, gut ischemia, renal failure or hemorrhage shock, or traumatic shock. 78. Compound according to any of the claims 1 – 46, in particular claim 33, or a pharmaceutically acceptable salt thereof, for use in the treatment of a chronic T cell disorder like multiple sclerosis and rheumatoid arthritis, or an acute inflammatory disorder in which T cells play a prominent role including transplant rejection, atopic dermatitis and delayed type hypersensitivity. 79. Compound according to any of the claims 1 – 46, in particular claim 34, or a pharmaceutically acceptable salt thereof, for use in the treatment of LCK/YES Novel Tyrosine Kinase (LYN) mediated disorders. 80. Compound according to any of the claims 1 – 46, in particular claim 34, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from cancer, Type II diabetes, fibrosis, chronic inflammation, chronic pancreatitis, pancreatic fibrosis, or inflammatory myofibroblastic tumors (IMTs).

81. Compound according to any of the claims 1 – 46, in particular claim 35, or a pharmaceutically acceptable salt thereof, for use in the treatment of Platelet Derived Growth Factor Receptor Beta (PDGFR-β) mediated disorders. 82. Compound according to any of the claims 1 – 46, in particular claim 35, or a pharmaceutically acceptable salt thereof, for use in the treatment a disease selected from cancer, vascular disorder and fibrotic diseases. 83. Compound according to any of the claims 1 – 46, in particular claim 36, or a pharmaceutically acceptable salt thereof, for use in the treatment of TEC Protein Tyrosine Kinase (TEC) mediated disorders. 84. Compound according to any of the claims 1 – 46, in particular claim 36, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection. 85. Compound according to any of the claims 1 – 46, in particular claim 37, or a pharmaceutically acceptable salt thereof, for use in the treatment of Vascular Endothelial Growth Factor Receptor (VEGFR) mediated disorders. 86. Compound according to any of the claims 1 – 46, in particular claim 37, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from cancer, angiogenesis and lymphangiogenesis, Parkinson’s and Alzheimer’s diseases, inflammatory diseases. 87. Compound according to any of the claims 1 – 46, in particular claim 38, or a pharmaceutically acceptable salt thereof, for use in the treatment of Mitogen-Activated Protein Kinase Kinase 1 (MEK1) mediated disorders. 88. Compound according to any of the claims 1 – 46, in particular claim 38, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, such as melanoma and non-small cell lung cancer (NSCLC). 89. Compound according to any of the claims 1 – 46, in particular claim 39, or a pharmaceutically acceptable salt thereof, for use in the treatment of Abelson Tyrosine Kinase (ABL) mediated disorders. 90. Compound according to any of the claims 1 – 46, in particular claim 39, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.

91. Compound according to any of the claims 1 – 46, in particular claim 40, or a pharmaceutically acceptable salt thereof, for use in the treatment of Axl Receptor Tyrosine Kinase (AXL) mediated disorders. 92. Compound according to any of the claims 1 – 46, in particular claim 40, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer. 93. Compound according to any of the claims 1 – 46, in particular claim 41, or a pharmaceutically acceptable salt thereof, for use in the treatment of hepatocyte growth factor receptor or tyrosine- protein kinase Met (c-MET) mediated disorders. 94. Compound according to any of the claims 1 – 46, in particular claim 41, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein preferably the cancer is selected from metastatic non-small cell lung cancer (NSCLC) and advanced renal cell carcinoma. 95. Compound according to any of the claims 1 – 46, in particular claim 42, or a pharmaceutically acceptable salt thereof, for use in the treatment of Fibroblast Growth Factor Receptor 3 (FGFR3) mediated disorders. 96. Compound according to any of the claims 1 – 46, in particular claim 42, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein preferably the cancer is selected from multiple myeloma, bladder cancer, non-small cell lung cancer, oral cancers, and oropharyngeal squamous cell carcinoma. 97. Compound according to any of the claims 1 – 46, in particular claim 43, or a pharmaceutically acceptable salt thereof, for use in the treatment of Insuline Like Growth Factor Receptor 1 (IGFR1) mediated disorders. 98. Compound according to any of the claims 1 – 46, in particular claim 43, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein preferably the cancer is selected from breast cancer, sarcoma, and non–small cell lung cancer (NSCLC). 99. Compound according to any of the claims 1 – 46, in particular claim 44, or a pharmaceutically acceptable salt thereof, for use in the treatment of "REarranged during Transfection" receptor tyrosine kinase (RET) mediated disorders. 100. Compound according to any of the claims 1 – 46, in particular claim 44, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, wherein preferably the cancer is selected from thyroid carcinomas and lung cancers.

101. Compound according to any of the claims 1 – 46, in particular claim 45, or a pharmaceutically acceptable salt thereof, for use in the treatment of non-receptor tyrosine kinase Src (SRC) mediated disorders. 102. Compound according to any of the claims 1 – 46, in particular claim 45, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease or disorder selected from cancers, osteoporosis, cardiovascular disorders and immune system dysfunction. 103. Compound according to any of the claims 1 – 46, in particular claim 46, or a pharmaceutically acceptable salt thereof, for use in the treatment of non-receptor tyrosine kinase Yes (YES) mediated disorders. 104. Compound according to any of the claims 1 – 46, in particular claim 46, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease or disorder selected from cancers, osteoporosis, cardiovascular disorders and immune system dysfunction.

Description:
Reversible macrocyclic kinase inhibitors

Field of the invention

The present invention relates to compounds. More specifically, the invention relates to macrocyclic compounds and compositions for use as kinase inhibitors, along with processes to prepare the compounds and uses of the compounds. Specifically, the invention relates to reversible macrocyclic kinase inhibitors. More in particular, the invention relates to reversible macrocyclic kinase inhibitors with long target residence time.

Background of the invention

Kinases are enzymes that transfer a phosphate group from ATP to a protein while phosphatases remove a phosphate group from protein. Together, these two enzymatic processes regulate cellular functions such as cell proliferation, subcellular translocation, apoptosis, inflammation and metabolism (Attwood M.M. et al (2021) Nat Rev Drug Discov). The human kinome is composed of over 500 kinases.

There is clinical evidence that supports the driver role of kinases in cancer owing to their aberrant activation by either translocations or activating mutations. Chromosomal translocations produce fusion proteins with abnormal localization that can be potentially oncogenic. Identification and characterization of these disease drivers has facilitated the design and approval of molecularly guided cancer therapies, beginning with the pioneering example of imatinib to treat CML driven by the BCR- ABL translocation, which results in a protein with elevated tyrosine kinase activity. Many of small- molecule kinase inhibitors approved by the FDA that target kinases have oncology indications. Recent developments of kinase inhibitors, including developments of kinase inhibitors for oncology, have been reported, see: Attwood et al. (2021) “Trends in kinase drug discovery targets, indications and inhibitor design.” Nat Rev Drug Discov 20, pages 839-861 (2021).

The recent development of small-molecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy. Among them are inhibitors for EGFR (afatinib, osimertinib), BTK (ibrutinib, acalabrutinib and zanubrutinib), RET (cabozantinib, selpercatinib), MET (capmatinib, tepotinib) and FLT-3 (gilteritinib, midostaurin). Nevertheless, many factors influence the clinical efficacy of these molecules.

Bruton's tyrosine kinase (BTK) is a member of the Src-related Tec family of protein kinases which are a large subset of kinases which play a central role in the regulation of a wide variety of cellular signaling processes. BTK plays a key role in the B-cell receptor signaling and a critical role in the regulation of survival, proliferation, activation and differentiation of B-lineage cells. Targeting of BTK with small molecule inhibitors such as the FDA approved irreversible BTK inhibitors ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib has proven to be efficacious in several B cell malignancies including Chronic Lymphocytic Leukemia (CLL), Mantle Cell Lymphoma (MCL), Waldenstrom's Macroglobulinemia (WM) and Small Lymphocytic Lymphoma SLL. Combinations of BTK inhibitors with other novel drugs or regimens results in more profound responses and much higher rates of minimal residual disease negativity. BTK is also expressed and plays also pro-tumorigenic roles in several solid tumors (Xianhui Wang et al. 2021). In prostate cancer cells BTK inhibition with ibrutinib or acalabrutinib inhibited cell growth (Kokabee et al 2015). Ibrutinib has also been shown to inhibit in vivo (xenograft) breast cancer cell growth (Wang et al., 2016) and inhibition of BTK with ibrutinib blocked gastric cancer cell growth (Wang et al., 2016). BTK inhibitors have also showed inhibition of cellular proliferation and migration, and induced apoptosis and autophagy in glioblastoma cell lines (Wei et al., 2016; Wang et al., 2017).

In addition to its role in BCR signaling, BTK is also involved in many other immunological pathways which provides a rationale for the targeting of BTK in the context of inflammatory and systemic autoimmune disease (Stefan F. H. Neys et al. 2021).

A drawback of the currently approved irreversible inhibitors is that drug resistance in malignant diseases can develop when BTK variations at the catalytic site and the gatekeeper of the BTK are not able to bind efficiently to irreversible inhibitors in patients treated with currently approved BTK inhibitors. This is a rather common event in patients treated with irreversible inhibitors and who experience relapse. A major mechanism for the acquired resistance is the emergence of BTK cysteine 481 (C481) mutations. These mutations hamper binding of irreversible inhibitors such as ibrutinib and acalabrutinib which form a covalent bond with this amino acid. Other mutations that can result in acquired resistance to both irreversible covalent and reversible non-covalent BTK inhibitors are BTK gatekeeper residue threonine 474 (T474) mutations which can reduce BTK inhibitor access to BTK (Rula Zain et al. 2021 , Shenqiu Wang et al. 2019).

Second-generation BTK inhibitors include acalabrutinib, zanubrutinib, and tirabrutinib which offer greater BTK selectivity. While these agents may limit off-target toxicity, they do not overcome common mechanisms of ibrutinib resistance.

Other kinase inhibitors are known, e.g. for the kinases LCK (Lymphocyte-Specific Protein Tyrosine Kinase), FGFR1 (Fibroblast Growth Factor Receptor 1), FLT3 (FMS-like tyrosine kinase 3), PDGFR-β (Platelet Derived Growth Factor Receptor Beta), FMS (Colony Stimulating Factor 1 Receptor), LYN (LCK/YES Novel Tyrosine Kinase), MEK1 (Mitogen-Activated Protein Kinase Kinase 1), AUR-B (Aurora B Kinase), ITK (IL2 inducible T cell kinase), VEGFR (Vascular Endothelial Growth Factor Receptor), EGFR (Epidermal Growth Factor Receptor), TEC (TEC Protein Tyrosine Kinase), ABL (Abelson Tyrosine Kinase), AXL (Axl Receptor Tyrosine Kinase), c-MET (hepatocyte growth factor receptor or tyrosine-protein kinase Met), FGFR3 (Fibroblast Growth Factor Receptor 3), IGFR1 (Insuline Like Growth Factor Receptor 1), RET ("REarranged during Transfection" receptor tyrosine kinase), SRC (non-receptor tyrosine kinase Src) and YES (non-receptor tyrosine kinase Yes). Mutations of one or more of these kinases are reported and are known to alter and /or disturb molecular pathways. In several studies the mutations of the particular kinases have been reported to be associated with disorders including cancer types. It has been found that advanced tumors find escape routes to circumvent target inhibition, leading to drug resistance. Drug resistance mechanisms related to mutations have been studied: drug resistance occurs primarily through four main mechanisms. Acquired drug resistance mutations most commonly affect the binding of the drug to its target. Acquired oncogenic amplifications or rearrangements can activate downstream signaling to bypass inhibition of the drug target. Mutations in downstream effectors can activate signaling pathways despite effective inhibition of an upstream kinase target. State transformation can lead to kinase inhibitor insensitivity (Cohen et al. 202, Kinase drug discovery 20 years after imatinib: progress and future directions; Nature Reviews Drug Discovery volume 20, pages 551-569 (2021)).

Kinase activity is commonly assessed by measuring the half-maximum inhibitory potency ( IC 50 ) or binding affinity (K D ) in a kinase enzyme activity assay or using Surface Plasmon Resonance (SPR) (Willemsen-Seegers N. et al (2016) J Mol Biol). An important aspect of these assays is that IC 50 or K D is measured in a system with a fixed concentration of inhibitor. In living systems, compounds and enzymes usually encounter each other in a compartment, such as the cell, or at the cell surface, where the compound diffuses in and out, or may be actively extruded by drug pumps. Thus, whereas the concentration of a target remains constant, the concentration of compound is continually changing. In such a system, it has been proposed that the time a compound resides on its target, the target residence time tau (T), is a more important determinant of its pharmacological activity than the IC 50 or K D measured at equilibrium (Copeland R.A. et al (2006) Nat Rev Drug Discov). In addition, if a drug demonstrates a long residence time on its target and a short residence time on its off-targets, selectivity is enhanced, providing advantages for drug safety (Barf T. and Kaptein A. (2012) J Med Chem). Thus, the biological action of drugs with long target residency can endure long after it is cleared from the systemic circulation. An extreme example of drugs with long target residence time is irreversible inhibitors, usually obtained by covalent binding to the target. Compounds with equipotent affinity or potency might possess different residence times on their target protein as was demonstrated by erlotinib, gefitinib and lapatinib on EGFR (Willemsen-Seegers N. et al (2016) J Mol Biol).

Therefore, an aim of the present invention is to provide inhibitors providing improved pharmacological activity towards kinases. In addition, the invention aims to provide mutant-inhibitors suitable for inhibiting mutant(s) of kinases.

Furthermore, it is an aim of certain embodiments of this invention to provide reversible (mutant) inhibitors with a long target residence time.

Another aim of certain embodiments of this invention is to provide cancer treatments. In particular, it is an aim of certain embodiments of this invention to provide compounds with have comparable activity to existing cancer treatments but are also effective against mutations.

Summary of the invention

The inventors have found that a compound of Formula (l-a) to (l-h): wherein said compound is as further described below, or a pharmaceutically acceptable salt and/or solvate thereof, provides improved kinase inhibition. The inventors have found that these compounds being a macrocyclic compound having any one of the scaffolds of Formula (l-a) to (l-h) provide improved kinase inhibition.

In a first aspect of the invention is provided a compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is selected from the group consisting of: W is a direct bond or an aryl group having 6-10 carbon or a heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;

V is selected from the group consisting of: a direct bond, O, -OCH 2 - , -CH(R 1v )-, -C(O)-, -C(O)-N(R 2v )-, -N(R 2v )-C(O)-,-NH-C(O)-NH-, -NH-C(O)-C(R 3v )(R 4v )-C(O)-NH-, -NH-SO 2 -, -NH-C(O)-O-, -CH(R 1v )-NH- C(O)-, -CH(R 1v )-C(O)-NH-, -C≡C-, and -CH 2 O- ; R 1v is hydrogen or (1 -2C)alkyl; R 2v is hydrogen or (1 -2C)alkyl; R 3v is hydrogen or (1 -2C)alkyl; R 4v is hydrogen or (1 -2C)alkyl; or R 3v and R 4v form together with the carbon atom they are attached to a (3-6C)cycloalkyl;

U is an aryl group having 6-10 carbon, a heteroaryl group having 1-9 carbon or a cycloalkyl group having 3-6 carbon; wherein any of said aryl group, heteroaryl group and cycloalkyl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1-6C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R 2 is of Formula (ll-a) to (ll-f) selected from the group consisting of: wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X 1 , X 2 and X 3 are independently selected from CH 2 , -CH 2 CH 2 -, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with one or more substituents selected from halogen, hydroxy, (1 -3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R 3 and R 4 together represent a linker having Formula ( III-1 to III-40) selected from the group consisting of: whereby the marks the position of R 3 in any one of Formula l-a to l-h, and whereby the marks the position of R 4 in any one of Formula I l-a to I l-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CD 3 , (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl, (3- 6C)cycloalkoxy or (1-6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein R 5 is hydrogen, NH 2 or Methyl.

In a second aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use as a medicament.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in therapy.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Bruton's Tyrosine Kinase (BTK) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Aurora B Kinase mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Epidermal Growth Factor Receptor (EGFR) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Fibroblast Growth Factor Receptor 1 (FGFR1) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of FMS related tyrosine kinase 3 (FLT- 3) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Colony Stimulating Factor 1

Receptor (FMS) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of IL2 inducible T cell kinase (ITK) mediated disorders. In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Lymphocyte-Specific Protein Tyrosine Kinase (LCK) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of LCK/YES Novel Tyrosine Kinase (LYN) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Platelet Derived Growth Factor Receptor Beta (PDGFR-β) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of TEC Protein Tyrosine Kinase (TEC) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Vascular Endothelial Growth Factor Receptor (VEGFR) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Mitogen-Activated Protein Kinase Kinase 1 (MEK1) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Abelson Tyrosine Kinase (ABL) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment ofAxI Receptor Tyrosine Kinase (AXL) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of hepatocyte growth factor receptor or tyrosine-protein kinase Met (c-MET) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Fibroblast Growth Factor Receptor 3 (FGFR3) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of Insuline Like Growth Factor Receptor 1 (IGFR1) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of “Rearranged during Transfection” receptor tyrosine kinase (RET) mediated disorders.

In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of non-receptor tyrosine kinase Src (SRC) mediated disorders. In another aspect of the invention is provided a compound according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of non-receptor tyrosine kinase Yes (YES) mediated disorders.

In another aspect of the invention is provided a compound of according to the invention or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.

In another aspect of the invention is provided a use of the compound according to the invention or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament.

In another aspect of the invention is provided a pharmaceutical composition which comprises the compound according to the invention or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.

In another aspect of the invention is provided a method for treating of cancer in a subject in need thereof comprising administering to the subject the compound according to the invention or a pharmaceutically acceptable salt thereof in an amount effective to treat cancer.

In another aspect of the invention is provided a method for treating a subject suffering with a Bruton's Tyrosine Kinase (BTK) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the BTK mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Aurora B Kinase mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Aurora B Kinase mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Epidermal Growth Factor Receptor (EGFR) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Epidermal Growth Factor Receptor (EGFR) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Fibroblast Growth Factor Receptor 1 (FGFR1) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Fibroblast Growth Factor Receptor 1 (FGFR1) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a FMS related tyrosine kinase 3 (FLT-3) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the FMS related tyrosine kinase 3 (FLT-3) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Colony Stimulating Factor 1 Receptor (FMS) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Colony Stimulating Factor 1 Receptor (FMS) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a IL2 inducible T cell kinase (ITK) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the IL2 inducible T cell kinase (ITK) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Lymphocyte-Specific Protein Tyrosine Kinase (LCK) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Lymphocyte-Specific Protein Tyrosine Kinase (LCK) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a LCK/YES Novel Tyrosine Kinase (LYN) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the LCK/YES Novel Tyrosine Kinase (LYN) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Platelet Derived Growth Factor Receptor Beta (PDGFR-β) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Platelet Derived Growth Factor Receptor Beta (PDGFR-β) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a TEC Protein Tyrosine Kinase (TEC) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the TEC Protein Tyrosine Kinase (TEC) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Vascular Endothelial Growth Factor Receptor (VEGFR)mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Vascular Endothelial Growth Factor Receptor (VEGFR)mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Mitogen-Activated Protein Kinase Kinase 1 (MEK1) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Mitogen-Activated Protein Kinase Kinase 1 (MEK1) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Abelson Tyrosine Kinase (ABL) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Abelson Tyrosine Kinase (ABL) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with an Axl Receptor Tyrosine Kinase (AXL) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Axl Receptor Tyrosine Kinase (AXL) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a hepatocyte growth factor receptor or tyrosine-protein kinase Met (c-MET) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the hepatocyte growth factor receptor or tyrosine-protein kinase Met (c-MET) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a Fibroblast Growth Factor Receptor 3 (FGFR3) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Fibroblast Growth Factor Receptor 3 (FGFR3) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with an Insuline Like Growth Factor Receptor 1 (IGFR1) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the Insuline Like Growth Factor Receptor 1 (IGFR1) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a “Rearranged during Transfection” receptor tyrosine kinase (RET) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the “Rearranged during Transfection” receptor tyrosine kinase (RET) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a non-receptor tyrosine kinase Src (SRC) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the non-receptor tyrosine kinase Src (SRC) mediated disorder.

In another aspect of the invention is provided a method for treating a subject suffering with a non-receptor tyrosine kinase Yes (YES) mediated disorder comprising administering to the subject the compound of the invention in an amount effective to treat the non-receptor tyrosine kinase Yes (YES) mediated disorder.

Each of the sub-formulas 1-62 of the compound is a preferred embodiment of the present application.

The present invention will be illustrated further by means of the following non-limiting examples.

Definitions

The term "pharmaceutical composition” as used herein has its conventional meaning and refers to a composition which is pharmaceutically acceptable.

The term "pharmaceutically acceptable” as used herein has its conventional meaning and refers to compounds, material, compositions and/or dosage forms, which are, within the scope of sound medical judgment suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.

The term "effective amount' as used herein, refers to an amount of the compound of the invention, and/or an additional therapeutic agent, or a composition thereof, that is effective in producing the desired therapeutic, ameliorative, inhibitory or preventative effect when administered to a subject suffering from a kinase-mediated disease or disorder. In the combination therapies of the present invention, as effective amount can refer to each individual agent or to the combination as a whole, wherein the amounts of all agents administered are together effective, but wherein the component agent of the combination may not be present individually in an effective amount.

A "subject" is a human or non-human mammal. In one embodiment, a subject is a human.

The term "controlling” is intended to refer to all processes wherein there may be a slowing, interrupting, arresting or stopping of the progression of the diseases and conditions affecting the mammal. However, "controlling” does not necessarily indicate a total elimination of all disease and condition symptoms, and is intended to include prophylactic treatment. The term "excipient” as used herein has its conventional meaning and refers to a pharmaceutically acceptable ingredient, which is commonly used in the pharmaceutical technology for preparing a granulate, solid or liquid oral dosage formulation.

The term "salt” as used herein has its conventional meaning and includes the acid addition and base salts of the compound of the invention.

The term "solvate” as used herein has its conventional meaning. One or more compounds of the invention or the pharmaceutically acceptable salts thereof may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. "Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding. Including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H 2 O and includes any hydrate of the compound or the salt of said compound.

The term "treatment” as used herein has its conventional meaning and refers to curative, palliative and prophylactic treatment.

The term "unit dosage form” has its conventional meaning and refers to a dosage form which has the capacity of being administered to a subject, preferably a human, to be effective, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose comprising the therapeutic agent, i.e. the compound of the invention.

The term "BTK” as used herein has its conventional meaning and refers to Bruton's Tyrosine Kinase. Bruton's tyrosine kinase (BTK) is a member of the Src-related Tec family of protein kinases which are a large subset of kinases which play a central role in the regulation of a wide variety of cellular signaling processes. BTK plays a key role in the B-cell receptor signaling and a critical role in the regulation of survival, proliferation, activation and differentiation of B-lineage cells. Targeting of BTK with small molecule inhibitors such as the FDA approved irreversible BTK inhibitors ibrutinib, acalabrutinib, zanubrutinib and tirabrutinib has proven to be efficacious in several B cell malignancies including Chronic Lymphocytic Leukemia (CLL), Mantle Cell Lymphoma (MCL), Waldenstrom's Macroglobulinemia (WM) and Small Lymphocytic Lymphoma SLL. Combinations of BTK inhibitors with other novel drugs or regimens results in more profound responses and much higher rates of minimal residual disease negativity.

The term "BTK inhibitor” as used herein has its conventional meaning and refers to an inhibitor for BTK. A BTK inhibitor may be a small molecule inhibitor. Inhibitors may be irreversible inhibitors, such as by forming a covalent bond, and may be reversible inhibitors, which may form a temporary interaction with BTK.

The term "mutant-BTK” as used herein has its conventional meaning and refers to mutations of BTK. Mutations of BTK may be referred to by an altered amino acid target (such as C as single-letter data-base code for cysteine) at a certain position of the BTK structure (such as 481). Additionally, the amino acid substitution at the mutation position may be referred to by an additional amino acid single- letter data-base code, such as C481 S for serine substitution and C481T for threonine substitution of cysteine at the 481 position.

A drawback of the currently approved irreversible inhibitors is that drug resistance in malignant diseases can develop when BTK variations at the catalytic site and the gatekeeper of the BTK are not able to bind efficiently to irreversible inhibitors in patients treated with currently approved BTK inhibitors. This is a rather common event in patients treated with irreversible inhibitors and who experience relapse. A major mechanism for the acquired resistance is the emergence of BTK cysteine 481 (C481) mutations. These mutations hamper binding of irreversible inhibitors such as ibrutinib and acalabrutinib which form a covalent bond with this amino acid. Other mutations that can result in acquired resistance to both irreversible covalent and reversible non-covalent BTK inhibitors are BTK gatekeeper residue threonine 474 (T474) mutations which can reduce BTK inhibitor binding to BTK.

The term "wt-BTK” or “WT-BTK” or “BTK WT ” as used herein has its conventional meaning and refers to wild-type Bruton's Tyrosine Kinase. A wild-type BTK has the regular meaning of a phenotype of the typical form of BTK as it occurs in nature. Originally, the wild-type was conceptualized as a product of the standard "normal" allele at a locus, in contrast to that produced by a non-standard, "mutant" allele.

The term "macrocycle” as used herein has its conventional meaning and refers to a part of a molecule containing a ring consisting of 12 or more ring atoms forming said ring. In an example, a twelve membered ring consist of 12 atoms forming said ring.

The term "binding affinity (K D )” as used herein has its conventional meaning and refers to the equilibrium dissociation constant which is an inverse measure of the affinity of a protein-ligand (small molecule) pair under equilibrium conditions. The value of K D is mathematically equivalent to the ratio k off /k on (or k d /k a ) measured using Surface Plasmon Resonance (SPR).

The term "association rate constant” or “on-rate (k on or k a )" as used herein has its conventional meaning and refers to a second-order rate constant that quantifies the rate at which a free ligand and free protein combine (through collisional encounters) to form a binary protein-ligand complex.

The term "dissociation rate constant” or “off-rate ( k off or k d )” as used herein has its conventional meaning and refers to a first-order rate constant that quantifies the rate at which a binary protein-ligand complex dissociates to the free ligand and free protein.

The term "target residence time tau (τ)” as used herein has its conventional meaning and refers to the time a compound resides on its target. Target residence time (T) can be determined according to the method as described below in the experimental section.

The term “ IC 50 ” as used herein has its conventional meaning and refers to the concentration of a substance that results in a 50% effect on some measure of biochemical function or substance-target binding interaction.

A bicyclic ringsystem, as used herein, refers to heterocyclic (heterocyclyl) groups, to cyclic groups having carbon groups only, i.e. without hetero atoms, within the cycle, and to combinations of a heterocyclic (heterocyclyl) group and a cyclic group having carbon groups only, i.e. without hetero atoms, within the cycle.

A monocylic ringsystem, as used herein, refers both to a heterocyclic (heterocyclyl) group, and to a cyclic group having carbon groups only, i.e. without hetero atoms, within the cycle. A heterocyclic (heterocyclyl) group, as used herein, refers to both heteroaryl groups and heterocycloalkyl groups.

A heterobicyclic group, as used herein, refers to a bicyclic group having one or more heteroatoms, which is saturated, partially unsaturated or unsaturated.

As used herein, aromatic groups (or aryl groups) include aromatic carbocyclic ring systems (e.g. phenyl) and fused polycyclic aromatic ring systems (e.g. naphthyl and 1 ,2,3,4-tetrahydronaphthyl).

The term "alkyl," as used herein, refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond having the specified number of carbon atoms. In different embodiments, an alkyl group contains, for example, from 1 to 6 carbon atoms (1 -6C)Alkyl or from 1 to 3 carbon atoms (1-3C)Alkyl. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl. In one embodiment, an alkyl group is linear. In another embodiment, an alkyl group is branched.

Unless specified otherwise, "alkyl" includes both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbon atoms; for example, "(1-6C)Alkyl" includes all of the hexyl alkyl and pentyl alkyl isomers as well as n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl. "Alkylene" refers to both branched- and straight-chain saturated aliphatic hydrocarbon groups, including all isomers, having the specified number of carbons, and having two terminal end chain attachments; for example, the term "A-C 4 alkylene-B" represents, for example, A-CH 2 -CH 2 -CH 2 -CH 2 -B, A-CH 2 -CH 2 -CH(CH 3 )-CH 2 -B, A-CH 2 -CH(CH 2 CH 3 )-B, A-CH 2 - C(CH 3 )(CH 3 )-B, and the like.

The term "alkylcarbonyl," as used herein, refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond attached to the carbonyl group, wherein the aliphatic hydrocarbon group has the specified number of carbon atoms. In different embodiments, an alkyl group or aliphatic hydrocarbon group contains, for example, from 1 to 6 carbon atoms (1-6C)Alkyl or from 1 to 3 carbon atoms (1-3C)Alkyl. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl. In one embodiment, an alkyl group is linear. In another embodiment, an alkyl group is branched.

Cycloalkyl means a cycloalkyl group having the recited number of carbon atoms, with the same meaning as previously defined, such as cyclopropyl, cyclobutyl, or cyclopentyl. "Cycloalkyl" refers to a cycloalkyl-group represented by an indicated number of carbon atoms; for example "(3-6C)cycloalkyl" includes cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

Heterocycloalkyl means a cycloalkyl group having the recited number of carbon atoms, and 1 - 3 heteroatoms selected from N, O and/or S, with the same meaning as previously defined.

Haloalkyl means a branched or unbranched alkyl group having the recited number of carbon atoms, in which one and up to all hydrogen atoms are replaced by a halogen; halogen is as defined herein. Examples of such branched or straight chained haloalkyl groups useful in the present invention include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl and n-butyl substituted independently with one or more halogens, e.g., fluoro, chloro, bromo and iodo. For example, a halo(1-3C)alkyl means a branched or unbranched alkyl group having 1 ,2, or 3 carbon atoms, in which at least one hydrogen atom is replaced by a halogen. Examples of "haloalkyl" include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 1 -fluoroethyl, 2-fluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, and perfluoro-n-propyl.

Alkoxy means an alkoxy group having the recited number of carbon atoms, the alkyl moiety having the same meaning as previously defined, e.g., "Alkoxy" refers to an alkyl-O-group represented by a linear or branched alkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "(1-6C)Alkoxy" includes -OCH 3 , -O-CH 2 CH 3 , -OCH(CH 3 ) 2 , -O(CH 2 ) 5 CH 3 , and the like.

Cycloalkoxy means a cycloalkyl group having the recited number of carbon atoms, with the same meaning as previously defined, attached via a ring carbon atom to an exocyclic oxygen atom, such as cyclopropoxyl, cyclobutoxyl.or cyclo pentoxyl. "Cycloalkoxy" refers to a cycloalkyl-O-group represented by a cycloalkyl group of indicated number of carbon atoms attached through an oxygen bridge; for example "(3-6C)cycloalkoxy" includes cyclopropyl-O-, cyclobutyl-O-, cyclopentyl-O-, or cyclohexyl-O-.

Heterocycloalkoxy means a cycloalkyl group having the recited number of carbon atoms, and 1-3 heteroatoms selected from N, O and/or S, with the same meaning as previously defined, attached via a ring carbon atom to an exocyclic oxygen atom.

Unless otherwise specifically noted as only "unsubstituted" or only "substituted", alkyl groups are unsubstituted or substituted with 1 to 3 substituents on each carbon atom.

It should be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.

The terms first, second, third and the like in the description and in the claims, are used for distinguishing between for example similar elements, compositions, constituents in a composition, or separate method steps, and not necessarily fordescribing a sequential or chronological order. The terms are interchangeable under appropriate circumstances and the embodiments of the invention can operate in other sequences than described or illustrated herein, unless specified otherwise.

Furthermore, the various embodiments, although referred to as “preferred” or “e.g." or “for example” or “in particular” and the like are to be construed as exemplary manners in which the invention may be implemented rather than as limiting the scope of the invention.

The term “comprising”, used in the claims, should not be interpreted as being restricted to for example the elements or the method steps or the constituents of a compositions listed thereafter; it does not exclude other elements or method steps or constituents in a certain composition. It needs to be interpreted as specifying the presence of the stated features, integers, (method) steps or components as referred to, but does not preclude the presence or addition of one or more other features, integers, steps or components, or groups thereof. Thus, the scope of the expression “a method comprising steps A and B” should not be limited to a method consisting only of steps A and B, rather with respect to the present invention, the only enumerated steps of the method are A and B, and further the claim should be interpreted as including equivalents of those method steps. Thus, the scope of the expression “a composition comprising components A and B” should not be limited to a composition consisting only of components A and B, rather with respect to the present invention, the only enumerated components of the composition are A and B, and further the claim should be interpreted as including equivalents of those components.

In addition, reference to an element or a component by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element or component are present, unless the context clearly requires that there is one and only one of the elements or components. The indefinite article "a" or "an" thus usually means "at least one".

Detailed description of the invention

The invention provides a new class of reversible macrocyclic kinase inhibitors with a long target residence time (T). Surprisingly, the inventors have found that compounds according to the invention provide an improved reversible binding activity towards various kinases, including BTK, LCK, FGFR1 , FLT3, PDGFR-β, FMS, LYN, MEK1 , AUR-B, ITK, VEGFR, EGFR, TEC, ABL, AXL, c-MET, FGFR3, IGFR1 , RET, SRC and YES including mutants of some of these kinases. The compounds according to the invention have any one of Formula (l-a) to (l-h), which contains a macrocyclic moiety, in combination with specific pharmacophores (e.g. based on ligands for binding to specific kinases) to provide a binding activity towards the kinase through improved reversible binding. In particular it has been found that the compound provides a considerable longer residence time than what is typically obtained with reversible kinase inhibitors.

Additionally, the inventors have surprisingly found that compounds of Formula l-a to l-h provide an enhanced binding activity towards mutant kinases forms, such as BTK mutant forms. In exemplary embodiments the inventors have demonstrated the enhanced binding activity towards BTK mutants BTK C481 S, BTK T316A, BTK T474I and BTKT474S. Based on these findings, and based on the macrocyclic moiety effect on the (BTK targeted) compounds of the invention, it is anticipated that binding activity to other kinase mutants is also enhanced.

