Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HETEROCYCLIC DERIVATIVES AS JANUS KINASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2024/052512
Kind Code:
A1
Abstract:
The present invention relates to a compounds of general formula (I) inhibiting the JAK family of non-receptor tyrosine protein kinases (JAK1, JAK2, JAK3, and TYK2); methods of preparing such compounds, pharmaceutical compositions containing them and therapeutic use thereof. 5 The compounds of the invention may be useful in the treatment of diseases or conditions associated with a dysregulation of the JAK family non-receptor kinases; in particular for the treatment of various inflammatory disease including asthma, COPD and other respiratory diseases.

Inventors:
ACCETTA ALESSANDRO (IT)
CUZZOLIN ALBERTO (IT)
RANCATI FABIO (IT)
RIZZI ANDREA (IT)
ELENKOV IVAYLO JIVKOV (IT)
MESIĆ MILAN (IT)
RADOŠEVIĆ ELA (IT)
Application Number:
PCT/EP2023/074691
Publication Date:
March 14, 2024
Filing Date:
September 08, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CHIESI FARM SPA (IT)
International Classes:
A61P35/00; A61K31/4353; A61P37/06; C07D471/04; C07D519/00
Domestic Patent References:
WO2012069202A12012-05-31
WO2008052734A12008-05-08
WO2018122212A12018-07-05
WO2022194779A12022-09-22
WO2022194781A12022-09-22
Other References:
HOWELL MICHAEL D. ET AL: "Targeting the Janus Kinase Family in Autoimmune Skin Diseases", vol. 10, 1 January 2019 (2019-01-01), pages 2342, XP055863286, Retrieved from the Internet DOI: 10.3389/fimmu.2019.02342
HU X ET AL., SIGNAL TRANSDUCT TARGET THER., vol. 26;6, no. 1, 2021, pages 402
HOWELL MD ET AL., FRONT. IMMUNOL., vol. 10, 2019, pages 2342
HODGE ET AL., CLIN EXP RHEUMATOL, vol. 34, no. 2, 2016, pages 318 - 28
HU X. ET AL., SIG TRANSDUCT TARGET THER, vol. 6, 2021, pages 402
ZHANG, Y ET AL., ANN. RHEUM. DIS., vol. 76, 2017, pages 1467 - 1475
WANG, W ET AL., SCLERODERMA RELAT. DISORD., vol. 5, 2020, pages 40 - 50
CONCA, W ET AL., FRONT. PHARMACOL., vol. 11, 2020, pages 5857619
D'ALESSANDRO M ET AL., INT. IMMUNOPHARMACOL., vol. 86, 2020, pages 106748
WIJAYA, I. ET AL., CLIN. EPIDEMIOL. GLOB. HEALTH, vol. 11, 2021, pages 100755
CAO Y. ET AL., J. ALLERGY CLIN. IMMUNOL., vol. 146, 2020, pages 137 - 146
STEVE NG ET AL., J ALLERGY CLIN IMMUNOL, vol. 148, 2021, pages 953 - 63
ISRAEL EREDDEL HK., N ENGL J MED, vol. 377, 2017, pages 965 - 76
WENZEL SE, AM J RESPIR CRIT CARE MED, vol. 203, 2021, pages 809 - 21
HINKS TSC ET AL., ERJ, vol. 57, no. 1, 2021, pages 2000528
ASHINO S ET AL., J ALLERGY CLIN IMMUNOL, vol. 133, 2014, pages 1162 - 74
DENGLER HS ET AL., SCI TRANSL MED, vol. 10, 2018, pages eaao2151
YOUNIS US ET AL., AAPS PHARMSCI-TECH, vol. 20, 2019, pages 167
DI STEFANO ET AL., EUR RESPIR J., vol. 24, no. 1, July 2004 (2004-07-01), pages 78 - 85
YEW-BOOTH L ET AL., EUR RESPIR J, vol. 46, no. 3, 2015, pages 843 - 5
BRINGMANN G ET AL., ANGEW. CHEMIE INT. ED., vol. 44, no. 34, 2005, pages 5384 - 5427
OKI M, TOPICS IN STEREOCHEMISTRY, vol. 14, 1983, pages 1 - 82
T. W. GREENEP. G. M. WUTS: "Strategic application of named reactions in organic synthesis", 2005
WACHTEL, H.KATTENBECK, S.DUNNE, S. ET AL., PULM THER, vol. 3, 2017, pages 19
Attorney, Agent or Firm:
CHIESI FARMACEUTICI S.P.A. (IT)
Download PDF:
Claims:
169 CLAIMS 1. A compound of formula I 5 R1 is a heteroaryl group selected from the group consisting of imidazo[1,2-b]pyridazin-3-yl, pyrazolo[1,5-a]pyrimidin-3-yl and 3-oxo- (3,4- dihydropyrazin-2-yl)amino; R2 is a group of formula 10 that is a substituent group link wherein V is absent (meaning it is a bond) or is a divalent group selected from O, S, N(R6), C(O)N(R6), N(R6)C(O); N(R6)C(O)O; N(R6)S(O)2; N(R6)C(O)N(R6); Q is selected from the group consisting of H, (C1-C6)alkyl, (C1-C6)hydroxyalkyl, (C1-15 C6)alkoxy, -(CH2)mNR4R5, (C3-C8)cycloalkyl, (C3-C10)heterocycloalkyl; -S-(C3- C6)heterocycloalkyl, and -N(R6)-(C3-C6)heterocycloalkyl; wherein said (C3-C8)cycloalkyl and (C3-C10)heterocycloalkyl are further optionally substituted by one or more substituent selected from the group consisting of -OH, oxo (i.e. (=O)), (C1-C10)alkyl, (C1-C6)alkoxy; halogen, (C1-C6)haloalkyl, alkanoyl, (C1-C6)hydroxyalkyl, (C1-C6)alkoxy(C1-C6)alkyl, - 20 N(R6)(CH2)mC(O)NR4R5, -(CO)NR4R5, -(CH2)mNR4R5, (C3-C8)cycloalkyl(C1-C6)alkyl; (C3-C6)heterocycloalkyl(C1-C6)alkyl, (C3-C6)heterocycloalkyl and hydroxyl-(C3- C6)heterocycloalkyl; R3 is a bicyclic moiety which is a substituent group linked to the scaffold graphically represented by 170 wherein the dashed line --- ind r a double bond X is selected from N, S,C; Y is selected from C, N; 5 Z is selected from C, N, O; K is absent (meaning a bond) or selected from O, C, S; G is absent (meaning a bond) or selected from C, O; is the point of attachment of a substituent group to the rest of the molecule d m are in each occurrence independently 0 or an integer selected from 1, 2, 3 10 and 4; R4 and R5, the same or different, are selected from the group consisting of -H, (C1-C6)alkyl, (C1-C6)haloalkyl, and 15 (C3-C6)heterocycloalkyl; R6 is in each occurrence independently selected from the group consisting of H, (C1- C6)alkyl and (C1-C6)hydroxyalkyl; R7 is one or more group (preferably 1 or 2) in each occurrence independently selected from the group consisting of -OH, oxo (that is =O), (C1-C6)alkyl, halogen, (C1-C6)haloalkyl, 20 (C1-C6)hydroxyalkyl, -(CH2)mNR4R5, (C1-C6)alkyl-S(O)2- and (C1-C6)alkyl-S(O)2N(R6)-; R8 is selected in the group consisting of (C1-C6)alkoxy, (C1-C6)haloalkoxy; single enantiomers, diastereoisomers and mixtures thereof in any proportion 25 or a pharmaceutically acceptable salt or solvate thereof. 2. A compound of formula I according to claim 1 wherein the bicyclic moiety R3 171 3 is selected from 3. A compound of for 5 wherein the bicyclic moiety R3 is selected in the group consisting of J1-J13 4. wherein R3 is J1 and R8 is methoxy; 10 the compound being thus represented by the formula Ia 172 wherein R1 is pyrazolo[1,5-a]pyrimidin-3-yl or (3-oxo-3,4-dihydropyrazin-2-yl)amino 5 V is a divalent group selected from C(O)N(R6), N(R6)C(O)O; Q is selected from the group consisting of (C1-C6)alkyl, (C1-C6)alkoxy, - (CH2)mNR4R5, (C3-C8)cycloalkyl and (C3-C12)heterocycloalkyl; wherein said (C3- C8)cycloalkyl and (C3-C12)heterocycloalkyl are further optionally substituted by one or more substituent selected from the group consisting of oxo (i.e. (=O)), (C1-C10)alkyl, Halogen,10 (C1-C6)hydroxyalkyl, -(CO)NR4R5; (C3-C8)cycloalkyl(C1-C6)alkyl; (C3- C6)heterocycloalkyl; n and m are in each occurrence independently 0 or an integer selected from 1, 2, 3 and 4; R4 and R5, the same or different, are in each occurrence independently selected from 15 the group consisting of -H, (C1-C6)alkyl, (C1-C6)haloalkyl; or a pharmaceutically acceptable salt or solvate thereof. 20 5. A compound of formula Ia according to claim 4 wherein V is N(R6)C(O)O; Q is selected from the group consisting of (C1-C6)alkyl, (C1-C6)alkoxy, - (CH2)mNR4R5, and (C3-C6)heterocycloalkyl being a grou wherein X1 is selected from CH , O, S, NH, NC 2 H3, (C=O) and S(=O)2; 25 single enantiomers, diastereoisomers and mixtures thereof in any proportion or a pharmaceutically acceptable salt or solvate thereof.

173 6. A compound of formula Ia according to claim 4 wherein V is C(O)N(R6), Q is selected from the group consisting of -(CH2)mNR4R5, and (C3-C6)heterocycloalkyl being a group wherein X1 is selected from the group consisting of CHR9, O, S, NH, 5 NCH3, CF2, (C ) alkoxy or (C1-C6)alkoxy(C1-C6)alkyl, wherein R9 is H or -(CO)NR4R5 single enantiomers, diastereoisomers and mixtures thereof in any proportion or a pharmaceutically acceptable salt or solvate thereof. 7. A compound of formula Ia according to claim 4 wherein Q is (C3-C6)heterocycloalkyl, -S-(C3-C6)heterocycloalkyl or -N(R6)-(C3- 10 C6)heterocycloalkyl selected from the group consisting of: 8. compoun accor ng to c a m se ecte rom: 6-(difluoromethoxy)-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]oxazine, 15 1-(6-methoxy-2-(methylsulfonyl)isoindolin-5-yl)-3-methyl-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine, 8-methoxy-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-1,3,4,5-tetrahydro-2H-benzo[b]azepin-2-one, 174 7-methoxy-6-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-3,4-dihydroquinolin-2(1H)-one, 7-methoxy-6-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-2,3-dihydroquinolin-4(1H)-one, 5 6-methoxy-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]thiazine, 6-methoxy-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-2H-benzo[b][1,4]thiazin-3(4H)-one, 6-methoxy-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- 10 c]pyridin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]thiazine 1,1-dioxide, 7-methoxy-8-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepine, 6-methoxy-5-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)indolin-2-one, 15 2,2-difluoro-6-methoxy-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)-2H-benzo[b][1,4]oxazin-3(4H)-one, 7-methoxy-3-methyl-6-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)-3,4-dihydroquinazolin-2(1H)-one, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-20 a]pyrimidin-3-yl)-N-(1-((tetrahydro-2H-pyran-4-yl)methyl)azetidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, N-(3-(4,4-difluoropiperidin-1-yl)propyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 3-carboxamide, 25 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(2-oxo-1-oxa-8- azaspiro[4.5]decan-8-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, N-(3-(dimethylamino)propyl)-1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7- yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide,30 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7-yl)-N-(1-methylazetidin-3-yl)- 6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide, (R)-1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7-yl)-N-((4- methylmorpholin-2-yl)methyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, 175 3-(2-oxomorpholino)propyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7- yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 2-(methyl(2-oxotetrahydrofuran-3-yl)amino)ethyl (1-(6-methoxy-3,4-dihydro-2H- 5 10 15 20 25 2-morpholinoethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 3-(4-methylpiperazin-1-yl)propyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]thiazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- yl)carbamate, 30 2-(piperidin-1-yl)ethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, (R)-1-methylpyrrolidin-3-yl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7- yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, (S)-1-methylpyrrolidin-3-yl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7- 35 yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 176 (4-(dimethylamino)tetrahydro-2H-pyran-4-yl)methyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]thiazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- yl)carbamate, (R)-2-(3-methoxypyrrolidin-1-yl)ethyl (1-(6-methoxy-3,4-dihydro-2H- 5 benzo[b][1,4]thiazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- yl)carbamate, 3-(1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7-yl)-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)-1,1-dimethylurea, 6-methoxy-7-(3-((2-(4-methylpiperazin-1-yl)ethyl)amino)-6-(pyrazolo[1,5- 10 a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-1-yl)-2H-benzo[b][1,4]thiazin-3(4H)-one, 7-(3-((2-(dimethylamino)ethyl)amino)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2H-benzo[b][1,4]thiazin-3(4H)-one, 1-(6-methoxybenzo[d]isothiazol-5-yl)-N-(2-morpholinoethyl)-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-amine, 15 6-methoxy-7-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]oxazine, 1-(6-methoxyindolin-5-yl)-3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridine, 7-methoxy-6-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- 20 c]pyridin-1-yl)-1,2,3,4-tetrahydroquinoline, N-(3-(dimethylamino)propyl)-1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7- yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(1-methylazetidin-3-yl)- 6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide,25 (R)-1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-((4- methylmorpholin-2-yl)methyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(4-methoxypiperidin- 1-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide,30 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(4- (methoxymethyl)piperidin-1-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, N-(2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 35 3-carboxamide, 177 N-(2-(2-oxa-6-azaspiro[3.5]nonan-6-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 3-carboxamide, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-((3aS,6aR)-1- 5 oxotetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridine-3-carboxamide, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(3-oxo-2-oxa-8- azaspiro[4.5]decan-8-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, 10 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(1-oxo-2-oxa-8- azaspiro[4.5]decan-8-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, N-(2-(4-(dimethylcarbamoyl)piperidin-1-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 15 3-carboxamide, N1-(1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)-N2,N2-dimethylethane-1,2-diamine, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-methyl-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-amine, 20 methyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 2-(dimethylamino)ethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 2-morpholinoethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6- 25 (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 3-(4-methylpiperazin-1-yl)propyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- yl)carbamate, 3-(dimethylamino)propyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)- 30 6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 3-morpholinopropyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 2-(1,1-dioxidothiomorpholino)ethyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- 35 yl)carbamate, 178 2-(4-methylpiperazin-1-yl)ethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin- 7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 2-thiomorpholinoethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 5 (S)-(4-methylmorpholin-2-yl)methyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- yl)carbamate, 2-(piperidin-1-yl)ethyl (1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 10 2-((2-oxotetrahydrofuran-3-yl)thio)ethyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- yl)carbamate, 2-((2-oxotetrahydrofuran-3-yl)amino)ethyl (1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3- 15 yl)carbamate, 2-(dimethylamino)ethyl (1-(7-methoxy-1,2,3,4-tetrahydroquinolin-6-yl)-6- (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate, 1-(1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)-3-(3-morpholinopropyl)urea,20 6'-methoxy-5'-(3-methyl-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)spiro[cyclopropane-1,3'-indolin]-2'-one, 5-methoxy-6-(3-((2-morpholinoethyl)amino)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)benzo[d]oxazol-2(3H)-one, N-(3-(dimethylamino)propyl)-6-(imidazo[1,2-b]pyridazin-3-yl)-1-(6-methoxy-3,4- 25 dihydro-2H-benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide, 3-((3-((2-(dimethylamino)ethyl)amino)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)pyrazin-2(1H)-one, N-(3-(dimethylamino)propyl)-1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7- yl)-6-((3-oxo-3,4-dihydropyrazin-2-yl)amino)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide,30 N-(3-(4,4-difluoropiperidin-1-yl)propyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-((3-oxo-3,4-dihydropyrazin-2-yl)amino)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, N-(2-(2,2-dimethyl-6-oxomorpholino)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 35 3-carboxamide, 179 1-(6-(difluoromethoxy)-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(3- (dimethylamino)propyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3- carboxamide, N-(2-(4-carbamoylpiperidin-1-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- 5 benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 3-carboxamide, (R)-N-(2-(3-carbamoylpyrrolidin-1-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 3-carboxamide, 10 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(7-oxo-6-oxa-2- azaspiro[3.4]octan-2-yl)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxamide, N-(2-(2-oxa-6-azaspiro[3.4]octan-6-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 15 3-carboxamide, 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-(2-methyl-6- oxomorpholino)ethyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-3- carboxamide, N-(1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5- 20 a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)-1-methylpiperidine-4-carboxamide, N-(2-(4-(3-hydroxyoxetan-3-yl)piperidin-1-yl)ethyl)-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine- 3-carboxamide, 2-((2-oxotetrahydrofuran-3-yl)thio)ethyl (1-(7-methoxy-1,2,3,4-tetrahydroquinolin-6- 25 yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate single enantiomers, diastereoisomers and mixtures thereof in any proportion or a pharmaceutically acceptable salt or solvate thereof. 9. A pharmaceutical composition comprising a compound as defined in any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, in admixture with one or more 30 pharmaceutically acceptable carrier or excipient. 10. A pharmaceutical composition according to claim 9 suitable to be administered by inhalation, selected from inhalable powders, propellant-containing metering aerosols or propellant-free inhalable formulations.

180 11. A device comprising the pharmaceutical composition according to claim 10, which may be a single- or multi-dose dry powder inhaler, a metered dose inhaler or a soft mist nebulizer. 12. A compound or a pharmaceutical composition according to any one of claims 1 to 10 for use as a medicament. 5 13. A compound or a pharmaceutical composition for use according to claim 12 in the prevention and /or treatment of a pulmonary disease selected from the group consisting of asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), acute lung injury and acute respiratory distress syndrome (ARDS). 14. A combination of a compound as defined in any one of the claims 1 to 8 with one or more 10 active ingredients selected from the classes currently used in the treatment of respiratory disorders, and known to the skilled person, such as beta2-agonists, antimuscarinic agents, corticosteroids, mitogen-activated kinases (P38 MAP kinases) inhibitors, PI3K inhibitors (phosphoinositide 3-kinases), nuclear factor kappa-B kinase subunit beta inhibitors (IKK2), Rho kinase inhibitors (ROCKi), human neutrophil elastase (HNE) inhibitors,15 phosphodiesterase 4 (PDE4) inhibitors, leukotriene modulators, non-steroidal anti- inflammatory agents (NSAIDs) and mucus regulators.

Description:
HETEROCYCLIC DERIVATIVES AS JANUS KINASE INHIBITORS

FIELD OF THE INVENTION

The present invention relates to chemical compounds that are derivatives useful as JAK inhibitors, such as JAK 1, useful for the treatment of various inflammatory disease including asthma, COPD and other respiratory diseases.

BACKGROUND OF THE INVENTION

The JAK family consists of non-receptor tyrosine protein kinases and has four main members, JAK1, JAK2, JAK3, and TYK2. More than 50 cytokines and growth factors bind to type I and II receptors noncovalently associated with different combinations of JAK kinases. The signalling triggered by the ligands consists in tyrosine phosphorylation of receptors by JAK and recruitment of one or more STATs proteins. Tyrosine-phosphorylated STATs dimerize and are then transported into the nucleus through the nuclear membrane to regulate specific genes. JAKs have seven homology domains (the JAK homology domain, JH). Starting from the carboxyl terminus, JH1 is the first JH, known as the kinase domain, and is composed of approximately 250 amino acid residues. JH1 encodes a kinase protein that constitutes the kinase structure domain that phosphorylates a substrate; JH2 is a pseudokinase domain which regulates the activity of the kinase domain. JAK3 is expressed in the bone marrow and lymphatic system, as well as endothelial cells and vascular smooth muscle cells; other members are expressed in almost all tissues (Hu X et al., Signal Transduct Target Ther. 2021, 26;6(l):402). Many cellular processes are downstream JAK/STAT signalling: hematopoiesis, immune balance, tissue repair, inflammation, apoptosis, and adipogenesis. Different biological responses are regulated by specific pairing of JAK isoforms. JAK1/JAK3 combination mediates IL-2, -4, -7, -9, -15, and -21 signalling that is relevant for growth/maturation of lymphoid cells, differentiation/homeostasis of T-cells/NK cells, B-cell class switching and other inflammatory processes. Combinations of JAK1/TYK2-JAK1/JAK2, regulate the signal associated with the innate immune response, such as IL-6 and the type I interferons, involved into naive T cell differentiation, T cell homeostasis, granulopoiesis and other inflammatory processes. (Howell MD et al., Front. Immunol. 2019, 10, 2342). JAK2 frequently associates with itself (JAK2/ JAK2) controlling the signalling of various cytokines and growth factors, such as IL-3, IL-5, granulocyte macrophage colony-stimulating factor (GM-CSF), erythropoietin (EPO), and thrombopoietin (TPO) (Hodge et al., Clin Exp Rheumatol 2016; 34(2):318-28).

Genetically modified mouse models and human diseases prove the importance of JAK/STAT pathways in immune fitness. In particular, overexpression or mutations involving some JAK isoforms as well as aberrant JAK/STAT signalling drive malignancies of hematopoietic and lymphoid tissues as well as inflammatory disorders. Currently, several Food and Drug Administration (FDA)- and/or EU- approved JAK inhibitors are in clinical use. Two (ruxolitinib and fedratinib) small molecules are in use for hematologic disorders as myelofibrosis and polycythemia vera; six JAK inhibitors (tofacitinib, baricitinib, ruxololitinib, filgotinib, upadicitinib and delgocitinib in Japan) result in use for immune-mediated disorders as rheumatoid arthritis, polyarticular juvenile idiopathic arthritis, atopic dermatitis, ulcerative colitis and acute graft-versus-host disease. Moreover, some of these drugs as well as others are currently under phase II and III of clinical trials for indications that span from autoimmune diseases (lupus, vitiligo, etc.), inflammatory bowel disease to Non-Hodgkin lymphoma and COVID-19 (Hu X. et al., Sig Transduct Target Ther 2021, 6: 402).

The small molecules targeting JAK/STAT represent an attractive option also for the therapy of fibrotic disorders. In fact, inflammatory cytokines (IL-4, IL-3, IL-6, IL-11, IL-31, etc) and growth factors (FGF, VEGF, etc.) involved in the fibrotic processes activate JAK/STAT pathway. Ruxolitinib tested in a bleomycin-induced fibrosis mouse model ameliorated the fibrotic lesions in lung, and reduced levels of fibrotic molecular markers (Zhang, Y et al., Ann. Rheum. Dis. 2017, 76, 1467-1475) while tofacitinib acted as a preventive agent in experimental dermal and pulmonary fibrosis (Wang, W et al., Scleroderma Relat. Disord. 2020, 5, 40-50). In patients, some case reports were studied. A single-case report corroborated the efficacy and safety of tofacitinib in combination with nintedanib in the management of an aggressive interstitial lung disease with poor prognosis (Conca, W et al., Front. Pharmacol. 2020, 11, 5857619). Baricitinib was demonstrated to be a safe immune modulator that reduces the biomarkers’ levels of lung fibrosis and inflammation in RA patients, including a subgroup with interstitial lung disease (D’ Alessandro M et al., Int. Immunopharmacol. 2020, 86, 106748).

In COVID- 19, there are some JAK inhibitors undergoing clinical trials, and they are tofacitinib, baricitinib, and ruxolitinib. Baricitinib and ruxolitinib were associated with a reduced risk of mortality. They reduced the use of invasive mechanical ventilation and had a borderline impact on the admission rate of the intensive care unit and the incidence of acute respiratory distress syndrome (ARDS). (Wijaya, I. et al. Clin. Epidemiol. Glob. Health 2021, 11, 100755). Ruxolitinib also was tested in COVID- 19 patients, and improved the clinical symptoms and chest computed tomography images (Cao Y. et al., J. Allergy Clin. Immunol. 2020 146, 137-146).

Asthma can be included in the plethora of immune-mediated diseases for which pathogenesis is characterized by an essential role of JAK/STAT signalling. Asthma is a chronic inflammatory disease of the airways due to a complex interplay between immune response, genetic susceptibility and nonspecific external stimuli like cold, allergens and exercise leading to hyperresponsiveness, remodelling of the airways, ultimately contributing to airflow limitation. Severe asthma affects 5% to 15% of the population with adult asthma (which is 300 million people worldwide) and represents a public health issue associated with increased mortality, increased hospitalizations, significant burden of symptoms, health care costs, and missed work and school (Steve NG et al., J Allergy Clin Immunol 2021;148:953-63). Severe asthma represents a subset of difficult-to-treat asthma and occurs in patients whose disease remains uncontrolled despite the use of high doses of inhaled corticosteroids (ICSs) combined with long-acting P-agonists or other controllers. To date, four types of biologies are licensed for severe asthma, i.e. omalizumab (anti-immunoglobulin E) antibody, mepolizumab and reslizumab (anti-interleukin [IL]-5antibody), benralizumab (anti-IL- 5 receptor a antibody) and dupilumab (anti-IL-4 receptor alpha antibody). Despite their efficacy, many patients continue to experience exacerbations or uncontrolled disease, indicating a need for more novel therapies (Israel E, Reddel HK. N Engl J Med 2017; 377:965-76).

Recently, the better understanding of asthma pathobiology brought to a shift from a phenotypic classification system to the introduction of the “endotype” concept. According to the latter, classification is performed on the basis of pathophysiologic mechanisms and clinical biomarkers associated with a given patient (Wenzel SE et a., Am J Respir Crit Care Med 2021;203:809-21). There are two major endotypes in asthma: type 2 and non-type 2. The type 2 pathway is defined by activation of cytokines derived from TH2 cells and group 2 innate lymphoid cells (ILC2s); these include IL-4, IL-5, and IL-13 that cause airway inflammation by activating eosinophils, B cells, airway epithelial cells, and other cell types. Biomarkers of type 2 asthma include blood/ sputum eosinophilia and elevated levels of fractional exhaled nitric oxide (FENO) and IgE. The type 2-low pathway is characterized by absence of type 2-high cytokines and biomarkers, and it manifests either increased levels of neutrophils in the airways or a paucigranulocytic profile, with normal levels of airway neutrophils and eosinophils. Type 2-low asthma is currently not well understood, and it likely encompasses multiple distinct endotypes. Potential mediators and/or biomarkers of T2 low endotypes under investigation include IL-6, IL- 17A/F, IL-23, Type I interferons, CXCL10, TNF, alarmins (TSLP, IL-25, IL-33), IL-ip, IL-8, IFN-y (Hinks TSC et al., ERJ 2021, 57 (1) 2000528).

Almost all the mediators mentioned above both for T2 and T2-low endotypes activate JAK/STAT pathway, here the rationale for the potential use of JAK inhibitors in both endotypes of severe asthma. Targeting simultaneously several cytokines by JAK inhibitors may offer advantage over the biologies (for no-responder patients) and standard therapies (for patients who remain uncontrolled) considering their administration on top of ICS.

Despite strong rationale of JAK inhibitors in asthma, safety concerns may arise by administration of systemic inhibitors or may limits administration into particular asthma subjects such as children. Considering that Asthma is a lung restricted disease, inhalatory route of administration for a JAK inhibitor may offers the advantage of therapeutic efficacy while limiting systemic exposure and correlated side effects. To date, some companies are developing inhaled JAK inhibitor for asthma treatment. Astrazeneca pipeline include AZD-0449 (completed Phase I clinical trial) and AZD-4604 (ongoing Phase I clinical trial); Theravance Biopharma is starting a new preclinical program on TD-8236 inhaled JAK inhibitor and Kinaset/Vectura is developing VR588 (ongoing Phase I clinical trial) as inhalatory compound. Many preclinical studies sponsored by the companies mentioned above demonstrated the efficacy of JAK inhibitors in the modulation of asthma. In the preclinical phase of drug development, JAK1/3 inhibitor R256 (now referred as AZD0449) orally given showed be effective in decreasing airway resistance, BAL eosinophilia, mucus production and if administered during sensitization, also TH2 cytokine responses (Ashino S et al., J Allergy Clin Immunol 2014;133:1162-74). iJak-381 from Genentech given as dry powder reduced BAL eosinophilia, CCL11, airway resistance, and Muc5AC in OVA- challenged mice. Moreover, it reduced BAL eosinophilia, neutrophilia, CCL11, and CXCL1 in a in mouse model of chronic exposure to AAH allergens (Dengler HS et al., Sci Transl Med 2018;10:eaao2151). Moreover, an oral JAK inhibitor as Tofacitinib, formulated for being administered as aerosol, reduced eosinophils count in a house dust mite mouse model of asthma (Younis US et al., AAPS PharmSci-Tech 2019;20:167).

Another respiratory disease that could benefit from lung restricted JAK inhibition is Chronic obstructive pulmonary disease (COPD), an inflammatory disease of the lung, most commonly resulting from cigarette smoke exposure, characterised by a largely irreversible and progressive airflow limitation. Despite inflammatory cytokines are drivers of chronic airway inflammation and some of them trigger JAK/STAT activation (IL-6, IFN-y, IL-2, etc.), the role of this pathway in COPD pathogenesis is poorly characterized. Phosphorylated-STAT4+ cells (Di Stefano A et al., Eur Respir J. 2004 Jul; 24(l):78-85) were found to be increased in COPD compared to non- smokers healthy controls. In another study, phosphorylated-STAT3+ and phosphorylated- STAT1+ cells counts were higher in lung biopsies of COPD patients than non-smokers controls while it was not possible to reproduce previous data on phosphorylated-STAT4 molecule (Yew- Booth L et al., Eur Respir J 2015; 46(3): 843-5). These data might also suggest a therapeutic use of JAK inhibitors also in COPD disease.

