Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
GCN2 MODULATING COMPOUNDS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2022/159745
Kind Code:
A1
Abstract:
Provided herein are compounds, compositions, and methods useful for modulating the activity of GCN2 and for treating related conditions, diseases, and disorders (e.g., cancer and neurodegenerative diseases).

Inventors:
RAMURTHY SAVITHRI (US)
MULVIHILL MARK J (US)
SHERBORNE BRADLEY (GB)
RAHEMTULLA BENJAMIN (GB)
TALBOT ERIC P A (GB)
THOMSON CHRISTOPHER G (GB)
Application Number:
PCT/US2022/013382
Publication Date:
July 28, 2022
Filing Date:
January 21, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HIBERCELL INC (US)
International Classes:
C07D401/12; A61K31/444; A61K31/4985; A61K31/5025; A61K31/5383; A61P25/28; A61P35/00; C07D471/04; C07D471/10; C07D487/04; C07D498/04
Domestic Patent References:
WO2008058341A12008-05-22
WO2008083027A12008-07-10
WO2020210828A12020-10-15
Foreign References:
EP0911333A11999-04-28
EP3498693A12019-06-19
Other References:
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 24 April 2019 (2019-04-24), XP002806002, Database accession no. 2305320-72-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 26 September 2014 (2014-09-26), XP002806003, Database accession no. 1626822-52-3
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 14 August 2011 (2011-08-14), XP002806004, Database accession no. 1317317-77-3
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 25 April 2019 (2019-04-25), XP002806005, Database accession no. 2305514-58-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 17 August 2011 (2011-08-17), XP002806006, Database accession no. 1319156-24-5
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 15 August 2011 (2011-08-15), XP002806007, Database accession no. 1317755-58-0
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 August 2011 (2011-08-12), XP002806008, Database accession no. 1316760-97-0
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 August 2011 (2011-08-12), XP002806009, Database accession no. 1316482-72-0
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 August 2011 (2011-08-12), XP002806010, Database accession no. 1316410-55-5
WEK, R.C. ET AL., BIOCHEM. SOC. TRANS., vol. 34, 2006, pages 7 - 11
YE, J. ET AL., EMBO J., vol. 29, no. 12, 2010, pages 2082 - 2096
MUNN, D. H. ET AL., IMMUNITY, vol. 22, 2005, pages 633 - 642
VAN DE VELDE, L-A. ET AL., CELL REPORTS, vol. 17, 2016, pages 2247 - 2258
WEI, C. ET AL., MOL. BIOL. CELL, vol. 26, no. 6, 2015, pages 1044 - 1057
T. W. GREENEP. G. M. WUTS: "Protecting Groups in Organic Synthesis", 1991, WILEY
"Current protocols in molecular biology", 1987
MARTIN: "Remington's Pharmaceutical Sciences", 1975, MACK PUBL. CO.
"Handbook of Pharmaceutical Salts. Properties, Selection and Use", 2002, WILEY-VCH
S. BERGE ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, no. 1, 1977, pages 1 - 19
P. GOULD, INTERNATIONAL J. OF PHARMACEUTICS, vol. 33, 1986, pages 201 - 217
ANDERSON ET AL.: "The Practice of Medicinal Chemistry", 1996, ACADEMIC PRESS
OHTA, K. ET AL., AUTOPHAGY, vol. 6, 2010, pages 345 - 352
WANG ET AL., REDOX BIOLOGY, vol. 17, 2018, pages 25 - 34
PARDOLL, NATURE REVIEWS CANCER, vol. 12, 2012, pages 252 - 264
Attorney, Agent or Firm:
MCCARTY, Catherine (US)
Download PDF:
Claims:
What is claimed is:

1. A compound represented by Formula (la): or a pharmaceutically acceptable salt thereof, wherein:

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R2;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocycyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R3; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl;

L is selected from the group consisting of C1-6alkylene, C1-6alkylene-O-, and C1- 6alkylene-N(RA)-;

R1 is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, C3-6cycloalkyl, cyano, hydroxyl, C1-6alkoxyl, -N(RA)(RB), -N(RA)-C(O)(RB), -(C1- 6alkylene)-N(RA)(RB), -CO2H, -CO2(C1-6alkyl), and -S-(C1-6alkyl), wherein the C1-6alkyl, C1- 6alkoxyl, and -S-(C1-6alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R2 is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, cyano, C1-6alkoxyl, and C3-6cycloalkyl, wherein the C1-6alkyl and C3-6cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R3 is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(RA)(RB), -N(RA)(RB), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl; or two R3 substituents together with the carbon to which they are attached form C3- 6cycloalkyl;

RA is independently, for each occurrence, hydrogen or C1-6alkyl;

RB is independently, for each occurrence, hydrogen or C1-6alkyl; and n = 0, 1, 2, 3, or 4; wherein the compound of Formula (la) is not

2. A compound represented by Formula (la): or a pharmaceutically acceptable salt thereof, wherein:

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R2;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R3; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl;

L is selected from the group consisting of C1-6alkylene, C1-6alkylene-O-, and C1- 6alkylene-N(RA)-;

R1 is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, C3-6cycloalkyl, cyano, hydroxyl, C1-6alkoxyl, -N(RA)(RB), -N(RA)-C(O)(RB), -(C1- 6alkylene)-N(RA)(RB), -CO2H, -CO2(C1-6alkyl), and -S-(C1-6alkyl), wherein the C1-6alkyl, C1- 6alkoxyl, and -S-(C1-6alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R2 is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, cyano, C1-6alkoxyl, and C3-6cycloalkyl, wherein the C1-6alkyl and C3-6cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R3 is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(RA)(RB), -N(RA)(RB), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl; or two R3 substituents together with the carbon to which they are attached form C3- 6cycloalkyl;

RA is independently, for each occurrence, hydrogen or C1-6alkyl;

RB is independently, for each occurrence, hydrogen or C1-6alkyl; and n = 1, 2, 3, or 4.

3. The compound of claim 1 or 2, wherein n is 2.

4. The compound of claim 3, wherein Ri is independently, for each occurrence, selected from the group consisting of halogen, C1-6alkyl, cyano, and C1-6alkoxyl.

5. The compound of claim 3 or 4, wherein Ri is independently, for each occurrence, selected from the group consisting of chloro, fluoro, cyano, CH3, and -O-CH3.

6. The compound of any one of claims 1-5, wherein B is phenylene or pyridinylene, wherein B may be optionally substituted with one, two, or three R2 substituents independently selected from halogen or cyano.

7. The compound of any one of claims 1-6, wherein B is selected from the group consisting * denotes the point of attachment to optionally substituted with one or two R2 substituents independently selected from logen or cyano.

8. The compound of claim 7, wherein B is , wherein B is substituted with one R2 substituent selected from halogen or cyano.

9. The compound of claim 8, wherein R2 is fluoro.

10. The compound of any one of claims 7-9, wherein B is

11. The compound of claim 7, wherein B is

, wherein B is substituted with two independent R2 substituents selected from halogen or cyano.

12. The compound of claim 11, wherein R2 is independently, for each occurrence, selected from the group consisting of chloro, fluoro, and cyano.

13. The compound of any one of claims 7 and 10-12, wherein B is selected from the group

14. The compound of any one of claims 1-13, wherein L is selected from the group consisting of -CH2-CH2-, -C(CH3)(H)-O-, -CH2-O-, -CH2-N(H)-, and -C(CH3)(H)-N(H)-.

15. The compound of any one of claims 1-14, wherein L is selected from the group consisting of -CH2-CH2-, #-C(CH3)(H)-O-##, #-CH2-O-##, #-O-CH2-##, #-CH2-N(H)- #-N(H)- CH2-##, and #-C(CH3)(H)-N(H)- wherein # denotes the point of attachment to

16. A compound represented by Formula (lb): or a pharmaceutically acceptable salt thereof, wherein:

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R3; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl;

L is selected from the group consisting of C1-6alkylene, C1-6alkylene-O-, and C1- 6alkylene-N(RA)-;

R3 is independently, for each occurrence, selected from the group consisting of C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, oxo, phenyl, -C(O)N(RA)(RB), -N(RA)(RB), 6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C1-6alkyl may each individually be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl; or two R3 substituents together with the carbon to which they are attached form C3- ecycloalkyl;

X1 is selected from CR6 and N;

X2 is selected from CH and N;

X3 is selected from CR7 and N;

R4 is selected from halogen and cyano;

R5 is selected from C1-6alkyl and C1-6alkoxyl;

R6 is selected from halogen and cyano;

R7 is halogen; and

RA and RB are independently, for each occurrence, hydrogen or C1-6alkyl.

17. The compound of claim 16, wherein R4 is selected from the group consisting of chloro, fluoro, and cyano.

18. The compound of claim 16 or 17, wherein R5 is CH3 or -O-CH3.

19. The compound of any one of claims 16-18, wherein X1 is CR6; X2 is CH; and X3 is CR7.

20. The compound of claim 19, wherein R6 is selected from the group consisting of chloro, fluoro, and cyano.

21. The compound of claim 19 or 20, wherein R7 is chloro or fluoro.

22. The compound of any one of claims 16-18, wherein X1 is CR6; X2 is N; and X3 is CR7.

23. The compound of claim 22, wherein R6 is chloro or fluoro.

24. The compound of claim 22 or 23, wherein R7 is fluoro.

25. The compound of any one of claims 16-18, wherein X1 is CR6; X2 is CH; and X3 is N.

26. The compound of claim 25, wherein R6 is fluoro.

27. The compound of any one of claims 16-18, wherein X1 is N; X2 is CH; and X3 is CR7.

28. The compound of claim 27, wherein R7 is fluoro.

29. The compound of any one of claims 16-28, wherein L is selected from the group consisting of -CH2-CH2-, -C(CH3)(H)-O-, -CH2-O-, -CH2-N(H)-, and -C(CH3)(H)-N(H)-.

30. The compound of any one of claims 16-29, wherein L is selected from the group consisting of -CH2-CH2-, #-C(CH3)(H)-O-##, #-CH2-O-##, #-O-CH2-##, #-CH2-N(H)- #-N(H)- CH2-##, and #-C(CH3)(H)-N(H)- wherein # denotes the point of attachment to

31. The compound of any one of claims 1-30, wherein C is selected from the group

available carbons by one, two, three, or more R3 substituents independently selected from the group consisting of C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, oxo, phenyl, - C(O)N(RA)(RB), -N(RA)(RB), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl; and

RA and RB are independently, for each occurrence, hydrogen or C1-6alkyl.

32. The compound of any one of claims 1-31, wherein C is selected from the group available carbons by one, two, three, or more independent R3 substituents selected from the group consisting of C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, oxo, phenyl, - C(O)N(RA)(RB), -N(RA)(RB), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl; and

RA and RB are independently, for each occurrence, hydrogen or C1-6alkyl.

33. The compound of any one of claims 1-32, wherein C is selected from the group

34. The compound of any one of claims 1-32, wherein C is selected from the group with one R3 substituent selected from the group consisting of C1-6alkyl, C2-6alkynyl, C3- ecycloalkyl, cyano, C1-6alkoxyl, oxo, phenyl, -C(O)N(RA)(RB), and -N(RA)(RB), wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents and RA and RB are independently, for each occurrence, hydrogen or C1-6alkyl.

35. The compound of any one of claims 1-32 and 34, wherein R3 is selected from the group consisting of cyano, oxo, CH3, CH2CH3, C(H)(CH3)2, -CCH, CF3, CH2CHF2, CF2CH3, CH2CH2F, -C(O)N(H)(CH3), -O-CH3, -NH2, -N(H)(CH3), cyclopropyl, and phenyl.

36. The compound of any one of claims 1-32, 34, and 35, wherein C is selected from the

37. The compound of any one of claims 1-32, wherein C is wherein C is substituted with two independent R3 substituents selected from the group consisting of C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, oxo, phenyl, - C(O)N(RA)(RB), -N(RA)(RB), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C1-6alkyl; and

RA and RB are independently, for each occurrence, hydrogen or C1-6alkyl.

38. The compound of any one of claims 1-32 and 37, wherein R3 is independently, for each occurrence, selected from the group consisting of CH3, -CH(CH3)2, cyclopropyl, cyclopentyl,

39. The compound of any one of claims 1-32, 37, and 38, wherein R3 is independently, for each occurrence, selected from the group consisting of CH3, -CH(CH3)2, cyclopropyl,

40. The compound of any one of claims 1-32 and 37-39, wherein C is selected from the

41. The compound of any one of claims 1-32, wherein C is wherein C is substituted with three independent R3 substituents and wherein two of the R3 substituents together with the carbon to which they are attached form C3-6cycloalkyl and the third R3 group is selected from the group consisting of C1-6alkyl, C2-6alkynyl, C3-6cycloalkyl, cyano, C1-6alkoxyl, oxo, phenyl, -C(O)N(RA)(RB), and -N(RA)(RB), wherein the C1-6alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents and RA and RB are independently hydrogen or C1-6alkyl.

42. The compound of any one of claims 1-32, and 41, C is wherein C is substituted with three independent R3, and wherein two of the R3 substituents together with the carbon to which they are attached form cyclopropyl and the third R3 group is oxo.

43. The compound of any one of claims 1-32, 41, and 42, wherein C is

44. The compound of any one of claims 1-32, wherein C is selected from the group

45. A compound of selected from any compound set forth in Table 1, or a pharmaceutically acceptable salt thereof.

46. A pharmaceutical composition comprising a compound of any one of claims 1-45 and a pharmaceutically acceptable carrier.

47. A method of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-45 or a pharmaceutical composition of claim 46.

48. The method of claim 47, wherein the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, sweat gland carcinoma, sebaceous gland carcinoma, thyroid cancer, kidney cancer, uterus cancer, esophagus cancer, liver cancer, head cancer, neck cancer, throat cancer, mouth cancer, bone cancer, chest cancer, lymph node cancer, eye cancer, mesothelioma, an acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, retinoblastoma, leukemia, lymphoma, or any combination thereof.

49. The method of claim 47, wherein the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, leukemia, or lymphoma.

50. A method of treating a neurodegenerative disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-45 or a pharmaceutical composition of claim 46.

51. The method of claim 50, wherein the neurodegenerative disease is Alzheimer's disease, Parkinson's Disease, Huntington's Disease, amyotrophic lateral sclerosis, or spinocerebellar ataxia.

52. A method of treating doxorubicin-induced cardiotoxicity in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1-45 or a pharmaceutical composition of claim 46.

53. The method of any one of claims 47-52, wherein the subject is a human.

54. A method of modulating the activity of GCN2, comprising exposing GCN2 to an effective amount of a compound of any one of claims 1-45 or a pharmaceutical composition of claim 46 to modulate the activity of said GCN2.

Description:
GCN2 MODULATING COMPOUNDSAND USES THEREOF

CROSS REFERENCE TO RELATED APPLICATION

[001] This application claims priority to U.S. Provisional Application No. 63/140,311 filed January 22, 2021, which is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

[002] Cancer continues to be a significant health problem despite the substantial research efforts and scientific advances reported in the literature for treating this disease. Some of the most frequently diagnosed cancers include prostate cancer, breast cancer, and lung cancer. Prostate cancer is the most common form of cancer in men. Breast cancer remains a leading cause of death in women. Current treatment options for these cancers are not effective for all patients and/or can have substantial adverse side effects. New therapies are needed to address this unmet need in cancer therapy.

[003] General control nonderepressible kinase 2 (GCN2) is a serine/threonine protein kinase that phosphorylates the a subunit of eukaryotic initiation factor 2 (eIF2a) in response to amino acid deficiency (see, for example, Wek, R.C. et al. in Biochem. Soc. Trans. 2006, 34(Pt 1), p. 7-11). Expression and activation of GCN2 have been shown to be elevated in human and mouse tumors, and reduction in the expression of GCN2 has been shown to inhibit tumor growth (see e.g., Ye, J. et al. in EMBO J. 2010, 29(12), p. 2082-2096). Tumors grow in an environment of amino acid deficiency which can be further depleted with chemotherapy inducing a dependence on autophagy which requires GCN2 activity. In addition, GCN2 mediates the induction of anergy in T cells in response to tryptophan depletion by indoleamine 2,3-dioxygenase (IDO) in the tumor microenvironment (Munn, D. H. et al in Immunity 2005, 22, p. 633-642) and is essential for the proliferative fitness of cytotoxic T cells in amino acid limiting environments (Van de Velde, L-A., et al. in Cell Reports 2016, 17, p. 2247-2258). Inhibition of GCN2 has been reported as a therapeutic approach for cancer therapy (see, e.g., Wei, C. et al. inMol. Biol. Cell. 2015, 26(6), p. 1044-1057). Accordingly, compounds having modulatory activity towards GCN2 are needed as therapeutic agents for treating cancer, with additional applications in the treatment of neurodegenerative diseases and doxorubicin-induced cardiotoxicity. SUMMARY OF THE INVENTION

[004] Provided herein are compounds and compositions for the modulation of GCN2 (e.g., the activation or inhibition of GCN2). In various embodiments, the compounds and compositions described herein are useful for the treatment of GCN2 mediated conditions, diseases, or disorders (e.g., cancers and neurodegenerative diseases).

[005] In one aspect, provided herein is a compound represented by Formula (la): or a pharmaceutically acceptable salt thereof, wherein:

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocycyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 3-6 cycloalkyl, cyano, hydroxyl, C 1-6 alkoxyl, -N(R A )(R B ), -N(R A )-C(O)(R B ), -(C 1-6 alkylene)-N(R A )(R B ), -CO 2 H, -CO 2 (C 1-6 alkyl), and -S-(C 1-6 alkyl), wherein the C 1-6 alkyl, C 1- 6 alkoxyl, and -S-( C 1-6 alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R 2 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, C 1-6 alkoxyl, and C 3-6 cycloalkyl, wherein the C 1-6 alkyl and C 3-6 cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents; R 3 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

R A is independently, for each occurrence, hydrogen or C 1-6 alkyl;

R B is independently, for each occurrence, hydrogen or C 1-6 alkyl; and n = 0, 1, 2, 3, or 4; wherein the compound of Formula (la) is not

[006] In another aspect, provided herein is a compound represented by Formula (la): or a pharmaceutically acceptable salt thereof, wherein:

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 3-6 cycloalkyl, cyano, hydroxyl, C 1-6 alkoxyl, -N(R A )(R B ), -N(R A )-C(O)(R B ), -(C 1- 6 alkylene)-N(R A )(R B ), -CO 2 H, -CO2(C 1-6 alkyl), and -S-(C 1-6 alkyl), wherein the C 1-6 alkyl, C 1- 6 alkoxyl, and -S-(C 1-6 alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R 2 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, C 1-6 alkoxyl, and C 3-6 cycloalkyl, wherein the C 1-6 alkyl and C 3-6 cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 3 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

R A is independently, for each occurrence, hydrogen or C 1-6 alkyl;

R B is independently, for each occurrence, hydrogen or C 1-6 alkyl; and n = 1, 2, 3, or 4.

[007] In another aspect, provided herein is a compound represented by Formula (lb): or a pharmaceutically acceptable salt thereof, wherein:

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 3 is independently, for each occurrence, selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may each individually be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

X 1 is selected from CR 6 and N;

X 2 is selected from CH and N;

X 3 is selected from CR 7 and N;

R 4 is selected from halogen and cyano;

R 5 is selected from C 1-6 alkyl and C 1-6 alkoxyl;

R 6 is selected from halogen and cyano;

R 7 is halogen; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[008] In some embodiments, a compound provided herein is selected from a compound set forth in Table 1, or a pharmaceutically acceptable salt thereof.

[009] In another aspect, provided herein is a pharmaceutical composition comprising a compound described herein and a pharmaceutically acceptable carrier.

[010] In another aspect, provided herein are methods of treating cancer in a subject in need thereof, the method generally comprises administering to the subject a therapeutically effective amount of a compound described herein. In some embodiments, the cancer is selected from the group consisting of colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, sweat gland carcinoma, sebaceous gland carcinoma, thyroid cancer, kidney cancer, uterus cancer, esophagus cancer, liver cancer, head cancer, neck cancer, throat cancer, mouth cancer, bone cancer, chest cancer, lymph node cancer, eye cancer, mesothelioma, an acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, retinoblastoma, leukemia, lymphoma, and a combination thereof. In some embodiments, the cancer is selected from the group consisting of colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, leukemia, lymphoma, and a combination thereof.

[011] In another aspect, provided herein are methods of treating a neurodegenerative disease in a subject in need thereof, the method generally comprises administering to the subject a therapeutically effective amount of a compound described herein. In some embodiments, the neurodegenerative disease is selected from the group consisting of Alzheimer's disease, Parkinson's Disease, Huntington's Disease, amyotrophic lateral sclerosis, and spinocerebellar ataxia.

[012] In another aspect, provided herein are methods of modulating (both activation and/or inhibition) the activity of GCN2, the method generally comprises exposing GCN2 to an effective amount of a compound described herein to modulate the activity of said GCN2.

DETAILED DESCRIPTION OF THE INVENTION

[015] The invention provides GCN2-interacting compounds and related compounds, pharmaceutical compositions, and their use in the treatment of medical conditions, such as cancer, neurodegenerative diseases, and doxorubicin-induced cardiotoxicity, and in modulating (inhibiting/activating) GCN2 activity. The practice of the present invention employs, unless otherwise indicated, conventional techniques of organic chemistry, pharmacology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology. Such techniques are explained in the literature, such as in "Comprehensive Organic Synthesis" (B.M. Trost & I. Fleming, eds., 1991-1992); "Handbook of experimental immunology" (D .M. Weir & C.C. Blackwell, eds.); "Current protocols in molecular biology" (F.M. Ausubel et al., eds., 1987, and periodic updates); and "Current protocols in immunology" (J.E. Coligan et al., eds., 1991), each of which is herein incorporated by reference in its entirety.

[016] Various aspects of the invention are set forth below in sections; however, aspects of the invention described in one particular section are not to be limited to any particular section. Further, when a variable is not accompanied by a definition, the previous definition of the variable controls.

Definitions

[017] The terms used herein have their ordinary meaning and the meaning of such terms is independent at each occurrence thereof. That notwithstanding and except where stated otherwise, the following definitions apply throughout the specification and claims. Chemical names, common names, and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name, and an ambiguity exists between the structure and the name, the structure predominates. These definitions apply regardless of whether a term is used by itself or in combination with other terms, unless otherwise indicated. Hence, the definition of "alkyl" applies to "alkyl" as well as the "alkyl" portions of "-O-alkyl" etc.

[018] The term "alkyl" refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-12, 1-10, or 1-6 carbon atoms, referred to herein as C 1 -C 12 alkyl, C 1 -C 10 alkyl, and C 1- C 6 alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l-propyl, 2-methyl-2-propyl, 2- methyl-1 -butyl, 3 -methyl -1 -butyl, 2-methyl-3 -butyl, 2,2-dimethyl-l-propyl, 2-methyl-l- pentyl, 3-methyl-l-pentyl, 4-methyl-l-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4- methyl-2-pentyl, 2,2-dimethyl-l -butyl, 3, 3 -dimethyl- 1 -butyl, 2-ethyl-l-butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc.

[019] The term "alkylene" refers to a diradical of an alkyl group. Exemplary alkylene groups include -CH 2 -, -CH 2 CH 2 -, and -CH 2 C(H)(CH 3 )CH 2 -. The term "-(C 0 alkylene)-" refers to a bond. Accordingly, the term "-(C 0-3 alkylene)-" encompasses a bond (i.e., C 0 ) and a -(C 1-3 alkylene) group.

[020] As used herein, "carbocyclyl" or "carbocyclic" refers to a radical of a non-aromatic cyclic hydrocarbon group having from 3 to 10 ring carbon atoms ("C3-10 carbocyclyl") and zero heteroatoms in the non-aromatic ring system. In certain embodiments, a carbocyclyl group has 3 to 8 ring carbon atoms. In certain embodiments, a carbocyclyl group has 3 to 7 ring carbon atoms ("C3-7 carbocycyl"). In certain embodiments, a carbocyclyl group has 3 to 6 ring carbon atoms ("C3-6 carbocyclyl"). In certain embodiments, a carbocyclyl group has 5 to 10 ring carbon atoms ("C5-10 carbocyclyl"). In certain embodiments, a carbocyclyl group has 5 to 10 ring carbon atoms ("C7-10 carbocyclyl"). Exemplary C3-6 carbocyclyl groups include, without limitation, cyclopropyl (C3), cyclobutyl (C4), cyclobutenyl (C4), cyclopentyl (C5), cyclopentenyl (C5), cyclohexyl (C6), cyclohexenyl (C6), cyclohexadienyl (C6), and the like. Exemplary C3-8 carbocyclyl groups include, without limitation, the aforementioned C3-6 carbocyclyl groups as well as cycloheptyl (C7), cycloheptenyl (C7), cycloheptadienyl (C7), cycloheptatrienyl (C7), cyclooctyl (C8), cyclooctenyl (C8), bicyclo[2.2.1]heptanyl (C7), bicyclo[2.2.2]octanyl (C8), and the like. Exemplary C3-10 carbocyclyl groups include, without limitation, the aforementioned C3-8 carbocyclyl groups as well as cyclononyl (C9), cyclononenyl (C9), cyclodecyl (CIO), cyclodecenyl (CIO), octahydro- IH-indenyl (C9), decahydronaphthalenyl (CIO), spiro[4.5]decanyl (CIO), and the like. As the foregoing examples illustrate, in certain embodiments, the carbocyclyl group is either monocyclic ("monocyclic carbocyclyl") or contain a fused, bridged or spiro ring system such as a bicyclic system ("bicyclic carbocyclyl") and can be saturated or partially unsaturated.

[021] The term "cycloalkyl" refers to a monovalent saturated cyclic, bicyclic, or bridged cyclic (e.g., adamantyl) hydrocarbon group of 3-12, 3-8, 4-8, or 4-6 carbons, referred to herein, e.g., as "C 3 -C 6 cycloalkyl," derived from a cycloalkane. Exemplary cycloalkyl groups include cyclohexyl, cyclopentyl, cyclobutyl, and cyclopropyl. The term "halocycloalkyl" refers to a cycloalkyl group that is substituted with at least one halogen.

[022] The term "cycloalkylene" refers to a diradical of a cycloalkyl group. Exemplary cycloalkylene groups include

[023] The term "haloalkyl" refers to an alkyl group that is substituted with at least one halogen. Exemplary haloalkyl groups include -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CF 3 , -CF 2 CF 3 , and the like.

[024] The term "hydroxyalkyl" refers to an alkyl group that is substituted with at least one hydroxyl. Exemplary hydroxyalkyl groups include -CH 2 CH 2 OH, - C(H)(OH)CH 3 , -CH 2 C(H)(OH)CH 2 CH 2 OH, and the like.

[025] The term "hydroxy fluoroalkyl" refers to a hydroxy alkyl that is substituted with at least one fluoro.

[026] The term "aralkyl" refers to an alkyl group substituted with an aryl group. Exemplary aralkyl groups include

[027] The term "heteroaralkyl" refers to an alkyl group substituted with a heteroaryl group. [028] The terms "alkenyl" and "alkynyl" are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.

[029] The term "cycloalkenyl" refers to a monovalent unsaturated cyclic, bicyclic, or bridged (e.g., adamantyl) carbocyclic hydrocarbon containing at least one C-C double bond. In certain embodiments, the cycloalkenyl contains 5-10, 5-8, or 5-6 carbons, referred to herein, e.g., as "C 5 -C 6 cycloalkenyl". Exemplary cycloalkenyl groups include cyclohexenyl and cyclopentenyl.

[030] The term "aryl" is art-recognized and refers to a carbocyclic aromatic group. Representative aryl groups include phenyl, naphthyl, anthracenyl, and the like. Unless specified otherwise, the aromatic ring may be substituted at one or more ring positions with, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, carboxylic acid, -C(O)alkyl, -CO 2 alkyl, carbonyl, carboxyl, alkylthio, sulfonyl, sulfonamido, sulfonamide, ketone, aldehyde, ester, heterocyclyl, aryl or heteroaryl moieties, -CF 3 , -CN, or the like. The term "aryl" also includes polycyclic aromatic ring systems having two or more carbocyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein all of the fused rings are aromatic rings, e.g., in a naphthyl group.

[031] The term "phenylene" refers to a diradical of a phenyl group. Exemplary phenylene groups include and

[032] The term "heteroaryl" refers to a radical of a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system (e.g., having 6 or 10 n electrons shared in a cyclic array) having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur ("5-10 membered heteroaryl"). In heteroaryl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems can include one or more heteroatoms in one or both rings. "Heteroaryl" also includes ring systems wherein the heteroaryl ring, as defined above, is fused with one or more aryl groups wherein the point of attachment is either on the aryl or heteroaryl ring, and in such instances, the number of ring members designates the number of ring members in the fused (aryl/heteroaryl) ring system. Bicyclic heteroaryl groups wherein one ring does not contain a heteroatom (e.g., indolyl, quinolinyl, carbazolyl, and the like) the point of attachment can be on either ring, i.e., either the ring bearing a heteroatom (e.g., 2-indolyl) or the ring that does not contain a heteroatom (e.g., 5 -indolyl). A heteroaryl group may be described as, e.g., a 6-10-membered heteroaryl, wherein the term "membered" refers to the non-hydrogen ring atoms within the moiety.

[033] In some embodiments, a heteroaryl group is a 5-10 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-10 membered heteroaryl"). In some embodiments, a heteroaryl group is a 5-8 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-8 membered heteroaryl"). In some embodiments, a heteroaryl group is a 5-6 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-6 membered heteroaryl"). In some embodiments, the 5- 6 membered heteroaryl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1 ring heteroatom selected from nitrogen, oxygen, and sulfur. Each instance of a heteroaryl group may be independently optionally substituted, i.e., unsubstituted (an "unsubstituted heteroaryl") or substituted (a "substituted heteroaryl") with one or more substituents. In certain embodiments, the heteroaryl group is unsubstituted 5-14 membered heteroaryl. In certain embodiments, the heteroaryl group is substituted 5-14 membered heteroaryl.

[034] Exemplary 5-membered heteroaryl groups containing one heteroatom include, without limitation, pyrrolyl, furanyl and thiophenyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, without limitation, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, without limitation, triazolyl, oxadiazolyl, and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, without limitation, tetrazolyl. Exemplary 6-membered heteroaryl groups containing one heteroatom include, without limitation, pyridinyl. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, without limitation, pyridazinyl, pyrimidinyl, and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, without limitation, triazinyl and tetrazinyl, respectively. Exemplary 7-membered heteroaryl groups containing one heteroatom include, without limitation, azepinyl, oxepinyl, and thiepinyl. Exemplary 5,6-bicyclic heteroaryl groups include, without limitation, indolyl, isoindolyl, indazolyl, benzotri azolyl, benzothiophenyl, isobenzothiophenyl, benzofuranyl, benzoisofuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzthiazolyl, benzisothiazolyl, benzthiadi azolyl, indolizinyl, and purinyl. Exemplary 6,6- bicyclic heteroaryl groups include, without limitation, naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl.

[035] The term "heteroarylene" refers to a diradical of a heteroaryl group. Exemplary heteroarylene groups include: phenylene, pyridinylene, pyridazinylene, pyrimidinylene, pyrazinylene,

[036] The terms ortho, meta, and para are art-recognized and refer to 1,2-, 1,3- and 1,4- disubstituted benzenes, respectively. For example, the names 1,2-dimethylbenzene and or/Ao-dimethylbenzene are synonymous.

[037] The term "heterocyclyl" or "heterocyclic" refers to a radical of a 3- to 10-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon ("3-10 membered heterocyclyl"). In heterocyclyl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits. A heterocyclyl group can either be monocyclic ("monocyclic heterocyclyl") or a fused, bridged or spiro ring system such as a bicyclic system ("bicyclic heterocyclyl"), and can be saturated or can be partially unsaturated. Heterocyclyl bicyclic ring systems can include one or more heteroatoms in one or both rings. "Heterocyclyl" also includes ring systems wherein the heterocyclyl ring, as defined above, is fused with one or more cycloalkyl groups wherein the point of attachment is either on the cycloalkyl or heterocyclyl ring, or ring systems wherein the heterocyclyl ring, as defined above, is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring, and in such instances, the number of ring members continue to designate the number of ring members in the heterocyclyl ring system. A heterocyclyl group may be described as, e.g., a 3-7-membered heterocyclyl, wherein the term "membered" refers to the non-hydrogen ring atoms, i.e., carbon, nitrogen, oxygen, sulfur, boron, phosphorus, and silicon, within the moiety. Each instance of heterocyclyl may be independently optionally substituted, i.e., unsubstituted (an "unsubstituted heterocyclyl") or substituted (a "substituted heterocyclyl") with one or more substituents. In certain embodiments, the heterocyclyl group is unsubstituted 3-10 membered heterocyclyl. In certain embodiments, the heterocyclyl group is substituted 3-10 membered heterocyclyl.

[038] In some embodiments, a heterocyclyl group is a 5-10 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon ("5-10 membered heterocyclyl"). In some embodiments, a heterocyclyl group is a 5-8 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-8 membered heterocyclyl"). In some embodiments, a heterocyclyl group is a 5-6 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-6 membered heterocyclyl"). In some embodiments, the 5-6 membered heterocyclyl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heterocyclyl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heterocyclyl has one ring heteroatom selected from nitrogen, oxygen, and sulfur. [039] Exemplary 3 -membered heterocyclyl groups containing one heteroatom include, without limitation, azirdinyl, oxiranyl, thiorenyl. Exemplary 4-membered heterocyclyl groups containing one heteroatom include, without limitation, azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclyl groups containing one heteroatom include, without limitation, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothiophenyl, pyrrolidinyl, dihydropyrrolyl and pyrrolyl-2, 5-dione. Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, without limitation, dioxolanyl, oxasulfuranyl, disulfuranyl, and oxazolidin-2-one. Exemplary 5-membered heterocyclyl groups containing three heteroatoms include, without limitation, triazolinyl, oxadiazolinyl, and thiadiazolinyl. Exemplary 6-membered heterocyclyl groups containing one heteroatom include, without limitation, piperidinyl, tetrahydropyranyl, dihydropyridinyl, and thianyl. Exemplary 6- membered heterocyclyl groups containing two heteroatoms include, without limitation, piperazinyl, morpholinyl, dithianyl, dioxanyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, without limitation, triazinanyl. Exemplary 7-membered heterocyclyl groups containing one heteroatom include, without limitation, azepanyl, oxepanyl and thiepanyl. Exemplary 8-membered heterocyclyl groups containing one heteroatom include, without limitation, azocanyl, oxecanyl and thiocanyl. Exemplary 5- membered heterocyclyl groups fused to a C6 aryl ring (also referred to herein as a 5,6- bicyclic heterocyclic ring) include, without limitation, indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinonyl, and the like. Exemplary 6- membered heterocyclyl groups fused to an aryl ring (also referred to herein as a 6,6-bicyclic heterocyclic ring) include, without limitation, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.

[040] The term "heterocycloalkyl" refers to a saturated heterocyclyl group having, for example, 3-7 ring atoms selected from carbon and heteroatoms (e.g., O, N, or S).

[041] The terms "amine" and "amino" are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formulas: wherein R 50 , R 51 , R 52 and R 53 each independently represent a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R 61 , or R 50 and R 51 , taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R 61 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a poly cycle; and m is zero or an integer in the range of 1 to 8. In certain embodiments, only one of R 50 or R 51 may be a carbonyl, e.g., R 50 , R 51 and the nitrogen together do not form an imide. In other embodiments, R 50 and R 51 (and optionally R 52 ) each independently represent a hydrogen, an alkyl, an alkenyl, or - (CH 2 ) m -R 61 .