Macrocyclic natural products have advanced to achieve numerous biochemical functions, and their pharmacological properties have led to their development as drugs. Macrocycles have been defined as a ring system consisting of 12 or more atoms (Driggers E.M. (2008) Nat Rev Drug Discov). A macrocycle provides diverse functionalities and stereochemical complexity in a conformationally pre- organized ring structure, which can result in superb physicochemical and pharmacological properties. By limiting the number of (bioactive) conformations available to the unbound molecule, there is a lower entropic cost when the molecule interacts with its target protein as compared to a non-macrocyclic compound. Macrocyclic ligands can be designed to displace ordered water molecules from a binding site, unoccupied by non-macrocyclic inhibitors, into bulk solvent. This is generally assumed to provide a second favorable entropic contribution (classical hydrophobic effect) (Mallinson J.M. and Collins I. (2012) Future Med Chem), leading to enhanced potencies of these inhibitors on their target protein.

Now, the inventors have found that compounds according to the invention, which comprise a macrocyclic moiety in addition to active binding parts, provide an improved binding activity towards one or more of BTK, LCK, FGFR1 , FLT3, PDGFR-β, FMS, LYN, MEK1 , AUR-B, ITK, VEGFR, EGFR TEC, ABL, AXL, c-MET, FGFR3, IGFR1 , RET, SRC and YES including mutants of some of these kinases, as compared to similar compounds, which provide a binding activity, but do not contain a macrocycle.

Embodiments

Compounds of the invention

Compounds of the invention are according to Formula (l-a) to (l-h): wherein said compound is as further described below, or a pharmaceutically acceptable salt and/or solvate thereof. The inventors have found that these compounds being a macrocyclic compound having any one of the scaffolds of Formula (l-a) to (l-h) provide improved kinase inhibition.

In a first aspect of the invention is provided a compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt and/or solvate thereof, wherein the compound is selected from the group consisting of: wherein R 1 is wherein :

W is a direct bond or an aryl group having 6-10 carbon or a heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;

V is selected from the group consisting of: a direct bond, O, -OCH 2 - , -CH(R 1v )-, -C(O)-, -C(O)-N(R 2v )-, -N(R 2V )-C(O)-,-NH-C(O)-NH-, -NH-C(O)-C(R 3V )(R 4v )-C(O)-NH-, -NH-SO 2 -, -NH-C(O)-O-, -CH(R 1v )-NH-C(O)-, -CH(R 1v )-C(O)-NH-, -C≡C-, and -CH 2 O- ; R 1v is hydrogen or (1 -2C)alkyl; R 2v is hydrogen or (1 -2C)alkyl;

R 3V is hydrogen or (1 -2C)alkyl; R 4v is hydrogen or (1 -2C)alkyl; or R 3v and R 4v form together with the carbon atom they are attached to a (3-6C)cycloalkyl;

U is an aryl group having 6-10 carbon, a heteroaryl group having 1-9 carbon or a cycloalkyl group having 3-6 carbon; wherein any of said aryl group, heteroaryl group and cycloalkyl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1-6C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R 2 is of Formula (I l-a) to (ll-f) selected from the group consisting of: wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X 1 , X 2 and X 3 are independently selected from CH 2 , -CH 2 CH 2 -, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with one or more substituents selected from halogen, hydroxy, (1 -3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; wherein R 3 and R 4 together represent a linker having Formula (III-1 to III-40) selected from the group consisting of: whereby the marks the position of R 3 in any one of Formula l-a to l-h, and whereby the marks the position of R 4 in any one of Formula I l-a to ll-f; wherein any of said linkers is optionally and independently substituted with one or more substituents selected from deuterium, halogen, oxo, hydroxy, CD 3 , (1-4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl, (3-6C)cycloalkoxy or (1- 6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein R 5 is hydrogen, NH 2 or Methyl.

(R 1 )

In an aspect of the invention the R 1 of the compounds of the invention has the formula: wherein:

W is a direct bond or an aryl group having 6-10 carbon or a heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;

V is selected from the group consisting of: a direct bond, O, -OCH 2 - , -CH(R 1v )-, -C(O)-, -C(O)-N(R 2v )-, -N(R 2V )-C(O)-,-NH-C(O)-NH-, -NH-C(O)-C(R 3v )(R 4v )-C(O)-NH-, -NH-SO 2 -, -NH-C(O)-O-, -CH(R 1v )-NH- C(O)-, -CH(R 1v )-C(O)-NH-, -C≡C-, and -CH 2 O- ; R 1v is hydrogen or (1 -2C)alkyl; R 2v is hydrogen or (1 -2C)alkyl; R 3v is hydrogen or (1 -2C)alkyl; R 4v is hydrogen or (1 -2C)alkyl; or

R 3V and R 4v form together with the carbon atom they are attached to a (3-6C)cycloalkyl;

U is an aryl group having 6-10 carbon, a heteroaryl group having 1-9 carbon or a cycloalkyl group having 3-6 carbon; wherein any of said aryl group, heteroaryl group and cycloalkyl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1-6C)alkyl, (1-6C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen;

In preferred embodiments, said heteroaryl group of W or U is a bicyclic heteroaryl group having 7-9 carbon.

In embodiments, R 1 is:

, wherein:

W is an aryl group having 6-10 carbon or a heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and

V is a direct bond.

In a preferred embodiment, R 1 is:

, wherein: the phenyl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and

U is an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1 -6C)alkyl, (1-6C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

Said R 1 is suitable for inhibiting various protein kinases, including AUR-B, EGFR, FLT3, FMS, VEGFR, PDGFR-β , ITK, LCK, ABL, AXL, c-MET, IGFR1 , RET, SRC, and YES.

In particular embodiments, V is any one of: -OCH 2 - , -C(O)-N(R 2v )-, -N(R 2v )-C(O)-,-NH-C(O)-NH-, -NH- C(O)-C(R 3V )(R 4v )-C(O)-NH-, -NH-SO 2 -, -NH-C(O)-O-, -CH(R 1v )-NH-C(O)-, -CH(R 1v )-C(O)-NH- ; R 1v is hydrogen or (1 -2C)alkyl; R 2v is hydrogen or (1 -2C)alkyl;

R 3V is hydrogen or (1 -2C)alkyl; R 4v is hydrogen or (1 -2C)alkyl; or R 3v and form together with the carbon atom they are attached to a (3-6C)cycloalkyl.

In more preferred embodiments, R 1 is any one of: wherein: the phenyl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and

U is an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon, wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1 -6C)alkyl, (1-6C)alkoxy, (3-6C)cycloalkyl and (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In another preferred embodiment, R 1 is:

In said preferred embodiment: W is a direct bond and V is a direct bond, and U is a heteroaryl group having 1-9 carbon, wherein said heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -6C)alkyl, and (1-6C)alkoxy.

In particular preferred embodiments, said heteroaryl group is a bicyclic heteroaryl group having 7-9 carbon, wherein more preferably R 1 is selected from any one of:

In another preferred embodiment, R 1 is: , wherein:

R 1w is selected from: hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy), (3-6C)cycloalkyl, (6-10C)aryl, and (1-5C)heteroaryl; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and

V is selected from the group consisting of: a direct bond, -CH(R 1v )-, -CH(R 1v )-NH-C(O)-, -CH 2 O- ; R 1v is hydrogen or (1 -2C)alkyl.

Said R 1 is suitable for inhibiting various protein kinases, including RET.

In particular embodiments, R 1 is: wherein : wherein R 1w is selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy, (3-6C)cycloalkyl, (6- 10C)aryl, and (1-5C)heteroaryl; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, (1 -2C)alkyl, (1- 2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and

V is a direct bond.

In another preferred embodiment, R 1 is: wherein:

W is a direct bond; and U is hydrogen or an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C) aryl, (1-5C)heteroaryl, (1 -6C)alkyl, (1- 6C)alkoxy, (3-6C)cycloalkyl and (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen. Said R 1 is suitable for inhibiting various protein kinases, including FGFR.

In another preferred embodiment, R 1 is:

W is a direct bond, and

U is hydrogen or an aryl group having 6-10 carbon or an heteroaryl group having 1-9 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (6-10C)aryl, (1-5C)heteroaryl, (1 -6C)alkyl, (1- 6C)alkoxy, (3-6C)cycloalkyl and (3-6C)heterocycloalkyl; wherein any of said aryl, heteroaryl, alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

Said R 1 is suitable for inhibiting various protein kinases, including EGFR.

In other preferred embodiments, R 1 is selected from the group consisting of: wherein:

R 1w and R 2w are independently selected from hydrogen, halogen, (1 -2C)alkyl, and (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;

V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R 1v )-NH-C(O)-, -CH(R 1v )- ;

R 1v is hydrogen or (1-2C)alkyl; and

U is an aryl group having 6-10 carbon or an heteroaryl group having 1-5 carbon; wherein any of said aryl group and heteroaryl group is optionally and independently substituted with one or more substituents selected from halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl and (3- 6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

Said R 1 is suitable for inhibiting various protein kinases, including BTK, mutant BTK C481 S, TEC, LCK, LYN, EGFR, ITK, AURB, VEGFR and MEK1.

In particular embodiments, R 1 is selected from the group consisting of: wherein:

R 1w and R 2w are independently selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro;

V is any one of O, -C(O)-NH-, -NH-C(O)-, -CH(R 1v )-NH-C(O)-, -CH(R 1v )- ;

R 1v is hydrogen or (1 -2C)alkyl; wherein R 1u and R 2u are independently selected from hydrogen, halogen, cyano, (1-4C)alkyl, (1- 5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein X u is selected from CH and N.

In particular embodiments, V is any one of O, -C(O)-NH-, -CH(R 1v )-NH-C(O)-, and -CH(R 1v )-; wherein R 1v is hydrogen or (1 -2C)alkyl.

In more preferred embodiments, R 1 is selected from the group consisting of: wherein R 1w and R 2w are independently selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; wherein R 1u and R 2u are independently selected from hydrogen, halogen, cyano, (1-4C)alkyl, (1- 5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen; and wherein X u is selected from CH and N.

In even more preferred embodiments, R 1 is selected from the group consisting of: wherein R 2w is selected from hydrogen, halogen, (1 -2C)alkyl, (1-2C)alkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro; and wherein R 3u is selected from hydrogen, halogen, cyano, (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro.

In most preferred embodiments, R 1 is: wherein R 2w is selected from hydrogen, fluoro, methyl or methoxy; wherein R 3u is selected from hydrogen, halogen, cyano, (1 -4C)alkyl, (1-2C)alkoxy, (3-6C)cycloalkyl or (3-6C)heterocycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three fluoro.

(R 2 )

In an aspect of the invention the R 2 of the compounds of the invention is of Formula (ll-a) to (ll-f) selected from the group consisting of: wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X 1 , X 2 and X 3 are independently selected from CH 2 , -CH 2 CH 2 -, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with one or more substituents selected from halogen, hydroxy, (1 -3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In embodiments, wherein R 2 is selected from the group consisting of: wherein Q is a monocyclic ring selected from a (3-7C)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X 1 , X 2 and X 3 are independently selected from CH 2 , -CH 2 CH 2 -, O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with halogen, hydroxy, (1 -3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In embodiments, wherein R 2 is selected from the group consisting of: wherein Q is a monocyclic ring selected from a (3-7)cycloalkyl and a (3-6C)heterocycloalkyl, wherein X 1 , X 2 and X 3 are independently selected from CH 2 , O, N and a direct bond; wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with halogen, hydroxy, (1 -3C)alkyl, (1-3C)alkoxy, (1-4C)alkylcarbonyl or (3-4C)cycloalkyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In preferred embodiments, at least one of the X 1 , X 2 and X 3 of R 2 is a nitrogen atom forming a secondary amine group, wherein the amine group is substituted by (1-4C)alkylcarbonyl, preferably by methylcarbonyl or ethylcarbonyl.

In preferred embodiments, R 2 is selected from the group consisting of: wherein any of said cycloalkyl, heterocycloalkyl and alkyl group is optionally and independently substituted with hydroxy, methyl or methoxy.

(Linker represented by R 3 and R 4 )

Each of the compounds of the invention comprises the linker represented by R 3 and R 4 . The linker is a part of the macrocycle of each of the compounds of the invention.

In the given Formulas of the linkers having Formula (III-1) to (III-40), the (i.e. the wavy with a star) marks the position of R 3 in any one of Formula l-a to l-h, and the (i.e. the wavy without a star) marks the position of R 4 in any one of Formula I l-a to ll-f. Thus, the linker is directly connected to the scaffold of any one of Formula l-a to l-h at the position of the (i.e. the wavy with a star).

The macrocycle is formed by the connections between the linker, R 2 and the scaffold of Formula (l-a) to (l-h). R 2 is directly connected to the linker at the position of R 4 . The scaffold of Formula (l-a) to (l-h) is connected to the linker, at another end of the linker, at the position of R 3 . The scaffold is a bicycle shown in the compounds of Formula (l-a) to (l-f), or the scaffold is a monocycle shown in the compounds of Formula (l-g) to (l-h).

In embodiments, the macrocycle may comprise at least 12 atoms forming said macrocycle, and may comprise any number of atoms from 12 - 18 forming said macrocycle, preferably from 13 - 15 atoms forming said macrocycle.

In an embodiment, wherein the linker represented by R 3 and R 4 is selected from the group consisting of:

whereby the marks the position of R 3 in any one of Formula l-a to l-h, and whereby the marks the position of R 4 in any one of Formula I l-a to I l-f; wherein any of said linker is optionally and independently substituted with one or more groups selected from deuterium, halogen, oxo, hydroxy, CD 3 , (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl, (3- 6C)cycloalkoxy or (1-6C)alkylcarbonyl; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In a preferred embodiment, the linker represented by R 3 and R 4 is selected from the group consisting of:

whereby the marks the position of R 3 in any one of Formula l-a to l-h, and whereby the marks the position of R 4 in any one of Formula I l-a to I l-f; wherein any of said linker is optionally and independently substituted with one or more substituents selected from deuterium, hydroxy, CD 3 , (1 -4C)alkyl, (1-5C)alkoxy, (3-6C)cycloalkyl or (3- 6C)cycloalkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In a more preferred embodiment, the linker represented by R 3 and R 4 is selected from the group consisting of:

whereby the marks the position of R 3 in any one of Formula l-a to l-h, and whereby the marks the position of R 4 in any one of Formula I l-a to I l-f; wherein any of said linker is optionally and independently substituted with one or more substituents selected from deuterium, hydroxy, CD 3 , (1 -2C)alkyl, (1-2C)alkoxy, (3-6C)cycloalkyl or (3- 6C)cycloalkoxy; wherein any of said alkyl and alkoxy group is optionally and independently substituted with one, two or three halogen.

In preferred embodiments, a secondary amine group of the linker represented by R 3 and R 4 is substituted by (1-6C)alkylcarbonyl, preferably by methylcarbonyl or ethylcarbonyl. In examples, the secondary amine group of any one of Formula (III-19) to (III-23) or (III-38) may be substituted by (1- 4C)alkylcarbonyl, such as by methylcarbonyl or ethylcarbonyl.

In preferred embodiments, a carbon group of the linker represented by R 3 and R 4 is substituted by (1- 4)alkyl, preferably by methyl or ethyl, thereby providing a tertiary carbon group.

(scaffold)

The compounds of the invention have a scaffold according to any one of Formula (l-a) to (l-h). BTK inhibitors which do not contain a macrocycle are generally known from the prior art, wherein said known BTK inhibitors have a scaffold according to any one of Formula (l-a) to (l-h):

See for compounds having a scaffold of Formula l-a: WO 2013/010380, W02016/210165;

See for compounds having a scaffold of Formula l-b: Boga S B et al (2017) Bioorg Med Chem Lett, 27, 3939-3943; Liu J et al (2016) ACS Med Chem Lett, 6 198-203;

See for compounds having a scaffold of Formula l-c: WO 2013/010380;

See for compounds having a scaffold of Formula l-d: BBA - General Subjects 1864 (2020) 129531 ;

See for compounds having a scaffold of Formula l-e: WO 2015/058084, WO2015/095099, WO2015/095102;

See for compounds having a scaffold of Formula l-f: WO 20130/81016;

See for compounds having a scaffold of Formula l-g: WO 2017/106429, WO 2019/091441 , WO 2017/103611 ;

See for compounds having a scaffold of Formula l-h: WO 2014/082598, WO 2014/025976.

Furthermore a review showing that various scaffolds have been used in BTK inhibitor compounds is provided in: Yifan Feng, Weiming Duan, Xiaochuan Cu, Chengyuan Liang & Minhang Xin (2019) Bruton's tyrosine kinase (BTK) inhibitors in treating cancer: a patent review (2010-2018), Expert Opinion on Therapeutic Patents, 29:4, 217-241.

All these prior art documents demonstrate that compounds providing BTK inhibition are found for each of the scaffolds according to any one of Formula (l-a) to (l-h).

The present invention concerns novel compounds having a scaffold according to any one of Formula (l-a) to (l-h), and further having a macrocycle as defined according to the embodiments of the invention.

In preferred embodiments, the compound comprises a bicyclic scaffold selected from: wherein R 5 is hydrogen, NH 2 or Methyl.

In more preferred embodiment, wherein the compound comprises a bicyclic scaffold according to Formula (l-a) to (l-f) selected from the group consisting of:

In an even more preferred embodiments, the compound comprises a bicyclic scaffold selected from:

In an alternative embodiment, wherein the compound comprises a monocyclic scaffold according to Formula (l-g) to (l-h) selected from the group consisting of:

In an alternative embodiment, wherein the compound comprises a scaffold according to any one of

Formula (l-a), Formula (l-b), Formula (l-c), Formula (l-d), Formula (l-e), Formula (l-f), Formula (l-g). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-a), Formula (l-b), Formula (l-c), Formula (l-d), Formula (l-e), Formula (l-f), Formula (l-h). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-a), Formula (l-b), Formula (l-c), Formula (l-d), Formula (l-e), Formula (l-g), Formula (l-h). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-a), Formula (l-b), Formula (l-c), Formula (l-d), Formula (l-f), Formula (l-g), Formula (l-h). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-a), Formula (l-b), Formula (l-c), Formula (l-e), Formula (l-f), Formula (l-g), Formula (l-h). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-a), Formula (l-b), Formula (l-d), Formula (l-e), Formula (l-f), Formula (l-g), Formula (l-h). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-a), Formula (l-c), Formula (l-d), Formula (l-e), Formula (l-f), Formula (l-g), Formula (l-h). In an alternative embodiment, wherein the compound comprises a scaffold according to any one of Formula (l-b), Formula (l-c), Formula (l-d), Formula (l-e), Formula (l-f), Formula (l-g), Formula (l-h). (specific compounds)

In an embodiment, wherein the compound has a sub-formula (1 - 62) selected from the group consisting of:

In a preferred embodiment, said compound is selected from the group consisting of:

Said preferred compounds are suitable for inhibiting Bruton's Tyrosine Kinase (BTK).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Bruton's Tyrosine Kinase C481S mutant (BTK C481S). In another preferred embodiment, said compound is selected from the group consisting of: Said compounds are suitable for inhibiting Aurora B Kinase (Aur-B).

In another preferred embodiment, said compound is selected from the group consisting of: Said compound are suitable for inhibiting Epidermal Growth Factor Receptor (EGFR).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Fibroblast Growth Factor Receptor 1 (FGFR1).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting FMS related tyrosine kinase 3 (FLT-3).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Colony Stimulating Factor 1 Receptor (FMS).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting IL2 inducible T cell kinase (ITK). In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Lymphocyte-Specific Protein Tyrosine Kinase (LCK). In another preferred embodiment, said compound is selected from the group consisting of:

Said compound is suitable for inhibiting LCK/YES Novel Tyrosine Kinase (LYN). In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Platelet Derived Growth Factor Receptor Beta (PDGFR-β). In another preferred embodiment, said compound is selected from the group consisting of: Said compounds are suitable for inhibiting TEC Protein Tyrosine Kinase (TEC).

In another preferred embodiment, said compound is selected from the group consisting of: Said compound are suitable for inhibiting Vascular Endothelial Growth Factor Receptor (VEGFR).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compound is suitable for inhibiting Mitogen-Activated Protein Kinase Kinase 1 (MEK1). In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Abelson Tyrosine Kinase (ABL).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Axl Receptor Tyrosine Kinase (AXL). In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting hepatocyte growth factor receptor or tyrosine-protein kinase Met (c-MET).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting Fibroblast Growth Factor Receptor 3 (FGFR3). In another preferred embodiment, said compound is selected from the group consisting of: Said compound is suitable for inhibiting Insuline Like Growth Factor Receptor 1 (IGFR1).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting “REarranged during Transfection” receptor tyrosine kinase (RET).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting non-receptor tyrosine kinase Src (SRC).

In another preferred embodiment, said compound is selected from the group consisting of:

Said compounds are suitable for inhibiting non-receptor tyrosine kinase Yes (YES).

Pharmaceutical composition

Pharmaceutical compositions in accordance with the present invention comprise, as the active ingredient (‘API'), compound of Formula (l-a) to (l-h) or a pharmaceutically acceptable salt, hydrate or solvate thereof.

As used herein, “a pharmaceutically acceptable salt” includes any salt that retains the activity of the active agent(s) and is acceptable for pharmaceutical use. A pharmaceutically acceptable salt also refers to any salt which may form in vivo as a result of administration of an acid, another salt, or a prodrug which is converted into an acid or salt. Preferably, the pharmaceutically acceptable salt is the HCI-salt of the compound of the invention. The pharmaceutically acceptable salt of the disclosed compounds may be prepared by methods of pharmacy well known to those skilled in the art.

Furthermore, the compositions can comprise compounds according to the invention in the form of a solvate, comprising a pharmaceutically acceptable solvent, such as water ('hydrate'), ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.

As used herein, the term “pharmaceutical composition” refers to a composition comprising a compound according to the invention or a salt or solvate thereof and, as the case may be, one or more additional, non-toxic ingredients, which composition is in a form suitable for administration to a (human) subject, through any route of administration, and which composition is physiologically tolerated upon such administration.

The compositions of the invention may thus comprise one or more additional ingredients. In a preferred embodiment, the composition comprises one or more carriers and/or excipients. As is known by those of average skill in the art, the appropriate choice of excipients is dependent on multiple factors, including the physicochemical properties of the API, the preferred pharmaceutical form, the preferred route of administration, the desired rate of release, etc. The compositions of the invention can be formulated for a variety of routes of administration, oral administration being particularly preferred. It is within the purview of those of average skill in the art to conceive and develop suitable formulations, relying on the common general knowledge as reflected in text books such as Remington's Pharmaceutical Sciences (Meade Publishing Co., Easton, Pa., 20.sup.th Ed., 2000), the entire disclosure of which is herein incorporated by reference, and routine development efforts.

In accordance with the various aspects of the invention, the composition is preferably provided in a unit dosage form. The term ‘unit dosage form' refers to a physically discrete unit suitable as a unitary dosage for human subjects, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with any suitable pharmaceutical carrier(s) and/or excipient(s). Exemplary, non-limiting unit dosage forms include a tablet (e.g., a chewable tablet), caplet, capsule (e.g., a hard capsule or a soft capsule), lozenge, film, strip, gelcap as well as any metered volume of a solution, suspension, syrup or elixir or the like, which may be contained, for instance in a vial, syringe, applicator device, sachet, spray, micropump etc. In accordance with particularly preferred embodiments of the invention, the unit dosage form, is a unit dosage form that is suitable for oral administration. Most preferably, it is a solid unit dosage form, such as a tablet.

Besides the compound according to the invention as such, pharmaceutically acceptable salts thereof may also be used. Pharmaceutically acceptable salts of compounds of the invention include the acid addition and base salts thereof, such as preferably the calcium, potassium or sodium salts. For a review on suitable salts, reference is made “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).

Pharmaceutically acceptable salts of compounds according to the invention may be readily prepared by mixing together solutions of compounds according to the invention and the desired acid or base, as appropriate. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionisation in the salt may vary from completely ionised to almost non-ionised.

Medical Use

The compounds and the pharmaceutical compositions of the present invention are useful as inhibitors of kinases, in particular tyrosine kinases. In particular, compounds of this invention are useful as inhibitors of tyrosine kinases that are important in hyper-proliferative diseases, especially in cancer and in the process of angiogenesis.

The compounds of the present invention are also useful in the treatment of cancer related indications such as solid tumors, sarcomas (especially Ewing's sarcoma and osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic malignancies, including leukaemia and lymphoma, tumor-induced pleural or pericardial effusions, and malignant ascites.

The compounds according to the invention having Formula (l-a) to (l-h) and pharmaceutical compositions thereof can be used to treat or prevent a variety of conditions, diseases or disorders mediated by any one of the kinases and mutants of these kinases: BTK, LCK, FGFR1 , FLT3, PDGFR- β, FMS, LYN, MEK1 , AUR-B, ITK, VEGFR, EGFR, TEC, ABL, AXL, c-MET, FGFR3, IGFR1 , RET, SRC and YES. Such conditions, diseases or disorders include: (1) arthritis, including rheumatoid arthritis, juvenile arthritis, psoriatic arthritis and osteoarthritis; (2) asthma and other obstructive airways diseases, including chronic asthma, late asthma, airway hyper-responsiveness, bronchitis, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, adult respiratory distress syndrome, recurrent airway obstruction, and chronic obstruction pulmonary disease including emphysema; (3) autoimmune diseases or disorders, including those designated as single organ or single cell-type autoimmune disorders, for example Hashimoto's thyroiditis, autoimmune hemolytic anemia, autoimmune atrophic gastritis of pernicious anemia, autoimmune encephalomyelitis, autoimmune orchitis, Goodpasture's disease, autoimmune thrombocytopenia including idiopathic thrombopenic purpura, sympathetic ophthalmia, myasthenia gravis. Graves' disease, primary biliary cirrhosis, chronic aggressive hepatitis, ulcerative colitis and membranous glomerulopathy, those designated as involving systemic autoimmune disorder, for example systemic lupus erythematosis, immune thrombocytopenic purpura, rheumatoid arthritis, Sjogren's syndrome, Reiter's syndrome, polymyositis-dermatomyositis, systemic sclerosis, polyarteritis nodosa, multiple sclerosis and bullous pemphigoid, and additional autoimmune diseases, which can be B-cell (humoral) based or T-cell based, including Cogan's syndrome, ankylosing spondylitis, Wegener's granulomatosis, autoimmune alopecia, Type I or juvenile onset diabetes, and thyroiditis;

(4) cancers or tumors, including alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer including mast cell tumor and squamous cell carcinoma, breast and mammary cancer, ovarian cancer, prostate cancer, lymphoma and leukemia (including but not limited to acute myelogenous leukemia, chronic myelogenous leukemia, mantle cell lymphoma, NHL B cell lymphomas (e.g. precursor B-ALL, marginal zone B cell lymphoma, chronic lymphocytic leukemia, diffuse large B cell lymphoma, Burkitt lymphoma, mediastinal large B-cell lymphoma), Hodgkin lymphoma, NK and T cell lymphomas; TEL-Syk and ITK-Syk fusion driven tumors, myelomas including multiple myeloma, myeloproliferative disorders kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer, brain cancer, melanoma including oral and metastatic melanoma, Kaposi's sarcoma, proliferative diabetic retinopathy, and angiogenic-associated disorders including solid tumors, and pancreatic cancer.

(5) diabetes, including Type I diabetes and complications from diabetes; (6) eye diseases, disorders or conditions including autoimmune diseases of the eye, keratoconjunctivitis, vernal conjunctivitis, uveitis including uveitis associated with Behcet's disease and lens-induced uveitis, keratitis, herpetic keratitis, conical keratitis, corneal epithelial dystrophy, keratoleukoma, ocular premphigus, Mooren's ulcer, scleritis, Grave's ophthalmopathy, Vogt-Koyanagi-Harada syndrome, keratoconjunctivitis sicca (dry eye), phlyctenule, iridocyclitis, sarcoidosis, endocrine ophthalmopathy, sympathetic ophthalmitis, allergic conjunctivitis, and ocular neovascularization; (7) intestinal inflammations, allergies or conditions including Crohn's disease and/or ulcerative colitis, inflammatory bowel disease, coeliac diseases, proctitis, eosinophilic gastroenteritis, and mastocytosis; (8) neurodegenerative diseases including motor neuron disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, cerebral ischemia, or neurodegenerative disease caused by traumatic injury, strike, glutamate neurotoxicity or hypoxia; ischemic/ reperfusion injury in stroke, myocardial ischemica, renal ischemia, heart attacks, cardiac hypertrophy, atherosclerosis and arteriosclerosis, organ hypoxia; (9) platelet aggregation and diseases associated with or caused by platelet activation, such as arteriosclerosis, thrombosis, intimal hyperplasia and restenosis following vascular injury; (10) conditions associated with cardiovascular diseases.

For the treatment of cancer a compound of the invention may be combined with one or more of an anticancer agents. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Heilman (editors), 6 th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.

BTK inhibition

BTK inhibition is a novel approach for treating many different human diseases associated with the inappropriate activation of B-cells, including B-cell proliferative disorders, B-cell malignancies, immunological disease for example autoimmune and inflammatory disorders.

In embodiments the condition treatable by inhibition of BTK may be selected from: cancer, lymphoma, leukemia, autoimmune diseases, inflammatory disorders, heteroimmune conditions, or fibrosis. Specific conditions treatable by the inhibition of BTK may be selected from: B-cell malignancy, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocyte leukemia, non-Hodgkin lymphoma for example ABC-DLBCL, mantle cell lymphoma, follicular lymphoma, hairy cell leukemia B-cell non- Hodgkin lymphoma, Waldenstrom's macroglobulinemia, Richter transformation, multiple myeloma, bone cancer, bone metastasis, follicular lymphoma, chronic lymphocytic lymphoma, B-cell prolymphocyte leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell lymphoma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, lymphomatoid granulomatosis, inflammatory bowel disease, arthritis, lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves'disease, Sjogren's syndrome, multiple sclerosis, Guillain-Barr syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylitisis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, coeliac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, endometriosis, interstitial cystitis, neuromyotonia, scleroderma, vulvodynia, graft versus host disease, transplantation, transfusion, anaphylaxis, allergy, type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, atopic dermatitis, asthma, appendicitis, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vulvitis, pulmonary fibrosis, idiopathic pulmonary fibrosis (IPF), usual interstitial pneumonitis (UIP), interstitial lung disease, cryptogenic fibrosing alveolitis (CFA), bronchiolitis obliterans, bronchiectasis, fatty liver disease, steatosis (e.g., nonalcoholic steatohepatitis (NASH)), cholestatic liver disease (e.g., primary biliary cirrhosis (PBC)), cirrhosis, alcohol-induced liver fibrosis, biliary duct injury, biliary fibrosis, cholestatis or cholangiopathies. In some embodiments, hepatic or liver fibrosis includes, but is not limited to, hepatic fibrosis associated with alcoholism, viral infection, e.g., hepatitis (e.g., hepatitis C, B or D), autoimmune hepatitis, nonalcoholic fatty liver disease (NAFLD), progressive massive fibrosis, exposure to toxins or irritants (e.g., alcohol, pharmaceutical drugs and environmental toxins), renal fibrosis (e.g., chronic kidney fibrosis), nephropathies associated with injury/fibrosis (e.g., chronic nephropathies associated with diabetes (e.g., diabetic nephropathy)), lupus, scleroderma of the kidney, glomerular nephritis, focal segmental glomerular sclerosis, IgA nephropathyrenal fibrosis associated with human chronic kidney disease (CKD), chronic progressive nephropathy (CPN), tubulointerstitial fibrosis, ureteral obstruction, chronic uremia, chronic interstitial nephritis, radiation nephropathy, glomerulosclerosis, progressive glomerulonephrosis (PGN), endothelial/thrombotic microangiopathy injury, HIV-associated nephropathy, or fibrosis associated with exposure to a toxin, an irritant, or a chemotherapeutic agent, fibrosis associated with scleroderma, radiation induced gut fibrosis, fibrosis associated with a foregut inflammatory disorder such as Barrett's esophagus and chronic gastritis, and/or fibrosis associated with a hindgut inflammatory disorder, such as inflammatory bowel disease (IBD), ulcerative colitis and Crohn's disease, age-related macular degeneration, diabetic retinopathy, retinopathy of prematurity and neovascular glaucoma.

In embodiments the condition treatable by the inhibition of BTK may be selected from: cancer, lymphoma, leukemia, autoimmune diseases and inflammatory disorders. Specific conditions treatable by the inhibition of BTK may be selected from: B-cell malignancy, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocyte leukemia, non-Hodgkin lymphoma for example ABC-DLBL, mantle cell lymphoma, follicular lymphoma, hairy cell leukemia B-cell non-Hodgkin lymphoma, Waldenstrom's macroglobulinemia, Richter transformation, multiple myeloma, bone cancer, bone metastasis, arthritis, multiple sclerosis, osteoporosis, irritable bowel syndrome, inflammatory bowel disease, Crohn's disease, lupus and Sjogren's syndrome.

B-cell malignancy, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocyte leukemia, non-Hodgkin lymphoma for example ABC-DLBCL, mantle cell lymphoma, follicular lymphoma, hairy cell leukemia B-cell non-Hodgkin lymphoma, Waldenstrom's macroglobulinemia, Richter transformation, multiple myeloma, bone cancer, bone metastasis, chronic lymphocytic lymphomas, B-cell prolymphocyte leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell lymphoma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, and lymphomatoid granulomatosis are examples of cancer, lymphoma and leukemia treatable by BTK inhibition.

B-cell malignancy, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocyte leukemia, non-Hodgkin lymphoma for example ABC-DLBCL, mantle cell lymphoma, follicular lymphoma, hairy cell leukemia B-cell non-Hodgkin lymphoma, Waldenstrom's macroglobulinemia, Richter transformation, multiple myeloma, bone cancer and bone metastasis are examples of cancer, lymphoma and leukemia treatable BTK inhibition.

Arthritis, multiple sclerosis, osteoporosis, irritable bowel syndrome, inflammatory bowel disease, Crohn's disease, lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, Sjogren's syndrome, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylitis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, coeliac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, endometriosis, interstitial cystitis, neuromyotonia, scleroderma, vulvodynia, asthma, appendicitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, vulvitis, graft versus host disease, transplantation, transfusion, anaphylaxis, allergy, type I hypersensitivity, allergic conjunctivitis, allergic rhinitis and atopic dermatitis are examples of immunological diseases treatable by BTK inhibition.

Arthritis, asthma, appendicitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, and vulvitis are examples of an inflammatory disorder treatable by BTK inhibition.

Lupus, rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, Ord's thyroiditis, Graves' disease, Sjogren's syndrome, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus-myoclonus syndrome, ankylosing spondylitis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, coeliac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, endometriosis, interstitial cystitis, neuromyotonia, scleroderma, and vulvodynia are examples of an autoimmune disease treatable by BTK inhibition.