In view of the number of pathological responses which are mediated by JAK enzymes, there is a continuing need for inhibitors of JAK enzymes which can be useful in the treatment of many disorders and particularly respiratory diseases.

Thus, the finding of novel and potent JAK inhibitor suitable for local administration to the lungs for treatment of asthma and respiratory disease still remains an important need. Strong need remains for JAK inhibitors, and particularly inhaled JAK inhibitors that have potential for giving compounds with an improved safety. Despite inhalatory administration, safety concerns may still arise by drug levels reaching systemic circulation following inhalation of a JAKi. In addition to a well suited profile for inhalation, a JAKi should preferably possess additional properties that may further limit the systemic exposure after inhalation.

SUMMARY OF THE INVENTION

Accordingly, it is one object of the present invention to provide compounds of formula (I)

Wherein Ri, R7, Rs, X, Y, Z, K, G, n, V and Q are as defined in the detailed description of the invention; or a pharmaceutically-acceptable salt thereof, that are useful as JAK kinase inhibitors.

It is another object of the present invention to provide pharmaceutical compositions comprising such compounds, methods of using such compounds to treat respiratory diseases, and processes and intermediates useful for preparing such compounds.

In one aspect, the present invention provides a compound of formula (I) for use as a medicament. In one aspect the present invention provides the use of a compound of the invention for the manufacture of a medicament.

In a further aspect, the present invention provides the use of a compound of the invention for the preparation of a medicament for the treatment of any disease associated with JAK enzyme mechanisms.

In another aspect, the present invention provides a method for prevention and/or treatment of any disease associated with JAK enzyme mechanisms as above defined, said method comprises administering to a patient in need of such treatment a therapeutically effective amount of a compound of the invention.

In a particular aspect the compounds of the invention are used alone or combined with other active ingredients and may be administered for the prevention and/or treatment of a pulmonary disease including asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), interstitial lung diseases and idiopathic pulmonary fibrosis (IPF), acute lung injury and acute respiratory distress syndrome (ARDS).

DETAILED DESCRIPTION OF THE INVENTION

Definitions

The term “Pharmaceutically acceptable salts” refers to derivatives of compounds of formula (I) wherein the parent compound is suitably modified by converting any of the free acid or basic group, if present, into the corresponding addition salt with any base or acid conventionally intended as being pharmaceutically acceptable.

Suitable examples of said salts may thus include mineral or organic acid addition salts of basic residues such as amino groups, as well as mineral or organic basic addition salts of acid residues such as carboxylic groups.

Cations of inorganic bases which can be suitably used to prepare salts of the invention comprise ions of alkali or alkaline earth metals such as potassium, sodium, calcium or magnesium. Those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt comprise, for example, salts of hydrochloric, hydrobromic, sulfuric, phosphoric, methane sulfonic, camphor sulfonic, acetic, oxalic, maleic, fumaric, succinic and citric acids.

Many organic compounds can form complexes with solvents in which they are reacted or from which they are precipitated or crystallized. These complexes are known as “solvates” which are a further object of the invention. Polymorphs and crystalline forms of compounds of formula (I), or of pharmaceutically acceptable salts, or solvates thereof are a further object of the invention.

The term “Halogen” or “halogen atoms” includes fluorine, chlorine, bromine, and iodine atom ; meaning Fluoro, Chloro, Bromo, Iodo as substituent.

The term “(Ci-CejAlkyl” refers to straight-chained or branched alkyl groups wherein the number of carbon atoms is in the range 1 to 6. Particular alkyl groups are for example methyl, ethyl, n-propyl, isopropyl, t-butyl, 3-methylbutyl and the like.

The expressions “(Ci-CejHaloalkyl” refer to the above defined “(Ci-Cejalkyl” groups wherein one or more hydrogen atoms are replaced by one or more halogen atoms, which can be the same or different from each other. Examples include halogenated, poly-halogenated and fully halogenated alkyl groups wherein all of the hydrogen atoms are replaced by halogen atoms, e.g. trifluoromethyl or difluoro methyl groups. By way of analogy, the terms “(C1-Cx) hydroxyalkyl” or “(C1-Cx) aminoalkyl” refer to the above defined “(C1-Cx) alkyl” groups wherein one or more hydrogen atoms are replaced by one or more hydroxy (OH) or amino group respectively. Thus, “(C 1 -C 6 )hydroxyalkyl” or “(C 1 - C6)aminoalkyl” refers to said hydroxy- or amino-alkyl groups wherein the number of carbon atoms is in the range 1 to 6. The definition of aminoalkyl encompasses alkyl groups (i.e. “(C1-C6)alkyl” groups) substituted by one or more amino groups (-NR 4 R 5 ). An example of aminoalkyl is a mono- aminoalkyl group such as R4R5N-(C1-C6)alkyl, or –(CH2)mNR4R5. Wherein R4 and R5 and m are as defined in the detailed description of the invention. With reference to the substituent R4 and R5 as above defined, it is here further explained that when either R 4 and R 5 are taken together with the nitrogen atom they are linked to form a 5 to 6 membered heterocyclic radical, at least one further ring carbon atom in the said heterocyclic radical may be replaced by at least one heteroatom or hetero-group (e.g. N, NH, S or O) or may bear an - oxo (=O) substituent group. The said heterocyclic radical might be further optionally substituted on the available points in the ring, namely on a carbon atom, or on an heteroatom or hetero-group available for substitution. Thus, Examples of said heterocycle radicals are 1-pyrrolidinyl, 1- piperidinyl, 1-piperazinyl, 4-morpholinyl, piperazin-4yl-2-one, 4-methylpiperazine-1-yl. The term “(C3-C10)cycloalkyl” likewise “(C3-C6)cycloalkyl” refers to saturated cyclic hydrocarbon groups containing the indicated number of ring carbon atoms. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl, and polycyclic ring systems such as adamantan-yl. The expression “Aryl” refers to mono, bi- or tri-cyclic carbon ring systems which have 6 to 20, preferably from 6 to 15 ring atoms, wherein at least one ring is aromatic. The expression “heteroaryl” refers to mono-, bi- or tri-cyclic ring systems with 5 to 20, preferably from 5 to 15 ring atoms, in which at least one ring is aromatic and in which at least one ring atom is a heteroatom (e.g. N, S or O). Examples of aryl or heteroaryl monocyclic ring systems include, for instance, phenyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furanyl radicals and the like. Examples of aryl or heteroaryl bicyclic ring systems include naphthalenyl, biphenylenyl, purinyl, pteridinyl, pyrazolopyrimidinyl, benzotriazolyl, benzoimidazole-yl, quinolinyl, isoquinolinyl, indolyl, isoindolyl, indazolyl, benzothiopheneyl, benzodioxinyl, dihydrobenzodioxinyl, indenyl, dihydro-indenyl, dihydrobenzo[1,4]dioxinyl, benzothiazole-2-yl, dihydrobenzodioxepinyl, benzooxazinyl, 1,2,3,4-tetrahydroisoquinoline-6-yl, 4,5,6,7- tetrahydrothiazolo[4,5-c]pyridine, 4,5,6,7-tetrahydrobenzo[d]thiazol-2-yl, 5,6,7,8-tetrahydro-1,7- naphthyridine, radicals and the like. Examples of aryl or heteroaryl tricyclic ring systems include fluorenyl radicals as well as benzocondensed derivatives of the aforementioned heteroaryl bicyclic ring systems. The derived expression “(C 3 -C 10 )heterocycloalkyl” likewise “(C 3 -C 6 )heterocycloalkyl” refers to saturated or partially unsaturated mono, bi- or tri- cycloalkyl groups of the indicated number of carbons, in which at least one ring carbon atom is replaced by at least one heteroatom (e.g. N, NH, S or O) and/or may bear an -oxo (=O) substituent group (e.g. C(=O), S(=O)2). Said heterocycloalkyl (i.e. heterocyclic radical or group) is further optionally substituted on the available points in the ring, namely on a carbon atom, or on an heteroatom available for substitution. Substitution on a carbon atom includes spiro disubstitution as well as substitution on two adjacent carbon atoms, in both cases thus form additional condensed 5 to 6 membered heterocyclic ring. The derived expression “hydroxyl-(C 3 -C 6 )heterocycloalkyl” refers to the said heterocycloalkyl groups substituted on the available points in the ring, namely on a carbon atom, or on an heteroatom available for substitution with an hydroxyl group. Examples of (C3-C6) heterocycloalkyl are represented by: oxetanyl, tetrahydro-furanyl, pyrrolidinyl, imidazolidinyl, thiazolidinyl, piperazinyl, piperidinyl, morpholinyl, thiomorpholinyl, dihydro- or tetrahydro-pyridinyl, tetrahydropyranyl, pyranyl, 2H- or 4H-pyranyl, dihydro- or tetrahydrofuranyl, dihydroisoxazolyl, pyrrolidin-2-one-yl, dihydropyrrolyl, 5-oxopyrrolidin-3-yl, (1R,5S,6r)-3-oxabicyclo[3.1.0]hexan-6-yl, 1,1-dioxidothiomorpholino, octahydrocyclopenta[c]pyrrol-5-yl, 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl; 4,5,6,7- tetrahydrothiazolo[5,4-c]pyridine-2-yl radicals and the like. Other Examples of said heterocycle radicals are 1-methyl-2-pyrrolidinyl, piperazin-4-yl-2- one, 4-methylpiperazine-1-yl, 1-methylpiperidin-4-yl, 4-metylpiperazine-1-yl-2-one, 7-methyl- 2,7-diazaspiro[3.5]nonan-2-yl, 2-methyl-2,9-diazaspiro[5.5]undecan-9-yl, 9-methyl-3,9- diazaspiro[5.5]undecan-3-yl, and (3aR,6aS)-5-methyl-octahydropyrrolo[3,4-c]pyrrol-2-yl, hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl, 2-oxa-6-azaspiro[3.4]octan-6-yl, 7-oxo-6-oxa-2- azaspiro[3.4]octan-2-yl, 3-hydroxyoxetan-3-yl. The term “Aryl(C1-C6)alkyl” refers to an aryl ring linked to a straight-chained or branched alkyl group wherein the number of constituent carbon atoms is in the range from 1 to 6, e.g. phenylmethyl (i.e. benzyl), phenylethyl or phenylpropyl. Likewise the term “Heteroaryl(C1-C6)alkyl” refers to an heteroaryl ring linked to a straight- chained or branched alkyl group wherein the number of constituent carbon atoms is in the range from 1 to 6, e.g. furanylmethyl. The term “alkanoyl”, refers to HC(O)- or to alkylcarbonyl groups (e.g. (C1-C6)alkylC(O)-) wherein the group “alkyl” has the meaning above defined. Examples include formyl, acetyl, propanoyl, butanoyl. The term “(C 1 -C 10 ) alkoxy” or “(C 1 -C 10 ) alkoxyl”, likewise “(C 1 -C 6 ) alkoxy” or “(C 1 -C 6 ) alkoxyl” etc., refers to a straight or branched hydrocarbon of the indicated number of carbons, linked to the rest of the molecule through an oxygen bridge.“(C 1 -C 6 )Alkylthio” refers to the above hydrocarbon linked through a sulfur bridge. The derived expression “(C 1 -C 6 )haloalkoxy” or “(C 1 -C 6 )haloalkoxyl” refers to the above defined haloalkyl, linked through an oxygen bridge. Example of (C1-C6)haloalkoxy is difluoromethoxy, trifluoromethoxy. Likewise derived expression “(C3-C6)heterocycloalkyl-(C1-C6)alkyl” and “(C3- C 6 )cycloalkyl-(C 1 -C 6 )alkyl” refer to the above defined heterocycloalkyl and cycloalkyl groups linked to the rest of the molecule via an alkyl group of the indicated number of carbons, for example piperidin-4-yl-methyl, cyclohexylethyl. The derived expression “(C1-C6)alkoxy (C1-C6)alkyl” refers to the above defined alkoxy group linked to the rest of the molecule via an alkyl group of the indicated number of carbons, for example methoxymethyl. Likewise “(C1-C6)haloalkoxy(C1-C6)alkyl” refers to the above defined (C1-C6)haloalkoxy” group linked to the rest of the molecule via an alkyl group of the indicated number of carbons, for example difluoromethoxypropyl. Likewise “(C 1 -C 6 )alkoxycarbonyl” refers to the above defined alkoxy group linked to the rest of the molecule via an carbonyl group. And “(C 1 -C 6 )alkoxycarbonyl-(C 1 -C 6 )alkyl” refers to the above defined alkoxy group linked to the rest of the molecule via an carbonyl group further enchained with an alkyl group of the indicated number of carbons, for example methoxycarbonylmethyl. And “(C1-C6)alkoxycarbonyl-(C1-C6)alkylthio consequently refer to enchained groups like methoxycarbonylmethylthio. An oxo moiety is represented by (O) as an alternative to the other common representation, e.g. (=O). Thus, in terms of general formula, the carbonyl group is herein preferably represented as –C(O)– as an alternative to the other common representations such as –CO–, –(CO)– or – C(=O)–. In general the bracketed group is a lateral group, not included into the chain, and brackets are used, when deemed useful, to help disambiguating linear chemical formulas; e.g. the sulfonyl group -SO2- might be also represented as–S(O)2– to disambiguate e.g. with respect to the sulfinic group –S(O)O–. When a numerical index the statement (value) “p is zero” or “p is 0” means that the substituent or group bearing the index p (e.g. Ip) is absent, that is to say no substituent, other than H when needed, is present. Likewise when the index is attached to a bridging divalent group (e.g. (CH2)n) the statement “n in each occurrence is zero…” or “n is 0” means that the bridging group is absent, that is to say it is a bond. Whenever basic amino or quaternary ammonium groups are present in the compounds of formula (I), physiological acceptable anions, selected among chloride, bromide, iodide, trifluoroacetate, formate, sulfate, phosphate, methanesulfonate, nitrate, maleate, acetate, citrate, fumarate, tartrate, oxalate, succinate, benzoate, p-toluenesulfonate, pamoate and naphthalene disulfonate may be present. Likewise, in the presence of acidic groups such as COOH groups, corresponding physiological cation salts may be present as well, for instance including alkaline or alkaline earth metal ions. Compounds of formula (I) when they contain one or more stereogenic center, may exist as optical stereoisomers. Where the compounds of the invention have at least one stereogenic center, they may accordingly exist as enantiomers. Where the compounds of the invention possess two or more stereogenic centers, they may additionally exist as diastereoisomers. It is to be understood that all such single enantiomers, diastereoisomers and mixtures thereof in any proportion are encompassed within the scope of the present invention. The absolute configuration (R) or (S) for carbon bearing a stereogenic center is assigned on the basis of Cahn-Ingold-Prelog nomenclature rules based on groups’ priorities. “Single stereoisomer”, “single diastereoisomer” or “single enantiomer”, when reported near the chemical name of a compound indicate that the isomer was isolated as single diastereoisomer or enantiomer (e.g via chiral chromatography) but the absolute configuration at the relevant stereogenic center was not determined/assigned. Atropisomers result from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers (Bringmann G et al, Angew. Chemie Int. Ed.44 (34), 5384-5427, 2005. doi:10.1002/anie.200462661). Oki defined atropisomers as conformers that interconvert with a half-life of more than 1000 seconds at a given temperature (Oki M, Topics in Stereochemistry 14, 1-82, 1983). Atropisomers differ from other chiral compounds in that in many cases they can be equilibrated thermally whereas in the other forms of chirality isomerization is usually only possible chemically. Separation of atropisomers is possible by chiral resolution methods such as selective crystallization. In an atropo-enantioselective or atroposelective synthesis one atropisomer is formed at the expense of the other. Atroposelective synthesis may be carried out by use of chiral auxiliaries like a Corey Bakshi Shibata (CBS) catalyst, an asymmetric catalyst derived from proline, or by approaches based on thermodynamic equilibration when an isomerization reaction favors one atropisomer over the other. Racemic forms of compounds of formula (I) as well as the individual atropisomers (substantially free of its corresponding enantiomer) and stereoisomer-enriched atropisomer mixtures are included in the scope of the present invention. The invention further concerns the corresponding deuterated derivatives of compounds of formula (I). In the context of the present invention, deuterated derivative means that at least one position occupied by a hydrogen atom is occupied by deuterium in an amount above its natural abundance. Preferably, the percent of deuterium at that position is at least 90%, more preferably at least 95%, even more preferably 99%. All preferred groups or embodiments described above and here below for compounds of formula (I) may be combined among each other and apply as well mutatis mutandis. As above mentioned, the present invention provides compounds of general formula (I), acting as JAK inhibitors, to processes for the preparation thereof, pharmaceutical compositions comprising them either alone or in combination with one or more active ingredient, in admixture with one or more pharmaceutically acceptable carriers. In a first aspect the present invention provides a class of compounds of formula (I) R 1 is a heteroaryl group imidazo[1,2-b]pyridazin-3-yl, pyrazolo[1,5-a]pyrimidin-3-yl, 3-oxo-(3,4-dihydropyrazin-2- yl)amino; R 2 is a group of formula that is a substituent group linked to the molecule scaffold, wherein V is absent (meaning it is a bond) or is a divalent group selected from O, S, N(R 6 ), C(O)N(R6), N(R6)C(O), N(R6)C(O)O, N(R6)S(O)2, N(R6)C(O)N(R6); Q is selected from the group consisting of H, (C 1 -C 6 )alkyl, (C 1 -C 6 )hydroxyalkyl, (C 1 - C6)alkoxy, -(CH2)mNR4R5, (C3-C8)cycloalkyl, (C3-C10)heterocycloalkyl, -S-(C3- C 6 )heterocycloalkyl, and -N(R 6 )-(C 3 -C 6 )heterocycloalkyl; wherein said (C 3 -C 8 )cycloalkyl and (C3-C10)heterocycloalkyl are further optionally substituted by one or more substituent selected from the group consisting of -OH, oxo (i.e. (=O)), (C 1 -C 10 )alkyl, (C 1 -C 6 )alkoxy; halogen, (C 1 - C6)haloalkyl, alkanoyl, (C1-C6)hydroxyalkyl, (C1-C6)alkoxy(C1-C6)alkyl, - N(R 6 )(CH 2 ) m C(O)NR 4 R 5 , -(CO)NR 4 R 5 , -(CH 2 ) m NR 4 R 5 , (C 3 -C 8 )cycloalkyl(C 1 -C 6 )alkyl; (C 3 - C6)heterocycloalkyl(C1-C6)alkyl, (C3-C6)heterocycloalkyl and, hydroxyl-(C3- C 6 )heterocycloalkyl; R 3 is a bicyclic moiety which is a substituent group linked to the scaffold graphically represented by wherein the dashed line --- indica double bond X is selected from N, S,C; Y is selected from C, N; Z is selected from C, N, O; K is absent (meaning a bond) or selected from O, C, S; G is absent (meaning a bond) or selected from C, O; (wherein each of X, Y, Z, K and G will be linked in the ring and with the right number of H or substituent according to their valency) is the point of attachment of a substituent group to the rest of the molecule (meaning the inking the substituent group R1, R2 or R3 to the molecule scaffold) n and m are in each occurrence independently 0 or an integer selected from 1, 2, 3 and 4 (n=0 found in (R7)n means there is no substituent R7, i.e. R7 being H, likewise n=0 or m=0 means there is no linking group -(CH 2 ) n - or -(CH 2 ) m , i.e. -(CH 2 ) n - or -(CH 2 ) m - being respectively a bond); R4 and R5, the same or different, are selected from the group consisting of -H, (C 1 -C 6 )alkyl, (C1-C6)haloalkyl, and (C 3 -C 6 )heterocycloalkyl; R6 is in each occurrence independently selected from the group consisting of H, (C1- C 6 )alkyl, (C 1 -C 6 )hydroxyalkyl; R7 is one or more group (preferably 1 or 2) in each occurrence independently selected from the group consisting of -OH, oxo (that is =O), (C 1 -C 6 )alkyl, halogen, (C 1 -C 6 )haloalkyl, (C 1 - C6)hydroxyalkyl, -(CH2)mNR4R5, (C1-C6)alkyl-S(O)2- and (C1-C6)alkyl-S(O)2N(R6)-; R 8 is selected in the group consisting of (C1-C6)alkoxy, (C 1 -C 6 )haloalkoxy; preferably R8 is methoxy, fluormethoxy, difluormetoxy; single enantiomers, diastereoisomers and mixtures thereof in any proportion or a pharmaceutically acceptable salt or solvate thereof. In a preferred embodiment the bicyclic moiety R 3 is selected from In a further preferred he group consisting of J1- J13

A group of p herein R 3 is J1 and R8 is methoxy Such preferred group of compounds are represented by the formula Ia

Wherein R 1 is pyrazolo[1,5-a]pyrimidin-3-yl or (3-oxo-3,4-dihydropyrazin-2-yl)amino V is a divalent group selected from C(O)N(R 6 ), N(R 6 )C(O)O; Q is selected from the group consisting of (C1-C6)alkyl,(C1-C6)alkoxy, -(CH2)mNR4R5, (C3- C 8 )cycloalkyl, and (C 3 -C 12 )heterocycloalkyl; wherein said (C 3 -C 8 )cycloalkyl and (C 3 - C12)heterocycloalkyl are further optionally substituted by one or more substituent selected from the group consisting of oxo (i.e. a group (=O)) , (C 1 -C 10 )alkyl, halogen, (C 1 -C 6 )hydroxyalkyl, - (CO)NR4R5; (C3-C8)cycloalkyl(C1-C6)alkyl; (C3-C6)heterocycloalkyl; n and m are in each occurrence independently 0 or an integer selected from 1, 2, 3 and 4; R 4 and R 5, the same or different, are selected from the group consisting of -H, (C1-C6)alkyl, (C1-C6)haloalkyl; single enantiomers, diastereoisomers and mixtures thereof in any proportion or a pharmaceutically acceptable salt or solvate thereof. Even more preferred in this group are compounds of the above formula Ia wherein V is N(R 6 )C(O)O; Q is selected from the group consisting of (C1-C6)alkyl, (C1-C6)alkoxy, -(CH2)mNR4R5, and (C 3 -C 6 )heterocycloalkyl; wherein said (C 3 -C 6 )heterocycloalkyl is a group wherein X 1 is selected from CH2, O, S, NH, NCH3, (C=O) and S(=O)2. Further preferred are compunds of the above formula Ia wherein V is C(O)N(R6), Q is selected from the group consisting of -(CH2)mNR4R5, and (C3-C6)heterocycloalkyl; wherein said (C 3 -C 6 )heterocycloalkyl is a group wherein X 1 is selected from the group consisting of CHR9, O, S, NH, NCH3, CF2, (C1-C6) alkoxy or (C1-C6)alkoxy(C1-C6)alkyl, wherein R 9 is H or -(CO)NR 4 R 5 . Further preferred are compounds of the above formula Ia wherein Q is (C3-C6)heterocycloalkyl, -S-(C3-C6)heterocycloalkyl or -N(R6)-(C3- C6)heterocycloalkyl selected from the group consisting of: W ere n pre era y suc tetra y ro- ,3 -5 - uro[3, -c]pyrro - -one s a 10 According to specific embodiments, the present invention provides the compounds of examples 1 to 86, as listed in the table below, or pharmaceutical acceptable salts and solvates thereof. 17 Example No Chemical Name - 18 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin-7-yl)-N-(1- 17 methylazetidin-3-yl)-6-(pyrazolo[15-a]pyrimidin-3-yl)-1H-pyr azolo[43- - 19 (R)-1-methylpyrrolidin-3-yl (1-(6-methoxy-3,4-dihydro-2H- 32 benzo[b][14]thiazin-7-yl)-6-(pyrazolo[15-a]pyrimidin-3-yl)-1 H- - 20 1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)-N-(2-( 4- 47 (methoxymethyl)piperidin-1-yl)ethyl)-6-(pyrazolo[15-a]pyrimi din-3-yl)-1H- - - - 21 2-(1,1-dioxidothiomorpholino)ethyl (1-(6-methoxy-3,4-dihydro-2H- 62 benzo[b][14]oxazin-7-yl)-6-(pyrazolo[15-a]pyrimidin-3-yl)-1H - - 22 N-(2-(2,2-dimethyl-6-oxomorpholino)ethyl)-1-(6-methoxy-3,4-d ihydro-2H- 77 benzo[b][14]oxazin-7-yl)-6-(pyrazolo[15-a]pyrimidin-3-yl)-1H - - - - be prepared from readily available starting materials using general methods and procedures as described in the experimental part below or by using slightly modified processes readily available to those of ordinary skill in the art. Although a particular embodiment of the present invention may 5 be shown or described herein, those skilled in the art will recognize that all embodiments or aspects of the present invention can be prepared using the methods described herein or by using other known methods, reagents and starting materials. When typical or preferred process conditions (i.e. reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. While the optimum reaction 10 conditions may vary depending on the particular reactants or solvent used, such conditions can be readily determined by those skilled in the art by routine optimization procedures. Processes of 23 preparation described below and reported in the following schemes should not be viewed as limiting the scope of the synthetic methods available for the preparation of the compounds of the invention. In some cases a step is needed in order to mask or protect sensitive or reactive moieties, 5 generally known protective groups (PG) could be employed, in accordance with general principles of chemistry (Protective group in organic syntheses, 3rd ed. T. W. Greene, P. G. M. Wuts). Compounds of formula (I), here reported again for clarity, including all here above listed, can be usually prepared according to the procedures shown in the schemes below. Where a specific detail or step differs from the general schemes it has been detailed in the specific examples, and/or 10 in additional schemes. Compounds of formula (I) can be prepared according to scheme 1. Compound IV is an intermediate where general group r1, r2 , and r3 can be converted into R1, R2 and R3 respectively 15 by means of procedures well known to those skilled in the art such as protective groups deprotection and/or functional group conversion that may involve more than one step. Said procedures can be applied to one or more of those groups (r1, r2 ,and r3) to allow the conversion of intermediate IV into compound of general formula I and they are detailed in the experimental section for specific examples. It is apparent that in the case such conversions are not needed (when 20 r 1, r 2 , and r 3 correspond to R 1 , R 2 and R 3 respectively), any general approach described below for the preparation of intermediate IV will provide a compound of general formula I.