[042] The terms "alkoxyl" or "alkoxy" are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An "ether" is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as may be represented by one of - O-alkyl, -O-alkenyl, -O-alkynyl, and -O-(CH 2 ) m -R 61 , where m and R 61 are described above. [043] The term "fluoroalkoxyl" refers to an alkoxyl group that is substituted with at least one fluoro group. Exemplary fluoroalkoxyl groups include -OCH 2 F, -OCHF2, -OCF 3 , - OCH 2 CF 3 , -OCF2CF 3 , and the like.

[044] The term "oxo" is art-recognized and refers to a "=O" substituent. For example, a cyclopentane substituted with an oxo group is cyclopentanone.

[045] The symbols " ", and indicate a point of attachment.

[046] The term "substituted" means that one or more hydrogens on the atoms of the designated group are replaced with a selection from the indicated group, provided that the atoms' normal valences under the existing circumstances are not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. The terms "stable compound" or "stable structure" refer to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.

[047] When any substituent or variable occurs more than one time in any constituent or the compound of the invention, its definition on each occurrence is independent of its definition at every other occurrence, unless otherwise indicated.

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

[049] One or more compounds of the invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. "Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H 2 O.

[050] Certain compounds contained in compositions of the present invention may exist in particular geometric or stereoisomeric forms. Further, certain compounds described herein may be optically active. The present invention contemplates all such compounds, including cis- and trans-i somers, R- and 5-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. The compounds may contain one or more stereogenic centers. For example, asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention, such as, for example, racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers, and it is intended that all of the possible optical isomers, diastereomers in mixtures, and pure or partially purified compounds are included within the ambit of this invention.

[051] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Alternatively, a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis. Still further, where the molecule contains a basic functional group (such as amino) or an acidic functional group (such as carboxylic acid) diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means known in the art, and subsequent recovery of the pure enantiomers.

[052] Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. Chiral center(s) in a compound of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. Further, to the extent a compound described herein may exist as an atropisomer (e.g., substituted biaryls), all forms of such atropisomer are considered part of this invention.

[053] As used herein, the terms "subject" and "patient" are used interchangeable and refer to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans.

[054] The term "IC 50 " is art-recognized and refers to the concentration of a compound that is required to achieve 50% inhibition of the target.

[055] As used herein, the term "effective amount" refers to the amount of a compound sufficient to effect beneficial or desired results (e.g., a therapeutic, ameliorative, inhibitory or preventative result). An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. As used herein, the term "treating" includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.

[056] As used herein, the term "pharmaceutical composition" refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo.

[057] As used herein, the term "pharmaceutically acceptable carrier" refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see e.g., Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975],

[058] As used herein, the term "pharmaceutically acceptable salt" refers to any pharmaceutically acceptable salt (e.g., acid or base) of a compound of the present invention which, upon administration to a subject, is capable of providing a compound of this invention or an active metabolite or residue thereof. As is known to those of skill in the art, "salts" of the compounds of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p- sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic, benzenesulfonic acid, and the like. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.

[059] Examples of bases include, but are not limited to, alkali metals (e.g., sodium) hydroxides, alkaline earth metals (e.g., magnesium), hydroxides, ammonia, and compounds of formula NW 3 , wherein W is C 1-4 alkyl, and the like.

[060] Examples of salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, fumarate, flucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate (also known as toluenesulfonate), undecanoate, and the like. Other examples of salts include anions of the compounds of the present invention compounded with a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a C 1-4 alkyl group), and the like. Further examples of salts include, but are not limited to, ascorbate, borate, nitrate, phosphate, salicylate, and sulfate. Further, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al., Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al., Journal of Pharmaceutical Sciences (1977) 66(1) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33 201-217; Anderson el al., The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference.

[061] Additional exemplary basic salts include, but are not limited to: ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen-containing groups may be quartemized with agents such as lower alkyl halides (e.g., methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g., decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenethyl bromides), and others.

[062] For therapeutic use, salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non- pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.

[063] In addition, when a compound of the invention contains both a basic moiety (such as, but not limited to, a pyridine or imidazole) and an acidic moiety (such as, but not limited to, a carboxylic acid) zwitterions ("inner salts") may be formed. Such acidic and basic salts used within the scope of the invention are pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts. Such salts of the compounds of the invention may be formed, for example, by reacting a compound of the invention with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.

[064] The present invention includes the compounds of the invention in all their isolated forms (such as any solvates, hydrates, stereoisomers, and tautomers thereof). Further, the invention includes compounds in which one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of the invention. For example, different isotopic forms of hydrogen (H) include protium ( 1 H) and deuterium ( 2 H). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds can be prepared without undue experimentation by conventional techniques known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.

[065] Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.

[066] The terms "a" and "an" as used herein mean "one or more" and include the plural unless the context is inappropriate.

[067] As a general matter, compositions specifying a percentage are by weight unless otherwise specified.

Compounds

[068] In one aspect, provided herein are compounds represented by Formula (I): or a pharmaceutically acceptable salt thereof, wherein:

A is phenyl or 5-6 membered heteroaryl, wherein the phenyl or the 5-6 membered heteroaryl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 1 ;

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 3-6 cycloalkyl, cyano, hydroxyl, C 1-6 alkoxyl, -N(R A )(R B ), -N(R A )-C(O)(R B ), -(C 1- 6 alkylene)-N(R A )(R B ), -CO 2 H, -CO 2 (C 1-6 alkyl), and -S-(C 1-6 alkyl), wherein the C 1-6 alkyl, C 1- 6 alkoxyl, and -S-(C 1-6 alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R 2 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, C 1-6 alkoxyl, and C 3-6 cycloalkyl, wherein the C 1-6 alkyl and C 3-6 cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 3 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- ecycloalkyl;

R A is independently, for each occurrence, hydrogen or C 1-6 alkyl; and

R B is independently, for each occurrence, selected from the group consisting of hydrogen, C 1-6 alkyl, and C 3-6 cycloalkyl; wherein the compound of Formula (I) is not

[069] In another aspect, provided herein are compounds represented by Formula (I): or a pharmaceutically acceptable salt thereof, wherein:

A is phenyl or 5-6 membered heteroaryl, wherein the phenyl or the 5-6 membered heteroaryl is substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 1 ;

B is phenylene or 5-6 membered heteroarylene, wherein the phenyl or the 5-6 membered heteroarylene is substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl, wherein the C 1-6 alkyl may optionally be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 2 is independently, for each occurrence, halogen or cyano;

R 3 is independently, for each occurrence, selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may each individually be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl. [070] In some embodiments, A is phenyl or pyridinyl, wherein A is substituted with one, two, or three independent R 1 substituents selected from the group consisting of halogen, C 1- 6 alkyl, cyano, and C 1-6 alkoxyl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents. [071] In some embodiments, A is phenyl or pyridinyl, wherein A is substituted with two R 1 substituents selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents. In some embodiments, A is phenyl or , wherein A is substituted with two independent Ri substituents selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents.

[072] In some embodiments, Ri is independently, for each occurrence, selected from the group consisting of chloro, fluoro, cyano, CH 3 , CF 3 , and -O-CH 3 .

[073] In some embodiments, A is selected from the group consisting of

[074] In another aspect, provided herein are compounds represented by Formula (I): or a pharmaceutically acceptable salt thereof, wherein:

A is pyridinyl, wherein the pyridinyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 1 ;

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 3-6 cycloalkyl, cyano, hydroxyl, C 1-6 alkoxyl, -N(R A )(R B ), -N(R A )-C(O)(R B ), -(C 1- 6 alkylene)-N(R A )(R B ), -CO 2 H, -CO 2 (C 1-6 alkyl), and -S-(C 1-6 alkyl), wherein the C 1-6 alkyl, C 1- 6 alkoxyl, and -S-(C 1-6 alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R 2 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, C 1-6 alkoxyl, and C 3-6 cycloalkyl, wherein the C 1-6 alkyl and C 3-6 cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 3 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

R A is independently, for each occurrence, hydrogen or C 1-6 alkyl; and

R B is independently, for each occurrence, selected from the group consisting of hydrogen, C 1-6 alkyl, and C 3-6 cycloalkyl; wherein the compound of Formula (I) is not

[075] In another aspect, provided herein are compounds represented by Formula (I): or a pharmaceutically acceptable salt thereof, wherein:

A is pyridinyl, wherein the pyridinyl is substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 1 ;

B is phenylene or 5-6 membered heteroarylene, wherein the phenyl or the 5-6 membered heteroarylene is substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl, wherein the C 1-6 alkyl may optionally be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 2 is independently, for each occurrence, halogen or cyano;

R 3 is independently, for each occurrence, selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may each individually be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[076] In some embodiments, A is pyridinyl, wherein the pyridinyl is substituted with two R 1 substituents selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl.

[077] In some embodiments, , wherein A is substituted with two independent Ri substituents selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl.

[078] In some embodiments, Ri is independently, for each occurrence, selected from the group consisting of chloro, fluoro, cyano, CH 3 , and -O- CH 3 .

wherein B may be optionally substituted with one or two independent R 2 substituents selected from halogen or cyano. In some embodiments, B is , h two independent R 2 substituents selected from halogen or cyano. In some embodiments, R 2 is independently, for each occurrence, selected from the group consisting of chloro, fluoro, and cyano. In some embodiments, B is selected from the group consisting of

[081] In some embodiments, L is selected from the group consisting of -CH 2 -CH 2 -, - C(CH 3 )(H)-O-, -CH 2 -O-, -CH 2 -N(H)-, and -C(CH 3 )(H)-N(H)-. In some embodiments, L is selected from the group consisting of -CH 2 -CH 2 -, # -C(CH 3 )(H)-O- ## , # -CH 2 -O- ## , # -O-CH 2 - ## , # -CH 2 -N(H)- # -N(H)-CH 2 - ## , and # -C(CH 3 )(H)-N(H)- wherein # denotes the point of attachment to and ## denotes the point of

[082] In another aspect, provided herein are compounds represented by Formula (la): or a pharmaceutically acceptable salt thereof, wherein:

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocycyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 3-6 cycloalkyl, cyano, hydroxyl, C 1-6 alkoxyl, -N(R A )(R B ), -N(R A )-C(O)(R B ), -(C 1- 6 alkylene)-N(R A )(R B ), -CO 2 H, -CO 2 (C 1-6 alkyl), and -S-(C 1-6 alkyl), wherein the C 1-6 alkyl, C 1- 6 alkoxyl, and -S-(C 1-6 alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R 2 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, C 1-6 alkoxyl, and C 3-6 cycloalkyl, wherein the C 1-6 alkyl and C 3-6 cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 3 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

R A is independently, for each occurrence, hydrogen or C 1-6 alkyl;

R B is independently, for each occurrence, hydrogen or C 1-6 alkyl; and n = 0, 1, 2, 3, or 4; wherein the compound of Formula (la) is not

[083] In another aspect, provided herein are compounds represented by Formula (la) or a pharmaceutically acceptable salt thereof, wherein:

B is phenylene or 5-6 membered heteroarylene, wherein the phenylene or the 5-6 membered heteroarylene may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 2 ;

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 3-6 cycloalkyl, cyano, hydroxyl, C 1-6 alkoxyl, -N(R A )(R B ), -N(R A )-C(O)(R B ), -(C 1- 6 alkylene)-N(R A )(R B ), -CO 2 H, -CO 2 (C 1-6 alkyl), and -S-(C 1-6 alkyl), wherein the C 1-6 alkyl, C 1- 6 alkoxyl, and -S-(C 1-6 alkyl) may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and hydroxyl;

R 2 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, C 1-6 alkoxyl, and C 3-6 cycloalkyl, wherein the C 1-6 alkyl and C 3-6 cycloalkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents;

R 3 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, hydroxyl, oxo, phenyl, - C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

R A is independently, for each occurrence, hydrogen or C 1-6 alkyl;

R B is independently, for each occurrence, hydrogen or C 1-6 alkyl; and n = 1, 2, 3, or 4.

[084] In some embodiments, n is 2. In some embodiments, R 1 is independently, for each occurrence, selected from the group consisting of halogen, C 1-6 alkyl, cyano, and C 1-6 alkoxyl. In some embodiments, Ri is independently, for each occurrence, selected from the group consisting of chloro, fluoro, cyano, CH 3 , and -O-CH 3 .

[085] In some embodiments, B is phenylene or pyridinylene, wherein B may be optionally substituted with one, two, or three R 2 substituents independently selected from halogen or cyano. In some embodiments, B is selected from the group consisting of , wherein * denotes the point of attachment to

substituents independently selected from halogen or cyano. In some embodiments, B is , wherein B is substituted with one

R 2 substituent selected from halogen or cyano. In some embodiments, R 2 is fluoro. In some embodiments, . In some embodiments, The compound of claim 7, wherein B is , wherein B is substituted with two independent R 2 substituents selected from halogen or cyano. In some embodiments, R 2 is independently, for each occurrence, selected from the group consisting of chloro, fluoro, and cyano. In some

[087] In another aspect, provided herein are compounds represented by Formula (lb): or a pharmaceutically acceptable salt thereof, wherein:

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 3 is independently, for each occurrence, selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may each individually be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; and wherein if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; or two R 3 substituents together with the carbon to which they are attached form C 3- 6 cycloalkyl;

X 1 is selected from CR 6 and N;

X 2 is selected from CH and N;

X 3 is selected from CR 7 and N;

R 4 is selected from halogen and cyano;

R 5 is selected from C 1-6 alkyl and C 1-6 alkoxyl;

R 6 is selected from halogen and cyano;

R 7 is halogen; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[088] In some embodiments, X 1 is CR 6 ; X 2 is CH; and X 3 is CR 7 . In some embodiments, R 6 is selected from the group consisting of chloro, fluoro, and cyano. In some embodiments, R 7 is chloro or fluoro.

[089] In some embodiments, X 1 is CR 6 ; X 2 is N; and X 3 is CR 7 . In some embodiments, R 6 is chloro or fluoro. In some embodiments, R 7 is fluoro.

[090] In some embodiments, X 1 is CR 6 ; X 2 is CH; and X 3 is N. In some embodiments, R 6 is fluoro.

[091] In some embodiments, X 1 is N; X 2 is CH; and X 3 is CR 7 . In some embodiments, R 7 is fluoro.

[092] In some embodiments, L is selected from the group consisting of -CH 2 -CH 2 -, - C(CH 3 )(H)-O-, -CH 2 -O-, -CH 2 -N(H)-, and -C(CH 3 )(H)-N(H)-. In some embodiments, L is selected from the group consisting of -CH 2 -CH 2 -, # -C(CH 3 )(H)-O- ## , # -CH 2 -O- ## , # -O-CH 2 - ## , # -CH 2 -N(H)- # -N(H)-CH 2 - ## , and # -C(CH 3 )(H)-N(H)- wherein # denotes the point of

[093] In another aspect, provided herein are compounds represented by Formula (Ic): or a pharmaceutically acceptable salt thereof, wherein:

C is 5-10 membered heteroaryl or 7-10 membered heterocyclyl, wherein the 5-10 membered heteroaryl or 7-10 membered heterocyclyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from R 3 ; and wherein, if the 5-10 membered heteroaryl or 7-10 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl;

L is selected from the group consisting of C 1-6 alkylene, C 1-6 alkylene-O-, and C 1- 6 alkylene-N(R A )-;

R 3 is independently, for each occurrence, selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), and - N(R A )(R B ), wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents; or two R 3 substituents together with the carbon to which they are attached form C 3- ecycloalkyl;

R 4 is halogen or cyano;

R 5 is C 1-6 alkyl or C 1-6 alkoxyl;

R 6 is halogen or cyano;

R 7 is halogen; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[094] In some embodiments, R 4 is selected from the group consisting of chloro, fluoro, and cyano.

[095] In some embodiments, R 5 is CH 3 or -O-CH 3 .

[096] In some embodiments, R 6 is selected from the group consisting of chloro, fluoro, and cyano.

[097] In some embodiments, R 7 is chloro or fluoro.

[098] In some embodiments, L is selected from the group consisting of -CH 2 -CH 2 -, - C(CH 3 )(H)-O-, -CH 2 -O-, -CH 2 -N(H)-, and -C(CH 3 )(H)-N(H)-. In some embodiments, L is selected from the group consisting of -CH 2 -CH 2 -, -C(CH 3 )(H)-O-, -CH 2 -O-, -CH 2 -N(H)-, and -C(CH 3 )(H)-N(H)-. In some embodiments, L is selected from the group consisting of -CH 2 - CH 2 -, # -C(CH 3 )(H)-O- ## , # -CH 2 -O- ## , # -O-CH 2 - ## , # -CH 2 -N(H)- ## , # -N(H)-CH 2 - ## , and # - C(CH 3 )(H)-N(H)- ## , wherein # denotes the point of attachment to denotes the point of attachment to

[099] In some embodiments, C is selected from the group consisting of wherein C may be optionally substituted on one or more available carbons by one, two, three, or more R 3 substituents independently selected from the group consisting of C 1-6 alkyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[0100] In some embodiments, C is selected from the group consisting of

wherein C may be optionally substituted on one or more available carbons by one, two, three, or more independent R 3 substituents selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl. [0101] In some embodiments, C is selected from the group consisting of

[0102] In some embodiments, C is selected from the group consisting of with one R 3 substituent selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3- 6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), and -N(R A )(R B ), wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents and R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[0103] In some embodiments, R 3 is selected from the group consisting of cyano, oxo, CH 3 , CH 2 CH 3 , C(H)(CH 3 ) 2 , -CCH, CF 3 , CH 2 CHF2, CF2CH 3 , CH 2 CH 2 F, -C(O)N(H)(CH 3 ), -O- CH 3 , -NH 2 , -N(H)(CH 3 ), cyclopropyl, and phenyl.

[0104] In some embodiments, C is selected from the group consisting of

[0105] In some embodiments, wherein C is substituted with two independent R 3 substituents selected from the group consisting of C 1-6 alkyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), -N(R A )(R B ), 5-6 membered heterocyclyl, and 5-6 membered heteroaryl, wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more substituents each independently selected from halogen and phenyl; if the 5-6 membered heterocyclyl contains a substitutable ring nitrogen atom, that ring nitrogen atom may be optionally substituted by C 1-6 alkyl; and

R A and R B are independently, for each occurrence, hydrogen or C 1-6 alkyl.

[0106] In some embodiments, R 3 is independently, for each occurrence, selected from the group consisting of CH 3 , -CH(CH 3 ) 2 , cyclopropyl, cyclopentyl, phenyl, pyridinyl,

[0107] In some embodiments, R 3 is independently, for each occurrence, selected from the

[0108] In some embodiments, R 3 is independently, for each occurrence, CH 3 or phenyl.

[0109] In some embodiments, C is selected from the group consisting of

, wherein C is substituted with three independent R 3 substituents and wherein two of the R 3 substituents together with the carbon to which they are attached form C 3-6 cycloalkyl and the third R 3 group is selected from the group consisting of C 1-6 alkyl, C 2-6 alkynyl, C 3-6 cycloalkyl, cyano, C 1-6 alkoxyl, oxo, phenyl, -C(O)N(R A )(R B ), and - N(R A )(R B ), wherein the C 1-6 alkyl may be optionally substituted on one or more available carbons by one, two, three, or more independent halogen substituents and R A and R B are independently hydrogen or C 1-6 alkyl.

[0112] In some embodiments, , wherein C is substituted with three independent R 3 , and wherein two of the R 3 substituents together with the carbon to which they are attached form cyclopropyl and the third R 3 group is oxo. [0113] In some embodiments, C is

[0114] In some embodiments, C is selected from the group consisting of

[0115] In another aspect, provided herein are compounds represented by Formula (Id): or a pharmaceutically acceptable salt thereof, wherein:

Xi is N or CR 9 ;

X 2 is N or CR 9 ;

X 3 is N or CR 9 ;

R 8 is halogen; and

R 9 is hydrogen or halogen.

[0116] In some embodiments, Xi is CR 9 ; X 2 is N; and X 3 is CR 9 . In some embodiments, R 9 is chloro or fluoro.

[0117] In some embodiments, Xi is CR 9 ; X 2 is CR 9 ; and X 3 is N. In some embodiments, R 9 is hydrogen or fluoro.

[0118] In some embodiments, Xi is N; X 2 is CR 9 ; and X 3 is CR 9 . In some embodiments, R 9 is fluoro.

[0119] In some embodiments, R 8 is chloro or fluoro.

[0120] In another aspect, the compound is any compound set forth in Table 1, or a pharmaceutically acceptable salt thereof. [0121] In another aspect, the compound is selected from the group consisting of 5-chloro-N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-ylo xy]methyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy)methyl]phenyl]-5 -fluoro -2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5- ylamino]methyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-(2,4-difluoro-3-[lH-pyrazolo[3,4-b]pyridin-5-y lmethoxy]phenyl)-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c][l, 4]oxazin-3-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5- ylmethyl]amino)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin- 5-yl]ethyl)phenyl]-2- methoxypyridine-3- sulfonamide;

5-[2-[3-(5-chloro-2-methoxypyridine-3-sulfonamido)-2,6-di fluorophenyl]ethyl]-N- methyl- lH-pyrazole-3-carboxamide;

N-(3-(2-(lH-pyrazolo[3,4-b]pyridin-5-yl)ethyl)-2,4-difluo rophenyl)-5-fluoro-2- methylpyridine-3 -sulfonamide;

5-cyano-N-[2,4-difluoro-3-(2-[1H-pyrazolo[3,4-b]pyridin-5 -yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[lH-pyrazolo[4,3-b]pyridin- 5-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[lH-pyrrolo[2,3-b]pyridin-5 -yl] ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[lH-pyrazolo[3,4-c]pyridin- 5-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b] pyridin-5-yl]oxy)methyl]phenyl]-

2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethy l)phenyl]-5-fhioro-2- methoxypyridine-3 -sulfonamide;

3-cyano-N-[2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5 -yl]ethyl)phenyl]-5- (trifluoromethyl) benzenesulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[2-methylpyrazolo[4,3-b]pyr idin-6-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[l-methylpyrazolo[4,3-b]pyr idin-6-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[imidazo[l,5-a] pyridin-7-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-[([4-methyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy)methyl]phenyl]-5-fluoro- 2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-yloxy]met hyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-[([4-methyl-lH-pyrazolo[3,4-b] pyridin-5-yl]oxy)methyl]phenyl]- 2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-[([4-m ethoxy- lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[2,4-difluoro-3-[([4-methoxy-lH-pyrazolo[3,4-b ]pyridin-5- yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[2,4-difluoro-3-(1-[lH-pyrazolo[3,4-b]pyridin- 5-yloxy]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

5-cyano-N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-y loxy]methyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

N-[3-[([4-cyano-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl ]-2,4-difluorophenyl]-5-fluoro-

2-methoxypyridine-3-sulfonamide;

5-chloro-N-[3-[([4-cyano-lH-pyrazolo[3,4-b]pyridin-5-yl]o xy)methyl]-2,4-difluorophenyl]-

2-methoxypyridine-3-sulfonamide;

5-chloro-N-[2,4-difluoro-3-(1-[lH-pyrazolo[3,4-b]pyridin- 5-ylamino]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

N-(3-(((lH-pyrazolo[3,4-b]pyridin-5-yl)oxy)methyl)-2,4-di fluorophenyl)-5-fluoro-2- methylpyridine-3 -sulfonamide;

5-chloro-N-(2,4-difluoro-3-(2-(8-(methylamino)imidazo[l,2 -a]pyrazin-3-yl)ethyl)phenyl)-2- methoxypyridine-3 -sulfonamide;

N-[3-(2-[8-aminoimidazo[l,2-a]pyrazin-3-yl]ethyl)-2,4-dif luorophenyl]-5-chloro-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6-yl]ethy l)-2,4-difluorophenyl]-2- methoxypyridine-3 -sulfonamide;

N-[3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6-yl]ethyl)-2,4-di fluorophenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-(2-[l-isopropylpyrazolo[4,3-b] pyridin-6-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-(2-[l-isopropylpyrazolo[4,3-b]pyridin-6 -yl]ethyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3-b]pyridin-6 -yl]ethyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy)methyl]phenyl]-5-fluoro-

2-methylpyridine-3 -sulfonamide;

N-[3-[([3-ethyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl ]-2,4-difluorophenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[3-[([3-ethyl-lH-pyrazolo[3,4-b]pyridin-5-yl]o xy)methyl]-2,4-difluorophenyl]-2- methoxypyridine-3 -sulfonamide;

5-cyano-N-[3-[([3-ethyl-lH-pyrazolo[3,4-b]pyridin-5-yl]ox y)methyl]-2,4-difluorophenyl]-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-[([l-methylpyrazolo[4,3-b]pyri din-6-yl]oxy)methyl]phenyl]-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-[([l-methylpyrazolo[4,3-b]pyridin-6-yl] oxy)methyl]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-([[3-(trifluoromethyl)-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]- 5-fluoro-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-([3-methyl-lH-pyrazolo[3,4-b]pyridin-5- yl]methoxy)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

5-chloro-N-[2,4-difluoro-3-([3-methyl-lH-pyrazolo [3,4-b]pyridin-5-yl]methoxy)phenyl]-2- methoxypyridine-3 -sulfonamide;

N-[3-[([3-ethyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl ]-2,4-difluorophenyl]-5-fluoro-2- methylpyridine-3 -sulfonamide;

5-cyano-N-(2,4-difluoro-3-((3-methyl-lH-pyrazolo[3,4-b]py ridin-5-yl)methoxy)phenyl)-2- methoxypyridine-3 -sulfonamide;

5-cyano-N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]p yridin-5- yl]methyl)amino]phenyl]-2-methoxypyridine-3 -sulfonamide;

5-cyano-N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]p yridin-5-yl]oxy)methyl]phenyl]-

2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-([3-methyl-lH-pyrazolo[3,4-b]pyridin-5- yl]methoxy)phenyl]-5-fluoro-2- methylpyridine-3 -sulfonamide;

N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4-b]pyridi n-5-yl]oxy)methyl]phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4 -b]pyridin-5- yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4-b]pyridi n-5-yl]oxy)methyl]phenyl]-5- fluoro-2-methylpyridine-3-sulfonamide;

5-cyano-N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4- b]pyridin-5- yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfonamide;

N-[3,5-difluoro-4-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy)methyl]pyridin-2-yl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]methyl)amino]phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b] pyridin-5- yl]methyl)amino]phenyl]-2-methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]methyl)amino]phenyl]-5- fluoro-2-methylpyridine-3-sulfonamide;

5-chloro-N-(2,4-difluoro-3-((imidazo[l,5-b]pyridazin-3-yl oxy)methyl)phenyl)-2- methoxypyridine-3 -sulfonamide;

N-[2-chloro-4-fluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

N-[2-cyano-4-fluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyrid in-5-yl]oxy)methyl]phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

5-fluoro-N-[3-fluoro-2-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]pyri din-4- yl]-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[3-fluoro-2-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]pyri din-4- yl]-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[3,5-difluoro-4-[([3-methyl-lH-pyrazolo[3,4-b] pyridin-5-yl]oxy)methyl]pyridin-

2-yl]-2-methoxypyridine-3-sulfonamide;

N-[3-chloro-5-fluoro-4-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]pyri din-2- yl]-5-fluoro-2-methoxypyridine-3-sulfonamide;

5-fluoro-N-[5-fluoro-6-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]pyridine-2- yl]-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[5-fluoro-6-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]pyri din-2- yl]-2-methoxypyridine-3-sulfonamide;

N-[3-[([3-ethynyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)meth yl]-2,4-difluorophenyl]-5-fluoro- 2-methoxypyridine-3-sulfonamide;

N-(3-(((3-cyclopropyl-lH-pyrazolo[3,4-b]pyridin-5-yl)oxy) methyl)-2,4-difluorophenyl)-5- fluoro-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-[([l -methyl -2-oxo-3H-imidazo[4,5-b]pyridin-6-yl]oxy)methyl]phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

N-(2,4-difluoro-3-(((2'-oxo-l',2'-dihydrospiro[cyclopropa ne-l,3'-pyrrolo[2,3-b]pyridin]-5'- yl)oxy)methyl)phenyl)-5-fluoro-2-methoxypyridine-3-sulfonami deN-(3-(((3-cyano-lH- pyrazolo[3,4-b]pyridin-5-yl)oxy)methyl)-2,4-difluorophenyl)- 5-fluoro-2-methoxypyridine-3- sulfonamide;

N-[2-chloro-4-fluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl]oxy)methyl]phenyl]-5- fluoro-2-methylpyridine-3-sulfonamide;

N-[3-([[3-(2,2-difluoroethyl)-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy]methyl)-2,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-([[3-(2-fluoroethyl)-lH-pyrazolo[3,4-b] pyridin-5-yl]oxy]methyl)phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

N-(4-chloro-2-fluoro-3-(((3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl)oxy)methyl)phenyl)-5- fluoro-2-methylpyridine-3-sulfonamide;

N-(4-chloro-2-fluoro-3-(((3-methyl-lH-pyrazolo[3,4-b]pyri din-5-yl)oxy)methyl)phenyl) -5- fluoro-2-methoxypyridine-3-sulfonamide;

5-chloro-N-[3-chloro-5-fluoro-4-[([3-methyl-lH-pyrazolo[3 ,4-b]pyridin-5- yl]oxy)methyl]pyridin-2-yl]-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-([[3-(2,2,2-trifluoroethyl)-lH-pyrazolo [3,4-b]pyridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de;

N-[3-([[3-(l,l-difluoroethyl)-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy]methyl)-2,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-[([5-methylimidazo[l,5-b]pyridazin-3-yl ]oxy)methyl]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide;

N-[2,4-difluoro-3-([[5-(trifluoromethyl)imidazo[l,5-b]pyr idazin-3-yl]oxy]methyl) phenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide;

N-[2,4-difluoro-3-[([3-phenyl-lH-pyrazolo[3,4-b]pyridin-5 -yl]oxy)methyl]phenyl]-5-fluoro- 2-methoxypyridine-3-sulfonamide,

N-[2, 4-difluoro-3-[([3-m ethyl -4-phenyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phenyl]- 5-fluoro-2-methoxypyridine-3-sulfonamide, N-[3-[([4-cyclopropyl-3-methyl-lH-pyrazolo[3,4-b]pyridin-5-y l]oxy)methyl]-2,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide, N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-4-yl)-lH-pyrazolo[3 ,4-b]pyridin-5- yl]°xy] m ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide , N-[2,4-difluoro-3-[([4-isopropyl-3-methyl-lH-pyrazolo[3,4-b] pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de,

N-[3-[([4-benzyl-3 -methyl- lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]-2,4-difluoropheny l]- 5-fluoro-2-methoxypyridine-3-sulfonamide, N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-2-yl)-lH-pyrazolo[3 ,4-b]pyridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de, N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-3-yl)-lH-pyrazolo[3 ,4-b]pyridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de, N-[3-[([4-cyclopentyl-3-methyl-lH-pyrazolo[3,4-b]pyridin-5-y l]oxy)methyl]-2,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide,

N-[2, 4-difluoro-3-([[3-m ethyl -4-(1-methylpiperidin-4-yl)-lH-pyrazolo[3,4-b]pyri din-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de, and N-[2,4-difluoro-3-([[3-methyl-4-(piperidin-l-yl)-lH-pyrazolo [3,4-b]pyridin-5- yl]°xy] m ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide , or a pharmaceutically acceptable salt thereof.

Therapeutic Applications of GCN2 Modulating (Inhibiting/Activating) Compounds [0122] It is contemplated that GCN2 modulating (inhibiting/activating) compounds and related compounds described herein, such as a compound of Formula I, provide therapeutic benefits to subjects suffering from cancer, neurodegenerative disease, and doxorubicin- induced cardiotoxicity. Accordingly, one aspect of the invention provides therapeutic methods for treating the foregoing diseases and conditions using GCN2 modulating compounds and related compounds described herein. Various aspects and embodiments of the therapeutic methods are described below.

Cancer

[0123] One aspect of the invention provides a method of treating cancer in a subject. The method comprises administering a therapeutically effective amount of a GCN2 modulating (inhibiting/activating) compound or related compound described herein, such as a compound of Formula 1 to a subject in need thereof to treat the cancer. In certain embodiments, the particular compound of Formula I, is a compound defined by one of the embodiments described above.

[0124] In certain embodiments, the cancer is a solid tumor, leukemia, or lymphoma. In certain embodiments, the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, sweat gland carcinoma, sebaceous gland carcinoma, thyroid cancer, kidney cancer, uterus cancer, esophagus cancer, liver cancer, head cancer, neck cancer, throat cancer, mouth cancer, bone cancer, chest cancer, lymph node cancer, eye cancer, mesothelioma, an acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, retinoblastoma, leukemia, or lymphoma. In certain embodiments, the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, leukemia, or lymphoma. In certain other embodiments, the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, lung cancer, leukemia, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, thyroid cancer, kidney cancer, uterus cancer, esophagus cancer, liver cancer, an acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, or retinoblastoma. In certain other embodiments, the cancer is small cell lung cancer, non-small cell lung cancer, melanoma, cancer of the central nervous system tissue, brain cancer, Hodgkin's lymphoma, nonHodgkin's lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, or diffuse large B-Cell lymphoma. In certain other embodiments, the cancer is breast cancer, colon cancer, small-cell lung cancer, non-small cell lung cancer, prostate cancer, renal cancer, ovarian cancer, leukemia, melanoma, or cancer of the central nervous system tissue. In certain other embodiments, the cancer is colon cancer, small-cell lung cancer, non-small cell lung cancer, renal cancer, ovarian cancer, renal cancer, or melanoma.

[0125] Additional exemplary cancers include fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, and hemangioblastoma.

[0126] In certain embodiments, the cancer is a neuroblastoma, meningioma, hemangiopericytoma, multiple brain metastases, glioblastoma multiforms, glioblastoma, brain stem glioma, poor prognosis malignant brain tumor, malignant glioma, anaplastic astrocytoma, anaplastic oligodendroglioma, neuroendocrine tumor, rectal adeno carcinoma, Dukes C & D colorectal cancer, unresectable colorectal carcinoma, metastatic hepatocellular carcinoma, Kaposi's sarcoma, karotype acute myeloblastic leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma, low grade follicular lymphoma, metastatic melanoma, localized melanoma, malignant mesothelioma, malignant pleural effusion mesothelioma syndrome, periton 6 al carcinoma, papillary serous carcinoma, gynecologic sarcoma, soft tissue sarcoma, scelroderma, cutaneous vasculitis, Langerhans cell histiocytosis, leiomyosarcoma, fibrodysplasia ossificans progressive, hormone refractory prostate cancer, resected high-risk soft tissue sarcoma, unrescectable hepatocellular carcinoma, Waldenstrom's macroglobulinemia, smoldering myeloma, indolent myeloma, fallopian tube cancer, androgen independent prostate cancer, androgen dependent stage IV non-metastatic prostate cancer, hormone-insensitive prostate cancer, chemotherapy -insensitive prostate cancer, papillary thyroid carcinoma, follicular thyroid carcinoma, medullary thyroid carcinoma, or leiomyoma. [0127] In certain embodiments, the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, sweat gland carcinoma, sebaceous gland carcinoma, thyroid cancer, kidney cancer, uterus cancer, esophagus cancer, liver cancer, head cancer, neck cancer, throat cancer, mouth cancer, bone cancer, chest cancer, lymph node cancer, eye cancer, mesothelioma, an acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, retinoblastoma, leukemia, or lymphoma.