Graft versus host disease, transplantation, transfusion, anaphylaxis, allergy, type I hypersensitivity, allergic conjunctivitis, allergic rhinitis and atopic dermatitis are examples of heteroimmune condition treatable by BTK inhibition.

Pulmonary fibrosis, idiopathic pulmonary fibrosis (IPF), usual interstitial pneumonitis (UIP), interstitial lung disease, cryptogenic fibrosing alveolitis (CFA), bronchiolitis obliterans, bronchiectasis, fatty liver disease, steatosis (e.g., nonalcoholic steatohepatitis (NASH)), cholestatic liver disease (e.g., primary biliary cirrhosis (PBC)), cirrhosis, alcohol-induced liver fibrosis, biliary duct injury, biliary fibrosis, cholestatis or cholangiopathies. In some embodiments, hepatic or liver fibrosis includes, but is not limited to, hepatic fibrosis associated with alcoholism, viral infection, e.g., hepatitis (e.g., hepatitis C, B or D), autoimmune hepatitis, nonalcoholic fatty liver disease (NAFLD), progressive massive fibrosis, exposure to toxins or irritants (e.g., alcohol, pharmaceutical drugs and environmental toxins), renal fibrosis (e.g., chronic kidney fibrosis), nephropathies associated with injury/fibrosis (e.g., chronic nephropathies associated with diabetes (e.g., diabetic nephropathy)), lupus, scleroderma of the kidney, glomerular nephritis, focal segmental glomerular sclerosis, IgA nephropathyrenal fibrosis associated with human chronic kidney disease (CKD), chronic progressive nephropathy (CPN), tubulointerstitial fibrosis, ureteral obstruction, chronic uremia, chronic interstitial nephritis, radiation nephropathy, glomerulosclerosis, progressive glomerulonephrosis (PGN), endothelial/thrombotic microangiopathy injury, HIV-associated nephropathy, or fibrosis associated with exposure to a toxin, an irritant, or a chemotherapeutic agent, fibrosis associated with scleroderma, radiation induced gut fibrosis, fibrosis associated with a foregut inflammatory disorder such as Barrett's esophagus and chronic gastritis, and/or fibrosis associated with a hindgut inflammatory disorder, such as inflammatory bowel disease (IBD), ulcerative colitis and Crohn's disease, age-related macular degeneration, diabetic retinopathy, retinopathy of prematurity and neovascular glaucoma are examples of fibrosis treatable by BTK inhibition.

Arthritis, multiple sclerosis, osteoporosis, irritable bowel syndrome, inflammatory bowel disease, Crohn's disease and lupus are examples of immunological diseases treatable by BTK inhibition. Arthritis is an examples of an inflammatory disorder treatable by BTK inhibition. Lupus and Sjogren's syndrome are examples of autoimmune diseases treatable by BTK inhibition. Any of the conditions disclosed above as being treatable by BTK inhibition may be treated by a compound of the invention, or may be treated in a method comprising administering a compound of the invention, or may be treated by a medication manufactured through the use of a compound of the present invention.

Aurora B - inhibition

Known Aurora B mediated disorders include cancer.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Aurora B mediated disorders, in particular used in the treatment of cancer.

EGFR - inhibition

Known Epidermal Growth Factor Receptor (EGFR) mediated disorders include cancer.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Epidermal Growth Factor Receptor (EGFR) mediated disorders, in particular for use in the treatment of cancer, wherein preferably the cancer is selected from a lung cancer, non-small cell lung cancer, a pancreatic cancer, a colon cancer, a breast cancer, colorectal cancer, a prostate cancer, a head and neck cancer, an ovarian cancer, a brain cancer, a kidney carcinoma, pancreatic cancer, ovarian cancer, gastric cancer, glioma or prostate cancer.

FGFR1- inhibition

Known Fibroblast Growth Factor Receptor 1 (FGFR1) mediated disorders include cancer.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Fibroblast Growth Factor Receptor 1 (FGFR1) mediated disorders, in particular for use in the treatment of cancer is selected from brain cancer, head and neck cancer, gastric cancer or ovarian cancer.

FLT-3- inhibition

Known FMS related tyrosine kinase 3 (FLT-3) mediated disorders include cancer.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating FMS related tyrosine kinase 3 (FLT-3) mediated disorders, in particular for use in the treatment of:

(i) a proliferative disease, such as cancer, preferably a proliferative disease selected from the group consisting of: presence or progression of solid tumor, sarcoma, lymphoma, B-cell lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B-cell lymphoma, intravascular large B-cell lymphoma, primary effusion lymphoma, Burkitt lymphoma, leukemias, chronic lymphocytic leukemia, B-cell prolymphocytic leukemia, plasma cell myeloma, plasmacytoma, lymphomatoid granulomatosis, melanoma, B-cell proliferative disease, brain cancer, kidney cancer, liver cancer, adrenal gland cancer, bladder cancer, breast cancer, breast ductal carcinoma, lobular carcinoma, stomach neoplasm, stomach cancer, esophagus cancer, ovarian cancer, colorectal cancer, prostate cancer, pancreas cancer, lung cancer, vagina cancer, membranous adenocarcinoma, thyroid cancer, neck cancer, CNS cancer, malignant glioma, myeloproliferative disease, glioblastoma, multiple myeloma, gastrointestinal cancer, colorectal carcinoma, head and neck neoplasms, brain tumor, epidermal hyperplasia, psoriasis, prostate hyperplasia, neoplasia, neoplasia of epithelial character, and a combination thereof;

(ii) a hematological malignancy selected from the group consisting of: myeloma, acute lymphocytic leukemia, acute myeloid leukemia, acute promyelocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, chronic neutrophilic leukemia, acute undifferentiated leukemia, anaplastic large-cell lymphoma, adult T-cell acute myelocytic leukemia, acute myelocytic leukemia with trilineage myelodysplasia, mixed lineage leukemia, myelodysplasia syndromes, myeloproliferative disorders, multiple myeloma, myeloid sarcoma and a combination thereof; the FMS related tyrosine kinase 3 (FLT- 3) mediated disorder is acute myeloid leukemia.

FMS- inhibition

Known Colony Stimulating Factor 1 Receptor (FMS) mediated disorders include cancer, cardiovascular diseases, inflammatory diseases, and autoimmune diseases, including chronic graft vs host disease.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Colony Stimulating Factor 1 Receptor (FMS) mediated disorders, in particular for use in the treatment of:

(i) Cancer, wherein cancer is selected from the group consisting of solid tumors, acute myeloid leukemia, myelodysplastic syndrome, acute lymphocytic leukemia, and chronic lymphocytic leukemia;

(ii) a disease selected from osteoporosis, Paget's disease, rheumatoid arthritis and other forms of inflammatory arthritis, osteoarthritis, prosthesis failure, osteolytic sarcoma, myeloma, and tumor metastasis to bone;

(iii) a disease selected from glomerulonephritis, inflammatory bowel disease, prosthesis failure, sarcoidosis, congestive obstructive pulmonary disease, idiopathic pulmonary fibrosis, asthma, pancreatitis, HIV infection, psoriasis, diabetes, tumor related angiogenesis, age-related macular degeneration, diabetic retinopathy, restenosis, schizophrenia and Alzheimer's dementia;

(iv) pain, including skeletal pain caused by tumor metastasis or osteoarthritis, or visceral, inflammatory, or neurogenic pain in a mammal;

(v) an autoimmune disease selected from the group consisting of systemic lupus erythematosus, rheumatoid arthritis and other forms of inflammatory arthritis, psoriasis, Sjogren's syndrome, multiple sclerosis, or uveitis.

ITK - inhibition

Known IL2 inducible T cell kinase (ITK) mediated disorders include cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating IL2 inducible T cell kinase (ITK) mediated disorders, in particular for use in the treatment of a disease selected from cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection.

LCK - inhibition

Known Lymphocyte-Specific Protein Tyrosine Kinase (LCK) mediated disorders include cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Lymphocyte-Specific Protein Tyrosine Kinase (LCK) mediated disorders, in particular for use in the treatment of:

(i) a disease or disorder mediated by immune cells selected from T lymphocytes, NK cells, B lymphocytes, e.g. acute or chronic rejection of organ or tissue allo- or xenografts, atheriosclerosis, vascular occlusion due to vascular injury such as angioplasty, restenosis, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, hypertension, heart failure, chronic obstructive pulmonary disease, CNS disease such as Alzheimer disease or amyotrophic lateral sclerosis, cancer, cholangiocarcinoma, cytokine release syndrome, lymphodepletion in combination with immunotherapy, such as immunotherapy using NK cells, infectious disease such as AIDS, septic shock or adult respiratory distress syndrome, ischemia/reperfusion injury e.g. myocardial infarction, stroke, gut ischemia, renal failure or hemorrhage shock, or traumatic shock;

(ii) a chronic T cell disorder like multiple sclerosis and rheumatoid arthritis, or an acute inflammatory disorder in which T cells play a prominent role including transplant rejection, atopic dermatitis and delayed type hypersensitivity.

LYN - inhibition Known LCK/YES Novel Tyrosine Kinase (LYN) mediated disorders include cancer, Type II diabetes, fibrosis, chronic inflammation, chronic pancreatitis, pancreatic fibrosis, or inflammatory myofibroblastic tumors (IMTs).

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating LCK/YES Novel Tyrosine Kinase (LYN) mediated disorders, in particular for use in the treatment of:

(i) cancer, Type II diabetes, fibrosis, chronic inflammation, chronic pancreatitis, pancreatic fibrosis, or inflammatory myofibroblastic tumors (IMTs);

(ii) a cancer disease selected from lymphomas, sarcomas, brain cancer, breast cancer, colon cancer, lung cancer, hepatocellular cancer, gastric cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, cancer of the urinary tract, thyroid cancer, renal cancer, carcinoma, melanoma, head and neck cancer, brain cancer, and prostate cancer. PDGFR-β - inhibition

Known Platelet Derived Growth Factor Receptor Beta (PDGFR-β) mediated disorders include cancer, vascular disorder and fibrotic diseases.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Platelet Derived Growth Factor Receptor Beta (PDGFR-β) mediated disorders, in particular for use in the treatment of:

(i) cancer, vascular disorder and fibrotic diseases;

(ii) wherein the cancer is selected from lung cancer, prostate cancer, renal cell carcinoma, chronic myelomonocytic leukemia (CMML) and glioblastoma;

(iii) wherein the vascular disorders is selected from atherosclerosis, restenosis and pulmonary hypertension.

TEC - inhibition

Known TEC Protein Tyrosine Kinase (TEC) mediated disorders include cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating TEC Protein Tyrosine Kinase (TEC) mediated disorders, in particular for use in the treatment of a disease selected from cancer, an inflammatory disease, an autoimmune disease or an pathogenic infection.

VEGFR - inhibition Known Vascular Endothelial Growth Factor Receptor (VEGFR) mediated disorders include cancer, angiogenesis and lymphangiogenesis, Parkinson's and Alzheimer's diseases, inflammatory diseases.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Vascular Endothelial Growth Factor Receptor (VEGFR) mediated disorders, in particular for use in the treatment of a disease selected from cancer, angiogenesis and lymphangiogenesis, Parkinson's and Alzheimer's diseases, inflammatory diseases.

MEK1 - inhibition

Known Mitogen-Activated Protein Kinase Kinase 1 (MEK1) mediated disorders include cancer, such as melanoma and non-small cell lung cancer (NSCLC).

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Mitogen-Activated Protein Kinase Kinase 1 (MEK1) mediated disorders, in particular for use in the treatment of cancer, such as melanoma and non-small cell lung cancer (NSCLC).

ABL - inhibition

Known Abelson Tyrosine Kinase (ABL) mediated disorders include cancers.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Abelson Tyrosine Kinase (ABL) mediated disorders, in particular for use in the treatment of cancer, wherein cancer is leukemia, myeloma or lymphoma.

In a preferred embodiment, the cancer is chronic myeloid leukemia (CML).

AXL - inhibition

Known Axl Receptor Tyrosine Kinase (AXL) mediated disorders include cancers.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Axl Receptor Tyrosine Kinase (AXL) mediated disorders, in particular for use in the treatment of cancer, wherein preferably the cancer is selected from hepatocellular cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, gastric cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, prostate cancer, esophageal cancer, gall bladder cancer, pancreatic cancer, thyroid cancer, skin cancer, leukemia, multiple myeloma, chronic lymphocytic lymphoma, adult T cell leukemia, B-cell lymphoma, acute myelogenous leukemia, Hodgkin's or non- Hodgkin's lymphoma, Waldenstrom's Macroglubulinemia, hairy cell lymphoma, Burkett's lymphoma, glioblastoma, melanoma, and rhabdosarcoma. c-MET - inhibition Known hepatocyte growth factor receptor or tyrosine-protein kinase Met (c-MET) mediated disorders include cancers.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating hepatocyte growth factor receptor or tyrosine-protein kinase Met (c- MET) mediated disorders, in particular for use in the treatment of cancer, wherein preferably the cancer is selected from metastatic non-small cell lung cancer (NSCLC) and advanced renal cell carcinoma.

FGFR3 - inhibition

Known Fibroblast Growth Factor Receptor 3 (FGFR3) mediated disorders include cancers.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Fibroblast Growth Factor Receptor 3 (FGFR3) mediated disorders, in particular for use in the treatment of cancer, wherein preferably the cancer is selected from multiple myeloma, bladder cancer, non-small cell lung cancer, oral cancers, and oropharyngeal squamous cell carcinoma.

IGFR1- inhibition

Known Insuline Like Growth Factor Receptor 1 (IGFR1) mediated disorders include cancers.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating Insuline Like Growth Factor Receptor 1 (IGFR1) mediated disorders, in particular for use in the treatment of cancer, wherein preferably the cancer is selected from breast cancer, sarcoma, and non-small cell lung cancer (NSCLC).

RET- inhibition

Known "REarranged during Transfection" receptor tyrosine kinase (RET) mediated disorders include cancers.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating "REarranged during Transfection" receptor tyrosine kinase (RET) mediated disorders, in particular for use in the treatment of cancer, wherein preferably the cancer is selected from thyroid carcinomas and lung cancers.

SRC- inhibition

Known non-receptor tyrosine kinase Src (SRC) mediated disorders include cancers, osteoporosis, cardiovascular disorders and immune system dysfunction.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating non-receptor tyrosine kinase Src (SRC) mediated disorders, in particular for use in the treatment of cancers, osteoporosis, cardiovascular disorders and immune system dysfunction.

YES- inhibition

Known non-receptor tyrosine kinase Yes (YES) mediated disorders include cancers, osteoporosis, cardiovascular disorders and immune system dysfunction.

In embodiments, the compounds of the invention or a pharmaceutically acceptable salt thereof, in particular are used for treating non-receptor tyrosine kinase Yes (YES) mediated disorders, in particular for use in the treatment of cancers, osteoporosis, cardiovascular disorders and immune system dysfunction.

Routes of Administration

Suitable routes of administration may, for example, include oral, eyedrop, rectal, transmucosal, topical, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.

Alternatively, one may administer the compound in a local rather than a systemic manner, for example, via injection of the compound directly into an edematous site, often in a depot or sustained release formulation.

Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with endothelial cell-specific antibody.

Composition/Formulation

The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.

Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.

For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.

For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.

Pharmaceutical preparations for oral use can be obtained by combining the active compound with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.

Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.

For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.

For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.

The compounds can be formulated for parenteral administration by injection, e.g. bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g. in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.

Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powderform for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.

The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides. In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly or by intramuscular injection). Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.

Synthesis of compounds

The compounds of the present invention can be prepared by methods well known in the art of organic chemistry. See, for example, J. March, ‘Advanced Organic Chemistry’ 4 th Edition, John Wiley and Sons. During synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This is achieved by means of conventional protecting groups, such as those described in T.W. Greene and P.G.M. Wutts ‘Protective Groups in Organic Synthesis’ 3 rd Edition, John Wiley and Sons, 1999. The protective groups are optionally removed at a convenient subsequent stage using methods well known in the art.

The products of the reactions are optionally isolated and purified, if desired, using conventional techniques, including but not limited to, filtration, distillation, crystallization, chromatography and the like. Such materials are optionally characterized using conventional means, including physical constants and spectral data.

Compounds of any one of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, can be prepared by the general synthetic route shown in any one of scheme I - XI.

Scheme I and II show a general synthetic route in relation to an exemplary compound of

Formula l-a.

4-Chloro-3-iodo-1 H -pyrazolo[4,3-c]pyridine II can be prepared from commercially available 4- chloro - 1H-pyrazolo[4,3-c]pyridine using N-iodosuccinimide in a solvent such as DMF at elevated temperatures. The resulting product can then be reacted with 2,4-dimethoxybenzylamine in an appropriate solvent like n-butanol, isopropanol or 2-pentanol at high temperatures to obtain N- [(2,4- dimethoxyphenyl)methyl]-3-iodo-1 H-pyrazolo[4,3-c]pyridin-4-amine III. Compound IV can, subsequently, be prepared from compound III and benzyl (1 R,5R)-5-hydroxycyclohex-3-ene-1- carboxylate using Mitsunobu conditions, for example DIAD/triphenylphosphine in THF at 0 °C.

Compound VI can be prepared from compound IV using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex or tetrakis(triphenylphosphine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Reduction of the double bond and deprotection of the benzylester can be accomplished by catalytic hydrogenation in the presence of a suitable catalyst system and solvent, for example palladium on charcoal in ethyl acetate and methanol to provide compounds of Formula VII.

Compounds of Formula VIII can be prepared from derivatives VII using diphenylphosphorylazide in toluene or THF and a suitable alcohol such as trimethylsilylethanol, benzylalcohol or tert-butanol. Subsequent halogenation can be performed using N-bromosuccinimide or N-iodosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula IX. Compounds of Formula X can be prepared from compound IX using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphos-phine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula XI can be prepared from derivatives of Formula X after deprotection of the amino function with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide. Macrocyclization towards compounds of Formula XII can be accomplished with an appropriate couplings reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. Finally conversion of compounds of Formula XII to compounds with Formula l-a can be accomplished using strong acids like HCI or TFA in the presence of water and a suitable cation scavenger like triisopropylsilane (TIS) at appropriate temperature.

Alternatively compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, can be prepared by the general synthetic route shown in scheme III. Scheme III shows a general synthetic route in relation to an exemplary compound of Formula l-a.

Halogenation of compounds of Formula VII can be performed using N-bromosuccinimide or N- iodosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula XIII. Compounds of Formula XIV can be prepared from compound XIII using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphos-phine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Macrocyclization towards compounds of Formula XV can be accomplished with an appropriate couplings reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. Finally conversion of compounds of Formula XV to compounds with Formula l-a can be accomplished using strong acids like HCI or TFA in the presence of water and TIS at appropriate temperature.

Another route to obtain compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme IV. Scheme IV shows a general synthetic route in relation to an exemplary compound of Formula l-a.

Compounds of Formula XVII can be prepared from compound III and amino-protected (chiral) aminoalcohols (XVI) using Mitsunobu conditions, for example DIAD/triphenylphosphine in THF at 0 °C. Alternatively compounds of Formula XVII can be obtained after activation of the alcohol with for example tosylchloride or mesylchloride to perform a substitution reaction in appropriate solvents such as DMF in the presence of an inorganic base like cesium carbonate or potassium carbonate. Compounds of Formula XVIII can be prepared from compound XVII using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(Il)chloride complex or tetrakis(triphenylphos- phine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Subsequent halogenation of compounds of Formula XVIII can be performed using N-bromosuccinimide or N- iodosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula XIX. Compounds of Formula XX can be prepared from compound XIX using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of a inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula XXI can be prepared from derivatives of Formula XX after amino deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide, subsequent macrocyclization towards compounds of Formula XXI can be accomplished with an appropriate couplings reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. Subsequent removal of the DMB-group using methods known by skilled organic chemists, such as TFA containing triethylsilane provides compounds of Formula l-a.

Yet another route to obtain compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme V. Scheme V shows a general synthetic route in relation to an exemplary compound of Formula l-a.

Compounds of Formula XXIII can be prepared from compound III and amino-protected (chiral) aminoalcohols (XXII) using Mitsunobu conditions, for example DIAD/triphenylphosphine in THF at 0 °C. Alternatively, compounds of Formula XXIII can be obtained after activation of the alcohol with for example tosylchloride or mesylchloride to perform a substitution reaction in appropriate solvents such as DMF in the presence of an inorganic base like cesium carbonate or potassium carbonate. Compounds of Formula XXIV can be prepared from compound XXIII using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex ortetrakis(triphenylphosphine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Subsequent halogenation of compounds of Formula XXIV can be performed using N- bromosuccinimide or N-iodosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula XXV. Compounds of Formula XXVI can be prepared from compound XXV using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula XXVII can be prepared from derivatives of Formula XXVI after deprotection of the amine group with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide, following macrocyclization towards compounds of Formula XXVII can be accomplished with an appropriate coupling-reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. Subsequent removal of the DMB- group using methods known by skilled organic chemists, such as TFA containing TIS provides compounds of Formula l-a.

Alternatively, compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme VI. Scheme VI shows a general synthetic route in relation to an exemplary compound of Formula l-a.

Compounds of Formula XXVIII can be prepared from compound XIX using 2-allyl-4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolane or potassium allyltrifluoroborate, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water.

Deprotection of derivatives of Formula XXVIII can accomplished using methods known by those skilled in the art, such as TBAF of a strong acid like TFA. Subsequently compounds of Formula XXIX can be obtained with terminal alkenes containing an acid functionality, using an appropriate coupling-reagent such as HATU of EDCI.HCI in a suitable solvent like DMF, THF or DCM at appropriate temperature. Ring closing metathesis towards compounds of Formula XXX can be accomplished with an appropriate Grubbs catalyst, molybdene or ruthenium catalyst in a suitable solvent like DCM or toluene at appropriate temperature. Subsequent removal of the DMB-group using methods known by skilled organic chemists, such as TFA containing triethylsilane provides isomeric mixtures of compounds of Formula l-a. The thus obtained mixtures of cis/trans isomers of Formula l-a could be separated using chromatographic techniques such as HPLC to obtain compounds of Formula l-a.

Another route to obtain compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme VII. Scheme VII shows a general synthetic route in relation to an exemplary compound of Formula l-a.

Compounds of Formula XXXI can be prepared from compound XIII using 2-allyl-4, 4,5,5- tetramethyl-1 ,3,2-dioxaborolane or potassium allyltrifluoroborate, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Subsequently compounds of Formula XXXII can be obtained from terminal alkenes containing an amine functionality, an appropriate coupling-reagent such as HATU of EDCI.HCI in a suitable solvent like DMF, THF or DCM at appropriate temperature. Ring closing metathesis towards compounds of Formula XXXIII can be accomplished with an appropriate Grubbs catalyst, molybdene or ruthenium catalyst in a suitable solvent like DCM ortoluene at appropriate temperature. Subsequent removal of the DMB-group using methods known by skilled organic chemists, such as TFA containing triethylsilane provides isomeric mixtures of compounds of Formula l-a. The thus obtained mixtures of cis/trans isomers of Formula l-a could be separated using chromatographic techniques such as HPLC to obtain compounds of Formula l-a. Yet another route to obtain compounds of Formula l-b to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme VIII. Scheme VIII shows a general synthetic route in relation to an exemplary compound of Formula l-b.

The reaction of (3-chloropyrazine-2-yl)methanamine. hydrochloride (XXXIV) can be carried out with an appropriately amine protected amino acid (XXXV) in a solvent such as DMF, THF or DCM in the presence of a base such as DIPEA, N-methylmorpholine, 4-DMAP or triethylamine and in the presence of a coupling reagent such as PyBOP, TBTU, EDCI or HATU to form compounds of Formula XXXVI. Cyclisation of chloropyrazine of with Formula XXXVI can be performed using condensation reagents like phosphorus oxychloride under heating conditions to provide compounds of Formula XXXVII. Subsequent bromination can be accomplished using bromine or N-bromosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula XXXVIII. 8-Aminoimidazo[1 ,5a]pyrazine derivatives XXXIX can be prepared from compounds of Formula XXXVIII using ammonia(gas) in isopropanol at elevated temperature in a pressure vessel or microwave (> 4 atm.) Compounds of Formula XL can be prepared from compound XXXIX using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex or tetrakis(triphenylphos- phine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Subsequent halogenation of compounds of Formula XL can be performed using N-chlorosuccinimide in a suitable solvent like acetic acid at appropriate temperature to obtain compounds of Formula XLL Compounds of Formula XLII can be prepared from compound XLI using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula I can be prepared from derivatives of Formula XLII after deprotection of the amine group with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide. Following macrocyclization towards compounds of Formula l-b can be accomplished with an appropriate coupling-reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature to obtain compounds of Formula l-b.

Yet another route to obtain compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme IX. Scheme IX shows a general synthetic route in relation to an exemplary compound of Formula l-b.

The reaction of (3-chloropyrazine-2-yl)methanamine. hydrochloride (XXXIV) can be carried out with an appropriately amine protected amino acid (XLIII) in a solvent such as DMF, THF or DCM in the presence of a base such as DIPEA, N-methylmorpholine, 4-DMAP or triethylamine and in the presence of a coupling reagent such as PyBOP, TBTU, EDCI or HATU to form compounds of Formula XLIV. Cyclisation of chloropyrazine of Formula XLIV can be performed using condensation reagents like phophorus oxychloride under heating conditions to provide compounds of Formula XLV. Subsequent bromination can be accomplished using bromine or N-bromosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula XLVI. 8- Aminoimidazo[1 ,5a]pyrazine derivatives XLVII can be prepared from compounds of Formula XLVI using ammonia (gas) in isopropanol at elevated temperature in a pressure vessel or microwave (> 4 atm.) Compounds of Formula XLVIII can be prepared from compound XLVII using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex or tetrakis(triphenylphos- phine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Subsequent halogenation of compounds of Formula XLVIII can be performed using N-chlorosuccinimide in a suitable solvent like acetic acid at appropriate temperature to obtain compounds of Formula XLIX. Compounds of Formula L can be prepared from compound XLIX using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula I can be prepared from derivatives of Formula L after deprotection of the amine group with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide. Following macrocyclization towards compounds of Formula l-b can be accomplished with an appropriate coupling-reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature to obtain compounds of Formula l-b.

Yet another route to obtain compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, is depicted in the general synthetic route shown in scheme X. Scheme X shows a general synthetic route in relation to an exemplary compound of Formula l-b.

3-Amino-6-bromo-pyrazine-2-carbonitrile (LIl) can be prepared from commercial available 2- amino-3,5-dibromo pyrazine (LI) using coppercyanide and sodiumcyanide in DMF at elevated temperature. The resulting product can then be converted to 6-bromo-3-chloro-pyrazine-2-carbonitrile (LIII) under Sandmeyer conditions with copperchloride, tert-butylnitrite in an appropriate solvent such as acetonitrile under heating. Reduction of derivative LIII can be accomplished by hydrogenation under elevated pressure in the presence of a suitable catalyst system and solvent, for example Raney-Nickel to provide (6-bromo-3-chloro-pyrazin-2-yl)methanamine (LIV). This can then be reacted with an appropriately amine protected amino acid (XXXV) in a solvent such as DMF, THF or DCM in the presence of a base such as DIPEA, N-methylmorpholine, 4-DMAP or triethylamine and in the presence of a coupling reagent such as PyBOP, TBTU, EDCI or HATU to form compounds of Formula LV. Compounds of Formula LVI can be prepared from compound LV using 2-allyl-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane or potassium allyltrifluoroborate, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Cyclisation chloropyrazine of Formula LVI can be performed using condensation reagents like phosphorus oxychloride under heating conditions to provide compounds of Formula LVII. Subsequent bromination can be accomplished using bromine or N- bromosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula LVIII. 8-Aminoimidazo[1 ,5-a]pyrazine derivatives LIX can be prepared from compounds of Formula LVIII using ammonia(gas) in isopropanol at elevated temperature in a pressure vessel or microwave (> 4 atm.) Compounds of Formula LX can be prepared from compound LIX using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex or tetrakis(triphenylphos-phine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Deprotection of derivatives of Formula LX can accomplished using methods known by those skilled in the art, such as TBAF of a strong acid like TFA. Subsequently, compounds of Formula LXI can be obtained with terminal alkenes containing an acid functionality, an appropriate coupling-reagent such as HATU of EDCI.HCI in a suitable solvent like DMF, THF or DCM at appropriate temperature. Ring closing metathesis towards compounds of Formula l-b can be accomplished with an appropriate Grubbs catalyst, molybdene or ruthenium catalyst in a suitable solvent like DCM or toluene at appropriate temperature. The thus obtained mixtures of cis/trans isomers of Formula l-b could be separated using chromatographic techniques such as HPLC to obtain compounds of Formula l-b.

Alternatively compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, can be prepared by the general synthetic route shown in scheme XI. Scheme XI shows a general synthetic route in relation to an exemplary compound of Formula l-a.

4-Chloro-3-bromo-1H -pyrazolo[4,3-c]pyridine LXII can be prepared from commercially available 4-chloro-1H -pyrazolo[4,3-c]pyridine using N- bromosuccinimide in a solvent such as acetonitrile or DMF at elevated temperatures. Compounds of Formula LXIII can, subsequently, be prepared from compound LXII and Compound XVI using Mitsunobu conditions, for example DIAD/triphenylphosphine in THF at 0 °C. 1H -pyrazolo[4,3-c]pyridin-4-amine derivatives LXIV can be prepared from compounds of Formula LXIII using ammonia(gas) in isopropanol or 25% aq. ammonia at elevated temperature in a pressure vessel or microwave (> 4 atm.). Compounds of Formula LXV can be prepared from compound LXIV using N- iodosuccinimide in a solvent such as acetonitrile or DMF at room temperature. Subsequent amine protection of compound LXV using for example Boc 2 O or Z- ONSu provide compounds of Formula LXVI. Compounds of Formula LXVII can be prepared from compound LXVI using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula LXVIII can be prepared from derivatives of Formula LXVII after deprotection of the amino group with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide. Macrocyclization towards compounds of Formula LXVIII can be accomplished with an appropriate couplings reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. Finally conversion of compounds of Formula LXVIII to compounds of Formula l-a can be accomplished using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex or tetrakis(triphenylphos-phine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Alternatively compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, can be prepared by the general synthetic route shown in scheme XII. Scheme XII shows a general synthetic route in relation to an exemplary compound of Formula l-b.

3-Amino-6-bromo-pyrazine-2-carbonitrile (LII) can be prepared from commercial available 2- amino-3,5-dibromo pyrazine (LI) using copper cyanide and sodium cyanide in DMF at elevated temperature. The resulting product can then be converted to 6-bromo-3-chloro-pyrazine-2-carbonitrile (LIII) under Sandmeyer conditions with copper chloride, tert-butylnitrite in an appropriate solvent such as acetonitrile under heating. Reduction of derivative LIII can be accomplished by hydrogenation under elevated pressure in the presence of a suitable catalyst system and solvent, for example Raney-Nickel to provide (6-bromo-3-chloro-pyrazin-2-yl)methanamine (LIV). This can then be reacted with an appropriately amine protected amino acid (XXXV) in a solvent such as DMF, THF or DCM in the presence of a base such as DIPEA, N-methylmorpholine, 4-DMAP or triethylamine and in the presence of a coupling reagent such as PyBOP, TBTU, EDCI or HATU to form compounds of Formula LV. Compounds of Formula LXIX can be prepared from compound LV using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Cyclisation chloropyrazine of Formula LXIX can be performed using condensation reagents like phosphorus oxychloride under heating conditions to provide compounds of Formula LXX. Subsequent bromination can be accomplished using bromine or N- bromosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula LXXI. 8-Aminoimidazo[1 ,5-a]pyrazine derivatives LXXII can be prepared from compounds of Formula LXXI using ammonia(gas) in isopropanol at elevated temperature in a pressure vessel or microwave (> 4 atm.) Compounds of Formula LXXIII can be prepared from compound LXXII using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex ortetrakis(triphenylphos-phine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula I can be prepared from derivatives of Formula LXXIII after deprotection of the amine group with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide. Following macrocyclization towards compounds of Formula l-b can be accomplished with an appropriate coupling- reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. After purification using chromatographic techniques such as HPLC compounds of Formula l-b could be obtained.

Alternatively compounds of Formula l-a to l-h, wherein R 1 to R 5 and W, V and U have the previously defined meanings, can be prepared by the general synthetic route shown in scheme XIII. Scheme XIII shows a general synthetic route in relation to an exemplary compound of Formula l-b.

3-Amino-6-bromo-pyrazine-2-carbonitrile (LII) can be prepared from commercial available 2- amino-3,5-dibromo pyrazine (LI) using copper cyanide and sodium cyanide in DMF at elevated temperature. The resulting product can then be converted to 6-bromo-3-chloro-pyrazine-2-carbonitrile (LIII) under Sandmeyer conditions with copper chloride, tert-butylnitrite in an appropriate solvent such as acetonitrile under heating. Reduction of derivative LIII can be accomplished by hydrogenation under elevated pressure in the presence of a suitable catalyst system and solvent, for example Raney-Nickel to provide (6-bromo-3-chloro-pyrazin-2-yl)methanamine (LIV). LIV can then be reacted with an appropriately amine protected amino acid (XXXV) in a solvent such as DMF, THF or DCM in the presence of a base such as DIPEA, N-methylmorpholine, 4-DMAP or triethylamine and in the presence of a coupling reagent such as PyBOP, TBTU, EDCI or HATU to form compounds of Formula LV. Compounds of Formula LXIX can be prepared from compound LV using an appropriate boronic acid or pinacolester, in the presence of a suitable palladium catalyst system, for example CataCXium® A Pd G3 or bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Cyclisation chloropyrazine of Formula LXIX can be performed using condensation reagents like phosphorus oxychloride under heating conditions to provide compounds of Formula LXX. Compounds of Formula LXXIV can be prepared from derivatives LXX using trimethylboroxine, in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)ch loride complex in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Subsequent bromination can be accomplished using bromine or N- bromosuccinimide in a suitable solvent like DCM or DMF at appropriate temperature to obtain compounds of Formula LXXV. 8-Methylimidazo[1 ,5-a]pyrazine derivatives LXXVI can be prepared from compound LXXV using an appropriate boronic acid or pinacolester (V), in the presence of a suitable palladium catalyst system, for example bis(diphenylphosphine)palladium(0) palladium(ll)chloride complex or tetrakis(triphenylphosphine)palladium(0) in the presence of an inorganic base like potassium carbonate, cesium carbonate or potassium phosphate in a suitable solvent system like combinations of dioxane and water. Derivatives of Formula I can be prepared from derivatives of Formula LXXVI after deprotection of the amine group with TBAF or a strong acid like TFA and subsequent carboxyl acid deprotection using a suitable inorganic base like lithium hydroxide or sodium hydroxide. Following macrocyclization towards compounds of Formula l-b can be accomplished with an appropriate coupling- reagent such as HATU of EDCI.HCI in a suitable solvent like DMF at appropriate temperature. After purification using chromatographic techniques such as HPLC compounds of Formula l-b could be obtained.