24 ) n 3 R 7 R ( Y X Z 8 r r 3 o / n o r s i a l o a v b 2 a n Rs i o a l r o s i r 3 e r i t 1 m o / p e r r e d n o ( r d e n s h t a r a e 2 R G r P u f r 1 h n w o r 3 N N r 2 V I N r 1 r o r r s r o s u r o s r c u u e c e c e d g e A r p r p r p z n i l R 1 2 3 y l a p RR t u a o r 1 I n a n a n a c c I d s I r r r s o 1 o 2 o 3 P o r c RRR d e t d c e t d e t e t c e o t c e t r o o p r p r p r 3 y l y b l y l 1 N N a t b i a t b a t u i u i E r 2 s a s u a s M E I I r o r a o r o R 1 R 2 R 3 H s C i s s S 1 i 2 i l N r r r 3 C 25 For synthetic convenience, cyclic and/or exocyclic NH moieties that may be present in r2 and/or r3 may need to be protected during the synthetic sequences. Suitable protective groups for protecting such NH moieties can be carbamate type protective groups such as Boc (tert- butoxycarbonyl) and/or hemiaminals such as SEM (2-(Trimethylsilyl)ethoxymethyl). A Boc 5 group can be inserted by reacting the NH derivative with Boc 2 O in the presence of a base like for example DMAP or pyridine, and in an organic solvent like THF or DMF at room temperature. A SEM group can be inserted by reacting the NH derivative with SEM-Cl (2- (Trimethylsilyl)ethoxymethyl chloride) in a suitable organic solvent like DMF and in the presence of a strong base like NaH or LiHMDS (lithium hexamethyldisilazide), at low temperature like 0ºC. 10 A Boc group can be easily removed by treating Boc protected intermediate IV (or from other convenient upstream intermediates) in acidic conditions with an organic or an inorganic strong acid. For example, Boc group can be cleaved by treating the intermediates with trifluoroacetic acid neat or in mixture with an organic solvent such as DCM, DCE, THF or similar, typically at room temperature for a few hours (typically 1 to 3 h). A SEM group can be removed by treating 15 intermediate IV (or from other convenient upstream intermediates) in acidic conditions similarly to Boc group removal. In some cases, acidic treatment may lead incomplete SEM removal, therefore a further treatment with concentrated ammonia may be necessary to completely remove formaldehyde adducts resistant to acidic treatment. It is understood that insertion and removal of NH protective groups in r2/r3 can be done where it is more convenient in the synthetic sequence. 20 Compound of formula I (or intermediate IV) can be obtained by direct introduction of R 1 (or r1) through a metal/palladium catalysed cross coupling reaction such as Suzuki coupling, Stille coupling, Buchwald-Hartwig or similar (Strategic application of named reactions in organic synthesis, L. Kurti, B. Czako, Ed.2005) by reaction of intermediate II with intermediate III. For example, a suitable palladium catalysed cross coupling for introducing R 1 , when it is an 25 pyrazolo[1,5-a]pyrimidin-3-yl, is a Suzuki coupling. Suzuki coupling can be performed by reacting intermediate II with the suitable boronic acid or boron pinacolate derivative (intermediate III, where r1 is pyrazolo[1,5-a]pyrimidin-3-yl and A is dihydroxyboryl or 4,4,5,5-tetramethyl- 1,3,2-dioxaborolanyl) in the presence of a Pd catalyst such as tetrakistriphenylphosphinepalladium(0), PdCl2(dppf)2, or a ligand-palladacycle precatalyst for30 example XPhos-Pd-G3 [(2-Dicyclohexylphosphino-2ƍ,4ƍ,6ƍ-triisopropyl-1,1ƍ-bip henyl)[2-(2ƍ- amino-1,1ƍ-biphenyl)]palladium(II) methanesulfonate], in an organic solvent such as 1,4-dioxane, THF, 1,2-dimethoxyethane, 2-propanol or DMF, with or without water, in the presence of an inorganic base such as an alkaline carbonate (for example Cs2CO3 or K2CO3) or an inorganic phosphate (for example K 3 PO 4 ), under heating (typically in the range of 50-100ºC) for a few hours 35 (typically 1 to 3h). Boronic acid and boronic pinacolate esters are generally commercially available 26 or may be readily prepared by those skilled in the art starting from commercially available reagents. A suitable palladium catalyzed cross coupling for introducing R 1 when it is an imidazo[1,2- b]pyridazin-3-yl, is a Stille coupling that can be performed by reacting intermediate II with the 5 suitable stannane reagent (intermediate III, where r 1 is imidazo[1,2-b]pyridazin-3-yl and A is tributylstannyl or a trimethylstannyl) in the presence of an appropriate palladium catalyst (such as Pd(PPh 3 ) 2 Cl 2 ) in a polar organic solvent (for example DMF or dioxane with or without additives (like base or lithium salt). Stannanes are generally commercially available or may be readily prepared by those skilled in the art starting from commercially available reagents. 10 A suitable palladium catalyzed cross coupling for introducing r 1 , when it is an (3-oxo-3,4- dihydropyrazin-2-yl)amino, is a Buchwald-Hartwig coupling. For synthetic convenience the lactam group of (3-oxo-3,4-dihydropyrazin-2-yl)amino needs to be masked as an alkoxyimmino group (such as methoxy immino, -C(OMe)=N-) and that is deprotected at the end of the synthesis from intermediate IV. Intermediate II and intermediate III (where r1 is 3-methoxypyrazin-2-15 aminyl and A is H) can be reacted to give intermediate IV (where r 1 is 3-methoxypyrazin-2- aminyl) in the presence of a suitable ligand palladacyle system such as XPhos-Pd-G3 (2- Dicyclohexylphosphino-2ƍ,4ƍ,6ƍ-triisopropyl-1,1ƍ-bipheny l)[2-(2ƍ-amino-1,1ƍ- biphenyl)]palladium(II) methanesulfonate) or RuPhos-Pd-G3 (2-Dicyclohexylphosphino-2ƍ,6ƍ- diisopropoxy-1,1ƍ-biphenyl)[2-(2ƍ-amino-1,1ƍ-biphenyl)]pa lladium(II) methanesulfonate) or in 20 general a suitable Pd source (for example Pd 2 (dba) 3 or Pd(OAc) 2 ) with a suitable phosphine ligand such as biphenylphosphine ligand type (RuPhos, X-Phos, or similar), in the presence of a strong organic base such as sodium tert-butoxide or an inorganic base such as Cs 2 CO 3 , in an organic solvent such 1,4-dioxane, THF or toluene, under heating at high temperature (typically 80-120ºC), for a few hours (typically overnight). Preparation of compound of formula I wherein R 1 is 3-oxo- 25 3,4-dihydropyrazin-2-yl)amino can be prepared by mean of demethylation of the corresponding methoxy-immino derivative by treatment of the protected precursor with TMS-Cl (trimethylsilyl chloride) and sodium iodide in acetonitrile for 1 to 5 h at 60 - 100ºC; these condition can also lead to Boc/SEM groups deprotection eventually present in r 2 /r 3 . The above described process may provide at least one non limiting synthetic route for the 30 preparation of examples of the invention starting from the appropriate intermediate II. A possible approach for the preparation of intermediate II is reported in scheme 2. Intermediate II can be prepared by means of N-arylation of intermediate V with an halide intermediate VI by using a copper catalyzed Ullmann type reaction. An Ullmann reaction between a NH heteroaryl and an aryl/heteroaryl halide (bromide or iodide) can be performed in the 35 presence of a suitable copper(I) catalyst/promoter such as CuI, Cu2O or CuTC (copper thiophene 27 carboxylate), ligandless or with a suitable ligand such as N,N-dimethylglicine, proline, phenantroline or dimethylcyclohexane-1,2-diamine (DMCHA), in the presence of an inorganic base such as K 2 CO 3 or Cs 2 CO 3 , by heating (typically 90-150ºC) in a polar organic solvent such as DMSO, DMF or DMA, overnight or longer. 5 In another approach, intermediate II when r3/R3 is J2, indicated as II i (for n:0) and II ii (for n:1, R 7 : 3-oxo) can be synthesized by means of a multistep synthesis according to scheme 3. Intermediate V and intermediate VIIa can undergo an aromatic nucleophilic substitution (SNAr) to give intermediate VIIIa, for example by reacting them in an organic solvent like DMF, DMSO 10 or 1,4-dioxane, in the presence of an organic base like DBU or DIPEA or an inorganic base like K2CO3 or Cs2CO3, at RT or higher (up to 120ºC), for a few hours (typically 1 to 4 hours). Similarly, intermediate VIIIb and VIIIc can be made from V by reaction with VIIb and VIIc respectively. Intermediate VIIIa can be reacted with mercaptoethanol (r5-SH, r5 is ethanol-2yl) under Pd- catalyzed C-S coupling conditions to give intermediate IXa. C-S coupling can be performed by 15 reacting aryl bromide VIIIa and mercaptoethanol in the presence of a suitable catalytic system like as Pd 2 (dba) 3 / Xantphos or another suitable palladium source / phosphine source, in an organic solvent as 1,4-dioxane, toluene or DMA, in the presence of an organic base like DIPEA or DBU, at temperature up to 100ºC for a few hours (typically 3 -5 hours). Intermediate IXb can be synthesized by aromatic nucleophilic substitution (SNAr) of intermediate 20 VIIIa with methyl 2-sulfanylacetate (r 5 -SH, r 5 is methyl ethanoat-2-yl) by reacting them in an organic solvent like acetonitrile or dioxane, under heating at high temperature (typically 120ºC), for a few hours (typically 3 to 6 hours). 29 Intermediate IXa can be converted into intermediate II i by a three steps process that involves: 1) nitro reduction, 2) chlorination and cyclization. Nitro reduction can be performed by treating intermediate IXa with a reducing agent like iron powder or zinc powder, in an organic solvent like methanol or ethanol, in the presence of an aqueous solution of a weak inorganic acid 5 salt like ammonium chloride, by heating at temperature up to 80ºC for a few hours (typically 4-5 hours). Subsequent conversion of alcohol to the corresponding chloride can be achieved by reaction with neat thionyl chloride or oxalyl chloride at low temperature (around 0ºC). Chloride intermediate can be cyclized to give intermediate II i by heating (typically 80-100ºC) in an appropriate organic solvent like DMF or 1,4-dioxane, in presence of an inorganic base like 10 potassium carbonate or sodium carbonate and in the presence of an additive like sodium iodide. Intermediate IXb can be converted into intermediate II ii by a process that involves nitro reduction and cyclization to lactam. Nitro reduction of intermediate IXb can be obtained similarly to what described for intermediate IXa. Cyclization can occur spontaneously during nitro reduction step or promoted in a separate step by treatment with a strong organic acid like TFA. 15 In an alternative approach, intermediate II ii can be converted into intermediate II i by reduction of lactam with borane like BH3*THF complex into the corresponding lactam. In another approach, intermediate II i can be converted into intermediate II iii by oxidation of sulfide to sulfone using an oxidazing agent like m-CPBA, in an organic solvent like DCM, at temperature around 0ºC. 20 In another way, intermediates II when r 3 /R 3 is J8, indicated as II iv in scheme 3, can be prepared by a multistep process starting from intermediate VIIIa. In the first step, intermediate VIIIa can undergo aromatic nucleophilic substitution with a malonate ester like diethyl malonate, by heating reagents (typically 60-70ºC) in an organic solvent like DMSO or DMF. In the second step, treatment with iron powder at high temperature (for example 90ºC) in acetic acid as a solvent 25 can promote one pot nitro reduction, cyclization and decarboxylation to give intermediate II iv . In another approach, intermediate II iv can be converted to II v by a two step process that involves 1) NH protection with SEM group and 2) cyclopropanation with a sulphur vinyl ylide like vinyl diphenyl sulfonium triflate. In a different way, intermediate II when r3/R3 is J6 (with n:0) indicated as intermediate II vi 30 and intermediate II when r 3 /R 3 is J1 (with n:3, R 7 : 3-oxo, 1,1-difluoro) indicated as intermediate II vii , can be prepared according to scheme 4. Intermediate VIIIb can be converted into intermediate X by a two step process that involves 1) de-allylation and 2) nitro reduction. De-allylation reaction can be performed by treating (typically at 50-70ºC) intermediate VIIIb with an inorganic base like potassium carbonate or 35 cesium carbonate, in an organic solvent like methanol or ethanol and in the presence of a Pd catalyst such as tetrakis(triphenylphosphine)palladium(0) or palladium chloride (II). Subsequent nitro reduction can be performed similarly to what described in scheme 3 for the reduction of intermediate IXa. Intermediate X can be selectively acylated on the anilinic nitrogen to form the corresponding amide, by transammidation of methyl 2-2,2-dibromo-difluoroacetate promoted by DABAL-Me 3 at high temperature (typically 130ºC) in an organic solvent like THF. Subsequent cyclization of amide intermediate, promoted by an organic base like DBU or DIPEA by heating (100-120 ºC) in an organic solvent like THF or dioxane, can give intermediate II vii .

32 In another approach, intermediate II vi can be obtained from intermediate VIIIc by a two step process that involve mesylation of the free hydroxyl group in r 4 (when r4 is propane-3-ol-1-oxyl) and followed by one pot nitro reduction and cyclization. Mesylation can be carried out by treatment of alcohol with methanesulfonyl chloride in an organic solvent like DMF at room temperature, 5 followed by one pot nitro reduction and cyclization by heating intermediate (around 80ºC) intermediate with iron, in an organic solvent like ethanol, in the presence of a weak inorganic acid like aqueous ammonium chloride. In a different way, intermediate II when r 2 is -NR 6 (CH 2 ) n Q', indicated as IIa, can be made according to scheme 5. Q' is a group that, if differs from Q, can be easily converted into Q by 10 means of procedures well known to those skilled in the art such as protective groups deprotection and/or functional group conversion that may involve more than one step.

33 Intermediate XI can be converted into intermediate Va by a multistep process that involves 1) NH protection, 2) C-N coupling with HNR 6 (CH 2 ) n Q' and 3) N-PG deprotection. A suitable protective group for this NH of intermediate XI is for example a THP group (tetrahydropyranyl). THP introduction can be achieved by heating around 40ºC intermediate XI with dihydropyran in an organic solvent like THF, DCM or mixture and in the presence of a sulfonic acid like methanesulfonic acid for several hours (12 h or more). A C-N coupling between THP protected intermediate XI and HNR6(CH2)n-Q' may be performed by heating (typically 60-100ºC) the heteroaromatic iodide and the amine in an organic solvent like DMF or DMSO, in the presence of copper(I) catalyst/promoter such as CuI, Cu2O or CuTC (copper thiophene carboxylate), ligandless or with a suitable ligand such as proline, N,N-dimethylglicine, dimethylcyclohexane- 1,2-diamine (DMCHA), and in the presence of an inorganic base as K2CO3 or Cs2CO3. Finally, THP removal can be performed by acidic treatment with an organic acid like TFA or methanesulphonic acid, in the presence of a silane scavenger like triethylsilane, in an organic solvent like DMF or THF for a few hours (around 2h) at room temperature. Conversion of intermediate Va to intermediate IIa can be performed using similar conditions to what described in scheme 2 for the conversion of intermediate V into intermediate II. In another way, intermediate XI and intermediate VIIa can be converted by SNAr into intermediate XIIa similarly to what described in scheme 3 for the conversion of V and VIIa to VIIIa. Conversion of intermediate XIIa into VIIIa' can be done using similar conditions that were reported above for the C-N coupling. Finally, conversion of VIIIa' to intermediate IIa (when r3/R3 is J2) can be done applying similar conditions already reported in scheme 3 for the conversion of IXb to II ii . In another approach, intermediates II can be obtained from further elaboration of r 2 group by general accepted methods and in accordance with principles of chemistry. In the following schemes, the most common transformations that can be used to obtain specific intermediates II have been detailed. For sake of clarity they were labelled with an additional letter index.

35 In one of these alternative approaches, intermediate IIc (intermediate II when r 2 is - C(O)N(R6)-(CH2)n-Q') can be prepared according to scheme 6 starting from intermediate IIb (intermediate II when r 2 is -C(O)OH) by amide coupling with HNR 6 (CH 2 ) n Q'. An amide coupling 5 can be performed by reacting the amine and the acid in an organic solvent like DMF, DCM, or THF, in the presence of a coupling agent like HATU((1-[Bis(dimethylamino)methylene]-1H- 1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate), HBTU (O-(Benzotriazol-1-yl)- N,N,Nƍ,Nƍ-tetramethyluronium hexafluorophosphate) or COMU ((1-Cyano-2-ethoxy-2- oxoethylidenaminooxy)dimethylamino-morpholino-carbenium) and in the presence of an organic 10 base like DIPEA, TEA, or pyridine. In a different way, when for synthetic convenience intermediate IIb is synthesized as methyl ester, it can be converted directly to amide by a transammidation reaction with the corresponding amine promoted by DABAL-Me3 at high temperature (typically 130ºC) in an organic solvent like THF. 15 In another approach, intermediate IIc can be prepared from intermediate IIe by displacement of Lg'' by alkylation with primary/secondary nitrogen present in Q' moiety by treating at room temperature or higher (for example 40ºC) for a few hours to overnight the amine Q' and halide IIe in an organic solvent like DMF, acetone or 1,4-dioxane, in the presence of an organic base like 36 DIPEA, TEA or an inorganic base like sodium carbonate together with an inorganic iodide like sodium iodide. Intermediate IIe can be obtained from intermediate IIb and HNR6(CH2)nLg'' by an amide coupling reaction using similar conditions to those reported above in this scheme for the conversion of intermediate IIb to intermediate IIc. 5 Intermediate IIb can be made by using conditions reported above for general synthesis of intermediate II in scheme 2, scheme 3 and scheme 4 starting from a suitable intermediate V where r 2 is -COOH or -COOMe. In another approach, reported in scheme 7, intermediate IIg (or IIh) may be prepared from intermediate IIf and HO(CH 2 ) n Q' (or HNR 6 (CH 2 ) n Q') by a two steps / one pot process that involve: 10 1) isocyanate formation and 2) reaction with alcohol (or amine) to form the corresponding carbamate (or urea). Intermediate IIf can be reacted with bis(trichloromethyl) carbonate in an organic solvent as DCM, THF or Me-THF, at low temperature like dry ice temperature (-78ºC), in the presence of an organic base like TEA or DIPEA for times up to 1 or 2 hours to form the corresponding isocyanate; addition of intermediate alcohol (or amine) and reaction at room 15 temperature lead to the formation of carbamate of formula IIg (or urea of formula IIh). An alternative method to form the isocyanate intermediate from IIf can use carbonildiimidazole (CDI) in an organic solvent like 2-Me-THF by heating at temperature around 90ºC overnight. In a different approach, carbamate IIg can be obtained by reaction of amine IIf can be reacted with the corresponding alcohol chloroformate (Cl-C(O)O(CH2)nQ') in an organic solvent 20 like DCM or THF and in the presence of a base like pyridine or DIPEA.

37 In an alternative method, intermediate of formula IIg (and IIh) may be obtained from intermediate IIb and alcohol intermediate HO(CH 2 ) n Q' (or amine intermediate HNR 6 (CH 2 ) n Q') through a two steps / one pot process that involves: 1) acylazide formation and Curtius 5 rearrangement to give the corresponding isocyanate intermediate and 2) one pot reaction of isocyanate with alcohol (or amine) to form the corresponding carbamate (or urea). The corresponding acyl azide of intermediate IIb can be prepared by reaction of IIb with an azide source as azido(trimethyl)silane, in the presence of a suitable coupling agent such as an alkyl phosphonic anhydride like T3P (2,4,6-Tripropyl-1,3,5,2,4,6-trioxatriphosphorinane-2,4,6-tr ioxide10 solution), with an organic base like TEA or DIPEA, and in an organic solvent such as 2-methyl- THF, DMF or toluene. The following Curtius rearrangement can be done by thermal degradation around solvent reflux temperature (generally from 50ºC to 100ºC) for few hours (generally 1-3 hours) to give the corresponding isocyanate; after isocyanate formation, addition of alcohol intermediate HO(CH 2 ) n Q' (or amine HNR 6 (CH 2 ) n Q') and continuing reflux overnight can lead to 15 the formation of the corresponding carbamate of formula IIg (or urea IIh). 38 Intermediate IIf can be made by using conditions reported above for general synthesis of intermediate II in scheme 2, scheme 3 and following scheme 4 starting from a suitable intermediate V where r 2 is -NH 2 . 5 In an alternative approach reported in scheme 8, intermediate IIa when exocyclic NH is protected by a Boc group, indicated as Boc-IIa, can be prepared by a two step process that involves 1)Boc insertion at exocyclic amine and 2) alkylation with Lg-(CH 2 ) n -Q'. Boc protection can be performed by reaction of IIf with Boc2O in an organic solvent like THF or 1,4-dioxane and in the presence of a strong base like LiHMDS (lithium hexamethyldisilazide) or NaH, at low temperature 10 like 0ºC. Alkylation can be performed by reacting Boc intermediate with Lg-(CH2)n-Q' in an organic solvent like DMF or THF, in the presence of a strong base like NaH, at low temperature like 0ºC. In a different approach, intermediate II can be obtained by further elaboration of Q' group to Q'' by means of the functional group transformation summarized in table 1, full details of 15 transformation were provided in the experimental section. Q'' is a group that, if differs from Q, can be easily converted into Q by means of procedures well known to those skilled in the art such as protective groups deprotection and/or functional group conversion that may involve more than one step.