[0128] In certain embodiments, the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, lung cancer, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, leukemia, or lymphoma. Neurodegenerative Disease

[0129] Another aspect of the invention provides a method of treating a neurodegenerative disease in a subject. The method comprises administering a therapeutically effective amount of a compound described herein, such as a compound of Formula I, to a subject in need thereof to treat the neurodegenerative disease. In certain embodiments, the neurodegenerative disease is Alzheimer's disease, Parkinson's Disease, Huntington's Disease, amyotrophic lateral sclerosis, or spinocerebellar ataxia.

[0130] Aberrant autophagic processes contribute to neurodegenerative diseases. For example, y-secretase activity is enhanced in autophagic vacuoles through signal transduction mediated by GCN2 phosphorylation of the a subunit of eukaryotic initiation factor 2 (eIF2a) (see, e.g., Ohta, K. et al. in Autophagy 2010, 6, 345-352). The y-secretase enhances amyloid- P synthesis and the progression of Alzheimer's disease. Accordingly, compounds having inhibitory activity towards GCN2 provide benefits to patients suffering from neurodegenerative diseases.

Doxorubicin-induced Cardiotoxicity

[0131] Another aspect of the invention provides a method of treating doxorubicin-induced cardiotoxicity in a subject. The method comprises administering a therapeutically effective amount of a compound described herein, such as a compound of Formula I, to a subject in need thereof suffering from doxorubicin-induced cardiotoxicity, to thereby treat the doxorubicin-induced cardiotoxicity.

[0132] Another aspect of the invention provides a method of preventing doxorubicin-induced cardiotoxicity in a subject. The method comprises administering a therapeutically effective amount of a compound described herein, such as a compound of Formula I, to a subject in need thereof that has received, or will receive, doxorubicin, to thereby prevent doxorubicin- induced cardiotoxicity.

[0133] Deficiency in GCN2 has been reported to ameliorate doxorubicin-induced cardiotoxicity. See, for example, Wang et al. in Redox Biology (2018) vol. 17, pages 25-34. Accordingly, compounds having inhibitory activity towards GCN2 provide benefits to patients suffering from or likely to suffer from doxorubicin-induced cardiotoxicity.

[0134] In certain embodiments, the subject is a human.

[0135] Another aspect of the invention provides for the use of a compound described herein (such as a compound of Formula I) in the manufacture of a medicament. In certain embodiments, the medicament is for treating a disorder described herein, such as cancer. [0136] Another aspect of the invention provides for the use of a compound described herein (such as a compound of Formula I) for treating a medical disorder, such a medical disorder described herein (e.g., cancer).

[0137] Further, it is contemplated that GCN2 modulators (inhibitors/activators) and related compounds described herein, such as a compound of Formula I, can inhibit/activate the activity of GCN2. Accordingly, another aspect of the invention provides a method of inhibiting/activating the activity of GCN2. The method comprises exposing a GCN2 to an effective amount of a GCN2 modulator (inhibitor/activator) or related compound described herein, such as a compound of Formula I, to inhibit/activate GCN2 activity. In certain embodiments, the particular compound of Formula I, is the compound defined by one of the embodiments described above.

Combination Therapy

[0138] Another aspect of the invention provides for combination therapy. GCN2 modulators (inhibitors/activators) and related compounds (e.g., a compound of Formula I) or their pharmaceutically acceptable salts may be used in combination with additional therapeutic agents to treat medical disorders, such as a cancer.

[0139] Exemplary therapeutic agents that may be used as part of a combination therapy in treating cancer, include, for example, mitomycin, tretinoin, ribomustin, gemcitabine, vincristine, etoposide, cladribine, mitobronitol, methotrexate, doxorubicin, carboquone, pentostatin, nitracrine, zinostatin, cetrorelix, letrozole, raltitrexed, daunorubicin, fadrozole, fotemustine, thymalfasin, sobuzoxane, nedaplatin, cytarabine, bicalutamide, vinorelbine, vesnarinone, aminoglutethimide, amsacrine, proglumide, elliptinium acetate, ketanserin, doxifluridine, etretinate, isotretinoin, streptozocin, nimustine, vindesine, flutamide, drogenil, butocin, carmofur, razoxane, sizofilan, carboplatin, mitolactol, tegafur, ifosfamide, prednimustine, picibanil, levamisole, teniposide, improsulfan, enocitabine, lisuride, oxymetholone, tamoxifen, progesterone, mepitiostane, epitiostanol, formestane, interferon- alpha, interferon-2 alpha, interferon -beta, interferon-gamma, colony stimulating factor- 1, colony stimulating factor-2, denileukin diftitox, interleukin-2, and leutinizing hormone releasing factor.

[0140] Radiation therapy may also be used as part of a combination therapy.

[0141] An additional class of agents that may be used as part of a combination therapy in treating cancer is immune checkpoint inhibitors (also referred to as immune checkpoint blockers). Immune checkpoint inhibitors are a class of therapeutic agents that have the effect of blocking immune checkpoints. See, for example, Pardoll in Nature Reviews Cancer (2012) vol. 12, pages 252-264. Exemplary immune checkpoint inhibitors include agents that inhibit one or more of (i) cytotoxic T-lymphocyte-associated antigen 4 (CTLA4), (ii) programmed cell death protein 1 (PD1), (iii) PDL1, (iv) LAB3, (v) B7-H3, (vi) B7-H4, and (vii) TIM3. The CTLA4 inhibitor Ipilumumab has been approved by the United States Food and Drug Administration for treating melanoma.

[0142] Yet other agents that may be used as part of a combination therapy in treating cancer are monoclonal antibody agents that target non-checkpoint targets (e.g., herceptin) and non- cytoxic agents (e.g., tyrosine-kinase inhibitors).

[0143] Yet other agents that may be used as part of a combination therapy in treating cancer are agents which deplete amino acids or other nutrients, radiation, and agents that provoke the integrated stress response or that promote autophagy. Such agents may include aspariginase, argininase inhibitors of kinases such a b-Raf, and cytotoxic agents such as cis-platin.

[0144] Accordingly, another aspect of the invention provides a method of treating cancer in a patient, where the method comprises administering to the patient in need thereof (i) a therapeutically effective amount of a GCN2 modulator (activator/inhibitor) compound described herein and (ii) a second anti-cancer agent, in order to treat the cancer, where the second therapeutic agent may be one of the additional therapeutic agents described above (e.g., mitomycin, tretinoin, ribomustin, gemcitabine, an immune checkpoint inhibitor, or a monoclonal antibody agent that targets non-checkpoint targets) or one of the following:

• an inhibitor selected from an ALK Inhibitor, an ATR Inhibitor, an A2A Antagonist, a Base Excision Repair Inhibitor, a Bcr-Abl Tyrosine Kinase Inhibitor, a Bruton's Tyrosine Kinase Inhibitor, a CDC7 Inhibitor, a CHK1 Inhibitor, a Cyclin-Dependent Kinase Inhibitor, a DNA- PK Inhibitor, an Inhibitor of both DNA-PK and mTOR, a DNMT1 Inhibitor, a DNMT1 Inhibitor plus 2-chloro-deoxyadenosine, an HD AC Inhibitor, a Hedgehog Signaling Pathway Inhibitor, an IDO Inhibitor, a JAK Inhibitor, a mTOR Inhibitor, a MEK Inhibitor, a MELK Inhibitor, a MTHl Inhibitor, a PARP Inhibitor, a Phosphoinositide 3-Kinase Inhibitor, an Inhibitor of both PARP1 and DHODH, a Proteasome Inhibitor, a Topoisomerase-II Inhibitor, a Tyrosine Kinase Inhibitor, a VEGFR Inhibitor, and a WEE1 Inhibitor;

• an agonist of 0X40, CD137, CD40, GITR, CD27, HVEM, TNFRSF25, or ICOS;

• a therapeutic antibody targeting one of the following: CD20, CD30, CD33, CD52, EpCAM, CEA, gpA33, a mucin, TAG-72, CAIX, PSMA, a folate-binding protein, a ganglioside, Le, VEGF, VEGFR, VEGFR2, integrin αVβ3, integrin α5β1, EGFR, ERBB2, ERBB3, MET, IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, FAP, tenascin, CD19, KIR, NKG2A, CD47, CEACAM1, c-MET, VISTA, CD73, CD38, BAFF, interleukin- 1 beta, B4GALNT1, interleukin-6, and interleukin-6 receptor;

• a cytokine selected from IL-12, IL-15, GM-CSF, and G-CSF;

• a therapeutic agent selected from sipuleucel-T, aldesleukin (a human recombinant interleukin-2 product having the chemical name des-alanyl- 1, serine- 125 human interleukin- 2), dabrafenib (a kinase inhibitor having the chemical name N-{3-[5-(2-aminopyrimidin-4- yl)-2-tert-butyl- 1 ,3-thiazol-4-yl]-2-fluorophenyl }-2,6-difluorobenzenesulfonamide), vemurafenib (a kinase inhibitor having the chemical name propane- 1 -sulfonic acid { 3-[5-(4- chlorophenyl)-1H-pyrrolo[2,3-b ]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide), and 2- chloro-deoxyadenosine; or

• a placental growth factor, an antibody-drug conjugate, an oncolytic virus, or an anticancer vaccine.

[0145] In certain embodiments, the second anti-cancer agent is an ALK Inhibitor. In certain embodiments, the second anti -cancer agent is an ALK Inhibitor comprising ceritinib or crizotinib. In certain embodiments, the second anti-cancer agent is an ATR Inhibitor. In certain embodiments, the second anti -cancer agent is an ATR Inhibitor comprising AZD6738 or VX-970. In certain embodiments, the second anti -cancer agent is an A2A Antagonist. In certain embodiments, the second anti -cancer agent is a Base Excision Repair Inhibitor comprising methoxyamine. In certain embodiments, the second anti-cancer agent is a Base Excision Repair Inhibitor, such as methoxyamine. In certain embodiments, the second anticancer agent is a Bcr-Abl Tyrosine Kinase Inhibitor. In certain embodiments, the second anticancer agent is a Bcr-Abl Tyrosine Kinase Inhibitor comprising dasatinib or nilotinib. In certain embodiments, the second anti -cancer agent is a Bruton's Tyrosine Kinase Inhibitor. In certain embodiments, the second anti -cancer agent is a Bruton's Tyrosine Kinase Inhibitor comprising ibrutinib. In certain embodiments, the second anti-cancer agent is a CDC7 Inhibitor. In certain embodiments, the second anti-cancer agent is a CDC7 Inhibitor comprising RXDX- 103 orAS-141.

[0146] In certain embodiments, the second anti-cancer agent is a CHK1 Inhibitor. In certain embodiments, the second anti-cancer agent is a CHK1 Inhibitor comprising MK-8776, ARRY-575, or SAR-020106. In certain embodiments, the second anti-cancer agent is a Cyclin-Dependent Kinase Inhibitor. In certain embodiments, the second anti-cancer agent is a Cyclin-Dependent Kinase Inhibitor comprising palbociclib. In certain embodiments, the second anti-cancer agent is a DNA-PK Inhibitor. In certain embodiments, the second anticancer agent is a DNA-PK Inhibitor comprising MSC2490484A. In certain embodiments, the second anti-cancer agent is Inhibitor of both DNA-PK and mTOR. In certain embodiments, the second anti -cancer agent comprises CC-115.

[0147] In certain embodiments, the second anti-cancer agent is a DNMT1 Inhibitor. In certain embodiments, the second anti -cancer agent is a DNMT1 Inhibitor comprising decitabine, RX-3117, guadecitabine, NUC-8000, or azacytidine. In certain embodiments, the second anti-cancer agent comprises a DNMT1 Inhibitor and 2-chloro-deoxyadenosine. In certain embodiments, the second anti -cancer agent comprises ASTX-727.

[0148] In certain embodiments, the second anti-cancer agent is a HD AC Inhibitor. In certain embodiments, the second anti -cancer agent is a HD AC Inhibitor comprising OBP-801, CHR- 3996, etinostate, resminostate, pracinostat, CG-200745, panobinostat, romidepsin, mocetinostat, belinostat, AR-42, ricolinostat, KA-3000, or ACY-241.

[0149] In certain embodiments, the second anti-cancer agent is a Hedgehog Signaling Pathway Inhibitor. In certain embodiments, the second anti-cancer agent is a Hedgehog Signaling Pathway Inhibitor comprising sonidegib or vismodegib. In certain embodiments, the second anti-cancer agent is an IDO Inhibitor. In certain embodiments, the second anticancer agent is an IDO Inhibitor comprising INCB024360. In certain embodiments, the second anti-cancer agent is a JAK Inhibitor. In certain embodiments, the second anti -cancer agent is a JAK Inhibitor comprising ruxolitinib or tofacitinib. In certain embodiments, the second anti-cancer agent is a mTOR Inhibitor. In certain embodiments, the second anticancer agent is a mTOR Inhibitor comprising everolimus or temsirolimus. In certain embodiments, the second anti -cancer agent is a MEK Inhibitor. In certain embodiments, the second anti-cancer agent is a MEK Inhibitor comprising cobimetinib or trametinib. In certain embodiments, the second anti -cancer agent is a MELK Inhibitor. In certain embodiments, the second anti-cancer agent is a MELK Inhibitor comprising ARN-7016, APTO-500, or OTS- 167. In certain embodiments, the second anti-cancer agent is a MTH1 Inhibitor. In certain embodiments, the second anti -cancer agent is a MTH1 Inhibitor comprising (S)-crizotinib, TH 2 87, or TH588.

[0150] In certain embodiments, the second anti-cancer agent is a PARP Inhibitor. In certain embodiments, the second anti -cancer agent is a PARP Inhibitor comprising MP-124, olaparib, BGB-290, talazoparib, veliparib, niraparib, E7449, rucaparb, or ABT-767. In certain embodiments, the second anti -cancer agent is a Phosphoinositide 3-Kinase Inhibitor. In certain embodiments, the second anti -cancer agent is a Phosphoinositide 3-Kinase Inhibitor comprising idelalisib. In certain embodiments, the second anti-cancer agent is an inhibitor of both PARP1 and DHODH (i.e., an agent that inhibits both poly ADP ribose polymerase 1 and dihydroorotate dehydrogenase).

[0151] In certain embodiments, the second anti-cancer agent is a Proteasome Inhibitor. In certain embodiments, the second anti -cancer agent is a Proteasome Inhibitor comprising bortezomib or carfilzomib. In certain embodiments, the second anti -cancer agent is a Topoisomerase-II Inhibitor. In certain embodiments, the second anti-cancer agent is a Topoisomerase-II Inhibitor comprising vosaroxin.

[0152] In certain embodiments, the second anti-cancer agent is a Tyrosine Kinase Inhibitor. In certain embodiments, the second anti-cancer agent is a Tyrosine Kinase Inhibitor comprising bosutinib, cabozantinib, imatinib or ponatinib. In certain embodiments, the second anti-cancer agent is a VEGFR Inhibitor. In certain embodiments, the second anticancer agent is a VEGFR Inhibitor comprising reg oraf enib. In certain embodiments, the second anti-cancer agent is a WEE1 Inhibitor. In certain embodiments, the second anticancer agent is a WEE1 Inhibitor comprising AZDI 775.

[0153] In certain embodiments, the second anti-cancer agent is an agonist of 0X40, CD137, CD40, GITR, CD27, HVEM, TNFRSF25, or ICOS. In certain embodiments, the second anticancer agent is a therapeutic antibody selected from the group consisting of rituximab, ibritumomab tiuxetan, tositumomab, obinutuzumab, ofatumumab, brentuximab vedotin, gemtuzumab ozogamicin, alemtuzumab, IGN101, adecatumumab, labetuzumab, huA33, pemtumomab, oregovomab, minetumomab, cG250, J591, Movl8, farletuzumab, 3F8, chl4.18, KW-2871, hu3S193, lgN311, bevacizumab, IM-2C6, pazopanib, sorafenib, axitinib, CDP791, lenvatinib, ramucirumab, etaracizumab, volociximab, cetuximab, panitumumab, nimotuzumab, 806, afatinib, erlotinib, gefitinib, osimertinib, vandetanib, trastuzumab, pertuzumab, MM-121, AMG 102, METMAB, SCH 900105, AVE1642, IMC-A12, MK-0646, R1507, CP 751871, KB004, IIIA-4, mapatumumab, HGS-ETR2, CS-1008, denosumab, sibrotuzumab, Fl 9, 81C6, MEDI551, lirilumab, MEDI9447, daratumumab, belimumab, canakinumab, dinutuximab, siltuximab, and tocilizumab.

[0154] In certain embodiments, the second anti-cancer agent is a placental growth factor. In certain embodiments, the second anti -cancer agent is a placental growth factor comprising ziv-aflibercept. In certain embodiments, the second anti-cancer agent is an antibody-drug conjugate. In certain embodiments, the second anti-cancer agent is an antibody-drug conjugate selected from the group consisting of brentoxumab vedotin and trastuzumab emtransine. [0155] In certain embodiments, the second anti-cancer agent is an oncolytic virus. In certain embodiments, the second anti -cancer agent is the oncolytic virus talimogene laherparepvec. In certain embodiments, the second anti-cancer agent is an anti-cancer vaccine. In certain embodiments, the second anti -cancer agent is an anti-cancer vaccine selected from the group consisting of a GM-CSF tumor vaccine, a STING/GM-CSF tumor vaccine, and NY-ESO-1. In certain embodiments, the second anti-cancer agent is a cytokine selected from IL-12, IL- 15, GM-CSF, and G-CSF.

[0156] In certain embodiments, the second anti-cancer agent is a therapeutic agent selected from sipuleucel-T, aldesleukin (a human recombinant interleukin-2 product having the chemical name des-alanyl- 1, serine- 125 human interleukin-2), dabrafenib (a kinase inhibitor having the chemical name N-{3-[5-(2-aminopyrimidin-4-yl)-2-tert-butyl-l,3-thiazol-4-y l]-2- fluorophenyl}-2,6-difluorobenzenesulfonamide), vemurafenib (a kinase inhibitor having the chemical name propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-1H -pyrrolo[2,3-b ]pyridine-3- carbonyl]-2,4-difluoro-phenyl}-amide), and 2-chloro-deoxyadenosine.

[0157] The doses and dosage regimen of the active ingredients used in the combination therapy may be determined by an attending clinician. In certain embodiments, the GCN2 modulator (inhibitor/activator) or related compound (e.g., a compound of any one of Formula I) and the additional therapeutic agent(s) are administered in doses commonly employed when such agents are used as monotherapy for treating the disorder. In other embodiments, the GCN2 modulator (inhibitor/activator) or related compound (e.g., a compound of any one of Formula I) and the additional therapeutic agent(s) are administered in doses lower than the doses commonly employed when such agents are used as monotherapy for treating the disorder. In certain embodiments, GCN2 modulator (inhibitor/activator) or related compound (e.g., a compound of any one of Formula I) and the additional therapeutic agent(s) are present in the same composition, which is suitable for oral administration.

[0158] In certain embodiments, GCN2 modulator (inhibitor/activator) or related compound (e.g., a compound of any one of Formula I) and the additional therapeutic agent(s) may act additively or synergistically. A synergistic combination may allow the use of lower dosages of one or more agents and/or less frequent administration of one or more agents of a combination therapy. A lower dosage or less frequent administration of one or more agents may lower toxicity of the therapy without reducing the efficacy of the therapy.

[0159] Another aspect of this invention is a kit comprising a therapeutically effective amount of the GCN2 modulator (inhibitor/activator) or related compound (e.g., a compound of any one of Formula I), a pharmaceutically acceptable carrier, vehicle or diluent, and optionally at least one additional therapeutic agent listed above.

Pharmaceutical Compositions and Dosing Considerations

[0160] As indicated above, the invention provides pharmaceutical compositions, which comprise a therapeutically-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. The pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally.

[0161] The phrase "therapeutically-effective amount" as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub -population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment.

[0162] The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[0163] Wetting agents, emulsifiers and lubricants, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.

[0164] Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.

[0165] In certain embodiments, a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, and a compound of the present invention. In certain embodiments, an aforementioned formulation renders orally bioavailable a compound of the present invention.

[0166] Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.

[0167] Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water- in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.

[0168] In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules, trouches and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers and/or any of the following: (1) fillers or extenders; (2) binders; (3) humectants; (4) disintegrating agents; (5) solution retarding agents; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants; (7) wetting agents; (8) absorbents; (9) lubricants; (10) coloring agents; and (11) controlled release agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents.

[0169] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using one or more binders, lubricants, inert diluents, preservatives, disintegrants, and/or surface-active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.

[0170] The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.

[0171] Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers.

[0172] Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.

[0173] Suspensions, in addition to the active compounds, may contain suspending agents. [0174] Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers which are solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.

[0175] Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.

[0176] Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.

[0177] Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.

[0178] Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.

[0179] Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

[0180] Proper fluidity of the pharmaceutical compositions can be maintained, for example, by the use of coating materials, by the maintenance of the required particle size in the case of dispersions, and/or by the use of surfactants.

[0181] These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents. It may also be desirable to include isotonic agents into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

[0182] In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

[0183] Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.

[0184] When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier.

[0185] The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred.

[0186] The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperiton 6 al, transtrach 6 al, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection and infusion.

[0187] The phrases "systemic administration," "administered systemically," "peripheral administration" and "administered peripherally" as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.

[0188] These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intraci sternally and topically, as by powders, ointments or drops, including buccally and sublingually.

[0189] Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. [0190] Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.

[0191] The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general h 6 alth and prior medical history of the patient being treated, and like factors well known in the medical arts. [0192] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.

[0193] In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Preferably, the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. When the compounds described herein are co-administered with another agent (e.g., as sensitizing agents), the effective amount may be less than when the agent is used alone. [0194] If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day.

[0195] The invention further provides a unit dosage form (such as a tablet or capsule) comprising an (aza)indazolyl-aryl sulfonamide or related compound described herein in a therapeutically effective amount for the treatment of a medical disorder described herein.

EXAMPLES

[0196] The representative examples that follow are intended to help illustrate the invention, and are not intended to, nor should they be construed to, limit the scope of the invention. [0197] The compounds provided herein can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where 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. Optimal reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization. [0198] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein.

[0199] The compounds provided herein may be isolated and purified by known standard procedures. Such procedures include recrystallization, filtration, flash chromatography, trituration, high pressure liquid chromatography (HPLC), or supercritical fluid chromatography

(SFC). Note that flash chromatography may either be performed manually or via an automated system. The compounds provided herein may be characterized by known standard procedures, such as nuclear magnetic resonance spectroscopy (NMR) or liquid chromatography mass spectrometry (LCMS). NMR chemical shifts are reported in part per million (ppm) and are generated using methods well known to those of skill in the art.

List of Abbreviations:

Ac acetyl

ACN acetonitrile

AcOH acetic acid

AIBN azobisisobutyronitrile

BAST bis(2-methoxyethyl)aminosulfur trifluoride

9-BBN 9-Borabicyclo[3.3.1]nonane Boc tert-butyl oxy carbonyl

BINAP 2,2' -bis(diphenylphosphino)-l,l ' -binaphthyl

BINOL 1,1' -bi-2-naphthol

BPO benzoyl peroxide

CDI carbonyldiimidazole

DAST diethylaminosulfur trifluoride dba dibenzylideneacetone

DBU l,8-diazabicyclo[5.4.0]undec-7-ene

DCE di chloroethane

DCM dichloromethane

DDQ 2,3-dichloro-5,6-dicyano-l,4-benzoquinone

DEAD Diethyl azodi carb oxy late

DIEA diisopropylethylamine

DMA dimethylacetamide

DMAP 4-dimethylaminopyridine

DMF dimethylformamide

DMSO dimethyl sulfoxide dppf 1, 1'-bis(diphenylphosphino)ferrocene

EA ethyl acetate

EDTA Ethylenediaminetetraacetic acid

EtOAc ethyl acetate

FA formic acid

HATU 1-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyri dinium-3-oxide hexafluorophosphate

HPLC high performance liquid chromatography

IBX 2-iodoxybenzoic acid

IPA isopropyl alcohol

KO Ac potassium acetate

LAH lithium aluminum hydride

LCMS liquid chromatography-mass spectrometry

LDA lithium diisopropylamide

MeCN acetonitrile

MTBE methyl tert-butyl ether NBS A-bromosuccinimide

NCS A-chlorosuccinimide

NIS A-iodosuccinimide

NMR nuclear magnetic resonance spectroscopy

PCy 3 tricyclohexylphosphine

PE petroleum ether

Pin pinacolato

PMB p-methoxybenzyl

Py pyridine

SEM trimethylsilylethoxymethyl

TBAF tetrabutylammonium fluoride

TBHP tert-butyl hydroperoxide

TBS tert-butyldimethylsilyl

TEA triethylamine

Tf triflate/trifluoromethanesulfonate

TFA trifluoroacetic acid

TFAA trifluoroacetic anhydride

TM AD N,N,N' ,N' -Tetramethyl azodi carb oxami de

General Schemes

[0200] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising one or more of Schemes 1-9 as set out below. Reaction steps represented by dashed arrows are to be understood to be optional. Unless otherwise specified, the variables of the Schemes are as defined herein. Reaction conditions should be understood to be exemplary and non-limiting, and may occur in the presence of an appropriate solvent.

[0201] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising Scheme 1, wherein Z is halide, Y is amino or a functional group that may be readily converted to amino (e.g., nitro), and R a is hydrogen or methyl when q is 1; p is 0 or 1, q is 0 or 1, and the sum of p and q is 1. Scheme 1

[0202] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising Scheme 2, wherein Z is halide and Y is halide or hydroxyl. Scheme 2

[0203] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising Scheme 3, wherein Z is a functional group that may be readily converted to amino (e.g., nitro) and Y is halide or hydroxyl.

Scheme 3 [0204] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising Scheme 4, wherein Z is halide.

Scheme 4

[0205] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising Scheme 5, wherein Z is halide.

Scheme 5

[0206] In some embodiments, compounds of the present disclosure may be manufactured using a process comprising Scheme 6, wherein Y is an amino or a functional group readily converted to amino (e.g., nitro) and R b is hydrogen or methyl. Scheme 6 reductive amination

[0207] In some embodiments, compounds of the present disclosure may be synthesized using a process comprising Scheme 7, wherein Z is halide.

Scheme 7

[0208] In some embodiments, compounds of the present disclosure may be synthesized using a process comprising Scheme 8, wherein Y is amino or a functional group that may be readily converted to amino (e.g., nitro). Scheme 8 reductive amination

[0209] In some embodiments, compounds of the present disclosure may be synthesized using a process comprising Scheme 9, wherein Y is amino or a functional group that may be readily converted to amino (e.g., nitro).

Scheme 9

Example 1: Synthesis of 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (Intermediate 1)

Intermediate 1

Synthesis of Int. 1-a: 3-(benzylsulfanyl)-5-fluoro-2-methoxypyridine

[0210] Into a 2000 mL 3 -necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-bromo-5-fluoro-2-methoxypyridine (150 g, 728 mmol, 1 equiv.), benzyl mercaptan (109 g, 878 mmol, 1.2 equiv), Pd 2 (dba) 3 (41.9 g, 36 mmol, 0.05 equiv.), Xantphos (30 g, 52 mmol, 0.07 equiv), DIEA (189 g, 1.46 mol) and toluene (1.2 L). The resulting solution was stirred for 2 h at 85 °C in an oil bath, then concentrated under vacuum. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 10) to give 3-(benzylsulfanyl)-5-fluoro-2-methoxypyridine (172 g, 95% yield) as a colorless oil.

Synthesis of Int. 1 : 5-fluoro-2-methoxypyridine-3-sulfonyl chloride

[0211] Into a 2000 mL 3 -necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-(benzylsulfanyl)-5-fluoro-2-methoxypyridine (172 g, 690 mmol, 1 equiv.) and CELCN (1000 mL). This was followed by the addition of HC1 (57 mL) at 10 °C. To this was added NCS (368.5 g, 2760 mmol, 4 equiv) in portions at 10 °C. The resulting solution was stirred for 30 min at 10-20 °C in a water/ice bath. The resulting solution was diluted with 2000 mL of H 2 O and extracted with 2 x 1.5 L of dichloromethane. The combined organics were washed with 2000 ml of brine and dried over anhydrous sodium sulfate, before being concentrated. The residue was applied to a silica gel column which was eluted with PE. 5-Fluoro-2-methoxypyridine-3-sulfonyl chloride (50.5 g, 32% yield) was isolated as a white solid. LC-MS: (ES, m/z): [M+H] + =284.0.

'H-NMR: (300 MHz, Chloroform-d, ppm): δ 8.37 (d, J= 3.0 Hz, 1H), 8.04-8.01 (m, 1H), 4.18 (s, 3H).

Example 2: Synthesis of 5-chloro-2-methoxypyridine-3-sulfonyl chloride (Intermediate 21

Intermediate 2

Synthesis of Int. 2-a: 3-(benzylsulfanyl)-5-chloro-2-methoxypyridine

[0212] Into a 3000 mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-bromo-5-chloro-2-methoxypyridine (150 g, 678 mmol, 1.0 equiv), toluene (1500 mL), phenylmethanethiol (92.6 g, 746 mmol, 1.1 equiv), DIEA (175.4 g, 1357 mmol, 2.0 equiv), XantPhos (3.92 g, 6 mmol, 0.01 equiv) and Pd2(dba)3-CHC13 (5.27 g, 5 mmol 0.0075 equiv). The resulting solution was stirred for 4 h at 110 °C in an oil bath. The solids were removed by filtration and the filtrate concentrated. The residue was applied to a silica gel column, eluting with PE: EA=20: 1. Concentration of the appropriate fractions gave a solid which was slurried with PE (3 V). The solid was removed by filtration and dried to give 3-(benzylsulfanyl)-5-chloro-2-methoxypyridine (130 g, 72% yield) as a yellow solid.

Synthesis of Int. 2: 5-chloro-2-methoxypyridine-3-sulfonyl chloride

[0213] Into a 4000 mL round-bottom flask, was placed 3-(benzylsulfanyl)-5-chloro-2- methoxypyridine (130 g, 490 mmol, 1 equiv), MeCN (2600 mL, 20 V), H 2 O (130 mL, 1 V), acetic acid (294 g, 491 mmol, 10 equiv) and NCS (196 g, 1470 mmol, 3.0 equiv). The resulting solution was stirred for 30 min at 25 °C. The reaction was quenched by the addition 1000 mL of water and extracted with 2 x 1000 mL of ethyl acetate. The combined organics were washed with H 2 O and concentrated. The resulting solution was diluted with 500 mL of diethyl ether and the solids removed by filtration. The filtrate was concentrated, and the residue purified by silica gel column chromatography, eluting with PE/THF (100: 1). The concentrated product was slurried with 300 mL hexane and kept at 0 °C for 1 h. The solids were removed by filtration and dried to give 5-chloro-2-methoxypyridine-3 -sulfonyl chloride (66.5 g, 56% yield) as a white solid.

LCMS: (ES, m/z): 313 [M+l] +

'H-NMR (300 MHz, CDCL, ppm): δ 8.45 (d, J= 2.5 Hz, 1H), 8.23 (d, J= 2.6 Hz, 1H), 4.21

(s, 3H).

Example 3 : Synthesis of 5-cvano-2-methoxypyridine-3-sulfonyl chloride (Intermediate

Synthesis of Int. 3-a: 5-bromo-6-methoxypyridine-3-carbonitrile

[0214] Into a 2 L round-bottom flask were added 6-methoxypyridine-3 -carbonitrile (100 g, 746 mmol, 1 equiv), HOAc (1000 mL), NaOAc (61.1 g, 746 mmol, 1 equiv) and Br 2 (235 g, 1490 mmol, 2 equiv) at room temperature. The resulting mixture was stirred for 48 h at 80 °C. The mixture was allowed to cool to room temperature and was diluted with ice water (3 L). The precipitated solids were collected by filtration and suspended in 1 L of PE:EA=5: 1 which was stirred for 1 h at room temperature. The precipitated solids were collected by filtration and dried to give 5-bromo-6-methoxypyridine-3-carbonitrile (70 g, 44% yield) as a light yellow solid.

Synthesis of Int. 3-b: 5-(benzylsulfanyl)-6-methoxypyridine-3-carbonitrile

[0215] Into a 3 L 3-necked round-bottom flask, were added 5-bromo-6-methoxypyridine-3- carbonitrile (70 g, 330 mmol, 1 equiv), toluene (1400 mL) and benzyl mercaptan (43 g, 347 mmol, 1.05 equiv) at room temperature. To this was added Pd2(dba)3.CHC13 (17 g, 16.5 mmol, 0.05 equiv), Xantphos (19 g, 33 mmol, 0.1 equiv) and DIEA (128 g, 990 mmol, 3 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 90 °C, then was cooled and filtered. The filtrate was concentrated under reduced pressure, and the residue purified by silica gel column chromatography, eluting with PE/THF (10: 1). 5- (Benzylsulfanyl)-6-methoxypyridine-3-carbonitrile (65 g, 77% yield) was obtained as a light brown solid.

Synthesis of Int. 3: 5-cyano-2-methoxypyridine-3-sulfonyl chloride

[0216] Into a 2 L 3-necked round-bottom flask were added 5-(benzylsulfanyl)-6- methoxypyridine-3 -carbonitrile (65 g, 2540 mmol, 1 equiv), MeCN (520 g), H 2 O (260 g) and HC1 (21 mL, 254 mmol, 1 equiv) at room temperature. To this was added NCS (101 g, 762 mmol, 3 equiv) in portions at 0 °C. The resulting mixture was stirred for 0.5 h at room temperature, then was cooled to 0 °C and stirred for 1 h. The precipitated solids were collected by filtration and dried to afford 5-cyano-2-methoxypyridine-3-sulfonyl chloride (25 g, 42% yield) as a white solid.

LC-MS: (ES, m/z): 302 [M-l]'

'H-NMR: (300 MHz, CDC13,ppm ): δ 8.78 (d, J= 2.2 Hz, 1H), 8.51 (d, J= 2.2 Hz, 1H), 4.31 (s, 3H).

Example 4: Synthesis of 5-fluoro-2-methylpyridine-3-sulfonyl chloride (Intermediate 4)

Int. 4-d Intermediate 4

Synthesis of Int. 4-a: tert-butyl N-(2-bromo-5-fluoropyridin-3-yl)-N-(tert- butoxycarbony1carbamate

[0217] Into a 500 mL 3-necked round-bottom flask was placed 2-bromo-5-fluoropyridin-3- amine (20 g, 1 equiv), DCM (220 mL) and TEA (44 mL, 3 equiv). This was followed by the addition of BOC 2 O (57 g, 2.5 equiv) in several batches. The resulting solution was stirred for 14 h at room temperature. The reaction mixture was concentrated, and the residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :5). This resulted in tert-butyl N-(2-bromo-5-fluoropyridin-3-yl)-N-(tert-butoxycarbonyl)carb amate (33 g) as a white solid.

Synthesis of Int. 4-b: tert-butyl N-(tert-butoxycarbonyl)-N-(5-fluoro-2-methylpyridin-3- y1carbamate

[0218] Into a 500 mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed tert-butyl N-(2-bromo-5-fluoropyridin-3-yl)-N-(tert- butoxycarbonyl)carbamate (33 g, 84 mmol, 1 equiv), 1,4-dioxane (150 mL), trimethyl- 1,3,5,2,4,6-trioxatriborinane (21.2 g, 169 mmol, 2 equiv), K 2 CO 3 (35 g, 253 mmol, 3 equiv) and Pd(dppPC12 (3.09 g, 4.2 mmol, 0.05 equiv). The resulting solution was stirred overnight at 110 °C under N2 atmosphere in an oil bath. The reaction mixture was cooled to room temperature and filtered. The filtrate was diluted with 200 mL of H 2 O and extracted with 3 x 100 mL of ethyl acetate. The combined organics were concentrated, and the residue applied to a silica gel column, eluting with ethyl acetate/petr oleum ether (1 : 10-1 :5). Tert-butyl N- (tert-butoxycarbonyl)-N-(5-fluoro-2-methylpyridin-3-yl)carba mate (16 g, 58% yield) was isolated as a yellow solid.