The invention is illustrated by the following examples.

Examples

The following examples are illustrative embodiments of the invention, not limiting the scope of the invention in any way. Reagents are either commercially available or are prepared according to procedures known in the literature.

Method LCMS (A)

LC-MS system equipped with a Waters 2998 Photodiode Array Detector, Waters Acquity QDa Detector, Waters 2767 autosampler and Waters 2545 binary gradient module was used for sample analyses with a XTerra ® MS C18 column (2.5 μm, 4.6 x 50 mm) for 10 min measurements.

The eluents used for this system are A (95/5 v/v% Milli-Q water/acetonitrile + 0.1 % formic acid) and B (acetonitrile + 0.1 % formic acid).

Method LCMS (A): 95% A to 95% B in 7 min, then 95% A. Method LCMS (B)

LC-MS system equipped with a Waters 2998 Photodiode Array Detector, Waters Acquity QDa Detector, Waters 2767 autosampler and Waters 2545 binary gradient module was used for sample analyses with a XTerra ® MS C18 column (2.5 μm, 4.6 x 50 mm) for 30 min measurements.

The eluents used for this system are A (95/5 v/v% Milli-Q water/acetonitrile + 0.1 % formic acid) and B (acetonitrile + 0.1 % formic acid).

Method LCMS (B): 95% A to 95% B in 22 min, then switched to 95% A.

Method Preparative HPLC

LC-MS system equipped with a Waters 2998 Photodiode Array Detector, Waters Acquity QDa Detector, Waters 2767 autosampler and Waters 2545 binary gradient module was used for Preparative reversed phase chromatography with a Luna ® 5 μm C18(2) 100 Å (150 x 21 mm).

The eluents used for this system are A (95/5 v/v% Milli-Q water/acetonitrile + 0.1 % formic acid) and B (acetonitrile + 0.1 % formic acid).

The following abbreviations are used throughout the application with respect to chemical terminology:

TFA Trifluoracetic acid

HATU O-(7-Azabenzotriazol-1-yl)-1 ,1 ,3,3-tetramethyluroniumhexafluorophosphate

DMF N,N- Dimethylformamide

THF Tetrahydrofuran

DCM Dichloromethane

TMS-CI Chlorotrimethylsilane

DiPEA N,N- Diisopropylethylamine

HPLC High Performance Liquid Chromatography

LCMS Liquid Chromatography with Mass Spectrometry detection

4-DMAP 4-Dimethylamino pyridine

Boc tert-Butyloxycarbonyl

Cbz Benzyloxycarbonyl

LiHMDS Lithium bis(trimethylsilyl)amide

DBU 1 ,8-Diazabicyclo[5.4.0]undec-7-ene

DEAD Diethyl azodicarboxylate o/n Overnight

Pd(dppf)CI 2 1 ,1 '-bis(diphenylphosphino)ferrocene palladium(ll) chloride

AIBN Azobisisobutyronitril

ZrCp 2 (H)CI zirconocene hydrochloride (Schwartz Reagent)

1 H-NMR Proton nuclear magnetic resonance

BOC 2 O Di-tert-butyl decarbonate

DPPA Diphenylphosphoryl azide T3P Propylphosphonic anhydride

NIS N- lodosuccinimide

PyBOP benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate

TBTU 2-(1 H-Benzotriazole-1-yl)-1 ,1 ,3,3-tetramethylaminium tetrafluoroborate

EDCI 1 -Ethyl-3-(3-dimethylaminopropyl)carbodiimide

2MeTHF 2-Methyltetrahydrofuran

Z-ONSu N- (Benzyloxycarbonyloxy)succinimide o/w Over the weekend

TCEP T ris(2-carboxyethyl)phosphine

Tris 2-Amino-2-(hydroxymethyl)propane-1 ,3-diol

The names of the final products in the intermediates and examples are generated using Biovia Draw (version 16.1). In cases were Biovia Draw could not generate a name, molecular structures are given.

Scaffold A N- [(2,4-dimethoxyphenyl)methyl1-3-iodo-1 /7-pyrazolo[4,3-clpyridin-4-amine

(a) 4-Chloro-3-iodo-1 /7-pyrazolo[4,3-clpyridine

To a solution of 4-chloro-1 /7-pyrazolo[4,3-c]pyridine (50 g, 325.6 mmol) in DMF (500 mL) was added N- iodosuccinimide (80.6 g, 353.1 mmol) and the mixture was stirred at 100 °C for 1 h. The mixture was cooled and added slowly to a mixture of 5% sodium thiosulfate I sodium bicarbonate solution I water (1 L / 500 mL I 500 mL). The mixture was transferred to a separation funnel and extracted with ethyl acetate (total of 3.5 L). The ethyl acetate layer was separated, washed with water (750 mL) and brine (500 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure (still contains >25 mL of DMF). To the resulting suspension was added 100 mL of ethyl acetate and under stirring 250 mL of hexane. The solvent was decanted and the resulting suspension was again treated with ethyl acetate (50 mL) and hexane 250 mL. The precipitate was filtered and dried under vacuum to give 67.7 g of the title compound as a powder (Yield: 74.4%).

(b) N- [(2,4-dimethoxyphenyl)methyl1-3-iodo-1 /7-pyrazolo[4,3-clpyridin-4-amine (Scaffold A)

To a suspension of 4-chloro-3-iodo-1 /7-pyrazolo[4,3-c]pyridine (67.7 g, 242.2 mmol) in 1-butanol (675 mL) at room temperature was added 2,4-dimethoxybenzylamine (121 .5 g, 726.6 mmol) and the mixture was heated at 120 °C and stirred overnight. After cooling to room temperature, the reaction solution was concentrated under reduced pressure. The precipitate was suspended in water and extracted with ethyl acetate. The ethyl acetate layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give the crude title compound. The crude was suspended/dissolved in dichloromethane (350 mL) and refluxed at 50 °C. Hexane (350 mL) was added dropwise under reflux and stirred for 1 h. After cooling, the precipitate formed was filtered, washed with dichloromethane/hexane = 1/1 v/v% and dried under vacuum at 40 °C to give 75.82 g of the title compound as an off-white powder (Yield: 76.3%).

Scaffold B

Benzyl (1 R,5S)-5-[4-[(2,4-dimethoxyphenyl)methylamino1-3-iodo-pyrazol o[4,3-c]pyridin-1-yl]cyclohex- 3-ene-1 -carboxylate (Scaffold B)

To an ice-cold (4 °C) suspension of N- [(2,4-dimethoxyphenyl)methyl]-3-iodo-1 H-pyrazolo[4,3- c]pyridin-4-amine (Scaffold A) (41.02 g, 100 mmol), benzyl (1 R,5R)-5-hydroxycyclohex-3-ene-1- carboxylate (Intermediate RP1) (25.54 g, 110 mmol) and triphenylphosphine (30.14 g, 115 mmol) in toluene (400 mL) was added dropwise a solution of diisopropyl azodicarboxylate (24.07 mL, 115 mmol) in toluene (100 mL). The mixture was stirred for 30 min at 4 °C and then stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and DCM/acetone = 97/3 to 95/5 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 50 g of benzyl (1 R,5S)-5-[4-[(2,4- dimethoxyphenyl)methylamino]-3-iodo-pyrazolo[4,3-c]pyridin-1 -yl]cyclohex-3-ene-1 -carboxylate (Scaffold B (yield 80 %).

(1R,3R)-3-[4-[(2,4-Dimethoxyphenyl)methylamino]-3-[4-[[4-(tr ifluoromethyl)-2-pyridyl]carba- moyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohexanecarboxyli c acid (Scaffold C) (a) benzyl (1R,5S)-5-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4-(tr ifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohe x-3-ene-1-carboxylate Benzyl (1R,5S)-5-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-pyraz olo[4,3-c]pyridine-1- yl]cyclohex-3-ene-1-carboxylate (15.88 g, 25.43 mmol) was dissolved in dioxane/water = 4/1 v/v% (125 mL) and potassium carbonate (10.54 g, 76.29 mmol) was added. The solution was purged with nitrogen for 5 min and tert-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-N-[4-(t rifluoromethyl)-2- pyridyl]benzamide (10.96 g, 27.97 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (1.03 g, 1.27 mmol) were added. The reaction mixture was stirred for 2 h at 80 °C. The reaction mixture was diluted with ethyl acetate and filtered over Decalite™. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and ethyl acetate/heptane = 1/4 to 3/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 18.5 g (Yield 95.0%). (b) (1R,3R)-3-[4-[(2,4-Dimethoxyphenyl)methylamino]-3-[4-[[4-(tr ifluoromethyl)-2-pyridyl]carba- moyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohexanecarboxyli c acid (Scaffold C) To a solution of benzyl (1R,5S)-5-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-1-yl]cyclohex-3-ene-1-carboxylate (18.0 g, 23.54 mmol) in ethyl acetate/methanol = 4/1 v/v% (350 mL) was added 1.8 g of 10% Pd/C. Catalytic hydrogenation was performed for 16 h at room temperature. The reaction was not completed. The benzylester was reduced completely but ~31% of a double-bond containing product remained. The palladium-catalyst was filtered and the filtrate was recharged with 10% Pd/C (1.8 g) and catalytic hydrogenation was continued for 24 h. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to give 14.74 g of the title compound (Yield: 85.0%). Benzyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (a) (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (R)-(+)-3-Cyclohexenecarboxylic acid (50.7 g, 402 mmol) was suspended in H 2 O (400 mL) under nitrogen. The mixture was cooled to 4 °C and sodium bicarbonate (101.3 g, 1.21 mol) was added, followed by a solution of potassium iodide (333 g, 2.01 mol) and iodine (107 g, 422 mmol) in H 2 O (400 mL). The reaction was allowed to come to room temperature and stirred o/n and then extracted with dichloromethane (4x100 mL). The combined organic layers were washed with sat. NaHSO 3 -solution (2x50 mL). The organic layer was protected from light, dried over Na 2 SO 4 , filtered and concentrated (20 mbar) to afford (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (90.1 g, 89.0 %) as an off-white solid. (b) (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (90.1 g, 358 mmol) was dissolved in dry THF (650 mL). Then, DBU (77 mL, 515 mmol) was added and the mixture was refluxed for 6 h. After cooling to room temperature, the suspension was filtered through Celite™, and concentrated in vacuo to ~250 mL. and was used directly in the next step. (c) Benzyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (Intermediate RP1) To the THF solution of (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one (0.4 mol) in methanol (300 mL) was added 2M NaOH-solution (300 mL) and the mixture was stirred for 15 min at room temperature. The reaction was quenched by addition of 3M HCl-solution (300 mL) and the water layer was saturated by addition of sodium chloride. The mixture was extracted with ethyl acetate (3x100 mL). The combined organic phases were washed with brine, dried over sodium sulfate, filtered and the solvent removed in vacuo. The residue was dissolved in DMF (800 ml), cesium carbonate (129 g, 0.4 mol) and benzyl bromide (57 mL, 0.48 mol) were added subsequently. The mixture was stirred at room temperature for 30 min. The precipitate formed was filtered and the precipitate was washed with diethylether. The filtrate was washed with water, brine, dried over sodium sulfate and evaporated under reduced pressure. The residue was purified by column chromatography (heptane/ethyl acetate = 95/5 to 45/55 v/v%) to give benzyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (57.1 g, 61.5% over 3 steps) as an oil. (a) (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (R)-(+)-3-Cyclohexenecarboxylic acid (50.7 g, 0.4 mol) was suspended in H 2 O (400 mL) under nitrogen. The reaction mixture was cooled to 4 °C and sodium bicarbonate (101 g, 1.2 mol) was added, followed by a solution of potassium iodide (333 g, 2 mol) and iodine (107 g, 0.42 mol) in H 2 O (400 mL). The reaction was allowed to come to room temperature and stirred o/n and then extracted with DCM (4x150 mL). The combined organic layers were washed with a solution of Na 2 S 2 O 3 (120 g) in H 2 O (600 mL). The aqueous layer was extracted with DCM (2x150 mL). The combined organic layers were protected from light, dried over Na 2 SO 4 , filtered and concentrated (20 mbar) to afford (1R,4R,5R)-4- iodo-6-oxabicyclo[3.2.1]octan-7-one (95.22 g, 94.5 %) as an off-white solid. (b) (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (95.22 g, 377.9 mmol) was dissolved in dry THF (700 mL). Then, DBU (86.3 g, 566.9 mmol) was added and the mixture was refluxed o/n. The reaction mixture was cooled to room temperature, diluted with diethylether (500 mL) and extracted with aq. HCl (1 L, 1 M) and brine (250 mL). The aqueous layers were extracted with diethylether (2 x 480 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated (350 mbar to afford (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one quantitatively as an oil which was used directly in the next step. (c) Ethyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (Intermediate RP2) To a stirred solution of (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one (377.9 mmol, theor.) in ethanol (750 mL) was added potassium carbonate (10.45 g, 75.6 mmol) at room temperature and the mixture stirred o/n. The reaction mixture was filtered through a Celite pad. Removal of ethanol under reduced pressure afforded the crude product that was purified by column chromatography plug filtration (eluent 40% EtOAc/heptane) to afford the title compound (41.38 g, 60.8% over 3 steps and column) as a yellow liquid. Intermediate RP3 Methyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (a) (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (R)-(+)-3-Cyclohexenecarboxylic acid (20.2 g, 160 mmol) was suspended in H 2 O (430 mL) under nitrogen. The reaction mixture was cooled to 0 °C and sodium bicarbonate (40.3 g, 480.3 mmol) was added, followed by a solution of potassium iodide (159.5 g, 961 mmol) and iodine (39.6 g, 168 mmol) in H 2 O (360 mL). The reaction was allowed to come to room temperature and stirred o/n and then extracted with DCM (3x150 mL). The combined organic layers were washed with a solution of Na 2 S 2 O 3 (120 g) in H 2 O (600 mL). The aqueous layer was extracted with DCM (2x150 mL). The combined organic layers were protected from light, dried over Na 2 SO 4 , filtered and concentrated (20 mbar) to afford (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (37.88 g, 93.9 %) as an off-white solid. (b) (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one (1R,4R,5R)-4-iodo-6-oxabicyclo[3.2.1]octan-7-one (37.88 g, 150.3 mmol) was dissolved in dry THF (750 mL). Then, DBU (34.3 g, 225.2 mmol) was added and the mixture was refluxed o/n. The reaction mixture was cooled to room temperature, diluted with diethylether (480 mL) and extracted with aq. HCl (1 L, 0.5 M) and brine (1 L). The aqueous layers were extracted with diethylether (2 x 480 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated (350 mbar to afford (1R,5R)-6-oxabicyclo[3.2.1]oct-3-en-7-one quantitatively as a yellowish oil which was used directly in the next step. (c) Methyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (Intermediate RP3) Sodium bicarbonate (37.88 g, 0.451 mol) was added to a solution (1R,5R)-6- oxabicyclo[3.2.1]oct-3-en-7-one (150.3 mmol theor.) in anhydrous MeOH (300 mL). After stirring for 1 week at room temperature the solvent was removed in vacuo (40 °C/300 mbar). The residue was diluted with water (500 mL) and extracted with dichloromethane (3x250 mL). The combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (21.2 g, 90.3%) as aliquid. tert-Butyl N-[(1R,3S)-3-hydroxycyclohexyl]carbamate (a) 2-Cyclohex-2-en-1-ylisoindoline-1,3-dione A mixture of potassium phthalimide (69.88 g, 377.2 mmol) and 3-bromocyclohexene (60.75 g, 377.2 mmol) in DMF (500 mL) was stirred at 30 °C and gradually warmed to 100 °C for 6 h and then o/n by room temperature. The reaction mixture was diluted with water (2 L), extracted with ethyl acetate (3x500 mL). The combined organic layers were washed with water (2x500 mL), brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography (dichloromethane) to afford 63.29 g of the title compound (Yield: 73.8%). (b) (±) 13-Bromo-12b-ethoxy-2,6-methano[1,3]oxazocino[2,3a]iso-indol -8-one N-Bromosuccinimide (61.95 g, 348 mmol) was added to a stirred solution of 2-cyclohex-2-en- 1-ylisoindoline-1,3-dione (63.29 g, 278.4 mmol) in chloroform (1000 mL) and ethanol (65 mL) and the mixture was stirred at room temperature o/n. The mixture was washed with 1M sodium thiosulphate solution (1.5 L). The organic layer was separated and dried over sodium sulphate, filtered and evaporated under reduced pressure. The residue was purified by column chromatography (Heptane/Ethyl acetate = 7/3 v/v%) to afford the title compound (97.0 g, 98.9%). (c) 1,2-Trans-2,3-trans-2-Bromo-3-N-phtalimidocyclohexanol 2M HCl-solution (150 mL) was added to a stirred solution of the orthoamide (86.44 g, 245.4 mmol) in methanol (600 mL) and the solution was stirred at room temperature for 30 min. Most of the solvent was removed and dichloromethane (300 mL) was added to the residue. This solution was then washed with water (2x50 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crystalline residue was recrystallised from ethyl acetate/hexane (200/500 mL @ 75°C) to give the title compound (70.04 g, 88.0%) (d) cis-3-Phthalimidocyclohexanol Tri-n-butyltin hydride (84 g, 288.7 mmol) was added to a stirred solution of 1,2-trans-2,3-trans- 2-bromo-3-N-phtalimidocyclohexanol (78 g, 240.6 mmol) and AIBN (0.2M in toluene, 60 mL, 12 mmol) in toluene (700 mL) and methanol (70 mL) and the mixture was stirred at reflux o/n. Additonal AIBN (2x5 mL) and tri-n-butyltin hydride (2x10 mL) were added and the reaction mixture was stirred at reflux o/n. Reaction proceeded slowly and additional AIBN (20 mL) and tri-n-butyltin hydride were added and the reaction mixture was stirred at reflux for 3h. Progress was followed by TLC. The mixture was then placed under nitrogen and additional AIBN (20 mL) and tri-butyltin hydride (20 g) were added subsequently and the mixture was stirred at reflux for 4 h. The reaction mixture was concentrated under reduced pressure and the residue was triturated with 750 mL heptane (2 x). The heptane layer was removed by suction under reduced pressure. Again 100 mL ethyl acetate and 650 mL heptane were added. The precipitate was filtered, washed with hexane and dried under vacuum at 40 °C o/n to give 49.07 g of the title compound (yield: 83.1%). (e) 2-[(1R,3S)-3-hydroxycyclohexyl]isoindoline-1,3-dione + [(1R,3S)-3-(1,3-dioxoisoindolin-2- yl)cyclohexyl] acetate To a solution of cis-3-phthalimidocyclohexanol (49 g, 200 mmol) in THF (600 mL) was added Lipase Novozyme 435 (25 g) and vinyl acetate (55.3 mL, 600 mmol). The resulting mixture was shaken at room temperature and 250 rpm o/n. The progress of the reaction was monitored by LC-MS. After o/n reaction, the enzyme was filtered off, washed with THF and the solvent was evaporated under reduced pressure. The residue was purified by column chromatography using SiO 2 and ethyl acetate/heptane = 1/1 to 10/0 and to ethyl acetate/dichloromethane = 3/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 21.21 g of 2-[(1R,3S)-3-hydroxycyclohexyl]isoindoline- 1,3-dione (yield 43.3 %) and 31 g of [(1R,3S)-3-(1,3-dioxoisoindolin-2-yl)cyclohexyl] acetate (yield 54.0 %). (f) (1S,3R)-3-aminocyclohexanol To a solution of 2-[(1R,3S)-3-hydroxycyclohexyl]isoidoline-1,3-dione (21.2 g, 86.4 mmol) in ethanol (280 mL) was added hydrazine hydrate (4.28 mL) and the reaction mixture was stirred at 100 °C o/n. The mixture was cooled to room temperature and the precipitate was filtered, washed with ethanol and the filtrate was concentrated under reduced pressure to give 9.87 g of the title compound as a yellow solid (yield: 59.9%). The crude precipitate 13.74 g still contains a lot of 2,3- dihydrophthalazine-1,4-dione and product (yield: 39.4%). (g) tert-Butyl N-[(1R,3S)-3-hydroxycyclohexyl]carbamate (Intermediate RP4) To a solution of (1S,3R)-3-aminocyclohexanol (9.87 g, 51.74 mmol) in dioxane (200 mL) was added di-tert-butyl dicarbonate (11.86 g) and the reaction mixture was stirred at room temperature o/n. Dioxane was partly evaporated and ethyl acetate (500 mL) was added to the suspension. The suspension was washed with NaOH-solution (4 g in 200 mL), water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 12.5 g of the title compound. Second batch: To a solution of (1S,3R)-3-aminocyclohexanol (13.74 g, 34.0 mmol) in dioxane (200 mL) was added di-tert-butyl dicarbonate (7.8 g) and the reaction mixture was stirred at room temperature o/w. Dioxane was partly evaporated and ethyl acetate (500 mL) was added to the suspension. The suspension was washed with NaOH-solution (4 g in 200 mL), water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 7.54 g of the title compound. Both batches were combined and suspended in ethyl acetate. Hexane was added and the precipitate formed was filtered, washed with hexane and dried under high vacuum at 40 °C to give 16.3 g of tert-butyl N-[(1R,3S)-3-hydroxycyclohexyl]carbamate (Intermediate RP4) (yield: 87.6%). (h) [(1S,3R)-3-(tert-butoxycarbonylamino)cyclohexyl] (2S)-3,3,3-trifluoro-2-methoxy-2-phenyl- propanoate To a solution of tert-butyl N-[(1R,3S)-3-hydroxycyclohexyl]carbamate (50 mg, 0.23 mmol) in dichloromethane (2 mL) was added triethylamine (35.6 µL, 0.26 mmol) and 4-dimethylaminopyridine (3 mg, 0.023 mmol) and the mixture was stirred for 5 min. (R)-(-)-α-methoxy-α-trifluoromethylphenylacetyl chloride (61.6 mg, 0.24 mmol) was added and the reaction mixture was stirred at room temperature o/n. Dichloromethane was distilled off under reduced pressure, the resulting residue was purified by column chromatography (heptane/ethyl acetate = 8/2 v/v%) to give 71.4 mg of the title compound (Yield: 72.0%). Both proton and fluor NMR showed that tert-butyl N-[(1R,3S)-3-hydroxycyclohexyl]carbamate (Intermediate RP4) had an diastereomeric excess of > 99.0%.

(1R,3R)-3-(Benzyloxycarbonylamino)cyclohexanecarboxylic acid (a) Methyl (1R,5S)-5-[bis(tert-butoxycarbonyl)amino]cyclohex-3-ene-1-ca rboxylate To an ice-cold (4 °C) solution of di-tert-butyl iminodicarboxylate (4.6 g, 21.2 mmol), methyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (Intermediate RP3) (3.31 g, 21.2 mmol) and triphenylphosphine (6.67 g, 25.4 mmol) in 2-MeTHF (180 mL) was added dropwise a solution of diisopropyl azodicarboxylate (6.26 mL, 31.8 mmol) in 2-MeTHF (30 mL). The mixture was stirred for 30 min at 4 °C and then allowed to warm to room temperature and stirred for 3 h. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 3/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 5.68 g of methyl (1R,5S)-5-[bis(tert-butoxycarbonyl)amino]cyclohex-3- ene-1-carboxylate (yield 75.4 %). (b) Methyl (1R,3R)-3-[bis(tert-butoxycarbonyl)amino]cyclohexanecarboxyl ate To a solution of methyl (1R,5S)-5-[bis(tert-butoxycarbonyl)amino]cyclohex-3-ene-1-ca rboxylate (2.48 g, 6.98 mmol) in ethanol (140 mL) was added 248 mg of 10% Pd/C. Catalytic hydrogenation was performed at room temperature for 16 h. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to give 2.6 g of the title compound (Yield: quantitative). (c) Methyl (1R,3R)-3-aminocyclohexanecarboxylate hydrochloride To methyl (1R,3R)-3-[bis(tert-butoxycarbonyl)amino]cyclohexanecarboxyl ate (2.6 g, 7.2 mmol) was added 4M HCl/dioxane solution (18 mL) and the mixture was stirred at room temperature o/n. The mixture was concentrated in vacuo to give 1.06 g the title compound (yield: 76%). (d) Methyl (1R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxylate Methyl (1R,3R)-3-aminocyclohexanecarboxylate hydrochloride (1.06 g, 5.47 mmol) was suspended in 10 mL water. Sodium bicarbonate (1.38 g, 16.4 mmol) in 10 mL water was added followed by a drop-wise addition of a solution N-(benzyloxycarbonyloxy)succinimide (1.50 g, 6.01 mmol) in dioxane (30 mL). The reaction mixture was stirred at room temperature o/n. The mixture was diluted with ethyl acetate (50 mL) and water (50 mL) and the bi-phasic system was stirred 30 minutes at room temperature. The layers were separated and the water layer was extracted with ethyl acetate (2x20 mL). The combined organic layers were washed with water (50 mL), 0.5N aq. HCl-solution (50 mL), water (50 mL), 5% aq. NaHCO 3 -solution (50 mL), water (50 mL) and brine (25 mL), dried (Na 2 SO 4 ), filtered and concentrated in vacuo to give 1.78 g of the title compound (yield: quantitative, crude). (e) (1R,3R)-3-(Benzyloxycarbonylamino)cyclohexanecarboxylic acid (Intermediate RP5) The crude product methyl (1R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxyate (1.45 g, 4.98 mmol) was dissolved in THF/dioxane/water = 4/1/1 v/v% (74 mL) and subsequently lithium hydroxide (358 mg, 14.9 mmol) was added. The mixture was stirred at room temperature o/n. Ethyl acetate (50 mL) and water (were added) and the pH of the mixture was adjusted to pH < 3 by addition of 2M HCl-solution. The organic phase was separated, washed with water, brine, dried over sodium sulfate , filtered and concentrated under reduced pressure to give 800 mg of (1R,3R)-3- (benzyloxycarbonylamino)cyclohexanecarboxylic acid (Intermediate RP5) (yield:57.9%). (1R,3R)-3-(Benzyloxycarbonylamino)cyclohexanecarboxylic acid (a) tert-Butyl N-(4-nitrophenyl)sulfonylcarbamate Triethylamine (10.4 mL, 74.62 mmol), 4-dimethylaminopyridine (605 mg, 4.95 mmol) and di- tert-butyl dicarbonate (13.5 g, 61.86 mmol) were added sequentially to a solution of 4-nitrobenzene sulfonamide (10 g, 49.46 mmol) in dichloromethane (100 mL). The reaction mixture was stirred for 30 minutes at room temperature. To the reaction mixture was added hydrochloric acid (1N aqueous solution) until it became acidic. The organic layer was separated and washed with saturated sodium chloride aqueous solution, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residue was dissolved in dichloromethane and purified by purification over silicagel (heptane to ethyl acetate = 10/0 to 0/10) to give 13.09 g of the title compound (yield: 87.5%). (b) Methyl (1R5S)-5-[tert-butoxycarbonyl-(4-nitrophenyl)sulfonyl-amino] -cyclohex-3-ene-1-carboxylate To a cold (-20 °C) solution of methyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (15 g, 96.0 mmol), tert-butyl N-(4-nitrophenyl)sulfonylcarbamate (29.0 g, 96.0 mmol) and triphenylphosphine (27.7 g, 105.6 mmol) in THF (300 mL) was added dropwise a solution of diisopropyl azodicarboxylate (20.8 mL, 105.6 mmol) in THF (100 mL). The reaction mixture was concentrated under reduced pressure to give a residue which was purified by column chromatography (hexane/ethyl acetate = 85/15 v/v%) to give 36 g of the title compound. (yield: 85.1%). (c) Methyl (1R,5S)-5-(tert-butoxycarbonylamino)cyclohex-3-ene-1-carboxy late To a stirred solution of methyl (1R,5S)-5-[tert-butoxycarbonyl-(4-nitrophenyl)sulfonyl-amino ]- cyclohex-3-ene-1-carboxylate (35.15 g, 79.8 mmol) in acetone (300 mL) was added DBU (23.85 mL, 159.6 mmol) and 2-mercaptoethanol (11.23 mL, 159.6 mmol). The reaction mixture was stirred for 3 h at room temperature. Acetone was removed under reduced pressure and the resulting residue was purified by column chromatography (hexane/ethyl acetate = 90/10 to 88/12 v/v%) to give 14.1 g of the title compound (Yield: 69.2%) as a crystalline white solid. (d) Methyl (1R,3R)-3-(tert-butoxycarbonylamino)cyclohexanecarboxylate To a solution of methyl (1R,5S)-5-(tert-butoxycarbonylamino)cyclohex-3-ene-1-carboxy late (14.9 g, 58.36 mmol) in methanol (300 mL) was added 1.5 g of 10% Pd/C. Catalytic hydrogenation was performed for 3 h at room temperature. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to afford the title compound in quantitative crude yield. (e) Methyl (1R,3R)-3-aminocyclohexanecarboxylate hydrochloride To a cooled (4 °C) solution of methyl (1R,3R)-3-(tert-butoxycarbonylamino)cyclo- hexanecarboxylate (15.2 g, 58.36 mmol) in methanol (300 mL) was added drop-wise acetyl chloride (42 mL, 583.6 mmol). The reaction mixture was stirred for 1 h. The mixture was concentrated under reduced pressure and dried in vacuo to give the title compound in quantitative crude yield. (f) Methyl (1R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxylate To a cooled (4 °C) solution of methyl (1R,3R)-3-aminocyclohexanecarboxylate hydrochloride (58.36 mmol) in dioxane/water = 1/1 v/v% (200 mL) was added portion-wise sodium bicarbonate (14.7 g, 175 mmol). To the resulting suspension was added drop-wise a solution of N- (benzyloxycarbonyloxy)succinimide (14.8 g, 59.02 mmol) in dioxane (150 mL) and the resulting mixture was stirred at room temperature o/w. LC-MS showed some starting material present. Additionally, 1.5 g of Z-ONSu was added as a solution in dioxane. The mixture was stirred at room temperature o/n. Ethyl acetate was added and the resulting mixture was washed with 0.5M HCl solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and concentrated under reduced pressure and dried in vacuo to give the title compound in quantitative crude yield. (g) (1R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxylic acid (Intermediate RP5) To a solution of methyl (1R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxylate (58.36 mmol) in THF/water = 4/1 v/v% (375 mL) was added lithium hydroxide (4.21 g, 175 mmol) and the reaction mixture was stirred at room temperature o/n. Ethyl acetate (300 mL) and water (300 mL) were added to the mixture and the aqueous phase was separated. The ethyl acetate layer washed extracted with water (100 mL). The combined aqueous phases were washed with dichloromethane (100 mL) and acidified (pH < 2) by addition of 2M HCl-solution (90 mL). The water layer was extracted with ethyl acetate (3x250 mL). The combined ethyl acetate layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure and dried in vacuo to give 15.72 g of the title compound (Yield: 96.7% over 4 steps). (1R,3R)-3-(tert-butoxycarbonylamino)cyclohexanecarboxylic acid (a) Ethyl (1R,5S)-5-(1,3-dioxoisoindolin-2-yl)cyclohex-3-ene-1-carboxy late To an ice-cold (0 °C) solution of ethyl (1R,5R)-5-hydroxycyclohex-3-ene-1-carboxylate (Intermediate RP2, 15.0 g, 88.13 mmol), phthalimide (14.26 g, 96.94 mmol) and triphenylphosphine (34.67 g, 132.2 mmol) in toluene (264 mL) was added dropwise diisopropyl azodicarboxylate (26.02 mL, 132.2 mmol) in 10 min. The reaction mixture was stirred at 0 °C for 30 min and then allowed to come to room temperature and stirred for 3 h. The mixture was concentrated under reduced pressure to give a yellow oil. Heptane/ethyl acetate = 7/3 v/v% (500 mL) was added and the mixture was heated to 70 °C. After cooling, the mixture was stirred for 72 h at room temperature. The solids were filtered, washed with heptane/ethyl acetate = 9/1 v/v% (2x50 mL) and the filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (heptane/ethyl acetate = 9/1 to 6/4 v/v%) to give 21.96 g of the title compound (Yield: 83.0%) as fluffy off-white solids. (b) Ethyl (1R,3R)-3-(1,3-dioxoisoindolin-2-yl)cyclohexanecarboxylate To a solution of ethyl (1R,5S)-5-(1,3-dioxoisoindolin-2-yl)cyclohex-3-ene-1-carboxy late (27.73 g, 97.19 mmol) in methanol (975 mL) was added 2.7 g of 10% Pd/C. Catalytic hydrogenation was performed for 3 h at room temperature. The palladium-catalyst was filtered and the filtrate was concentrated in vacuo to afford 27.52 g the title compound (Yield: 94%). (c) Ethyl (1R,3R)-3-aminocyclohexanecarboxylate To a solution of ethyl (1R,3R)-3-(1,3-dioxoisoindolin-2-yl)cyclohexanecarboxylate (26.5 g, 87.95 mmol) in ethanol (440 mL) was added drop-wise hydrazine hydrate (64% in water, 4.68 mL, 96.74 mmol). The reaction mixture was stirred for 30 min. at room temperature and then refluxed for 3 h. Additional hydrazine hydrate (425 µL) was added and stirring under reflux was continued for 2 h. The mixture was concentrated under reduced pressure and dried in vacuo to give the title compound in quantitative crude yield. (d) Ethyl (1R,3R)-3-(tert-butoxycarbonylamino)cyclohexanecarboxylate To a cold (0 °C) stirred suspension of ethyl (1R,3R)-3-aminocyclohexanecarboxylate (91.27mmol, theoretical) in dichloromethane (456ml) was added portion-wise di-tert-butyl dicarbonate (21.91g, 100.4 mmol). The reaction mixture was stirred for 15 min at 0 °C, then allowed to come to room temperature. The mixture was washed with cold 0.5N NaOH-solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (heptane/ethyl acetate = 8/2 to 6/4 v/v%) to give 20.5 g of the title compound (Yield: 83.0% over two steps) as an off-white solid. (e) (1R,3R)-3-(tert-butoxycarbonylamino)cyclohexanecarboxylic acid To a solution of ethyl (1R,3R)-3-(tert-butoxycarbonylamino)cyclohexanecarboxylate (20.5 g, 75.56 mmol) in THF (300 mL) was added a solution of lithium hydroxide (1.8 g, 75.56 mmol) in water (150 mL) and the reaction mixture was stirred at room temperature o/n. Additional lithium hydroxide (0.9 g) was added and stirring was continued for 24 h. at room temperature. The mixture was acidified (pH < 2) by addition of 1M HCl-solution (131 mL). The water layer was separated and extracted with dichloromethane (2x100 mL). The combined organic layers were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was dried in vacuo to give 16.81 g (91%) of (1R,3R)-3-(tert-butoxycarbonylamino)cyclo-hexanecarboxylic acid (Intermediate RP5b). tert-Butyl N-[(1R,3S)-3-hydroxy-1-methyl-butyl]carbamate (a) tert-Butyl N-[(1R)-3-[methoxy(methyl)amino]-1-methyl-3-oxo-propyl]carba mate DiPEA (22.3 mL, 132 mmol) followed by T3P (50 wt% in EtOAc, 34 mL, 57 mmol) and N,O- dimethylhydroxylamine (6.43 g, 66 mmol) were added sequentially to a solution of (R)-N-Boc-3- aminobutyric acid (8.94 g, 44 mmol) in DMF (90 mL). The reaction mixture was stirred at room temperature o/n. Water (50 mL) was added and the aqueous mixture was stirred for 1 h. and then extracted with ethyl acetate. The organic extracts were combined, washed with 5% aq. NaHCO 3 - solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo to give 10.84 g (100%) of the title compound (b) tert-Butyl N-[(1R)-1-methyl-3-oxo-butyl]carbamate Methylmagnesium bromide (3 M in Et 2 O, 32.3 mL, 96.8 mmol) was added dropwise to a solution of tert-butyl N-[(1R)-3-[methoxy(methyl)amino]-1-methyl-3-oxo-propyl]carba mate (10.84 g, 44 mmol) in THF (132 mL) at -15 °C under nitrogen. After stirring at this temperature for 15 min, the mixture was allowed to come to room temperature and stirring was continued for 1 h. The mixture was cooled to 0 °C and sat. aq. NH 4 Cl-solution (40 mL) was added carefully. The aqueous mixture was extracted with ethyl acetate. The organic extracts were combined, washed with water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 3.3 g of tert-butyl N-[(1R)-1-methyl-3-oxo-butyl]carbamate (yield 37%). (c) tert-Butyl N-[(1R,3S)-3-hydroxy-1-methyl-butyl]carbamate (Intermediate RP6) Sodium borohydride (744 mg, 19.68 mmol) was added to a cold (0 °C) solution of tert-butyl N- [(1R)-1-methyl-3-oxo-butyl]carbamate (3.3 g, 16.4 mmol) in ethanol (82 mL) under nitrogen. After stirring at this temperature for 5 min, the mixture was allowed to come to room temperature and stirring was continued for 1 h. The mixture was cooled to 0 °C and sat. aq. NH 4 Cl-solution was added carefully. The aqueous mixture was extracted with ethyl acetate. The organic extracts were combined, washed with water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography using SiO 2 and dichloromethane/TBME = 10/0 to 6/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.93 g (50%) of tert- butyl N-[(1R,3S)-3-hydroxy-1-methyl-butyl]carbamate (Intermediate RP6) and 1.82 g (47%) of tert-butyl N-[(1R,3R)-3-hydroxy-1-methyl-butyl]carbamate. Intermediate RP7 (1R,3R)-3-(tert-butoxycarbonylamino)cyclopentanecarboxylic acid The title compound was prepared according to procedures described in WO2019/236631 starting from (1S)-(+)-2-azabicyclo[2.2.1]hept-5-en-3-one (10 g.) to give 7.79 g (37.1% over 4 steps) of (1R,3R)-3-(tert-butoxycarbonylamino)cyclopentanecarboxylic acid (Intermediate RP7).