39 Table 1 -V( n type / steps group to Q'' by means of the functional group transformation summarized in table 2, full details of 5 transformation were provided in the experimental section. Q'' is a group that, if differs from Q, can be easily converted into Q by means of procedures well known to those skilled in the art such as protective groups deprotection and/or functional group conversion that may involve more than one step. 40 Table 2 -V(CH 2 ) n Q' -V(CH 2 ) n Q'' Reaction type / steps inase activity, in particular. inhibiting JAK kinase activity for the treatment of JAK-dependent diseases. 5 In one aspect the invention provides compounds according to the invention, i.e. a compound of formula (I) or a pharmaceutical composition thereof, for use as a medicament, preferably for the prevention and /or treatment of respiratory and specifically pulmonary disease. In a further aspect the invention provides the use of a compound (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of disorders 10 associated with JAK mechanisms, particularly for the treatment of disorders such as respiratory and pulmonary diseases. In particular, the invention provides compounds of formula (I) for use in the prevention and /or treatment of pulmonary disease selected from the group consisting of asthma, chronic obstructive pulmonary disease COPD, idiopathic pulmonary fibrosis (IPF)acute lung injury and 15 acute respiratory distress syndrome (ARDS). Moreover, the invention provides a method for the prevention and/or treatment of disorders associated with JAK mechanisms, said method comprising administering to a patient in need of 41 such treatment a therapeutically effective amount of a compound of the invention. In particular, the invention provides methods for the prevention and/or treatment wherein the disorder is a respiratory disease selected from asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), acute lung injury and acute respiratory distress 5 syndrome (ARDS). Preferred is the use of the compounds of the invention for the prevention of the aforesaid disorders. Equally preferred is the use of the compounds of the invention for the treatment of the aforesaid disorders. 10 Generally speaking, compounds which are JAK inhibitors may be useful in the treatment of many disorders associated with JAK enzyme mechanisms. In one embodiment, the disorder that can be treated by the compound of the present invention is selected from the group consisting of asthma, chronic obstructive pulmonary disease (COPD) and interstitial lung disease such as idiopathic pulmonary fibrosis (IPF), acute lung injury and 15 acute respiratory distress syndrome (ARDS). In a further embodiment, the disorder is selected from asthma and chronic obstructive pulmonary disease (COPD). The methods of treatment of the invention comprise administering an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof to a patient in need thereof. 20 As used herein, " effective amount" in reference to a compound of formula (I) or a pharmaceutically acceptable salt thereof or other pharmaceutically-active agent means an amount of the compound sufficient to treat the patient's condition but low enough to avoid serious side effects and it can nevertheless be routinely determined by the skilled artisan. The compounds of formula (I) or pharmaceutically acceptable salts thereof may be administered once or according to 25 a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. Typical daily dosages may vary depending upon the particular route of administration chosen. The invention also provides pharmaceutical compositions of compounds of formula (I) in admixture with one or more pharmaceutically acceptable carrier or excipient, for example those 30 described in Remington’s Pharmaceutical Sciences Handbook, XVII Ed., Mack Pub., N.Y., U.S.A. The present invention is also directed to use of the compounds of the invention and their pharmaceutical compositions for various route of administration. Administration of the compounds of the invention and their pharmaceutical compositions may be accomplished according to patient needs, for example, orally, nasally, parenterally 35 (subcutaneously, intravenously, intramuscularly, intrasternally and by infusion), by inhalation, 42 rectally, vaginally, topically, locally, transdermally, and by ocular administration. Various solid oral dosage forms can be used for administering compounds of the invention including such solid forms as tablets, gelcaps, capsules, caplets, granules, lozenges and bulk powders. The compounds of the present invention can be administered alone or combined with 5 various pharmaceutically acceptable carriers, diluents (such as sucrose, mannitol, lactose, starches) and known excipients, including suspending agents, solubilizers, buffering agents, binders, disintegrants, preservatives, colorants, flavorants, lubricants and the like. Time release capsules, tablets and gels are also advantageous. Various liquid oral dosage forms can also be used for administering compounds of the 10 invention, including aqueous and non-aqueous solutions, emulsions, suspensions, syrups, and elixirs. Such dosage forms can also contain suitable known inert diluents such as water and suitable known excipients such as preservatives, wetting agents, sweeteners, flavorants, as well as agents for emulsifying and/or suspending the compounds of the invention. The compounds of the present invention may be formulated as injectable composition, for example to be injected intravenously, 15 in the form of an isotonic sterile solution. Other preparations are also possible. Suppositories for rectal administration of the compounds of the invention can be prepared by mixing the compound with a suitable excipient such as cocoa butter, salicylates and polyethylene glycols. Formulations for vaginal administration can be in the form of cream, gel, paste, foam, or 20 spray formula containing, in addition to the active ingredient, such as suitable carriers, are also known. For topical administration the pharmaceutical composition can be in the form of creams, ointments, liniments, lotions, emulsions, suspensions, gels, solutions, pastes, powders, sprays, and drops suitable for administration to the skin, eye, ear or nose. Topical administration may also 25 involve transdermal administration via means such as transdermal patches. Compounds of the invention may exhibit profile suitable for oral route administration. Optimization of drugs for oral delivery needs certain characteristics that allow orally administered compound to be absorbed by GI (gastrointestinal) tract and to be poorly cleared in order to give a good bioavailability (F%), thus to maintain a sufficient concentration in plasma and target tissues 30 for a time adequate to sustain pharmacological effect. To enhance oral bioavailability, one or more features of the compounds need to be optimized such as, and not limited to, maximizing membrane permeability and reducing metabolic hot spots (optimizing in-vitro clearance). For the treatment of the diseases of the respiratory tract, the compounds according to the invention, as above said, may also preferably be administered by inhalation. 35 Some preferred compounds of the invention exhibit profile suitable for inhalatory route 43 administration. Drugs optimized for inhaled delivery require certain characteristics that allow the compound, when administered to the lung to maintain a sufficient local concentration (lung retention) to exert a pharmacological effect of the desired duration, with minimal drug absorption in the GI tract for 5 the swallowed fraction and in general non-relevant levels in unwanted compartments (i.e. plasma). For this purpose, one or more features of a compounds were optimized such as, and not limited to, membrane permeability, dissolution rate and the compound’s basicity to enhance its binding to the phospholipid-rich lung tissue or through lysosomal trapping. In some embodiments, compounds of invention show one or more of the features above in the range desirable for an inhaled 10 compound. An inhaled JAKi should preferably possess additional properties that may further limit the systemic exposure after inhalation. A way to limit systemic exposure, after local administration, might be to design soft-drug that means the introduction of specific moieties, like for example cyclic esters or lactones, in the present invention, which favour a controlled rapid systemic metabolism (in the liver and/or in the 15 blood) into predicted functionally less active, metabolites compared to parent compound. For this purpose, one way resulted in the optimization of 'suitably designed lactones derivatives' which can be substrates of liver and/or blood esterases that may be beneficial for achieving an enhanced clearance in-vivo. Thus, preferred compounds of the invention showed one or more of the following properties: 20 high biochemical activity on target, strong functional activity (like cell based activity) and a rapid clearance in representative assays (stability in liver microsomes and/or hepatocytes, plasma stability) so that they have potential for giving compounds with an improved safety. For the treatment of the diseases of the respiratory tract, the compounds according to the invention, as above said, may be administered by inhalation. 25 Inhalable preparations include inhalable powders, propellant-containing metering aerosols or propellant-free inhalable formulations and may be administered through a suitable inhalation device which may be respectively selected from dry powder inhaler, pressurized metered dosed inhaler, or a nebulizer. For administration as a dry powder, single- or multi-dose inhalers known from the prior art 30 may be utilized. In that case the powder may be filled in gelatine, plastic or other capsules, cartridges or blister packs or in a reservoir. A diluent or carrier, e.g. lactose or any other additive suitable for improving the respirable fraction may be added to the powdered compounds of the invention. Inhalation aerosols containing propellant gas such as hydrofluoroalkanes may contain the 35 compounds of the invention either in solution or in dispersed form. The propellant-driven 44 formulations may also contain other ingredients such as co-solvents, stabilizers and optionally other excipients. The propellant-free inhalable formulations comprising the compounds of the invention may be in the form of solutions or suspensions in an aqueous, alcoholic or hydroalcoholic medium and 5 they may be delivered by jet or ultrasonic nebulizers known from the prior art or by soft-mist nebulizers such as Respimat £ ,a registered trademark of Boehringer Ingelheim Pharmaceuticals (Wachtel, H., Kattenbeck, S., Dunne, S. et al. Pulm Ther (2017) 3: 19. The compounds of the invention, regardless of the route of administration, can be administered as the sole active agent or in combination (i.e. as co-therapeutic agents administered 10 in fixed dose combination or in combined therapy of separately formulated active ingredients) with other pharmaceutical active ingredients. The compounds of the invention can be administered as the sole active agent or in combination with other pharmaceutical active ingredients including those currently used in the treatment of respiratory disorders, and known to the skilled person, such as beta2-agonists, 15 antimuscarinic agents, corticosteroids, mitogen-activated kinases (P38 MAP kinases) inhibitors, PI3K inhibitors (phosphoinositide 3-kinases), nuclear factor kappa-B kinase subunit beta inhibitors (IKK2), Rho kinase inhibitors (ROCKi), human neutrophil elastase (HNE inhibitors, phosphodiesterase 4 (PDE4) inhibitors, leukotriene modulators, non-steroidal anti-inflammatory agents (NSAIDs) and mucus regulators). 20 The invention is also directed to a kit comprising the pharmaceutical compositions of compounds of the invention alone or in combination with or in admixture with one or more pharmaceutically acceptable carriers and/or excipients and a device which may be a single- or multi-dose dry powder inhaler, a metered dose inhaler or a nebulizer. The dosages of the compounds of the invention depend upon a variety of factors including 25 the particular disease to be treated, the severity of the symptoms, the route of administration, the frequency of the dosage interval, the particular compound utilized, the efficacy, toxicology profile, and pharmacokinetic profile of the compound. A pharmaceutical composition comprising a compound of the invention suitable to be administered by inhalation is in various respirable forms, such as inhalable powders (DPI), 30 propellant-containing metering aerosols (PMDI) or propellant-free inhalable formulations (e.g. UDV). The invention is also directed to a device comprising the pharmaceutical composition comprising a compound according to the invention, which may be a single- or multi-dose dry powder inhaler, a metered dose inhaler and a nebulizer particularly soft mist nebulizer. 35 The following examples illustrate the invention in more detail. 45 The features of the invention will become apparent in the course of the following descriptions of exemplary embodiments which are given for illustration of the invention and are not intended to be limiting thereof. PREPARATION OF INTERMEDIATES AND EXAMPLES 5 General Experimental details Chemical Names of the compounds were generated with Structure To Name Enterprise 10.0 Cambridge Software or latest. When reactions are conducted at a temperature above solvent boiling point, it is intended a reaction caried out in a closed vessel by conventional heating or by microwave heating. 10 Purification by 'chromatography' or 'flash chromatography' refers to purification using a Biotage SP1, or Interchim puriFlash purification system, or Biotage Isolera Four purification system equipped with Biotage Dalton 2000 mass detector, or equivalent MPLC using a pre-packed polypropylene column containing stationary phase (cartridge). Where products were purified using a Si cartridge, this refers to an Interchim (or equivalent) pre-packed polypropylene column 15 containing unbounded activated silica with spherical particles with average size of 15 ^m or Isolute® (or equivalent) pre-packed polypropylene column containing unbounded activated silica with irregular particles with average size of 50 ^m. Fractions containing the desired product (identified by TLC and/or LCMS analysis) were pooled and concentrated in vacuo. Purification by 'reverse phase chromatography' or 'reverse phase flash flash chromatography' refers to 20 purification on MPLC instrument with with C18 functionalized silica cartridges, as Biotage Sfar C18 or equivalents. Where an SCX-2 cartridge was used, ‘SCX-2 cartridge’ refers to a Bond Elut® pre-packed polypropylene column containing a non-end-capped propylsulphonic acid functionalised silica strong cation exchange sorbent. Where preparative HPLC-MDAP was used for purification (MDAP: mass directed automatic purification) fractions containing the desired 25 product were pooled and the solvent removed by evaporation or alternatively lyophilised. NMR Methods NMR spectra were obtained on a Bruker Avance III 600 (5 mm RT inverse probe head), Bruker DRX 500, Bruker Avance AV 400 (5 mm RT direct probehead) or Bruker DPX 300 spectrometers using standard Bruker pulse sequences. Alternatively, NMR spectra were recorded 30 with Varian MR-400 Mhz spectrometer operating at 400 Mhz or a Varian Unity Inova 400 spectrometer with a 5 mm inverse detection triple resonance probe operating at 400 MHz. DMSO- d 6 or CDCl 3 were used as solvents. Chemical shifts are given in relative to internal standard tetramethylsilane or solvent residual peak. All experiments were recorded at 298 K, unless stated 46 differently. Coupling constants, (J values) are given in hertz (Hz) and multiplicities are reported using the following abbreviation: s=singlet, d=doublet, t=triplet, q=quartet, m=multiplet, br=broad, nd=not determined. LCMS Methods 5 Method 1 Acquity UPLC coupled with SQD mass spectrometer; Column: Acquity UPLC BEH C 18 (50mm x 2.1mm i.d., 1.7^m packing diameter), mobile phase A: 0.1% formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; Gradient – Time Flow (mL/min) A % B% 000 09 97 3 Column MS conditions: 10 Ionisation Mode: alternate-scan Positive and Negative Electrospray (ES + /ES-), Scan Range: 100 to 1000 AMU. Method 2 Acquity UPLC coupled with SQD mass spectrometer; Column: Acquity UPLC BEH C18 (50mm x 2.1mm i.d., 1.7^m packing diameter), mobile phase A: 10 mM aqueous solution of 15 ammonium bicarbonate (adjusted to pH 10 with ammonia), mobile phase B: acetonitrile; Gradient-Time Flow (mL/min) A % B% 000 09 97 3 Column MS conditions: Ionisation Mode: alternate-scan Positive and Negative Electrospray (ES + /ES-), Scan Range: 100 to 1000 AMU. Method 3 20 Acquity UPLC coupled with SQD mass spectrometer; Column: Acquity UPLC BEH C18 (50mm x 2.1mm i.d., 1.7^m packing diameter), mobile phase A: 0.1% v/v formic acid in water, mobile phase B: 0.1% v/v formic acid in acetonitrile; 47 Gradient-Time Flow (mL/min) A % B% 000 06 97 3 Colum MS conditions: Ionisation Mode: alternate-scan Positive and Negative Electrospray (ES + /ES-), Scan Range: 100 to 1500 AMU. Method 4 5 AGILENT LC 1260 Infinity with SFC and Agilent 6540 UHD Accurate-Mass Q-TOF LC/MS; Column: Acquity UPLC BEH C18 (100mm x 2.1mm i.d., 1.7^m packing diameter), mobile phase A: 0.05 % aqueous ammonia, mobile phase B: acetonitrile; Gradient-Time Flow (mL/min) A % B% 000 05 97 3 Column MS conditions: Ionisation Mode: alternate-scan Positive and Negative Electrospray (ES + /ES-), Scan Range: 100 10 to 1000 AMU. Method 5 Shimadzu LCMS-2020 Single Quadrupole Liquid Chromatograph Mass Spectrometer; Column: Acquity UPLC BEH C18 (100mm x 2.1mm i.d., 1.7^m packing diameter), mobile phase A: 0.1 % formic acid in water, mobile phase B: 0.1% formic acid in acetonitrile; Gradient-Time Flow (mL/min) A % B% 010 05 80 20 15 Column itions: Ionisation Mode: alternate-scan Positive and Negative Electrospray (ES + /ES-), Scan Range: 100 to 1000 AMU. 48 HPLC-MDAP Method 1 Agilent 1290 Infinity II Purification System; Column: Waters XBridge® (C18, 100 mm x 19 mm i.d., 5 ^m), mobile phase A: 0.1% (v/v) formic acid in water, mobile phase B: acetonitrile; Gradient – Time Flow (mL/min) A % B% 5 Abbrev AIBN=Azobisisobutyronitrile; aq.=aqueous; Boc 2 O=Di- tert-butyl dicarbonate; CDI=carbonyl diimidatzole; DABAL-Me3=Bis(trimethylaluminum)-1,4- diazabicyclo[2.2.2]octane adduct; DBU=1,8-Diazabicyclo[5.4.0]undec-7-ene; DCC=Dicyclohexylcarbodiimine; DCE=1,2-Dichloroethane; DCM=Dichloromethane;10 DIPEA=N,N-Diisopropylethylamine; DMAP=4-dimethylaminopyridine; DMCHDA=trans-N,Nƍ- Dimethylcyclohexane-1,2-diamine; DMF=N,N-Dimethylformamide; DMSO=Dimethylsulfoxide; EtOAc=Ethyl acetate; HATU=(1-[Bis(dimethylamino)methylene]- 1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate); LCMS=Liquid chromatography-mass spectrometry; LiHMDS=Lithium bis(trimethylsilyl)amide; NBS=N- 15 Bromosuccinimide; ^W=microwave; 1 H-NMR=Proton nuclear magnetic resonance; RM=Reaction mixture; Rt = Retention time; RT = Room temperature; sat.=saturated; T3P ® =Propylphosphonic anhydride; TEA=Triethylamine; TFA – Trifluoroacetic acid; THF=Tetrahydrofuran; Xphos–Pd-G3-(2-Dicyclohexylphosphino-2ƍ,4ƍ,6ƍ-triisopro pyl-1,1ƍ- biphenyl)[2-(2ƍ-amino-1,1ƍ-biphenyl)]palladium(II) methanesulfonate. 20 In the procedures that follow, some of the starting materials are identified through an “Intermediate” or “Example” number with indications on Step number. This is provided merely for assistance to the skilled chemist. A “similar” or “analogous” procedure means that such a procedure may involve minor variations, for example reaction temperature, reagent/solvent amount, reaction time, work-up 25 conditions and/or chromatographic purification conditions. 49 The stereochemistry of the compounds in the Examples, where indicated, has been assigned on the assumption that absolute configuration at resolved stereogenic centres of starting materials is maintained throughout any subsequent reaction conditions. Unless otherwise stated, where absolute configuration (R) or (S) is reported in the compound 5 name, ee% has to be considered equal or greater than 90%. PREPARATION OF INTERMEDIATES Intermediate 1 Step 1 10 4-(Difluoromethoxy)-2-nitroph eno ( ntermediate 1-1) A solution of 4-(difluoromethoxy)phenol (385 μL, 3.12 mmol) in DCE (3 mL) was cooled at 0°C. Concentrated nitric acid 63% w/w (456 ^L) was added dropwise and RM stirred at 0 °C for 3 h. RM was poured on ice/water mix and extracted with DCE (10 mL). Organic layer was separated, dried and evaporated in vacuo to afford the desired product (630 mg) that was used in 15 the next steps without further purification. LCMS (Method 1): Rt = 0.40 min 1H-NMR (500 MHz, DMSO-d 6 ) į: 11.09 (bs, 1H), 7.73 (d, J=2.9 Hz, 1H), 7.42 (dd, J=9.0, 2.9 Hz, 1H), 7.18 (t, J=73.1 Hz, 1H), 7.17 (d, J=9.0 Hz, 1H). Step 2 20 1-(2-Bromoethoxy)-4-(diflu oromet oxy)- -n trobenzene (Intermediate 1-2) A mixture of intermediate 1-1 (630 mg, 3.07 mmol) and K 2 CO 3 (1.27 g, 9.21 mmol) in DMF (10 mL) and 1,2-dibromoethane (265 ^L, 3.07 mmol) was stirred at 60 °C overnight. After cooling to RT, RM was diluted with water and extracted twice with EtOAc. Combined organics were 25 washed with sat aq. NaHCO 3 , dried and concentrated in vacuo. The crude material was purified by flash chromatography on Si cartridge by eluting with 0-20 % DCM/MeOH/NH4OH (90:9:0.5) in DCM to give the title product (466 mg). LCMS (Method 1): Rt = 1.15 min, No MS data 50 1H-NMR (500 MHz, DMSO-d6) į: 7.80 (d, J=2.4 Hz, 1H), 7.44-7.52 (m, 2H), 7.23 (t, J=73.1 Hz, 1H), 4.50 (t, J=5.2 Hz, 2H), 3.79 (t, J=5.4 Hz, 2H). Step 3 5 6-(Difluoromethoxy)-3,4-dihyd ro- -benzo[b][1,4]oxazine (Intermediate 1-3) Intermediate 1-2 (466 mg, 0.78 mmol) was dissolved in ethanol (42 mL) and heated at 80 °C, then a solution of NH4Cl (166 mg, 3.10 mmol) in water (5mL) was added and followed by the addition of iron (434 mg, 7.80 mmol). RM was stirred at 80 °C for 3 h. After cooling to RT, RM was diluted with water and extracted twice with DCM. Combined organic layers were dried over 10 Na 2 SO 4 and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-20 % EtOAc in cyclohexane to afford the title product (88 mg). LCMS (Method 1): Rt = 0.99 min, ES + m/z 202.1 [M+H] + . Step 4 15 7-Bromo-6-(difluoromethoxy)-3 , -d ydro- H-benzo[b][1,4]oxazine (Intermediate 1) To a 0°C cooled solution of intermediate 1-3 (44.0 mg, 0.22 mmol) in EtOAc (1 mL), NBS (38.9 mg, 0.22 mmol) was added and RM stirred at RT for 5 h. RM was diluted with EtOAc and quenched with sat. aq. NaHCO 3 . The layers were separated and aqueous layer additionally 20 extracted with EtOAc. Combined organic layers were washed with sat .aq. NaCl, passed through a phase separator and concentrated in vacuo to afford the desired product (63mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.14 min, ES + m/z 280.0/282.1 [M+H] + . Intermediate 2 25 Step 1 51 7-Bromo-6-methoxy-3,4-dihydro-2H-benzo[b][1,4]oxazine (Intermediate 2-1) A solution of 6-methoxy-3,4-dihydro-2H-1,4-benzoxazine (3.0 g, 18.20 mmol) in EtOAc (30.0 mL) was cooled to 0°C. 1,3-Dibromo-5,5-dimethyl-imidazolidine-2,4-dione (2.6 g, 9.08 mmol) was added portion-wise during 15 minutes. RM was stirred for additional 30 min at 0°C 5 and quenched with an aqueous K 2 CO 3 solution (10% w/w; 60 mL). The organic layer was separated, washed with sat. aq. NaCl and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-30 % EtOAc in cyclohexane to afford the title product (3.5 g). LCMS (Method 1): Rt = 0.97 min, ES + m/z 243.9/245.9 [M+H] + . 10 Step 2 tert-Butyl 7-bromo-6-methox y- , - y ro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 2) THF (15 mL) was added to a mixture of intermediate 2-1 (1.4 g, 5.74 mmol), DMAP (840.9 15 mg, 6.88 mmol) and Boc 2 O (2.80 g, 13.19 mmol), then RM stirred at RT overnight. RM was partitioned between EtOAc (50 mL) / water (30 mL). The organic layer was washed with 2M aq. citric acid (2x20 mL), sat. aq. NaCl (20 mL) and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-10 % EtOAc in cyclohexane to afford the title compound (1.26 g). 20 LCMS (Method 1): Rt = 1.27 min 1H-NMR (300 MHz, CDCl 3 ) į: 7.56 (brs, 1H), 7.05 (s, 1H), 4.14-4.18 (m, 2H), 3.82 (s, 3H), 3.78-3.82 (m, 2H), 1.53 (s, 9H). Intermediate 3 Step 1 25 tert-Butyl 7-methoxy-3,4-dihydroqu no ne- ( H)-carboxylate (Intermediate 3-1) A solution of 7-methoxy-1,2,3,4-tetrahydroquinoline (500 mg, 3.06 mmol), DMAP (449 mg, 3.68 mmol) and Boc 2 O (1.54 g, 7.05 mmol) in THF (10 mL) was stirred at RT overnight. A further 52 equivalent of Boc2O was added and stirring proceed at RT. RM was partitioned between EtOAc (50 mL) and water (30 mL). The organic layer was washed with aq.2M citric acid (2x15 mL), sat. aq. NaCl (20 mL) and solvent evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-10 % EtOAc in cyclohexane to afford the title 5 product (228 mg). LCMS (Method 1): Rt = 1.35 min 1H-NMR (300 MHz, DMSO-d 6 ) į: 7.18 (d, J=2.5 Hz, 1H), 6.98 (d, J=8.6 Hz, 1H), 6.57 (dd, J=8.5, 2.6 Hz, 1H), 3.69 (s, 3H), 3.56-3.60 (m, 2H), 2.63 (t, J=6.46 Hz, 2H), 1.74-1.82 (m, 2H), 1.45 (s, 9H). 10 Step 2 tert-Butyl 6-bromo-7-methoxy-3 , -d ydroquinoline-1(2H)-carboxylate (Intermediate 3) A solution of intermediate 3-1 (228 mg, 0.86 mmol) in EtOAc (10 mL) was cooled to 0°C. 15 1,3-dibromo-5,5-dimethyl-imidazolidine-2,4-dione (124 mg, 43.3 mmol) was added portionwise over 15 min. RM was stirred at 0 °C for 20 min, then quenched with 10 % (w/w) aq. K2CO3 (20 mL). The organic layer was separated, washed with sat. aq. NaCl (20 mL) and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-15 % EtOAc in cyclohexane to afford the title product (240 mg). 20 LCMS (Method 1): Rt = 1.46 min, ES + m/z 285.9/287.9 [M+H] + . Intermediate 4 7-Bromo-8-methoxy-1,3,4,5-tetr a ydro- -benzo[b]azepin-2-one (Intermediate 4) 1,3-dibromo-5,5-dimethyl-imidazolidine-2,4-dione (123 mg, 0.43 mmol) was added 25 portionwise over 10 minutes to a solution of 8-methoxy-1,3,4,5-tetrahydro-1-benzazepin-2-one (165 mg, 0.86 mmol) in EtOAc (4 mL) at 0°C and. RM was stirred at RT overnight. RM was quenched with sat. aq. NaHCO3 and extracted twice with EtOAc. Combined organic layers were 53 washed with sat. aq. NaCl, passed through a phase separator and concentrated in vacuo to give the title product (230 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 0.93, ES + m/z 270.0/272.0 [M+H] + . Intermediate 5 5 Step 1 tert-Butyl 7-methoxy-4-oxo-3, 4-dihydroquinoline-1(2H)-carboxylate (Intermediate 5- 1) A solution of 7-methoxy-2,3-dihydro-1H-quinolin-4 e (100.0 mg, 0.56 mmol), DMAP 10 (20.7 mg, 0.17 mmol) and BoC2O (147.8 mg, 0.68 mmol) in THF (1.0 mL) was stirred at RT for 2 h. RM was partitioned between EtOAc and water. The organic layer was washed with sat. aq. NaCl, passed through a phase separator and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-10 % EtOAc in cyclohexane to afford the title product (138 mg). 15 LCMS (Method 1): Rt = 1.11 min 1H-NMR (500 MHz, DMSO-d6) į: 7.79 (d, J=8.6 Hz, 1H), 7.27 (d, J=2.0 Hz, 1H), 6.79 (dd, J=8.4, 2.0 Hz, 1H), 4.07 (t, J=6.1 Hz, 2H), 3.82 (s, 3H), 2.66 (t, J=6.1 Hz, 2H), 1.51 (s, 9H). Step 2 20 tert-Butyl 6-bromo-7-m et oxy- -oxo-3,4-dihydroquinoline-1(2H)-carboxylate (Intermediate 5) A solution of intermediate 5-1 (100.0 mg, 0.36 mmol) in EtOAc (2.0 mL s cooled to 0°C prior the addition og NBS (64.2 mg, 0.36 mmol). RM was warmed to RT and heated at 65 °C for 30 h. After cooling to RT, RM was diluted sat. aq. NaHCO 3 and extracted twice with EtOAc. 25 Combined organic layers were washed with sat. aq. NaCl and passed through a phase separator. Solvent was removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-40 % EtOAc in cyclohexane to give the title product (153 mg). 54 LCMS (Method 1): Rt = 1.30 min. 1H-NMR (500 MHz, DMSO-d6) į: 7.93 (s, 1H), 7.46 (s, 1H), 4.08 (t, J=6.0 Hz, 2H), 3.93 (s, 3H), 2.69 (t, J=6.0 Hz, 2H), 1.52 (s, 9H). Intermediate 6 5 Step 1 tert-Butyl 6-methoxyindoline-1 -carboxyate (Intermediate 6-1) A solution of 6-methoxyindoline (1.0 g, 6.70 mmol), BoC2O (1.76 mg, 8.04 mmol) and DMAP (819 mg, 6.70 mmol) in THF (20 mL) was stirred at RT for 6 h. RM was partitioned 10 between EtOAc (50 mL) and water (30 mL) and organic layer washed with aq. 10% w/w citric acid (2x50 mL), sat. aq. NaHCO 3 (50 mL) and sat. aq. NaCl (50 mL). Organic phase was evaporated in vacuo to dryness. The residue was purified by flash chromatography on Si cartridge by eluting with 0-15 % EtOAc in cyclohexane to afford the title product (1.16 g). LCMS (Method 1): Rt = 1.25 min, ES + m/z 250.0 [M+H] + . 15 Step 2 tert-Butyl 5-bromo-6-methoxyi ndo ne- -carboxylate (Intermediate 6) 1,3-dibromo-5,5-dimethyl-imidazolidine-2,4-dione (0.66 g, 2.3 mmol) was added portion- wise over 15 minutes to a solution of intermediate 6-1 (1.16 g, 4.65 mmol) in EtOAc (70 mL) at 20 0°C. RM was stirred at RT for 2 h, then quenched with 10% w/w aq. K2CO3 (75 mL). Organic layer was separated, washed with sat. aq. NaCl (35 mL) and concentrated in vacuo to afford the desired product (1.52 g) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.40 min, ES + m/z 228.0/230.0 [M+H] + . Intermediate 7 25 Step 1 55 tert-butyl 6-metho xy- ,3-d ydro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 7-1) The title compound was prepared in a similar manner of intermediate 3-step 1 starting from 5 6-methoxy-3,4-dihydro-2H-1,4-benzoxazine. LCMS (Method 5): Rt = 2.5 min 1H-NMR (300 MHz, DMSO-d 6 ) į: 7.47 – 7.38 (m, 1H), 6.77 (d, J = 8.9 Hz, 1H), 6.57 (dd, J = 8.9, 3.0 Hz, 1H), 4.18 – 4.09 (m, 2H), 3.80 – 3.72 (m, 2H), 3.68 (s, 3H), 1.50 (s, 9H). Step 2 10 tert-butyl 7-iodo-6-metho xy- ,3-d ydro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 7) Intermediate 7-1 (5.9g, 22.1 mmol) was dissolved in DMF (60mL), then N- Iodohydroxysuccinimide (12.7g, 111 mmol) added and RM stirred at 40°C overnight. RM was 15 quenched in cold water and extracted with EtOAc. Combined organic layers were evaporated to dryness and the residue purified by chromatography on silica gel by grdient eluition from 1:1 to 3:2 DCM-Hexane to afford the title compound (7.38g). LCMS (Method 1): Rt = 2.8 min 1H-NMR (300 MHz, DMSO-d 6 ) į: 7.50 (s, 1H), 7.24 (s, 1H), 4.14 (dd, J = 5.3, 3.8 Hz, 2H), 20 3.80 – 3.75 (m, 2H), 3.74 (s, 3H), 1.51 (s, 9H). Intermediate 8 Step 1 5-Methoxybenzo[d]oxazol-2(3H )-one (Intermediate 8-1) 56 CDI (2.40 g, 14.8 mmol) was added portionwise over 1 h to a refluxing solution of 2-amino- 4-methoxy-phenol (1.00 g, 14.4 mmol) in THF (20 mL) and RM refluxed for further 1 h. RM was cooled to RT and solvent removed in vacuo. The residue was dissolved in EtOAc (100 mL), washed with water (3x30mL) and sat. aq. NaCl (2x30mL). Organic layer was dried over MgSO4 5 and solvent removed in vacuo. The crude material was purified by flash chromatography on Si cartridge by eluting with 0-80 % DCM/MeOH/NH4OH (90:5:0.5) in DCM to afford the title product (700 mg). LCMS (Method 1): Rt = 0.70 min, ES + m/z 166.1 [M+H] + . Step 2 10 6-Iodo-5-methoxybenzo[d]oxaz o- (3 )-one ( ntermediate 8-2) A solution of intermediate 8-1 (400 m 2.42 mmol) and NIS (817 mg, 3.63 mmol) in DMF (5 mL) was stirred at RT for 1 h. reaction was quenched with water (30 mL) and extracted with EtOAc (3x20mL). Combined organic layers were washed with water (3x20mL) and sat. aq. NaCl 15 (2x10mL). Organic phase was evaporated in vacuo to afford the title product (626 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 0.93 min, ES + m/z 291.9 [M+H] + . Step 3 20 6-Iodo-5-methoxy-3-((2-(trimet y s y )et oxy)methyl)benzo[d]oxazol-2(3H)-one (Intermediate 8) A solution intermediate 8-2 (300 mg, 1.03 mmol) in DMF (9 mL) was added to a 0 °C cooled suspension of NaH (60.0 % in mineral oil, 41.2 mg, 1.03 mmol) in DMF (5 mL). RM was stirred for 30 min reaching RT. After cooling RM to 0 °C, 2-(chloromethoxy)ethyl-trimethyl-silane (192 25 ^L, 1.03 mmol) was added dropwise. RM was stirred for 2 h reaching RT. RM was quenched with sat. aq. NaHCO3 (15 mM) and extracted with EtOAc (3x15 mL). Combined organic layers were washed with water (20mL), sat. aq. NaCl (20mL), dried over MgSO 4 and concentrated in vacuo 57 to afford the desired product (300 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.49 min. 1H-NMR (300 MHz, DMSO-d6) į: 7.78 (s, 1H), 7.13 (s, 1H), 5.25 (s, 2H), 3.83 (s, 3H), 3.61 5 (t, J=8.2 Hz, 2H), 0.86 (t, J=8.0 Hz, 2H), -0.06 (s, 9H). Intermediate 9 Step 1 1-Bromo-4,5-bis(bromomethyl )- -me oxy enzene (Intermediate 9-1) 10 1-Bromo-2-methoxy-4,5-dimethyl-benzene (200 mg, 0.93 mmol) was dissolved in Į,Į,Į- trifluorotoluene (10.0 mL). NBS (331 mg, 1.86 mmol) and AIBN (30.5 mg, 0.19 mmol) were added and RM was stirred at 90°C for 3 h. After cooling to RT, RM was diluted with EtOAc, washed with water, and sat. aq. NaCl, dried over phase separator and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-30 % EtOAc 15 in cyclohexane to afford the title product (246 mg). LCMS (Method 1): Rt = 1.34 min. 1H-NMR (300 MHz, DMSO-d 6 ) į: 7.74 (s, 1H), 7.26 (s, 1H), 4.80 (s, 2H), 4.79 (s, 2H), 3.87 (s, 3H). Step 2 20 5-Bromo-6-methoxy-2-(methyls u ony) so ndoline (Intermediate 9) NaH (60.0 %, 56.6 mg, 1.42 mmol) was added to a solution of methanesulfonamide (135 mg, 1.42 mmol) in DMF (10.0 mL) and RM stirred for 1 h. Intermediate 9-1 (240 mg, 0.64 mmol) in DMF (10.0 mL) was added dropwise and the resulting solution stirred at 50 °C for 3 h and at 25 RT overnight. RM was quenched with water and extracted with EtOAc. Combined organic layers were washed with water and sat. aq. NaCl, passed through a phase separator and concentrated in 58 vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-30 % EtOAc in cyclohexane to afford the title product (160 mg). LCMS (Method 1): Rt = 1.34 min. 1H-NMR (300 MHz, DMSO-d6) į: 7.56 (s, 1H), 7.12 (s, 1H), 4.57 (s, 2H), 4.55 (s, 2H), 3.83 5 (s, 3H), 2.96 (s, 3H). Intermediate 10 Step 1 5-Bromo-2-( tert-butylthio)-4-me t oxybenza dehyde (Intermediate 10-1) 10 A mixture of 5-bromo-2-fluoro-4-methoxy-benzaldehyde (1.0 g, 4.29 mmol), 2-methyl-2- propanethiol (726 μL, 6.44 mmol) and K 2 CO 3 (949 mg, 6.87 mmol) in DMF (15 mL) was stirred at 80 °C for 3 h. RM was cooled to RT and poured in water (30 mL). The formed precipitate was collected by filtration, washed with water (2x10 mL), and dried at 45 °C for 3 h to afford the title product (1 g). 15 LCMS (Method 1): Rt = 1.40 min. 1H-NMR (300 MHz, CDCl3) į: 10.6 (s, 1H), 8.20 (s, 1H), 7.08 (s, 1H), 3.98 (s, 3H), 1.32 (s, 9H). Step 2 20 5-Bromo-2-( tert-butylthio)-4-m et oxybenza dehyde oxime (Intermediate 10-2) A mixture of intermediate 10-1 (0.9 g, 0.3 mmol), hydroxylamine chloride (413 mg, 0.59 mmol) and sodium acetate (584 mg, 7.12 mmol) in EtOH (9 mL) was stirred at RT for 1 h. RM was filtered to remove residual solids and filtrate evaporated in vacuo. The residue was dissolved in EtOAc (25 mL), washed with water (15 mL), sat. aq. NaCl (15 mL), dried over MgSO4 and 25 solvent removed in vacuo to afford the desired product (997 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.26 min. 1H-NMR (300 MHz, DMSO-d6) į: 11.34 (s, 1H), 8.90 (s, 1H), 7.99 (s, 1H), 7.16 (s, 1H), 3.89 (s, 3H), 1.24 (s, 9H). 