Synthesis of Int. 4-c: 3-bromo-5-fluoro-2 -methylpyridine

[0219] Into a 2 L 3-necked round-bottom flask, was placed tert-butyl N-(tert- butoxycarbonyl)-N-(5-fluoro-2-methylpyridin-3-yl)carbamate (50 g, 153 mmol, 1 equiv) and HBr (1 L, 48%) . This was followed by the addition of a solution of NaNO 2 (11.6 g, 169 mmol, 1.1 equiv) in H 2 O (100 mL) dropwise with stirring at 0-5 °C. The resulting solution was stirred for 30 min in an ice bath. To this was added CuBr (24.2 g, 169 mmol, 1.1 equiv) at 0 °C. The resulting solution was stirred for 1 h at room temperature, then quenched by the addition of 1 L water/ice. The pH of the solution was adjusted to 8 with Na 2 CO 3 and the resulting mixture was extracted with 3x200 mL of ethyl acetate. The combined organics were dried over anhydrous sodium sulfate and concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :50). 3-Bromo-5-fluoro-2- methylpyridine (13 g, 45% yield) was isolated as a white solid.

Synthesis of Int. 4-d: 3-(benzylsulfanyl)-5-fluoro-2-methylpyridine

[0220] Into a 250 mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed 3-bromo-5-fluoro-2-methylpyridine (13 g, 68 mmol, 1 equiv), toluene (130 mL), benzyl mercaptan (12.8 g, 103 mmol, 1.5 equiv), DIEA (17.7 g, 137 mmol, 2 equiv), XantPhos (3.96 g, 6.8 mmol, 0.1 equiv) and Pd 2 (dba) 3 (3.13 g, 3.4 mmol, 0.05 equiv). The resulting solution was stirred for 4 h at 115 °C, then cooled and concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :20) to give 3-(benzylsulfanyl)-5-fluoro-2-methylpyridine (11 g, 69% yield) as a yellow solid.

Synthesis of Intermediate 4: 5-fluoro-2-methylpyridine-3-sulfonyl chloride

[0221] Into a 2000 mL 3-necked round-bottom flask, was placed 3-(benzylsulfanyl)-5-fluoro- 2-methylpyridine (45 g, 193 mmol, 1 equiv), HO Ac (700 mL) and H 2 O (200 mL). This was followed by the addition of NCS (103 g, 772 mmol, 4 equiv), the temperature being maintained under 20 °C. The resulting solution was stirred for 2 h at room temperature. The reaction was quenched by the addition of 700 mL of water, and the resulting solution was extracted with 3 x 300 mL of di chloromethane. The combined organics were concentrated, and the residue applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :10). 5-Fluoro-2-methylpyridine-3-sulfonyl chloride (25.4 g, 63% yield) was obtained as a yellow oil.

LCMS: (ES, m/z): 210 [M+l] +

'H-NMR: (300 MHz, CDCl 3 , ppm): δ 8.72-8.71 (d, J= 3.0 Hz, 1H), 8.11-8.08 (dd, J= 3.0 Hz, 1H), 3.01 (s, 3H).

Example 5: Synthesis of 5-chloro-N-[3.,5-difluoro-4-[1-(lH-imidazol-2-yl)imidazo[l., 5- a] pyridin-6-yl] pyridin-2-yl]-2-methoxypyridine-3-sulfonamide (Intermediate 5)

Int. 2 Intermediate 5

Synthesis of Intermediate 5: N-(3-bromo-2,4-difluorophenyl)-5-chloro-2-methoxypyridine-3- sulfonamide

[0222] To a solution of 3-bromo-2,4-difluoroaniline (5 g, 24 mmol, 1 eq ) in DCM (100 mL) were added 5-chloro-2-methoxypyridine-3-sulfonyl chloride (8.73 g, 36 mmol, 1.5 eq ) and pyridine (5.7 g, 72 mmol, 3 eq ). The resulting solution was stirred for 1 hour at room temperature. The reaction was concentrated and purified by column chromatography over silica gel (eluent: PE:EA = 8: 1) to afford N-(3-bromo-2,4-difluorophenyl)-5-chloro-2- methoxypyridine-3 -sulfonamide (7 g, 70% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 412

'H NMR (300 MHz, Chloroform-d) δ 8.30 (d, J= 2.6 Hz, 1H), 8.05 (d, J= 2.6 Hz, 1H), 7.56 (td, J= 8.9, 5.4 Hz, 1H), 7.28 (d, J= 3.4 Hz, 1H), 6.96 (ddd, J= 9.4, 7.6, 2.1 Hz, 1H), 4.16 (s, 3H). Example 6: Synthesis of 3-ethynyl-2.,4-difluoroaniline & 5-chloro-N-(3-ethynyl-2.,4- difluorophenyl)-2-methoxypyridine-3-sulfonamide (Intermediate 6 & Intermediate 7) I nt. 6/7 -a Intermediate 6

Intermediate 7

Synthesis of Int. 6/7-a: 2,4-difluoro-3-[2-(trimethylsilyl)ethynyl1aniline

[0223] Into a 100 mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 2, 4-difluoro-3 -iodoaniline (1.8 g, 6.9 mmol, 1 equiv), THF (14 mL), DMF (14 mL), TEA (2.1 g, 20.6 mmol, 3 equiv), Cui (0.1 g, 0.7 mmol, 0.1 equiv), Pd(PPh 3 ) 2 C12 (0.5 g, 0.7 mmol, 0.1 equiv) and trimethylsilylacetylene (2.0 g, 20.6 mmol, 3 equiv). The resulting solution was stirred overnight at 60 °C in an oil bath. The reaction mixture was cooled and diluted with 50 mL of water. The resulting solution was extracted with 2 x 50 mL of ethyl acetate and the organic layers combined. This was washed with 3 x 50 ml of water, dried over anhydrous sodium sulfate and concentrated. The residue was applied onto a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 10). The appropriate fractions were combined and concentrated to give 2,4-difluoro-3-[2- (trimethylsilyl)ethynyl]aniline (2 g, 97% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 226 Synthesis of Int. 6: 3-ethynyl- 2,4-difluoroaniline

[0224] Into a 50 mL round-bottom flask, was placed 2,4-difluoro-3-[2- (trimethylsilyl)ethynyl]aniline (1.7 g, 7.5 mmol, 1 equiv). TBAF (15 mL, 1 M in THF) was added and the solution was stirred for 30 min at 25 °C. The resulting mixture was concentrated, and the residue applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :5). The collected fractions were combined and concentrated to give 3-ethynyl-2,4-difluoroaniline (1.05 g, 91% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 154 Synthesis of Int. 7: 5-chloro-N-(3-ethynyl-2,4-difluorophenyl)-2-methoxypyridine- 3- sulfonamide

[0225] Into an 8 mL vial was placed 3-ethynyl-2,4-difluoroaniline (300 mg, 2 mmol, 1 equiv), DCM (10 mL), pyridine (1.5 g, 19.6 mmol, 10 equiv) and 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (569 mg, 2.4 mmol, 1.2 equiv). The resulting solution was stirred for 1 h then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlashTM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 5-95% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give 5-chloro- N-(3-ethynyl-2,4-difluorophenyl)-2-methoxypyridine-3-sulfona mide (560 mg, 80% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 359

Example 7: Synthesis of 2,6-difluoro-3-nitrobenzaldehyde (Intermediate 8)

Example 7: Synthesis of 2,6-difluoro-3 -nitrobenzaldehyde (Intermediate 8)

[0226] To a stirred solution of 2,6-difluorobenzaldehyde (1 g, 7 mmol, 1 equiv) in H 2 SO4 (2.5 mL) was added HNO 3 (1 mL, 68%) dropwise at 0 °C under air atmosphere. The resulting mixture was stirred for 1 h at 0 °C. The reaction was quenched with water/ice at 0 °C, and filtered. The filter cake was washed with water (2 x 10 mL), and the resulting solid was dried under infrared light to afford crude 2,6-difluoro-3-nitrobenzaldehyde (1 g, 76% yield) as a yellow solid which was used in next step directly without further purification.

Example 8: 2,4-difluoro-3-(2-[lH-pyrazolo[3.,4-b]pyridin-5-yl]ethyl)ani line

(Intermediate 9)

Synthesis of Int. 9-a: L3-difluoro-2-iodo-4-nitrobenzene

[0227] To a stirred suspension of l,3-difluoro-2-iodobenzene (25 g, 100 mmol, 1 equiv) in H 2 SO 4 (100 mL) was added fuming HNO 3 (16.4 g, 260 mmol, 2.5 equiv) dropwise at 0 °C. The mixture was stirred for 4 h at room temperature. The reaction mixture was poured into ice water (1 L) and the resulting mixture extracted with EA (3 x 500 mL). The combined organic layers were washed with saturated aqueous NaHCO 3 (2 x 200 mL), brine (2 x 200 mL), dried over anhydrous Na 2 SO 4 and filtered. The filtrate was concentrated under reduced pressure to afford 1,3-difluoro-2-iodo-4-nitrobenzene (28.3 g, 95% yield) as a yellow solid which was used directly in next step without further purification. Synthesis of Int. 9-b: 2,4-difluoro-3 -iodoaniline

[0228] To a stirred suspension of l,3-difluoro-2-iodo-4-nitrobenzene (26 g, 92 mmol, 1 equiv) in EtOH (300 mL) was added SnCl 2 2H 2 O (62 g, 280 mmol, 3 equiv) portion wise at room temperature. The resulting solution was stirred for 1 h at 85 °C, cooled to room temperature and concentrated under reduced pressure. The residue was basified to pH > 9 with saturated aqueous NaOH and the resulting mixture extracted with EtOAc (3 x 500 mL). The combined organic extracts were washed with brine (2 x 200 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EtOAc (9: 1 to 4: 1) to afford 2,4-difluoro-3- iodoaniline (19 g, 80% yield) as a black solid.

Synthesis of Int. 9-c: 5-[2-(trimethylsilyl)ethynyl1-1H-pyrazolo[3,4-b1pyridine

[0229] To a mixture of 5-bromo-lH-pyrazolo[3,4-b]pyridine (7.6 g, 38 mmol, 1 equiv) and trimethylsilylacetylene (11.3 g, 115 mmol, 3 equiv) in THF (180 mL) and DMF (90 mL) under a nitrogen atmosphere was added Cui (730 mg, 3.8 mmol, 0.1 equiv), Pd(PPh3)2C12 (2.7 g, 3.8 mmol, 0.1 equiv) and TEA (38.6 g, 382 mmol, 10 equiv). The resulting mixture was stirred for 5 h at 70 °C under nitrogen atmosphere. The mixture was allowed to cool to room temperature, then poured onto the ice water (500 mL) and the resulting mixture diluted with EtOAc (500 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic extracts were washed with water (3 x 200 mL) and brine (1 x 100 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (20: 1 to 1 :2) to afford 5-[2-(trimethylsilyl)ethynyl]-lH-pyrazolo[3,4-b]pyridine (8 g, 97% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 216

Synthesis of Int. 9-d: 5-ethynyl-1H-pyrazolo[3,4-b1pyridine

[0230] To a stirred solution of 5-[2-(trimethylsilyl)ethynyl]-lH-pyrazolo[3,4-b]pyridine (8.0 g, 37 mmol, 1 equiv) in MeOH (190 mL) was added K2CO3 (10 g, 74 mmol, 2 equiv) in portions under a nitrogen atmosphere. The resulting mixture was stirred for 4 h at 60 °C and then allowed to cool to room temperature. The resulting mixture was filtered and the filter cake washed with MeOH (3 x 100 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1 to 0: 1) to afford 5-ethynyl-lH-pyrazolo[3,4-b]pyridine (4.1 g, 77% yield) as a light yellow solid. LCMS (ES, m/z): [M+H] + : 144

Synthesis of Int. 9-e: 2,4-difluoro-3-(2-[1H-pyrazolo[3,4-b1pyridin-5-yl1ethynyl)an iline

[0231] Into a 250 mL round-bottom flask were added 5-ethynyl-lH-pyrazolo[3,4-b]pyridine (1.5 g, 11 mmol, 1.2 equiv), 2, 4-difluoro-3 -iodoaniline (2.3 g, 8.8 mmol, 1 equiv), Cui (170 mg, 0.9 mmol, 0.1 equiv) and Pd(PPh 3 )2C12 (620 mg, 0.9 mmol, 0.1 equiv), followed by DMF (80 mL) at room temperature. The mixture was sparged with N2 for 10 min and then TEA (2.7 g, 27 mmol, 3 equiv) was added. The resulting mixture was stirred for 1 h at 65 °C and then allowed to cool to room temperature. The reaction was poured into the ice water (200 mL) and diluted with EtOAc (200 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL) and the combined organic layers were washed with water (3 x 100 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1 to 1 : 1) to afford 2,4-difluoro- 3-(2-[1H-pyrazolo[3,4-b]pyridin-5-yl]ethynyl)aniline (1.7 g, 71% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 271

Synthesis of Intermediate 9: 2A-difluoro-3-(2-[1H[-pyrazolo[3,4-b]pyridin-5-yl]ethyl)anil ine

[0232] To a stirred solution of 2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5- yl]ethynyl)aniline (1.1 g, 4 mmol, 1 equiv) in MeOH (180 mL) was added Pd/C (220 mg). The mixture was stirred at 100 °C for 36 h under a hydrogen atmosphere (2MPa). The mixture was allowed to cool down to 30 °C and the resulting mixture filtered. The filter cake was washed with hot MeOH (3 x 150 mL) and the filtrate concentrated under reduced pressure to afford 2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethyl)anil ine (600 mg, 54% yield).

LCMS (ES, m/z): [M+H] + : 275

Example 9: Synthesis of 2,4-difluoro-3-(4.,4.,5.,5-tetramethyl-l.,3.,2-dioxaborolan- 2- vDaniline (Intermediate 10)

Intermediate 10

Synthesis of Intermediate 10: 2,4-difluoro-3-(4,4,5,5-tetramethyl-L3,2-dioxaborolan-2- y1aniline

[0233] 3-Bromo-2,4-difluoroaniline (10 g, 48 mmol, 1 equiv), Pd(dppf)C12 (3.5 g, 4.8 mmol, 0.1 equiv), bis(pinacolato)diboron (18.3 g, 72 mmol, 1.5 equiv) and KOAc (14.2 g, 144.2 mmol, 3 equiv) were dissolved in dioxane (240 mL). The resulting solution was stirred overnight at 100 °C in an oil bath. The reaction mixture was cooled and the solids removed by filtration. The filtrate was concentrated and diluted with DCM (100 mL), then washed with 2 x 100 mL of water and 1 x 100 mL of brine. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1/10). 2,4-Difluoro-3- (4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-yl)aniline (8 g, 65% yield) was isolated as a yellow solid.

LCMS (ES, m/z): [M+H] + : 256

Example 10: Synthesis of 3,5-difluoro-4-iodopyridin-2-amine (Intermediate 11)

Intermediate 11

Synthesis of Intermediate 11 : 3,5-difluoro-4-iodopyridin-2-amine

[0234] To a stirred solution of 3,5-difluoropyridin-2-amine (5 g, 38 mmol, 1 equiv) in THF (200 mL) was added LDA (61 mL, 123 mmol, 3 equiv) dropwise at -78 °C under N2 atmosphere. The solution was stirred for 1.5 h at -78 °C. To the resulting mixture was added a solution of I2 (34 g, 135 mmol, 3.5 equiv) in THF (50 mL) dropwise at -78

°C. This mixture was stirred for additional 0.5 h at -78 °C, then was quenched with saturated aqueous Na 2 S 2 O 3 (100 mL). The reaction was extracted with EA (3 x 50 mL), and the combined organics were washed with brine (2 x 50 mL) and dried over anhydrous sodium sulfate before being concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (3: 1) to afford 3,5-difluoro-4-iodopyridin-2- amine (7.8 g, 79% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 257

Example 11: Synthesis of 4-chloro-3-fluoropicolinaldehyde (Intermediate 12)

Intermediate 12

Synthesis of Intermediate 12: 4-chloro-3-fluoropicolinaldehyde

[0235] To a stirred solution of n-BuLi (38 mL, 92 mmol, 1.2 equiv) in THF (100 mL) was added 2,2,6,6-tetramethylpiperidine (13 g, 92 mmol, 1.2 equiv) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was stirred for 10 min at -78 °C, then 4-chloro-3- fluoropyridine (10 g, 76 mmol, 1 equiv) was added dropwise over 10 min. The resulting mixture was stirred for additional 0.5 h at -78 °C, before DMF (6.1 g, 84 mmol, 1.1 equiv) was added dropwise over 10 min. The resulting mixture was stirred for 0.5 h at -78 °C then quenched by the addition of saturated aqueous NaHCCL solution (50 mL). The resulting mixture was extracted with EA (3 x 100 mL) and the combined organics were washed with brine (1 x 100 mL), then dried over anhydrous sodium sulfate. The solution was concentrated under reduced pressure to give crude 4-chl oro-3 -fluoropicolinaldehy de (10 g) as a yellow oil, which was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 160

Example 12: Synthesis of 5-chloro-N-[2.,4-difluoro-3-([lH-pyrazolo[3.,4-b]pyridin-5- yloxy]methyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 1)

Synthesis of 1-a: (2,6-difluoro-3-nitrophenyl)methanol

[0236] To a stirred solution of 2,6-difluoro-3-nitrobenzaldehyde (1 g, 5.3 mmol, 1 equiv) in MeOH (10 mL) was added NaBH4 (0.4 g, 10.6 mmol, 2 equiv) in portions at 0°C under nitrogen atmosphere and the mixture stirred for 1 h. The resulting mixture was concentrated under vacuum, and the residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford (2,6-difluoro-3-nitrophenyl)methanol (720 mg, 71% yield) as a yellow oil. Synthesis of 1-b: 5-bromo-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1p yridine [0237] To a stirred solution of 5-bromo-lH-pyrazolo[3,4-b]pyridine (1 g, 5 mmol, 1 equiv) in DMF (15 mL) was added NaH (157 mg, 6.5 mmol, 1.3 equiv, 60% in oil) in portions at 0 °C under nitrogen atmosphere. The reaction was stirred for 15 min then SEMC1 (1 g, 6 mmol, 1.2 equiv) was added dropwise at 0 °C. The resulting mixture was stirred for additional 4 h at room temperature. The reaction was quenched with water at room temperature and the aqueous layer extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (2 x 10 mL) and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford 5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1.2 g, 72% yield) as a yellow solid. LCMS (ES, m/z): [M+H] + : 328

Synthesis of 1-c: 1-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5 -ol [0238] To a stirred mixture of 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridine (1 g, 3 mmol, 1 equiv) and B(OMe)i (633 mg, 6 mmol, 2 equiv) in dioxane (20 mL) were added K2CO3 (1 g, 7.6 mmol, 2.5 equiv) and Pd(dppf)C12 (223 mg, 0.3 mmol, 0.1 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 100 °C for 3 h , then was allowed to cool to room temperature. To this was added H 2 O 2 (30%) (207 mg, 6 mmol, 2 equiv) dropwise over 5 min at room temperature. The resulting mixture was stirred for additional 1 h, then concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with hexane/EtOAc (1 : 1) to afford l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5 -ol (0.9 g, 92% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 266

Synthesis of 1-d: 5-[(2,6-difluoro-3-nitrophenyl)methoxy1-1-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine

[0239] To a stirred mixture of (2,6-difluoro-3-nitrophenyl)methanol (1.2 g, 6.4 mmol, 2 equiv) and l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5 -ol (860 mg, 3.2 mmol, 1 equiv) in toluene (50 mL) was added PPI13 (2.1 g, 8 mmol, 2.5 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 20 min, then DEAD (1.7 g, 9.6 mmol, 3 equiv) was added dropwise over 5 min. The resulting mixture was stirred for 5 h at room temperature, then diluted with water (50 mL). This was extracted with EtOAc (2 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure and the residue was purified by silica gel column chromatography, eluting with PE/EtOAc (1 :3) to afford 5-[(2,6-difluoro-3-nitrophenyl)methoxy]-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (560 mg, 31% yield) as a yellow solid. LCMS (ES, m/z): [M+H] + : 437

Synthesis of 1-e: 2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy1methyl1pyrazo lol3,4- b1pyridin-5-yl)oxy1methyl1aniline

[0240] To a stirred solution of 5-[(2,6-difluoro-3-nitrophenyl)methoxy]-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (540 mg, 1.2 mmol, 1 equiv) in EtOH (10 mL) was added SnCl 2 (469 mg, 2.4 mmol, 2 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 1 h at 50 °C, then was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (1 : 1) to afford 2,4-difluoro-3 -[[( 1 -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (280 mg, 55% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 407

Synthesis of 1-f: 5-chloro-N-(2,4-difluoro-3-(((1-((2-(trimethylsilyl)ethoxy)m ethyl)-1H- pyrazolo[3, 4-blpyri din-5 -yl)oxy)methyl)phenyl)-2-methoxypyridine-3 -sulfonamide [0241] To a stirred solution of 2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (260 mg, 0.64 mmol, 1 equiv) in pyridine (5 mL) was added 5 -chi oro-2-methoxypyridine-3 -sulfonyl chloride (232 mg, 0.96 mmol, 1.5 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h, then concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (1 : 1) to afford 5-chloro-A-(2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy]m ethyl]pyrazolo[3,4- b]pyridin-5-yl)oxy]methyl]phenyl)-2-methoxypyridine-3-sulfon amide (360 mg, 92% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 612

Synthesis of Compound 1 : 5-chloro-N-[2,4-difluoro-3-([1H-pyrazolo[3,4-b1pyridin-5- yloxy1methyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0242] To a stirred solution of 5-chloro-A-(2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-2- methoxypyridine-3 -sulfonamide (250 mg, 0.4 mmol, 1 equiv) in DCM (0.5 mL) was added TFA (0.5 mL, 6.7 mmol, 16 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 1 h, then quenched with sat. NaHCO 3 (aq.). The organics were dried over sodium sulphate, concentrated under vacuum and the residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm mobile phase, Mobile Phase A: 0.1% FA in Water, Mobile Phase B: CAN (10% up to 60% in 10 min) Detector, 220 nm. 5-Chloro-A-[2,4-difluoro-3-([lH-pyrazolo[3,4- b]pyridin-5-yloxy]methyl)phenyl]-2-methoxypyridine-3-sulfona mide (73 mg, 37% yield) was isolated as a white solid.

LCMS (ES, m/z): [M+H] + : 482

1 H NMR (300 MHz, DMSO-d6) δ 13.55 (s, 1H), 10.45 (s, 1H), 8.44 (d, J= 2.6 Hz, 1H), 8.23 (d, J= 2.7 Hz, 1H), 8.08-7.98 (m, 2H), 7.84 (d, J= 2.8 Hz, 1H), 7.35 (td, J= 9.0, 6.0 Hz, 1H), 7.11 (t, J = 8.9 Hz, 1H), 5.12 (s, 2H), 3.90 (s, 3H).

Example 13: Synthesis of N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl] phenyl] -5 -fluoro -2- methoxypyridine-3-sulfonamide (Compound 2)

Synthesis of 2-a: (3-amino-2,6-difluorophenyl)methanol

[0243] To a solution of (2,6-difluoro-3-nitrophenyl)methanol (11.6 g, 61 mmol, 1 equiv) in

EtOAc (240 mL) was added Pd/C (10%, 3.8 g). The mixture was hydrogenated at room temperature for 2 h under hydrogen atmosphere using a hydrogen balloon. After completion, the reaction was filtered through a Celite pad and the filtrate was concentrated under reduced pressure to afford crude (3-amino-2,6-difluorophenyl)methanol (9.5 g, 97% yield) as a light brown solid which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 160.

Synthesis of 2-b: 5-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine

[0244] A solution of 5-bromo-3-methyl-lH-pyrazolo[3,4-b]pyridine (5 g, 23.5 mmol, 1 equiv), dihydropyran (9.9 g, 117 mmol, 5 equiv) and DL-camphorsulfonic acid (0.8 g, 3.6 mmol, 0.15 equiv) in THF (50 mL) was stirred for 2 h at 70 °C. The mixture was allowed to cool to room temperature. The resulting mixture was diluted with EtOAc (250 mL) and washed with 100 mL of aqueous NaHCO 3 . The organic layer was washed with water (2 x 100 mL) and brine (100 mL), then dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with EtOAc/PE (1 :4) to afford 5-bromo-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (6.7 g, 96% yield) as a colorless solid.

Synthesis of 2-c: 3-methyl-l-(tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl -L3,2- dioxaborolan-2-yl) -1H-pyrazolo[3,4-b1pyridine

[0245] A solution of 5-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (7 g, 23.6 mmol, 1 equiv), KO Ac (4.6 g, 47.2 mmol, 2 equiv), Pd(dppf)Cl 2 .CH 2 Cl 2 (96 mg, 1.18 mmol, 0.05 equiv) and bis(pinacolato)diboron (9 g, 35.5 mmol, 1.5 equiv) in dioxane (70 mL) was stirred at 90 °C for 2 h under nitrogen atmosphere. The mixture was allowed to cool to room temperature; the resulting mixture was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 344.

Synthesis of 2-d: 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-ol

[0246] To the reaction solution of (3-methyl-l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl) pyrazolo[3,4-b]pyridine (23.6 mmol, 1 equiv) were added THF (80 mL), NaOH (2%, 80 mL) and H 2 O 2 (30%, 8 g, 70.8 mmol, 3 equiv) dropwise at room temperature. The resulting mixture was stirred at room temperature for 2 h under air atmosphere. The reaction was quenched with aqueous Na 2 SO 4 (200 mL). The resulting mixture was extracted with EtOAc (3 x 150 mL). The combined organic layers were washed with water (2 x 150 mL) and brine (150 mL), then dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with EtOAc/PE (1 : 1) to afford 3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-ol (4.5 g, 81% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 234.

Synthesis of 2-e: 2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyrid in-5- yl]oxy]methyl)aniline

[0247] A solution of 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (5.4 g, 23 mmol, 1 equiv), TMAD (5.1 g, 29 mmol, 1.3 equiv), (3-amino-2,6-difluorophenyl)methanol (4 g, 25 mmol, 1.1 equiv) and PPh 3 (7.8 g, 29 mmol, 1.3 equiv) in DCM (60 mL) was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was diluted with water (100 mL) and extracted with DCM (3 x 100 mL). The combined organic layers were washed with water (2 x 150 mL) and brine (150 mL), then dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with EtOAc/PE (1 : 1) to afford 2,4-difluoro-3-([[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b] pyridin-5-yl]oxy]methyl)aniline (6.4 g, 74% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 375.

Synthesis of 2-f: N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1py ridin-5- ylloxylmethyl) phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0248] To a stirred solution of 2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (5.5 g, 14.7 mmol, 1 equiv) and pyridine (4.65 g, 58 mmol, 4 equiv) in DCM (110 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (4.97 g, 22 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for 4 h at room temperature, then diluted with water (100 mL). The mixture was extracted with DCM (3 x 100 mL) and the combined organic layers were washed with water (2 x 150 mL) and brine (150 mL), then dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with EtOAc/PE (0-50%) to afford N-[2,4-difluoro-3-([[3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5- fluoro-2-methoxypyridine-3- sulfonamide (6.4 g, 77% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 564.

Synthesis of Compound 2: N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1oxy)methyl1phenyl1-5 -fluoro -2- methoxypyridine-3 -sulfonamide

[0249] To a stirred mixture of N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl) phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide(6.4 g, 11.6 mmol, 1 equiv) was added HC1 in MeOH (64 mL, 256 mmol, 22 equiv ,4 M) dropwise at 0 - 5 °C . The resulting mixture was stirred for 1 h at room temperature. After concentration under vacuum, the residue was neutralized to pH 7 with NH 3 .H 2 O (5%) and this was dissolved in DMF (30 mL). The crude product was purified by Prep-HPLC with the following conditions: Column: Welch XB-C18, 50x250mm , mobile phase: 200 mL/min, Mobile Phase A: 0.1%FA in Water, Mobile Phase B: ACN (20% up to 65% in 30 min). N- [2,4-Difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]o xy)methyl]phenyl]-5-fluoro -2- methoxypyridine-3 -sulfonamide (5.1 g, 94%) was isolated as a white solid.

LCMS (ES, m/z): [M+H] + : 480.

1 H NMR (300 MHz, DMSO-d6) δ 13.09 (s, 1H), 10.36 (s, 1H), 8.44 (d, J= 3.0 Hz, 1H), 8.18 (d, J= 2.7 Hz, 1H), 7.98 (dd, J= 13, 3.0 Hz, 1H), 7.86 (d, J= 2.8 Hz, 1H), 7.38 (td, J= 8.9, 6.0 Hz, 1H), 7.16 (td, J= 9.0, 1.6 Hz, 1H), 5.14 (s, 2H), 3.91 (s, 3H), 2.48 (s, 3H).

Example 14: Synthesis of 5-chloro-N-[2.,4-difluoro-3-([lH-pyrazolo[3.,4-b]pyridin-5- ylamino]methyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 3)

Compound 3

Synthesis of 3-a: 2,6-difluoro-3-nitrobenzaldehyde

[0250] To a stirred solution of 2,6-difluorobenzaldehyde (1.0 g, 7.0 mmol, 1 equiv) in H 2 SO4 (2.5 mL) was added HNO3 (1.0 mL, 68%) dropwise at 0 °C under air atmosphere.

The resulting mixture was stirred for 1 h at 0 °C and then poured onto ice water. The resulting mixture was filtered and the filter cake washed with water (2 x 10 mL). The resulting solid was dried under infrared light to afford crude 2,6-difluoro-3-nitrobenzaldehyde (1 g, 76% yield) as a yellow solid which was used in next step directly without further purification. Synthesis of 3-b: N-[(2,6-difluoro-3-nitrophenyl)methyl]-1H-pyrazolo[3,4-b]pyr idin-5-amine [0251] NaBH(OAc) 3 (566 mg, 2.6 mmol, 2 equiv) was added portion wise to a stirred solution of 2, 6-difluoro-3 -nitrobenzaldehyde (250 mg, 1.3 mmol, 1 equiv) and 1H- pyrazolo[3,4-b]pyri din-5 -amine (179 mg, 1.3 mmol, 1 equiv) in MeOH (10 mL) and HO Ac (8 mg, 0.13 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The reaction mixture was stirred for 3 h and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford A-[(2,6- difluoro-3-nitrophenyl)methyl]-lH-pyrazolo[3,4-b]pyridin-5-a mine (250 mg, 61% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 306

Synthesis of 3-c: N-[(3-amino-2,6-difluorophenyl)rnethyl]-1H-pyrazolo[3,4-b]py ridin-5- amine

[0252] Pd/C (14 mg, 0.013 mmol, 0.02 equiv, 10% w/w) was added portion wise to a stirred solution of 7V-[(2,6-difluoro-3-nitrophenyl)methyl]-lH-pyrazolo[3,4-b]py ridin-5-amine (200 mg, 0.65 mmol, 1 equiv) and HCOONH 4 (206 mg, 3.3 mmol, 5 equiv) in MeOH (10 mL) at room temperature. The reaction mixture was stirred for 2 h at room temperature and then filtered. The filter cake was washed with MeOH (2 x 10 mL) and the filtrate concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (1 : 1) to afford A-[(3-amino-2,6-difluorophenyl)methyl]-lH- pyrazolo[3,4-b]pyri din-5 -amine (160 mg, 88% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 276

Synthesis of Compound 3: 5-chloro-N-[2,4-difluoro-3-([1H-pyrazolo[3,4-b1pyridin-5- ylamino1methyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0253] To a stirred solution of A-[(3-amino-2,6-difluorophenyl)methyl]-lH-pyrazolo[3,4- b]pyridin-5-amine (200 mg, 0.73 mmol, 1 equiv) in pyridine (6 mL) was added 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (350 mg, 1.5 mmol, 2 equiv) portion wise at room temperature under a nitrogen atmosphere. The reaction was stirred for 2 h at 50 °C and then concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 10-50% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N-[2,4- difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-ylamino]methyl)phen yl]-2-methoxypyridine-3- sulfonamide (57 mg, 16% yield) as a white solid.

LCMS (ES, m/z): [M+H]+ : 481

1 H NMR (300 MHz, DMSO-d6) δ 13.17 (s, 1H), 10.31 (s, 1H), 8.33 (d, J= 2.6 Hz, 1H), 8.10 (d, J= 2.6 Hz, 1H), 7.97 (d, J= 2.6 Hz, 1H), 7.84 (d, J= 1.4 Hz, 1H), 7.17 (d, J = 7.1 Hz, 2H), 6.96 (t, J= 9.1 Hz, 1H), 5.99 (t, J= 5.9 Hz, 1H), 4.21 (d, J= 5.7 Hz, 2H), 3.85 (s, 3H).

Example 15: Synthesis of 5-chloro-N-(2.,4-difluoro-3-[lH-pyrazolo[3.,4-b]pyridin-5- ylmethoxy]phenyl)-2-methoxypyridine-3-sulfonamide (Compound 4) Synthesis of 4-a: 2,6-difluorophenyl ethyl carbonate

[0254] Ethyl carb onochlori date (3.7 g, 34 mmol, 1.5 equiv) was added to a solution of 2,6- difluorophenol (3.0 g, 23 mmol, 1 equiv) in DCM (30 mL) and NEt 3 (4.7 g, 46 mmol, 2 equiv) at 0 °C. The resulting solution was stirred at room temperature for 1 h and then quenched by the addition of ice water (10 mL). The resulting mixture was extracted with DCM (3 x 40 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure to afford crude 2,6-difluorophenyl ethyl carbonate (3.6 g, 77% yield) as a light yellow oil, which was used directly in next step without further purification.

LCMS (ES, m/z): [M+H] + : 203

Synthesis of 4-b: 2,6-difluoro-3 -nitrophenyl ethyl carbonate

[0255] HNO3 (13 mL) was added to a solution of 2,6-difluorophenyl ethyl carbonate (3.5 g, 17 mmol, 1 equiv) in H 2 SO4 (5 mL) at 0 °C. The resulting solution was stirred at -10 °C for 30 min and then diluted with H 2 O (20 mL). The resulting solution was extracted with EtOAc (3 x 20 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure to afford crude 2, 6-difluoro-3 -nitrophenyl ethyl carbonate (4.8 g) as light yellow oil, which was used directly in next step without further purification.