tert-Butyl N-[(3S,5R)-5-hydroxytetrahydropyran-3-yl]carbamate (a) 1-Allyloxybut-3-en-2-ol To a cold (0 °C) solution of 2-vinyloxirane (2 mL, 25 mmol) and prop-2-en-1-ol (3.4 mL, 50 mmol) in DMF (50 mL) was added carefully portion-wise sodium hydride (60% in mineral oil, 2 g, 50 mmol). After stirring at 0 °C for 30 min, the mixture was stirred at 50 °C o/n. The mixture was cooled to 0 °C and quenched by addition of 1N HCl-solution (100 mL) and stirred for 1 h, allowing the temperature to come to room temperature. The mixture was extracted with diethyl ether (3x100 mL). The combined organic extracts were washed with 10% w/w LiCl-solution (100 mL) and brine. The organic layer was separated, dried over sodium sulfate, filtered and concentrated under reduced pressure (bath temperature 35 °C, 600 mbar). The resulting residue was purified by column chromatography (pentane/diethyl ether = 95/5 to 1/1 v/v%) to give 1.38 g of the title compound (Yield: 43.0%) as a colourless oil. (b) 3,6-Dihydro-2H-pyran-3-ol To a solution of 1-allyloxybut-3-en-2-ol (1.38 g, 10.8 mmol) in dichloromethane (100 mL) was added Grubbs 1 st generation catalysator (178 mg, 0.22 mmol) and the reaction mixture was stirred at room temperature o/n. The mixture was concentrated under reduced pressure (bath temperature 35 °C, 600 mbar). The resulting residue was purified by column chromatography (pentane/diethyl ether = 95/5 to 1/1 v/v%) to give 566 mg of the title compound (Yield: 52.0%) as a colourless oil. (c) 2-(3,6-Dihydro-2H-pyran-3-yl)isoindoline-1,3-dione To a cold (0 °C) solution of 3,6-dihydro-2H-pyran-3-ol (380 mg, 3.8 mmol), phthalimide (373 mg, 2.53 mmol) and triphenylphosphine (798 mg, 3.03 mmol) in THF (15 mL) was added dropwise a solution of di-tert-butyl azodicarboxylate (698 mg, 3.03 mmol) in THF (4 mL). The reaction mixture was stirred at 0 °C for 30 min and then allowed to come to room temperature and stirred for 3 h. The mixture was evaporated under reduced pressure to give a yellow oil. The resulting residue was purified by column chromatography (dichloromethane/ethyl acetate = 10/0 to 1/9 v/v%) to give 456 mg of the title compound (Yield: 79%). (d) tert-Butyl N-[(3S,5R)-5-hydroxytetrahydropyran-3-yl]carbamate (Intermediate RP8) This compound was prepared in an analogous manner as described Intermediate RP4 steps b to h, using 2-(3,6-dihydro-2H-pyran-3-yl)isoindoline-1,3-dione to afford 220 mg of the title compound (~90% e.e.). Intermediate BP1 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-N-[4-(triflu oromethyl)-2-pyridyl]benzamide (Intermediate BP1) (a) 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)benzoyl chloride To a cold (0 °C) solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoic acid (24.8 g, 100 mmol) in dichloromethane (300 mL) was added a catalytic amount of DMF. A solution of oxalyl chloride (12.9 mL, 150 mmol) was added dropwise. After stirring for 60 min at 0 °C, the reaction mixture was allowed to warm to room temperature and the mixture was stirred o/n. The reaction mixture was concentrated to give 26.33 g of crude 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoyl chloride (yield: 99%). (b) 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-N-[4-(triflu oromethyl)-2-pyridyl]benzamide (Intermediate BP1) To a cold (0 °C) solution of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzoyl chloride (26.33 g, 100 mmol) in acetonitrile (300 mL) was subsequently added 4-(trifluoromethyl)pyridin-2-amine (19.45 g, 120 mmol) and 4-DMAP (14.66 g, 120 mmol) The mixture was stirred under nitrogen atmosphere at 0°C and allowed to warm to room temperature overnight. The reaction mixture was concentrated in vacuo. The crude oily solids were then dissolved in dichloromethane (300 mL) and washed with 5% citric acid (3x, 300 mL), 5% NaHCO 3 (2x300 mL) and brine (200 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The residue was triturated in refluxing heptane (300 mL) for 1-2 hours. The mixture was filtrated and evaporated under reduced pressure. The residue was stirred in 6 N NaOH (140 mL) and THF (140 mL) at room temperature for 4 hrs. Then 250 mL EtOAc was added and the layers were separated. The organic layer was washed with water, 5% citric acid and brine, dried over sodium sulfate and concentrated in vacuo to give 24.4 g of 4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-N-[4-(trifluoromethyl)- 2-pyridyl]benzamide (Intermediate BP1) (yield: 62%) as off-white solids. [4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]boronic acid (Intermediate BP2) To a suspension of 4-aminomethylphenylboronic acid hydrochloride (4.41 g, 23.5 mmol) and 5- fluoro-2-methoxybenzoic acid (4 g, 23.5 mmol) in anhydrous THF (100 mL), under a nitrogen atmosphere, was added successively, N,N-diisopropylethylamine (19.4 mL, 118 mmol) and 1- propanephosphonic acid cyclic anhydride (50 wt% in EtOAc, 23 mL, 35.3 mmol). The reaction mixture was heated under reflux at 70 °C o/n. The mixture was diluted with water and dichloromethane, then partitioned. The aqueous layer was extracted with DCM (2x). The combined organic extracts were filtered over a PE-filter and concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and dichloromethane/methanol = 99/1 to 95/5 v/v%. All fractions containing the title compound were collected and evaporated in vacuo to give 5.07 g of [4-[[(5-fluoro-2-methoxy- benzoyl)amino]methyl]phenyl]boronic acid (Intermediate BP2) (Yield 71%). 4-Methoxy-N-[2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxabor olan-2-yl)phenyl]-1-methyl-indole-2- carboxamide (Intermediate BP3) To 4-methoxy-1-methyl-1H-indole-2-carboxylic acid (24.4 mmol, 5 g) and oxalyl chloride (24.4 mmol, 2.3 mL) in dichloromethane (60 mL) N,N-dimethylformamide (1.22 mmol, 95 µL) was added and the mixture was stirred at room temperature until it formed a clear solution (approximately 4 hours). The mixture was concentrated in vacuo. The residue, 4-methoxy-1-methyl-1H-indole-2-carbonyl chloride, was added to a solution of 4-amino-3-methoxyphenylboronic acid pinacol ester (24.2 mmol, 6.03 g) and 4-dimethylaminopyridine (2.419 mmol, 0.296 g) in pyridine (30 mL) and dichloromethane (30 mL). After stirring at room temperature overnight the reaction mixture was diluted with dichloromethane and 2N hydrochloric acid. The organic layer was separated and the aqueous layer extracted with dichloromethane. The combined organic layers were dried (sodium sulfate) and concentrated in vacuo to give 10.55 g of the title compound. N-(4-Isopropyl-3-methyl-phenyl)-3-(4,4,5,5-tetramethyl-1,3,2 -dioxaborolan-2-yl)benzamide (Intermediate BP4) A solution of 4-DMAP cat (33 mg, 0.27 mmol), EDCI.HCl (516 mg, 191.71 mmol), 3- carboxyphenylboronic acid, pinacol ester (635 mg, 2.56 mmol) and 4-isopropyl-3-methylaniline hydrochloride (500 mg, 2.69 mmol) in DCM (25 mL) was stirred at room temperature o/n.3% aq. Citric acid solution (25 mL) and ethyl acetate (25 mL) were added to the reaction mixture and the mixture was stirred for 5 min at room temperature. The organic layer was separated, washed with 1% aq. citric acid solution (25 mL) and brine (25 mL), dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and ethyl acetate/heptane = 1/4 to 1/1 v/v%. All fractions containing the title compound were collected and concentrated under reduced pressure to give 630 mg (Yield 61.7%). 1-(5-tert-Butylisoxazol-3-yl)-3-[4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)phenyl]urea (Intermediate BP5) (a) 2,2,2-Trichloroethyl N-(5-tert-butylisoxazol-3-yl)carbamate To a cold (0 °) solution of 3-amino-5-tert-butylisoxazole ( 1.o g, 7.13 mmol) and pyridine (632 µL, 7.84 mmol) in THF (40 mL) was added 2,2,2-trichloroethyl chloroformate (1.08 mL, 7.84 mmol) drop- wise within 10 minutes. The resulting suspension was stirred for 1h at 0 °C and a further 4 h at room temperature. The mixture was diluted with ethyl acetate (20 mL) and water (20 mL) was added. The resulting bi-phasic system was stirred 30 min at room temperature. After separation of the layers, the water layer was extracted with EtOAc (2x20 mL). The combined organic layers were washed with water (2x50 mL), 50 mL sat. aq. NaHCO 3 -solution and brine (25 mL), dried (Na 2 SO 4 ), filtered and concentrated to give 2.27 g of the title compound (yield >95%). This was used without further purification. (b) 1-(5-tert-Butylisoxazol-3-yl)-3-[4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)phenyl]urea (Intermediate BP5) 4-Aminophenylboronic acid pinacol ester (183 mg, 0.83 mmol) and N,N-diisopropylethylamine (533 µL, 3.0 mmol) were added to a solution of 2,2,2-trichloroethyl N-(5-tert-butylisoxazol-3-yl)carba- mate (315 mg, 1.0 mmol) in DMSO (20 mL). The reaction mixture was stirred at 100 °C for 24 h. The mixture was poured in a stirred mixture of water (100 mL), brine (100 mL) and EtOAc (50 mL). The resulting bi-phasic system was stirred 30 min at room temperature. After separation of the layers, the water layer was extracted with EtOAc (2x20 mL). The combined organic layers were washed with water/brine = 1/1 v/v% (100 mL), 0.2N HCl-solution (2x100 mL) and brine (50 mL), dried (Na 2 SO 4 ), filtered and concentrated under reduced pressure. The crude residue was triturated with dichloromethane (10 mL) to give 100 mg of the title compound (yield: 25%). 1-(3-Fluorophenyl)-3-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2-yl)phenyl]urea (Intermediate BP6) This compound was prepared in an analogous manner as described in Intermediate BP5 starting from 3-fluoroaniline to afford the title compound (42 mg, 28.9%). Intermediate BP7 N1-(4-fluorophenyl)-N1'-[3-fluoro-4-(4,4,5,5-tetramethyl-1,3 ,2-dioxaborolan-2-yl)phenyl]cyclopropane- 1,1-dicarboxamide (Intermediate BP7) This compound was prepared in an analogous manner as described in Bioorg. Med. Chem. (2015), 23(3), 564-578 starting from commercially available 1-((4-fluorophenyl)carbamoyl)cyclo- propanecarboxylic acid and 4-amino-2-fluorophenylboronic acid pinacol ester to afford the title compound (587 mg, quantitative). Intermediate BP8 N-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-3- (trifluoromethyl)benzamide (Intermediate BP8) This compound was prepared in an analogous manner as described in Bioorg. Med. Chem. (2015), 23(3), 564-578 starting from commercially available 3-(frifluoromethyl)benzoic acid and 4- aminophenylboronic acid pinacol ester to afford the title compound (393 mg, 90%). Intermediate BP9 1-(4-Fluorophenyl)-N-[3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl]-2-oxo-pyridine-3- carboxamide (Intermediate BP9) This compound was prepared in an analogous manner as described in WO2010011538 starting from commercially available methyl 2-oxo-1,2-dihydropyridine-3-carboxylate, 4-fluorophenylboronic acid and 4-amino-2-fluorophenylboronic acid pinacol ester to afford the title compound (390 mg, 86%). 2-[3-Chloro-4-[(3-fluorophenyl)methoxy]phenyl]-4,4,5,5-tetra methyl-1,3,2-dioxaborolane (Intermediate BP10) This compound was prepared in an analogous manner as described in J. Med. Chem. (2017), 60(7), 2944-2962 starting from commercially available 4-bromo-2-chlorophenol and 3-fluorobenzyl bromide to afford the title compound (1.42 g, 93%). 2-Fluoro-N-[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborol an-2-yl)phenyl]benzenesulfonamide (Intermediate BP11) To a stirred solution of 4-amino-3-fluorophenylboronic acid pinacol ester (237mg, 1.0 mmol) and pyridine (242 µL, 3.0 mmol) in DCM (5 mL) was added 2-fluorobenzenesulfonyl chloride (194 mg, 1.0mmol). The reaction mixture was stirred for at room temperature o/n.The mixture was concentrated under reduced pressure and the crude product was purified by chromatography on SiO 2 (heptane to ethyl acetate = 100% to 30%) to give 2-fluoro-N-[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborol an-2- yl)phenyl]benzenesulfonamide (247 mg, 63%). 2-Chloro-4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)aniline (Intermediate BP12) This compound was prepared in an analogous manner as described in ACS Med. Chem. Letters (2021) 12, 1912−1919 starting from commercially available 5-bromo-2-chloro-4-fluoroaniline and bis(pinacolato)diboron to afford the title compound (214 mg, 79%). 4-Methoxy-2-phenyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)quinoline (Intermediate BP13) This compound was prepared in an analogous manner as described in ACS Med. Chem. Letters (2013) 4(7), 627-631 starting from 7-chloro-2-phenyl-quinolin-4-ol to afford the title compound (350 mg, 63%). N-(5-tert-Butylisoxazol-3-yl)-2-[4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)phenyl]acetamide (Intermediate BP14) This compound via an HATU coupling starting from commercially available phenylacetic acid- 4-boronic acid pinacol ester and 3-amino-5-tert-butylisoxazole to afford the title compound (139 mg, 28%).

tert-Butyl N-[(E)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hex-5- enyl]carbamate (Intermediate L1) (a) tert-Butyl N-hex-5-ynylcarbamate A solution of 6-heptynoic acid (0.800 mL, 6.32 mmol), diphenylphosphoryl azide (1.65 mL, 7.66 mmol), and Et 3 N (1.75 mL, 12.6 mmol) in tert-BuOH (6.3 mL) was stirred at reflux for 48 h. The reaction mixture was cooled to rt and diluted with Et 2 O (50 mL) and H 2 O (50 mL). The layers were separated and the aqueous phase was extracted with Et 2 O (2 x 50 mL). The combined organic extracts were washed with H 2 O (100 mL) and brine (100 mL), dried over MgSO 4 , filtered, and concentrated under reduced pressure to give a dark yellow oil. The oil was purified by chromatography on SiO 2 (15% EtOAc in hexanes) to give tert-butyl N-hex-5-ynylcarbamate (0.54 g, 43.3%) as a clear, colourless oil. (b) tert-Butyl N-[(E)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hex-5- enyl]carbamate (Intermediate L1) A solution of tert-butyl N-hex-5-ynylcarbamate (0.54 g, 2.74 mmol), ZrCp 2 (H)Cl (211 mg, 0.82 mmol), Et 3 N (385 µL, 2.76 mmol), and pinacolborane (596 µL, 4.11 mmol) in dichloromethane (2.75 mL) was stirred at reflux for 3 h. The reaction mixture was cooled to room temperature and quenched by dropwise addition of methanol (3.15 mL). The organic solvents were evaporated under reduced pressure to give a cloudy white oil which was taken up in diethyl ether (5 mL) and filtered through a thin pad of boron-doped SiO 2 . After “rinsing” with diethyl ether (25 mL) the filtrate was evaporated under reduced pressure to give 0.79 g (88.7%) a clear, colorless oil. 1 H-NMR showed the presence of reduced acetylene for ~25% therefor the crude product was purified by flash chromatography on boron-doped silica gel in hexane/ethyl acetate = 95/5 to 8/2 v/v% as eluent. The fractions containing the title compound were pooled and concentrated to obtain 481 mg of the title compound (yield: 54.0%). tert-Butyl N-[(E)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hept-6 -enyl]carbamate (Intermediate L2) This compound was prepared in an analogous manner as described in Intermediate L1 starting from 7-heptynoic acid to afford the title compound (742.6 mg, 28.9%). tert-Butyl N-[2-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)allyl]amino]ethyl]-carbamate (Intermediate L3) (a) tert-Butyl N-[2-[methyl(prop-2-ynyl)amino]ethyl]carbamate To a solution of propargyl p-toluenesulfonate (1.05 g, 4.98 mmol) in acetonitrile (25 mL) was subsequently added N-tert-butoxycarbonyl-2-methylamino-ethylamine hydrochloride (4.75 mmol) and potassium carbonate (1.31g, 9.49 mmol) and the reaction mixture was refluxed for 4 h. Water and 5% sodium bicarbonate solution were added to the mixture and the mixture was extracted with ethyl acetate. The combined organic layers were washed with brine and concentrated under reduced pressure. The residue was purified by chromatography on SiO 2 (ethyl acetate) to give tert-butyl N-[2-[methyl(prop-2- ynyl)amino]ethyl]carbamate (0.88 g, 87.3%) as a clear, yellow oil. (b) tert-Butyl N-[2-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)allyl]amino]ethyl]- carbamate (Intermediate L3) A solution of tert-Butyl N-[2-[methyl(prop-2-ynyl)amino]ethyl]carbamate (0.88 g, 4.15 mmol), ZrCp2(H)Cl (321 mg, 1.24 mmol), Et 3 N (584 µL, 4.19 mmol), and pinacolborane (902 µL, 4.19 mmol) in dichloromethane (5 mL) was stirred at reflux for 3 h. The reaction mixture was cooled to room temperature and quenched by dropwise addition of methanol (5.3 mL). The organic solvents were evaporated under reduced pressure to give a cloudy white oil which was taken up in diethyl ether (5 mL) and filtered through a thin pad of boron-doped SiO 2 . After “rinsing” with diethyl ether (25 mL) the filtrate was concentrated under reduced pressure to give a clear, yellow oil. The crude product was purified by flash chromatography on boron-doped silica gel in ethyl acetate as eluent. The fractions containing the title compound were pooled and concentrated to obtain 0.88 g of the title compound. (Yield: 62.4%) tert-Butyl N-[2-[methyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)but-3-enyl]amino]- ethyl]carbamate (Intermediate L4) This compound was prepared in an analogous manner as described in Intermediate L3, starting from N-tert-butoxycarbonyl-2-methylamino-ethylamine hydrochloride and 3-butynyl p- toluenesulfonate to afford the title compound (1.07 g, 52.6%). Methyl 2-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3- enoxy]acetate (Intermediate L5) (a) Methyl 2-but-3-ynoxyacetate To a cold (0 °C) suspension of NaH (60% dispersion in mineral oil, 428 mg, 10.7 mmol) in THF (7 mL) was added but-3-yn-1-ol (624 mg, 8.92 mmol mmol) in THF (1 mL). After stirring at room temperature for 30 min, the mixture was cooled to 0 °C and a solution of methyl 2-bromoacetate (1.36 g, 8.92 mmol) in THF (1 mL) was added dropwise. The reaction mixture was stirred o/n allowing the temperature to come to room temperature. Diethyl ether (25 mL) was added to the mixture and washed with water (25 mL). The water layer was extracted with diethyl ether (2x25 mL). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by chromatography on SiO 2 (pentanes/diethyl ether = 10/0 to 1/1 v/v%) to give methyl 2-but-3-ynoxyacetate (620 mg, 48%) as a clear, colourless oil. (b) Methyl 2-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3- enoxy]acetate (Intermediate L5) This compound was prepared in an analogous manner as described in Intermediate L1 step b, starting from methyl 2-but-3-ynoxyacetate to afford the title compound (480 mg, 42%). Methyl (E)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hex-5-eno ate (Intermediate L6) This compound was prepared in an analogous manner as described in Intermediate L1 step b, starting from methyl 5-hexynoate to afford the title compound (558 mg, 44%). Intermediate L7

Methyl 2-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)allyl]amino]acetate (Intermediate L7) (a) Methyl 2-(prop-2-ynylamino)acetate To a solution of propargylamine (2.0 mL, 31.2 mmol) and triethylamine (4.8 mL, 34.4 mmol) was added slowly methyl bromoacetate (3.25 mL, 34.4 mmol). The reaction mixture was stirred at 50 °C for 12 h. The crude product, obtained after concentration of the mixture in vacuo, was purified by chromatography on SiO 2 (dichloromethane/methanol = 10/0 to 95/5 v/v%) to give methyl 2-(prop-2- ynylamino)acetate (2.63 g, 67%) as an orange coloured oil. (b) Methyl 2-[tert-butoxycarbonyl(prop-2-ynyl)amino]acetate To a cold (0 °C) solution of methyl 2-(prop-2ynylamino)acetate (1.50 g, 11.8 mmol) and DiPEA (3.90 mL, 23.6 mmol) in DCM (40 mL) was added Boc 2 O (2.32 g, 10.6 mmol) and the mixture was stirred o/n. The mixture was washed with 1N KHSO 4 (2x60 mL), brine, dried over sodium sulfate, filtered and concentrated in vacuo to yield the title compound (2.42 g, 86%). (c) Methyl 2-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)allyl]amino]acetate (Intermediate L7) This compound was prepared in an analogous manner as described in Intermediate L1 step b, starting from methyl 2-[tert-butoxycarbonyl(prop-2-ynyl)amino]acetate to afford the title compound (831 mg, 53%). Methyl 2-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyl]amino]acetate (Intermediate L8) This compound was prepared in an analogous manner as described in Intermediate L7, starting from N-methyl-N-prop-2ynyl-amine and methyl bromoacetate to afford the title compound (1.22 g 64%)

Methyl 4-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyl]amino]butanoate (Intermediate L9) This compound was prepared in an analogous manner as described in Intermediate L7, starting from N-methyl-N-prop-2ynyl-amine and methyl 4-bromobutyrate to afford the title compound (344 mg, 16%). Methyl (E,6S)-6-[tert-butyl(dimethyl)silyl]oxy-8-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2-yl)oct-7- enoate (Intermediate L10) (a) Methyl 6-oxo-8-trimethylsilyl-oct-7-ynoate A solution of bis(trimethylsilyl)acetylene (12.8 g, 96 mmol) and methyl 6-chloro-6-oxo- hexanoate (80 mmol) in dichloromethane (100 mL) was added dropwise to a suspension of aluminium chloride (12.8 g, 96 mmol) in dichloromethane (100 mL) at 0 °C and stirred for 2 h allowing the temperature to come to room temperature. The reaction mixture was quenched with ice and saturated aq. citric acid (100 mL) and extracted with diethyl ether. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The brown residue was purified by column chromatography (hexane/ethyl acetate = 99:1 to 9/1 v/v%) on silica gel to afford the title compound (4.86 g, 25.3%) as a yellowish oil. Crude fractions were again purified by column chromatography (hexane/ethyl acetate = 99:1 to 95/5 v/v%) on silica gel to afford the title compound (5.37 g, 27.9%) as a yellowish oil. (b) Methyl (6S)-6-hydroxy-8-trimethylsilyl-oct-7-ynoate A mixture of (1S,2S)-(+)-N-(4-toluenesulfonyl)-1,2-diphenylethylenediamin e (295 mg, 0.806 mmol), dichloro(p-cymene)ruthenium(II)dimer (248 mg, 0.40 mmol) and potassium hydroxide (363 mg, 6.47 mmol) in dichloromethane (10 mL) was stirred at room temperature for 10 min. The solution was treated with water (10 mL) and the colour changed from orange to deep purple. The organic layer was separated and dried over MgSO 4 , filtered and concentrated in vacuo. The residue was dissolved in dichloromethane (2 mL) and added to a solution of methyl 6-oxo-8-trimethylsilyl-oct-7-ynoate (4.86 g, 20.22 mmol) in degassed isopropanol (50 mL) at room temperature. After stirring o/n, the solution was recharged with same amount of pre-treaded Ru-cat. and stirred at room temperature for 2 h., concentrated under reduced pressure and the residue was filtered over silica gel (hexane/ethyl acetate = 98/2 to 9/1 v/v%) to afford the title compound (4.27 g, 87.1%) as a yellow/orange oil. (c) Methyl (6S)-6-hydroxyoct-7-ynoate Methyl (6S)-6-hydroxy-8-trimethylsilyl-oct-7-ynoate (4.27 g, 17.6 mmol) was dissolved in DMF (40 mL) and treated with a solution of potassium fluoride (2.05 g, 35.2 mmol) in water (5 mL) at room temperature. After 30 min, 1 M hydrochloric acid (50 mL) was added and the product was extracted with diethyl ether (3x50 mL). The combined organic layers were washed with brine (30 mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by column chromatography on silica gel (hexane/ethyl acetate = 6/4 v/v%) affording methyl (6S)-6-hydroxyoct-7-ynoate (2.4 g, 80.0%) as a yellowish oil. (d) Methyl (6S)-6-[tert-butyl(dimethyl)silyl]oxyoct-7-ynoate To a solution of methyl (6S)-6-hydroxyoct-7-ynoate (2.4 g, 14.1 mmol) in dichloromethane (25 mL) under nitrogen atmosphere was added at 0 °C tert-butyldimethylsilyl chloride (2.34 g, 15.51 mmol) and imidazole (1.92 g, 28.2 mmol). The mixture was stirred at room temperature o/n and water was added. The aqueous layer was separated over a PE-filter and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (hexane/ethyl acetate = 99/1 to 9/1 v/v%) affording methyl (6S)-6-[tert-butyl(dimethyl)silyl]oxyoct-7-ynoate (3.79 g, 75.6% over two steps) as a colourless oil. (e) Methyl (E,6S)-6-[tert-butyl(dimethyl)silyl]oxy-8-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2-yl)oct-7- enoate (Intermediate L10) To an oven-dried 10 mL Schlenk-tube equipped with a magnetic stirring bar were added Schwartz's reagent (90.6 mg, 0.35 mmol), methyl (6S)-6-[tert-butyl(dimethyl)silyl]oxyoct-7-ynoate (1 g, 3.51 mmol), Et 3 N (49 μL, 0.35 mmol) and pinacolborane (552 µL, 3.69 mmol), under an inert nitrogen atmosphere. The tube was then sealed and the mixture was stirred at 60 °C for 24 hours. The reaction was allowed to cool to room temperature, diluted with diethyl ether, passed through a pad of silica gel and concentrated under reduced pressure at room temperature. The crude mixture was purified by column chromatography using SiO 2 and hexane/ethyl acetate = 99/1 to 9/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 311.6 mg of the title compound (yield: 21.5%). Methyl (E,6R)-6-[tert-butyl(dimethyl)silyl]oxy-8-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2-yl)oct-7- enoate (Intermediate L11) This compound was prepared in an analogous manner as described in Intermediate L10, starting from methyl 6-oxo-8-trimethylsilyl-oct-7-ynoate and of (1R,2R)-(+)-N-(4-toluenesulfonyl)-1,2- diphenylethylenediamine to afford the title compound (295 mg, 20.4%). Methyl (E,5S)-5-[tert-butyl(dimethyl)silyl]oxy-7-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2-yl)hept-6- enoate (Intermediate L12) This compound was prepared in an analogous manner as described in Intermediate L10, starting from methyl 5-oxo-7-trimethylsilyl-hept-6-ynoate and of (1S,2S)-(+)-N-(4-toluenesulfonyl)-1,2- diphenylethylenediamine to afford the title compound (367.6 mg, 20.1%). Methyl (E,5R)-5-[tert-butyl(dimethyl)silyl]oxy-7-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2-yl)hept-6- enoate (Intermediate L13) This compound was prepared in an analogous manner as described in Intermediate L10, starting from methyl 5-oxo-7-trimethylsilyl-hept-6-ynoate and of (1R,2R)-(+)-N-(4-toluenesulfonyl)-1,2- diphenylethylenediamine to afford the title compound (400.9 mg, 21.8%). Intermediate L14

Methyl 4-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)allyl]amino]- butanoate (Intermediate L14) (a) 2-Nitro-N-prop-2-ynyl-benzenesulfonamide To a solution of propargylamine (3.2 mL, 50 mmol) and triethylamine (17.5 mL, 125 mmol) in dichloromethane (100 mL) was added 2-nitrophenylsulfonyl chloride (10.5 g, 47.2 mmol). The reaction mixture was stirred at room temperature o/n. The mixture was washed, after addition of dichloromethane (100 mL), with 1N HCl-solution, water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was triturated with ethyl acetate/heptane to give 2-nitro-N-prop-2-ynyl- benzenesulfonamide (9.57 g, 79%). (b) Methyl 4-[(2-nitrophenyl)sulfonyl-prop-2-ynyl-amino]butanoate To a solution of 4-nitro-N-prop-2-ynyl-benzenesulfonamide (1 g, 4.16 mmol) and K 2 CO 3 (1.15 g, 8.32 mmol) in DMF (12 mL) was added t-butyl-3-bromopropionate (0.9 g, 5 mmol) and the mixture was stirred at 40 °C for 4 h. The reaction mixture was diluted with EtOAc (100 mL) and washed with water (3x100 mL), brine and dried over sodium sulfate, filtered and concentrated in vacuo. The crude mixture was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.5 g of the title compound (yield: 85%). (c) Methyl 4-[tert-butoxycarbonyl(prop-2-ynyl)amino]butanoate To a solution of methyl 4-[(2-nitrophenyl)sulfonyl-prop-2-ynyl-amino]butanoate (0.950 g, 2.79 mmol) and cesium carbonate (1.82 g, 5.58 mmol) in MeCN (15 mL) was added 2-mercaptoethanol (235 µL, 3.35 mmol) This was heated to 40°C for 1 d. Subsequently di-tert-butyl bicarbonate (0.245 g, 1.12 mmol) was added and stirred at rt for 1 h. After addition of ethyl acetate (100 mL) the mixture was washed with 5% aq. NaHCO 3 -solution, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude mixture was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 0.73 g of the title compound (yield: 72%). (d) Methyl 4-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)allyl]- amino]butanoate (Intermediate L14) This compound was prepared in an analogous manner as described in Intermediate L1 step b, starting from methyl 4-[tert-butoxycarbonyl(prop-2-ynyl)amino]butanoate to afford the title compound (304 mg 28%)