59 Step 3 5-Bromo-6-methoxybenzo[d]isoth azoe ( ntermediate 10) A solution of intermediate 10-2 (0.87 g, 2.73 mmol) and TsOH (52 mg, 0.27 mmol) in i- 5 PrOH (8 mL) was stirred at 100 °C overnight, then in a ^W reactor at 110 °C for 30 min. A second equivalent of TsOH was added and RM further stirred in ^W reactor at 120 °C for 1h for two cycle. After cooling to RT, the formed precipitate was collected by filtration and dried at 45 °C. to afford the desired product (280 mg). LCMS (Method 1): Rt = 1.10 min. 10 1 H-NMR (500 MHz, DMSO-d6) į: 8.92 (s, 1H), 8.46 (s, 1H), 7.93 (s, 1H), 3.96 (s, 3H). Intermediate 11 Step 1 Methyl (4-bromo-2-formyl-5- met oxyp eny )carbamate (Intermediate 11-1) 15 Methyl chloroformate (170 ^L, 2.2 mmol) was added dropwise to a 0 o C cooled solution of 2-amino-5-bromo-4-methoxy-benzaldehyde (200 mg, 0.87 mmol) and DIPEA (0.61 mL, 3.5 mmol) in DCM (5 mL). RM was stirred at 40 °C for 2 days. RM was evaporated in vacuo and the residue purified by flash chromatography on Si cartridge, by eluting with 0-25 % DCM/MeOH (9:1) in DCM, to afford the title product (190 mg). 20 LCMS (Method 1): Rt = 1.16 min, ES + m/z 288.0/290.0 [M+H] + Step 2 6-Bromo-7-methoxy-3-methyl-3,4-dihydroquinazolin-2(1H)-one (Intermediate 11) A solution of intermediate 11-1 (185 mg, 0.64 mmol) and methanolic methyl amine (9.8 M, 25 97 ^L, 0.95 mmol) in AcOH (2mL) was stirred in a ^W reactor at 130 °C for 15 min. RM was cooled to RT and formic acid (1.2 mL, 32 mmol) added. RM was stirred at 150 C° for 30 min. RM was evaporated in vacuo and the residue purified by flash chromatography on a Si cartridge, by 60 eluting with 0-50 % DCM/MeOH/NH4OH (90:5:0.5) in DCM, to afforfd the title product (154 mg). LCMS (Method 1): Rt = 0.88 min, ES + m/z 271.1/273. [M+H] + Intermediate 12 5 3-(Tributylstannyl)imidazo[1, -b]pyr daz ne ( ntermediate 12) A solution of 3-bromoimidazo[1,2-b]pyridazine (400 mg, 2.0 mmol) in THF (5 mL) was cooled to 0 °C then i-PrMgCl LiCl complex (1.3 M solution in THF, 4.7 mL, 6.1 mmol) added dropwise over 5 min. RM was stirred at 0°C for 15 min then tri-n-butyltin chloride (685 μL, 2.5 10 mmol) added dropwise, and RM further stirred at RT for 30 min. RM was cooled in an ice bath and quenched with water (5 mL), added with aq. sat. NH4Cl (10 mL) and extracted with EtOAc (15 mL). Organic layer was washed with aq. sat. NaCl (10 mL), dried over MgSO4 and evaporated in vacuo. The residue was purified by chromatography on neutral allumina (Al 2 O 3 ) by eluting with mixtures of cyclohexane and EtOAc to afford the title product (285 mg). 15 LCMS (Method 2): Rt = 1.82 min, ES + m/z (most abundant isotope) 410.0 [M+H] + Intermediate 13 3-((2-Hydroxyethyl)thio)dihyd ro uran- (3 )-one (Intermediate 13) A solution of 3-bromotetrahydrofuran-2-one (822 ^L, 9.09 mmol), 2-sulfanylethanol (2.55 20 mL, 36.4 mmol), and DIPEA (1.74 mL, 10.0 mmol) in THF (4 mL) was stirred at RT overnight. RM was evaporated in vacuo and the residue purified by flash chromatography on Si cartridge by eluting with 0-50 % EtOAc in DCM to afford the title product (1.43 g). 1H-NMR (300 MHz, CDCl3) į: 4.42 (dt, J=8.9, 7.4 Hz, 1H), 4.31 (ddd, J=9.1, 8.0, 4.8 Hz, 1H), 3.82-3.87 (m, 2H), 3.63 (dd, J=8.7, 5.5 Hz, 1H), 3.07 (ddd, J=14.3, 5.9, 4.8, 1H), 2.75-2.78 25 (m, 1H), 2.67 (ddd, J=15.9, 13.8, 8.0, 1H), 2.08-2.16 (m, 1H). Intermediate 14 3-((2-Hydroxyethyl)(methyl)a m no)d ydro uran-2(3H)-one (Intermediate 14) 61 A solution of 3-bromotetrahydrofuran-2-one (782 ^L, 8.65 mmol), 2-(methylamino)ethanol (1.39 mL, 17.3 mmol) in THF (4 mL) was stirred at RT overnight. Solvent was removed in vacuo and the residue purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH (9:1) in DCM to afford the title product (295 mg). 5 1 H-NMR (300 MHz, CDCl 3 ) į: 4.37 (dt, J=9.1, 2.1 Hz, 1H), 4.18 (ddd, J=10.2, 9.6, 6.6 Hz, 1H), 3.73 (dd, J=10.9, 8.9 Hz, 1H), 3.63 (t, J=5.3, 2H), 2.76-2.81 (m, 2H), 2.42 (s, 3H), 2.11-2.39 (m, 1H). Intermediate 15 Step 1 10 Ethyl 1-(2-((tert-butoxyc arbony )am no)et y )p peridine-4-carboxylate (Intermediate 15-1) A suspension of tert-butyl N-(2-bromoethyl)carbamate (3 g, 13.0 mmol), ethyl piperidine- 4-carboxylate (2.1 mL, 13.0 mmol) and K2CO3 (3.7 g, 27 mmol) in DMF (30.0 mL) was stirred at 15 65 °C for 24 h. After cooling to RT, RM was diluted with water (80 mL) and extracted with EtOAc (3 x 80 mL). Combined organics were washed with sat. aq. NaHCO3 (80 mL) and sat. aq. NaCl (50 mL). Organic solvent was removed in vacuo and residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM:MeOH (93:7) in DCM to afford the title product (3.24 g). 20 LCMS (Method 2): Rt = 1.05 min, ES + m/z 301.3 [M+H] + . Step 2 Ethyl 1-(2-aminoethyl)pip er d ne- -carboxy ate ( ntermediate 15) A solution of intermediate 15-1 (1.77 g, 5.9 mmol) and TFA (8.8 mL, 118 mmol) in DCM 25 (14 mL) was stirred at RT overnight. Volatiles were removed in vacuo and crude product passed through an SCX cartridge, washed with EtOH (500 mL) and eluted with 15% aq. NH3 in EtOH (100 mL) to afford the title product (1.16 g). 62 1H-NMR (300 MHz, CDCl3) į: 4.13 (q, J=6.9 Hz, 2H), 2.84-2.88 (m, 2H), 2.78 (t, J=6.2 Hz, 2H), 2.39 (t, J=6.2 Hz, 2H), 2.24-230 (m, 1H), 2.02 (t, J=11.5 Hz, 2H), 1.87-1.91 (m, 2H), 1.71- 1.79 (m, 2H), 1.25 (t, J=6.9 Hz, 3H). Intermediate 16 5 4-(3-Hydroxypropyl)morpho n- -one ( ntermediate 16) A mixture of 3-(2-hydroxyethylamino)propan-1-ol (393 mg, 3.30 mmol), methyl 2- bromoacetate (344 ^L, 3.63 mmol) and K 2 CO 3 (502 mg, 3.63 mmol) in dry acetonitrile (14 mL) was stirred at RT overnight. RM was filtered and concentrated in vacuo. Residue was dissolved in 10 DCM (20 mL) and washed with water (10 mL). Combined organic layers were passed through phase separator and solvent removed in vacuo to afford the desired product (120 mg) that was used in the next synthetic steps without further purification. 1H-NMR (300 MHz, CDCl3) į: 4.37-4.40 (m, 2H), 3.67-3.82 (m, 2H), 3.34 (s, 2H), 3.05 (m, 1H), 2.73 (t, J=5.1 Hz, 2H), 2.62 (t, J=6.2 Hz, 2H), 1.74 (quint, J=5.5 Hz, 2H). 15 Intermediate 17 6-Chloro-3-iodo-1-(tetrahydro - -pyran- -y )-1H-pyrazolo[4,3-c]pyridine (Intermediate 17) Dihydropyran (9.79 mL, 107 mmol) and methanesulfonic acid (464 ^L, 7.16 mmol) were 20 added to 6-chloro-3-iodo-1H-pyrazolo[4,3-c]pyridine (10 g, 35.8 mmol) in DCM (100 mL) and THF (50 mL). RM was stirred at 40 °C for 4 h, then at RT overnight. RM was evaporated to dryness and the residue purified by flash chromatography on a Si cartridge by eluting with 0-40% of EtOAc in cyclohexane to afford the title product (7.5 g). LCMS (Method 2): Rt = 1.22 min, ES + m/z 364.0/366.0 [M+H] + . 25 Intermediate 18a Step 1 63 6-Chloro-N-(2-morpholino et y)- -(tetra ydro- H-pyran-2-yl)-1H-pyrazolo[4,3- c]pyridin-3-amine (Intermediate 18a-1) A degassed mixture of intermediate 17 (900 mg, 2 . mmol), 2-morpholinoethanamine (1.30 5 mL, 12.7 mmol), K 2 CO 3 (2.05 g, 14.9 mmol), proline (85.5 mg, 0.74 mmol) and CuI (94.3 mg, 0.50 mmol) in DMF (10 mL) was stirred under nitrogen at 100°C for 2 h. After cooling to RT, RM was diluted with water (30mL) and extracted with EtOAc (4x15mL). Combined organics were washed with aq. ammonia 10% w/w (2x15 mL), water (2x20mL) and sat. aq. NaCl (40mL). Organic phase was evaporated in vacuo and the residue purified by flash chromatography on Si 10 cartridge by eluting with 0-80 % DCM/MeOH/NH4OH (90:5:0.5) in DCM to afford the title product (750 mg). LCMS (Method 2): Rt = 0.94 min, ES + m/z 366.3/368.3 [M+H] + . Step 2 15 6-Chloro-N-(2-morpholino et y)- -pyrazo o[ ,3-c]pyridin-3-amine (Intermediate 18a) A solution of intermediate 18a-1 (635 mg, 1.88 mmol), TFA (2.66 mL, 34.7 mmol) and triethylsilane (0.9 mL, 5.64 mmol) in DCM (20 mL) was stirred at RT for 2 h. RM was quenched with water (15 mL) and pH adjusted to 9.5. Aqueous phase was further extracted with DCM (3x20 20 mL), and combined organic layers were evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100% DCM/MeOH/NH 4 OH (90:5:0.5) in DCM to give the title product (250 mg). LCMS (Method 1): Rt = 0.42 min, ES + m/z 282.0/283.9 [M+H] + Intermediate 18b 25 Step 1 64 6-Chloro-N-methyl-1-(tetrahyd ro- -pyran-2-yl)-1H-pyrazolo[4,3-c]pyridin-3-amine (Intermediate 18b-1) The title product was prepared on a similar manner to intermediate 18a-1(step 1) starting 5 from intermediate 17 (230 mg, 0.63 mmol) and methylamine hydrochloride (171 mg, 2.5 mmol). DMSO was used as reaction solvent instead of DMF. LCMS (Method 2): Rt = 0.94 min, ES + m/z 267.1/269.1 [M+H] + . Step 2 10 6-chloro-N-methyl-1H-pyrazol o[ ,3-c]pyr d n-3-amine (Intermediate 18b) The title compound was prepared similarly to intermediate 18a (step 2) starting from intermediate 18b-1. LCMS (Method 2): Rt = 0.51 min, ES + m/z 183.1/185.1 [M+H] + Intermediate 19 15 6-(6-Chloro-3-((2-morphol noet y )am no)- -pyrazolo[4,3-c]pyridin-1-yl)-5- methoxy-3-((2-(trimethylsilyl)ethoxy)methyl)benzo[d]oxazol-2 (3H)-one (Intermediate 19) A degassed mixture of intermediate 18a (102 mg, 0.36 mmo , ntermediate 8 (85.0 mg, 0.18 20 mmol), K 2 CO 3 (151 mg, 1.09 mmol), proline (6.27 mg, 0.05 mmol) and CuI (6.9 mg, 36 ^mol) in DMF (2 mL) was stirred under nitrogen at 100°C for 8 h. After cooling to RT, RM was diluted with water (10mL) and extracted with EtOAc (5x5mL). Combined organic layers were washed with aq. Ammonia 10% w/w (2x10 mL), water (2x20mL) and sat. aq. NaCl (30mL). Organic phase 65 was evaporated in vacuo and the residue purified by flash chromatography on Si cartridge by eluting with 0-80 % DCM/MeOH/NH4OH (90:5:0.5) in DCM to afford the title product (15 mg). LCMS (Method 1): Rt = 1.07 min, ES + m/z 575.4/577.4 [M+H] + . Intermediate 20 5 6-Chloro-1-(6-methoxybenz o[d] sot azo -5-y)-N-(2-morpholinoethyl)-1H- pyrazolo[4,3-c]pyridin-3-amine (Intermediate 20) A degassed mixture of intermediate 18a (120 mg, 0.43 mmol), intermediate 10 (156 mg, 0.64 mmol), CuI (40.6 mg, 0.21 mmol), N,N-dimethylglycine (43.9 mg, 0.43 mmol) and K 2 CO 3 10 (118 mg, 0.85 mmol) in DMSO (2.5 mL) was heated overnight at 100 °C under nitrogen. After cooling to RT, RM was diluted with EtOAc (15 mL), washed with water (2x10 mL) and sat. aq. NaCl (10 mL). RM was evaporated in vacuo and the residue purified by flash chromatography on Si cartridge by eluting with 0-80% DCM/MeOH/NH4OH (90:5:0.5) in DCM to give the title product (105 mg). 15 LCMS (Method 1): Rt = 0.99 min, ES + m/z 445.2/447.2 [M+H] + Intermediate 21 Step 1 Methyl 1-(5-bromo-2-methoxy - -n trop eny)-6-chloro-1H-pyrazolo[4,3-c]pyridine-3- 20 carboxylate (Intermediate 21-1) 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid (3.00 g, 15.2 mmol) was added to a solution of 1-bromo-5-fluoro-4-methoxy-2-nitro-benzene (3.80 g, 20.2 mmol) and DBU (5.88 mL, 45.6 mmol) in DMF (48 mL). RM was stirred at RT for 4 h. Iodomethane (3.78 mL, 60.7 mmol) was added, and RM stirred at RT overnight. RM was quenched into water (200 mL), the formed 25 precipitate was collected by filtration and dried. The crude material was triturated in EtOAc and 66 washed with EtOAc to give the title product (5.83 g) that was used in the next synthetic step without further purification. LCMS (Method 1): Rt = 1.08 min, ES + m/z 426.9/428.9/430.9 [M+H] + . Step 2 5 Methyl 6-chloro-1- (5-(( - ydroxyet yl)thio)-2-methoxy-4-nitrophenyl)-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 21-2) A degassed mixture of intermediate 21-1 (5.0 g, 11 mmol), 2-sulfanylethanol (1.13 mL, 12 mmol), DIP . L, 20 mmol), Xantphos (820 mg, 1.4 mmol) and Pd 2 (dba) 3 (330 mg, 0.57 10 mmol) in 1,4-dioxane (100 mL) was stirred at 100°C under argon for 3 h. After cooling to RT, reaction was quenched with water. The formed precipitate was collected by filtration, washed with a small amount of EtOAc and dried to afford the title compound (5.3 g) that was used in the next synthetic step without further purification. LCMS (Method 1): Rt = 1.08 min, ES + m/z 439.0/441.0 [M+H] + . 15 Step 3 Methyl 1-(4-amino-5- (( - ydroxyet yl)thio)-2-methoxyphenyl)-6-chloro-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 21-3) A solution of intermediate 21-2 (5.3 g, 12 mmol) in MeOH (200 mL) was stirred at 80 °C, 20 then followed by the addition of a solution of NH4Cl (6.5 g, 121 mmol) in water (20 mL) and iron powder (6.7 g, 121 mmol). RM was stirred at 80 °C for 4.5 h, then filtered while still hot and the filtrate concentrated in vacuo. The crude material was diluted with sat. aq NaHCO3, the resulting suspension sonicated, and the solids collected by filtration. The solid material was washed several times with water and dried to afford the title product (3.13 g). 25 LCMS (Method 1): Rt = 0.92 min, ES + m/z 409.1/411.1 [M+H] + . 67 Step 4 Methyl 1-(4-amino- 5-(( -c oroet yl)thio)-2-methoxyphenyl)-6-chloro-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 21-4) 5 Intermediate 21-3 (4.4 g, 11 mmol) was mixed with thionyl chloride (12 mL, 165 mmol) at 0 °C. RM was warmed to RT and stirred for 2 h, then volatiles removed in vacuo and the residue taken with sat. aq. NaHCO 3 . The formed slurry was sonicated and solid collected by filtration, washed with water, and dried. The resulting crude was purified by flash chromatography on Si cartridge by eluting with 0-10 % EtOAc in DCM to afford the title product (2.7 g). 10 LCMS (Method 1): Rt = 1.24 min, ES + m/z 427.0/429.0/431.0 [M+H] + . Step 5 Methyl 6-chloro-1-(6-m et oxy-3, -d hydro-2H-benzo[b][1,4]thiazin-7-yl)-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 21-5) 15 A mixture of intermediate 21-4 (2.70 g, 6.32 mmol), K 2 CO 3 (2.62 g, 19 mmol) and NaI (189 mg, 1.26 mmol) in DMF (80 mL) was stirred at 90°C overnight. After cooling to RT, RM was diluted with water (200 mL). The formed precipitate was filtered, washed several times with water and dried. The resulting material was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/Acetonitrile (9:1) in DCM to afford the title product (730 mg). 20 LCMS (Method 1): Rt = 1.13 min, ES + m/z 391.1/393.1 [M+H] + . Step 6 68 6-Chloro-1-(6-methoxy-3,4-dih ydro- -benzo[b][1,4]thiazin-7-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxylic acid (Intermediate 21) Intermediate 21-5 (730 mg, 1.87 mmol) was suspended in THF (15 mL), and added with a 5 solution of LiOH (1.0 M in water, 9 mL, 9 mmol). RM was stirred at 40 °C for 2.5 h. Organic solvent was removed in vacuo and the residue diluted with water. The pH was adjusted to 2.5 using aq.1M HCl. The formed precipitate was filtered, washed several times with water and dried to afford the title compound (698 mg). LCMS (Method 1): Rt = 0.95 min, ES + m/z 377.1/379.0 [M+H] + . 10 Intermediate 22 Step 1 Methyl 6-chloro-1-(2-met oxy-5-(( -met oxy-2-oxoethyl)thio)-4-nitrophenyl)-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 22-1) 15 Intermediate was prepared similarly to intermediate 21-2 starting from intermediate 21-1 and methyl 2-sulfanylacetate. LCMS (Method 1): Rt = 1.18 min, ES + m/z 467.1/469.1 [M+H] + . Step 2 69 Methyl 1-(4-amino-2-methoxy-5-((2-methoxy-2-oxoethyl)thio)phenyl)-6 -chloro-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 22-2) A solution of intermediate 22-1 (265 mg, 0.51 mmol) in ethanol (6.0 mL) was stirred at 80 °C, then added with a solution of NH4Cl (109 mg, 2.0 mmol) in water (3.0 mL) and iron (340 mg, 5 6.1 mmol). RM was stirred at 80 °C for 16h. After cooling to RT, RM was filtered, and the filter further washed with EtOAc. Combined organic layers were evaporated in vacuo to afford the desired product (1.03 g) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.09 min, ES + m/z 437.2/439.1 [M+H] + . Step 3 10 Methyl 6-chloro-1-(6-metho xy-3-oxo-3, -d ydro-2H-benzo[b][1,4]thiazin-7-yl)-1H- pyrazolo[4,3-c]pyridine-3-carboxylate (Intermediate 22-3) TFA (1.3 mL, 17.0 mmol) was added to a suspension of intermediate 22-2 (1.0 g, 1.7 mmol) in DCM (12.0 mL). RM was stirred at RT for 24 h. RM was evaporated in vacuo and the residue 15 purified by flash chromatography on Si cartridge by eluting with 0-60 % DCM/MeOH (20:1) in DCM to give the title product (341.5 mg). LCMS (Method 1): Rt = 0.99 min, ES + m/z 405.2/407.1 [M+H] + . Step 4 20 6-Chloro-1-(6-methoxy-3-oxo- 3, -d ydro- -benzo[b][1,4]thiazin-7-yl)-1H- pyrazolo[4,3-c]pyridine-3-carboxylic acid (Intermediate 22) An aqueous solution of LiOH (1.0 M, 7.5 mL, 7.5 mmol) was added to a suspension of intermediate 22-3 (340 mg, 0.84 mmol) in THF (10 mL). RM was stirred at RT for 2 h. THF was removed in vacuo. The residue was diluted with water and acidified to pH 2.5 by using aq.1M 70 HCl. The formed precipitate was collected by filtration, washed with water and dried to afford the title product (320 mg). LCMS (Method 2): Rt = 0.48 min, ES + m/z 391.0/393.0 [M+H] + . Intermediate 23a 5 Step 1 1-(5-bromo-2-methoxy-4-nitro p eny)-6-c oro-3-iodo-1H-pyrazolo[4,3-c]pyridine (Intermediate 23a-1) A mixture of 6-chloro-3-iodo-1H-pyrazolo[4,3-c]pyridine (1.50 g, 5.37 mmol), 1-bromo-5- 10 fluoro-4-methoxy-2-nitro-benzene (1.34 g, 5.37 mmol) and K2CO3 (2.23 g, 16.1 mmol) in DMF (30 mL) was stirred at 65 °C for 2 h. Reaction was repeated on 17.4 mmol scale and combined with previous after cooling to RT. Combined RM were poured into water (400 mL), and the precipitate collected by filtration. Collected solids were washed thoroughly with water and azeotroped with toluene to afford title product (11.1 g). 15 LCMS (Method 2): Rt = 1.36 min, ES + m/z 508.9/510.9/512.8 [M+H] + . Step 2 1-(5-Bromo-2-methoxy-4-n trop eny )-6-c oro-N-(2-(4-methylpiperazin-1-yl)ethyl)- 1H-pyrazolo[4,3-c]pyridin-3-amine (Intermediate 23a-2) 20 A degassed mixture of intermediate 23a-1 (800 mg, 1.57 mmol), 2-(4-methylpiperazin-1- y )ethanamine (366 ^L, 2.83 mmol), proline (90.4 mg, 0.79 mmol), K 2 CO 3 (1.3 g, 9.42 mmol) and CuI (89.7 mg, 0.47 mmol) in dry DMSO (6 mL) was stirred at 75 °C for 5 h. A second equivalent of CuI and proline was added. After cooling to RT, solvent was removed in vacuo. The residue was partitioned between EtOAc and water. Aqueous layer was further extracted with AcOEt. 25 Combined organic layers were washed with sat. aq. NaCl (20 mL) and concentrated in vacuo. The 71 residue was purified twice by flash chromatography on Si cartridge by eluting with DCM/MeOH/NH4OH (90:15:1.5) to afford the title product (243 mg). LCMS (Method 2): Rt = 1.08 min, ES + m/z 524.1/526.1/528.1 [M+H] + . Step 3 5 Methyl 2-((5-(6-chloro-3 -(( -( -me y p peraz n-1-yl)ethyl)amino)-1H-pyrazolo[4,3- c]pyridin-1-yl)-4-methoxy-2-nitrophenyl)thio)acetate (Intermediate 23a-3) A solution of intermediate 23a-2 (240 mg, 0.46 mmol), DIPEA (199 ^L, 1.1 mmol) and methyl 2-sulfanylacetate (102 μL, 1.1 mmol) in dry acetonitrile (14 mL) was stirred at 120 °C 10 overnight. The solvent was removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH/NH 4 OH (90:15:1.5) in DCM to afford the title product (87 mg). LCMS (Method 2): Rt = 0.97 min, ES + m/z 550.2/552.2 [M+H] + . Step 4 15 7-(6-Chloro-3-((2-(4-methy p peraz n- -y)et y)amino)-1H-pyrazolo[4,3-c]pyridin-1- yl)-6-methoxy-2H-benzo[b][1,4]thiazin-3(4H)-one (Intermediate 23a) A solution of intermediate 23a-3 (85 mg, 0.15 mmol) in EtOH (20 mL) was stirred at 80 °C, then added with a solution of NH 4 Cl (70 mg, 1.3 mmol) in water (5 mL) and iron (0.16 g, 2.9 20 mmol). RM was stirred at 80 °C for 4.5 h. A second equivalent of iron and NH4Cl were added and RM further stirred at 80 °C for 1 h. RM was quenched with aq.2 N HCl (1 mL) and stirred at 80 °C for 2 h. After cooling to RT, RM was filtered and filter cake washed with EtOH. The filtrate was evaporated in vacuo. The residue was treated with sat. aq. NaHCO 3 (10 mL) and extracted with EtOAc (2x20 mL). Combined organic layers were washed with sat. aq. NaCl (10 mL), dried 25 over Na 2 SO 4 , and solvent evaporated in vacuo. The residue was purified by flash chromatography 72 on Si cartridge by eluting with 0-100 % DCM/MeOH/NH4OH (90:15:1.5) in DCM to afford the title product (40 mg). LCMS (Method 2): Rt = 0.80 min, ES + m/z 488.2/490.2 [M+H] + . Intermediate 23b 5 Step 1 N 1 -(1-(5-Bromo-2-methoxy- -n trop eny )-6-c oro-1H-pyrazolo[4,3-c]pyridin-3-yl)- N 2 ,N 2 -dimethylethane-1,2-diamine (Intermediate 23b-1) The title intermediate was prepared on a similar manner to intermediate 23a-2 starting from 10 mediate 23a-1 and N',N'-dimethylethane-1,2-diamine. LCMS (Method 2): Rt = 1.18 min, ES + m/z 469.1/471.1/473.1 [M+H] + Step 2 Methyl 2-((5-(6-chloro-3-((2 -(d met yam no)et yl)amino)-1H-pyrazolo[4,3-c]pyridin- 15 1-yl)-4-methoxy-2-nitrophenyl)thio)acetate (Intermediate 23b-2) The title intermediate was prepared on a similar manner to intermediate 23a-3 starting from intermediate 23b-1 and methyl 2-sulfanylacetate to give the title product (122 mg). LCMS (Method 2): Rt = 1.12 min, ES + m/z 495.1/497.1 [M+H] + Step 3 20 73 7-(6-Chloro-3-((2-(dimethylamino)ethyl)amino)-1H-pyrazolo[4, 3-c]pyridin-1-yl)-6- methoxy-2H-benzo[b][1,4]thiazin-3(4H)-one (Intermediate 23b) The title compound was prepared on a similar manner to intermediate 23a-4 starting from intermediate 23b-2. 5 LCMS (Method 2): Rt = 1.04 min, ES + m/z 433.2/434.2 [M+H] + Intermediate 24 1-(4-(tert-Butoxycarbonyl)-6 -met oxy-3, -d ydro-2H-benzo[b][1,4]oxazin-7-yl)-6- chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid (Intermediate 24) 10 Method 1 A degassed mixture of 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid (400 mg, 2.02 mmol), intermediate 2 (733 mg, 2.13 mmol), Cs2CO3 (2.31 g, 7.09 mmol) and thiophene-2- carbonyloxycopper (278 mg, 1.46 mmol) in anhydrous DMSO (7 mL) was stirred under nitrogen at 110 °C overnight. After cooling to RT, RM was filtered. The filtrate was diluted with MeCN 15 (50 mL) thus the formed precipitate collected by filtration and triturated with MeOH. The crude material was purified by flash chromatography on Si cartridge by eluting with 0-100 % MeOH in EtOAc to afford the title product (150 mg). Method 2 A degassed mixture of 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid (9.4 g, 47.520 mmol), intermediate 7 (18.6 g, 47.5 mmol), Cs2CO3 (54 g, 166.3 mmol) and thiophene-2- carbonyloxycopper (9 g, 47.5 mmol) in DMSO (100 mL) was stirred under argon at 110 °C for 24h. RM was quenched in water and extracted with DCM. Combined organic layers was washed with aq 10% w/w citric acid, aq. sat. NaCl and evaporated to dryness. The residue was purified by cromatography on silica gel by eluting with DCM to DCM (1% v/v MeOH+1%AcOH v/v). The 25 material thus obatined was triturated in ethyl ether to afford the title compound (3.14g). LCMS (Method 1): Rt = 1.22 min, ES + m/z 461.2/463.2 [M+H] + . Intermediate 25 74 6-Chloro-1-(6-methoxy-3,4-dihy dro- -benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3- c]pyridine-3-carboxylic acid (Intermediate 25) The title intermediate was prepared in a similar manner to intermediate 24 method 1 starting 5 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid and intermediate 2-1. LCMS (Method 1): Rt = 0.86 min, ES + m/z 360.9/362.8 [M+H] + . Intermediate 26 6-Chloro-1-(7-methoxy-2-oxo- , ,3, -tetra ydroquinolin-6-yl)-1H-pyrazolo[4,3- 10 c]pyridine-3-carboxylic acid (Intermediate 26) The title intermediate was prepared on similar manner like intermediate 24 method 1 starting from 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid and 6-bromo-7-methoxy-3,4- dihydro-1H-quinolin-2-one. LCMS (Method 1): Rt = 0.77 min, ES + m/z 373.1/375.1 [M+H] + . 15 Intermediate 27a 1-(1-(tert-Butoxycarbonyl)-7 -met oxy- , ,3, -tetrahydroquinolin-6-yl)-6-chloro-1H- pyrazolo[4,3-c]pyridine-3-carboxylic acid (Intermediate 27a) A mixture of intermediate 3 (1.04 g, 3.04 mmol), 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3- 20 carboxylic acid (300 mg, 1.52 mmol), thiophene-2-carbonyloxycopper (290 mg, 1.52 mmol) and 75 Cs2CO3 (1.48 g, 4.56 mmol) in DMSO (7 mL) was stirred for 72 h at 100 °C under nitrogen. After cooling to RT, RM was added dropwise to water under stirring. The pH was adjusted to 4 (with aq. HCl 1M) and the formed precipitate filtered, washed with water, and dried. The crude material was purified by flash chromatography on Si cartridge by eluting with 0-50% DCM:MeOH:HCO2H 5 (90:10:0.3) in DCM to afford the title product (330 mg). LCMS (Method 1): Rt = 1.24 min, ES + m/z 458.9/460.9 [M+H] + . Intermediate 27b tert-Butyl 6-(6-chloro -3-(met oxycarbonyl)-1H-pyrazolo[4,3-c]pyridin-1-yl)-7- 10 methoxy-3,4-dihydroquinoline-1(2H)-carboxylate (Intermediate 27b) A mixture of intermediate 3 (346 m 101 mmol) 6-Chloro-1H-pyrazolo[4,3-c]pyridine-3- carboxylic acid (100 mg, 0.506 mmol), thiophene-2-carbonyloxycopper (97 mg, 0.5 mmol) and Cs2CO3 (0.49 g, 1.52 mmol) in DMSO (2.3 mL) was stirred for 72 h at 100 °C under nitrogen. After cooling to RT, iodomethane (126 ^l, 2.02 mmol) was added and the mixture stirred at RT 15 for further 2h more. RM was quenched with water and extracted three times with DCM. Combined organic layers were dried over Na 2 SO 4 , filtered, and evaporated to dryness. The residue was purified by flash chromatography on Si cartridge by eluting with 0-10 % cyclohexane/EtOAc (1:1) in cyclohexane to afford the title product (118 mg). LCMS (Method 1): Rt = 1.41 min, ES + m/z 473.0/475.0 [M+H] + . 20 Intermediate 28 tert-Butyl 7-(3-amino -6-c oro- -pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3- dihydro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 28) A degassed mixture of intermediate 2 (408 mg, 1.19 mmol), 6-chloro-1H-pyrazolo[4,3- 25 c]pyridin-3-amine (200 mg, 1.19 mmol), Cs2CO3 (980 mg, 3.01 mmol), DMCHDA (93.5 μL, 0.59 76 mmol) and CuI (113 mg, 0.59 mmol) in DMSO (4.9 mL) was stirred under argon at 110 °C overnight. After cooling to RT, RM was quenched with sat. aq. NaHCO3 (50 mL) and extracted with EtOAc (5x50 mL). Combined organic layers were washed with sat. aq. NaCl, dried over Na2SO4 and solvent removed in vacuo. The crude residue was purified by flash chromatography 5 on Si cartridge by eluting with 0-50% DCM/MeOH/NH 4 OH (90:5:0.5) in DCM to afford the title product (282 mg). LCMS (Method 1): Rt = 1.15 min, ES + m/z 431.9/433.9 [M+H] + . Intermediate 29 10 6-(3-Amino-6-chloro-1H-pyra zo o[ , -c]pyr n-1-yl)-7-methoxy-3,4-dihydroquinolin- 2(1H)-one (Intermediate 29) A degassed solution of 6-bromo-7-methoxy-3,4-dihydro-1H-quinolin-2-one (304 mg, 1.19 mmol), 6-chloro-1H-pyrazolo[4,3-c]pyridin-3-amine (100 mg, 0.59 mmol), K2CO3 (246 mg, 1.78 mmol), N,N-dimethylglycine (93.5 μL, 0.59 mmol) and CuI (56.5 mg, 0.30 mmol) in DMSO (3.6 15 mL) was stirred under argon at 110 °C overnight. After cooling to RT, RM was quenched with sat. aq. NaHCO3 (50 mL) and extracted with EtOAc (5x50 mL). Combined organic layers were washed with sat. aq. NaCl, dried over Na 2 SO 4 and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH/NH4OH (90:5:1) in DCM to give the title product (68.6 mg). 20 LCMS (Method 2): Rt = 0.68 min, ES + m/z 344.1/345.9 [M+H] + . Intermediate 30 6-(6-Chloro-3-(methylamino)- -pyrazo o[ ,3-c]pyridin-1-yl)-7-methoxy-3,4- dihydroquinolin-2(1H)-one (Intermediate 30) 77 The title compound was prepared similarly to intermediate 29 starting from 6-bromo-7- methoxy-3,4-dihydroquinolin-2(1H)-one and intermediate 18b. LCMS (Method 2): Rt = 0.79 min, ES + m/z 358.1/360.1 [M+H] + . Intermediate 31a 5 Step 1 tert-Butyl 7-(3-(( tert-buto xycarbony )am no)-6-chloro-1H-pyrazolo[4,3-c]pyridin-1- yl)-6-methoxy-2,3 -dihydro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 31a-1) 10 LiHMDS (1.30 M in THF, 463 μL, 0.60 mmol) was added to a solution of intermediate 28 (130 mg, 0.30 mmol) in THF (7 mL) at 0°C under nitrogen. RM was stirred for 15 min. A solution of Boc 2 O (131 mg, 0.60 mmol) in THF (1 mL) was added dropwise and RM stirred at RT for 3 h. RM was diluted with water and extracted with EtOAc. Combined organic layers were washed with sat. aq. NaCl, dried over Na 2 SO 4 and solvent removed in vacuo. The residue was purified by flash 15 chromatography on Si cartridge by eluting with 0-50 % EtOAc in cyclohexane. The isolated product was dissolved in methanol (1 mL), added with K 2 CO 3 (76.5 mg, 0.55 mmol) and stirred at RT overnight. RM was diluted with sat. aq. NaHCO3 and extracted with DCM. Combined organic layers were washed with sat. aq. NaCl, dried over Na 2 SO 4 and solvent removed in vacuo to afford the title product (107 mg) that was used in the next synthetic step without further 20 purification. LCMS (Method 2): Rt = 1.49 min, ES + m/z 532.3/534.3 [M+H] + . Step 2 78 tert-Butyl 7-(3-(( tert-butoxycarbonyl)(2-(dimethylamino)ethyl)amino)-6-chloro- 1H- pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[ b][1,4]oxazine-4-carboxylate (Intermediate 31a) A solution of intermediate 31a-1 (85.0 mg, 0.16 mmol) in ry DMF (1.0 mL) was cooled 5 under argon at 0 °C. NaI (23.9 mg, 0.16 mmol) and NaH, (60 % dispersion in mineral oil, 19.2 mg, 0.48 mmol) were added and RM stirred at 0 °C for 30 min.2-bromo-N,N-dimethyl-ethanamine hydrobromide (55.8 mg, 0.24 mmol) was added and stirring proceeded at RT for 60 h. RMs was diluted with water and extracted with EtOAc (3x10 mL). Combined organics were passed through a phase separator cartridge and solvent was evaporated in vacuo. The residue was purified by flash 10 chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH/NH4OH (90:4:1) in DCM to afford the title product. LCMS (Method 2): Rt = 1.54 min, ES + m/z 603.3/605.3 [M+H] + . Intermediate 31b 15 tert-Butyl 7-(3-(( tert-b u oxycar ony )(me hyl)amino)-6-chloro-1H-pyrazolo[4,3- c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazin e-4-carboxylate (Intermediate 31b) The title compound was prepared similarly to intermediate 31a starting from intermediate 31a-1 and iodomethane. 20 LCMS (Method 2): Rt = 1.54 min, ES + m/z 546.2/548.2 [M+H] + . Intermediate 32a tert-Butyl 7-(6-chlor o-3-met y- -pyrazoo[4,3-c]pyridin-1-yl)-6-methoxy-2,3- dihydro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 32a) 79 To a degassed mixture of intermediate 2 (657 mg, 1.9 mmol), 6-chloro-3-methyl-1H- pyrazolo[4,3-c]pyridine (200 mg, 1.2 mmol), CuI (114 mg, 0.6 mmol), N,N-dimethylglycine (123 mg, 1.2 mmol) and K 2 CO 3 (330 mg, 2.4 mmol) was added DMSO (5 mL). RM was stirred at 100 °C overnight. After cooling to RT, RM was diluted with EtOAc (25 mL) and washed with 15% 5 w/w aq. NH 4 OH (3x15 mL) and sat. aq. NaCl (15 mL). The organic layers were dried over Na 2 SO 4 and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-15 % EtOAc in DCM to afford the title product (312 mg). LCMS (Method 2): Rt = 1.32 min, ES + m/z 431.1 /433.1 [M+H] + . Intermediates 32b to 32i 10 The following intermediates were prepared in a similar manner to intermediate 32a from the indicated starting materials. It is additionally stated if base, ligand/catalyst, solvent and/or temperature were varied. Intermediate Structure Starting material LCMS t t t 80 t t t t t 81 6-(6-Chloro-3-methyl-1H-py razoo[ ,3-c]pyr d n-1-yl)-7-methoxy-2,3- dihydroquinolin-4(1H)-one (Intermediate 32j) TFA (173 ^L, 2.26 mmol) was added to a solution of intermediate 32h (40.0 mg, 0.09 mmol) 5 in DCM (1.0 mL). RM was stirred at RT overnight, then concentrated in vacuo. The residue was loaded on SCX cartridge, washed with methanol, and eluted with 2M methanolic ammonia to afford the title product (32 mg). LCMS (Method 1): Rt = 0.87 min, ES + m/z 342.9/344.9 [M+H] + Intermediate 33 10 Step 1 1-(5-Bromo-2-methoxy-4-ni trop eny )-6-c oro-3-methyl-1H-pyrazolo[4,3-c]pyridine (Intermediate 33-1) A mixture of 6-chloro-3-methyl-1H-pyrazolo[4,3-c]pyridine (300 mg, 1.79 mmol), 1- 15 bromo-5-fluoro-4-methoxy-2-nitro-benzene (448 g, 1.79 mmol) and K 2 CO 3 (742 g, 5.37 mmol) was suspended in DMF (6.0 mL) and stirred at 80 °C for 1 h. After cooling to RT, RM was diluted with water (80 mL). The formed precipitate was collected by filtration, washed with water (3x30 mL) and dried to afford the title product (620 mg) that was used in the next synthetic steps without further purification. 20 LCMS (Method 1): Rt = 1.26 min, ES + m/z 397.1/399.1/401.1 [M+H] + Step 2 82 Methyl 2-((5-(6-chloro-3-methyl-1H-pyrazolo[4,3-c]pyridin-1-yl)-4-m ethoxy-2- nitrophenyl)thio)acetate (Intermediate 33-2) A mixture of intermediate 33-1 (350 mg, 0.88 mmol), methyl 2-sulfanylacetate (118 μL, 1.3 mmol) and DIPEA (230 μL, 1.3 mmol) in dry acetonitrile (15.0 mL) was stirred at 120 °C in a μM 5 reactor for 3 h. A new equivalent of methyl 2-sulfanylacetate (16 μL, 0.17 mmol) was added and RM was further stirred at 120 °C for 2 h. After cooling to RT, solvent was removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-80 % (DCM/MeOH (9:1) in DCM to afford the desired product (170 mg). LCMS (Method 1): Rt = 1.16 min, ES + m/z 423.2/425.2 [M+H] + 10 Step 3 O H 7-(6-Chloro-3-methyl-1H-pyr azoo[ ,3-c]pyr d n-1-yl)-6-methoxy-2H- benzo[b][1,4]thiazin-3(4H)-one (Intermediate 33-3) To a stirring solution of intermediate 33-2 (160 mg, 0.38 mmol) in e nol (5 mL) w at 80 15 °C, a solution of NH 4 Cl (81 mg, 1.5 mmol) in water (2 mL) and iron (0.25 g, 4.5 mmol) were added. RM was stirred at 80 °C for 6h. After cooling to RT, RM was diluted with EtOAc (20 mL), filtered through a bed of diacematous earth and washed thoroughly with EtOAc. The filtrate was concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-35 % DCM/MeOH (95:5) in DCM to afford the title product (50 mg). 