LCMS (ES, m/z): [M+H] + : 248

Synthesis of 4-c: 2, 6-difluoro-3 -nitrophenol

[0256] Into a 250 mL round-bottom flask, was placed 2, 6-difluoro-3 -nitrophenyl ethyl carbonate (4.7 g, 19 mmol, 1 equiv), NaHCO 3 (2.4 g, 28 mmol, 1.5 equiv), MeOH (20 mL) and H 2 O (20 mL). The resulting solution was stirred at room temperature overnight. The solids were filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE/EtOAc (3: 1), to afford 2,6-difluoro-3- nitrophenol (2.3 g, 69% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 176

Synthesis of 4-d: 5-bromo-l-[[2-(trimethylsilyl)ethoxy1 methyl1pyrazolo[3,4-b1pyridine [0257] NaH (0.6 g, 25.2 mmol, 2 equiv) was added portion wise to a solution of 5-bromo- IH-pyrazolo [3,4-b]pyridine (2.5 g, 12.6 mmol, 1 equiv) in DMF (50 mL) at 0 °C. SEMC1 (3.2 g, 19 mmol, 1.5 equiv) was added at 0 °C and the reaction mixture stirred at room temperature for 40 minutes, then quenched with ice water (100 mL). The mixture was extracted with EA (3 x 100 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PEZEA (5: 1), to afford 5-bromo-l-[[2-(trimethylsilyl)ethoxy] methyl]pyrazolo[3,4-b]pyridine (2.1 g, 50% yield) as light yellow oil. LCMS (ES, m/z): [M+H] + : 328

Synthesis of 4-e: methyl-l-[[2-(trimethylsilyl)ethoxy1 methyl1pyrazolo[3,4-b1pyridine-5- carb oxy late

[0258] Into a 50 mL s 6 aled tube purged and maintained with an inert atmosphere of nitrogen was placed 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]p yridine (2.1 g, 6.4 mmol, 1 equiv), Pd(dppf)C12 (0.94 g, 1.28 mmol, 0.2 equiv), TEA (3.2 g, 32 mmol, 5 equiv), MeOH (20 mL), dioxane (5 mL), and CO. The resulting solution was stirred at 100 °C for 10 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PEZEA (7: 1), to afford methyl-l-[[2-(trimethylsilyl)ethoxy] methyl]pyrazolo[3,4-b]pyridine-5-carboxylate (1.4 g, 71% yield) as light yellow oil.

LCMS (ES, m/z): [M+H] + : 308

Synthesis of 4-f: (l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin- 5-yl)methanol [0259] LAH (0.18 g, 4.7 mmol, 1.2 equiv) was added to a stirred solution of methyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-5- carboxylate (1.2 g, 3.9 mmol, 1 equiv) in THF (10 mL) at 0 °C. The reaction mixture was stirred at 0 °C for 30 min and then quenched by the addition of ice water (15 mL). The solution was adjusted to pH 9 with aqueous NaOH (2 M, 10 mL). The mixture was extracted with EA (3 x 50 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PEZEA (5: 1), to afford (1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)me thanol (460 mg, 42% yield) as light yellow oil.

LCMS (ES, m/z): [M+H] + : 280

Synthesis of 4-g: 5-(2, -difluoro-3-nitrophenoxymethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine [0260] PPh 3 (470 mg, 1.8 mmol, 1.2 equiv) and DEAD (390 mg, 2.2 mmol, 1.5 equiv) were added to a solution of (1-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin- 5- yl)methanol (42- mg, 1.5 mmol, 1 equiv) and 2, 6-difluoro-3 -nitrophenol (260 mg, 1.5 mmol, 1 equiv) in toluene (12 mL) under an atmosphere of nitrogen. The reaction mixture was stirred at 80 °C for 2 h and then concentrated. The residue was purified by silica gel chromatography, eluting with PEZEA (2: 1), to afford 5-(2, -difluoro-3-nitrophenoxymethyl)- l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (320 mg, 48% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 437 Synthesis of 4-h: 2,4-difluoro-3-[(l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazol o[3,4- b1pyridin-5-yl)methoxy1aniline

[0261] Zn (90 mg, 1.4 mmol, 2 equiv) was added to a solution of 5-(2,6-difluoro-3- nitrophenoxymethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyra zolo[3,4-b]pyridine (300 mg, 0.7 mmol, 1 equiv) in HOAc (6 mL) and then stirred at 25 °C overnight. The resulting mixture was concentrated and the residue diluted with water (5 mL). The solution was adjusted to pH 8 with saturated aqueous NaHCO 3 , and then extracted with EA (3 x 15 mL). The combined organic extracts were dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE/EA (L 1), to afford 2,4-difluoro-3-[(1-[[2-(trimethylsilyl)ethoxy]methyl]pyrazol o[3,4- b]pyridin-5-yl)methoxy]aniline (138 mg, 49% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 407

Synthesis of 4-i: 5-chloro-N-[2,4-difluoro-3-[(l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)me thoxy]phenyl]-2- methoxypyridine-3 -sulfonamide

[0262] Into an 8 mL vial was placed 2,4-difluoro-3-[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)me thoxy]aniline (120 mg, 0.3 mmol, 1 equiv), DCM (3 mL), pyridine (230 mg, 3 mmol, 10 equiv) and 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (110 mg, 0.45 mmol, 1.5 equiv). The reaction mixture was stirred at 25 °C for 30 min and then diluted with water (15 mL). The mixture was extracted with DCM (3 x 15 mL) and the combined organic extracts dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford crude 5-chloro-A-[2,4- difluoro-3-[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5- yl)methoxy]phenyl]-2-methoxypyridine-3-sulfonamide (150 mg, 83% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 612

Synthesis of Compound 4: 5-chloro-N-(2,4-difluoro-3-[iH-pyrazolo[3,4-b1pyridin-5- ylmethoxy1phenyl)-2-methoxypyridine-3-sulfonamide

[0263] A solution of 5-chloro-A-[2,4-difluoro-3-[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo [3,4-b]pyridin-5-yl)methoxy]phenyl]-2- methoxypyridine-3 -sulfonamide (130 mg) in DCM (2 mL) and TFA (2 mL) was stirred at room temperature for 45 min and then then quenched with ice water (10 mL). The solution was adjusted to pH 8 with saturated aqueous NaHCO 3 and extracted with DCM (3 x 10 mL). The combined organic extracts were dried over anhydrous Na 2 SO 4 , filtered, and concentrated. The residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-50% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N -(2,4-difluoro-3-[lH-pyrazolo[3,4- b]pyridin-5-ylmethoxy]phenyl)-2-methoxypyridine-3-sulfonamid e (32 mg, 31% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 482

1 H NMR (300 MHz, DMSO-d6) δ 13.72 (s, 1H), 10.31 (s, 1H), 8.46 (s, 2H), 8.12 (s, 2H), 7.92 (s, 1H), 7.05 (d, J= 10.2 Hz, 1H), 6.91 (s, 1H), 5.19 (s, 2H), 3.91 (s, 3H).

Example 16: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[5H.,6H.,8H-iniidazo[2,l- c][l.,4]oxazin-3-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfon amide (Compound 5)

Synthesis of 5-a: 3-ethynyl-2,4-difluoroaniline

[0264] Into a 50 mL s 6 aled tube purged and maintained with an inert atmosphere of nitrogen, were placed 3-bromo-2,4-difluoroaniline (2 g, 9.7 mmol, 1 equiv), trimethylsilylacetylene (1.4 g, 14.5 mmol, 1.5 equiv), Cui (180 mg, 0.97 mmol, 0.1 equiv), Pd2(dba)3 (560 mg, 0.48 mmol, 0.05 equiv), K2CO3 (4.0 g, 29 mmol, 3 equiv), t-BusP/Toluene (390 mg, 0.97 mmol, 0.1 equiv), and THF (15 mL). The resulting solution was stirred for 24 h at 100 °C and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (10: 1) to afford 3-ethynyl-2,4-difluoroaniline (900 mg, 70% yield) as a red solid.

Synthesis of 5-b: 2,4-difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c1[L41oxazin-3- yllethynyl aniline

[0265] Into a 30 mL s 6 aled tube purged and maintained with an inert atmosphere of nitrogen, were placed 3-ethynyl-2,4-difluoroaniline (300 mg, 2.0 mmol, 1 equiv), 3-bromo-5H,6H,8H- imidazo[2,l-c][l,4]oxazine (400 mg, 2.0 mmol, 1 equiv), Cui (37 mg, 0.2 mmol, 0.1 equiv), Pd2(dba) 3 (110 mg, 0.1 mmol, 0.05 equiv), K2CO3 (810 mg, 5.9 mmol, 3 equiv), t- BusP/Toluene (79 mg, 0.2 mmol, 0.1 equiv) and THF (5 mL). The reaction mixture was stirred for 24 h at 100 °C and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 : 1) to afford 2,4- difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c][l,4]oxazin-3-yl]ethyn yl)aniline (190 mg, 80% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + :276

Synthesis of 5-c: 2,4-difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c1[L41oxazin-3-yl1et hyl)aniline [0266] Into a 25 mL pressure tank reactor, was placed 2,4-difluoro-3-(2-[5H,6H,8H- imidazo[2,l-c][l,4]oxazin-3-yl]ethynyl)aniline (70 mg, 0.25 mmol, 1 equiv), 10% Pd/C (7 mg, 0.025 mmol, 0.1 equiv) and MeOH (5 mL). The mixture was stirred at 100 °C for 12 hours under H 2 (30 atm). The mixture was filtered and the filtrate concentrated under reduced pressure to afford 2,4-difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c][l,4]oxazin-3-yl]e thyl)aniline (70 mg, 90% yield) as a light yellow solid that was used directly in the next step without further purification.

LCMS (ES, m/z): [M+H] + : 280

Synthesis of Compound 5: 5-chloro-N-[2,4-difluoro-3-(2-[5H,6H,8H-imidazo[2,l- c][1,4oxazin-3-yl1ethyl)phenyl1-2-methoxypyridine-3-sulfonam ide

[0267] 5 -Chloro-2-methoxypyridine-3-sulfonyl chloride (60 mg, 0.25 mmol, 1 equiv) was added to a solution of 2,4-difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c][l,4]oxazin-3- yl]ethyl)aniline (70 mg, 0.25 mmol, 1 equiv) in pyridine (40 mg, 0.5 mmol, 2 equiv) and DCM (5 mL). The reaction mixture was stirred for 1 h at room temperature and then concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 35-70% MeCN / 0.05% aqueous NH4HCO3; Detector: 220 nm; to afford 5-chloro-N-[2,4- difluoro-3-(2-[5H,6H,8H-imidazo[2,l-c][l,4]oxazin-3-yl]ethyl )phenyl]-2-methoxypyridine- 3-sulfonamide (78 mg, 97% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 485 1 H NMR (300 MHz, DMSO-d6) δ 10.28 (s, 1H), 8.51 (s, 1H), 8.00 (s, 1H), 7.12-6.98 (m, 2H), 6.48 (s, 1H), 4.66 (s, 2H), 3.96 (s, 5H), 3.82 (s, 2H), 2.81-2.50 (m, 4H).

Example 17: Synthesis of 5-chloro-N-[2.,4-difluoro-3-([lH-pyrazolo[3.,4-b]pyridin-5- ylmethyl]amino)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 6)

Synthesis of 6-a: 5-bromo-l-[[2-(trimethylsilyl)ethoxy1 methyl1pyrazolo[3,4-b1pyridine [0268] NaH (1.2 g, 50 mmol, 2 equiv) was added portion wise to a solution of 5-bromo-lH- pyrazolo [3, 4-b] pyridine (5.0 g, 25 mmol, 1 equiv) in DMF (100 mL) at 0 °C. SEMCI (6.3 g, 38 mmol, 1.5 equiv) was then added at 0 °C and the reaction mixture stirred for 40 min at room temperature. The reaction was quenched with ice water (20 mL) and extracted with EA (3 x 200 mL). The combined organic extracts were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (5: 1), to afford 5-bromo-l-[[2-(trimethylsilyl)ethoxy] methyl]pyrazolo[3,4-b]pyridine (5.2 g, 63% yield) as light yellow oil.

LCMS (ES, m/z): [M+H]+ : 328

Synthesis of 6-b: l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1 pyridine-5-carbonitrile [0269] DMF (20 mL) was added to a flask containing 5-bromo-l-[[2-

(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (2.0 g, 6.0 mmol, 1 equiv), Zn(CN)2 (1.0 g, 9.0 mmol, 1.5 equiv), Pd(PPh3)4 (1.4 g, 1.2 mmol, 0.2 equiv) under an atmosphere of N2. The reaction mixture was stirred at 90 °C for 3 h and then concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE:EA (7: 1), to afford l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b] pyridine-5 -carbonitrile (1.2 g, 74% yield) as brown oil.

LCMS (ES, m/z): [M+H] + : 275

Synthesis of 6-c: l-(l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridi n-5- yDmethanamine

[0270] Into a 50 mL s 6 aled tube were placed 1 -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-5-carb onitrile (1.1 g, 4.0 mmol, 1 equiv), EtOH (30 mL), NH3.H 2 O (0.5 mL), Raney-Ni (0.2 g) and the mixture was stirred at room temperature overnight under an atmosphere of H 2 (10 atm). The mixture was filtered carefully and the filtrate was concentrated to afford l-(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)me thanamine (0.8 g, 72% yield) as a brown solid which was used in the next step without further purification.

LCMS (ES, m/z): [M+H] + : 279

Synthesis of 6-d: 5-chloro-N-(2,4-difluoro-3-[[(l-[[2-

(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl )methyl1amino1phenyl)-2- methoxypyridine-3 -sulfonamide

[0271] DMF (15 mL) was added to a l-(1-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridin-5-yl)methanamine (500 mg, 1.8 mmol, 1 equiv), A-(3-bromo-2, 4-difluorophenyl)- 5-chloro-2-methoxypyridine-3-sulfonamide (740 mg, 1.8 mmol, 1 equiv), BINOL (770 mg, 2.7 mmol, 1.5 equiv), Cu (110 mg, 1.8 mmol, 1 equiv), Cui (340 mg, 1.8 mmol, 1 equiv), CS2CO3 (1.2 g, 3.6 mmol, 2 equiv) under an atmosphere of N2 and the resulting solution was stirred for at 120 °C for 6 h. The reaction mixture was cooled to room temperature and then filtered. The filtrate was concentrated under reduced pressure and the residue purified by prep-HPLC with the following conditions Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 10-65% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N -(2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy]methyl]pyra zolo[3,4- b]pyridin-5-yl)methyl]amino]phenyl)-2-methoxypyridine-3-sulf onamide (72 mg, 6.6% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 611

Synthesis of Compound 6: 5-chloro-N-12,4-difluoro-3-([1H-pyrazolo[3,4-b1pyridin-5- ylmethyl1amino)phenyl1-2-methoxypyridine-3-sulfonamide [0272] A solution of 5-chloro-A-(2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)me thyl]amino]phenyl)-2- methoxypyridine-3 -sulfonamide (59 mg, 0.097 mmol, 1 equiv) in DCM (2 mL) and TFA (2 mL) was stirred at room temperature for 30 min. The reaction mixture was quenched with ice water (10 mL). The solution was adjusted to pH 8 with saturated aqueous NaHCO, and extracted with DCM (3 x 10 mL). The combined organic extracts were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18; 50 x 250 mm, 10 μm, Mobile Phase: 10-50% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5- chloro-N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-ylmet hyl]amino)phenyl]-2- methoxypyridine-3 -sulfonamide (7 mg, 15% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 481

1 H NMR (300 MHz, DMSO-d6) δ 13.56 (s, 1H), 10.03 (s, 1H), 8.45-8.37 (m, 2H), 8.08 (s, 1H), 7.99 (s, 1H), 7.91 (d, J= 2.6 Hz, 1H), 6.81 (t, J= 10.2 Hz, 1H), 6.56-6.40 (m, 1H), 6.03 (s, 1H), 4.43 (d, J= 6.9 Hz, 2H), 3.88 (3H, s)

Example 18: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[lH-pyrazolo[3.,4-b]pyridin-5 yl]ethyl)phenyl]-2-methoxypyridine-3- sulfonamide (Compound 7) p

Synthesis of Compound 7: 5-chloro-N-[2,4-difluoro-3-(2-[ lH-pyrazolo[3,4-b]pyridin-5- yl1ethyl)phenyl1-2-methoxypyridine-3- sulfonamide

[0273] To a solution of 2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethyl)anil ine (500 mg , 1.8 mmol, 1 equiv ) in DCM (10 mL) was added pyridine (430 mg, 5.5 mmol, 3 equiv ) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (660 mg, 2.7 mmol, 1.5 eq ). The reaction was stirred at room temperature for 1 h and then the solution was concentrated. The residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase 10-65% MeCN / 0.1% aqueous formic acid;

Detector: 220 nm; to afford 5-chloro-N -[2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5- yl]ethyl)phenyl]-2-methoxypyridine-3- sulfonamide (200 mg, 22% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 480

^ NMR (300 MHz, DMSO-d 6 ) δ 13.51 (s, 1H), 10.23 (s, 1H), 8.51 (d, J= 2.6 Hz, 1H), 8.21 (d, J= 2.1 Hz, 1H), 8.03 (d, J= 1.2 Hz, 1H), 7.99 (d, J= 2.6 Hz, 1H), 7.83 (d, J= 2.1 Hz, 1H), 7.11 (td, J= 8.9, 5.9 Hz, 1H), 6.94 (td, J= 9.0, 1.6 Hz, 1H), 3.95 (s, 3H), 2.87 (m, 4H).

Example 19: Synthesis of 5-[2-[3-(5-chloro-2-methoxypyridine-3-sulfonamido)-2.,6- difluorophenyl]ethyl]-N- methyl-1H-pyrazole-3-carboxamide (Compound 8) Synthesis of 8-a: ethyl 5-iodo-1H-pyrazole-3-carboxylate

[0274] To a suspension of ethyl 3-amino-lH~pyrazole-5-carboxylate (10 g) in diiodomethane (200 mL) at -10 °C under Nz atmosphere, was slowly added isoamyl nitrite (78 ml) over 20 min. The mixture was stirred at room temperature for 1 h, then heated at 90 °C and stirred at that temperature for Ih. After cooling, the mixture was diluted with AcOEt and washed with 1a 2 SO 4 and IM HC1. The organic phases collected were dried and evaporated under vacuum. The residue was purified by silica chromatography eluting with 0-30% AcOEt/petroleum ether to give the ethyl 5-iodo-lH-pyrazole-3-carboxylate (7.5 g) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 267

Synthesis of 8-b: ethyl 5-iodo-l-(4-methoxybenzyl)-1H-pyrazole-3-carboxylate [0275] To a suspension of NaH (1.7 g, 42 mmol, 1.5 equiv, 60%) in DMF (10 ml) was added a solution of ethyl 5-iodo-lH-pyrazole-3-carboxylate (7.5 g, 28 mmol, 1 equiv) in DMF (100 ml) dropwise over 10 min. The resulting solution was stirred for 30 min at room temperature, then a solution of PMB-C1 (5.3 g, 34 mmol, 1.20 equiv) in DMF (10 ml) was added over 10 min. The resulting solution was stirred overnight at room temperature. The reaction mixture was quenched with water/ice (100 ml). The mixture was extracted three times with 200 ml of ether and the organic layers combined and dried over Na 2 SO 4 . The residue was purified by eluting through a silica gel column with 1 :5 EtOAc/PE. This resulted in ethyl 5-iodo-l-(4- methoxybenzyl)-lH-pyrazole-3-carboxylate (6.5 g, 60% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 387

Synthesis of 8-c: ethyl 5-[2-(3-amino-2,6-difhjorophenyl)ethynyl]-l-[(4- methoxyphenyl methyl] pyrazole- 3 -carboxylate

[0276] Into a 10 mL microwave tube was placed ethyl 5-iodo-l-[(4- methoxyphenyl)methyl]pyrazole-3-carboxylate (300 mg, 0.8 mmol, 1 equiv), 3-ethynyl-2,4- difluoroaniline (119 mg, 0.8 mmol, 1 equiv), Pd(dppf)C12 (57 mg, 0.08 mmol, 0.1 equiv), DBU (12 mg, 0.08 mmol, 0.1 equiv), CS 2 CO 3 (253 mg, 0.8 mmol, 1 equiv) and DMF (3 mL). The resulting solution was stirred for 10 min at 150 °C in a microwave reactor. The residue was purified on silica gel eluting with ethyl acetate/petroleum ether (1 :2). This gave ethyl 5- [2-(3-amino-2,6-difluorophenyl) ethynyl]-l-[(4-methoxyphenyl)methyl]pyrazole-3- carboxylate (200 mg, 63% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 412

Synthesis of 8-d: ethyl 5-[2-(3-amino-2,6-difluorophenyl)ethyl]-l-[(4- methoxyphenyl)methyl]pyrazole- 3 -carboxylate

[0277] Into a 25 mL pressure tank reactor, was placed ethyl 5-[2-(3-amino-2,6- difluorophenyl)ethynyl] -l-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate (200 mg, 0.5 mmol, 1 equiv), MeOH (5 mL), 10% Pd/C (20 mg, 0.1 equiv) and H 2 (2 MPa). The resulting solution was stirred for 12 h at 60 °C in an oil bath. The solids were filtered off and the resulting mixture was concentrated under vacuum to give ethyl 5-[2-(3-amino-2,6- difluorophenyl)ethyl]-l-[(4-methoxyphenyl)methyl]pyrazole-3- carboxylate (171 mg, 85% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 416

Synthesis of 8-e: ethyl 5-[2-[3-(5-chloro-2-methoxypyridine-3-sulfonamido)-2,6- difluorophenyllethyll -l-l(4-methoxyphenyl)methyl1pyrazole-3-carboxylate [0278] Into a 25 mL 3-necked round-bottom flask, was placed ethyl 5-[2-(3-amino-2,6- difluorophenyl)ethyl]- l-[(4-methoxyphenyl)methyl]pyrazole-3-carboxylate (171 mg, 0.4 mmol, 1 equiv), pyridine (98 mg, 1.2 mmol, 3 equiv), 5-chloro-2-methoxypyridine-3-sulfonyl chloride (100 mg, 0.4 mmol, 1 equiv) and DCM (10 mL). The resulting solution was stirred for 2 h at room temperature. The mixture was concentrated under vacuum and the residue purified on a silica gel column eluting with ethyl acetate/petroleum ether (1:3). Ethyl 5-[2-[3- (5-chloro- 2-methoxypyridine-3-sulfonamido)-2,6-difluorophenyl]ethyl]-l -[(4- methoxyphenyl)methyl]pyrazole-3-carboxylate (194 mg, 76% yield) was isolated as a white solid.

LCMS (ES, m/z): [M+H] + : 621

Synthesis of 8-f: 5-[2-[3-(5-chloro-2-methoxypyridine-3-sulfonamido)-2,6- difluorophenyllethyll- l-l(4-methoxyphenyl)methyl1-N-methylpyrazole-3-carboxamide [0279] Into a 25 mL 3-necked round-bottom flask, was placed ethyl 5-[2-[3-(5-chloro-2- methoxypyridine- 3-sulfonamido)-2,6-difluorophenyl]ethyl]-l-[(4- methoxyphenyl)methyl]pyrazole-3-carboxylate (194 mg, 0.3 mmol, 1 equiv), and methylamine solution (5 mL, 30% in EtOH). The resulting solution was stirred for 12 h at room temperature. The mixture was concentrated under vacuum to give 5-[2-[3-(5-chloro-2- methoxypyridine-3-sulfonamido)-2,6-difluorophenyl]ethyl]-l-[ (4-methoxyphenyl)methyl]-N- methylpyrazole-3 -carboxamide (130 mg, 69% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 606

Synthesis of Compound 8: 5-[2-[3-(5-chloro-2-methoxypyridine-3-sulfonamido)-2,6- difluorophenyllethyll-N- methyl-1H-pyrazole-3-carboxamide

[0280] 5-[2-[3-(5-Chloro-2-methoxypyridine-3-sulfonamido)-2,6-diflu orophenyl]ethyl]-l- [(4-methoxyphenyl)methyl]-N-methylpyrazole-3-carboxamide (130 mg, 0.2 mmol, 1 equiv) in trifluoroacetic acid (3 ml) was stirred for 2 h at 70 °C in an oil bath. The resulting mixture was concentrated under vacuum. The crude product (200 mg) was purified by Flash-Prep- HPLC with the following conditions: welch Vltimate XB-C18, 50 x 250 mm, 10 μm mobile phase, Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN (10-60% over 15 min) ; Detector, 220 nm. 5-[2-[3-(5-Chloro-2-methoxypyridine-3-sulfonamido)-2,6- difluoropheny 1 ] ethyl ] -N -methyl - 1 H-py razol e-3 - carboxamide (43 mg, 41 % yield) was isolated as a white solid.

LCMS (ES, m/z): [M+H] + : 486

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.25 (s, 1H), 8.50 (d, J= 2.6 Hz, 1H), 8.01 (d, J= 2.6 Hz, 1H), 7.96 (s, 1H), 7.14 (td, J= 8.7, 5.9 Hz, 1H), 7.07-6.95 (m, 1H), 6.28 (s, 1H), 3.93 (s, 3H), 2.86 (s, 2H), 2.72 (d, J= 4.8 Hz, 5H).

Example 20: Synthesis of N-(3-(2-(lH-pyrazolo[3.,4-b]pyridin-5-yl)ethyl)-2.,4- difluorophenyl)-5-fluoro-2-methylpyridine-3-sulfonamide (Compound 9)

Compound 9 Synthesis of 9-a: 2,4-difluoro-3-(2-[ lH-pyrazolo[3,4-b1pyridin-5-yl1ethynyl)aniline

[0281] To a solution of 5-bromo-lH-pyrazolo[3,4-b]pyridine (500 mg, 2.5 mmol, 1 equiv) in DMF (10 mL) was added 3-ethynyl-2,4-difluoroaniline (463 mg, 3.0 mmol, 1.2 equiv), TEA (766 mg, 7.5 mmol, 3 equiv), Cui (48 mg, 0.2 mmol, 0.1 equiv) and Pd(PPh 3 )2Cl 2 (177 mg, 0.2 mmol, 0.1 equiv) at room temperature under a N2 atmosphere. The resulting mixture was stirred at 100 °C for 0.5 h. After cooling to room temperature, water (20 mL) was added and mixture was extracted with ethyl acetate (3 x 20 mL). The combined organic extracts were washed with brine (2 x 10 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with PE:EA = 3: 1) to afford 2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethynyl)an iline (500 mg, 73% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 271

Synthesis of 9-b: 2,4-difluoro-3-(2-[ lH-pyrazolo[3,4-b1pyridin-5-yl1ethyl)aniline

[0282] 10% Pd/C (20 mg) was added to a solution of 2,4-difluoro-3-(2-[lH-pyrazolo[3,4- b]pyridin-5-yl]ethynyl)aniline (200 mg, 0.7 mmol, 1 equiv) in MeOH (20 mL) in a pressure tank. The mixture was heated to 100 °C under H 2 (20 atm) for 16 h, and then filtered through a Celite pad and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE:EA (2: 1) to afford 2,4-difluoro-3-(2-[lH-pyrazolo[3,4- b]pyridin-5-yl]ethyl)aniline (100 mg, 49% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 274

Synthesis of Compound 9: N-(3-(2-(lH-pyrazolo[3,4-b1pyridin-5-yl)ethyl)-2,4- difluorophenyl)-5-fluoro-2-methylpyridine-3-sulfonamide

[0283] Pyridine (86 mg, 1 mmol, 3 equiv) and then 5-fluoro-2-methylpyridine-3-sulfonyl chloride (130 mg, 0.45 mmol, 1.5 equiv) were added to a solution of 2,4-difluoro-3-(2-[lH- pyrazolo[3,4-b]pyri din-5 -yl]ethyl)aniline (100 mg, 0.3 mmol, 1 equiv) in DCM (5 mL) at room temperature. The reaction mixture was stirred for 1 h at room temperature and then concentrated under reduced pressure and the crude residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-50% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford N-(3-(2-(lH- pyrazolo[3,4-b]pyridin-5-yl)ethyl)-2,4-difluorophenyl)-5-flu oro-2-methylpyridine-3- sulfonamide (60 mg, 34% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 448

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.51 (s, 1H), 8.67 (d, J= 2.8 Hz, 1H), 8.21 (d, J= 2.1 Hz, 1H), 8.03 (s, 1H), 7.90 - 7.75 (m, 2H), 7.08 (td, J= 9.0, 6.0 Hz, 1H), 6.89 (td, J= 9.0, 1.7 Hz, 1H), 2.85 (t, J= 4.8 Hz, 4H), 2.73 (d, J= 1.2 Hz, 3H).

Example 21: Synthesis of 5-cyano -N-[2.4-dinuoro-3-(2-|l//-pyra zolo[3.4-b|pyridin-5- yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 10) p

Synthesis of Compound 10: 5-cyano-N -[2,4-difluoro-3-(2-[1H -pyrazolo[3,4-b1pyridin-5- yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide

[0285] To a stirred solution of 2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5- yl]ethyl)aniline (100 mg, 0.4 mmol, 1 equiv) in DCM (10 mL) was added 5-cyano-2- methoxypyridine-3 -sulfonyl chloride (85 mg, 0.4 mmol, 1 equiv) and pyridine (87 mg, 1 mmol, 3 equiv). The resulting solution was stirred for 2 h at room temperature and then concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 10-65% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-cyano-N-[2,4- difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethyl)phenyl]- 2-methoxypyridine-3- sulfonamide (59 mg, 35% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 471

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.52 (s, 1H), 10.33 (s, 1H), 8.93 (d, J= 2.3 Hz, 1H), 8.42 (d, J= 2.3 Hz, 1H), 8.21 (d, J= 2.1 Hz, 1H), 8.04 (s, 1H), 7.84 (d, J= 2.1 Hz, 1H), 7.12 (td, J = 8.9, 5.9 Hz, 1H), 6.94 (t, J= 9.0 Hz, 1H), 4.03 (s, 3H), 2.87 (p, J= 7.2, 6.0 Hz, 4H). Example 22: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[lH-pyrazolo[4.,3-b]pyridin-5 - yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 11)

Synthesis of 11-a: 2,4-difluoro-3-(2-[iH-pyrazolo[4,3-b1pyridin-5-yl1ethynyl)an iline [0286] Cui (0.25 g, 1.3 mmol, 0.1 equiv) and Pd(PPh 3 )2Cl 2 (0.92 g, 1.3 mmol, 0.1 equiv) were added to a stirred solution of 5-chloro-lH-pyrazolo[4,3-b]pyridine (2 g, 13 mmol, 1 equiv), TEA (3.30 g, 32.6 mmol, 2.5 equiv) and 3-ethynyl-2,4-difluoroaniline (2 g, 13 mmol, 1 equiv) in DMF (20 mL) under a nitrogen atmosphere. After stirring for 0.5 h at 80 °C under nitrogen atmosphere, the reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE:EA (1 : 1-0: 1) to afford 2,4- difluoro-3-(2-[lH-pyrazolo[4,3-b]pyridin-5-yl]ethynyl)anilin e (2.2 g, 62% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 271

Synthesis of 11-b: 2A-difluoro-3-(2-|TE[-pyrazolor4,3-b]pyridin-5-yl]ethyl)anil ine

[0287] 10% Pd/C (78 mg) was added to a solution of 2,4-difluoro-3-(2-[lH-pyrazolo[4,3- b]pyridin-5-yl]ethynyl)aniline (200 mg , 0.7 mmol, 1 equiv) in MeOH (20 mL). The mixture was heated to 80 °C for 4 h under H 2 (20 atm). The reaction mixture was cooled to room temperature, filtered through a Celite pad and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluting with PE:EA = 1 : 1) to afford 2,4- difluoro-3-(2-[lH-pyrazolo[4,3-b]pyridin-5-yl]ethyl)aniline (90 mg, 44% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 275

Synthesis of Compound 11 : 5-chloro-N-[2,4-difluoro-3-(2-[iH-pyrazolo[4,3-b1pyridin-5- yl1ethyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0288] Pyridine (86 mg, 1.1 mmol, 3 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (132 mg, 0.45 mmol, 1.5 equiv) were added to a solution of 2,4-difluoro-3-(2-[lH- pyrazolo[4,3-b]pyridin-5-yl]ethyl)aniline (100 mg, 0.3 mmol, 1 equiv) in DCM (5 mL) at room temperature. The reaction mixture was stirred for 2 hrs and then concentrated under reduced pressure. The crude residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18; 50 x 250 mm; 10 μm; Mobile Phase: 25-50% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N-[2,4-difluoro-3- (2-[lH-pyrazolo[4,3-b]pyridin-5-yl]ethyl)phenyl]-2-methoxypy ridine-3-sulfonamide (40 mg, 23% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 480

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.20 (s, 1H), 8.47 (d, J= 2.6 Hz, 1H), 8.18 (s, 1H), 7.98 (d, J = 2.6 Hz, 1H), 7.87 (dd, J= 8.6, 1.1 Hz, 1H), 7.11 (dq, J = 8.9, 3.2 Hz, 2H), 6.94 (t, J = 8.8 Hz, 1H), 3.92 (s, 3H), 2.98 (s, 4H).

Example 23: Synthesis of 5-chloro-N -[2.,4-difluoro-3-(2-[lH-pyrrolo[2.,3-b]pyridin-5-yl] ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 12)

12-b Compound 12 Synthesis of 12-a: 2,4-difluoro-3-(2-[lH-pyrrolo [2,3-b1pyridin-5-yl1ethynyl)aniline

[0289] 5-Bromo-lH-pyrrolo[2,3-b]pyridine (196 mg, 0.1 mmol, 1 equiv), DMF (2.5 mL), THF (2.5 mL), NEt 3 (302 mg, 3 mmol, 3 equiv), Cui (19 mg, 0.1 mmol, 0.1 equiv), Pd(PPh 3 ) 2 Cl 2 (70 mg, 0.1 mmol, 0.1 equiv) and 3-ethynyl-2,4-difluoroaniline (183 mg, 1.2 mmol, 1.2 equiv) were placed into a 40 mL vial. The reaction mixture was stirred at 50 °C for 16 h and then diluted with water (20 mL). The resulting solution was extracted with EtOAc (2 x 20 mL) and the combined organic extracts washed water (3 x 20 mL), dried over anhydrous sodium sulfate and concentrated. The crude residue was purified by Flash-Prep- HPLC with the following conditions: Column: WelFlash TM C18-I; Spherical C18 20-40 μm; 120 g; mobile phase 5-80% MeCN / 0.1% aqueous formic acid; Detector 220 nm; to afford 2,4-difluoro-3-(2-[lH-pyrrolo [2,3-b]pyridin-5-yl]ethynyl) aniline (105 mg, 39% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 270

Synthesis of 12-b: 2,4-difluoro-3-(2-[iH-pyrrolo[2,3-b1pyridin-5-yl1 ethyl aniline

[0290] 10% Pd/C (45 mg, 0.4 mmol, 1.3 equiv) was added to a solution of 2,4-difluoro-3-(2- [lH-pyrrolo[2,3-b]pyridin-5-yl]ethynyl)aniline (90 mg, 0.3 mmol, 1 equiv) in MeOH (5 mL) in a 50 mL s 6 aled tube. The suspension was heated to 60 °C for 2 h under H 2 (20 atm). The reaction mixture was cooled to room temperature, filtered, and concentrated under reduced pressure to afford 2,4-difluoro-3-(2-[lH-pyrrolo[2,3-b]pyridin-5-yl] ethyl)aniline (80 mg, 88% yield) as a white solid which was used directly in next step without further purification. LCMS (ES, m/z): [M+H] + : 274

Synthesis of Compound 12: 5-chloro-N-[2,4-difluoro-3-(2-[iH-pyrrolo[2,3-b1pyridin-5-yl 1 ethyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0291] Pyridine (230 mg, 3.0 mmol, 10 equiv) and then 5-chloro-2-methoxypyridine-3- sulfonyl chloride (99 mg, 0.4 mmol, 1.4 equiv) were added to a solution of 2,4-difluoro-3-(2- [lH-pyrrolo[2,3-b]pyridin-5-yl]ethyl)aniline (80 mg, 0.3 mmol, 1 equiv) in DCM (3 mL) and the reaction mixture stirred for 3 hours at room temperature. The reaction mixture was concentrated and the crude residue purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 42-65% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-(2-[lH- pyrrolo[2,3-b]pyridin-5-yl] ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (41 mg, 29% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 479

1 H NMR (300 MHz, DMSO-d 6 ) δ 11.48 (s, 1H), 8.48 (d, J= 2.6 Hz, 1H), 8.17 (s, 1H), 7.99 (d, J= 2.6 Hz, 1H), 7.90 (d, J= 2.1 Hz, 1H), 7.61 (d, J= 2.0 Hz, 1H), 7.41 (dd, J= 3.4, 2.5 Hz, 1H), 7.10 (td, J= 8.9, 5.9 Hz, 1H), 6.91 (td, J= 9.1, 1.6 Hz, 1H), 6.34 (dd, J= 3.4, 1.8

Hz, 1H), 3.94 (s, 3H), 2.86 (m, 2H), 2.80 (m, 2H).