Methyl (E)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)oct-7-eno ate (Intermediate L15) (a) Methyl oct-7-ynoate To a solution of oct-7-ynoic acid (1.82 g, 13 mmol) in methanol (15 ml) was added conc.-H 2 SO 4 (4 drops). The reaction mixture was stirred for 5 h. at 70 °C. After cooling of the mixture, ethyl acetate (150 mL) was added and the organic phase was washed with sat. aq. NaHCO 3 -solution, water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1.86 g of methyl oct-7-ynoate (yield: 93%). (b) Methyl 2-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3- enoxy]acetate (Intermediate L15) This compound was prepared in an analogous manner as described in Intermediate L10 step e, starting from methyl oct-7-ynoate to afford the title compound (881 mg, 47%). Methyl (E)-9-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)non-8-eno ate (Intermediate L16) This compound was prepared in an analogous manner as described in Intermediate L15, starting from non-8-ynoic acid to afford the title compound (785 mg, 45%). Ethyl 3-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyl]amino]propanoate (Intermediate L17) (a) Ethyl 3-[methyl(prop-2-ynyl)amino]propanoate N-methyl propargylamine (1.0 g, 18.15 mmol) was added to ethyl acrylate (1.21 g, 12.1 mmol) followed by addition of acidic alumina (24.2 mmol, 2 eq.) and the mixture was stirred at 75°C in a sealed tube for 3 h. The mixture was purified directly by column chromatography using SiO 2 and heptane/ethyl acetate = 10/0 to 3/7 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 2.41 g of the title compound (yield: 106%). (b) Ethyl 3-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyl]amino]propanoate (Intermediate L17) This compound was prepared in an analogous manner as described in Intermediate L10 step e, starting from ethyl 3-[methyl(prop-2-ynyl)amino]propanoate to afford the title compound (112 mg, 24%). Methyl 2-[(E)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pent-4 -enoxy]acetate (Intermediate L18) (a) Methyl 2-pent-4-ynoxyacetate Sodium hydride (60% dispersion in mineral oil, 428 mg, 10.7 mmol) was suspended in THF (7 mL) and cooled to 0 °C. Next, a solution of the 4-pentyn-1-ol (750 mg, 8.92 mmol) in THF (1 mL) was added dropwise. The reaction mixture was allowed to come to room temperature and stirred for 30 min. The resulting suspension was cooled to 0 °C again and methyl 2-bromoacetate (1.36 g, 8.92 mmol) in THF (1 mL) was added dropwise. The resulting mixture was allowed to come to room temperature and stirred o/n. The reaction mixture was diluted with diethyl ether (25 mL) followed by addition of sat. aq. ammonium chloride solution (10 mL). The aqueous layer was extracted twice with diethyl ether (10 mL). The combined organic layers were, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. (first 46 °C, 750 mbar, followed by rt, 70 mbar for 15 minutes) to give an orange/brown liquid. The mixture was purified directly by column chromatography using SiO 2 and pentanes/diethyl ether = 95/5 to 1/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 480 mg of the title compound (yield: 34%). (b) Methyl 2-[(E)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pent-4 -enoxy]acetate (Intermediate L18) This compound was prepared in an analogous manner as described in Intermediate L10 step e, starting from methyl 2-pent-4-ynoxyacetate to afford the title compound (430 mg, 49%). Intermediate L19 Ethyl 5-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyl]amino]pentanoate (Intermediate L19) This compound was prepared in an analogous manner as described in Intermediate L9, starting from N-methylprop-2-yn-1-amine and ethyl 5-bromopentanoate to afford the title compound (550 mg, 30%). Ethyl 3-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)allyl]amino]-propanoate (Intermediate L20) This compound was prepared in an analogous manner as described in Intermediate L17 step a and Intermediate 7 step b and c, starting from propargylamine and ethyl acrylate to afford the title compound (594 mg, 19%). Ethyl 5-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)allyl]amino]-pentanoate (Intermediate L21) This compound was prepared in an analogous manner as described in Intermediate L14, starting from propargylamine and ethyl 5-bromopentanoate to afford the title compound (537 mg, 25%). Methyl (E)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hept-6-en oate (Intermediate L22) This compound was prepared in an analogous manner as described in Intermediate L15, starting from 6-heptynoic acid to afford the title compound (2.15 g, 25%). Methyl 3-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allylo xy]propanoate (Intermediate L23) (a) tert-Butyl 3-prop-2-ynoxypropanoate To a solution of propargyl alcohol (1.36 mL, 23.4 mmol) in THF (20 mL) was added a small lump of sodium (~20.08 mg, 0.874 mmol) and the reaction mixture was heated at 60°C until complete solubilization (30-45 min) of sodium. The reaction mixture was cooled to room temperature and tert- butyl acrylate (2.29 mL, 15.6 mmol) in THF (3 mL) was added dropwise over 10 minutes. After completion of addition, the reaction mixture was stirred 3 h at room temperature. Water (25 mL) was added to the reaction mixture and the bi-phasic system was stirred 30 minutes at room temperature. The layers were separated and the aqueous phase was extracted with ethyl acetate (2 x 25 mL). The combined organic layers were washed with water (2x20 mL), brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give 2.2 g (yield: 76%) of tert-butyl 3-prop-2-ynoxypropanoate as a colourless oil. (b) Methyl 3-prop-2-ynoxypropanoate To a solution of tert-butyl 3-prop-2-ynoxypropanoate (2.2 g, 11.9 mmol) in dichloromethane (50 mL) was added trifluoroacetic acid (8 mL, 119 mmol). The reaction mixture was stirred at room temperature o/n. The mixture was concentrated and traces of trifluoroacetic acid were co-evaporated with toluene and DCM. The residue was purified by flash column chromatography using SiO 2 and dichloromethane/methanol = 10/0 to 9/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo. The residue was dissolved in methanol (5 mL) and conc. H 2 SO 4 (4 drops) was added. The reaction mixture was stirred at 70 °C o/n. After cooling of the mixture, ethyl acetate (150 mL) was added and the organic phase was washed with sat. aq. NaHCO 3 -solution, water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1.34 g of methyl 3- prop-2-ynoxypropanoate (yield: 91%). (c) Methyl 3-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allylo xy]propanoate (Intermediate L23) This compound was prepared in an analogous manner as described in Intermediate L10 step e, starting from methyl 3-prop-2-ynoxypropanoate to afford the title compound (920 mg, 37%). Methyl 3-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3- enoxy]propanoate (Intermediate L24) This compound was prepared in an analogous manner as described in Intermediate L23, starting from but-3-yn-1-ol and tert-butyl acrylate to afford the title compound (1.32 g, 38%). Ethyl 4-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allylo xy]butanoate (Intermediate L25) (a) 3-(2-Bromoethoxy)prop-1-yne Triphenylphosphine (3.93 g, 15 mmol) was added at room temperature, portion-wise to a stirred solution of carbon tetrabromide (4.97 g, 15 mmol) and 2-prop-2-ynoxyethanol (1.00 g, 10 mmol) in DCM (33 mL). The reaction mixture was stirred at room temperature o/n. The mixture was concentrated and the residue was purified by column chromatography using SiO 2 and pentanes/diethyl ether = 95/5 to 8/2 v/v%. All fractions containing the title compound were collected and concentrated in vacuo (500 mbar) to give 730 mg of the title compound (yield: 46%). (b) Diethyl 2-(2-prop-2-ynoxyethyl)propanedioate NaH (60% dispersion in mineral oil, 122 mg, 3.0 mmol) was added carefully to a stirred solution of diethyl malonate (418 μL, 2.75 mmol) in THF (23 mL) at room temperature. The reaction mixture was stirred for 30 min after which a solution of 3-(2-bromoethoxy)prop-1-yne (450 mg, 2.75 mmol) in THF (4.5 mL) was added dropwise, followed by sodium iodide (405 mg, 2.75 mmol). The resulting mixture was stirred at 54 °C for 24 h. The mixture was diluted with water (25 mL) and diethyl ether (25 mL) was added. The organic phase was separated and the water layer was extracted with diethyl ether (2x20 mL). The combined organic extracts were washed with water (2x30 mL), brine (15 mL), dried (Na 2 SO 4 ) filtered and concentrated in vacuo to give 500 mg of the title compound which was used directly in the next step. (c) Ethyl 4-prop-2-ynoxybutanoate To a solution of diethyl 2-(2-prop-2-ynoxyethyl)propanedioate (635 mg, 2.62 mmol) in DMSO (1.3 mL) was added water (94 µL, 5.24 mmol) and lithium chloride (331 mg, 7.87 mmol). The resulting mixture was stirred at 170 °C for 3 h. Water (50 mL), brine (50 mL) and diethyl ether (50 mL) were added to the cooled mixture. After stirring 30 minutes at room temperature, the mixture was filtered over Decalite ® and the layers of the filtrate were separated. The water layer was extracted with diethyl ether (2x25 mL). The combined organic extracts were washed with water (100 mL), brine (25 mL), dried (Na 2 SO 4 ) filtered and concentrated in vacuo (500 mbar). The mixture was concentrated and the residue was purified by column chromatography using SiO 2 and pentanes/diethyl ether = 99/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo (300 mbar) to give 103 mg of the title compound (yield: 46% taking into account content of 50%). (d) Ethyl 4-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allylo xy]butanoate (Intermediate L25) This compound was prepared in an analogous manner as described in Intermediate L10 step e, starting from ethyl 4-prop-2-ynoxybutanoate to afford the title compound (50 mg, 28%). Methyl (2R)-2-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)a llyloxy]propanoate (Intermediate L26) This compound was prepared in an analogous manner as described in Intermediate L18, starting from propargyl bromide and (R)-(+)-methyl lactate to afford the title compound (380 mg, 44%). Methyl 2-[methyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)but-3-enyl]amino]acetate (Intermediate L27) This compound was prepared in an analogous manner as described in Intermediate L3, starting from 3-butynyl p-toluenesulfonate and sarcosine methyl ester, HCl to afford the title compound (372 mg, 37%). Methyl 2-[tert-butoxycarbonyl-[(E)-5-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)pent-4- enyl]amino]acetate (Intermediate L28) This compound was prepared in an analogous manner as described in Intermediate L14, starting from 4-pentyn-1-ol and glycine methyl ester hydrochloride to afford the title compound (1.18 g, 88%). Intermediate L29 Methyl 2-[tert-butoxycarbonyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)but-3- enyl]amino]acetate (Intermediate L29) This compound was prepared in an analogous manner as described in Intermediate L14, starting from but-3-yn-1-ol and glycine methyl ester hydrochloride to afford the title compound (615 mg, 65%). Methyl 3-[tert-butoxycarbonyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)but-3- enyl]amino]propanoate (Intermediate L30) This compound was prepared in an analogous manner as described in Intermediate L14, starting from but-3-yn-1-ol and methyl 3-aminopropanoate hydrochloride to afford the title compound (544 mg, 47%). Methyl (2S)-1-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)b ut-3-enyl]azetidine-2-carboxylate (Intermediate L31) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl (2S)-azetidine-2-carboxylate hydrochloride and but- 3-ynyl 4-methylbenzenesulfonate to afford the title compound (348 mg, 36%). Methyl 3-[methyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)but-3-enyl]amino]-propanoate (Intermediate L32) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl 3-(methylamino)propanoate and but-3-ynyl 4- methylbenzenesulfonate to afford the title compound (574 mg, 40%). Intermediate L33 Methyl 2-[tert-butoxycarbonyl-[(E)-2-[tert-butyl(dimethyl)silyl]oxy -4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)but-3-enyl]amino]acetate (Intermediate L33) This compound was prepared in an analogous manner as described in Intermediate 7 and Intermediate L10, starting from the Weinreb amide of Boc-Gly-OH and TMS-acetylene to afford the title compound (241 mg, 35%). Intermediate L34 Methyl (3R)-3-[methyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)but-3-enyl]amino]-butanoate (Intermediate L34) (a) Methyl (3R)-3-[(2-nitrophenyl)sulfonylamino]butanoate To a solution of (R)-3-amino-butyric acid methyl ester hydrochloride (3 g, 25.6 mmol) and triethylamine (8.5 mL, 64 mmol) in dichloromethane (75 mL) was added 2-nitrophenylsulfonyl chloride (5.8 g, 26.2 mmol). The reaction mixture was stirred at room temperature o/n. The mixture was washed, after addition of dichloromethane (75 mL), with 0.1N HCl-solution, 5% aq. NaHCO 3 -solution, water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude mixture was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 9/1 to 4/6 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give methyl (3R)- 3-[(2-nitrophenyl)sulfonylamino]butanoate (5.6 g, 95%). (b) Methyl (3R)-3-[methyl-(2-nitrophenyl)sulfonyl-amino]butanoate To a solution of methyl (3R)-3-[(2-nitrophenyl)sulfonylamino]butanoate (4.09 g, 12.9 mmol) and K 2 CO 3 (3.57 g, 25.8 mmol) in acetonitrile (44 mL) was added methyl iodide (0.84 mL, 13.5 mmol) and the mixture was stirred at 50 °C for 2 d. The reaction mixture was diluted with EtOAc (100 mL) and washed with water (3x100 mL), brine and dried over sodium sulfate, filtered and concentrated in vacuo to give 4.13 g of the title compound (yield: 101%). (c) Methyl (3R)-3-(methylamino)butanoate To a solution of methyl (3R)-3-[methyl-(2-nitrophenyl)sulfonyl-amino]butanoate (1.69 g, 5.34 mmol) and cesium carbonate (3.49 g, 10.7 mmol) in acetonitrile (20 mL) was added 2-mercaptoethanol (1.1 mL, 15.66 mmol) This was heated to 40°C o/n. The reaction mixture was filtered and acidified with 2 N HCl-solution until pH 1 was reached. Then mixture was then loaded over a SCX-2 column (20 g). The column was washed with acetonitrile until the column was colourless, followed by a solvent switch to methanol and then the product was eluted with 2 N NH3/MeOH to yield methyl (3R)-3- (methylamino)butanoate (0.569 g, 81% ). (d) Methyl (3R)-3-[methyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)but-3-enyl]amino]- butanoate (Intermediate L34) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl (3R)-3-(methylamino)butanoate and but-3-ynyl 4- methylbenzenesulfonate to afford the title compound (980 mg, 57%). Methyl 2-[methyl-[(E)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)pent-4-enyl]amino]acetate (Intermediate L35) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from sarcosine methyl ester hydrochloride and pent-4-ynyl methanesulfonate to afford the title compound (760 mg, 37%). Methyl (2R)-1-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)b ut-3-enyl]azetidine-2-carboxylate (Intermediate L36) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl (2R)-azetidine-2-carboxylate hydrochloride and but- 3-ynyl 4-methylbenzenesulfonate to afford the title compound (1.1 g, 91%). Methyl 4-[methyl-[(E,1S)-1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-diox aborolan-2-yl)allyl]amino]- butanoate (Intermediate L37) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl 4-(methylamino)butanoate hydrochloride and [(1R)-1- methylprop-2-ynyl] 4-methylbenzenesulfonate to afford the title compound (78 mg, 23%). Methyl (3R)-3-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)allyl]amino]butanoate (Intermediate L38) This compound was prepared in an analogous manner as described in Intermediate L34, starting from methyl (3R)-3-(methylamino)butanoate and propargyl p-toluenesulfonate to afford the title compound (215 mg, 24%). Methyl (3R)-3-[tert-butoxycarbonyl-[(E)-4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)but-3- enyl]amino]butanoate (Intermediate L39) This compound was prepared in an analogous manner as described in Intermediate L14, starting from but-3-yn-1-ol and (R)-3-amino-butyric acid methyl ester hydrochloride to afford the title compound (759 mg, 55%). Methyl 2-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allylo xy]acetate (Intermediate L40) This compound was prepared in an analogous manner as described in Intermediate L18, starting from propargyl alcohol and methyl bromoacetate to afford the title compound (359 mg, 41%). Methyl (2S)-1-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)a llyl]azetidine-2-carboxylate (Intermediate L41) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl (2S)-azetidine-2-carboxylate hydrochloride and prop- 2-ynyl 4-methylbenzenesulfonate to afford the title compound (1.86 g, 80%). Methyl 2-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allyl] sulfanylacetate (Intermediate L42) (a) 2-prop-2-ynylsulfanylacetic acid A solution of propargyl bromide in toluene (18.4 mL, 213 mmol) was added to a cold (0 °C) solution of mercaptoacetic acid (13.08 g, 142 mmol) in aqueous ammonia (24%, 250 mL). The reaction mixture was stirred at 0 °C for 40 min. The solution was concentrated, filtered and a sat. aq. NaHCO 3 - solution was added. The solution was washed with dichloromethane. The aqueous phase was carefully acidified with concentrated HCl and extracted with dichloromethane. The organic phase was separated over a PE-filter and concentrated under reduced pressure giving 13.53 g of 2-prop-2-ynylsulfanylacetic acid as a slightly green oil which crystalized slowly to form off-white crystals (Yield: 73.2%). (b) 2-prop-2-ynylsulfanylacetic acid To a solution of 2-prop-2-ynylsulfanylacetic acid (13.53 g, 104 mmol) in methanol (150 mL) was added 10 drops of H 2 SO 4 (conc). The reaction mixture was stirred at reflux for 3 h. The mixture was concentrated under reduced pressure and ethyl acetate was added. The organic was washed carefully with 5% aq. NaHCO 3 -solution, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 14.11 g of the title compound as a slightly brown oil (yield: 94.1%). (c) Methyl 2-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)allyl] sulfanylacetate (Intermediate L42) This compound was prepared in an analogous manner as described in Intermediate 10 step e, starting from 2-prop-2-ynylsulfanylacetic acid to afford the title compound (455 mg, 25%). Methyl (E)-10-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)dec-9-en oate (Intermediate L43) This compound was prepared in an analogous manner as described in Intermediate L15, starting from dec-9-ynoic acid to afford the title compound (801 mg, 39%). Methyl (E)-11-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)undec-10 -enoate (Intermediate L44) This compound was prepared in an analogous manner as described in Intermediate L15, starting from undec-10-ynoic acid to afford the title compound (278 mg, 14%). Ethyl 3-[(E)-1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)but-3-enoxy]propanoate (Intermediate L45) This compound was prepared in an analogous manner as described in Intermediate L23, starting from 4-pentyn-2-ol and tert-butyl acrylate to afford the title compound (1.08 g, 87%). Methyl (E,7S)-7-[tert-butyl(dimethyl)silyl]oxy-9-(4,4,5,5-tetrameth yl-1,3,2-dioxaborolan-2-yl)non-8- enoate (Intermediate L46) This compound was prepared in an analogous manner as described in Intermediate L10, starting from ethyl 7-chloro-7-oxo-heptanoate and of (1S,2S)-(+)-N-(4-toluenesulfonyl)-1,2- diphenylethylenediamine to afford the title compound (477 mg, 22.2%). Ethyl (E)-6,6-dideuterio-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)oct-7-enoate (Intermediate L47) (a) Methyl 6,6-dideuterio-6-hydroxy-hexanoate To a cold (0 °C) solution of adipic acid monomethyl ester (958 mg, 5.98 mmol) and triethylamine (917 µL, 6.58 mmol) in THF (9.5 mL) was added drop-wise solution of ethyl chloroformate (629 µL, 6.58 mmol) in THF (7.0 mL). The mixture was stirred for 1 h. allowing the temperature to come to room temperature. The mixture was filtrated and the residue washed with THF (5 mL). The combined filtrates were added dropwise to a solution of NaBD 4 (500 mg, 11.9 mmol) in water (14 mL) at 0 °C. The reaction mixture was stirred for 1 h. The mixture was acidified with 2N aq. HCl-solution (10 mL) to adjust the pH to 3–4 and diethyl ether was added (20 mL). The resulting mixture was stirred 30 minutes at room temperature. The water layer was separated and extracted with diethyl ether (2x10 mL). The combined organic layers were washed with 0.5N NaOH (2x20 mL), water (20 mL) and brine (5.0 mL), dried (Na 2 SO 4 ) and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and pentane/diethyl ether = 4/1 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give methyl 6,6-dideuterio-6-hydroxy- hexanoate (280 mg, 32%). (b) 6,6-Dideuterio-6-hydroxy-hexanoic acid Methyl 6,6-dideuterio-6-hydroxy-hexanoate (280 mg, 1.89 mmol) was dissolved in THF/water = 1/1 v/v% (18 mL) and subsequently lithium hydroxide (50 mg, 2.08 mmol) was added. The mixture was stirred at room temperature o/n. Ethyl acetate (50 mL) and water (were added) and the pH of the mixture was adjusted to pH < 3 by addition of 2M HCl-solution. The organic phase was separated, washed with water, brine, dried over sodium sulfate , filtered and concentrated under reduced pressure to give 230 mg of 6,6-dideuterio-6-hydroxy-hexanoic acid (yield: 91%). (c) 6-Bromo-6,6-dideuterio-hexanoic acid Triphenylphosphine (563 mg, 2.15 mmol) was dissolved in dichloromethane (8 mL) and cooled to -78 °C. N-Bromosuccinimide (382 mg, 2.15 mmol) was added in one portion to the mixture and stirring was continued for 30 minutes at -78 °C. Next, 6,6-dideuterio-6-hydroxy-hexanoic acid (230 mg, 1.72 mmol) dissolved in dichloromethane (8 mL) was added dropwise and the mixture was stirred for 45 min at - 78 °C allowing to come to room temperature. The mixture was diluted with water and thoroughly stirred, 15 minutes at rt. The layers were separated and the water layer was extracted with dichloromethane (2x10 mL). The organic layers were combined and washed with 10% aq. Na 2 S 2 O 4 - solution (20 mL), 0.2N aq. NaOH-solution.2N aq. HCl-solution was added to the alkaline water layer to adjust pH < 3. This water layer was extracted with dichloromethane. The combined organic layers were filtered over a PE filter and concentrated under reduced pressure to afford 230 mg of the title compound (yield: 54%). (d) 6,6-Dideuteriooct-7-ynoic acid A solution of 6-bromo-6,6-dideuterio-hexanoic acid (405 mg, 2.05 mmol) in DMSO (1 mL) was added dropwise to a cold (0 °C) suspension of lithium acetylide ethylenediamine complex (560 mg, 6.09 mmol) in DMSO (2 mL). The resulting mixture was allowed to come to room temperature and stirred 1.5 h. The mixture was cautiously poured, at 0 °C, in a mixture of ice-water (35 mL) and brine (20 mL) and stirred 30 min at 0 °C. At this temperature, 2 N aq. HCl-solution (10 mL) was added followed by ethyl acetate (20 mL) and after stirring 30 minutes at room temperature, the layers were separated. The water layer was extracted with ethyl acetate (2x50 mL). The combined organic layers were washed with brine (25 mL), dried (Na 2 SO 4 ), filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 1/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 6,6-dideuteriooct-7-ynoic acid (200 mg, 69%). (e) Ethyl (E)-6,6-dideuterio-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)oct-7-enoate (Intermediate L47) This compound was prepared in an analogous manner as described in Intermediate L10, starting from ethyl 6,6-dideuteriooct-7-ynoate to afford the title compound (320 mg, 86%). [(E)-3,3-Difluoro-7-methoxy-7-oxo-hept-1-enyl]boronic acid (Intermediate L48) This compound was prepared according to procedures described in Org. Lett. (2020) 22, 2991−2994 afford the title compound. O1-tert-butyl O2-methyl (2R)-4-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)a llyl]piperazine-1,2- dicarboxylate (Intermediate L49) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from 1-tert-butyl 2-methyl (2R)-piperazine-1,2-dicarboxylate and propargyl p-toluenesulfonate to afford the title compound (300 mg, 72%). Methyl 2-[tert-butoxycarbonyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)allyl]amino]- propanoate (Intermediate L50) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from Boc-D-Ala-OMe and propargyl bromide to afford the title compound (520 mg, 40%). Methyl (3R)-1-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)a llyl]pyrrolidine-3-carboxylate (Intermediate L51) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from methyl (3R)-pyrrolidine-3-carboxylate hydrochloride and propargyl p-toluenesulfonate to afford the title compound (410 mg, 40%). tert-Butyl N-[(2R)-2-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyloxy]propyl]carba-mate (Intermediate L52) This compound was prepared in an analogous manner as described in Intermediate L3 step a and Intermediate 10 step e, starting from N-Boc-(R)-1-amino-2-propanol and propargyl bromide to afford the title compound (578 mg, 41%). Methyl (3S)-3-[tert-butoxycarbonyl-[(E)-4-(4,4,5,5-tetramethyl-1,3, 2-dioxaborolan-2-yl)but-3- enyl]amino]butanoate (Intermediate L53) This compound was prepared in an analogous manner as described in Intermediate L14, starting from but-3-yn-1-ol and (S)-3-amino-butyric acid methyl ester hydrochloride to afford the title compound (600 mg, 50%). Methyl (3S)-3-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)allyl]amino]butanoate (Intermediate L54) This compound was prepared in an analogous manner as described in Intermediate L34, starting from (S)-3-amino-butyric acid methyl ester hydrochloride and propargyl p-toluenesulfonate to afford the title compound (630 mg, 95%). Methyl (3S)-3-[methyl-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)but-3-enyl]amino]bu-tanoate (Intermediate L55) This compound was prepared in an analogous manner as described in Intermediate L34, starting from (S)-3-amino-butyric acid methyl ester hydrochloride and 3-butynyl p-toluenesulfonate to afford the title compound (350 mg, 35%). Intermediate L56 tert-Butyl N-[(2R)-2-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxabor olan-2-yl)allyl]amino]pro- pyl]carbamate (Intermediate L56) This compound was prepared in an analogous manner as described in Intermediate L10 step e, starting from tert-butyl N-[(2R)-2-[methyl(prop-2-ynyl)amino]propyl]carbamate to afford the title compound (272 mg, 87%). Ethyl 3-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3- enoxy]butanoate (Intermediate L57) This compound was prepared in an analogous manner as described in Intermediate 23, starting from but-3-yn-1-ol and tert-butyl crotonate to afford the title compound (380 mg, 34%). Ethyl 3-[(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3- enoxy]propanoate (Intermediate L58) This compound was prepared in an analogous manner as described in Intermediate 23, starting from but-3-yn-1-ol and tert-butyl acrylate to afford the title compound (3.89 g, 40%).

Example 1 (a) (1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-7-iodo-3-[4- [[4-(trifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohe xanecarboxylic acid (Scaffold D) To a cold (0 °C) solution of (1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-1-yl]cyclohexanecarboxylic acid (Scaffold C) (1.72 g, 2.55 mmol) in DMF (13 mL) was added N-Iodosuccinimide (544 mg, 2.42 mmol). The reaction mixture was stirred o/n allowing the temperature to raise to room temperature. Still ~6% of starting material present. Additional NIS (29 mg) and the mixture was stirred at room temperature o/n. The mixture was diluted with ethyl acetate and washed with sodium thiosulfate solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and dichloromethane/methanol/acetic acid = 99/1 to 95/5 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.75 g of the title compound (Yield: 86%). (b) (1R,3R)-3-[7-[(E)-6-(tert-butoxycarbonylamino)hex-1-enyl]-4- [(2,4-dimethoxyphenyl)methyl-amino]- 3-[4-[[4-(trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazo lo[4,3-c]pyridin-1-yl]cyclo-hexanecarboxylic acid A mixture of (1R,3R)-3-[7-[(E)-6-(tert-butoxycarbonylamino)hex-1-enyl]-4- [(2,4- dimethoxyphenyl)methyl-amino]-3-[4-[[4-(trifluoromethyl)-2-p yridyl]carbamoyl]phenyl]pyrazolo-[4,3- c]pyridin-1-yl]cyclo-hexanecarboxylic acid (100 mg, 0.125 mmol), tert-butyl N-[(E)-6-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)hex-5-enyl]carbamate (Intermediate L1) (49 mg, 0.15 mmol) and cesium carbonate (122 mg, 0.375 mmol) in DMF/water = 9/1 v/v% (1 mL) was degassed with nitrogen for 5 min at 30 °C. CataCXium® A Pd G3 (4 mg, 0.006 mmol) was added and the mixture was again degassed with nitrogen for 5 min at 30 °C. The reaction mixture was stirred at 65 °C o/n. After cooling ethyl acetate was added and the mixture was stirred for 5 min. The mixture was filtered over Decalite™ and the filtrate was washed with 5% citric acid solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol = 10/0 to 94/6 v/v%) to give 328 mg of the title compound (yield: 81.6%). (c) (1R,3R)-3-[4-Amino-7-[(E)-6-aminohex-1-enyl]-3-[4-[[4-(trifl uoromethyl)-2-pyridyl]carba- moyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohexanecarboxyli c acid hydrochloride (1R,3R)-3-[7-[(E)-7-(tert-butoxycarbonylamino)hept-1-enyl]-4 -[(2,4-dimethoxyphenyl)- methylamino]-3-(3-quinolyl)pyrazolo[4,3-c]pyridin-1-yl]cyclo hexanecarboxylic acid (324 mg, 0.38 mmol) was dissolved in dichloromethane (3 mL).4M HCl/dioxane solution (3 mL) was added and the mixture was stirred at room temperature o/n. The solvent was decantated and the residue triturated with dichloromethane, filtered and dried in vacuo, to give 340 mg the title compound in quantitative crude yield. (d) Example 1 (1R,3R)-3-[4-Amino-7-[(E)-6-aminohex-1-enyl]-3-[4-[[4-(trifl uoromethyl)-2-pyridyl]car- bamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohexanecarboxy lic acid hydrochloride (170 mg, 0.188 mmol) was dissolved in DMF (7.6 mL) and N-ethylmorpholine (119 µL, 0.94 mmol) was added. This solution was added at via a syringe pump at a rate of 0.7 rpm, to a stirred solution of HATU (214 mg, 0.564 mmol) and N-ethylmorpholine (72 µL, 0.564 mmol) in DMF (11.4 mL). Ethyl acetate (50 mL) was added to the reaction mixture and the mixture was washed with 5% NaHCO 3 -solution and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography dichloromethane/methanol = 10/0 to 94/6 v/v%). Purification was performed using preparative HPLC to afford the title compound (41 mg, 36%). Data: LCMS (B) R t : 9.43 min; m/z 604.4 [M+H] + .

Example 2 (a) 2-Trimethylsilylethyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4- (trifluoro-methyl)- 2-pyridyl]carbamoyl]phenyl]pyrazolo[43-c]pyridin-1-yl]cycloh exyl]carbamate Diphenylphosphoryl azide (0.69 mL, 3.2 mmol) was added to a suspension of (1R,3R)-3-[4- [(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4-(trifluoromethyl )-2-pyridyl]carbamoyl]phenyl]- pyrazolo[4,3-c]pyridin-1-yl]cyclohexanecarboxylic acid (Scaffold C) (4.95 g, 7.30 mmol), triethylamine (1.44 mL, 10.3 mmol) in toluene (52 mL) and stirred for 30 min at 100 °C. After cooling to room temperature, 2-(trimethylsilyl)ethanol (1.22 g, 10.3 mmol) was added and the reaction mixture was again stirred for 10 h. at 100 °C. The reaction mixture was diluted with water (50 mL) and the mixture was stirred 30 minutes at room temperature. The layers were separated and the water layer was extracted with toluene (2x100 mL, slow settlement of layers). The combined organic layers were washed with water (100 mL), 1N NaOH (2x100 mL), water (100 mL) and concentrated and the residue was subjected to high vacuum to give 6.10 g 2-trimethylsilylethyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]- 3-[4-[[4-(trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]-pyraz olo[4,3-c]pyridin-1-yl]cyclohexyl]carbamate as a light brown foam (yield >97%). (b) 2-Trimethylsilylethyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-7-iodo-3- [4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-1-yl]cyclohexyl]carbamate To a cold (0 °C) solution of 2-trimethylsilylethyl N-[(1R,3R)-3-[4-[(2,4-dimethoxy- phenyl)methylamino]-3-[4-[[4-(trifluoromethyl)-2-pyridyl]car bamoyl]phenyl]-pyrazolo[4,3-c]pyridin-1- yl]cyclohexyl]carbamate (Scaffold C) (6.1 g, 7.7 mmol) in DMF (35 mL) was added N-Iodosuccinimide (1.91 g, 8.5 mmol). The reaction mixture was stirred 2 h allowing the temperature to raise to room temperature. The mixture was diluted with ethyl acetate and washed with sodium thiosulfate solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 6/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 5.2 g of the title compound (Yield: 74%). (c) 4-[1-[(1R,3R)-3-aminocyclohexyl]-4-[(2,4-dimethoxyphenyl)met hylamino]-7-iodo-pyrazolo-[4,3- c]pyridin-3-yl]-N-[4-(trifluoromethyl)-2-pyridyl]benzamide (Scaffold E) 2-Trimethylsilylethyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-7-iodo-3- [4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-1-yl]cyclohexyl]carbamate (5.4 g, 5.90 mmol) was dissolved in MeCN (59 mL) to give a bright yellow solution. Tetrabutylammonium fluoride, 1M solution in THF (6.5 mL; 6.5 mmol) was added and the resulting mixture was stirred at 60 °C for 18h. The reaction mixture was added dropwise to a stirred -mixture of sat. NaHCO 3 -solution/ethyl acetate (1/1, v/v, 400 mL). The resulting biphasic system was stirred 30 minutes at room temperature. The layers were separated and the water layer was extracted with ethyl acetate (50 mL). The combined organic layers were washed with 5% NaHCO 3 -solution (3x200 mL), brine (75 mL), dried over sodium sulfate, filtered and concentrated to give 4.8 g of the title compound. (Yield: quantitative crude). (d) 2-[(E)-4-[1-[(1R,3R)-3-Aminocyclohexyl]-4-[(2,4-dimethoxyphe nyl)methylamino]-3-[4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-7-yl]but-3-enoxy]acetic acid A mixture of 4-[1-[(1R,3R)-3-aminocyclohexyl]-4-[(2,4-dimethoxyphenyl)met hylamino]-7-iodo- pyrazolo-[4,3-c]pyridin-3-yl]-N-[4-(trifluoromethyl)-2-pyrid yl]benzamide (100 mg, 0.13 mmol), methyl 2- [(E)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)but-3-en oxy]acetate (Intermediate L5) (53 mg, 0.20 mmol) and potassium phosphate tribasic (83 mg, 0.39 mmol) in dioxane/water = 4/1 v/v% (2 mL) was degassed with nitrogen for 5 min at 30 °C. CataCXium® A Pd G3 (4.7 mg, 6.5 µmol) was added and the mixture was again degassed with nitrogen for 5 min at 30 °C. The reaction mixture was stirred at 65 °C o/n. After cooling ethyl acetate was added and the mixture was stirred for 5 min. The mixture was filtered over Decalite™ and lyophilized to give 150 mg of the title compound (yield: quantitative crude). (e) Example 2 2-[(E)-4-[1-[(1R,3R)-3-Aminocyclohexyl]-4-[(2,4-dimethoxyphe nyl)methylamino]-3-[4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-7-yl]but-3-enoxy]acetic acid (150 mg, 0.13 mmol) was added portion-wise to a stirred solution of HATU (148 mg, 0.39 mmol) and N- ethylmorpholine (83 µL, 0.65 mmol) in DMF (11.4 mL). and the reaction mixture was stirred for 4 h. at room temperature. Ethyl acetate (50 mL) was added to the reaction mixture and the mixture was washed with 5% NaHCO 3 -solution and brine, dried over sodium sulfate, filtered and evaporated under reduced pressure. The residue was purified by column chromatography dichloromethane/ethyl acetate = 10/0 to 0/10 v/v%) to give 30 mg of DMB-protected macrocycle Example 2. The product was dissolved in TFA/TIS/Water 90/5/5 v/v% (1 mL) and the mixture was stirred at room temperature for 2 h. The mixture was concentrated and the residue was co-evaporated with DCM (2 mL). The residue was dissolved in DCM/MeOH = 9/1 v/v% (3 mL) and 5% aq. NaHCO 3 -solution (4 mL) was added (pH>8). The layers were separated and the water layer was extracted with DCM/MeOH = 9/1 v/v% (2x3 mL). The combined organic layers were filtered over a PE-filter and concentrated under reduced pressure. Purification was performed using preparative HPLC to afford the title compound (7 mg). Data: LCMS (B) R t : 9.17 min; m/z 606.4 [M+H] + .