20 LCMS (Method 1): Rt = 0.96 min, ES + m/z 361.2/363.2 [M+H] + Step 4 7-(6-Chloro-3-methyl-1H-pyr azoo[ ,3-c]pyr d n-1-yl)-6-methoxy-3,4-dihydro-2H- benzo[b][1,4]thiazine (Intermediate 33-4) 83 To a 0 °C cooled solution of BH3.THF (1.0 M in THF, 166.3 μL, 0.16 mmol), a solution of intermediate 33-3 (30.00 mg, 0.08 mmol) in THF (1 mL) was added and RM stirred at 25 °C for 2h. A second equivalent of BH 3 .THF was added and RM further stirred for 2h. RM was quenched with sat aq. NH4Cl (20mL) and extracted with EtOAc (2x10 mL). Combined organic layers were 5 washed with sat. aq. NaHCO 3 (3x10 mL) and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH (98:2) in DCM to afford the title product (18mg). LCMS (Method 1): Rt = 1.11 min, ES + m/z 347.2/349.2 [M+H] + Step 5 10 7-(6-Chloro-3-methyl-1H-py razoo[ ,3-c]pyr d n-1-yl)-6-methoxy-3,4-dihydro-2H- benzo[b][1,4]thiazine 1,1-dioxide (Intermediate 33) To a 0 o C cooled solution of intermediate 33-4 (25.0 mg, 67.8 ^mol) in dry DCM (1 mL), m-CPBA (70.0 %, 33.4 mg, 0.136 mmol) was added and RM stirred at 0°C for 1 h. After warming 15 to RT, RM was diluted with DCM and washed with sat. aq. NaHCO3. Organic phase was evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-60 % DCM/MeOH/NH4OH (90:5:0.5) in DCM to give the title product (13 mg). LCMS (Method 1): Rt = 0.82 min, ES + m/z 378.9/380.8 [M+H] + Intermediate 34 20 Step 1 Diethyl 2-(5-(6-chlor o-3-met y - -pyrazolo[4,3-c]pyridin-1-yl)-4-methoxy-2- nitrophenyl)malonate (Intermediate 34-1) To a mixture of NaH (60 % suspension in mineral oil, 80 mg, 2.0 mmol) in DMSO (4.0 mL), 25 diethyl malonate (186 ^L, 1.3 mmol) was slowly added and RM stirred at 65 °C. After 20 min, 84 intermediate 33-1 (195 mg, 0.49 mmol) was added and RM was at 100 °C for 1 h. After cooling to RT, RM was poured on ice/water and was extracted with EtOAc (4x10 mL). Combined organic layers were washed with water (10x10 mL) and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-45 % DCM/MeOH (98:2) in DCM to afford 5 the title product (75 mg). LCMS (Method 1): Rt = 1.26 min, ES + m/z 477.2/479.2 [M+H] + Step 2 5-(6-chloro-3-methyl-1H-pyr azoo[ ,3-c]pyr d n-1-yl)-6-methoxyindolin-2-one 10 (Intermediate 34-2) To a stirring solution of intermediate 34-1 (75 mg, 0.16 mmol) in acetic acid (4.0 mL) at 90 oC, iron (97 mg, 1.7 mmol) was added in portions and RM stirred at 90 °C for 80 min. After cooling to RT, RM was filtered, filtrate was diluted with water (5 mL) and extracted EtOAc (3x10 mL). Combined organics were washed with sat. aq. NaHCO 3 (10 mL), dried over Na 2 SO 4 and 15 concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-75 % DCM/MeOH/NH 4 OH (90:5:0.5) in DCM to afford the title product (21.4 mg). LCMS (Method 1): Rt = 0.82 min, ES + m/z 329.2/331.2 [M+H] + Step 3 20 5-(6-Chloro-3-methyl-1H -pyrazoo[ ,3-c]pyr d n- -yl)-6-methoxy-1-((2- (trimethylsilyl)ethoxy)methyl)indolin-2-one (Intermediate 34-3) To a stirring suspension of NaH (60.0 %, 14.9 mg, 0.37 mmol) in dry DMF (0.3 mL), a solution of intermediate 34-2 (180 mg, 0.37 mmol) in DMF (3 mL) was added and RM stirred for 30 min reaching RT. 2-(chloromethoxy)ethyl-trimethyl-silane (139 ^L, 0.75 mmol) was added 85 dropwise at 0 °C. RM was stirred overnight at RT, then quenched with sat. aq. NaHCO3 (15 mL), and extracted with EtOAc (3x15 mL). Combined organic layers were washed with water (20mL), sat. aq. NaCl (20mL) and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-40 % EtOAc/DCM (1:9) in DCM to afford the 5 title product (100 mg). LCMS (Method 2): Rt = 1.35 min, ES + m/z 459.2/461.2 [M+H] + Step 4 5'-(6-Chloro-3-methyl-1H-p yrazoo[ , -c]pyr n-1-yl)-6'-methoxy-1'-((2- 10 (trimethylsilyl)ethoxy)methyl)spiro[cyclopropane-1,3'-indoli n]-2'-one (Intermediate 34) To a stirring mixture of intermediate 34-3 (60.0 mg, 0.12 mmol) diphenyl(vinyl)sulfonium trifluoromethanesulfonate (51.7 mg, 0.14 mmol) and zinc trifluoromethanesulfonate (43.2 mg, 0.12 mmol) in dry DMF (3 mL), DBU (53.3 ^L, 0.36 mmol) was added. RM was stirred at RT for 2 h. RM was quenched with sat. aq. NH4Cl (10 mL) and extracted with EtOAc (3x15 mL). 15 Combined organic layers were washed with water (2x10 mL), dried over Na 2 SO 4 and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-25 % EtOAc/DCM (1:9) in DCM to afford the title product (48 mg). LCMS (Method 2): Rt = 1.44 min, ES + m/z 485.2/487.2 [M+H] + Intermediate 35 20 Step 1 1-(Allyloxy)-5-chloro-4-methox y- -n trobenzene (Intermediate 35-1) Suspension of 5-chloro-4-methoxy-2-nitro-phenol (1.00 g, 4.91 mmol), 3-bromoprop-1-ene (509 ^L, 5.89 mmol) and K2CO3 (1018 mg, 7.37 mmol) in acetonitrile (10mL) was stirred at 75 25 °C overnight. RM was cooled to RT and diluted with water and DCM. Layers were separated and aqueous layer was washed with DCM. Combined organics were dried and concentrated to get a crude product that was used in the next synthetic step without further purification (1.08 g). 86 LCMS (Method 1): Rt = 1.18 min 1H-NMR (300 MHz, CDCl3) į: 7.48 (s, 1H), 7.13 (s, 1H), 5.95-6.07 (m, 1H), 5.47 (dq, J=17.3, 1.3 Hz, 1H), 5.33 (dq, J=10.4, 1.3 Hz, 1H), 4.62 (dt, J=5.0, 1.3 Hz, 2H), 3.90 (s, 3H). Step 2 5 1-(5-(Allyloxy)-2-methoxy-4-n rop eny )- -c oro-3-methyl-1H-pyrazolo[4,3- c]pyridine (Intermediate 35-2) A suspension of 6-chloro-3-methyl-1H-pyrazolo[4,3-c]pyridine (700 mg, 4.18 mmol), intermediate 35-1 (1.08 g, 4.42 mmol) and K 2 CO 3 (1.73 g, 12.5 mmol) in DMSO (10.0 mL) was 10 stirred at 120 °C for 5 h. RM was cooled to RT and diluted with water. The formed precipitate was collected by filtration, washed with water, and dried to afford the desired product (950 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.19 min, ES + m/z 374.9/376.9 [M+H] + . Step 3 15 5-(6-Chloro-3-methyl-1H-pyr azoo[ ,3-c]pyr d n-1-yl)-4-methoxy-2-nitrophenol (Intermediate 35-3) To a degassed mixture of intermediate 35-2 (500 mg, 1.33 mmol) and K2CO3 (553 mg, 4.00 mmol) in MeOH (15 mL), Pd(PPh 3 ) 4 (50.0 mg, 0.04 mmol) was added and RM stirred at 60°C 20 for 30 min. After cooling to RT, RM was concentrated and suspended in water, acidified to pH 3 and extracted with DCM. Combined organic layers were dried over Na 2 SO 4 , and evaporated to dryness. The residue was triturated in DCM/MeOH to afford the desired product (200 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.09 min, ES + m/z 335.1/337.1 [M+H] + . 25 Step 4 87 2-Amino-5-(6-chloro-3-methy - -pyrazoo[ ,3-c]pyridin-1-yl)-4-methoxyphenol (Intermediate 35-4) To a warm refluxing solution of intermediate 35-3 (500 mg, 1.49 mmol) in MeOH (30 mL), 5 a solution of NH4Cl (320 mg, 5.98 mmol) in water (5 mL) and iron (834 mg, 14.9 mmol) were added. RM was heated at reflux temperature for 2 days. After cooling to RT, RM was filtered through a pad of diacematous earth. Filtrate was evaporated in vacuo and the residue taken in DCM, washed with water. The aqueous phase was back-extracted with DCM (3 × 50 mL) and combined organic layers were washed with sat. aq. NaCl and passed through a phase separator. 10 Organic phase was evaporated in vacuo to afford the desired product (126.9 mg) that was used in the next steps without further purification. LCMS (Method 1): Rt = 0.72 min, ES + m/z 305.0/306.9 [M+H] + . Step 5 15 2-Bromo-N-(4-(6-chloro-3- met y - -pyrazoo[ ,3-c]pyridin-1-yl)-2-hydroxy-5- methoxyphenyl)-2,2-difluoroacetamide (Intermediate 35-5) A solution of intermediate 35-4 (125 mg, 0.34 mmol), DABAL-Me3 (131 mg, 0.51 mmol) and methyl 2-bromo-2,2-difluoro-acetate (71 mg, 0.37 mmol) in THF (3 mL) was stirred under nitrogen and microwave irradiation at 130 °C for 15 min. After cooling to RT, reaction was 20 cautiously quenched with 4M HCl in 1,4-dioxane (2 mL). The resulting mixture was poured into sat. aq. NaHCO3 and extracted with DCM. Organic phase was evaporated in vacuo and the residue purified by flash chromatography on Si cartridge by eluting with 0-90 % DCM/MeOH (20:1) in DCM to afford the title product (25 mg). LCMS (Method 1): Rt = 1.09 min, ES + m/z 461.0/463.1/465.0 [M+H] + . 88 Step 6 7-(6-Chloro-3-methyl-1H-pyr azoo[ ,3-c]pyr d n-1-yl)-2,2-difluoro-6-methoxy-2H- benzo[b][1,4]oxazin-3(4H)-one (Intermediate 35) 5 A solution of intermediate 35-5 (24 mg, 52 ^mol) and DBU (13.0 ^L, 0.10 mmol) in THF (15 mL) was stirred a 0 °C for 16 h. After cooling to RT, RM was evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH (20:1) in DCM to afford the title product (12.4 mg). LCMS (Method 1): Rt = 1.09 min, ES + m/z 381.1/383.1 [M+H] + . 10 Intermediate 36 Step 1 3-(5-Chloro-4-methoxy-2-nit rop enoxy)propan-1-ol (Intermediate 36-1) 5-Chloro-4-methoxy-2-nitro-phenol (1.00 g, 4.91 mmol), 3-iodopropan-1-ol (685 ^L, 7.37 15 mmol) and K2CO3 (2.04 mg, 14.7 mmol) were suspended in DMF (15mL) and RM stirred at 70 °C for 5 h. After cooling to RT, RM was diluted with water (30 mL) and extracted with EtOAc (4 x 20mL). Combined organic layers were washed with water (40mL) and sat. aq. NaCl (40mL), and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-40 % DCM/MeOH (99:1) in DCM to afford the title product (1.08 g). 20 LCMS (Method 1): Rt = 0.93 min, ES + m/z 262.1 [M+H] + . Step 2 89 3-(5-(6-chloro-3-methyl-1H-py razoo[ ,3-c]pyridin-1-yl)-4-methoxy-2- nitrophenoxy)propan-1-ol (Intermediate 36-2) A suspension of intermediate 36-1 (1.01 g, 3.86 mmol), 6-chloro-3-methyl-1H-pyrazolo[4,3- 5 c]pyridine (610 mg, 3.64 mmol), and K 2 CO 3 (1.51 g, 10.9 mmol) in DMSO (10.0 mL) was stirred at 120 ºC for 5 h. After cooling to RT, RM was diluted with water. The formed precipitate was collected by filtration, washed with water, and dried to afford the title product (700 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.00 min, ES + m/z 393.2/395.2 [M+H] + . 10 Step 3 3-(5-(6-Chloro-3-methyl-1H-p yrazo o[ ,3-c]pyr din-1-yl)-4-methoxy-2- nitrophenoxy)propyl methanesulfonate (Intermediate 36-3) To a 0 °C cooled solution of intermediate 36-2 (200 mg, 0.51 mmol) in DMF (5mL), 15 methanesulfonyl chloride (59 ^L, 0.76 mmol) was added dropwise and RM stirred at RT for 2h. RM was poured into water (20 mL) and the formed precipitate collected by filtration, washed with water, and dried to afford the title product (225 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.11 min, ES + m/z 471.1/473.1 [M+H] + . 20 Step 4 90 8-(6-Chloro-3-methyl-1H-pyra zoo[ ,3-c]pyr d n-1-yl)-7-methoxy-2,3,4,5- tetrahydrobenzo[b][1,4]oxazepine (Intermediate 36) To a stirrin solution of intermediate 36-3 (210 mg, . mmo n e anol (10 mL) at 80 5 , a sou o o 4 g, 1.7 mmol) in water (5 mL) and Fe (0.28 g, 5.10 mmol) were added. RM was stirred at 80 °C overnight. After cooling to RT, RM was diluted with EtOAc (20 mL), filtered through a bed of diacematous earth and washed thoroughly with EtOAc. Filtrate was concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH (95:5) in DCM to afford the title product (65 mg). 10 LCMS (Method 1): Rt = 0.99 min, ES + m/z 345.2 [M+H] + . Intermediate 37a tert-Butyl 7-(6-chloro -3-((3-(d met yam no)propyl)carbamoyl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazin e-4-carboxylate 15 (Intermediate 37a) To a solution of intermediate 24 (100 mg, 0.22 mmol), DIPEA (76 ^L, 0.43 mmol) and HATU (91 mg, 0.26 mmol) in DMF (2.0 mL), N',N'-dimethylpropane-1,3-diamine (33 ^L, 0.26 mmol) was added and RM stirred at RT for 1 h. RM was diluted with EtOAc (20 mL) and washed with sat. NaHCO3 (4x4 mL), and sat. aq. NaCl (10 mL). Combined organic layers were 20 concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-85 % DCM/MeOH/NH4OH (95:9:1.5) in DCM to afford the title product (63.6 mg). LCMS (Method 1): Rt = 0.92 min, ES + m/z 545.4/547.3 [M+H] + . Intermediates 37b to 37k 91 The following intermediates were prepared in a similar manner to intermediate 37a from the indicated starting materials. I ntermediate Structure Starting m aterials LCMS 92 93 Step 1 1-(2-(1-(4-(tert-butoxyca rbony)-6-met oxy-3, -d ydro-2H-benzo[b][1,4]oxazin-7- 5 yl)-6-chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxamido)ethyl) piperidine-4-carboxylic acid (Intermediate 37m-1) To a solution of intermediate 37l (1.0 g, 1.48 mmol) in THF (10 mL), an aq. solution 1M of LiOH (10.6 mL, 11.8 mmol) was added and RM stirred at RT for 16h. RM was evaporated in 94 vacuo and the residue taken with water and the pH adjusted to 6.5 using aq.1M HCl. The aqueous phase was extracted with DCM (3x10 mL), then combined organic layers were evaporated to dryness in vacuo to afford the title product (756 mg). LCMS (Method 1): Rt = 0.99 min, ES + m/z 615.1/617.1 [M+H] + 5 Step 2 tert-butyl 7-(6-chloro-3-(( -( -(d met y carbamoyl)piperidin-1-yl)ethyl)carbamoyl)- 1H-pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-ben zo[b][1,4]oxazine-4- carboxylate (Intermediate 37m) 10 A mixture of intermediate 37m-1 (34.0 mg, 55.3 ^mol), dimethylamine hydrochloride (9.11 mg, 111 ^mol), HATU (25.2 mg, 66.3 ^mol) and DIPEA (48.1 ^L, 0.28 mmol) in DMF (1 mL) was stirred at RT for 1.5 h. RM was quenched with water (20 mL) and extracted twice with EtOAc. Combined organic layers were washed with sat. aq. NaCl (20 mL) and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-70 % 15 DCM/MeOH/NH4OH (90:5:0.5) in DCM to afford the title product (26 mg). LCMS (Method 2): Rt = 0.95 min, ES + m/z 642.4/644.4 [M+H] + Intermediate 37n Step 1 20 N-(azetidin-3-yl)-6-chloro-1-( 6-met oxy-3, -d ydro-2H-benzo[b][1,4]oxazin-7-yl)- 1H-pyrazolo[4,3-c]pyridine-3-carboxamide (Intermediate 37n-1) A solution of intermediate 37k (100.0 mg, 0.16 mmol) and TFA (362 μL, 4.88 mmol) in DCM (4 mL) was stirred at RT for 4.5 h. RM was evaporated in vacuo. The residue was dissolved in MeOH and passed through a on a SCX cartridge, washed with methanol and eluted with 25 methanolic ammonia (7M) to afford the title product (63 mg). LCMS (Method 2): Rt = 0.68 min, ES + m/z 415.2/4.17.2 [M+H] + 95 Step 2 6-Chloro-1-(6-methoxy-3,4 -d ydro- -benzo[b][ ,4]oxazin-7-yl)-N-(1-((tetrahydro- 2H-pyran-4-yl)methyl)azetidin-3-yl)-1H-pyrazolo[4,3-c]pyridi ne-3-carboxamide 5 (Intermediate 37n) A mixture of intermediate 37n-1(50 mg, 0.12 mmol), 4-(bromomethyl)tetrahydropyran (33.3 mg, 0.13 mmol) and K 2 CO 3 (33.3 mg, 0.24 mmol) in DMF (1.5 mmol) was stirred at 80 °C for 7h. A second equivalent of 4-(bromomethyl)tetrahydropyran was added and RM further stirred for 48 hours. After cooling to RT, reaction was quenched with water (10 mL) and extracted with EtOAc 10 (2x15 mL). Combined organic layers were washed with sat. aq. NaCl (10 mL) and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-15 % DCM/MeOH/NH4OH (90:9:0.5) in DCM to afford the title product (48 mg). LCMS (Method 2): Rt = 0.72, ES + m/z 557.2/559.2. Intermediate 37o 15 (R)-6-chloro-1-(6-methoxy- , - y ro- - enzo[b][1,4]thiazin-7-yl)-N-((4- methylmorpholin-2-yl)methyl)-1H-pyrazolo[4,3-c]pyridine-3-ca rboxamide (Intermediate 37o) A mixture of intermediate 21-5 (30 mg, 77 ^mol), DABAL-Me3 (30 mg, 0.12 mmol), THF 20 (1 mL) and [(2R)-4-methylmorpholin-2-yl]methanamine (15 mg, 0.12 mmol) was heated under argon atmosphere, at 130 °C for 45 min, under ^W irradiation. After cooling to RT, RM was quenched with 4M HCl in dioxane (2 mL) and poured into sat. aq. NaHCO 3 . The mixture was extracted with DCM and combined organic layers evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH (9:1) in DCM to 25 afford the title product (26 mg). 96 LCMS (Method 1): Rt = 0.81 min, ES + m/z 489.2/491.2 [M+H] + Intermediate 37p 6-Chloro-N-(3-(dimethylam no)propy )- -(7-met oxy-1,2,3,4-tetrahydroquinolin-6- 5 yl)-1H-pyrazolo[4,3-c]pyridine-3-carboxamide (Intermediate 37p) The title compound was prepared on a similar manner to intermediate 37o starting from intermediate 27b and N’,N’-dimethylpropane-1,3-diamine. LCMS (Method 1): Rt = 0.77 min, ES + m/z 443.0/445.0 [M+H] + . Intermediate 37q 10 Step 1 tert-Butyl 7-(3-((2-brom oet y )carbamoy)-6-c oro-1H-pyrazolo[4,3-c]96yridine-1- yl)-6-methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazine-4-carboxyl ate (Intermediate 37q-1) 15 Intermediate 37q-1 was prepared in a similar manner to intermediate 37a starting from intermediate 24 and 2-bromoethylamine hydrochloride. LCMS (Method 2): Rt = 1.38 min, ES + m/z 566.6/568.1/570.1 [M+H] + Step 2 97 tert-Butyl 7-(6-chloro-3-((2-(4-methoxypiperidin-1-yl)ethyl)carbamoyl)- 1H- pyrazolo[4,3-c]97yridine-1-yl)-6-methoxy-2,3-dihydro-4H-benz o[b][1,4]oxazine-4- carboxylate (Intermediate 37q) A solution o of intermediate 37q-1 (80.0 mg, 0.14 mmol), 4-methoxypiperidine (19.5 mg, 5 0.17 mmol) and DIPEA (98.3 μL, 0.57 mmol) in dry DMF (1.3 mL) was stirred under argon, overnight at RT. RM was quenched by the addition of sat. aq. NH4Cl, then extracted with EtOAc (x2). Combined organic layers were washed with water and sat. aq. NaCl, dried over Na 2 SO 4 and evaporated to dryness. The residue was purified by flash chromatography on Si cartridge by eluting with DCM/MeOH mixture (from 20:1 to 10:1) to afford the title product (50.0 mg). 10 LCMS (Method 1): Rt = 0.95 min, ES + m/z 601.0/603.2 [M+H] + Intermediates 37q to 37w The following intermediates were prepared in a similar manner to Intermediate 37q from the indicated starting materials. Intermediate Structure Starting materials LCMS 98 Step 1 N-(2-bromoethyl)-6-chloro- -(6-met oxy-3, -d ydro-2H-benzo[b][1,4]oxazin-7-yl)- 5 1H-pyrazolo[4,3-c]pyridine-3-carboxamide (Intermediate 37x-1) A solution of intermediate 37q-1 (215.0 mg, 0.37 mmol) and TFA (1.38 mL, 18.6 mmol) in DCM (2.0 mL) was stirred at 16 °C for 16 h. RM was neutralized with sat. aq. NaHCO3 (20 mL) and layers were separated. Organic phase was washed with sat. aq. NaHCO 3 , passed through a 99 phase separator and evaporated in vacuo to afford the desired product (223.6 mg) that was used in the next synthetic steps without further purification. LCMS (Method 1): Rt = 1.07 min, ES + m/z 466.1/468.1/470.0 [M+H] + Step 2 5 6-Chloro-1-(6-methoxy -3, -d ydro- -benzo[b][ , ]oxazin-7-yl)-N-(2-(2-oxo-1-oxa- 8-azaspiro[4.5]decan-8-yl)ethyl)-1H-pyrazolo[4,3-c]pyridine- 3-carboxamide (Intermediate 37x ) To a mixture of intermediate 37x-1 (40.0 mg, 81 ^mol) in acetone (0.5 mL), 1-oxa-8- 10 azaspiro[4.5]decan-2-one hydrochloride (49 mg, 240 ^mol), Na2CO3(8.6 mg, 81 ^mol) and NaI (1.3 mg, 8.1 ^mol) in acetone (1.0 mL) were added and RM stirred at 40 °C for 24 h. RM was evaporated in vacuo and the residue partitioned between water (10 mL) and DCM (10 mL). After additional extraction of aqueous phase with DCM (3x10 mL), combined organic layers were passed through a phase separator and solvent evaporated in vacuo. The residue was purified by 15 flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH/NH 4 OH (90:5:0.5) in DCM to afford the title product (22 mg). LCMS (Method 1): Rt = 0.71 min, ES + m/z 541.1/543.1 [M+H] + Intermediate 38a 20 tert-Butyl 7-(6-chloro-3-((m et oxycarbony )am no)-1H-pyrazolo[4,3-c]pyridin-1-yl)-6- methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 38a) To a 0 °C cooled solution of intermediate 28 (60 mg, 0.14 mmol) in DCM (0.9 mL), pyridine (15 μL, 0.18 mmol) w as added followed by methyl chloroformate (13 μL, 0.17 mmol). RM was stirred at 0 o C for 30 min and at RT for 1 h. RM was quenched with sat aq. NaHCO 3 and extracted 100 with DCM (4 × 15 mL). Combined organic layers were washed with sat. aq. NaHCO3 and sat. aq. NaCl, dried over Na2SO4 and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH/NH 4 OH (90:9:1.5) in DCM to afford the title product (30.6 mg). 5 LCMS (Method 1): Rt = 1.27 min, ES + m/z 490.1/492.0 [M+H] + Intermediate 38b tert-Butyl 7-(6 -c oro-3-((( -(d met yamino)ethoxy)carbonyl)amino)-1H- pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[ b][1,4]oxazine-4-carboxylate 10 (Intermediate 38b) To a -78 o C cooled solution of intermediate 28 (250 mg, 0.5 mmol) and TEA (242 μL, 1.74 mmol) in DCM (15 mL), bis(trichloromethyl) carbonate (172 mg, 0.58 mmol) was added in one portion. RM was stirred at -78 o C for 1 h, followed by the addition of a solution of 2- (dimethylamino)ethanol (293 μL, 2.89 mmol) and TEA (161 μL, 1.16 mmol) in DCM (7.5 mL). 15 RM was stirred at -78°C for further 1 h, then allowed to warm to RT and quenched with sat. aq. NaHCO3. Aqueous phase was further extracted with DCM (3 × 25 mL) and combined organic layers were dried over Na2SO4 and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH/NH 4 OH (90:9:1) in DCM to afford the title product (310 mg). 20 LCMS (Method 2): Rt = 1.23 min, ES + m/z 547.3/549.2 [M+H] + Intermediates 38c to 38k The following intermediates were prepared in a similar manner to intermediate 38b from the indicated starting materials. It is additionally stated if base, solvent or temperature were varied. 101 Intermediate Structure Starting materials LCMS 102 103 2-(Dimethylamino)ethyl (6-chloro-1-(7-methoxy-2-oxo-1,2,3,4-tetrahydroquinolin-6- yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate (Intermediate 38l) A solution of CDI (75 mg, 0.47 mmol), intermediate 29 (40 mg, 0.12 mmol) and imidazole (2 mg, 0.35 mmol) in 2-Me-THF (1.3 mL) was stirred under argon at 90°C overnight. 2- 5 (dimethylamino)ethanol (35 μL, 0.35 mmol) was added and RM stirred at 90°C for further 5 h. After cooling to RT, RM was diluted with water and extracted with DCM/iPrOH (3×15 mL). Combined organic layers were washed with sat. aq. NaCl, dried over Na 2 SO 4 and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH/NH 4 OH (90:9:0.5) in DCM to afford the title product (19.5 mg). 10 LCMS (Method 2): Rt = 0.94 min, ES + m/z 459.1/461.1 [M+H] + Intermediate 38m 2-(Dimethylamino)ethyl (6 -c oro- -(6-met oxy-3,4-dihydro-2H-benzo[b][1,4]thiazin- 7-yl)-1H-pyrazolo[4,3-c]pyridin-3-yl)carbamate (Intermediate 38m) 15 A mixture of intermediate 21 (200 mg, 0.53 mmol), T3P ® (50 %, in EtOAc, 620 μL, 1.1 mmol), azido(trimethyl)silane (141 μL, 1.1 mmol) and TEA (222 μL, 1.6 mmol) in 2-Me THF (20 mL) was refluxed for 1 h. A solution of 2-(dimethylamino)ethanol (95 mg, 1.1 mmol) in 2-Me THF (1 mL) was added and RM refluxed for further 4 h. After cooling to RT, RM was diluted with EtOAc (35 mL), and washed with sat. aq. NaHCO3 (3x10 mL) and sat. aq. NaCl (10 mL). 20 Organic layer was dried over Na 2 SO 4 and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH/NH4OH (90:9:1.5) in DCM to afford the title product (116 mg). LCMS (Method 1): Rt = 0.75 min, ES + m/z 463.2/465.2 [M+H] + Intermediates 38n to 38x 25 The following intermediates were prepared in a similar manner to intermediate 38m from the indicated starting materials. 104 I ntermediate Structure Starting m aterials LCMS 105 I di 21 106 tert-buty 7-(6-c oro-3-((( -( ,3-d oxosondo n- -y)et oxy)carbony)am no)-1H- 5 pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[ b][1,4]oxazine-4-carboxylate (Intermediate 38y-1) To a -78 o C cooled solution of intermediate 28 (100 mg, 0.23 mmol) in DCM (5 mL), TEA (97 ^L, 0.70 mmol) and bis(trichloromethyl) carbonate (68.7 mg, 0.23 mmol) were added and RM stirred at -78°C for 1 h. 2-(2-Hydroxyethyl)isoindoline-1,3-dione (88 ^L, 0.926 mmol) and TEA 10 (65 ^L, 0.46 mmol) were added and RM stirred at RT overnight. RM was quenched with sat. aq. NaHCO3 (10 mL) and extracted with DCM (15 mL). Combined organic layers were washed with sat. aq. NaCl and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-10 % EtOAc in DCM to afford the title product (182 mg). LCMS (Method 2): Rt =1.32 min, ES + m/z 649.3/661.3 [M+H] + 15 Step 2 107 tert-Butyl 7-(3-(((2-aminoethoxy)carbonyl)amino)-6-chloro-1H-pyrazolo[4 ,3- c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazin e-4-carboxylate (Intermediate 38y-2) A solution of intermediate 38y-1 (155 mg, 0.24 mmol) and hydrazine hydrate (310 ^L, 6.39 5 mmol) in EtOH (5 mL) was stirred at RT for 2h. The precipitate removed by filtration and the filtrate partitioned between EtOAc (30 mL) and water (10 mL). Combined organic layers were washed with sat. aq. NaCl (10 mL) and solvent removed in vacuo to afford the desired product (81.7 mg) that was used in the next synthetic steps without further purification. LCMS (Method 2): Rt = 0.92 min, ES + m/z 519.2/521.2 [M+H] + 10 Step 3 tert-Butyl 7-(6-c oro-3-(((2-((2-oxotetrahydrofuran-3- yl)amino)ethoxy)carbonyl)amino)-1H-pyrazolo[4,3-c]pyridin-1- yl)-6-methoxy-2,3-dihydro- 4H-benzo[b][1,4]oxazine-4-carboxylate (Intermediate 38y) 15 A solution of intermediate 38y-2 (75.0 mg, 0.145 mmol), 3-bromotetrahydrofuran-2-one (14.4 ^L, 0.16 mmol) and DIPEA (101 ^L, 0.58 mmol) in DMF (3 mL) was stirred at RT for 72 h. RM was diluted with EtOAc (15 mL) and washed with water (2x10mL), and sat. aq. NaCl (15 mL). Solvents were removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-5 % DCM/MeOH/NH 4 OH (90:5:0.5) in DCM to afford the title product 20 (30 mg) LCMS (Method 2): Rt = 0.95 min, ES + m/z 603.3/605.3 [M+H] + Intermediate 39a 108 3-(6-chloro-1-(6-methoxy-3,4-dihydro-2H-benzo[b][1,4]thiazin -7-yl)-1H-pyrazolo[4,3- c]pyridin-3-yl)-1,1-dimethylurea (Intermediate 39a) A solution of intermediate 21 (40.0 mg, 0.11 mmol), T3P ® (50.0 % solution in DMF, (124 ^L, 0.21 mmol), azido(trimethyl)silane (28.2 ^L, 0.21 mmol) and TEA (44.4 ^L, 0.32 mmol) in 5 2-Me THF (0.5 ml) was refluxed overnight. After cooling to RT, RM was diluted with EtOAc (25 mL) and washed with sat. aq. NaHCO3 (3x15 mL) and sat. aq. NaCl (15 mL). Organic layer was dried over Na 2 SO 4 and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-30 % DCM/MeOH (90:1) in DCM to afford the title product (10 mg). 10 LCMS (Method 1): Rt = 1.01 min, ES + m/z 419.1/421.1 [M+H] + Intermediate 39b tert-Butyl 7-(6-chloro-3-( 3-(3-morp o nopropy )ureido)-1H-pyrazolo[4,3-c]pyridin-1- yl)-6-methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazine-4-carboxyl ate (Intermediate 39b) 15 To a -78 °C cooled solution of intermediate 28 (50.0 mg, 0.12 mmol) in DCM (3 mL), DIPEA (48.4 μL, 0.35 mmol) and bis(trichloromethyl) carbonate (34.4 mg, 0.12 mmol) were added and RM stirred for 2 h. A solution of 3-morpholinopropan-1-amine (59.2 μL, 0.40 mmol) and DIPEA (32.3 μL, 0.23 mmol) in DCM (2 mL) was added dropwise and RM stirred at -78°C for further 2 h. RM was allowed to warm to RT and diluted with sat. aq. NaHCO3. Aqueous layer 20 was further extracted with DCM (3×15 mL), and combined organic layers dried over Na 2 SO 4 and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH/NH 4 OH (90:9:0.5) in DCM to afford the title product (19.4 mg). LCMS (Method 2): Rt =1.24 min, ES + m/z 602.3/604.3 [M+H] + Intermediate 40 25 Step 1 109 tert-Butyl 7-oxo-6-oxa-2-azaspiro[3.4]octane-2-carboxylate (Intermediate 40-1) To a solution of tert-butyl 6-oxo-2-azaspiro[3.3]heptane-2-carboxylate (1 g, 4.64 mmol) in DCM (10 mL) at 0°C, NaHCO 3 (1.68 g) and m-CPBA (1.43 g, 7.89 mmol) were added and RM stirred at RT for 3 h. RM was diluted with DCM (10 mL) and sat. aq. NaHCO3 (25 mL). The 5 phases were separated, and aqueous layer extracted with DCM (2 × 25 mL). Combined organic layers were washed with brine (20 mL) and solvent removed in vacuo. Residue was slurried in a mixture DCM/ MeOH, filtered to remove insoluble solids and the filtrate evaporated in vacuo to afford the title product (1.11 g). 1H-NMR (500 MHz, DMSO-d 6 ) į: 4.39 (s, 2H), 3.87 (bs, 4H), 2.83 (s, 2H), 1.37 (s, 9H). 10 Step 2 6-Oxa-2-azaspiro[3.4]octan-7-one acetate (intermediate 40- 2) A solution of Intermediate 40-1 (200 mg, 0.88 mmol) and TFA (9.81 mL, 132.0 mmol) in DCM (10.0 mL) was stirred at RT for 3h. The volatiles were evaporated in vacuo and residue 15 triturated in diethyl ether to afford the title product (198.9 mg). 1H-NMR (500 MHz, DMSO-d 6 ) į: 8.80 (bs, 2H), 4.40 (s, 2H), 4.04 (bs, 4H), 2.90 (s, 2H). Step 3 tert-Butyl (2-(7-oxo-6-oxa-2 -azasp ro[3. ]octan-2-yl)ethyl)carbamate (Intermediate 20 40-3) A mixture of N-Boc-2-aminoacetaldehyde (109.0 mg, 0.68 mmol, Intermediate 40-2 (128.0 mg, 0.57 mmol), TEA (79.2 ^L, 0.57 mmol) and powdered molecular sieves (4 Å, 200 mg) in methanol (4.0 mL) was stirred at RT for 1h. Na(CN)BH3 (143.0 mg, 2.27 mmol) and acetic acid (48.8 ^L, 0.85 mmol) were added and RM stirred at RT for 16h. RM was quenched with water (10 25 mL), diluted with EtOAc (10 mL), filtered and layers separated through a phase separator. Organic layer was evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH (10:1) in DCM to afford the title product. LCMS (Method 2): Rt = 0.75 min, ES + m/z 271.2 [M+H] + 110 Step 3 2-(2-Aminoethyl)-6-oxa-2-az asp ro[3. ]octan-7-one trifluoroacetate (Intermediate 40) A Solution of Intermediate 40-3 (50 mg, 0.185 mmol) and TFA (2.06 mL, 27.7 mmol) in 5 DCM (2.0 mL) was stirred at RT for 16 h. RM was evaporated to dryness and triturated in diethyl ether to afford the desired product (93 mg) that was used in the next synthetic step without further purifications. LCMS (Method 2): Rt = 0.34 min, ES + m/z 171.2 [M+H] + Intermediate 41 10 Step 1 Methyl (R)-1-(2-((te rt-butoxycarbony )am no)ethyl)pyrrolidine-3-carboxylate (Intermediate 41-1) A mixture of (R)-pyrrolidine-3-carboxylate hydrochloride (406 mg, 2.5 mmol), K2CO3 (925 15 mg, 6.7 mmol) and tert-butyl N-(2-bromoethyl)carbamate (500 mg, 2.2 mmol) in MeCN (2.5 mL) was stirred at 70 o C overnight. After cooling to RT, RM was filtered and solvent evaporated. The crude was dissolved in EtOAc and washed with water. Aqueous solution was extracted with EtOAc (2x 15 mL) and combined organic layers washed further with water (3x15 mL), sat aq NH4Cl (15 mL) and sat. aq. NaCl (15 mL). Organic layer was passed through a phase separator 20 and solvent evaporated to afford the title product (422 mg) that was used in the next synthetic steps without further purifications. 1H-NMR (300 MHz, CDCl 3 ) į: 4.95 (brs, 1H), 3.66 (s, 3H), 3.18-3.23 (m, 2H), 2.94-3.04 (m, 1H), 2.82 (t, J=7.8 Hz, 1H), 2.49-2.72 (m, 5H), 2.01-2.10 (m, 2H), 1.24 (s, 9H). Step 2 25 111 Methyl (R)-1-(2-aminoethyl)pyrrolidine-3-carboxylate (Intermediate 41) To a solution of Intermediate 41-1 (420 mg, 1.54 mmol) in dry DCM (3 ml) at 0 o C, TFA (1.77 mL, 23.1 mmol) was added dropwise. RM was warmed to RT and stirred for 2h. RM was loaded onto a preconditioned SCX column, washed with MeOH and eluted with 2N methanolic 5 ammonia (50 mL). Evaporation of methanolic fractions afforded the desired product (263 mg). 1H-NMR (300 MHz, CDCl3) į: 3.66 (s, 3H), 2.95-3.06 (m, 1H), 2.76-2.87 (m, 3H), 2.61-2- 68 (m, 2H), 2.43-2.58 (m, 3H), 2.02-2.16 (m, 2H). Intermediates 42a-b The following intermediates were prepared in a similar manner to intermediate 37a from the 10 indicated starting materials. Intermediate Structure Starting materials LCMS 112 tert-Butyl 7-(6-chloro-3-(1-methylpiperidine-4-carboxamido)-1H-pyrazolo [4,3- c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[b][1,4]oxazin e-4-carboxylate (Intermediate 43) A solution of Intermediate 28 (50.0 mg, 0.12 mmol), TEA (97 ^L, 0.70 mmol) and 1- 5 methylpiperidine-4-carbonyl chloride hydrochloride (34.4 mg, 0.17 mmol) in DCM (2 mL) was stirred at RT for 2 h. A second equivalent of 1-methylpiperidine-4-carbonyl chloride hydrochloride was added and stirring proceeded overnight. RM was diluted with DCM (10 mL), washed with sat. aq. NaHCO3 and sat. aq. NH4Cl (10 mL each). Organic layer was separated through a phase separator and solvent removed in vacuo. The residue was purified by flash chromatography on Si 10 cartridge by eluting with 0-40 % DCM/MeOH/NH4OH (90:9:1.5) in DCM to afford the desired product (44 mg). LCMS (Method 2): Rt = 1.21 min, ES + m/z 557.3/559.3 [M+H] + Intermediate 44 Step 1 15 tert-Butyl 6-(3-amino- 6-c oro- -pyrazolo[4,3-c]pyridin-1-yl)-7-methoxy-3,4- dihydroquinoline-1(2H)-carboxylate (Intermediate 44-1) A Solution of Intermediate 27a (500 mg, 1.1 mmol), T3P ® (50 % in EtOAc, 1.27 mL, 2.2 mmol), azido(trimethyl)silane (289 μL, 2.2 mmol) and TEA (456 μL, 3.3 mmol) in 2-Me THF (20 20 ml) was refluxed for 30 min. Water (2.51 mL, 139 mmol) was added and RM was refluxed overnight. After cooling to RT, RM was diluted with EtOAc (25mL) and sat. aq. NaHCO 3 . Aqueous layer was extracted with EtOAc (3 × 25 mL). Combined organic layers were washed with sat. aq. NaCl, dried over Na2SO4 and concentrated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-35 % DCM/MeOH/NH 4 OH (90:9:1.5) in DCM 25 to afford the title product (279 mg). LCMS (Method 1): Rt = 1.20 min, ES + m/z 430.2/432.2 [M+H] + 113 Step 2 tert-Butyl 6-(6-c oro-3-(((2-((2-oxotetrahydrofuran-3- yl)thio)ethoxy)carbonyl)amino)-1H-pyrazolo[4,3-c]pyridin-1-y l)-7-methoxy-3,4- 5 dihydroquinoline-1(2H)-carbox late (Intermediate 44) Solution of Intermediate 44 (100 mg, 0.23 mmol), TEA (97.3 μL, 0.70 mmol) in DCM (5 mL) was cooled -78°C and bis(trichloromethyl) carbonate (69.0 mg, 0.23 mmol) was added in one portion. RM was stirred under the same conditions for 1h. A solution of Intermediate 13 (151 mg, 0.93 mmol) and TEA (64.8 μL, 0.47 mmol) in DCM (2.5 mL) was added to RM at -78°C and 10 stirring proceeded for 2h under cooling and overnight at RT. Reaction was quenched with sat. aq. NaHCO 3 and added with DCM and water. Layers were separated, and aqueous one extracted with DCM (3 × 20 mL). Combined organic layers were washed with sat. aq. NaCl, dried over Na2SO4 and solvent removed in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-35 % DCM/EtOH/NH4OH (90:4:1) in DCM to afford the title product (177 mg). 15 LCMS (Method 2): Rt = 1.28 min, ES + m/z 618.2/620.2 [M+H] + Intermediates 45a to 45d The following intermediates were prepared in a similar manner to step 2 of Intermediate 37q from the indicated starting materials. Intermediate Structure Starting materials LCMS 114 tert-Butyl 7-(6-c oro-3-((-(-met y-6-oxomorpholino)ethyl)carbamoyl)-1H- pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H-benzo[ b][1,4]oxazine-4-carboxylate 5 (Intermediate 45e) 115 Mixture of Intermediate 45a (42.0 mg, 0.07 mmol), K2CO3 (11.4 mg, 0.08 mmol) and ethyl 2-bromoacetate (9.1 μL, 0.08 mmol) in dry MeCN (9 mL) was stirred at RT overnight, then heated in pressure vial at 110 °C for 9 h. After cooling to RT, solvent was removed in vacuo and residue taken with DCM (20 mL), washed with sat. aq. NaHCO3 (3x10 mL) and sat. aq. NaCl (10 mL). 5 Organic layer evaporated to afford the title product (45 mg,) that was used in the next step without further purifications. LCMS (Method 2): Rt = 1.25 min, ES + m/z 601.3/603.3 [M+H] + Intermediate 45f Step 1 10 6-Chloro-N-(2-((2-hydro xy- -met y propy)am no)ethyl)-1-(6-methoxy-3,4-dihydro- 2H-benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3-c]pyridine-3-ca rboxamide (Intermediate 45f- 1) A solution of Intermediate 45d (80 mg, 0.14 mmol) and TFA (517 μL, 7.0 mmol) in DCM 15 (9.5 mL) was stirred at RT overnight. Solvent was removed in vacuo and residue purified on a SCX column by elution with methanol and methanolic ammonia to afford the title compound (55.1 mg) that was used in the next step without further purification. LCMS (Method 1): Rt = 0.70 min, ES + m/z 475.2/477.1 [M+H] + Step 2 20 6-Chloro-N-(2-(2,2-dime t y -6-oxomorp o no)et y )-1-(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3-c]pyridine-3-carbo xamide (Intermediate 45f) 116 A solution of Intermediate 45f-1 (55 mg, 0.12 mmol) in toluene/acetonitrile was added to a precooled mixture of oxaldehyde (40 %, 15 μL, 0.12 mmol) in toluene (0.3 mL) at 10 °C. RM was stirred for 3 h at 10 °C and RT overnight. RM was diluted with water and extracted with DCM (4 × 10 mL). Combined organic layers were passed through phase separator and solvent was 5 evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH/NH4OH (90:5:0.5) in DCM to afford the title product (32 mg). LCMS (Method 2): Rt = 0.99 min, ES + m/z 515.2/517.1 [M+H] + PREPARATION OF EXAMPLES Example 1 10 6-(Difluoromethoxy)-7-(3- met y -6-(pyrazo o[ ,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]oxazine (Example 1) To a degassed solution of intermediate 32b (33.0 mg, 0.09 mmol), 3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyrimidine (30.9 mg, 0.13 mmol) and K3PO4 (38.2 mg, 15 0.18 mmol) THF (0.5 mL) and water (0.2 mL), XPhos Pd G3 (7.62 mg, 9.0 ^mol) was added and RM stirred at 75 °C for 2 h. After cooling to RT, RM was diluted with water and extracted with DCM (3x5 mL). Combined organic layers were passed through phase separator and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-50 % DCM/MeOH/NH 4 OH (90:9:0.5) in DCM, and purified through an SCX column to afford the title 20 compound (17 mg). LCMS (Method 4): Rt = 4.42 min, ES + m/z 450.1 [M+H] + 1 H-NMR (500 MHz, DMSO-d6) į: 9.22 (br d, J=7.0 Hz, 1H), 9.12 (s, 1H), 8.89 (s, 1H), 8.72 (d, J=2.7 Hz, 1H), 8.18 (s, 1H), 7.16 (m, 1H), 6.88 (s,1H), 6.88 (t, J=72.4 Hz, 1H), 6.65 (s, 1H), 6.57 (s, 1H), 4.19 (br s, 2H), 3.39 (br s, 2H), 2.61 (s, 3H). 25 Examples 2 to 39 The following examples were prepared in a similar manner to Example 1 from the indicated intermediate. 117 Example Structure / Name Interme 1 d iate H-NMR LCMS 118 (600 MHz DMSO-d 6 ) į: 119 120