Example 24: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[lH-pyrazolo[3.,4-c]pyridin-5 - yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 13)

Synthesis of 13-a: 3-((lH-pyrazolo[3,4-c]pyridin-5-yl)ethynyl)-2,4-difluoroanil ine [0292] 3-Ethynyl-2,4-difluoroaniline (770 mg, 5.1 mmol, 1 equiv), TEA (1.5 g, 15 mmol, 3 equiv), Cui (96 mg, 0. 5 mmol, 0.1 equiv) and Pd(PPh 3 ) 2 Cl 2 (350 mg, 0.5 mmol, 0.1 equiv) were added to a solution of 5-bromo-lH-pyrazolo[3,4-c]pyridine(1.0 g, 5 mmol, 1 equiv) in DMF (10 mL) under an atmosphere of nitrogen. The reaction mixture was heated to 80 °C for 16 hours. After cooling to room temperature, water (200 mL) was added and the mixture extracted with ethyl acetate (3 x 200 mL). The combined organic extracts were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude residue was purified by silica gel chromatography, eluting with PE:EA =1 : 1, to afford 3-((lH- pyrazolo[3,4-c]pyridin-5-yl)ethynyl)-2,4-difluoroaniline (1.0 g, 73% yield) as a brown solid. LCMS (ES, m/z): [M+H] + : 271

Synthesis of 13-b: 2A-difluoro-3-(2-[iH-pyrazolo[3,4-c]pyridin-5-yl]ethyl)anili ne [0293] 10% Pd/C (78 mg) was added to a solution of 2,4-difluoro-3-(2-[lH-pyrazolo[3,4- c]pyridin-5-yl]ethynyl)aniline (200 mg, 0.7 mmol, 1 equiv) in MeOH (20 mL) and THF (3 mL). The suspension was stirred at 70 °C for 2 h under a H 2 atmosphere (2 MPa). The mixture was filtered through Celite, concentrated under reduced pressure, and the residue purified by silica gel chromatography, eluting with PE:EA =1 : 1, to afford product 2,4- difluoro-3-(2-[lH-pyrazolo[3,4-c]pyridin-5-yl]ethyl)aniline (100 mg, 49% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 275

Synthesis of Compound 13: 5-chloro-N-[2,4-difluoro-3-(2-[iH-pyrazolol3,4-c1pyridin-5- yl1ethyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0294] Pyridine (52 mg, 0.6 mmol, 3 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (80 mg, 0.3 mmol, 1.5 equiv) were added to a solution of 2,4-difluoro-3-(2-[lH- pyrazolo[3,4-c]pyridin-5-yl]ethyl)aniline (60 mg , 0.2 mmol, 1 equiv) in DCM (5 mL) and the reaction mixture stirred at room temperature for 1 h. The mixture was concentrated under reduced pressure and the residue was purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18; 50 x 250 mm; 10 μm mobile phase; Mobile Phase: 25- 40% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N-[2,4- difluoro-3-(2-[lH-pyrazolo[3,4-c]pyridin-5-yl]ethyl)phenyl]- 2-methoxypyridine-3- sulfonamide (15 mg , 14% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 480

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.50 (s, 1H), 8.96 (s, 1H), 8.38 (d, J= 2.5 Hz, 1H), 8.10 (s, 1H), 7.96 (d, J= 2.6 Hz, 1H), 7.43 (s, 1H), 7.05 (q, J= 8.4, 7.7 Hz, 1H), 6.83 (t, J= 9.1 Hz, 1H), 3.89 (s, 3H), 2.95 (s, 4H).

Example 25: Synthesis of 5-chloro-N-[2.,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (Compound 14)

Compound 14 Synthesis of 14-cl & 14-c2: 5-bromo-3-methyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine and 5-bromo-3-methyl-2-((2- (trimethylsilyl)ethoxy)methyl)-2H-pyrazolo[3,4-b1pyridine

[0295] NaH (0.23 g, 9.5 mmol, 2 equiv) was added portion wise to a stirred solution of 5- bromo-3-methyl-lH-pyrazolo[3,4-b]pyridine (1 g, 4.7 mmol, 1 equiv) and [2- (chloromethoxy)ethyl]trimethylsilane (1.6 g, 9.4 mmol, 2 equiv) in THF (30 mL) at 0 °C. The reaction mixture was stirred at room temperature for 5 hours and then quenched with water (50 mL). The mixture was extracted with EA (3 x 30 mL) and the combined organic extracts washed with brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue was purified by silica gel chromatography, eluting with PE:EA (5: 1-2: 1) to afford 5-bromo-3-methyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (0.8 g, 50% yield) as colorless oil and 5-bromo-3-methyl-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-pyr azolo[3,4-b]pyridine (0.64 g, 37% yield).

LCMS (ES, m/z): [M+H] + : 342

Synthesis of 14-a: 3-methyl-5-(4,4,5,5-tetramethyl-L3,2-dioxaborolan-2-yl)-l-[[ 2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine

[0296] 5 -Bromo-3-methyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo [3,4-b]pyridine (0.8 g, 2.3 mmol, 1 equiv), bis(pinacolato)diboron (1.2 g, 4.7 mmol, 2 equiv), KO Ac (0.46 g, 4.7 mmol, 2 equiv), Pd(dppf)C12 (0.32 g, 0.45 mmol, 0.2 equiv) were suspended in dioxane (12 mL) under an atmosphere of nitrogen and stirred at 90 °C for 16 h. The reaction mixture was cooled to room temperature, diluted with water (50 mL), and extracted with EA (2 x 30 mL). The combined organic extracts were washed with water (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (3: 1), to afford 3-methyl-5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1.6 g, 172%, 60% purity) as colorless oil.

LCMS (ES, m/z): [M+H] + : 390

Synthesis of 14-b: 3-methyl-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1 pyridin-5-ol [0297] NaOH (1 M, 16 mL, 16 mmol) and aqueous H 2 O 2 (30%, 1.5 g, 14 mmol, 6 equiv) were added to a solution of 3-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l-[ [2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1.6 g, 2.5 mmol, 1 equiv) in THF (16 mL). The resulting solution was stirred for 3 h at 25 °C and then diluted with water (50 mL). The mixture was extracted with EA (2 x 30 mL) and the combined organic extracts washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE:EA (2: 1) to afford 3-methyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b] pyridin-5-ol (0.25 g, 37% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 280

Synthesis of 14-c: 2,4-difluoro-3-[[(3-methyl-l-[[2-

(trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5-yl )oxy1methyl1aniline

[0298] PPI13 (300 mg, 1.1 mmol, 1.5 equiv) and TMAD (190 mg, 1.1 mmol, 1.5 equiv) were added to a solution of 3-methyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b] pyridin- 5-ol (210 mg, 0.8 mmol, 1 equiv) and (3-amino-2,6-difluorophenyl)methanol (130 mg, 0.8 mmol, 1.1 equiv) in DCM (10 mL) under an atmosphere of nitrogen. The reaction mixture was stirred for 3 h at 25 °C and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford 2,4- difluoro-3-[[(3-methyl-l-[[2-(trimethylsilyl)ethoxy]methyl]p yrazolo[3,4-b]pyridin-5- yl)oxy]methyl]aniline (316 mg, 100% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 421

Synthesis of 14-d: 5-chloro-N-(2,4-difluoro-3-[[(3-methyl-l-[[2-

(trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5-yl )oxy1methyl1phenyl)-2- methoxypyridine-3 -sulfonamide

[0299] Pyridine (98 mg, 1.2 mmol, 4 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (135 mg, 0.6 mmol, 1.8 equiv) were added to a solution of 2,4-difluoro-3-[[(3- methyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]py ridin-5-yl)oxy]methyl]aniline (130 mg, 0.3 mmol, 1 equiv) in DCM (8 mL) and the reaction mixture was stirred for 48 h at 25 °C. The resulting mixture was concentrated under reduced pressure and the residue purified by prep-HPLC with the following conditions: Column: WelFlash TM C18-1; Spherical C18 20-40 μm; 120 g; Mobile Phase: 10-80% MeCN / 0.1% aqueous formic acid; Detector 220 nm; to afford 5-chloro-N-(2,4-difluoro-3-[[(3-methyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-2- methoxypyridine-3 -sulfonamide (90 mg, 47% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 626

Synthesis of Compound 14: 5-chloro-N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-2-methoxypyridine-3-sulfon amide

[0300] A solution of 5-chloro-N-(2,4-difluoro-3-[[(3-methyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-2- methoxypyridine-3 -sulfonamide (90 mg, 0.14 mmol, 1 equiv) in TFA (0.8 mL) and DCM (5 mL) was stirred at room temperature for 3 h and then concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-[([3-methyl-lH- pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyri dine-3-sulfonamide (44 mg, 64% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 496

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.38 (s, 1H), 8.46 (d, J= 2.6 Hz, 1H), 8.18 (d, J = 2.7 Hz, 1H), 8.03 (d, J= 2.6 Hz, 1H), 7.86 (d, J= 2.7 Hz, 1H), 7.37 (q, J= 8.5, 8.1 Hz, 1H), 7.14 (t, J= 8.9 Hz, 1H), 5.13 (s, 2H), 3.91 (s, 3H), 2.47 (s, 3H).

Example 26: Synthesis of N-[2,4-difluoro-3-(2-[lH-pyrazolo[3.,4-b]pyridin-5- yl]ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 15)

Synthesis of Compound 15: N-[2,4-difluoro-3-(2-[iH-pyrazolol3,4-b1pyridin-5- yl1ethyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0301] Pyridine (93 mg, 1.2 mmol, 4 equiv) and 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (99 mg, 0.4 mmol, 1.5 equiv) were added to a solution of 2,4-difluoro-3-(2-[lH- pyrazolo[3,4-b]pyri din-5 -yl]ethyl)aniline (80 mg, 0.3 mmol, 1 equiv) in DCM (5 mL) and the reaction mixture stirred for 2 days at 25°C. The reaction mixture was concentrated and the residue purified by prep-HPLC with the following conditions: Column: welch Vltimate XB- C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford N-[2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5- yl]ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (38 mg, 28% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 464 1 H NMR (300 MHz, DMSO-d 6 ) δ 13.51 (s, 1H), 10.22 (s, 1H), 8.45 (d, J= 3.0 Hz, 1H), 8.19 (s, 1H), 8.02 (s, 1H), 7.93 (dd, J= 7.4, 3.1 Hz, 1H), 7.82 (s, 1H), 7.09 (s, 1H), 6.92 (t, J= 9.1 Hz, 1H), 3.93 (s, 3H), 2.88 (s, 4H).

Example 27: Synthesis of 3-cyano-N-[2.,4-difluoro-3-(2-[lH-pyrazolo[3.,4-b]pyridin-5- yl]ethyl)phenyl]-5-(triflnoromethyl) benzenesulfonamide (Compound 16)

Compound 16

Synthesis of 16-a: 3-(benzylsulfanyl)-5- (trifluoromethyl)benzonitrile

[0302] A solution of 3-fluoro-5-(trifluoromethyl)benzonitrile (2.0 g, 10 mmol, 1 equiv), LiOH (0.5 g, 21 mmol, 2 equiv) and benzyl mercaptan (1.6 g, 13 mmol, 1.2 equiv) in DMF (50 mL) was stirred for 1 h at room temperature. The reaction mixture was diluted with water (100 mL) and then extracted with EA (2 x 100 mL). The combined organic extracts were washed with water (3 x 100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. This afforded 3-(benzylsulfanyl)-5-(trifluoromethyl)benzonitrile (3.0 g, 97% yield) as a yellow oil, which was used directly in next step without further purification. Synthesis of 16-b: 3-cyano-5-(trifluoromethyl)benzenesulfonyl chloride

[0303] NCS (910 mg, 6.8 mmol, 4 equiv) was added portion wise to a mixture of MeCN (7.5 mL) and aqueous HC1 (6 M, 1.5 mL) at 0 °C. To this was added 3-(benzylsulfanyl)-5- (trifluoromethyl)benzonitrile (500 mg, 1.7 mmol, 1 equiv) portion wise at 0 °C and the reaction mixture stirred at 0 °C for 1 hour. The reaction mixture was quenched with ice water (20 mL) and extracted with DCM (2 x 20 mL). The combined organic extracts were washed with water (2 x 20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to afford of 3-cyano-5-(trifluoromethyl)benzenesulfonyl chloride (700 mg, 152% yield, 60% purity) as yellow oil which was used directly in next step without further purification.

Synthesis of Compound 16: 3-cyano-N-[2,4-difluoro-3-(2-[iH-pyrazolo[3,4-b1pyridin-5- yl]ethyl)phenyl]-5-(trifluoromethyl) benzenesulfonamide

[0304] Pyridine (170 mg, 2.2 mmol, 5 equiv) and 3-cyano-5- (trifluoromethyl)benzenesulfonyl chloride (350 mg, 1.3 mmol, 3 equiv) were added to a solution of 2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethyl)anil ine (120 mg, 0.4 mmol, 1 equiv) in DCM (2 mL) and the reaction mixture stirred at room temperature for 30 minutes. The reaction mixture was concentrated and the residue purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 33-53% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 3-cyano-N- [2,4-difluoro-3-(2-[lH-pyrazolo[3,4-b]pyridin-5-yl]ethyl)phe nyl]-5- (trifluoromethyl)benzenesulfonamide (85 mg, 38% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 508

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.51 (s, 1H), 10.51 (s, 1H), 8.75 (s, 1H), 8.42 (d, J= 1.7 Hz, 1H), 8.24-8.15 (m, 2H), 8.03 (d, J= 1.3 Hz, 1H), 7.86 (d, J= 2.1 Hz, 1H), 7.10 (td, J= 8.8, 5.9 Hz, 1H), 7.03-6.92 (m, 1H), 2.93-2.81 (m, 4H).

Example 28: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[2-methylpyrazolo[4.,3- b]pyridin-6-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 17) Synthesis of 17-a: 6-bromo-2-methylpyrazolo[4,3-b]pyridine and 6-bromo-l-methyl-1H- pyrazolo[4,3 -bipyridine

[0305] NaH (60% in mineral oil, 218 mg, 9.1 mmol, 2 equiv) was added to a solution of 6- bromo-2H-pyrazolo[4,3-b]pyridine (900 mg, 4.5 mmol, 1 equiv) and methyl iodide (774 mg, 5.4 mmol, 1.2 equiv) in DMF (20 mL) and the reaction mixture stirred at room temperature for 3 hours. The reaction was quenched with water (50 mL) and then extracted EA (3 x 30 mL). The organic layer was washed with 50 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column: WelFlash TM C18-1; Spherical C18 20- 40 μm; 120 g; Mobile Phase: 5-55% MeCN / 0.1% aqueous formic acid; Detector 220 nm; to afford 6-bromo-2-methylpyrazolo[4,3-b]pyridine (210 mg, 22% yield) as a white solid and 6- bromo-l-methyl-lH-pyrazolo[4,3-b]pyridine (150 mg, 15% yield) as a white solid.

LC-MS (ES, m/z): [M+H] + = 212

Synthesis of 17-b: 2,4-difluoro-3-12-(2-methylindazol-6-yl)ethynyl1aniline

[0306] A solution of 6-bromo-2-methylindazole (180 mg, 0.9 mmol, 1 equiv), Cui (17 mg, 0.09 mmol, 0.1 equiv 3-ethynyl-2,4-difluoroaniline (131 mg,), TEA (259 mg, 2.6 mmol, 3 equiv) and Pd(PPh 3 )2C12 (60 mg, 0.09 mmol, 0.1 equiv) in DMF (10 mL) under a nitrogen atmosphere was heated to 80 °C for 2 h. The reaction was then quenched by the addition of water (50 mL). The solids were collected by filtration and dried to give 2,4-difluoro-3-[2-(2- methylindazol-6-yl)ethynyl]aniline (260 mg, 110% yield, 80% purity) as a light yellow solid. LC-MS (ES, m/z): [M+H] + = 285

Synthesis of 17-c: 5-chloro-N-[2,4-difluoro-3-(2-[2-methylpyrazolo[4,3-b1pyridi n-5- yl1ethynyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0307] 5 -Chloro-N-[2,4-difluoro-3-(2-[2-methylpyrazolo[4,3-b]pyridin -6-yl]ethynyl)phenyl]- 2-methoxypyridine-3 -sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 10% Pd/C (30 mg) in MeOH (10 mL) were stirred at room temperature for 1 h under a H 2 atmosphere (1 atm). The reaction mixture was filtered and the filtrate concentrated under reduced pressure. The crude residue was purified by Prep-HPLC with the following conditions: Column: WelFlash TM C18-I; Spherical C18 20-40 μm; 120 g; mobile phase 10-70% MeCN / 0.1% aqueous formic acid Detector 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-(2-[2-methylpyrazolo[4,3- b]pyridin-6-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (14 mg, 14% yield) as a white solid.

LC-MS (ES, m/z): [M+H] + = 289 Synthesis of Compound 17: 5-chloro-N-[2,4-difluoro-3-(2-[2-methylpyrazolor4,3-b1pyridi n- 6-yl1ethyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0308] Pyridine (150 mg, 1.9 mmol, 3 equiv) and then 5-chloro-2-methoxypyridine-3- sulfonyl chloride (150 mg, 0.6 mmol, 1 equiv) were added to a solution of 2,4-difluoro-3-(2- [2-methylpyrazolo[4,3-b]pyridin-5-yl]ethynyl)aniline (180 mg, 0.6 mmol, 1 equiv) in DCM (8 mL). The reaction mixture was stirred at 30 °C for 16 hours, then concentrated and the residue purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB- C18, 50 x 25 0 mm, 10 μm; Mobile Phase: 35-65% MeCN / 0.1% aqueous formic acid; Detector: 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-(2-[2-methylpyrazolo[4,3-b]pyridi n- 6-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (100 mg, 32% yield) as a white solid. LC-MS (ES, m/z): [M+H] + 494

H NMR (300 MHz, DMSO-d 6 ) δ 8.54 (s, 1H), 8.45 (d, J= 2.6 Hz, 1H), 8.22 (s, 1H), 7.98 (d, J= 2.6 Hz, 1H), 7.63 (s, 1H), 7.14-7.00 (m, 1H), 6.87 (t, J = 9.0 Hz, 1H), 4.18 (s, 3H), 3.92 (s, 3H), 2.95-2.84 (m, 4H).

Example 29: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[l-methylpyrazolo[4.,3- b]pyridin-6-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 18)

Compound 18

Synthesis of 18-a: 2,4-difluoro-3-(2-[i-methylpyrazolol4,3-b1pyridin-6-yl1ethyn yl)aniline [0309] A solution of 6-bromo-l -methylindazole (360 mg, 1.7 mmol, 1 equiv), 3-ethynyl-2,4- difluoroaniline (261 mg, 1.7 mmol, 1 equiv), TEA (518 mg, 5.1 mmol, 3 equiv), Pd(PPh 3 )2Cl 2 (120 mg, 0.17 mmol, 0.1 equiv), and Cui (33 mg, 0.17 mmol, 0.1 equiv) in DMF (15 mL) under N2 atmosphere was stirred at 80 °C for 1 h. The reaction mixture was then quenched by the addition of water and the mixture filtered. The filter cake was dried and afforded 2,4- difluoro-3-(2-[l-methylpyrazolo[4,3-b]pyridin-6-yl]ethynyl)a niline (490 mg, crude) as a light yellow solid.

LC-MS (ES, m/zy. [M+H] + 285 Synthesis of 18-b: 2,4-difluoro-3-(2-[i-methylpyrazolo[4,3-b1pyridin-6-yl1ethyl )aniline [0310] A suspension of 2,4-difluoro-3-(2-[l-methylpyrazolo[4,3-b]pyridin-6- yl]ethynyl)aniline (100 mg, 0.35 mmol, 1 equiv), and 10% Pd/C (100 mg, 0.94 mmol, 2.7 equiv) in MeOH (5 mL) was stirred at 60 °C for 1 h under H 2 (30 atm). The mixture was filtered and the filtrate concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: WelFlash TM C18-I, Spherical C18 20- 40 μm, 120 g; mobile phase 25-68% MeCN / 0.1% aqueous formic acid; Detector 220 nm; to afford 2,4-difluoro-3-(2-[l-methylpyrazolo[4,3-b]pyridin-6-yl]ethyl )aniline (20 mg, 20% yield) as a white solid.

LC-MS (ES, m/z). [M+H] + 289

Synthesis of Compound 18: 5-chloro-N-[2,4-difluoro-3-(2-[i-rnethylpyrazolo[4,3-b]pyrid in- 6-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide

[0311] Pyridine (9 mg, 0.1 mmol, 3 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (13 mg, 0.05 mmol, 1.5 equiv) were added to a solution of 2,4-difluoro-3-(2-[l- methylpyrazolo[4,3-b]pyridin-6-yl]ethyl)aniline (10 mg, 0.035 mmol, 1 equiv) in DCM (1 mL) and the reaction mixture stirred at 30 °C for 16 hours. The reaction mixture was concentrated under reduced pressure and the residue purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile phase: 5-40% MeCN / 0.1% aqueous NH 3 ; Detector: 220 nm; to afford 5-chloro-N-[2,4-difluoro-3- (2-[l-methylpyrazolo[4,3-b]pyridin-6-yl]ethyl)phenyl]-2-meth oxypyridine-3-sulfonamide (2.3 mg, 13% yield) as a white solid.

LC-MS (ES, m/z) [M+H] + 494

1 H NMR (300 MHz, Methanol-d 4 ) δ 8.31 (d, J= 2.6 Hz, 1H), 8.24 (d, J= 1.8 Hz, 1H), 8.13 (d, J= 1.0 Hz, 1H), 7.97 (d, J= 2.6 Hz, 1H), 7.77 (s, 1H), 7.39-7.20 (m, 1H), 6.93-6.80 (m, 1H), 4.03 (d, J= 14.2 Hz, 6H), 3.05 (s, 4H).

Example 30: Synthesis of 5-chloro-/V-[2.,4-difluoro-3-(2-[imidazo[l.,5-a] pyridin-7- yl]ethyl)phenyl]-2- methoxypyridine-3-sulfonamide (Compound 19)

Compound 19

Synthesis of 19-a: l-(4-bromopyridin-2-yl)methanamine

[0312] BH 3 THF (82 mL, 1 M, 82 mmol, 5 equiv) was added dropwise to a stirred solution of 4-bromopyridine-2-carbonitrile (3.0 g, 16 mmol, 1 equiv) in THF (33 mL) at 0 °C under N2 atmosphere and stirred at room temperature for 16 hours. The mixture was cooled to 0 °C, and 2 M aqueous HC1 (83 mL, 170 mmol, 10 eq) was added dropwise. The mixture was heated to reflux for 30 min and then cooled to room temperature. The mixture was basified to pH 8 with 2 M aqueous NaOH (100 mL).

The resulting mixture was extracted with THF (50 mL x 3) and the combined organic layers washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford crude l-(4-bromopyri din-2 - yl)methanamine (3 g, crude) as a yellow oil which was used directly in next step without further purification. LCMS (ES, m/z): [M+H] + : 187

Synthesis of 19-b: A-[(4-bromopyridin-2-yl)methyl]formamide

[0313] l-(4-Bromopyridin-2-yl)methanamine (2.8 g, 15 mmol, 1 equiv) was dissolved in formic acid (8 mL) and stirred at 100 °C for 3 h. The mixture was concentrated under reduced pressure, saturated NaHCO 3 aqueous (45 mL) was added, and the resulting mixture extracted with DCM (3 x 45 mL). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The crude residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford N-[(4-bromopyridin-2-yl)methyl]formamide (1.0 g, 30% yield for 2 steps) as a grey solid. LCMS (ES, m/z): [M+H] + : 215

Synthesis of 19-c: 7-bromoimidazo[1,5-a]pyridine

[0314] POCL (784 mg, 5 mmol, 1.1 equiv) was added to a stirred solution of N-[(4- bromopyridin-2-yl)methyl]formamide (1.0 g, 4.6 mmol, 1 equiv) in toluene (10 mL) and the reaction mixture stirred at 110 °C for 2 h. The solution was concentrated under reduced pressure and the residue purified by silica gel chromatography, eluting with PE:EA (1 : 1) to afford 7-bromoimidazo[l,5-a]pyridine (750 mg, 82% yield) as a grey solid.

LCMS (ES, m/z): [M+H] + : 197

Synthesis of 19-d: 2,6-difluoro-3-(2-[imidazo[1,5-a1pyridin-7-yl1ethynyl)anilin e [0315] TEA (340 mg, 3.3 mmol, 3 equiv), Cui (21 mg, 0.11 mmol, 0.1 equiv) and Pd(PPh 3 ) 2 C12 (78 mg, 0.11 mmol, 0.1 equiv) were added to a stirred solution of 7- bromoimidazo[l,5-a]pyridine (220 mg, 1.1 mmol, 1 equiv) and 3-ethynyl-2,6-difluoroaniline (200 mg, 1.3 mmol, 1.2 equiv) in DMF (5 mL) and the mixture purged with N 2 three times. The reaction mixture was then heated to 50 °C for 2 hours and then filtered. The filtrate was diluted with H 2 O (10 mL) and extracted with EA (3 x 10 mL). The combined organic extracts were dried over anhydrous Na 2 SO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with PE:EA (1 :1) to afford 2,6- difluoro-3-(2-[imidazo[l,5-a]pyridin-7-yl]ethynyl)aniline (270 mg, 90% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 270

Synthesis of 19-e: 5-chloro-N-[2,4-difluoro-3-(2-[imidazo[L5-a1pyridin-7-yl1eth ynyl)phenyl1 -2-methoxypyridine-3 -sulfonamide

[0316] Pyridine (240 mg, 3 mmol, 3 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (290 mg, 1.2 mmol, 1.2 equiv) were added to a stirred solution of 2,4-difluoro-3-(2- [imidazo[l,5-a]pyridin-7-yl]ethynyl)aniline (270 mg, 1.0 mmol, 1 equiv) in DCM (5 mL) and the reaction mixture stirred at room temperature for 16 hours. The mixture was concentrated under reduced pressure and the residue purified by prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; 20-60% MeCN / 0.1% aqueous formic acid; Detector 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-(2-[imidazo[l,5- a]pyridin-7-yl]ethynyl)phenyl]-2-methoxypyridine-3-sulfonami de (150 mg, 32% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 475.

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.48 (s, 1H), 8.53 (d, J= 2.6 Hz, 1H), 8.49 (s, 1H), 8.38 (d, J= 13 Hz, 1H), 8.09 (d, J= 2.6 Hz, 1H), 7.94 (s, 1H), 7.53 (s, 1H), 7.40-7.33 (m, 1H), 7.23 (td, J= 8.9, 1.5 Hz, 1H), 6.69 (dd, J= 13, 1.6 Hz, 1H), 3.96 (s, 3H).

Synthesis of Compound 19: 5-chloro-N-[2,4-difluoro-3-(2-[imidazo[1,5-a] pyridin-7- yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide

[0317] 10% Pd/C (10 mg) was added to a solution of 5-chloro-N-[2,4-difluoro-3-(2- [imidazo[l,5-a]pyridin-7-yl]ethynyl)phenyl]-2 -methoxypyridine-3- sulfonamide (50 mg, 0.1 mmol, 1 equiv) in EA (5 mL) in a pressure tank. The mixture was stirred at room temperature for 2 h under H 2 (50 psi), then filtered through a Celite pad and concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-60% MeCN / aqueous formic acid; Detector 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-(2-[imidazo[l,5-a] pyridin-7-yl]ethyl)phenyl]-2- methoxypyridine-3 -sulfonamide (15 mg, 30% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 479.

'H NMR (300 MHz, Methanol-d 4 ) δ 8.32 (d, J= 2.6 Hz, 1H), 8.25 (s, 1H), 8.11 (d, J = 7.2 Hz, 1H), 7.98 (d, J= 2.6 Hz, 1H), 7.31-7.23( m, 1H), 7.21 (s, 1H), 7.06 (s, 1H), 6.86 (td, J= 9.0, 1.9 Hz, 1H), 6.46 (dd, J= 13, 1.7 Hz, 1H), 4.03 (s, 3H), 2.95 (t, J= 13 Hz, 2H), 2.75 (t, J = 13 Hz, 2H).

Example 31: Synthesis of N-[2,4-difluoro-3-[([4-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (Compound 20)

Synthesis of 20-a: 4-chloro-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine

[0318] Into a 1000 mL 3-necked round-bottom flask, was placed 4-chloro-lH-pyrazolo[3,4- b]pyridine (40 g, 260 mmol, 1 equiv), DCM (500 mL) and /?-toluenesulfonic acid (9 g, 52 mmol, 0.2 equiv). This was followed by the addition of dihydropyran (65.7 g, 781 mmol, 3 equiv) dropwise with stirring at 0 °C. The resulting solution was stirred for 16 h at room temperature, then concentrated under vacuum. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 1) to afford 4-chloro-l-(oxan-2-yl)pyrazolo[3,4- b]pyridine (39 g, 57% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 238

Synthesis of 20-b: 4-azido-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine

[0319] Into a 500 mL 3-necked round-bottom flask, was placed 4-chloro-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (39 g, 164 mmol, 1 equiv), DMF (200 mL) and sodium azide (16 g, 0.25 mmol, 1.5 equiv). The resulting solution was stirred for 12 h at 100 °C in an oil bath. The resulting solution was diluted with 600 mL of H 2 O and extracted with 2 x 300 mL of ethyl acetate. The combined organics were washed with 2 x 500 ml of brine and dried over anhydrous sodium sulfate, then concentrated under vacuum. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 1) to afford 4-azido-l-(oxan- 2-yl)pyrazolo[3,4-b]pyridine (24 g, 54% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 245

Synthesis of 20-c: l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-4-amine

[0320] Into a 500 mL round-bottom flask, was placed 4-azido-l-(oxan-2-yl)pyrazolo[3,4- b]pyridine (24 g, 98 mmol, 1 equiv), methanol (200 mL) and Pd/C (2.4 g, 10%). This was hydrogenated at 4 atm hydrogen for 12 hrs, then filtered. The filtrate was concentrated under vacuum, and the residue applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 1) to afford l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-4-amine (13 g, 55% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 219

Synthesis of 20-d: 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-4-amine

[0321] To a solution of l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-4-amine (5 g , 23 mmol, 1 equiv) in DCM (50 mL) was added NBS (4 g, 23 mmol, 1 equiv) in portions at 0 °C. The resulting solution was stirred for 0.5 h at 0 °C, then concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA =1 : 1) to afford 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-4-amine (6 g, 88% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 297

Synthesis of 20-e: 5-bromo-4-iodo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine

[0322] A mixture of Cui (7.7 g, 40 mmol, 2 equiv) in MeCN (150 mL) was heated at 50 °C followed by addition of /BuONO (10.4 g, 101 mmol, 5 equiv). The reaction mixture was stirred at 50 °C for 0.5 h, when 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-4-amine (6 g , 20 mmol, 1 equiv) was added. The temperature was raised to 80 °C for 2 h, then cooled. The reaction was quenched with an aqueous saturated solution of NaHCO 3 (500 mL) and extracted with ethyl acetate (3 x 200 mL). The combined organics were washed with brine (2 x 100 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 4: 1) to afford 5-bromo-4-iodo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (6 g, 73% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 408

Synthesis of 20-f: 5-bromo-4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine

[0323] To a solution of 5-bromo-4-iodo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (2 g, 4.9 mmol, 1 equiv) in dioxane (50 mL) were added a solution of trimethylboroxine in THF (1.23 g, 4.9 mmol, 1 equiv, 50%), K2CO3 (1.03 g, 14.7 mmol, 3 equiv) and Pd(PPh3)4 (566 mg, 0.49 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The reaction mixture was stirred under nitrogen atmosphere at 100°C for 16 h. The mixture was cooled to room temperature, water (200 mL) was added and this extracted with ethyl acetate (3 x 60 mL). The combined organics were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The crude product was purified by column chromatography over silica gel (eluent: PE:EA = 3: 1) to afford 5-bromo-4-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (1.2 g, 82% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 296

Synthesis of 20-g: 4-methyl-l-(tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl -L3,2- dioxaborolan-2-yl)-1H-pyrazolo[3,4-b1pyridine

[0324] To a solution of 5-bromo-4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (1.2 g, 4.1 mmol, 1 equiv) in dioxane (50 mL) were added bis(pinacolato)diboron (2.06 g, 8.1 mmol, 2 equiv), KO Ac (795 mg, 8.1 mmol, 2 equiv) and Pd(dppf)C12 (297 mg, 0.41 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The reaction mixture was heated at 100 °C for 8 h, then cooled. The mixture was diluted with water (100 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organics were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford 4-methyl-l-(tetrahydro- 2H-pyran-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-y l)-lH-pyrazolo[3,4-b]pyridine (2 g, crude) as a black solid which was used in the next step without further purification. LCMS (ES, m/z): [M+H] + : 344

Synthesis of 20-h: 4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-ol

[0325] To a solution of 4-methyl-l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxabor olan-2- yl)pyrazolo[3,4-b]pyridine (1.4 g, 4.1 mmol, 1 equiv) in THF (20 mL) and H 2 O (20 mL) were added NaHCO 3 (860 mg, 10 mmol, 2.5 equiv) followed by addition of H 2 O 2 (4.6 g, 41 mmol, 10 equiv, 30%) dropwise at room temperature. The resulting solution was stirred at room temperature for 1 h, then water (200 mL) was added. The mixture was extracted with ethyl acetate (3 x 60 mL), and the combined organics were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 1 : 1) to afford 4-methyl- l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (600 mg, 63% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 234

Synthesis of 20-i: 2A-difluoro-3-([[4-rnethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyrid in-5- yl]oxy]methyl)aniline

[0326] To a solution of 4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (400 mg , 1.72 mmol, 1 equiv) in DCM (10 mL) were added (3-amino-2,6-difluorophenyl)methanol (272 mg, 1.72 mmol, 1 equiv) and PPh 3 (674 mg, 2.6 mmol, 1.5 equiv) followed by addition of TMAD (442 mg, 2.57 mmol, 1.5 equiv) in portions at room temperature. The resulting solution was stirred at room temperature for 1 h. Water (200 mL) was added and the mixture extracted with ethyl acetate (3 x 80 mL). The combined organics were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 1 : 1) to afford 2,4- difluoro-3-([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5 -yl]oxy]methyl)aniline (300 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 375

Synthesis of 20-j : N-[2,4-difluoro-3-([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1py ridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de

[0327] To a solution of 2,4-difluoro-3-([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyrid in-5- yl]°xy]methyl)aniline (150 mg, 0.4 mmol, 1 equiv) in DCM (5 mL) was added pyridine (95 mg, 1.2 mmol, 3 equiv) and 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (99 mg, 0.4 mmol, 1.1 equiv). The resulting solution was stirred at room temperature for 1 h, then concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 2: 1) to afford N-[2,4-difluoro-3-([[4-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (80 mg, 35% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 564 Synthesis of Compound 20: N-[2,4-difluoro-3-[([4-methyl-1H-pyrazolol3,4-b1pyridin-5- yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0328] To a solution of N-[2,4-difluoro-3-([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]py ridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (80 mg, 0.14 mmol, 1 equiv) in DCM (4 mL) was added TFA (1 mL) at room temperature. The resulting solution was stirred for 1 h, then concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-60% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to afford N-[2,4-difluoro-3-[([4-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl ]oxy)methyl]phenyl]-5- fluoro-2-methoxypyridine-3 -sulfonamide (33 mg, 48% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 480

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.47 (s, 1H), 10.36 (s, 1H), 8.45 (d, J= 3.0 Hz, 1H), 8.34 (s, 1H), 8.12 (d, J= 1.3 Hz, 1H), 7.94 (dd, J= 7.3, 3.0 Hz, 1H), 7.35 (td, J= 8.9, 6.0 Hz, 1H), 7.11 (td, J= 9.0, 1.6 Hz, 1H), 5.13 (s, 2H), 3.91 (s, 3H), 2.25 (s, 3H).