Example 3 (a) Benzyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4- (trifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohe xyl]carbamate Diphenylphosphoryl azide (0.39 mL, 1.43 mmol) was added to a suspension of (1R,3R)-3-[4- [(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4-(trifluoromethyl )-2-pyridyl]carba-moyl]phenyl]- pyrazolo[4,3-c]pyridin-1-yl]cyclohexanecarboxylic acid (Scaffold C) (963 mg, 1.43 mmol), triethylamine (0.219 mL, 1.57 mmol) in toluene (15 mL) and stirred for 1 h. at 100 °C. After cooling to room temperature, benzyl alcohol (0.74 mL, 7.15 mmol) was added and the reaction mixture was again stirred for 2 h. at 80 °C. The reaction mixture was diluted with DCM and washed with 5% citric acid solution, 5% aq. NaHCO 3 -solution, brine, dried over sodium sulfate, filtered and evaporated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 to 1/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 652 mg of the title compound (Yield: 58%). (b) Benzyl N-[(1R,3R)-3-[7-bromo-4-[(2,4-dimethoxyphenyl)methylamino]-3 -[4-[[4-(trifluoro-methyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohe xyl]carbamate To a cold (0 °C) solution of benzyl N-[(1R)-3-[(1R)-4-[(2,4-dimethoxyphenyl)-methylamino]-3- [4-[[4-(trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo [4,3-c]pyridin-1-yl]cyclohexyl]carbamate (932 mg, 1.2 mmol) in acetonitrile (35 mL) was added N-bromosuccinimide (2x202 mg, 1.14 mmol). The reaction mixture was stirred 2 h allowing the temperature to raise to room temperature. The mixture was diluted with ethyl acetate and washed with sodium thiosulfate solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 6/4 v/v%. All fractions containing the title compound were collected and evaporated in vacuo to give 856 mg of the title compound (Yield: 83%). (c) 4-[4-Amino-1-[(1R,3R)-3-aminocyclohexyl]-7-bromo-pyrazolo[4, 3-c]pyridin-3-yl]-N-[4- (trifluoromethyl)-2-pyridyl]benzamide Benzyl N-[(1R,3R)-3-[7-bromo-4-[(2,4-dimethoxyphenyl)methylamino]-3 -[4-[[4-(trifluo- romethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin- 1-yl]cyclohexyl]carbamate (855 mg 1 mmol) was dissolved in TFA/H 2 O = 9/1 v/v% (10 mL). The reaction mixture was stirred for 5 h. at 60 °C. The mixture was concentrated under reduced pressure and traces of TFA were co-evaporated with toluene (3x 25 mL). Dark purple oil was dissolved in DCM (50 mL) and water (50 mL) was added. After stirring for 15 min at room temperature, the water layer was separated. Water layer was basified using 2N NaOH-solution, stirred for 15 min. and extracted with DCM/sec-BuOH = 3/2 v/v% (2x50 mL). The combined organic layers was separated over an PE-filter and concentrated under reduced pressure to give 490 mg of the title compound (Yield: 85%). (d) tert-Butyl N-[(1R,3R)-3-[4-amino-7-bromo-3-[4-[[4-(trifluoromethyl)-2-p yridyl]carbamoyl]- phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohexyl]carbamate To a suspension of 4-[4-amino-1-[(1R,3R)-3-aminocyclohexyl]-7-bromo-pyrazolo[4, 3-c]pyridin- 3-yl]-N-[4-(trifluoromethyl)-2-pyridyl]benzamide (485 mg, 0.84 mmol) in DCM (23 mL) was added subsequently Et 3 N (235 µl, 1.69 mmol) and a solution of Boc 2 O (203 mg, 0.93 mmol) in DCM (2 mL). The reaction mixture was stirred for 2 h. at room temperature. DCM (10 mL) was added to the mixture and the organic phase was washed with water and brine. The DCM-layer was separated by filtration over an PE-filter and concentrated under reduced pressure to give 590 mg (100%) of the title compound. (e) Methyl (E)-7-[4-amino-1-[(1R,3R)-3-(tert-butoxycarbonylamino)cycloh exyl]-3-[4-[[4-(trifluoromethyl)- 2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-7-yl]hept- 6-enoate A mixture of tert-butyl N-[(1R,3R)-3-[4-amino-7-bromo-3-[4-[[4-(trifluoromethyl)-2-p yri- dyl]carbamoyl]-phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohexyl ]carbamate (150 mg, 0.22 mmol), methyl (E)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hept-6-en oate (Intermediate L22) (78 mg, 0.29 mmol) and potassium phosphate tribasic (140 mg, 0.66 mmol) in dioxane/water = 4/1 v/v% (2.6 mL) was degassed with nitrogen for 5 min at 30 °C. CataCXium® A Pd G3 (8 mg, 0.012 mmol) was added and the mixture was again degassed with nitrogen for 5 min at 30 °C. The reaction mixture was stirred for 1 h at 100 °C under microwave radiation. After cooling ethyl acetate was added and the mixture was stirred for 5 min. The mixture was filtered over Decalite™ and the filtrate was washed with 5% citric acid solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol = 10/0 to 9/1 v/v%) to give 282 mg of the title compound (yield: 92%). (f) Lithium (E)-7-[4-amino-1-[(1R,3R)-3-aminocyclohexyl]-3-[4-[[4-(trifl uoromethyl)-2-pyridyl]carba- moyl]phenyl]pyrazolo[4,3-c]pyridin-7-yl]hept-6-enoate To a solution of methyl (E)-7-[4-amino-1-[(1R,3R)-3-(tert-butoxycarbonylamino)cycloh exyl]-3- [4-[[4-(trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo [4,3-c]pyridin-7-yl]hept-6-enoate (282 mg 0.38 mmol) in dioxane (2.1 mL) was added 4N HCl/dioxane (2.1 mL). The reaction mixture was stirred for 3 h at room temperature. The mixture was concentrated under reduced pressure and the residual oil dissolved in DCM (50 mL) and water (50 mL) was added. After stirring for 15 min at room temperature, the water layer was separated. The Water layer was basified using 2N NaOH-solution, stirred for 15 min. and extracted with DCM/sec-BuOH = 3/2 v/v% (2x50 mL). The combined organic layers was separated over an PE-filter and concentrated under reduced pressure. The crude product was dissolved in dioxane (3.8 mL) and subsequently water (630 µL) and 0.5N LiOH-solution (630 µL) were added. The mixture was stirred at room temperature o/n. The mixture was lyophilized to give 226 mg of the title compound (yield: quantitative crude). (g) Example 3 Lithium (E)-7-[4-amino-1-[(1R,3R)-3-aminocyclohexyl]-3-[4-[[4-(trifl uoromethyl)-2-pyridyl]car- bamoyl]phenyl]pyrazolo[4,3-c]pyridin-7-yl]hept-6-enoate (226 mg, 0.3 mmol) was dissolved in DMF (12 mL) and N-ethylmorpholine (76 µL, 0.6 mmol) was added. This solution was added at batchwise (20 x 600 µL every 5 min), to a stirred solution of HATU (342 mg, 0.9 mmol) and N-ethylmorpholine (114.5 µL, 0.9) in DMF (4.6 mL). The mixture was stirred at room temperature for 30 min after addition was complete. Ethyl acetate (200 mL) was added to the reaction mixture and the mixture was washed with 5% NaHCO 3 -solution and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography dichloromethane/methanol = 10/0 to 9/1 v/v%). Purification was performed using preparative HPLC to afford the title compound (95 mg, 52%). Data: LCMS (B) R t : 9.15 min; m/z 604.4 [M+H] + .

Example 4 (a) [(1S,3R)-3-(tert-butoxycarbonylamino)cyclohexyl] 4-methylbenzenesulfonate To a solution of tert-butyl N-[(1R,3S)-3-hydroxycyclohexyl]carbamate (Intermediate RP4) (1 g, 4.64 mmol) in dichloromethane (10 mL) was added triethylamine (712 µL, 5.11 mmol) and 4-DMAP (57 mg, 0.46 mmol) and the mixture was stirred for 5 min. p-toluenesulfonyl chloride (5.15 g, 27.0 mmol) was added portion-wise over a period of 4 h and the reaction mixture was stirred at room temperature for 7 days. The mixture was washed with 5% citric acid solution, filtered over a PE-filter and concentrated under reduced pressure.The resulting residue was purified by column chromatography (heptane/ethyl acetate = 8/2 v/v%) to give 1.3 g of the title compound (Yield: 75.8%). (b) tert-butyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-py razolo[4,3-c]pyri-din-1- yl]cyclohexyl]carbamate To a solution of N-[(2,4-dimethoxyphenyl)methyl]-3-iodo-1H-pyrazolo[4,3-c]pyr idin-4-amine (Scaffold A) (1.62 g, 3.95 mmol) in DMF (20 mL) was added cesium carbonate (1.93 g, 5.93 mmol) and the mixture was stirred for 30 min at room temperature. The mixture was warmed up to 70 °C and a solution of [(1S,3R)-3-(tert-butoxycarbonylamino)cyclohexyl] methanesulfonate (1.46 g, 3.95 mmol) in DMF (10 mL) was added dropwise. The mixture was stirred at 80 °C for 3 h. The mixture was diluted with ethyl acetate and washed with water. The organic layer was separated, washed with 5% citric acid, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol = 98/2 to 9/1 v/v%) to give the title compound (512 mg, 21.4%). (c) tert-Butyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[(5 -fluoro-2-methoxy- benzoyl)amino]methyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cycl ohexyl]carbamate tert-butyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-py razolo[4,3-c]pyridin-1- yl]cyclohexyl]carbamate (512 mg, 0.84 mmol) was dissolved in dioxane/water = 2/1 v/v% (6.3 mL) and potassium carbonate (583 mg, 4.22 mmol) was added. The solution was purged with nitrogen for 5 min and [4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]-phenyl]boroni c acid (Intermediate BP2) (282 mg, 0.93 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (35 mg, 0.084 mmol) were added. The reaction mixture was stirred microwave irradiation for 20 min at 120 °C. The reaction mixture was diluted with ethyl acetate and filtered over Decalite™. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 10/0 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 484 mg of the title compound (Yield 78%). (d) Example 4 This compound was prepared in an analogous manner as described in Method C steps b-g, using tert-butyl N-[(1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[(5 -fluoro-2-methoxy- benzoyl)amino]methyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cycl ohexyl]-carbamate and finally methyl (E)- 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hept-6-enoate (Intermediate L22) to afford crude Example 4. Purification was performed using preparative HPLC to afford the title compound (27.7 mg, 48.2%). Data: LCMS (B) R t : 8.07 min; m/z 597.4 [M+H] + . Example 5 (a) tert-Butyl N-[(1R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-pyraz olo[4,3-c]pyridin-1-yl]-1- methyl-propyl]carbamate To an ice-cold (4 °C) suspension of N-[(2,4-dimethoxyphenyl)methyl]-3-iodo-1H-pyrazolo[4,3- c]pyridin-4-amine (Scaffold A) (5 g, 12.19 mmol), tert-butyl N-[(1R)-3-hydroxy-1-methyl- propyl]carbamate (2.77 g, 14.63 mmol) and triphenylphosphine (3.84 g, 14.63) in THF (122 mL) was added dropwise a solution of diisopropyl azodicarboxylate (2.88 mL, 14.63 mmol) in THF (25 mL). The mixture was stirred for 30 min at 4 °C and then allowed to warm to room temperature and stirred o/n. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 1/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 5.73 g of tert-butyl N-[(1R)-3-[4-[(2,4- dimethoxyphenyl)methylamino]-3-iodo-pyrazolo[4,3-c]pyridin-1 -yl]-1-methyl-propyl]carbamate (yield: 81%). (b) tert-Butyl N-[(1R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4-(tr ifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]-1-meth yl-propyl]carbamate tert-Butyl N-[(1R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-pyraz olo[4,3-c]pyridin-1-yl]- 1-methyl-propyl]carbamate (800 mg, 1.38 mmol) was dissolved in dioxane/water = 3/1 v/v% (25 mL) and potassium carbonate (951 mg, 4.22 mmol) was added. The solution was purged with nitrogen for 5 min and 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-N-[4-(triflu oromethyl)-2-pyridyl]benzamide (Intermediate BP1) (593 mg, 6.88 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (56 mg, 0.07 mmol) were added. The reaction mixture was stirred for 2 h at 100 °C. The reaction mixture was diluted with ethyl acetate and filtered over Decalite™. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 776 mg of the title compound (Yield 78%). (d) Example 5 This compound was prepared in an analogous manner as described in Method C steps b-g, using tert-butyl N-[(1R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4-(tr ifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]-1-meth yl-propyl]-carbamate and finally methyl (E)- 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)hept-6-enoate (Intermediate L22) to afford crude Example 5. Purification was performed using preparative HPLC to afford the title compound (23 mg, yield 36%). Data: LCMS (B) R t : 8.11 min; m/z 578.4 [M+H] + .

Example 6 (a) tert-Butyl (3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-pyrazolo [4,3-c]pyridin-1- yl]piperidine-1-carboxylate To an ice-cold (4 °C) suspension of N-[(2,4-dimethoxyphenyl)methyl]-3-iodo-1H-pyrazolo[4,3- c]pyridin-4-amine (Scaffold A) (1 g, 2.44 mmol), tert-butyl (3S)-3-hydroxy-piperidine-1-carboxylate (638 mg, 3.17 mmol) and triphenylphosphine (1 g, 3.83) in toluene (19 mL) was added dropwise a solution of di-2-methoxyethyl azodicarboxylate (792 mg, 3.83 mmol) in toluene (5 mL). The mixture was stirred for 30 min at 4 °C and then allowed to warm to room temperature and stirred o/n. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 45/55 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 480 mg of tert-butyl (3R)-3-[4-[(2,4- dimethoxyphenyl)methylamino]-3-iodo-pyrazolo[4,3-c]pyridin-1 -yl]piperidine-1-carboxylate (yield: 33%). (b) tert-Butyl (3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4-(trifl uoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]piperid ine-1-carboxylate tert-Butyl (3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-iodo-pyrazolo [4,3-c]pyridin-1- potassium carbonate (580 mg, 4.2 mmol) was added. The solution was purged with nitrogen for 5 min and 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-N-[4-(triflu orome-thyl)-2-pyridyl]benzamide (Intermediate BP1) (603 mg, 1.54 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (57 mg, 0.07 mmol) were added. The reaction mixture was stirred for 1 h at 80 °C. The reaction mixture was diluted with ethyl acetate and filtered over Decalite™. The filtrate was concentrated under reduced pressure to give 1.1 g of the title compound (quant.). (c) tert-Butyl (3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-7-iodo-3-[4-[[4 -(trifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]piperid ine-1-carboxylate To a cold (0 °C) solution of tert-butyl (3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-3-[4-[[4- (trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c] pyridin-1-yl]piperidine-1-carboxylate (1.1 g, 1.40 mmol) in DMF (15 mL) was added N-iodosuccinimide (346 mg, 1.54 mmol). The reaction mixture was stirred for 2 h allowing the mixture to warm up to room temperature. The mixture was diluted with ethyl acetate and washed with sodium thiosulfate solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 1/1 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.06 g of the title compound (Yield: 88%). (d) Example 6 This compound was prepared in an analogous manner as described in Method C steps c-g, using tert-butyl (3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-7-iodo-3-[4-[[4 -(trifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]piperid ine-1-carboxylate and finally methyl (E)-9- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)non-8-enoate (Intermediate L16) to afford crude Example 6. Purification was performed using preparative HPLC to afford the title compound (10 mg, 16.2%). Data: LCMS (B) R t : 10.15 min; m/z 618.5 [M+H] + .

Example 7 (a) (1R,3R)-3-[7-allyl-4-[(2,4-dimethoxyphenyl)methylamino]-3-[4 -[[4-(trifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohe xanecarboxylic acid A mixture of (1R,3R)-3-[4-[(2,4-dimethoxyphenyl)methylamino]-7-iodo-3-[4- [[4-(trifluoromethyl)- 2-pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclo hexanecarboxylic acid (Scaffold D) (736 mg, 0.92 mmol), 2-allyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (541 mg, 3.22 mmol) and cesium fluoride (559 mg, 3.68 mmol) in dioxane (9 mL) was degassed with nitrogen. CataCXium® A Pd G3 (29 mg, 0.04 mmol) was added the reaction mixture was stirred for 2 h at 65 °C. After cooling ethyl acetate was added and the mixture was stirred for 5 min. The mixture was filtered over Decalite™ and the filtrate was washed with 5% citric acid solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residue was triturated with diethyl ether to give 948 mg of the title compound ( yield: 101%) (b) 4-[7-Allyl-4-[(2,4-dimethoxyphenyl)methylamino]-1-[(1R,3R)-3 -(hex-5-enylcarbamoyl)cyclohexyl]- pyrazolo[4,3-c]pyridin-3-yl]-N-[4-(trifluoromethyl)-2-pyridy l]benzamide (1R,3R)-3-[7-allyl-4-[(2,4-dimethoxyphenyl)methylamino]-3-[4 -[[4-(trifluoromethyl)-2- pyridyl]carbamoyl]phenyl]pyrazolo[4,3-c]pyridin-1-yl]cyclohe xanecarboxylic acid (250 mg, 0.35 mmol) and 1-amino-5-hexene (35 mg, 0.35 mmol) were suspended in DMF (1.5 ml). N-ethylmorpholine (89 µl, 0.70 mmol) and HATU (148 mg, 0.39 mmol) were added subsequently and the mixture stirred at room temperature o/n. The mixture was washed with 5% NaHCO 3 -solution/brine. The organic layer was separated and washed with 5% aq. Citric acid solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and dichloromethane/ethyl acetate = 10/0 to 1/9 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 238 mg of the title compound (Yield: 85%). (c) mixture of DMB-protected trans/cis macrocycle A solution of 4-[7-allyl-4-[(2,4-dimethoxyphenyl)methylamino]-1-[(1R,3R)-3 -(hex-5-enylcarba- moyl)cyclohexyl]pyrazolo[4,3-c]pyridin-3-yl]-N-[4-(trifluoro methyl)-2-pyridyl]benzamide (50 mg, 0.06 mmol) was dissolved in dichloromethane (1.5 mL) and added dropwise to a solution of Hoveyda-Grubbs Catalyst 2nd Generation (5 mg, 6 µmol) in dichloromethane (4.5 mL). The resulting solution was stirred 24 h at reflux in a sealed tube (5 mL). After TLC indicated that the reaction was not complete thehis procedure was repeated. The mixture was concentrated under reduced pressure and the residue was purified by column chromatography using SiO 2 and dichloromethane/ethyl acetate = 10/0 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 36 mg of a mixture of DMB-protected trans- and cis-macrocycle. (48%). (d) Example 7a and Example 7b Deprotection of the DMB-protected trans/cis-macrocycle was performed as described in Example 2 step e using TFA/TIS/water = 90/5/5 v/v%. Purification was performed using preparative HPLC to afford the two separated trans and cis-isomers of the title compound. Example 7a is the first eluting isomer, which corresponds with the trans-isomer (16 mg, 32.2%) Data: LCMS (B) R t : 9.76 min; m/z 618.5 [M+H] + . Example 7b is the last eluting isomer, which corresponds with the cis-isomer (16 mg, 32.2%) Data: LCMS (B) R t : 10.34 min; m/z 618.5 [M+H] + .

Example 8 This compound was prepared in an analogous manner as described in Method G steps a-d, starting from 4-[1-[(1R,3R)-3-aminocyclohexyl]-4-[(2,4-dimethoxyphenyl)met hyl-amino]-7-iodo- pyrazolo[4,3-c]pyridin-3-yl]-N-[4-(trifluoromethyl)-2-pyridy l]benzamide (Scaffold E) to afford crude Example 8. Purification was performed using preparative HPLC to afford the two separated trans and cis-isomers of the title compound. Example 8a is the first eluting isomer, which corresponds with the trans-isomer (8 mg, 24.4%) Data: LCMS (B) R t : 8.91 min; m/z 618.5 [M+H] + . Example 8b is the last eluting isomer, which corresponds with the cis-isomer (6 mg, 15.5%) Data: LCMS (B) R t : 9.60 min; m/z 618.5 [M+H] + .

Example 9 To a solution of Example 7a and 7b (15 mg, 0.022 mmol) in methanol (15 mL) was added 14 mg of 10% Pd/C. Catalytic hydrogenation was performed at room temperature for 16 h. The palladium- catalyst was filtered and the filtrate was concentrated in vacuo to give 11 mg of the title compound (Yield: quantitative). Data: LCMS (B) R t : 10.17 min; m/z 620.5 [M+H] + .

Example 10 (a) Benzyl (3R)-3-[(3-chloropyrazin-2-yl)methylcarbamoyl]piperidine-1-c arboxylate To a solution of (R)-piperidine-1,3-dicarboxylic acid 1-benzyl ester (8.3 g, 31.6 mmol) in dichloromethane (143 mL) was added subsequently, triethylamine (15.4 mL, 110 mmol) and HATU (12.0 g, 31.6 mmol). The resulting suspension was stirred at room temperature for 1 h. after which (3- chloropyrazin-2-yl)methanamine hydrochloride (7.68 g, 42.7 mmol) was added. The resulting mixture was stirred at room temperature o/n. The reaction mixture was filtered over a Buechner filter. The filtrate was washed with 5% aq. NaHCO 3 -solution (150 mL), 5% aq. citric acid solution (150 mL), water (150 mL), dried over sodium sulfate filtered and concentrated under reduced pressure to give 14.5 g of the title compound (quant. crude yield). The product was used directly in the next step. (b) Benzyl (3R)-3-(8-chloroimidazo[1,5-a]pyrazin-3-yl)piperidine-1-carb oxylate Benzyl (3R)-3-[(3-chloropyrazin-2-yl)methylcarbamoyl]piperidine-1-c arboxylate (14.5 g, 31.6 mmol) was dissolved in acetonitrile (130 mL), phosphorus oxychloride (14.7 mL, 158 mmol) was added and the mixture was stirred for 7 h. at 80 °C and at room temperature o/n. The mixture was added carefully to 25% ammonia (400 mL) and crushed ice (700 mL) keeping the temperature below 0 °C. Ethyl acetate (400 mL) was added and the resulting mixture was stirred for 30 min. The water layer was separated and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated in vacuo to give 9 g of the title compound (yield: 77%). (c) Benzyl (3R)-3-(1-bromo-8-chloro-imidazo[1,5-a]pyrazin-3-yl)piperidi ne-1-carboxylate N-Bromosuccinimide (4.74 g, 26.6 mmol) was added to a cold (0 °C) solution of benzyl (3R)-3- (8-chloroimidazo[1,5-a]pyrazin-3-yl)piperidine-1-carboxylate (9 g, 24.2 mmol) in DMF (98 mL). The reaction mixture was stirred for 3 h. at 0 °C. The mixture was quenched with 10% aq. Na 2 S 2 O 4 - solution/5% aq. NaHCO 3 -solution/brine and ethyl acetate. The phases were separated and the water layer was extracted with ethyl acetate. The combined organic phases were washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 to 1/4 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 9.4 g of the title compound (Yield: 86%). (d) Benzyl (3R)-3-(8-amino-1-bromo-imidazo[1,5-a]pyrazin-3-yl)piperidin e-1-carboxylate Benzyl (3R)-3-(1-bromo-8-chloro-imidazo[1,5-a]pyrazin-3-yl)piperidi ne-1-carboxylate (1 g, 2.22 mmol) was suspended in 25% ammonia (6 mL, 40 mmol) and placed in a sealed tube. The mixture was stirred at 120 °C o/n. After cooling, the mixture was concentrated in vacuo, dissolved in ethyl acetate and washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 860 mg (90%) of the title compound. (e) Benzyl (3R)-3-[8-amino-1-[4-[[4-(trifluoromethyl)-2-pyridyl]carbamo yl]phenyl]imidazo[1,5-a]pyrazin- 3-yl]piperidine-1-carboxylate Benzyl (3R)-3-(8-amino-1-bromo-imidazo[1,5-a]pyrazin-3-yl)piperidin e-1-carboxylate (860 mg, 2.0 mmol) was dissolved in dioxane/water = 4/1 v/v% (25 mL) and potassium carbonate (1.38 g, 10.0 mmol) was added. The solution was purged with nitrogen for 5 min and 4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-N-[4-(trifluoromethyl)-2-pyridyl]benzamid e (Intermediate BP1) (788 mg, 2.0 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (41 mg, 0.05 mmol) were added. The reaction mixture was stirred for 4 h at 90 °C. The reaction mixture was diluted with ethyl acetate after cooling to room temperature and filtered over Decalite™. The filtrate was washed with water, 0.2M NaOH-solution, water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.0 g of the title compound (Yield: 81%). (f) Benzyl (3R)-3-[8-amino-5-chloro-1-[4-[[4-(trifluoromethyl)-2-pyridy l]carbamoyl]phenyl]-imidazo[1,5- a]pyrazin-3-yl]piperidine-1-carboxylate To a solution of benzyl (3R)-3-[8-amino-1-[4-[[4-(trifluoromethyl)-2-pyridyl]carbamo yl]- phenyl]imidazo[1,5-a]pyrazin-3-yl]piperidine-1-carboxylate (1.00 g, 1.62 mmol) in acetic acid (6 mL) was added N-chlorosuccinimide (217 mg, 1.62 mmol) and the mixture was stirred at 80 °C for 1 h. After cooling to room temperature, the mixture was added dropwise to a stirred mixture of water (240 mL) and ethyl acetate (60 mL) and stirred 30 minutes at room temperature. The layers were separated and the water layer was extracted with ethyl acetate (2x60 mL). The combined organic layers were washed with water (300 mL), aq. Na 2 CO 3 -solution (10.6 g, 100 mmol in 50 mL water), brine (25 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 640 mg of the title compound (Yield: 61%). (g) 4-[8-Amino-5-chloro-3-[(3R)-3-piperidyl]imidazo[1,5-a]pyrazi n-1-yl]-N-[4-(trifluoromethyl)-2- pyridyl]benzamide To benzyl (3R)-3-[8-amino-5-chloro-1-[4-[[4-(trifluoromethyl)-2-pyridy l]carbamoyl]- phenyl]imidazo[1,5-a]pyrazin-3-yl]piperidine-1-carboxylate (640 mg, 0.98 mmol) was added 33% HBr/acetic acid solution (5.9 mL, 34 mmol) and the mixture was left at room temperature for 4 h. The mixture was diluted with water/brine (80 mL) and extracted with dichloromethane. The aqueous phase was neutralized with 1N NaOH-solution (60 mL), and then extracted with dichloromethane/methanol = 9/1 v/v%. The organic layer was separated over a PE-filter. The filtrate was concentrated in vacuo to afford 340 mg of the title compound (yield: 67%). (h) 2-Trimethylsilylethyl (3R)-3-[8-amino-5-chloro-1-[4-[[4-(trifluoromethyl)-2-pyridy l]carbamoyl]- phenyl]imidazo[1,5-a]pyrazin-3-yl]piperidine-1-carboxylate DiPEA (234 µL, 1.32 mmol) was added to a solution of 4-[8-amino-5-chloro-3-[(3R)-3- piperidyl]imidazo[1,5-a]pyrazin-1-yl]-N-[4-(trifluoromethyl) -2-pyridyl]benzamide (340 mg, 0.66 mmol) in dichloromethane (6.6 mL). Next, 1-[(2-trimethylsilyl)ethoxycarbonyloxy]pyrrolidin-2,5-dione (188 mg, 0.73 mmol) was added and the reaction mixture was stirred at room temperature o/n. The mixture was diluted with dichloromethane (30 mL) and water (30 mL0 was added and the mixture stirred for 30 minutes at room temperature. The aqueous layer was separated and extracted with dichloromethane. The combined organic layers were washed with water (25 mL), filtered over a PE filter and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and ethyl acetate. All fractions containing the title compound were collected and concentrated in vacuo to give 320 mg of the title compound (Yield: 73%). (i) 2-Trimethylsilylethyl (3R)-3-[8-amino-5-[(E)-3-[(5-ethoxy-5-oxo-pentyl)-methyl-ami no]prop-1-enyl]-1- [4-[[4-(trifluoromethyl)-2-pyridyl]carbamoyl]phenyl]imidazo[ 1,5-a]pyrazin-3-yl]piperidine-1-carboxylate A mixture of 2-trimethylsilylethyl (3R)-3-[8-amino-5-chloro-1-[4-[[4-(trifluoromethyl)-2- pyridyl]carbamoyl]-phenyl]imidazo[1,5-a]pyrazin-3-yl]piperid ine-1-carboxylate (160 mg, 0.24 mmol), ethyl 5-[methyl-[(E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)allyl]amino]pentanoate (Intermediate L19) (157 mg, 0.48 mmol) and potassium carbonate (99.4 mg, 0.72 mmol) in dioxane/water = 4/1 v/v% (4.5 mL) was degassed with nitrogen for 5 min at 30 °C. CataCXium® A Pd G3 (11 mg, 0.015 mmol) was added and the mixture was again degassed with nitrogen for 5 min at 30 °C. The reaction mixture was stirred at 100 °C for 60 min under microwave irradiation. After cooling ethyl acetate was added and the mixture was stirred for 5 min. The mixture was filtered over Decalite™ and the filtrate was washed with 5% citric acid solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated under reduced pressure. The residue was purified by column chromatography (dichloromethane/methanol = 10/0 to 9/1 v/v%) to give 40 mg of the title compound. (j) Example 10 This compound was prepared in an analogous manner as described in Method C steps f and g, using 2-trimethylsilylethyl (3R)-3-[8-amino-5-[(E)-3-[(5-ethoxy-5-oxo-pentyl)-methyl-ami no]prop-1- enyl]-1-[4-[[4-(trifluoromethyl)-2-pyridyl]carbamoyl]phenyl] imidazo[1,5-a]pyrazin-3-yl]piperidine-1- carboxylate to afford crude Example 10. Purification was performed using preparative HPLC to afford the title compound (3 mg, 10%). Data: LCMS (B) R t : 6.52 min; m/z 633.4 [M+H] + .