121 (600 MHz, DMSO-d6) į: 9.44 (d, J=1.2 Hz, 1H),

122 (600 MHz , DMSO-d 6 ) 123 (300 MHz, DMSO-d6) į:

124 (600 MHz, DMSO-d 6 ) į: 1046 (br s 1H) 924-

125 (300 MHz, DMSO-d 6 ) į:

126 (300 MHz, DMSO-d 6 ) į: 127 (500 MHz, DMSO-d6) į: 128 (500 MHz, DMSO) į: 129 (500 MHz, DMSO-d6) į:

130 (500 MHz, DMSO-d6) į: 10.42-10.49 (br s, 1H), 131 (500 MHz, DMSO-d 6 ) į: Step 1 tert-Butyl 6-methoxy-7- (3-met y -6-(pyrazoo[ ,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3- 5 c]pyridin-1-yl)-2,3-dihydro-4H-benzo[b][1,4]oxazine-4-carbox ylate (Example 40 – step 1) A degassed mixture of intermediate 32a (172 mg, 0.40 mmol), 3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazolo[1,5-a]pyrimidine (117 mg, 0.48 mmol), K 3 PO 4 (169 mg, 0.80 mmol) and XPhos PdG3 (17 mg, 20 ^mol) in water (1 mL) / THF (2 mL) was stirred under nitrogen at 55 132 °C for 1.5 h. After cooling to RT, RM was diluted with water (5 mL) and extracted with DCM (3x10 mL). Combined organic layers were washed with water and sat. aq. NaCl (10 mL each), dried over Na 2 SO 4 and evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-80 % DCM/MeOH/NH4OH (90:9:0.5) in DCM to afford the title 5 product (158.4 mg). LCMS (Method 2), Rt = 1.22, ES + m/z 514.3. Step 2 6-Methoxy-7-(3-methyl-6-( pyrazo o[ ,5-a]pyr m din-3-yl)-1H-pyrazolo[4,3-c]pyridin- 10 1-yl)-3,4-dihydro-2H-benzo[b][1,4]oxazine (Example 40) TFA (0.7 mL, 9.23 mmol) was added to a solution of intermediate examp e - step 1 (158 mg, 0.31 mmol) in DCM (5.0 mL) and stirred at RT overnight. Solvents were removed in vacuo, and the residue loaded on a SCX cartridge, washed with methanol, and eluted with methanolic ammonia (7M) to afford the title product (78.7 mg) 15 LCMS (Method 3), Rt = 2.67 min, ES + m/z 414.1 [M+H] + 1 H-NMR (600 MHz, DMSO-d6) į: 9.21 (dd, J=6.9, 1.8 Hz, 1H); 9.09 (d J=1.2 Hz, 1H); 8.88 (s, 1H); 8.71 (dd, J=4.0, 1.7 Hz, 1H); 8.14 (d J=1.2 Hz, 1H); 7.15 (dd, J=6.9,4.1Hz, 1H); 6.73 (s, 1H); 6.46 (s, 1H); 6.26 (brs, 1H); 4.13 (t, J=3.3Hz, 2H); 3.62 (s, 3H); 3.36 (m, 2H); 2.60 (s, 3H). Examples 41 to 70 20 The following examples were prepared by a two step process similarly to example 40 starting from the indicated intermediate.