Example 32: Synthesis of N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5- yloxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamid e (Compound 21)

Synthesis of 21-a: 5-(4,4,5,5-tetramethyl-L3,2-dioxaborolan-2-yl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine [0329] Into a 500 mL vial was placed 5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (8 g, 24 mmol, 1 equiv), dioxane (200 mL), bis(pinacolato)diboron (12.4 g, 49 mmol, 2 equiv), Pd(dppf)C12 (1.78 g, 2 mmol, 0.1 equiv) and KO Ac (4.8 g, 50 mmol, 2 equiv). The resulting solution was stirred for 2 h at 80 °C under N2 atmosphere. The solids were removed by filtration and the filtrate was concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :3) to give 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (11 g, crude) as an oil. The crude product was used directly.

LC-MS (ES, m/z): 376 [M+H] + Synthesis of 21-b: l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5 -ol [0330] Into a 500 mL vial, was placed 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (12 g, 32 mmol, 1 equiv), MeOH (100 mL), Na2SO3 (252 mg, 2 mmol, 0.06 equiv), H 2 O (100 mL) and H 2 O 2 (35% in water) (32.8 g, 964 mmol, 30 equiv). The resulting solution was stirred overnight, then quenched by the addition of water (100 mL). The solids were collected by filtration and dried to give 1 -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (3.7 g, 44% yield) as a white solid. LC-MS (ES, m/z): 266 [M+H] +

Synthesis of 21-c: 2,4-difluoro-3-[[(l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazo lo[3,4- b1pyridin-5-yl)oxy1methyl1aniline

[0331] Into a 40 mL vial was placed l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridin-5-ol (133 mg, 0.5 mmol, 1 equiv), DCM (15 mL), (3-amino-2,6- difluorophenyl)methanol (88 mg, 0.6 mmol, 1.1 equiv), PPI13 (197 mg, 0.8 mmol, 1.5 equiv) and TMAD (130 mg, 0.8 mmol, 1.5 equiv). The resulting solution was stirred overnight, then concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 10-80% MeCN / 0.1% aqueous formic acid; to afford 2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (130 mg, 64% yield) as a white solid.

LCMS (ES, m/z): 407 [M+H] +

Synthesis of 21-d: N-(2,4-difluoro-3-[[(l-[[2-(trimethylsilyl)ethoxy1methyl1pyr azolo[3,4- b1pyridin-5-yl)oxy1methyl1phenyl)-5-fluoro-2-methoxypyridine -3-sulfonamide [0332] Into a 40 mL vial was placed 2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (130 mg, 0.3 mmol, 1 equiv), DCM (8 mL), pyridine (79 mg, 1 mmol, 3 equiv) and 5-fluoro-2- methoxypyridine-3 -sulfonyl chloride (108 mg, 0.5 mmol, 1.5 equiv). The resulting solution was stirred overnight, then concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 5-85% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give N-(2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy]methyl]pyr azolo[3,4- b]pyridin-5-yl)oxy]methyl]phenyl)-5-fluoro-2-methoxypyridine -3-sulfonamide (110 mg, 58% yield) as a white solid.

LCMS (ES, m/z): 596 [M+H] + Synthesis of Compounds 21 : N-[2,4-difluoro-3-(riH-pyrazolol3,4-b1pyridin-5- yloxy1methyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamid e [0333] Into a 40 mL vial, was placed N-(2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide (130 mg, 0.2 mmol, 1 equiv), DCM (5 mL) and TFA (1 mL). The resulting solution was stirred for 5 h, then concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-yloxy]methyl )phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (53 mg, 52% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 466

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.56 (s, 1H), 10.45 (s, 1H), 8.42 (d, J= 3.0 Hz, 1H), 8.23 (d, J= 2.7 Hz, 1H), 8.05 (s, 1H), 7.96 (dd, J= 7.4, 3.0 Hz, 1H), 7.84 (d, J= 2.8 Hz, 1H), 7.42-7.29 (m, 1H), 7.19-7.07 (m, 1H), 5.13(s, 2H), 3.90 (s, 3H).

Example 33: Synthesis of 5-chloro-N-[2.,4-difluoro-3-[([4-methyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (Compound 22)

Compound 22

Synthesis of 22-a: 5-chloro-N-[2A-difluoro- methyl-l-(oxan-2-yl)pyrazolo[3,4- b1pyridin-5-yl1oxy1methyl)phenyl1-2-methoxypyridine-3-sulfon amide

[0334] To a stirred solution of 2,4-difluoro-3-([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (150 mg, 0.4 mmol, 1 equiv) in DCM (5 mL) were added pyridine (95 mg, 1.2 mmol, 3 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (145 mg, 0.6 mmol, 1.5 equiv) at room temperature. The resulting solution was stirred for 1 h, then concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 2: 1) to afford 5-chloro-N-[2,4-difluoro-3- ([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]met hyl)phenyl]-2- methoxypyridine-3 -sulfonamide (150 mg, 65% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 580

Synthesis of Compound 22: 5-chloro-N-[2,4-difluoro-3-[([4-methyl-1H-pyrazolol3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-2-methoxypyridine-3-sulfon amide

[0335] To a stirred solution of 5-chloro-N-[2,4-difluoro-3-([[4-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-2-methoxyp yridine-3-sulfonamide (160 mg, 0.276 mmol, 1 equiv) in DCM (8 mL) was added TFA (2 mL) at room temperature. The resulting solution was stirred for 1 h, then concentrated under reduced pressure. The residue was purified by prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 50-80% MeCN 0.1% aqueous formic acid; Detector, 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-[([4-methyl-lH-pyrazolo[3,4-b]pyr idin-5- yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfonamide (80 mg, 58% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 496

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.47 (s, 1H), 10.38 (s, 1H), 8.49 (d, J= 2.6 Hz, 1H), 8.35 (s, 1H), 8.12 (s, 1H), 8.00 (d, J= 2.6 Hz, 1H), 7.36 (td, J= 8.9, 5.9 Hz, 1H), 7.12 (td, J= 8.9, 1.6 Hz, 1H), 5.13 (s, 2H), 3.93 (s, 3H), 2.25 (s, 3H).

Example 34: Synthesis of N-[2,4-difluoro-3-[([4-methoxy-1H-pyrazolo[3.,4-b]pyridin-5- yl] methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 23)

Compound 23

Synthesis of 23-a: 5-bromo-4-methoxy-l-(oxan-2-yl)pyrazolol3,4-b]pyridine [0336] To a solution of 5-bromo-4-iodo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (1 g , 2.5 mmol, 1 equiv) in MeOH (20 mL) was added a solution of 30% NaOMe in MeOH (1.32 g, 7.35 mmol, 3 equiv) at room temperature. The resulting solution was stirred for 10 h then concentrated under reduced pressure. Water (100 mL) was added to the residue and this was extracted with ethyl acetate (3 x 60 mL). The combined organics were washed with brine (2 x 100 mL), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 4: 1) to afford 5-bromo-4-methoxy-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (700 mg, 92% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 312 Synthesis of 23-b: 4-methoxy-l-(oxan-2-yl)-5-(4A5,5-tetramethyl-L3,2-dioxaborol an-2- yl)pyrazolo[3,4-b1pyridine

[0337] To a solution of 5-bromo-4-methoxy-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (600 mg, 1.9 mmol, 1 equiv) in dioxane (20 mL) were added bis(pinacolato)diboron (976 mg, 3.8 mmol, 2 equiv) and KOAc (377 mg, 3.8 mmol, 2 equiv) followed by addition of Pd(dppf)C12 (140 mg, 0.2 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The resulting solution was stirred at 100 °C for 8 h. After the reaction mixture was cooled and quenched with water (300 mL), it was extracted with ethyl acetate (3 x 100 mL). The combined organics were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure to afford 4-methoxy-l-(oxan-2-yl)-5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine (600 mg) as a brown solid which was used in the next step directly without purification.

LCMS (ES, m/z): [M+H] + : 360

Synthesis of 23-c: 4-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol

[0338] To a stirred solution of 4-methoxy-l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine (400 mg, 1.1 mmol, 1 equiv) in THF (5 mL) and water (5 mL) were added NaHCO 3 (233 mg, 2.7 mmol, 2.5 equiv) followed by addition of H 2 O 2 (1.26 g, 11.1 mmol, 10 equiv, 30%) dropwise at room temperature. The resulting mixture was stirred for 0.5 h, then water (200 mL) was added. The mixture was extracted with ethyl acetate (3 x 60 mL). The combined organics were washed with brine (2 x 20 mL), dried over anhydrous ISfeSCL, and concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 1 : 1) to afford 4-methyl- l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (270 mg, 97% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 250

Synthesis of 23-d: 2,4-difluoro-3-([[4-methoxy-l-(oxan-2-yl)pyrazolo[3,4-b1pyri din-5- ylloxylmethyDaniline

[0339] To a solution of 4-methoxy-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (400 mg, 1.6 mmol, 1 equiv) in DCM (10 mL) were added (3-amino-2,6-difluorophenyl)methanol (255 mg, 1.6 mmol, 1 equiv) and PPh 3 (631 mg, 2.4 mmol, 1.5 equiv) followed by TMAD (414 mg, 2.4 mmol, 1.5 equiv) in portions at room temperature. The resulting solution was stirred for 1 h. Water (50 mL) was added and extracted with DCM (3 x 50 mL). The combined organics were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 1 : 1) to afford 2,4-difluoro-3-([[4-methoxy-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)aniline (300 mg, 47% yield) as a yellow solid. LCMS (ES, m/z): [M+H] + : 391

Synthesis of 23-e: N-12,4-difluoro-3-([[4-methoxy-l-(oxan-2-yl)pyrazolol3,4-b1p yridin-5- yl1oxy1methyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0340] To a stirred solution of 2,4-difluoro-3-([[4-methoxy-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (150 mg, 0.3 mmol, 1 equiv) in DCM (5 mL) were added pyridine (91 mg, 1 mmol, 3 equiv) and 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (130 mg, 0.45 mmol, 1.5 equiv) at room temperature. The resulting solution was stirred 2 h, then concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 2: 1) to afford N-[2,4-difluoro-3-([[4-methoxy-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (180 mg, 81% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 580

Synthesis of Compound 23: N-[2,4-difluoro-3-[([4-methoxy-1H-pyrazolo[3,4-b1pyridin-5- yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0341] To a stirred solution of N-[2,4-difluoro-3-([[4-methoxy-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (100 mg , 0.17 mmol, 1 equiv) in DCM (8 mL) was added TFA (2 mL) at room temperature. The resulting solution was stirred for 1 h, then concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-40% MeCN / 0.1% aqueous ammonia; Detector: 220; nm to afford N-[2,4-difluoro-3-[([4-methoxy-lH-pyrazolo[3,4-b]pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (40 mg, 29% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 496

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.46 (s, 1H), 10.32 (s, 1H), 8.45 (d, J= 3.0 Hz, 1H), 8.38 (d, J= 1.4 Hz, 1H), 7.94 (q, J= 3.3 Hz, 2H), 7.30 (td, J= 8.8, 5.8 Hz, 1H), 7.14-6.94 (m, 1H), 5.04 (s, 2H), 4.25 (s, 3H), 3.92 (s, 3H). Example 35: Synthesis of 5-chloro-N-[2.,4-difluoro-3-[([4-methoxy-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (Compound 24)

Compound 24

Synthesis of 24-a: 5-chloro-N-[2A-difluoro-3-([[4-methoxy-l-(oxan-2-yl)pyrazolo [3,4- b1pyridin-5-yl1oxy1methyl)phenyl1-2-methoxypyridine-3-sulfon amide

[0342] To a stirred solution of 2,4-difluoro-3-([[4-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (150 mg, 0.4 mmol, 1 equiv) in DCM (5 mL) was added pyridine (95 mg, 1.2 mmol, 3 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (139 mg, 0.5 mmol, 1.5 equiv) at room temperature. The resulting solution was stirred for 1 hour, then concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 2: 1) to afford 5-chloro-N-[2,4-difluoro-3- ([[4-methoxy-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]me thyl)phenyl]-2- methoxypyridine-3 -sulfonamide (170 mg, 74% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 596

Synthesis of Compound 24: 5-chloro-N-[2,4-difluoro-3-[([4-methoxy-1H-pyrazolo[3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-2-methoxypyridine-3-sulfon amide

[0343] To a stirred solution of 5-chloro-N-[2,4-difluoro-3-([[4-methoxy-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-2-methoxyp yridine-3-sulfonamide (100 mg, 168 μmol, 1 equiv) in DCM (8 mL) was added TFA (2 mL) at room temperature. The resulting solution was stirred for 1 hour then concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-40% MeCN / 0.1% aqueous ammonia; Detector, 220 nm; to afford 5-chloro-N-[2,4-difluoro-3-[([4-methoxy-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (40 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 512

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.46 (s, 1H), 10.34 (s, 1H), 8.49 (d, J= 2.6 Hz, 1H), 8.38 (d, J= 1.3 Hz, 1H), 8.01 (d, J= 2.6 Hz, 1H), 7.96 (s, 1H), 7.38-7.22 (m, 1H), 7.07 (t, J= 9.0 Hz, 1H), 5.04 (s, 2H), 4.25 (s, 3H), 3.92 (s, 3H).

Example 36: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(l-[lH-pyrazolo[3.,4-b]pyridin-5 - yloxy]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 25)

Compound 25

Synthesis of 25-a: tert-butyl N-(2,4-difluorophenyl)carbamate

[0344] Into a 250 mL vial was placed 2,4-difluoroaniline (8 g, 62 mmol, 1 equiv), MeOH (150 mL) and di-tert-butyl dicarbonate (14.9 g, 74 mmol, 1.2 equiv). The resulting solution was stirred overnight at 40 °C, then concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 :5) to give tert-butyl N-(2,4- difluorophenyl)carbamate(13 g, 92% yield) as a light yellow solid.

LC-MS (ES, m/z): 230 [M+H] +

Synthesis of 25-b: tert-butyl N-[2,4-difluoro-3-(l-hydroxyethyl)phenyl1carbamate

[0345] Into a 250 mL vial was placed tert-butyl N-(2,4-difluorophenyl)carbamate (5 g, 22 mmol, 1 equiv) in THF (100 mL), and 2.5M n-BuLi in hexane (20 mL, 50 mmol, 2.3 equiv) was added dropwise at -78 °C. The reaction was stirred for 1 h at -78 °C, then acetaldehyde (880 mg, 22 mmol, 1 equiv) was dropwise at -78 degrees. The resulting solution was stirred for 1 h at -78 °C. The reaction was quenched with water (50 mL) at low temperature, and the resulting solution extracted with ethyl acetate (50 mL x 3). The combined organics were dried (Na 2 SO 4 ) and concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/hexane (1 :5) to give tert-butyl N-[2,4-difluoro-3-(1-hydroxyethyl)phenyl]carbamate (2.8 g, 47% yield) as a light yellow solid.

LC-MS (ES, m/z): 273 [M+H] +

Synthesis of 25-c: tert-butyl-N-(2,4-difluoro-3-[l-[(l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5-yl)ox y1ethyl1phenyl)carbamate [0346] Into a 40 mL vial was placed l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridin-5-ol (1.7 g, 6.4 mmol, 1 equiv), DCM (25 mL), tert-butyl N-[2,4-difluoro-3-(1- hydroxyethyl)phenyl]carbamate (1.9 g, 7.1 mmol, 1.1 equiv), TMAD (1.65 g, 10 mmol, 1.5 equiv) and PPh 3 (2.5 g, 10 mmol, 1.5 equiv). The resulting solution was stirred for 2 h then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 10- 80% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give tert-butyl N-(2,4-difluoro- 3-[l-[(1-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]py ridin-5- yl)oxy]ethyl]phenyl)carbamate (1.6 g, crude) as a light yellow oil.

LC-MS (ES, m/z): 521 [M+H] +

Synthesis of 25-d: 2,4-difluoro-3-(l-[ lH-pyrazolo[3,4-b1pyridin-5-yloxy1ethyl)aniline [0347] Into a 100 mL 3-necked flask was placed tert-butyl N-(2,4-difluoro-3-[l-[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]ethyl]phenyl)carbamate (1.4 g, 2.7 mmol, 1 equiv), DCM (30 mL) and TFA (5 mL). The resulting solution was stirred for 20 min then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 10-80% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give 2,4-difluoro-3- (1-[lH-pyrazolo[3,4-b]pyridin-5-yloxy]ethyl)aniline (310 mg, 40% yield) as a white solid. LC-MS (ES, m/z): 291 [M+H] +

Synthesis of Compound 25: Synthesis of 5-chloro-N-[2,4-difluoro-3-(l-[iH-pyrazolo[3,4- b1pyridin-5-yloxy1ethyl)phenyl1-2-methoxypyridine-3-sulfonam ide

[0348] Into a 40 mL vial was placed 2,4-difluoro-3-(1-[lH-pyrazolo[3,4-b]pyridin-5- yloxy]ethyl)aniline (60 mg, 0.2 mmol, 1 equiv), pyridine (4 mL) and 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (100 mg, 0.4 mmol, 2 equiv). The resulting solution was stirred for 1 h then concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10 μm; Mobile Phase: 25-65% MeCN / 0.1% aqueous formic acid; Detector, 220 nm. This resulted in 5-chloro-N-

[2,4-difluoro-3-(1-[lH-pyrazolo[3,4-b]pyridin-5-yloxy]eth yl)phenyl]-2-methoxypyridine-3- sulfonamide (32 mg, 31% yield) as a white solid.

LC-MS (ES, m/z): 496 [M+H] + 1 H NMR (300 MHz, DMSO-d 6 ) δ 13.51 (s, 1H), 10.32 (s, 1H), 8.34 (d, J = 2.6 Hz, 1H), 8.19-

8.11 (m, 1H), 7.98 (s, 1H), 7.89 (d, J= 2.6 Hz, 1H), 7.55 (d, J = 2.7 Hz, 1H), 7.29-7.14 (m, 1H), 7.01 (t, J= 9.5 Hz, 1H), 5.80-5.68 (m, 1H), 3.84 (s, 3H), 1.66 (d, J= 6.5 Hz, 3H).

Example 37: Synthesis of 5-cyano-N-[2.,4-difluoro-3-([lH-pyrazolo[3.,4-b]pyridin-5- yloxy]methyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 26)

Compound 26

Synthesis of 26-a: 5-cyano-N-(2,4-difluoro-3-[[(l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5-yl)ox y1methyl1phenyl)-2- methoxypyridine-3 -sulfonamide

[0349] Into a 40 mL vial was placed 2,4-difluoro-3-[[(l -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (150 mg, 0.37 mmol, 1 equiv), pyridine (4 mL) and 5-cyano-2-methoxypyridine-3-sulfonyl chloride (172 mg, 0.74 mmol, 2 equiv). The resulting solution was stirred for 1 h then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 10-85% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give 5-cyano-N-(2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-2- methoxypyridine-3 -sulfonamide (170 mg, 76% yield) as light yellow oil.

Synthesis of Compound 26: 5-cyano-N-12,4-difluoro-3-([1H-pyrazolo[3,4-b]pyridin-5- yloxy1methyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0350] Into a 40 mL vial was placed 5-cyano-N-(2,4-difluoro-3-[[(1- [[(trimethylsilyl)methoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl )oxy]methyl]phenyl)-2- methoxypyridine-3 -sulfonamide (140 mg, 0.24 mmol, 1 equiv), DCM (5 mL) and TFA (1 mL). The resulting solution was stirred for 1 h then concentrated, and the pH of the residue was adjusted to 8 with ammonia (7 M in MeOH). The resulting solution was purified by Prep- HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-30% MeCN / 0.1% aqueous ammonia; Detector, 220 nm; to give 5- cyano-N-[2,4-difluoro-3-([lH-pyrazolo[3,4-b]pyridin-5-yloxy] methyl)phenyl]-2- methoxypyridine-3 -sulfonamide (60 mg, 53% yield) as a white solid.

LC-MS (ES, m/z): [M+H] + = 473

'H NMR (300 MHz, DMSO-d 6 ) δ 13.56 (s, 1H), 10.48 (s, 1H), 8.91 (d, J= 2.2 Hz, 1H), 8.46 (d, J = 2.2 Hz, 1H), 8.23 (d, J= 2.7 Hz, 1H), 8.05 (s, 1H), 7.85 (d, J= 2.7 Hz, 1H), 7.45-7.31 (m, 1H), 7.21-7.09 (m, 1H), 5.12 (s, 2H),.4.01(s,3H).

Example 38: Synthesis of N-[3-[([4-cyano-1H-pyrazolo[3.,4-b]pyridin-5-yl]oxy)methyl]-

2,4-difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonam ide (Compound 27)

Synthesis of 27-a: methyl 5-bromo-l-(oxan-2-yl)pyrazolol3,4-b1pyridine-4-carboxylate [0351] To a solution of 5-bromo-4-iodo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (1.5 g, 3.6 mmol, 1 equiv) in MeOH (20 mL) were added TEA (1.1 g, 11 mmol, 3 equiv) and Pd(dppf)C12 (268 mg, 0.3 mmol, 0.1 equiv). The resulting solution was stirred at 60 °C under carbon monoxide atmosphere for 6 hrs. The mixture was filtered through celite and concentrated, and the residue was purified by column chromatography over silica gel (eluent: PE:EA =4: 1) to afford methyl 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carboxylate (1.2 g, 96% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 340 Synthesis of 27-b: 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine-4-carboxamide [0352] A solution of methyl 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carboxylate (1.2 g, 3.5 mmol, 1 equiv) in 7 M ammonia solution in MeOH (20 ml) was stirred at 100 °C for 16 h. After cooling to room temperature, it was concentrated to give 5-bromo-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine-4-carboxamide (800 mg, 70% yield) as a brown solid, which was used in next step directly without any purification.

LCMS (ES, m/z): [M+H] + : 325

Synthesis of 27-c: 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine-4-carbonitrile

[0353] To a solution of 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carboxamide (800 mg, 2.4 mmol, 1 equiv) in THF (10 mL) were added TEA (622 mg, 6.1 mmol, 2.5 equiv) and TFA (1033 mg, 4.9 mmol, 2 equiv). The resulting solution was stirred for 2 hr, then concentrated. The residue was purified by column chromatography over silica gel (eluent: PE:EA =1 : 1) to afford 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carbonitrile (700 mg, 93% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 307

Synthesis of 27-d: l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-L3,2-dioxaborolan-2- yl)pyrazolo[3,4-b1pyridine-4-carbonitrile

[0354] To a solution of 5-bromo-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carbonitrile (800 mg, 2.6 mmol, 1 equiv) in dioxane (10 mL) were added bis(pinacolato)diboron (1.3 g, 5.2 mmol, 2 equiv), KO Ac (511 mg, 5.2 mmol, 2 equiv) and Pd(dppf)C12 (190 mg, 0.3 mmol, 0.1 equiv). The resulting solution was stirred at 100 °C under nitrogen for 4 h. After cooling, water (300 mL) was added and the mixture extracted with ethyl acetate (3x100 mL). The organic phase was washed with brine, dried over anhydrous Na 2 SO 4 , and concentrated to give the crude l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl )pyrazolo[3,4- b]pyridine-4-carbonitrile (850 mg) as a brown solid which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 355

Synthesis of 27-e: 5-hydroxy-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine-4-carbonitril e [0355] To a solution of l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[3,4-b]pyridine-4-carbonitrile (800 mg, 2.2 mmol, 1 equiv) in THF (5 mL) and H 2 O (5 mL) were added NaHCO 3 (474 mg, 5.6 mmol, 2.5 equiv) and 30% H 2 O 2 (2.5 g, 22.5 mmol, 10 equiv). The resulting solution was stirred for 4 hours, then diluted with water (200 mL). The mixture was extracted with ethyl acetate (3 x 100 mL), and the organics were washed with brine (3 x 50 mL), dried over anhydrous Na 2 SO 4 , before being concentrated. The residue was purified by column chromatography over silica gel (eluent: PE:EA =1 : 1) to give 5-hydroxy-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carbonitril e (500 mg, 91% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 245

Synthesis of 27-f: 5-[(3-amino-2,6-difluorophenyl)methoxy]-l-(oxan-2-yl)pyrazol o[3,4- blpyridine-4-carbonitrile

[0356] To a solution of 5-hydroxy-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carbonitril e (300 mg, 1.2 mmol, 1 equiv) in DCM (10 mL) were added (3-amino-2,6-difluorophenyl)methanol (234 mg, 1.4 mmol, 1.2 equiv), TMAD (317 mg, 1.8 mmol, 1.5 equiv) and PPh 3 (483 mg, 1.8 mmol, 1.5 equiv). The resulting solution was stirred at room temperature for 1 hour. The solution was concentrated, water (300 mL) was added and the mixture extracted with ethyl acetate (3 x 100 mL). The combined organics were washed with brine (3 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 2: 1) to give 5-[(3-amino-2,6-difluorophenyl)methoxy]-l- (oxan-2-yl)pyrazolo[3,4-b]pyridine-4-carbonitrile (400 mg, 85% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 386

Synthesis of 27-g: N-[3-([[4-cyano-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1oxy 1methyl)- 2,4-difluorophenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0357] To a solution of 5-[(3-amino-2,6-difluorophenyl)methoxy]-l-(oxan-2-yl)pyrazol o[3,4- b]pyridine-4-carbonitrile (150 mg , 389 μmol, 1 equiv) in DCM (5 mL) were added 5-fluoro- 2-methoxypyridine-3 -sulfonyl chloride (131 mg, 584 μmol, 1.5 equiv) and pyridine (93 mg, 1.2 mmol, 3 equiv). The resulting solution was stirred for 1 hour at room temperature. The resulting solution was concentrated and purified by column chromatography over silica gel (eluent: PE:EA = 4: 1) to afford N-[3-([[4-cyano-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]oxy]methyl)-2,4-difluorophenyl]-5-fluoro-2-methoxypyridin e-3-sulfonamide (130 mg, 58% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 575

Synthesis of Compound 27: N-[3-[([4-cyano-1H -pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]- 2,4-difluorophenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0358] To a solution of N-[3-([[4-cyano-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]oxy]methyl)-2,4-difluorophenyl]-5-fluoro-2-methoxypyridin e-3-sulfonamide (100 mg , 174 μmol, 1 equiv) in DCM (8 mL) was added TFA (2 mL). The resulting solution was stirred at room temperature for 1 h. The mixture was concentrated, and the residue purified by prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-50% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to afford N-[3-[([4-cyano-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]-2 ,4-difluorophenyl]-5-fluoro- 2-methoxypyridine-3 -sulfonamide (40 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 491

1 H NMR (300 MHz, DMSO-d 6 ) δ 14.23 (s, 1H), 10.38 (s, 1H), 8.85 (s, 1H), 8.40 (d, J= 3.0 Hz, 1H), 8.25 (s, 1H), 7.94 (dd, J= 13, 3.0 Hz, 1H), 7.39 (td, J= 9.0, 6.0 Hz, 1H), 7.17 (t, J = 9.0 Hz, 1H), 5.47 (s, 2H), 3.87 (s, 3H).

Example 39: Synthesis of 5-chloro-N-[3-[([4-cvano-1H-pyrazolo[3.,4-b]pyridin-5- yl] methyl]-2.,4-difluorophenyl]-2-methoxypyridine-3-sulfonamide (Compound 28)

Compound 28

Synthesis of 28-a: 5-chloro-N-[3-([[4-cyano-l-(oxan-2-yl)pyrazolol3,4-b1pyridin -5- yl1oxy1methyl)-2,4-difluorophenyl1-2-methoxypyridine-3-sulfo namide

[0359] To a solution of 5-[(3-amino-2,6-difluorophenyl)methoxy]-l-(oxan-2-yl)pyrazol o[3,4- b]pyridine-4-carbonitrile (150 mg, 0.4 mmol, 1 equiv) in DCM (5 mL) were added 5-chloro- 2-methoxypyridine-3 -sulfonyl chloride (141 mg, 584 μmol, 1.5 equiv) and pyridine (93 mg, 1.2 mmol, 3 equiv). The resulting solution was stirred for 1 h at room temperature then concentrated under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: PE:EA = 5: 1) to afford 5-chloro-N-[3-([[4-cyano-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)-2,4-difluoropheny l]-2-methoxypyridine-3- sulfonamide (130 mg, 56% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 591 Synthesis of Compound 28: 5-chloro-N-[3-[([4-cyano-1H-pyrazolol3,4-b1pyridin-5- yl1oxy)methyl1-2,4-difluorophenyl1-2-methoxypyridine-3-sulfo namide

[0360] To a solution of 5-chloro-N-[3-([[4-cyano-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin -5- yl]oxy]methyl)-2,4-difluorophenyl]-2-methoxypyridine-3-sulfo namide (120 mg, 0.2 mmol, 1 equiv) in DCM (8 mL) was added TFA (2 mL) at room temperature. The resulting solution was stirred for 1 h then concentrated under reduced pressure. The residue was purified by prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-55% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to afford 5-chloro-N-[3-[([4-cyano-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy) methyl]-2,4-difluorophenyl]- 2-methoxypyridine-3 -sulfonamide (25 mg, 24% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 507

1 H NMR (300 MHz, DMSO-d 6 ) δ 14.23 (s, 1H), 10.40 (s, 1H), 8.85 (s, 1H), 8.43 (d, J= 2.6 Hz, 1H), 8.25 (s, 1H), 8.00 (d, J= 2.6 Hz, 1H), 7.51-7.32 (m, 1H), 7.17 (t, J= 9.0 Hz, 1H), 5.47 (s, 2H), 3.88 (s, 3H).

Example 40: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(l-[lH-pyrazolo[3.,4-b]pyridin-5 - ylamino]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 29)

Compound 29

Synthesis of 29-a: tert-butyl N-(2,4-difluorophenyl)carbamate

[0361] Into a 250 mL vial was placed 2,4-difluoroaniline (8 g, 62 mmol, 1 equiv), MeOH (150 mL) and tert-butyl 4,4-dimethyl-3-oxopentanoate (14.9 g, 74 mmol, 1.2 equiv). The resulting solution was stirred overnight at 40 °C. The reaction mixture was concentrated and the residue was applied onto a silica gel column, eluting with ethyl acetate/petroleum ether (1 :5) to give tert-butyl N-(2,4-difluorophenyl)carbamate (13 g, 92% yield) as a light yellow solid.

LC-MS (ES, m/z): [M+H] + = 230

Synthesis of 29-b: tert-butyl N-(3-acetyl-2,4-difluorophenyl)carbamate

[0362] Into a 50 mL 3-necked round-bottom flask, purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl N-(2,4-difluorophenyl)carbamate (1 g, 4.4 mmol, 1 equiv) and THF (20 mL). 2.5 M n-BuLi in hexane (3.9 mL, 9.6 mmol, 2.2 equiv) was added dropwise with stirring at -78 °C. To this was added a solution of N-methoxy-N- methylacetamide (0.67 g, 6.5 mmol, 1.5 equiv) in THF (5 mL) dropwise with stirring at - 78 °C. The resulting solution was stirred for 5 h at 25 °C. The reaction was then quenched by the addition of 50 mL of water. The resulting solution was extracted with 3 x 30 mL of ethyl acetate, and the combined organics washed with 50 ml of water and 50 mL of brine, then dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography, eluting with PEZEA (8: 1) to afford tert-butyl N-(3-acetyl-2,4- difluorophenyl)carbamate (0.22 g, 19% yield) as colorless oil.

LCMS (ES, m/z): [M+H] + : 272

Synthesis of 29-c: tert-butyl N-[2,4-difluoro-3-[(1E)-l-[iH-pyrazolo[3,4-b1pyridin-5- yliminolethyllphenyllcarbamate

[0363] Into a 40 mL vial was placed tert-butyl N-(3-acetyl-2,4-difluorophenyl)carbamate (420 mg, 1.5 mmol, 1 equiv), lH-pyrazolo[3,4-b]pyridin-5-amine (270 mg, 2 mmol, 1.3 equiv), DCE (15 mL) and Ti(Oi-Pr) 3 Cl (1.2 g, 4.6 mmol, 3 equiv). The resulting solution was stirred for 48 h at 70 °C, then cooled and diluted with 30 mL of MeOH. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford tert-butyl N-[2,4-difluoro-3-[(lE)-l-[lH- pyrazolo[3,4-b]pyri din-5 -ylimino]ethyl]phenyl]carbamate (330 mg, 55% yield) as an off- white solid.

LCMS (ES, m/z): [M+H] + : 388

Synthesis of 29-d: tert-butyl N-[2,4-difluoro-3-(l-[iH-pyrazolo[3,4-b1pyridin-5- ylaminolethyDphenyllcarbamate

[0364] Into a 100 mL 3-necked round-bottom flask was placed tert-butyl N-[2,4-difluoro-3- [(lE)-l-[lH-pyrazolo[3,4-b]pyridin-5-ylimino]ethyl]phenyl]ca rbamate (300 mg, 0.8 mmol, 1 equiv), THF (15 mL) and MeOH (15 mL). NaBH4 (117 mg, 3.1 mmol, 4 equiv) was added in portions at room temperature. The resulting solution was stirred overnight, then quenched by the addition of 10 mL of MeOH. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 : 1) to afford tert-butyl N-[2,4-difluoro-3-(1-[lH-pyrazolo[3,4-b]pyridin-5- ylamino]ethyl)phenyl]carbamate (100 mg, 33% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 390 Synthesis of 29-e: N-[i-(3-amino-2,6-difluorophenyl)ethyl1-1H-pyrazolo[3,4-b1py ridin-5- amine

[0365] Into a 40 mL vial was placed tert-butyl N-[2,4-difluoro-3-(1-[lH-pyrazolo[3,4- b]pyridin-5-ylamino]ethyl)phenyl]carbamate (100 mg, 0.26 mmol, 1 equiv), DCM (5 mL) and 4M HC1 in 1,4-dioxane (1 mL, 4 mmol). The resulting solution was stirred for 5 h then concentrated under vacuum. This resulted in N-[l-(3-amino-2,6-difluorophenyl)ethyl]-lH- pyrazolo[3,4-b]pyri din-5 -amine hydrochloride (80 mg, crude) as a light brown solid.