Example 11 (a) (6-Bromo-3-chloro-pyrazin-2-yl)methanamine hydrochloride (6-bromo-3-chloro-pyrazin-2-yl)methanamine hydrochloride was prepared according to procedures described in WO 2013/010380 A1. (b) Benzyl N-[(1R,3R)-3-[(6-bromo-3-chloro-pyrazin-2-yl)methylcarbamoyl ]cyclohexyl]-carbamate To a solution of (1R,3R)-3-(benzyloxycarbonylamino)cyclohexanecarboxylic acid (Intermediate RP5) (800 mg, 2.88 mmol) in dichloromethane (15 mL) was added subsequently, triethylamine (1.2 mL, 8.65 mmol) and HATU (1.1 g, 2.88 mmol). The resulting suspension was stirred at room temperature for 1 h. after which (6-bromo-3-chloro-pyrazin-2-yl)methanamine hydrochloride (747 mg, 2.88 mmol) was added. The resulting mixture was stirred at room temperature o/n. The reaction mixture was filtered over a Buechner filter. The filtrate was washed with 5% aq. NaHCO 3 - solution, 5% aq. citric acid solution, water, dried over sodium sulfate filtered and concentrated under reduced pressure to give 1.73 g of the title compound (quant. crude yield). The product was used directly in the next step. (c) Benzyl N-[(1R,3R)-3-[(6-allyl-3-chloro-pyrazin-2-yl)methylcarbamoyl ]cyclohexyl]carbamate A mixture of benzyl N-[(1R,3R)-3-[(6-bromo-3-chloro-pyrazin-2-yl)methylcarbamoyl ]- cyclohexyl]carbamate (1.73 g, 3.33 mmol), 2-allyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (560 mg, 2.91 mmol) and cesium fluoride (1.52 g, 9.99 mmol) in dioxane (33 mL) was degassed with nitrogen. Pd(dppf)Cl 2 .CH 2 Cl 2 (135 mg, 0.17 mmol) was added the reaction mixture was stirred for 2 h at 100 °C. After cooling ethyl acetate was added and the mixture was stirred for 5 min. The mixture was filtered over Decalite™ and the filtrate was washed with 5% citric acid solution, water and brine. The organic layer was separated, dried over sodium sulfate, filtered and then concentrated under reduced pressure to give 1.62 g of the title compound (yield: quantitative crude). (d) Benzyl N-[(1R,3R)-3-(5-allyl-1-bromo-8-chloro-imidazo[1,5-a]pyrazin -3-yl)cyclohexyl]carba-mate This compound was prepared in an analogous manner as described in Method J steps b and c, using benzyl N-[(1R,3R)-3-[(6-allyl-3-chloro-pyrazin-2-yl)methylcarbamoyl ]cyclo-hexyl]carbamate to afford 680 mg of benzyl N-[(1R,3R)-3-(5-allyl-1-bromo-8-chloro-imidazo-[1,5-a]pyrazi n-3- yl)cyclohexyl]carbamate (Yield: 40%). (e) Benzyl N-[(1R,3R)-3-(5-allyl-8-amino-1-bromo-imidazo[1,5-a]pyrazin- 3-yl)cyclohexyl]carba-mate Benzyl N-[(1R,3R)-3-(5-allyl-1-bromo-8-chloro-imidazo[1,5-a]pyrazin -3-yl)cyclohexyl]- carbamate (680 mg, 1.35 mmol) was suspended in 2N ammonia/isopropanol (18 mL) and placed in a sealed tube. The mixture was stirred at 120 °C for 14 h under microwave irradiation. After cooling, the mixture was concentrated in vacuo, dissolved in ethyl acetate and washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 1/1 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 515 mg of the title compound (Yield: 79%). Example 11 This compound was prepared in an analogous manner as described in Method J step e and g, Method H steps b-d using benzyl N-[(1R,3R)-3-(5-allyl-8-amino-1-bromo-imidazo[1,5-a]pyrazin- 3- yl)cyclohexyl]carba-mate and 5-hexenoic acid to afford crude Example 11. Purification was performed using preparative HPLC to afford the two separated trans and cis-isomers of the title compound. Example 11a is the first eluting isomer, which corresponds with the trans-isomer (5 mg, 9%) Data: LCMS (B) R t : 8.16 min; m/z 604.4 [M+H] + . Example 11b is the last eluting isomer, which corresponds with the cis-isomer (5 mg, 9%) Data: LCMS (B) R t : 8.54 min; m/z 604.4 [M+H] + . Example 15 (a) 3-Bromo-4-chloro-1H-pyrazolo[4,3-c]pyridine To a suspension of 4-chloro-1H-pyrazolo[4,3-c]pyridine (1.61 g, 10.5 mmol) in acetonitrile (50 mL) was added N-bromosuccinimide (1.87 g, 10.5 mmol) and the reaction mixture was stirred under reflux for 3 h. After cooling, the mixture was stirred at room temperature o/n upon which a precipitate formed. The mixture was concentrated and the residue was stirred 1h at room temperature in water/ethanol = 1/9 v/v% (20 mL). Next, additional water/ethanol = 9/1 v/v% was added dropwise and stirring was continued 30 minutes at room temperature. The solids were filtered and the residue was dried in vacuo to give 1.3 g of 3-bromo-4-chloro-1H-pyrazolo[4,3-c]pyridine (yield: 53%). (b) tert-Butyl N-[(1R)-3-(3-bromo-4-chloro-pyrazolo[4,3-c]pyridin-1-yl)-1-m ethyl-propyl]carba-mate To an ice-cold (0 °C) solution of tert-butyl N-[(1R)-3-hydroxy-1-methyl-propyl]carbamate (1.3 g, 6.86 mmol), 3-bromo-4-chloro-1H-pyrazolo[4,3-c]pyridine (1.33 g, 5.72 mmol) and triphenylphosphine (1.80 g, 6.86 mmol) in THF (57 mL) was added dropwise a solution of di-2-methoxyethyl azodicarboxylate (1.61 g, 6.86 mmol) in THF (10 mL). The mixture was stirred for 30 min at 4 °C and then allowed to warm to room temperature and stirred o/n. To the mixture was added water/5% aq. citric acid solution/ethyl acetate=1/1/1 v/v% (150 mL). The resulting mixture was stirred for 30 min at room temperature. The layers were separated and the water layer was extracted with ethyl acetate (50 mL). The combined organic layers were washed with 5% aq. citric acid solution (2x75 mL), water (75 mL), brine (25 mL), dried (Na 2 SO 4 ) filtered and concentrated under reduced pressure. The residue was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.43 g of tert- butyl N-[(1R)-3-(3-bromo-4-chloro-pyrazolo[4,3-c]pyridin-1-yl)-1-m ethyl-propyl]carbamate (isomeric mixture in a 84/16 ratio) (yield: 62%). (c) tert-Butyl N-[(1R)-3-(4-amino-3-bromo-pyrazolo[4,3-c]pyridin-1-yl)-1-me thyl-propyl]carba-mate tert-Butyl N-[(1R)-3-(3-bromo-4-chloro-pyrazolo[4,3-c]pyridin-1-yl)-1-m ethyl-propyl]carbamate (715 mg, 1.77 mmol) was suspended in 2N ammonia/isopropanol (9 mL) and 25% ammonia solution (9 mL) and placed in a sealed tube. The mixture was stirred at 120 °C for 20 h under microwave irradiation. After cooling, the mixture was concentrated in vacuo, dissolved in ethyl acetate and washed with water, brine, dried over sodium sulfate, filtered and concentrated under reduced pressure The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 470 mg of the title compound (Yield: 35%). (d) tert-Butyl N-[(1R)-3-(4-amino-3-bromo-7-iodo-pyrazolo[4,3-c]pyridin-1-y l)-1-methyl-propyl]- carbamate To a cold (0 °C) solution of tert-butyl N-[(1R)-3-(4-amino-3-bromo-pyrazolo[4,3-c]pyridin-1-yl)- 1-methyl-propyl]carbamate (220 mg, 0.57 mmol) in DMF (5.7 mL) was added N-iodosuccinimide (192 mg, 0.85 mmol). The reaction mixture was stirred o/n allowing the mixture to warm up to room temperature. The mixture was diluted with ethyl acetate and washed with sodium thiosulfate solution, water, brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 250 mg of the title compound (Yield: 86%). (e) tert-Butyl N-[3-bromo-1-[(3R)-3-(tert-butoxycarbonylamino)butyl]-7-iodo -pyrazolo[4,3-c]pyridin-4-yl]- N-tert-butoxycarbonyl-carbamate To a suspension of tert-butyl N-[(1R)-3-(4-amino-3-bromo-7-iodo-pyrazolo[4,3-c]pyridin-1-y l)-1- methyl-propyl]carbamate (250 mg, 0.49 mmol) and 4-DMAP (1.5 mg, 0.01 mmol) in dichloromethane (4.9 mL) was added di-tert-butyl dicarbonate (160 mg, 0.74 mmol). The reaction mixture was stirred at room temperature o/n. Dichloromethane (10 mL) and sat. aq. NaHCO 3 -solution were added. The water layer was separated and extracted with dichloromethane (10 mL). The combined organic layers were washed with water (10 mL), 5% aq. citric acid solution (10 mL), water (10 mL), filtered over a PE filter and concentrated to give 340 mg tert-butyl N-[3-bromo-1-[(3R)-3-(tert-butoxycarbonylamino)butyl]-7- iodo-pyrazolo[4,3-c]pyridin-4-yl]-N-tert-butoxycarbonyl-carb amate (yield: 95%). (f) Methyl (E)-8-[4-[bis(tert-butoxycarbonyl)amino]-3-bromo-1-[(3R)-3-( tert-butoxycarbonyl- amino)butyl]pyrazolo[4,3-c]pyridin-7-yl]oct-7-enoate N-[3-bromo-1-[(3R)-3-(tert-butoxycarbonylamino)butyl]-7-iodo -pyrazolo[4,3-c]pyridin-4-yl]-N- tert-butoxycarbonyl-carbamate (240 mg, 0.34 mmol) was dissolved in dioxane/water = 4/1 v/v% (12 mL) and cesium carbonate (326 mg, 1.0 mmol) was added. The solution was purged with nitrogen for 5 min and methyl (E)-8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)oct-7-eno ate (Intermediate L15) (120 mg, 0.43 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (29 mg, 0.034 mmol) were added. The reaction mixture was stirred for 4 h at 82 °C. The reaction mixture was diluted with ethyl acetate after cooling to room temperature and filtered over Decalite™. The filtrate was washed with water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 95/5 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 220 mg of the title compound (Yield: 69%). (g) Methyl (E)-8-[4-amino-1-[(3R)-3-aminobutyl]-3-bromo-pyrazolo[4,3-c] pyridin-7-yl]oct-7-enoate Methyl (E)-8-[4-[bis(tert-butoxycarbonyl)amino]-3-bromo-1-[(3R)-3-( tert-butoxycarbonyl- amino)butyl]pyrazolo[4,3-c]pyridin-7-yl]oct-7-enoate (218 mg, 0.30 mmol) was stirred in a mixture of dichloromethane/TFA = 1/1 v/v% (5 mL) at room temperature o/n. The mixture was concentrated in vacuo. The crude product was co-evaporated with dichloromethane (3x10 mL). The resulting oil was partitioned between dichloromethane (10 mL) and 5% aq. NaHCO 3 -solution (10 mL). The organic layer was separated, washed with water, brine, separated over a PE-filter and concentrated under reduced pressure to give 146 mg of the title product. (h) Lithium (E)-8-[4-amino-1-[(3R)-3-aminobutyl]-3-bromo-pyrazolo[4,3-c] pyridin-7-yl]oct-7-enoate Methyl (E)-8-[4-amino-1-[(3R)-3-aminobutyl]-3-bromo-pyrazolo[4,3-c] pyridin-7-yl]oct-7-enoate (146 mg, 0.3 mmol) was dissolved in dioxane (2 mL) and water (0.5 mL) and subsequently lithium hydroxide (16 mg, 0.66 mmol) was added. The mixture was stirred at room temperature o/n. The mixture was lyophilized to give the title compound in a quantitative crude yield. (i) Macrocyclisation of lithium (E)-8-[4-amino-1-[(3R)-3-aminobutyl]-3-bromo-pyrazolo[4,3-c] pyridin-7- yl]oct-7-enoate Lithium (E)-8-[4-amino-1-[(3R)-3-aminobutyl]-3-bromo-pyrazolo[4,3-c] pyridin-7-yl]oct-7-enoate (149 mg, 0.34 mmol) was dissolved in DMF (20 mL) and N-ethylmorpholine (130 µL, 1.02 mmol) was added. This solution was added at via a syringe pump at a rate of 0.7 rpm, to a stirred solution of HATU (395 mg, 1.04 mmol) and N-ethylmorpholine (216 µL, 1.7 mmol) in DMF (14 mL) and the mixture was stirred at room temperature for 3.5 h. Ethyl acetate (50 mL) was added to the reaction mixture and the mixture was washed with 5% NaHCO 3 -solution and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to afford the macrocyclised compound (65 mg, 47%). (j) Example 15 Macrocycle (20 mg, 0.05 mmol) from step I was dissolved in dioxane/water = 9/1 v/v% (2.2 mL) and cesium carbonate (53 mg, 0.14 mmol) was added. The solution was purged with nitrogen for 5 min and 4-methoxy-N-[2-methoxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxabor olan-2-yl)phenyl]-1-methyl-indole- 2carboxamide (Intermediate BP3) (26 mg 006 mmol) and Pd(dppf)Cl 2 CH 2 Cl 2 (4 mg 0005 mmol) were added. The reaction mixture was stirred for 18 h at 110 °C. The reaction mixture was diluted with ethyl acetate after cooling to room temperature and filtered over Decalite™. The filtrate was washed with water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to afford crude Example 15. Purification was performed using preparative HPLC to afford the title compound (12 mg, 38.3%). Data: LCMS (B) R t : 10.05 min; m/z 636.5 [M+H] + . The following Examples were synthesized following methods as depicted in the tables below.

Example 52 (a) Ethyl 3-[(E)-4-[1-bromo-3-[(1R,3R)-3-(tert-butoxycarbonylamino)cyc lopentyl]-8-chloro-imidazo[1,5- a]pyrazin-5-yl]but-3-enoxy]propanoate This compound was prepared in an analogues manner as described in Method K step a-d, and Method J steps b and c, using Intermediate RP7, Intermediate L58 and (6-bromo-3-chloro-pyrazin-2- yl)methanamine hydrochloride to afford 1.58 g of the title compound. (b) Ethyl 3-[(E)-4-[3-[(1R,3R)-3-aminocyclopentyl]-1-bromo-8-[(2,4-dim ethoxyphenyl)methylamino]- imidazo[1,5-a]pyrazin-5-yl]but-3-enoxy]propanoate To a suspension of ethyl 3-[(E)-4-[1-bromo-3-[(1R,3R)-3-(tert-butoxycarbonylamino)cyc lo- pentyl]-8-chloro-imidazo[1,5-a]pyrazin-5-yl]but-3-enoxy]prop anoate (1.58 g, 2.7 mmol) in 1-butanol (20 mL) was added 2,4-dimethoxybenzylamine (1.21 mL, 8.09 mmol) and the mixture was stirred at 90 °C o/n. The mixture was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate/water 3/1 v/v% (100 mL). The organic layers was separated and washed with 5% NaHCO 3 - solution (25 mL) and brine (10 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and heptane/ethyl acetate = 4/1 to 0/10 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 1.62 g of the title compound (Yield: 83%). (c) Example 52 This compound was prepared in an analogues manner as described in Method J, K and L, starting from ethyl 3-[(E)-4-[3-[(1R,3R)-3-aminocyclopentyl]-1-bromo-8-[(2,4-dim ethoxyphenyl)methyl- amino]imidazo[15a]pyrazin5yl]but3enoxy]propanoate and using pyrrolo[23b]pyridine5boronic acid pinacol ester in the last step to afford crude Example 52. Purification was performed using preparative HPLC to afford the title compound (6 mg, 13%). Data: LCMS (B) R t : 4.35 min; m/z 458.3 [M+H] + . Example 62 (a) Ethyl 3-[(E)-4-[3-[(1R,3R)-3-(tert-butoxycarbonylamino)cyclopentyl ]-8-chloro-imidazo[1,5-a]pyrazin- 5-yl]but-3-enoxy]propanoate This compound was prepared in an analogues manner as described in Method K step a-d, and Method J step, using Intermediate RP7, Intermediate L58 and (6-bromo-3-chloro-pyrazin-2- yl)methanamine hydrochloride to afford 1.42 g of the title compound. (b) Ethyl 3-[(E)-4-[3-[(1R,3R)-3-(tert-butoxycarbonylamino)cyclopentyl ]-8-methyl-imidazo[1,5-a]pyrazin- 5-yl]but-3-enoxy]propanoate Ethyl 3-[(E)-4-[3-[(1R,3R)-3-(tert-butoxycarbonylamino)cyclopentyl ]-8-chloro-imidazo[1,5-a]py- razin-5-yl]but-3-enoxy]propanoate (100 mg, 0.20 mmol) was suspended in dioxane (1 mL) and potassium carbonate (41 mg, 0.30 mmol) was added The solution was purged with nitrogen for 5 min and trimethylboroxine 50% w/w soln. in THF (84 µL, 0.30 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (16 mg, 0.02 mmol) were added. The reaction mixture was stirred for 2 h at 100 °C. The reaction mixture was diluted with ethyl acetate after cooling to room temperature and filtered over Decalite™. The filtrate was washed with water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography using SiO 2 and dichloromethane/methanol = 95/5 v/v%. All fractions containing the title compound were collected and concentrated in vacuo to give 80 mg of the title compound (Yield: 83%). (c) Example 62 This compound was prepared in an analogues manner as described in Method J, K and L, starting from ethyl 3-[(E)-4-[3-[(1R,3R)-3-(tert-butoxycarbonylamino)cyclopentyl ]-8-methyl-imidazo[1,5- a]pyrazin-5-yl]but-3-enoxy]propanoate and using 4-methoxy-N-[2-methoxy-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)phenyl]-1-methyl-indole-2-carboxamid e (Intermediate 3) in the last step to afford crude Example 62. Purification was performed using preparative HPLC to afford the title compound (6 mg, 21.5%). Data: LCMS (B) R t : 9.35 min; m/z 649.5 [M+H] + . The following Examples were synthesized following methods as depicted in the tables below.

Example A Binding kinetics assay wt-BTK, BTK C481S, LCK, FGFR1, FLT3, PDGFR-β, FMS, LYN, MEK1, AUR- B, ITK, VEGFR, EGFR, FGFR3, AXL, c-MET, SRC, YES, ABL, RET and IGFR1 (Surface Plasmon Resonance) Streptavidin-coated chips (Cat. No. BR100531), disposables and maintenance kits for Biacore were purchased from Cytiva (Eindhoven, The Netherlands). Biotinylated wt-BTK enzym (Carna Biosciences, cat. no.08-480-20N), BTK C481S (Carna Biosciences, cat. no.08-417-20N), LCK (Carna Biosciences, cat. no. 08-470-20N), FGFR1 (Carna Biosciences, cat. no. 08-435-20N), FLT3 (Carna Biosciences, cat. no.08-454-20N), PDGFR-β (Carna Biosciences, cat. no.08-458-20N), FMS (Carna Biosciences, cat. no. 08-455-20N), LYN (Carna Biosciences, cat. no. 08-471-20N), MEK1 (Carna Biosciences, cat. no. 07-441-10-20N), AUR-B (Carna Biosciences, cat. no. 05-402-21N), ITK (Carna Biosciences, cat. no.08-481-20N), VEGFR (Carna Biosciences, cat. no.08-491-20N), EGFR (Carna Biosciences, cat. no. 08-415-20N), FGFR3 (Carna Biosciences, cat. no. 08-433-20N), AXL (Carna Biosciences, cat. no. 08-407-20N), c-MET (Carna Biosciences, cat. no. 08-451-20N), SRC (Carna Biosciences, cat. no. 08-473-20N), YES (Carna Biosciences, cat. no. 08-475-20N), ABL (Carna Biosciences, cat. no. 08-401-20N), RET (Carna Biosciences, cat. no. 08-459-20N) or IGFR1 (Carna Biosciences, cat. no.08-441-20N) were immobilized on a streptavidin-coated chip to a level of about 8000 resonance units (RU) using Biacore buffer (50 mM Tris pH 7.5, 0.05 % (v/v) Tween-20, 150 mM NaCl and 5 mM MgCl 2 ) + 1 mM TCEP. Remaining streptavidin was blocked with biocytin. Immobilization was performed at 4°C. Subsequent assay steps were conducted at 22°C. After changing buffer to Biacore buffer with 1 % (v/v) dimethylsulfoxide (DMSO), a pre-run was performed for a period of at least 30 min at a flow rate of 30 µl/min to obtain a stable surface. The kinetic constants of the compounds were determined with single cycle kinetics with five consecutive injections with an increasing compound concentration with ranges of 3.16 – 316 nM. Experiments were performed with an association time of 100 s per concentration and a dissociation time of 1200 s, except for compounds with a long target residence time, such as irreversible inhibitors, where dissociation time was increased. To circumvent problems of mass transport limitation, a flow rate of 30 µl/min was used. A blank run with the same conditions was performed before the compound was injected. The SPR sensorgrams were analyzed with Biacore Evaluation Software by using a method of double referencing. First the reference channel was subtracted from the channel containing immobilized protein. Subsequently, the reference curve obtained with buffer injections was subtracted. The resulting curve was fitted with a 1:1 binding model. Compounds that bound according to an induced fit model were fitted with a two-state reaction model. The kinetic constants (k a , k d , K D ) of duplicates were geometrically averaged. Target residence time (τ) for the 1:1 binding model was calculated from the dissociation constant k d with the formula τ = 1/ k d . Target residence for an induced fit model was calculated as described (Tummino and Copeland, 2008). Example B Binding kinetics assay TEC (Surface Plasmon Resonance) An aliquot of N-terminal His-tagged TEC (Eurofins DiscoverX, cat. no.14-801) was thawed, diluted to 4.8 μg/mL in pre-cooled Biacore buffer (10 mM Tris pH 7.5, 0.05%Tween, 150 mM NaCl and 5 mM MgCl 2 ) and immobilized on a Ni-NTA chip with His-tag capturing, followed by amine-coupling. First, the surface of the chip was first washed for 420 seconds with 0.35 M EDTA, subsequently loaded with 1 mM NiCl for 420 seconds, followed by activation for 1000 seconds with a 1:1 dilution of 0.2 M 1-ethyl- 3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and 50 mM N-hydroxysuccinimide (NHS). His-tagged protein was immobilized on the surface until an immobilization level of 4000 RU was obtained. Remaining active groups on the sensor surface were blocked by injection of 1 M ethanolamine for 420 seconds. The immobilization procedure was performed at 22 °C. Channel 1 was immobilized as described above without injection of His-tagged protein and used as a reference channel to correct for buffer effects, other channels were immobilized as described above. After immobilization, buffer was changed to Biacore buffer containing 1% DMSO. A pre-run was performed for a period of ≥ 30 min at a flow rate of 30 μL/min until a stable surface was obtained. The kinetic constants of the compounds were determined with single cycle kinetics with five consecutive injections with an increasing compound concentration with ranges of 3.16 – 316 nM. Experiments were performed with an association time of 100 s per concentration and a dissociation time of 1200 s, except for compounds with a long target residence time, such as irreversible inhibitors, where dissociation time was increased. To circumvent problems of mass transport limitation, a flow rate of 30 µl/min was used. A blank run with the same conditions was performed before the compound was injected. The SPR sensorgrams were analyzed with Biacore Evaluation Software by using a method of double referencing. First the reference channel was subtracted from the channel containing immobilized protein. Subsequently, the reference curve obtained with buffer injections was subtracted. The resulting curve was fitted with a 1:1 binding model. Compounds that bound according to an induced fit model were fitted with a two-state reaction model. The kinetic constants (k a , k d , K D ) of duplicates were geometrically averaged. Target residence time (τ) for the 1:1 binding model was calculated from the dissociation constant k d with the formula τ = 1/k d . Target residence for an induced fit model was calculated as described (Tummino and Copeland, 2008). Results of the SPR binding affinity (K D and Target Residence Time ( τ) of the exemplified compounds on the different kinase targets are shown in Table 1 below. K D : A means IC50 < 5 nM B means IC50 between 5 and 50 nM C means IC50 > 50 nM and < 500 nM Target Residence Time ( ^) : A means τ between 0.2 h and 1 h B means τ between 1 and 3 h C means τ > 3 h A target residence time ( τ) between 0.2 h and 1 h is considerably longer than what is generally known to be a target residence time ( τ) for common reversible kinase inhibitors. The target residence time ( τ) between 1 h and 3 h and the target residence time ( τ) longer than 3 h indicate an even further enhanced target residence of the macrocycle compounds of the invention. Table 1: Binding Kinetics (SPR measurements) Target residence times ( τ (h)) and K D for the different kinases.

Example C Biochemical kinase assay wt-BTK To determine the inhibitory activity of compounds on wt-BTK enzyme activity, the IMAP® assay (Molecular Devices) was used. Compounds were serially diluted in dimethylsulfoxide (DMSO) and subsequently in 4% DMSO in IMAP reaction buffer, which consists of 10 mM Tris-HCl, pH 157.5, 10 mM MgCl 2 , 0.01 % Tween-20, 0.1 % NaN3 and 1 mM freshly prepared dithiotreitol (DTT). Compound solution was mixed with an equal volume of full-length wt-BTK enzyme (Carna Biosciences, cat. no.08- 180) in IMAP reaction buffer. After pre-incubation of 1 hour in the dark at room temperature, fluorescein- labeled MBP-derived substrate peptide (Molecular Devices, cat. no. RP 7123) was added, followed by ATP to start the reaction. Final enzyme concentration was 1.2 nM, final substrate concentration 50 nM, and final ATP concentration was 4 μM. The reaction was allowed to proceed for 2 hours at room temperature in the dark. The reaction was stopped by quenching with IMAP progressive binding solution according to the protocol of the manufacturer (Molecular Devices). Fluorescein polarization was measured on an Envision multimode reader (Perkin Elmer, Waltham, MA, U.S.A.). IC 50 were calculated using XLfit™5 software (ID Business Solutions, Ltd., Surrey, U.K.). Compounds of examples 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 17, 31, 32, 33, 34 and 35 showed an IC 50 value < 5 nM. Example D Biochemical kinase assay BTK C481S To determine the inhibitory activity of compounds on BTK C481S enzyme activity, the IMAP® assay (Molecular Devices) was used. Compounds were serially diluted in dimethylsulfoxide (DMSO) and subsequently in 4 % DMSO in IMAP reaction buffer, which consists of 10 mM Tris-HCl, pH 157.5, 10 mM MgCl 2 , 0.01 % Tween-20, 0.1 % NaN3 and 1 mM freshly prepared dithiotreitol (DTT). Compound solution was mixed with an equal volume of full-length BTK C481S enzyme (Carna Biosciences, cat. no. 08-547) in IMAP reaction buffer. After pre-incubation of 1 hour in the dark at room temperature, fluorescein-labeled MBP-derived substrate peptide (Molecular Devices, cat. no. RP 7123) was added, followed by ATP to start the reaction. Final enzyme concentration was 1.2 nM final substrate concentration 50 nM, and final ATP concentration was 7 μM. The reaction was allowed to proceed for 2 hours at room temperature in the dark. The reaction was stopped by quenching with IMAP progressive binding solution according to the protocol of the manufacturer (Molecular Devices). Fluorescein polarization was measured on an Envision multimode reader (Perkin Elmer, Waltham, MA, U.S.A.). IC 50 were calculated using XLfit™5 software (ID Business Solutions, Ltd., Surrey, U.K.). Compounds of examples 17, 31, 32 and 34 showed an IC 50 value < 5 nM. Example E Biochemical kinase assay LCK To determine the inhibitory activity of compounds on LCK enzyme activity, the IMAP® assay (Molecular Devices) was used. Compounds were serially diluted in dimethylsulfoxide (DMSO) and subsequently in 4 % DMSO in IMAP reaction buffer, which consists of 10 mM Tris-HCl, pH 157.5, 10 mM MgCl 2 , 0.01 % Tween-20, 0.1 % NaN 3 and 1 mM freshly prepared dithiotreitol (DTT). Compound solution was mixed with an equal volume of full-length Lck enzyme (Carna Biosciences, cat. no.08- 170) in IMAP reaction buffer. After pre-incubation of 1 hour in the dark at room temperature, fluorescein- labeled MBP-derived substrate peptide (Molecular Devices, cat. no. RP 7123) was added, followed by ATP to start the reaction. Final enzyme concentration was 2.9 nM final substrate concentration 100 nM, and final ATP concentration was 4 μM. The reaction was allowed to proceed for 2 hours at room temperature in the dark. The reaction was stopped by quenching with IMAP progressive binding solution according to the protocol of the manufacturer (Molecular Devices). Fluorescein polarization was measured on an Envision multimode reader (Perkin Elmer, Waltham, MA, U.S.A.). IC 50 were calculated using XLfit™5 software (ID Business Solutions, Ltd., Surrey, U.K.). Compounds of examples 35, 61 and 62 showed an IC 50 value < 5 nM. Example F Cell proliferation assay REC-1 mantle cell lymphoma cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen (cat. no. ACC 584, DSMZ) and cultured in RPMI-1640 cell culture medium (cat. no.61870036, Life Technologies), supplemented with 18% (v/v) fetal bovine calf serum and 1% penicillin/streptavidin.1200 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for at least 5 hours at 37 °C, 95 % humidity, and 5 % CO 2 .5 µl compound solution was added to the cells and incubation was continued for 120 hours (5 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC 50 . Compounds of examples 1, 2, 3, 4, 5, 6, 7, 8, 9, 17, 31, 32, 33 and 35 showed an IC 50 value < 50 nM. Example G Cell proliferation assay CCRF-HSB-2 T acute lymphoblastic leukemia cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen (cat. no. ACC 435, DSMZ). Frozen stocks were thawed and cells were diluted in RPMI-1640 cell culture medium (cat. no.61870036, Life Technologies), supplemented with 10% (v/v) bovine calf serum and 1% penicillin/streptavidin. 3200 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for 24 hours at 37 °C, 95 % humidity, and 5 % CO 2 . 5 µl compound solution was added to the cells and incubation was continued for 72 hours (3 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill- parameter and IC 50 . Compounds of examples 15, 24, 35 and 47 showed an IC 50 value < 50 nM. Example H Cell proliferation assay CTV-1 T acute lymphoblastic leukemia cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen (cat. no. ACC 40, DSMZ). Frozen stocks were thawed and cells were diluted in RPMI-1640 cell culture medium (cat. no.61870036, Life Technologies), supplemented with 10% (v/v) bovine calf serum and 1% penicillin/streptavidin.1600 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for 24 hours at 37 °C, 95 % humidity, and 5 % CO 2 .5 µl compound solution was added to the cells and incubation was continued for 72 hours (3 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC 50 . Compounds of examples 15, 24, 35 and 47 showed an IC 50 value < 50 nM. Example I Cell proliferation assay MV4-11 acute myelogenous leukemia cells were purchased from American Type Culture Collection (cat. no. CRL-9591, ATCC). Frozen stocks were thawed and cells were diluted in IMDM cell culture medium (cat. no.31980022, Life Technologies), supplemented with 10% (v/v) bovine calf serum and 1% penicillin/streptavidin.1600 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for 24 hours at 37 °C, 95 % humidity, and 5 % CO 2 .5 µl compound solution was added to the cells and incubation was continued for 72 hours (3 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC 50 . Compounds of examples 16, 48, 56, 57, 59 and 60 showed an IC 50 value < 50 nM. Example J Cell proliferation assay MOLM-13 acute myelogenous leukemia cells were purchased from Deutsche Sammlung von Mikroorganismen und Zellkulturen (cat. no. ACC 554, DSMZ). Frozen stocks were thawed and cells were diluted in RPMI-1640 cell culture medium (cat. no.61870036, Life Technologies), supplemented with 10% (v/v) bovine calf serum and 1% penicillin/streptavidin.800 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for 24 hours at 37 °C, 95 % humidity, and 5 % CO 2 .5 µl compound solution was added to the cells and incubation was continued for 72 hours (3 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC 50 . Compounds of examples 16, 48, 56, 57, 59 and 60 showed an IC 50 value < 75 nM. Example K Cell proliferation assay TT thyroid squamous cell carcinoma cells were purchased from American Type Culture Collection (cat. no. CRL-1803, ATCC). Frozen stocks were thawed and cells were diluted in F 12 NUTRIENT MIX cell culture medium (cat. no.21127022, Life Technologies), supplemented with 10% (v/v) bovine calf serum and 1% penicillin/streptavidin.800 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for 24 hours at 37 °C, 95 % humidity, and 5 % CO 2 .5 µl compound solution was added to the cells and incubation was continued for 72 hours (3 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC 50 . Compounds of examples 16, 44, 48, 50, 52, 53, 56, 57, 59 and 60 showed an IC 50 value < 100 nM. Example L Cell proliferation assay KG-1 acute myeloid leukemia cells were purchased from American Type Culture Collection (cat. no. CCL-246, ATCC). Frozen stocks were thawed and cells were diluted in IMDM cell culture medium (cat. no. 31980022, Life Technologies), supplemented with 20% (v/v) fetal calf serum and 1% penicillin/streptavidin.3200 cells per well (in 45 µl) were seeded in a white 384-well culture plate (cat. no.781080, Greiner Bio-One) and allowed to rest for 24 hours at 37 °C, 95 % humidity, and 5 % CO 2 . 5 µl compound solution was added to the cells and incubation was continued for 72 hours (3 days), followed by addition of 24 µl ATPlite 1Step™ (PerkinElmer, Groningen, The Netherlands) solution was added to each well. Luminescence was recorded on an Envision multimode reader. The cell signal at the start of incubation was recorded separately in order to distinguish between cell population growth and cell death. In addition, maximum growth was determined by incubation of a duplicate without compound in the presence of 0.3 % DMSO. Percentage growth was used as the main y-axis signal. IC 50s were fitted by non-linear regression using IDBS XLfit™5 using a 4-parameter logistic curve, yielding a maximum signal, minimum signal, hill-parameter and IC 50 . Compounds of examples 44, 50, 52, 53, 54, 55, 59 and 60 showed an IC 50 value < 300 nM. Example M Biochemical kinase assay RET The kinase activity of RET was assayed at Reaction Biology Corporation. The substrate in the RET reaction, TRK-C derived peptide (Genscript, Cat.# U4552FL280_1), was prepared in fresh reaction buffer (20 mM HEPES (pH 7.5), 10 mM MgCl 2 , 1 mM EGTA, 0.01% Brij35, 0.02 mg/mL BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT, 1% DMSO). RET (Invitrogen, Cat.# PV3819) was delivered into the substrate solution and mixed gently. The final concentrations of RET and the substrate in the reaction were 2 nM and 20 µM respectively. Compounds were tested in 10-point concentration/response mode with 3-fold serial dilution steps starting at 10 µM. Compounds in 100% DMSO were delivered into the kinase reaction mixture by acoustic liquid delivery technology (ECHO550; nanoliter range) and incubated for 20 min at room temperature.10 µM [ 33 P]-ATP (ATP: Sigma, Cat#: A7699; [ 33 P]-ATP: Hartmann Analytic, Cat#: SCF-301-12) was delivered into the reaction mixture to initiate the reaction. The mixture was incubated for 120 min at room temperature. Radioactivity was detected utilizing a proprietary filter-binding method. Kinase activity data were expressed as the percent remaining activity in test samples compared to vehicle (DMSO) reactions. IC 50 values and curve fits were obtained using Prism4 Software (GraphPad). Compounds of examples 44, 50, 52, 53 and 59 showed an IC 50 value < 5 nM.

REFERENCES • Attwood et al. (2021) “Trends in kinase drug discovery targets, indications and inhibitor design.” Nat Rev Drug Discov 20, pages 839–861 (2021). • Xianhui Wang et al. (2021) Bruton’s Tyrosine Kinase and Its Isoforms in Cancer. Front. Cell Dev. Biol.9:668996 • Kokabee L. et al., Bruton's Tyrosine kinase is a potential therapeutic target in prostate cancer cells (2015) Cancer Biology & Therapy 16;11, 1604-1615 • Wang et al, Bruton's Tyrosine Kinase inhibitors prevent therapeutic escape in breast cancer cells; Mol Cancer Ther. (2016) 15(9) 2198–2208 • Wei et al. (2016) Oncotarget 7(43), 69961-69975 • Wang et al. (2017) J Exp Clin Cancer Res, 36, pp96 • Stefan F.H. Neys et al., Targeting Bruton's Tyrosine Kinase in inflammatory and autoimmune pathalogies;(2021) Front Cell Dev Biol, 9:668131 • Rula Zain et al., Structure-Function relationships of covalent and non-covalent BTK inhibitors (2021) Front Immunol 12:694853 • Wang et al. (2019) Noncovalent inhibitors reveal BTK gatekeeper and auto-inhibitory residues that control its transforming activity : JCI Insight.2019;4(12):e127566. • Cohen et al.2021, Kinase drug discovery 20 years after imatinib: progress and future directions; Nature Reviews Drug Discovery volume 20, pages 551–569 (2021)) • Willemsen-Seegers N. et al (2016) J Mol Biol (2017) 429, 574–586 • Copeland R.A. et al (2006) Nat Rev Drug Discov, 5, 730 • Barf T. and Kaptein A. (2012) J Med Chem, 55(14):6243-62 • Willemsen-Seegers N. et al (2016) J Mol Biol (2017) 429, 574–586 • Driggers, EM et al (2008) Nature Reviews Drug Discovery, 7(7), 608–624 • Mallinson J et al (2012) Future Med. Chem.4(11), 1409–1438; Macrocycles in new drug discovery • P.J. Tummino, R.A. Copeland, Residence time of receptor–ligand complexes and its effect on biological function, Biochemistry 47 (2008) 5481–5492.