133 : ) 4 2 z / : ) 4 1 z / 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 Example 71 Step 1 6'-Methoxy-5'-(3-methyl -6-(pyrazo o[ ,5-a]pyr m dn-3-yl)-1H-pyrazolo[4,3- 5 c]pyridin-1-yl)-1'-((2-(trimethylsilyl)ethoxy)methyl)spiro[c yclopropane-1,3'-indolin]-2'-one (Example 71 – step 1) The title compound was prepared similarly to example 40 – step 1 starting from intermediate 34 and 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5- a]pyrimidine. LCMS (Method 1), Rt = 1.32, ES + m/z 568.3 10 Step 2 6'-Methoxy-5'-(3-methyl-6-( pyrazo o[ , -a]pyr midin-3-yl)-1H-pyrazolo[4,3- c]pyridin-1-yl)spiro[cyclopropane-1,3'-indolin]-2'-one (Example 71) An ice bath cooled solution of intermediate example 71 – step 1 (32.0 mg, 0.05 mmol) in 15 dry DCM (1 mL) was treated with TFA (371 ^L, 4.85 mmol) and stirred at RT overnight. RM was evaporated in vacuo and the residue partitioned between DCM (2x10 mL)/ NaHCO3 (15 mL). Combined organic layers were evaporated in vacuo and the residue taken in DCM (1mL) and treated with 7N methanolic ammonia (200^L), then stirred at RT for 4 h. RM was diluted with DCM (8 mL) and washed with sat. aq. NaHCO3 (10 mL), and sat. aq. NaCl (10 mL). Organic layer 20 was evaporated in vacuo and the residue purified by flash chromatography on Si cartridge, by eluting with 0-100 % DCM/MeOH/NH4OH (90:5:0.5) in DCM, to afford the title product (12.5 mg). LCMS (Method 4): Rt = 3.89 min, ES + m/z 438.2 [M+H] + 149 1H-NMR (600 MHz, DMSO-d6) į: 10.83 (s, 1H), 9.22 (dd, J=6.9, 1.7 Hz, 1H), 9.12 (d, J=1.1 Hz, 1H), 8.88 (s, 1H), 8.70 (dd, J=4.0, 1.7 Hz, 1H), 8.17 (d, J=1.1 Hz, 1H), 7.14-7.16 (m, 2H), 6.86 (s, 1H), 3.78 (s, 3H), 2.63 (s, 3H), 1.58-1.61 (m, 2H), 1.44-1.47 (m, 2H). Example 72 5 Step 1 5-Methoxy-6-(3-((2-morph o noe y)am no)- -(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)-3-((2-(trimethylsilyl)ethoxy)me thyl)benzo[d]oxazol-2(3H)-one (Example 72 – step 1) 10 The title compound was prepared similarly to intermediate example 72 – step 1 starting from intermediate 19 and 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5- a]pyrimidine. LCMS (Method 1), Rt = 0.79, ES + m/z 658.5 Step 2 15 5-Methoxy-6-(3-((2-morp o noet y)am no)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridin-1-yl)benzo[d]oxazol-2(3H)-one (Example 72) The title compound was prepared similarly to example 72 (step 2) starting from intermediate example 72 – step 1. LCMS (Method 4): Rt = 3.52 min, ES + m/z 528.2 [M+H] + 20 1 H-NMR (500 MHz, DMSO-d6) į: 11.87 (s, 1H), 9.20 (d, J=7.1 Hz, 1H), 9.06 (s, 1H), 8.85 (s, 1H), 8.67 (m, 1H), 8.04 (s, 1H), 7.42 (s, 1H), 7.13 (dd, J=7.5, 3.4 Hz, 1H), 7.02 (s, 1H), 6.68 (t, J=5.3 Hz, 1H), 3.82 (s, 3H), 3.56-3.63 (m, 4H), 3.44 (q, J=6.7 Hz, 2H), 2.60-2.65 (m, 2H); 2.42- 2.48 (m, 2H, overlapped with DMSO); 2.35-2.38 (m, 2H). Example 73 150 Step 1 tert-Butyl 7-(3-((3-(dim ethylamino)propyl)carbamoyl)-6-(imidazo[1,2-b]pyridazin-3- yl)-1H-pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H -benzo[b][1,4]oxazine-4- 5 carboxylate (Example 73 – step 1) To a degassed mixture of intermediate 37a (70 mg, 0.13 mmol), bis(triphenylphosphine)palladium (II) chloride (14 mg, 19 ^mol) in DMF (1 mL), intermediate 12 (270 mg, 0.33 mmol) in DMF (1 mL) was added and RM stirred at 110 °C overnight. After cooling to RT, RM was partitioned between EtOAc (25 mL) and water (15 mL). Aqueous phase 10 was brought to pH 9 with aq. 2N NaOH and extracted with EtOAc (15 mL). Combined organic layers were evaporated in vacuo. The residue was purified by flash chromatography on Si cartridge by eluting with 0-90 % DCM/MeOH/NH4OH (90:9:1.5) in DCM to afford the title product (38 mg). LCMS (Method 1), Rt = 0.86, ES + m/z 628.4 15 Step 2 N-(3-(Dimethylamino)p ropy )-6-( m dazo[ , -b]pyr dazin-3-yl)-1-(6-methoxy-3,4- dihydro-2H-benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3-c]pyrid ine-3-carboxamide (Example 73) 20 The title compound was prepared similarly to example 40 (step 2). LCMS (Method 4), Rt = 3.94 min, ES + m/z 528.2 [M+H] + 1 H-NMR (600 MHz, DMSO-d6) į: 9.51 (s, 1H), 8.76 (br s, 2H), 8.59 (s, 1H), 8.52 (s, 1H), 8.30 (d, J=9.2 Hz, 1H), 8.24 (s, 1H), 7.36 (dd, J=9.3, 4.4 Hz, 1H), 6.91 (s, 1H), 6.50 (s, 1H), 4.14 151 (br s, 2H), 3.67 (s, 3H), 3.41-3.35 (m, 4H), 2.40 (br t, J=7.2 Hz, 2H), 2.23 (s, 6H), 1.78-1.72 (m, 2H). Example 74 Step 1 5 tert-Butyl 7-(3-(( tert-butoxycarbony )( -(dimethylamino)ethyl)amino)-6-((3- methoxypyrazin-2-y l)amino)-1H-pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihyd ro-4H- benzo[b][1,4]oxazine-4-carboxylate (Example 74 – step 1) A degassed mixture of intermediate 31a (25 mg, 41 ^mol), 2-amino-3-methoxypyrazine (6.7 10 mg, 54 ^mol), sodium tert-butoxide (6.0 mg, 62 ^mol) and XPhos PdG3 (3.5 mg, 4.1 ^mol) in dioxane (600 ^L) was stirred at 100°C overnight. After cooling to RT, RM was chromatographed on Si cartridge by eluting with 0-30 % DCM/MeOH/NH 4 OH (90:15:1.5) in DCM to afford the title product (11 mg). LCMS (Method 2): Rt = 1.59 min, ES + m/z 692.4 [M+H] + 15 Step 2 3-((3-((2-(dimethylamino )et y )am no)- -(6-met oxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-1H-pyrazolo[4,3-c]pyridin-6-yl)ami no)pyrazin-2(1H)-one (Example 74) 20 A solution of intermedia e example 74 – step 1 (11.0 mg, 8 ^mol), NaI (3.58 mg, 24 ^mol) and TMS-Cl (9.0 μL, 72 ^mol) in acetonitrile (0.5 mL) was stirred at 85 °C for 2 h. After cooling 152 to RT, RM was chromatographed on Si cartridge by eluting with 0-100 % DCM/MeOH/NH4OH (90:15:1.5) in DCM to afford the title product (2 mg). 1H-NMR (600 MHz, DMSO-d 6 ) į: 8.74 (d, J=0.9 Hz, 1H), 8.64 (s, 1H), 7.90 (d, J=0.9 Hz, 1H), 6.93 (d, J=4.4 Hz, 1H), 6.86 (d, J=4.4 Hz, 1H), 6.68 (s, 1H), 6.52 (m, 1H), 6.42 (s, 1H), 6.12 5 (s, 1H), 4.11 (t, J=4.2 Hz, 2H), 3.65 (s, 3H), 2.65 (m, 2H), 2.30 (br s, 6H). LCMS (Method 4): Rt = 4.34, ES + m/z 478.1 [M+H] + Examples 75 to 76 The following examples were prepared by a two step process similarly to example 74 starting from the indicated intermediate.

153 = = 2 p e t S 1 p e t S y ( n l n e p 2 - - 6 i b r a di - 4 y ) - l y i d m o o r - ) a r n p ) d l m y a )l c - r 3 e , 3 7 - - 2 i r o y h - 7 y - - p e i - p y x n i - n y p e d e o z i z ] c i / n r i i d - n 2 - n i o r h a x a r - m a u t m- c a u l 4 , i y 3 z n - a i x z d i o o a r r y u t l f e o y p ] i d m ] - 4 y 3 , x 6 , p 4 [ o 1 o r o l b r r t h S t e y x ] p c - 4 ( - [ ] d y o z a c m i o 4 , 1 o r - 3 , , 4 1 - b [ h i a r d h t e [ ] d y 4 [ (- ) l o z d - y (- 3 m b [ h ( i o l 3 ( - y p n e 4 , p - - o z n d - o z N o r p b - 3 - o H 1 N e 4 b , 3 a r y ) p l y H 2 x o e l e p l e m p l a m5 p m 6 x a 7 a 7 E x E x E 154 Example 77 N-(2-(2,2-dimethyl-6 -oxomorp o no)et y )- -(6-met oxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)- 1H-pyrazolo[4,3-c]pyridine-3- 5 carboxamide (Example 77) The title compound was prepared in a similar manner to Example 1 from Intermediate 45f and 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5- a]pyrimidine. LCMS (Method 3), Rt = 4.51 min, ES + m/z 598.3 [M+H] + 1 H-NMR (500 MHz, DMSO-d6) į: 9.45 (d, J=1.2 Hz, 1H), 9.24 (dd, J=6.9, 1.7 Hz, 1H), 8.91 10 (s, 1H), 8.74 (dd, J=4.1, 1.7 Hz, 1H), 8.53 (t, J=6.0 Hz, 1H), 8.25 (d, J=0.9 Hz, 1H), 7.17 (dd, J=7.0, 4.3 Hz, 1H), 6.86 (s, 1H), 6.49 (s, 1H), 6.36-6.41 (m, 1H), 4.14 (t, J=4.3 Hz, 2H), 3.63 (s, 3H), 3.48 (q, J=6.3 Hz, 2H), 3.36-3.42 (m, 2H), 3.29 (s, 2H), 2.57-2.66 (m, 4H), 1.33 (s, 6H). Example 78 Step 1 15 1-(4-(tert-Butoxycarbonyl)- 6-(d uoromet oxy)-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-chloro-1H-pyrazolo[4,3-c]pyridin e-3-carboxylic acid (Example 78-step 1) 155 Title compound was prepared similarly to Intermediate 24 (Method 1) starting from of 6- chloro-1H-pyrazolo[4,3-c]pyridine-3-carboxylic acid and Intermediate 1. LCMS (Method 1), Rt = 1.22 min, ES + m/z 497.1/499.0 [M+H] + Step 2 5 1-(4-(tert-Butoxycarbony )-6-(d uoromet oxy)-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)- 1H-pyrazolo[4,3-c]pyridine-3- carboxylic acid (Example 78-step 2) To a degassed mixture of Example 78-step 1 (200 mg, 0.40 mmol), 3-(4,4,5,5-tetramethyl- 10 1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyrimidine (138 mg, 0.56 mmol) and K3PO4 ( 0.5 M in water, 1.61 mL, 0.81 mmol) in THF/water (5.61 mL), XPhos Pd G3 (19 mg, 22 μmol) was added and RM heated at 80 °C for 1 h. Warm reaction mixture was filtrated and THF evaporated in vacuo. Water was added to RM until solids dissolved, filtered and added with sat. aq. NH 4 Cl to form a precipitate that was collected by filtration to give the title product (77 mg) used in the next 15 step without further purifications. LCMS (Method 1), Rt = 1.01 min, ES + m/z 580.2 [M+H] + Step 3 tert-Butyl 6- (d uoromet oxy)-7-(3-((3-(d met ylamino)propyl)carbamoyl)-6-20 (pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine-1 -yl)-2,3-dihydro-4H- benzo[b][14]oxazine-4-carbox late (Exam le 78-ste 3) 156 Title compound was prepared similarly to Intermediate 37a starting from Example 78-step 2 and N',N'-dimethylpropane-1,3-diamine. LCMS (Method 1), Rt = 0.86 min, ES + m/z 664.3 [M+H] + Step 4 5 1-(6-(Difluoromethoxy)- 3, -d ydro- -benzo[b][ , ]oxazin-7-yl)-N-(3- (dimethylamino)propyl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H- pyrazolo[4,3-c]pyridine-3- carboxamide (Exam le 78) 10 mple 78-step 3. LCMS (Method 4): Rt = 3.92, ES + m/z 564.2 [M+H] + 1 H-NMR (500 MHz, DMSO-d6) į: 9.48 (d, J=1.2 Hz, 1H), 9.24 (dd, J=7.0, 1.8 Hz, 1H), 8.92 (s, 1H), 8.74 (dd, J=4.1, 1.7 Hz, 1H), 8.70 (t, J=6.0 Hz, 1H), 8.29 (d, J=0.9 Hz, 1H), 7.18 (dd, 15 J=7.0, 4.3 Hz, 1H), 7.06 (s, 1H), 6.95 (t, J=73.9 Hz, 1H), 6.66-6.71 (m, 2H), 4.20 (t, J=4.3 Hz, 2H), 3.39-3.44 (m, 2H), 3.34-3.38 (m, 2H), 2.28 (t, J=7.0 Hz, 2H), 2.14 (s, 6H), 1.70 (quin, J=7.1 Hz, 2H). Example 79 Step 1 20 157 1-(2-(1-(4-(tert-Butoxycarbonyl)-6-methoxy-3,4-dihydro-2H-be nzo[b][1,4]oxazin-7-yl)- 6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridine -3- carboxamido)eth l)piperidine-4-carbox lic acid (Example 79-step 1) l), 3-(4,4,5,5-tetramethyl- 5 1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyrimidine (202 mg, 0.82 mmol), K 3 PO 4 (233 mg, 1.1 mmol) and XPhos PdG3 (46 mg, 0.055 mmol)in THF/water (2:1, 9 mL) was stirred at 70°C for 45min After cooling to RT, RM was partitioned between EtOAc (30 mL) and water (30 mL). After adjusting pH from 9.5 to 6.5, aqueous layer was extracted with DCM/isopropanol mix 4/1 (4x20 mL). Combined organic layers were passed through a phase separator and evaporated in vacuo. 10 The residue was purified by flash chromatography on Si cartridge by eluting with 0-100 % DCM/MeOH:HCO 2 H (90:9:2) in DCM to afford the title product (320 mg). LCMS (Method 1): Rt = 0.74 min, ES + m/z 698.4 [M+H] + Step 2 15 tert-Butyl 7-(3-((2 -( -carbamoyp perd n- -y)et y )carbamoyl)-6-(pyrazolo[1,5- a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy- 2,3-dihydro-4H- benzo[b][14]oxazine-4-carbox late (Example 79-step 2) mg, 0.14 mmol), HATU (65.4 mg, 0.17 mmol) and DIPEA (74.9 ^L, 0.43 mmol) in DMF (2.0 mL), NH 4 Cl (61.3 mg, 0.57 mmol) and 20 DIPEA (99.9 ^L, 0.57 mmol) were added and RM stirred at RT for 1h. A second portion of of HATU (15.0 mg, 0.04 mmol) was added and the mixture stirred for further 1h. Volatiles were removed in vacuo and residue purified by flash chromatography on Si cartridge by eluting with 0- 100 % DCM/MeOH/NH 4 OH (90:9:0.5) in DCM to afford the title product (32 mg). LCMS (Method 1): Rt = 0.74 min, ES + m/z 697.4 [M+H] + 25 Step 3 158 N-(2-(4-Carbamoylpipe r d n- -y)et y )- -(6-met oxy-3, -dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)- 1H-pyrazolo[4,3-c]pyridine-3- carboxamide (Exam le 79) 5 Title com ound was prepared similarly to step 2 of Example 40 starting from Example 79- step 2. LCMS (Method 4): Rt = 3.61, ES + m/z 597.2 [M+H] + 1 H-NMR (500 MHz, DMSO-d6) į: 9.46 (d, J=0.9 Hz, 1H), 9.23 (dd, J=7.0, 1.8 Hz, 1H), 8.91 (s, 1H), 8.73 (dd, J=4.1, 1.7 Hz, 1H), 8.44 (t, J=5.8 Hz, 1H), 8.24 (d, J=0.9 Hz, 1H), 7.19 (br s, 10 1H), 7.17 (dd, J=7.0, 4.0 Hz, 1H), 6.89 (s, 1H), 6.69 (br s, 1H), 6.49 (s, 1H), 6.38 (s, 1H), 4.14 (t, J=4.3 Hz, 2H), 3.63 (s, 3H), 3.44 (q, J=6.7 Hz, 2H), 3.39 (m, 2H), 2.93 (m, 2H), 2.48 (m, 2H), 2.00-2.08 (m, 1H), 1.90-1.99 (m, 2H), 1.66 (m, 2H), 1.50-1.58 (m, 2H). Example 80 Step 1 15 1- - 5- 20 tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyrimidin e . LCMS (Method 2): Rt = 1.17 min, ES + m/z 698.3 [M+H] + Step 2 159 (R)-1-(2-(1-(4-(tert-butox ycarbony )-6-met oxy-3, -dihydro-2H-benzo[b][1,4]oxazin- 7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)-1H-pyrazolo[4,3-c]py ridine-3- carboxamido)ethyl)pyrrolidine-3-carboxylic acid (Example 80-step 2) 5 A solution of Example 80-step 1 (574 mg, 0.82 mmol) and LiOH (1.00 M in water, 4.11 mL, 4.11 mmol) in THF (10 mL) was stirred at RT overnight. RM was diluted with water and washed with EtOAc, then pH of the aqueous layer adjusted to 5 and aqueous layer extracted with DCM:i- PrOH 8:1 (4x15 mL). Combined organic layers were dried over Na2SO4 and evaporated to dryness to afford the title compound (553 mg) that was used in the next step without further purification. 10 LCMS (Method 1): Rt = 0.82 min, ES + m/z 684.4 [M+H] + Step 3 tert-Butyl (R)-7-(3-((2-(3 -carbamoy pyrro d n- -y )ethyl)carbamoyl)-6-(pyrazolo[1,5- a]pyrimidin-3- l)-1H-pyrazolo[4,3-c]pyridin-1-yl)-6-methoxy-2,3-dihydro-4H- 15 benzo[b ][ , ]oxazine-4-carboxylate (Example 80-step 2) Title compound was prepared similarly to step 2 of Example 79 starting from Example 80- step . LCMS (Method 2): Rt = 1.00 min, ES + m/z 683.4 [M+H] + Step 4 160 (R)-N-(2-(3-Carbamoylp yrro d n- -y )et y )- -(6-methoxy-3,4-dihydro-2H- benzo[b][1,4]oxazin-7-yl)-6-(pyrazolo[1,5-a]pyrimidin-3-yl)- 1H-pyrazolo[4,3-c]pyridine-3- carboxamide (Example 80) 5 Title compound was prepared similarly to step 2 of Example 40 starting from Example 80- step 3. LCMS (Method 4): Rt = 3.56, ES + m/z 583.1 [M+H] + 1 H-NMR (500 MHz, DMSO-d6) į: 9.46 (d, J=0.9 Hz, 1H), 9.24 (dd, J=7.0, 1.5 Hz, 1H), 8.91 (s, 1H), 8.74 (dd, J=4.1, 1.7 Hz, 1H), 8.47 (t, J=5.8 Hz, 1H), 8.23-8.26 (m, 1H), 7.25 (br s, 1H), 10 7.17 (dd, J=7.0, 4.3 Hz, 1H), 6.89 (s, 1H), 6.76 (br s, 1H), 6.49 (s, 1H), 6.36-6.41 (m, 1H), 4.14 (br t, J=4.1 Hz, 2H), 3.63 (s, 3H), 3.43 (q, J=6.6 Hz, 2H), 3.37-3.41 (m, 2H), 2.84-2.90 (m, 1H), 2.79 (quin, J=7.8 Hz, 1H), 2.66-2.73 (m, 1H), 2.56-2.65 (m, 2H), 2.37-2.48 (m, 2H), 1.87 (q, J=7.3 Hz, 2H). Examples 81 to 86 15 The following examples were prepared by a two steps process similarly to example 40 starting from the indicated intermediate. It is additionally stated if base, catalyst, solvent or temperature were varied.

161 : ) 4 z / : 2 m + ) 4 1 z / m + - - - 7 H ( i - 2 - d 3 - 6 o 2 n i e - n r - H a d y 6 - 1 - - 2 ( o - a t mi r n i r N c d t c h i ) l )l y - 3 e d - o ] y p i r y o ] d - y - 7 - - - 3 - e n m y ) l a h i y 4 - . ] a n / d 7 3 [ - p 5 ] 4 . 3 4 , , c - e d [ 3 n i n i i d o - y z a d i i r e d e - - r 4 , n o i r z i 1 p [ 3 , i m ri x x mi y p i m s o l 4 [ a p s o h o t ] r y ] c - a u t 3 c - a u y r t x x o a o z o l x o a z e 4 , x ] z a a r o z b r a - m - 1 [ p ] ] a 3 , 4 o [ b r S o h t 4 e , - 1 2 y a r a c 6 - 6 ( b [ - 5 o l a c m [ - p( y a - ] a x - p - x - 1 o z , 1 [ o z 6 b ( [ o - 6 -) o l H - - 2 ) l n e o l o a r y - o z 6 - o y 1 h -) ( y b l - 2 h t - z a r p 1 n e b x t o e ) l y - ( e y 3 - N ) l H y 2 y p ( e l e l e p p l m a m1 p 2 x a x 8 m a 8 E E x E 162 : ) 4 z / : 9 m + ) 4 4 z / m + t n m y e I d o S : i d x o n r p o a e d P ) 3 t n I d - - - - H- - 2 - 2( ) H l 1 - - 2( 6 H - H- 1 - - 1 e d o - y )l r y - 3 2 - o 6- )l - ) i l m d N y - h ) t l e ) - h o 3 - - e r d ) l n y y - 3 y - a x i y i n i d y h i - 7 - n i 3 - o b d - n i - 7 4 - l d i i r e d d - - n i d i n i r a , n o h mi y p i m 4 , z d a mi i r c - 4 3 i - z p r r y ] c - a x 3 - x o r y y p - e y a x x o p ] 3 , o y b x ] 4 o o ] m , p ] ] c - n i d h o a - 4 [ t 4 , o r a o h t 1 [ a - 3 , i r e 1 x o 5 , 1 l o c e ] b 5 , 1 4 [ o e p i m [ - ] - 6 b 6 ( [ - [ o - o l y l z o a r m - [ 6 o [ o l l o p l z y p (- z n 1 z n h t e o z z a r y h e e a r 1 b m y ( - b a r p y y p t e m ( N p ( e l e p l m a 3 p x 8 m a 4 8 E x E 163 164 PHARMACOLOGICAL ACTIVITY OF THE COMPOUNDS OF THE INVENTION (1-86). Biochemical Potency on JAK1, JAK2, JAK3 and Tyk2 Assay principle 5 The objective of this study was to assess the capability of compounds to inhibit all 4 JAK isoforms activity in a cell-free environment. Assay for JAK 1, JAK 2, JAK 3 and TYK2 were performed by Time-resolved fluorescence resonance energy transfer (TR-FRET) technology. It consists in the interaction of two labelled binding partners detected by the energy transfer from an excited donor to an acceptor dye and measurement of light emission by the acceptor dye. LANCE 10 Ultra kinase assay was used. In presence of JAK 1, JAK 2, JAK 3 and TYK2 kinases and ATP (corresponding to Km), the ULight peptide substrate (LANCE Ulight-JAK-1 (Tyr1023) Peptide, Perkin Elmer, TRF0121) is phosphorylated. It is then captured by Eu-anti-phospho-substrate antibody (LANCE Eu-W1024 Anti-phosphotyrosine (PT66), Perkin Elmer, AD0069), which bring the Eu-chelate donor and ULight acceptor dyes into close proximity. Upon excitation at 320 nm, 15 the Eu-chelate transfers its energy to the ULight dye, resulting in a fluorescent light emission at 665 nm. Compound testing Serial dilutions of compounds in pure DMSO are prepared from 10 mM DMSO stock solutions. Compounds were tested in 384-well plate for 11 consecutive 5-fold dilutions starting 20 from 20 μM highest concentration (20 μM – 2 pM). 200 nL of compound were transferred from mother plate to test plate by using Mosquito (TTP labtech). Assay was performed in 384-well Perkin Elmer test plate in 20 μL assay volume (kinase reaction) and 40 μL total volume (stopping reagent and antibody detection reagents). In 10 μL of substrate solution (peptide + ATP) 30/50/20/10nM of peptide and 20/0.7/0.2/12μM of ATP were added for JAK 1, JAK 2, JAK 3 and 25 TYK2 respectively. 10 μL of enzyme solution was added to kinase reaction at these concentrations: 0.15/0.083/0.025/0.144 ng/μL of JAK 1, JAK 2, JAK 3 and TYK2 respectively. After shaking and 1.5h of incubation at r.t., 20 μL of Stop (10 μL EDTA) and Detection mixture (10 μL Europium-anti-phospho antibody, final: 0.5 nM) were added. Reading was performed after 1h of incubation on a EnVision 2104 reader (Perkin Elmer). 30 Calculation of IC50 data, curves and QC analysis was performed by using Excel tool and GraphPadPrism software, v9. Briefly, individual concentration-effect curves are generated by plotting the logarithm of the tested concentration of tested compounds (X) vs. corresponding percent inhibition values (Y) using least squares (ordinary) fit. Best fit IC50 values are calculated using Log(inhibitor) vs. normalized response - Variable slope equation, where 165 Y=100/(1+10^((LogIC50-X)*HillSlope)). QC criteria parameters (Z’, S:B, R2, HillSlope) were checked for every IC50 curve. Calculation of IC50 data, curves and QC analysis were made using Excel tools and GraphPadPrism software. QC criteria parameters: Z' ^ 0.5, Hill Slope range 0.5 to 5, S:B > 2. 5 Compounds according to the invention showed values in terms of pIC50 higher than 6 with respect to their inhibitory activity on all JAK isoforms corresponding to ^ 1 μM in terms of inhibitory concentration. Most compounds preferably showed values higher than 7.3, even more preferably higher than 8.3, at least with respect to their inhibitory activity on JAK1; corresponding to ^50 nM, even more preferably ^5 nM, in terms of inhibitory concentration. 10 Data for compounds 1-76 are reported in the table hereinbelow Example No JAK1 JAK2 JAK3 TYK2 1 +++ +++ +++ ++ 166 31 +++ +++ +++ +++ 32 +++ +++ +++ +++ 167 75 +++ +++ +++ ++ 76 +++ +++ +++ ++ The compo with respect to their inhibitory activity on JAK1, JAK2, JAK3 and Tyk2 isoforms according to the following classification criterion: + + + : pIC50 ^ 8.3 5 + + : 8.3 > pIC50 ^ 7.3 + : pIC 50 < 7.3 Inhibition of IL-13 induced pSTAT6 in BEAS BEAS-2B human cell line was seeded (100.000 cells/well) and incubated for 48h at 37°C, 5% CO2, 95% humidity. Compounds were added and incubated 30 min followed by IL-13 as10 trigger. After 30 min incubation, cells were lysed and pSTAT6 determined by Fastscan phospho- stat6 (Tyr641) sandwich ELISA kit (cell signaling). Inhibitors were tested at 11 consecutive 5-fold dilutions starting from 10 μM (10 μM – 40 pM) in duplicate. Calculation of IC50 data, curves and QC analysis were made using Excel tools and GraphPadPrism software. QC criteria parameters: Z' ^ 0.35, Hill Slope range 0.5 to 5, S:B > 2. E xample No pIC50 pIC50 pIC50 ( BEAS) Example No ( BEAS)^ Example No ( BEAS)^ 168 E xample No pIC50 pIC50 pIC50 ( BEAS)^ Example No ( BEAS)^ Example No ( BEAS)^ The compounds are classified in the table above in term of potency with respect to their functional activity in BEAS according to the following classification criterion: §§§§ : pIC50 ^ 8.3 5 §§§ : 8.3 > pIC50 ^ 7.3 §§ : 7.3 > pIC50 ^ 6.3 § : pIC50 < 6.3 Where a numerical limit or range is stated herein, the endpoints are included. Also, all values and subranges within a numerical limit or range are specifically included when not explicitly 10 written out. As used herein the words “a” and “an” and the like carry the meaning of “one or more.” Obviously, numerous modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that, within the scope of the appended claims, the invention may be practiced otherwise than as specifically described herein.