LCMS (ES, m/z): [M+H] + : 290

Synthesis of Compound 29: 5-chloro-N-[2,4-difluoro-3-(1-[1H-pyrazolo[3,4-b1pyridin-5- ylamino1ethyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0366] Into a 40 mL vial was placed N-[1-(3-amino-2,6-difluorophenyl)ethyl]-lH- pyrazolo[3,4-b]pyri din-5 -amine (70 mg, crude, 1 equiv), DCM (5 mL), Pyridine (79 mg, 1 mmol, 4 equiv) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (70 mg, 0.3 mmol, 1.2 equiv). The resulting solution was stirred overnight, then concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 10-40% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[2,4-difluoro-3-(1-[lH-pyrazolo[3,4-b]pyridin-5- ylamino]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (14 mg, 11% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 495

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.13 (s, 1H), 10.20 (s, 1H), 8.32 (d, J= 2.6 Hz, 1H), 8.08 (d, J= 2.6 Hz, 1H), 7.87 (d, J= 2.6 Hz, 1H), 7.77 (s, 1H), 7.20-6.94 (m, 2H), 6.81 (s, 1H), 6.10 (d, J= 8.6 Hz, 1H), 4.85-4.74 (m, 1H), 3.86 (s, 3H), 1.52 (d, J= 6.8 Hz, 3H).

Example 41: Synthesis of N-(3-(((lH-pyrazolo[3.,4-b]pyridin-5-yl)oxy)methyl)-2.,4- difluorophenyl)-5-fluoro-2-methylpyridine-3-sulfonamide (Compound 30)

Compound 30

Synthesis of 30-a: N-(2,4-difluoro-3-(((l-((2-(trimethylsilyl)ethoxy)methyl)-1H - pyrazolo[3,4-b1pyridin-5-yl)oxy)methyl)phenyl)-5-fluoro-2-me thylpyridine-3-sulfonamide [0367] Into a 40 mL vial was placed 2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (150 mg, 0.37 mmol, 1 equiv), pyridine (4 mL) and 5-fluoro-2-methylpyridine-3-sulfonyl chloride (155 mg, 0.74 mmol, 2 equiv). The resulting solution was stirred for 1 h then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 5-85% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give N-(2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-5-fluoro-2- methylpyridine-3 -sulfonamide (170 mg, 80% yield) as light yellow oil.

LCMS (ES, m/z): [M+H] + : 580

Synthesis of Compound 30: N-(3-(((lH-pyrazolo[3,4-b1pyridin-5-yl)oxy)methyl)-2,4- difluorophenyl)-5-fluoro-2-methylpyridine-3-sulfonamide

[0368] Into a 40 mL vial was placed 5-cyano-N-(2,4-difluoro-3-[[(1- [[(trimethylsilyl)methoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl )oxy]methyl]phenyl)-2- methoxypyridine-3 -sulfonamide (140 mg, 0.24 mmol, 1 equiv), DCM (5 mL) and TFA (1 mL). The resulting solution was stirred for 1 h then concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 15-70% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give N-(3-(((lH-pyrazolo[3,4-b]pyridin-5-yl)oxy)methyl)-2,4-diflu orophenyl)-5- fluoro-2-methylpyridine-3-sulfonamide (60 mg, 54% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 450

1 H NMR (300 MHz, DMSO-d6) 3 13.55 (s, 1H), 10.71 (s, 1H), 8.70 (d, J= 2.9 Hz, 1H), 8.21 (d, J= 2.7 Hz, 1H), 8.05 (s, 1H), 7.87 (dd, J= 8.3, 2.9 Hz, 1H), 7.82 (d, J= 2.8 Hz, 1H), 7.42-7.28 (m, 1H), 7.16 (t, J= 9.1 Hz, 1H), 5.11 (s, 2H), 2.75 (d, J= 1.3 Hz, 3H).

Example 42: Synthesis of 5-chloro-N-(2.,4-difluoro-3-(2-(8-(methylamino)imidazo[l.,2- a]pyrazin-3-yl)ethyl)phenyl)-2-methoxypyridine-3-sulfonamide (Compound 31)

Synthesis of 31-a: 3-bromo-N-methylimidazo[L2-a1pyrazin-8-amine

[0369] A solution of 3-bromo-8-chloroimidazo[l,2-a]pyrazine (500 mg, 2.2 mmol, 1 equiv) in 33 wt. % CH 3 NH 2 /EtOH (5 mL) was heated by microwave at 150 °C for 0.5 h. After the mixture was cooled to room temperature, the mixture was filtered to afforded 3-bromo-N- methylimidazo[l,2-a]pyrazin-8-amine (450 mg, 92% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 227

Synthesis of 31-b: 5-chloro-N-(2,4-difluoro-3-[2-[8-(methylamino)imidazo[1,2a]p yrazin-3- yl1ethynyl1phenyl)-2-methoxypyridine-3-sulfonamide

[0370] To a solution of 5-chloro-N-(3-ethynyl-2,4-difluorophenyl)-2-methoxypyridine- 3- sulfonamide (500 mg, 1.4 mmol, 1 equiv) in DMF (10 mL) was added TEA (423 mg, 4.18 mmol, 3 equiv), 3-bromo-N-methylimidazo[l,2-a]pyrazin-8-amine (632 mg, 2.8 mmol, 2 equiv), CuI (26 mg, 139 μmol, 0.1 equiv) and Pd(PPh 3 ) 2 Cl 2 (97 mg, 139 μmol, 0.1 equiv). The reaction mixture was stirred at 100 °C for 0.5 h, then cooled and quenched with water (200 mL). This was extracted with ethyl acetate (200 mL), and the organics washed with brine, dried over Na 2 SO 4 , and concentrated. The residue was purified by column chromatography over silica gel (eluent: PE:EA=1:1) to afford 5-chloro-N-(2,4-difluoro-3-[2- [8-(methylamino)imidazo[l,2-a]pyrazin-3-yl]ethynyl]phenyl)-2 -methoxypyridine-3- sulfonamide (300 mg, 42% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 505

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.53 (s, 1H), 8.50 (d, J= 2.6 Hz, 1H), 8.08 (d, J= 2.6 Hz, 1H), 8.00 (s, 1H), 7.76 (q, J= 4.7 Hz, 1H), 7.58 (d, J= 4.6 Hz, 1H), 7.53 (d, J= 4.6 Hz, 1H), 7.39 (td, J= 8.9, 5.9 Hz, 1H), 7.27-7.18 (m, 1H), 3.94 (s, 3H), 2.96 (d, J= 4.7 Hz, 3H).

Synthesis of Compound 31 : 5-chloro-N-(2,4-difluoro-3-(2-(8-(methylamino)imidazolL2- a1pyrazin-3-yl)ethyl)phenyl)-2-methoxypyridine-3 -sulfonamide

[0371] To a solution of 5-chloro-N-(2,4-difluoro-3-[2-[8-(methylamino)imidazo[l,2- a]pyrazin-3-yl]ethynyl]phenyl)-2-methoxypyridine-3-sulfonami de (100 mg, 0.2 mol, 1 equiv) in THF (10 mL) was added 10% Pd/C (43 mg). The resulting solution was stirred at room temperature under H 2 for 2 hours. The mixture was filtered through celite and concentrated, and the residue purified by HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-60% MeCN / 0.5% aqueous ammonia; to afford 5-chloro-N-(2,4-difluoro-3-(2-(8-(methylamino)imidazo[l,2-a] pyrazin-3- yl)ethyl)phenyl)-2-methoxypyridine-3-sulfonamide (20 mg, 20% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 509

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.27 (s, 1H), 8.47 (s, 1H), 7.99 (d, J= 2.6 Hz, 1H), 7.47 (d, J= 4.7 Hz, 1H), 7.34 (d, J= 4.9 Hz, 1H), 7.27 (d, J= 4.7 Hz, 1H), 7.11 (d, J= 6.9 Hz, 1H), 7.06 (s, 1H), 6.92 (t, J= 9.0 Hz, 1H), 3.93 (s, 3H), 2.95 (m, 7H). Example 43: Synthesis of N-[3-(2-[8-aminoimidazo[l.,2-a]pyrazin-3-yl]ethyl)-2.,4- difluoroDhenyl]-5-chloro-2-methoxyDyridine-3-sulfonamide (Compound 32)

Synthesis of 32-a: 3-bromoimidazo[L2-a1pyrazin-8-amine

[0372] A solution of 3-bromo-8-chloroimidazo[l,2-a]pyrazine (1 g, 4.3 mmol, 1 equiv) and NH 3 H 2 O (14 mL) in IPA (3 mL) was stirred for 3.5 h at 120 °C in a microwave reactor. The mixture was cooled, and the resulting solids were collected by filtration and washed with DCM (3 x 10 mL), then dried under vacuum to afford 3-bromoimidazo[l,2-a]pyrazin-8- amine (526 mg, 57% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 213

Synthesis of 32-b: N-13-(2-[8-aminoimidazo[1,2a]pyrazin-3-yl1ethynyl)-2,4- difluorophenyl1-5-chloro-2-methoxypyridine-3-sulfonamide

[0373] To a stirred solution of 3-bromoimidazo[l,2-a]pyrazin-8-amine (100 mg, 0.5 mmol, 1 equiv), TEA (143 mg, 1.41 mmol, 3 equiv) and 5-chloro-N-(3-ethynyl-2,4-difluorophenyl)-2- methoxypyridine-3 -sulfonamide (169 mg, 0.5 mmol, 1 equiv) in dimethylformamide (2 mL) were added Pd(PPh 3 ) 2 Cl 2 (33 mg, 0.05 mmol, 0.1 equiv) and Cui (18 mg, 0.1 mmol, 0.2 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for Ih at 90 °C then cooled. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 10-55% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to afford N-[3-(2-[8-aminoimidazo[l,2- a]pyrazin-3-yl]ethynyl)-2,4-difluorophenyl]-5-chloro-2-metho xypyridine-3-sulfonamide (33 mg, 14% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 491

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.53 (s, 1H), 8.54 (d, J= 2.6 Hz, 1H), 8.10 (d, J= 2.7 Hz, 1H), 8.03 (s, 1H), 7.66-7.59 (m, 1H), 7.41 (td, J= 8.9, 5.9 Hz, 1H), 7.26 (dd, J= VIA, 6.7 Hz, 3H), 3.96 (s, 3H), 2.48 (s, 1H).

Synthesis of Compound 32: N-13-(2-[8-aminoimidazolL2-a1pyrazin-3-yl1ethyl)-2,4- difluorophenyl1-5-chloro-2-methoxypyridine-3-sulfonamide

[0374] To a solution of N-[3-(2-[8-aminoimidazo[l,2-a]pyrazin-3-yl]ethynyl)-2,4- difluorophenyl]-5-chloro-2-methoxypyridine-3-sulfonamide (100 mg, 0.2 mol, 1 equiv) in THF (10 mL) was added 10% Pd/C (108 mg). The resulting solution was stirred at room temperature under a balloon of H 2 for 2 hours. The mixture was filtered through celite and the filtrate concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-60% MeCN / 0.5% aqueous NH4HCO3; Detector 254 nm; to afford N-[3-(2-[8-aminoimidazo[l,2- a]pyrazin-3-yl]ethyl)-2,4-difluorophenyl]-5-chloro-2-methoxy pyridine-3-sulfonamide (50 mg, 50% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 495

1 H NMR (300 MHz, DMSO-d 6 ) δ 8.51 (d, J = 2.6 Hz, 1H), 8.15 (s, 1H), 8.00 (d, J = 2.6 Hz, 1H), 7.50 (d, J= 4.7 Hz, 1H), 7.21 (d, J= 4.7 Hz, 1H), 7.18-7.06 (m, 2H), 7.01-6.92 (m, 1H), 6.78 (s, 2H), 3.95 (s, 3H), 2.96 (q, J= 4.9 Hz, 4H).

Example 44: Synthesis of 5-chloro-N-[3-(2-[l-ethylpyrazolo[4.,3-b]pyridin-6-yl]ethyl) -

2,4-difluorophenyl]-2-methoxypyridine-3-sulfonamide (Compound 33)

Synthesis of 33-a: 6-bromo-l-ethylpyrazolo[4,3-b1pyridine

[0375] Into a 50 mL round-bottom flask was placed 6-bromo-lH-pyrazolo[4,3-b]pyridine (1 g, 5 mmol, 1 equiv), DMF (25 mL), NaH (404 mg, 10 mmol, 2 equiv, 60%) and ethyl iodide (945 mg, 6 mmol, 1.2 equiv). The resulting solution was stirred for 3 h at room temperature, then concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford 6-bromo-l-ethylpyrazolo[4,3-b]pyridine (470 mg, 41% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 226

Synthesis of 33-b: 3-(2-|T-ethylpyrazolor4,3-b]pyridin-6-yl]ethynyl)-2,4-difluo roaniline [0376] Into a 50 mL round-bottom flask was placed 6-bromo-l-ethylpyrazolo[4,3-b]pyridine (200 mg, 0.9 mmol, 1 equiv), DMF (10 mL), TEA (269 mg, 2.7 mmol, 3 equiv), Cui (17 mg, 0.09 mmol, 0.1 equiv), Pd(PPh 3 ) 2 Cl 2 (62 mg, 0.09 mmol, 0.1 equiv) and 3-ethynyl-2,4- difluoroaniline (271 mg, 1.8 mmol, 2 equiv) in portions at room temperature under N2 atmosphere. The reaction was stirred for 1 h at 80 °C, then was cooled and diluted with 10 mL of H 2 O. The resulting solution was extracted with 2 x 10 mL of ethyl acetate and the organics combined, washed with 3 x 10 mL of brine, dried over anhydrous sodium sulfate, then concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford 3 -(2- [ 1 -ethylpyrazolo[4,3-b]pyridin-6- yl]ethynyl)-2,4-difluoroaniline (210 mg, 80% yield) as a dark yellow oil.

LCMS (ES, m/z): [M+H] + : 299 Synthesis of 33-c: 3-(2-[i-ethylpyrazolor4,3-b1pyridin-6-yl1ethyl)-2,4-difluoro aniline [0377] Into a 30 mL s 6 aled tube was placed 3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6- yl]ethynyl)-2,4-difluoroaniline (210 mg, 0.7 mmol, 1 equiv), MeOH (4 mL), Pd/C (100 mg) and H 2 (20 atm.). The reaction was stirred for 2 h at room temperature. The mixture was filtered, and the filtrate concentrated under vacuum to give crude 3-(2-[l-ethylpyrazolo[4,3- b]pyridin-6-yl]ethyl)-2,4-difluoroaniline (202 mg) as a colorless oil which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 303

Synthesis of Compound 33: 5-chloro-N-[3-(2-[i-ethylpyrazolo[4,3-b]pyridin-6-yl]ethyl)- 2,4- difluorophenyl1-2-methoxypyridine-3-sulfonamide

[0378] Into a 50 mL round-bottom flask was placed 3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6- yl]ethyl)-2,4-difluoroaniline (100 mg, 0.3 mmol, 1 equiv), pyridine (5 mL), 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (80 mg, 0.3 mmol, 1 equiv) and DCM (0.1 mL). The reaction was stirred for 2 h at room temperature, then concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-60% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6-yl]ethyl)- 2,4-difluorophenyl]-2- methoxypyridine-3 -sulfonamide (30 mg ,18% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 508

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.24 (s, 1H), 8.50 (d, J= 2.6 Hz, 1H), 8.22 (dd, J= 10.9, 1.4 Hz, 2H), 7.99 (d, J= 2.6 Hz, 1H), 7.91-7.84 (m, 1H), 7.13 (td, J= 8.9, 5.9 Hz, 1H), 6.98 (td, J= 9.0, 1.6 Hz, 1H), 4.41 (q, J= 7.2 Hz, 2H), 3.94 (s, 3H), 2.97-2.87 (m, 4H), 1.37 (t, J= 7.2 Hz, 3H).

Example 45: Synthesis of N-[3-(2-[l-ethylpyrazolo[4.,3-b]pyridin-6-yl]ethyl)-2.,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 34) Synthesis of Compound 34: N-[3-(2-[i-ethylpyrazolol4,3-b1pyridin-6-yl1ethyl)-2,4- difluorophenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0380] Into an 8 mL vial was placed 3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6-yl]ethyl)-2,4- difluoroaniline (100 mg, 0.3 mmol, 1 equiv), pyridine (2 mL) and 5-fluoro-2- methoxypyridine-3 -sulfonyl chloride (112 mg, 0.5 mmol, 1.5 equiv) in DCM (0.1 mL). The resulting solution was stirred for 1 h at room temperature, then concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-50% MeCN / 0.1% aqueous formic acid; to afford N-[3-(2-[l-ethylpyrazolo[4,3-b]pyridin-6-yl]ethyl)-2,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (30 mg, 18% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 492

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.22 (s, 1H), 8.46 (d, J= 3.0 Hz, 1H), 8.21 (dd, J= 8.9, 1.4 Hz, 2H), 7.97-7.84 (m, 2H), 7.12 (td, J= 8.8, 5.9 Hz, 1H), 6.97 (td, J= 9.0, 1.6 Hz, 1H), 4.40 (q, J= 7.2 Hz, 2H), 3.93 (s, 3H), 2.94 (d, J= 10.8 Hz, 4H), 1.36 (t, J= 7.2 Hz, 3H).

Example 46: Synthesis of 5-chloro-N-[2.,4-difluoro-3-(2-[l-isopropylpyrazolo[4.,3- b]pyridin-6-yl]ethyl)phenyl]-2-methoxypyridine-3-sulfonamide (Compound 35)

Synthesis of 35-a: 6-bromo-l-isopropylpyrazolo[4,3-b]pyridine and 6-bromo-2-isopropyl- 2H-pyrazolo[4,3 -bipyridine

[0381] Into a 50 mL round-bottom flask was placed 6-bromo-lH-pyrazolo[4,3-b]pyridine (1 g, 5 mmol, 1 equiv), THF (25 mL), CS 2 CO 3 (3.3 g, 10 mmol, 2 equiv) and 2-iodopropane (1 g, 6 mmol, 1.2 equiv). The resulting solution was stirred for 12 h at 60 °C in an oil bath, then cooled and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford 6-bromo-l-isopropylpyrazolo[4,3- b]pyridine (600 mg, 50% yield) and 6-bromo-2-isopropyl-2H-pyrazolo[4,3-b]pyridine (450 mg) as light brown solids.

LCMS (ES, m/z): [M+H] + : 240

Synthesis of 35-b: 2,4-difluoro-3-(2- isopropylpyrazolo[4,3-b]pyridin-6-yl1ethynyl)aniline [0382] Into a 50 mL round-bottom flask was placed 6-bromo-l-isopropylpyrazolo[4,3- b]pyridine (600 mg, 2.5 mmol, 1 equiv), DMF (30 mL), Cui (47 mg, 0.25 mmol, 0.1 equiv), Pd(PPh 3 ) 2 Cl 2 (175 mg, 0.25 mmol, 0.1 equiv), TEA (758 mg, 7.5 mmol, 3 equiv) and 3- ethynyl-2,4-difluoroaniline (765 mg, 5 mmol, 2 equiv) in portions at room temperature under N2 atmosphere. The reaction was stirred for 2 h at 80 °C in an oil bath, then cooled and diluted with 10 mL of H 2 O. The resulting solution was extracted with 2 x 10 mL of ethyl acetate and the organics combined, washed with 3 x 10 mL of brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30- 80% MeCN / 0.1% aqueous formic acid; to afford 2,4-difluoro-3-(2-[l- isopropylpyrazolo[4,3-b]pyridin-6-yl]ethynyl)aniline (570 mg, 73% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 313

Synthesis of 35-c: 2,4-difluoro-3-(2-[i-isopropylpyrazolo[4,3-b1pyridin-6-yl1et hyl)aniline [0383] Into a 30 mL s 6 aled tube was placed 2,4-difluoro-3-(2-[l-isopropylpyrazolo[4,3- b]pyridin-6-yl]ethynyl)aniline (560 mg, 1.8 mmol, 1 equiv), MeOH (20 mL) and 10% Pd/C (300 mg) under H 2 (20 atm.). The reaction was stirred for 2 h at room temperature, then filtered and the filtrate concentrated. Crude 2,4-difluoro-3-(2-[l-isopropylpyrazolo[4,3- b]pyridin-6-yl]ethyl)aniline (470 mg) was isolated as a white solid which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 317

Synthesis of Compound 35: 5-chloro-N-[2,4-difluoro-3-(2-[i-isopropylpyrazolo[4,3- b1pyridin-6-yl1ethyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0384] Into a 40 mL vial was placed 2,4-difluoro-3-(2-[l-isopropylpyrazolo[4,3-b]pyridin-6- yl]ethyl)aniline (100 mg, 0.3 mmol, 1 equiv), pyridine (5 mL), DCM (0.1 mL) and 5-chloro- 2-methoxypyridine-3 -sulfonyl chloride (115 mg, 0.5 mmol, 1.5 equiv). The reaction was stirred for 30 min then concentrated under vacuum. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-65% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[2,4- difluoro-3-(2-[l-isopropylpyrazolo[4,3-b]pyridin-6-yl]ethyl) phenyl]-2-methoxypyridine-3- sulfonamide (30 mg, 18% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 522

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.24 (s, 1H), 8.49 (d, J= 2.6 Hz, 1H), 8.21 (dd, J= 8.9, 1.3 Hz, 2H), 7.99 (d, J= 2.6 Hz, 1H), 7.87 (dd, J= 1.9, 1.0 Hz, 1H), 7.12 (td, J= 8.9, 6.0 Hz, 1H), 6.96 (td, J= 9.0, 1.6 Hz, 1H), 4.94 (h, J= 6.7 Hz, 1H), 3.94 (s, 3H), 2.94 (s, 4H), 1.45 (d, J = 6.7 Hz, 6H). Example 47: Synthesis of N-[2,4-difluoro-3-(2-[l-isopropylpyrazolo[4.,3-b]pyridin-6- yl]ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 36)

Synthesis of Compound 36: N-[2,4-difluoro-3-(2-[i-isopropylpyrazolol4,3-b1pyridin-6- yl1ethyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0386] Into a 40 mL vial was placed 2,4-difluoro-3-(2-[l-isopropylpyrazolo[4,3-b]pyridin-6- yl]ethyl)aniline (100 mg, 0.3 mmol, 1 equiv), pyridine (4 mL), DCM (0.1 mL) and 5-fluoro- 2-methoxypyridine-3 -sulfonyl chloride (107 mg, 0.5 mmol, 1.5 equiv). The resulting solution was stirred for 1 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-60% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-(2-[l- isopropylpyrazolo[4,3-b]pyridin-6-yl]ethyl)phenyl]-5-fluoro- 2-methoxypyridine-3- sulfonamide (30 mg, 19% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 506

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.21 (s, 1H), 8.45 (d, J= 3.0 Hz, 1H), 8.25-8.11 (m, 2H), 7.97-7.84 (m, 2H), 7.11 (td, J= 8.9, 6.0 Hz, 1H), 6.95 (t, J= 8.9 Hz, 1H), 4.92 (p, J= 6.6 Hz, 1H), 3.92 (s, 3H), 2.94 (s, 4H), 1.45 (d, J= 6.6 Hz, 6H).

Example 48: Synthesis of N-[2,4-difluoro-3-(2-[2-isopropylpyrazolo[4.,3-b]pyridin-6- yl]ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 37)

Synthesis of 37-a: 2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3-b1pyridin-6-yl1et hynyl)aniline [0387] Into a 50 mL round-bottom flask was placed 6-bromo-2-isopropylpyrazolo[4,3- b]pyridine (410 mg, 1.7 mmol, 1 equiv), DMF (10 mL), Cui (33 mg, 0.17 mmol, 0.1 equiv), Pd(PPh 3 ) 2 Cl 2 (120 mg, 0.17 mmol, 0.1 equiv), TEA (518 mg, 5 mmol, 3 equiv) and 3- ethynyl-2,4-difluoroaniline (392. mg, 2.6 mmol, 1.5 equiv) at room temperature under N2 atmosphere. The reaction was stirred for 2 h at 80 °C then cooled and diluted with 10 mL of H 2 O. The resulting solution was extracted with 2 x 10 mL of ethyl acetate and the organics combined, washed with 3 x 10 mL of brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified by prep-HPLC Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30- 80% MeCN / 0.1% aqueous formic acid; to afford 2,4-difluoro-3-(2-[2- isopropylpyrazolo[4,3-b]pyridin-6-yl]ethynyl)aniline (400 mg, 75% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 313

Synthesis of 37-b: 2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3-b]pyridin-6-yl]et hyl)aniline [0388] Into a 30 mL s 6 aled tube was placed 2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3- b]pyridin-6-yl]ethynyl)aniline (400 mg, 1.3 mmol, 1 equiv), CH 3 OH (10 mL) and 10% Pd/C (200 mg) under H 2 (20 atm.). The resulting solution was stirred for 2 h then filtered through celite. The filtrate was concentrated to give 2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3- b]pyridin-6-yl]ethyl)aniline (350 mg, 86% yield) as a white solid which was used in next step directly without further purification. LCMS (ES, m/z): [M+H] + : 317

Synthesis of Compound 37: N-[2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3-b1pyridin-6- yl1ethyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0389] Into a 40 mL vial was placed 2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3-b]pyridin-6- yl]ethyl)aniline (110 mg, 0.3 mmol, 1 equiv), pyridine (8 mL) and 5-fluoro-2- methoxypyridine-3 -sulfonyl chloride (108 mg, 0.6 mmol, 1.5 equiv). The reaction was stirred for 30 min then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-50% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-(2-[2-isopropylpyrazolo[4,3- b]pyridin-6-yl]ethyl)phenyl]-5-fluoro-2-methoxypyridine-3-su lfonamide (30 mg, 17% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 506

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.22 (s, 1H), 8.62 (d, J= 2.8 Hz, 1H), 8.45 (t, J= 3.2 Hz, 1H), 8.20 (q, J= 1.9 Hz, 1H), 7.93 (dt, J= 6.8, 3.2 Hz, 1H), 7.67 (s, 1H), 7.08 (td, J= 7.6, 6.7, 3.5 Hz, 1H), 6.99-6.87 (m, 1H), 4.88-4.77 (m, 1H), 3.97-3.90 (m, 3H), 2.94-2.83 (m, 4H), 1.65-1.51 (m, 6H).

Example 49: Synthesis of N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine-3-sulfonamid e (Compound 38)

Compound 38

Synthesis of 38-a: N-[2,4- difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5 - ylloxylmethyl) phenyl1-5-fluoro-2-methoxypyridine-3- sulfonamide

[0390] To a stirred solution of 2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (100 mg, 0.26 mmol, 1 equiv) and pyridine (85 mg, 1.1 mmol, 4 equiv) in DCM (2 mL) was added 5-fluoro-2-methylpyridine-3 -sulfonyl chloride (84 mg, 0.4 mmol, 1.5 equiv) at room temperature. The reaction was stirred for 4 h and diluted with water (2 mL). The resulting mixture was extracted with DCM (3 x 10 mL), and the combined organics washed with water (2 x 10 mL), brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with EtOAc/PE (1 : 1) to afford N-[2,4- difluoro-3 -([[3 -methyl- l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (110 mg, 73% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 548.

Synthesis of Compound 38: N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine-3-sulfonamid e

[0391] A-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]py ridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methylpyridine-3-sulfonamid e (90 mg, 0.16 mmol, 1 equiv) was added to 4 M HC1 in MeOH (1 mL) at 0 °C and the resulting mixture warmed to room temperature over 2 h. The resulting mixture was concentrated under vacuum, and the residue was neutralized to pH 7 with NH3.H 2 O (5%). This was purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; mobile phase: 90 mL/min, 20-50% MeCN / 0.1% aqueous formic acid; to afford A-[2,4-difluoro-3- [([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phenyl ]-5-fluoro-2-methylpyridine- 3 -sulfonamide (37 mg, 48% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 464.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.72 (s, 1H), 8.69 (d, J= 2.8 Hz, 1H), 8.16 (d, J= 2.7 Hz, 1H), 7.93 -7.80 (m, 2H), 7.37 (td, J= 8.9, 5.9 Hz, 1H), 7.16 (t, J= 9.0 Hz, 1H), 5.11 (s, 2H), 2.76 (d, J = 1.2 Hz, 3H), 2.48 (s, 3H).

Example 50: Synthesis of N-[3-[([3-ethyl-1H-pyrazolo[3.,4-b]pyridin-5-yl]oxy)methyl]-

2,4-difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonam ide (Compound 39)

Compound 39

Synthesis of 39-a: l-(5-bromo-2-fluoropyridin-3-yl)propan-l-one

[0392] To a stirred solution of diisopropylamine (15 g, 148 mmol, 1.3 equiv) in THF (300 mL) was added 2.5 M butyllithium in hexanes (59 mL, 148 mmol, 1.3 equiv) dropwise at - 78 °C under nitrogen atmosphere. The LDA solution was stirred for 0.5 h at -78 °C, then 5- bromo-2-fluoropyridine (20 g, 114 mmol, 1 equiv) was added dropwise over 10 min at - 78 °C. The resulting mixture was stirred for 1 h at -65 °C, then N-methoxy-N- methylpropanamide (14.6 g, 125 mmol, 1.1 equiv) in THF (20 mL) was added dropwise over 10 min at -78 °C. The solution was stirred for 1 hr at -30 °C, before being quenched with sat. NH4CI (aq.) at low temperature. The resulting mixture was extracted with EtOAc (3 x 300 mL), and the combined organics washed with brine (500 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (8: 1) to afford l-(5-bromo-2-fluoropyridin-3- yl)propan-l-one (25 g, 95% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 232

Synthesis of 39-b: 5-bromo-3-ethyl-1H-pyrazolo[3,4-b1pyridine

[0393] To a stirred solution of l-(5-bromo-2-fluoropyridin-3-yl)propan-l-one (25 g, 108 mmol, 1 equiv) in EtOH (500 mL) was added NH 2 NH 2 .H 2 O (16.2 g, 323 mmol, 3 equiv) dropwise. The reaction was stirred for 16 h at 80 °C under nitrogen atmosphere, then cooled and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (6: 1) to afford 5-bromo-3-ethyl-lH-pyrazolo[3,4- b]pyridine (12 g, 49% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 226.

Synthesis of 39-c: 5-bromo-3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine

[0394] To a stirred solution of 5-bromo-3-ethyl-lH-pyrazolo[3,4-b]pyridine (12g, 53 mmol, 1 equiv) and dihydropyran (22 g, 265 mmol, 5 equiv) in THF (200 mL) was added DL- Camphor sulfonic acid (1.85 g, 8 mmol, 0.15 equiv). The reaction mixture was stirred for 2 h at 60 °C then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (8: 1) to afford 5-bromo-3-ethyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (11 g, 67% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 310.

Synthesis of 39-d: 3-ethyl-l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-L3,2-dioxaborol an-2-yl) pyrazolo [3,4-blpyridine

[0395] To a solution of 5-bromo-3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (10 g, 32 mmol, 1 equiv) and bis(pinacolato)diboron (12 g, 48 mmol, 1.5 equiv) in dioxane (150 mL) were added KO Ac (6.3 g, 64 mmol, 2 equiv) and Pd(dppf)C12 (1.32 g, 1.6 mmol, 0.05 equiv). After stirring for 2 h at 100 °C, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (8: 1) to afford 3-ethyl-l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaboro lan-2- yl)pyrazolo[3,4-b]pyridine (10g, 87% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 358.

Synthesis of 39-e: 3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-ol

[0396] To a stirred solution of 3-ethyl-l-(oxan-2-yl)-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine (2.6 g, 7.3 mmol, 1 equiv) in THF (20 mL) and NaOH (20 mL, 2% aq.) was added 30% H 2 O 2 (8.2 g, 73 mmol, 10 eq) dropwise. The reaction mixture was stirred for 2 h then quenched with sat. Na 2 S 2 O 3 (aq.). The resulting mixture was extracted with EtOAc (2 x 50 mL), and the combined organics were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (4: 1) to afford 3-ethyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-ol (1.2 g, 67% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 248.

Synthesis of 39-f: 3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1me thyl)-2,4- difluoroaniline

[0397] To a stirred solution of 3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (1.2 g, 4.8 mmol, 1 equiv), (3-amino-2,6-difluorophenyl)methanol (0.93 g, 5.8 mmol, 1.2 equiv) and PPh 3 (1.9 g, 7.3 mmol, 1.5 equiv) in DCM (30 mL) was added TMAD (1.25 g, 7.3 mmol, 1.5 equiv). The reaction was stirred for 2 h then diluted with water (50 mL) and extracted with DCM (2 x 50 mL). The combined organics were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (2: 1) to afford 3-([[3-ethyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)-2,4-difluoroanili ne (1.1 g, 58% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 389.

Synthesis of 39-g: N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1oxy 1methyl)-2,4- difluorophenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0398] To a stirred solution of 3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]oxy]methyl)-2,4-difluoroaniline (100 mg, 0.26 mmol, 1 equiv) and pyridine (81 mg, 1 mmol, 4 equiv) in DCM (3 mL) was added 5-fluoro-2-methoxypyridine-3 -sulfonylchloride (75 mg, 0.33 mmol, 1.3 equiv) in DCM (0.5 mL) dropwise. The resulting mixture was stirred for 16 h then diluted with water (10 mL). This was extracted with DCM (3 x 10 mL), and the combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (2: 1) to afford N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]oxy]methyl)-2,4-difluorophenyl]-5-fluoro-2-methoxypyridin e-3-sulfonamide (130 mg, 87% yield) as a white solid. Synthesis of Compound 39: N-[3-[([3-ethyl-1H-pyrazolo[3,4-b1pyridin-5-yl1oxy)methyl1-

2.4-difluorophenyl1-5-fluoro-2-methoxypyridine-3-sulfonam ide

[0399] A mixture of N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl] oxy]methyl)-

2.4-difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonam ide (110 mg, 0.19 mmol, 1 equiv) in 4 M HC1 in MeOH (2 mL) was stirred for 2 h then diluted with water (10 mL). The resulting mixture was extracted with EtOAc (2 x 10 mL). The combined organics were dried (MgSCU), concentrated under reduced pressure and the residue purified by Prep-HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; mobile phase: 20 mL/min, 2-27% MeCN / 0.05% aqueous ammonia; to afford N-[3-[([3-ethyl-lH- pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]-2,4-difluorophenyl]- 5-fluoro-2-methoxypyridine-3- sulfonamide (55 mg, 59% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 494.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.36 (s, 1H), 8.44 (d, J= 2.9 Hz, 1H), 8.17 (d, J= 2.7 Hz, 1H), 7.97 (dd, J= 13, 3.0 Hz, 1H), 7.87 (d, J= 2.8 Hz, 1H), 7.44-7.30 (m, 1H), 7.21-7.09 (m, 1H), 5.14 (s, 2H), 3.90 (s, 3H), 2.90 (q, J= 7.6 Hz, 2H), 1.32 (t, J= 7.6 Hz, 3H).

Example 51: Synthesis of 5-chloro-N-[3-[([3-ethyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]-2.,4-difluorophenyl]-2-methoxypyridine-3-sulf onamide (Compound 40)

Compound 40

Synthesis of 40-a: 5-chloro-N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin -5- ylloxylmethyl) -2,4-difluorophenyl1-2-methoxypyridine-3-sulfonamide

[0400] To a stirred solution of 3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]°xy]methyl)-2,4-difluoroaniline (100 mg, 0.26 mmol, 1 equiv) and pyridine (82 mg, 1.1 mmol, 4 equiv) in DCM (3 mL) was added 5-chloro-2-methoxypyridine-3-sulfonyl chloride(81 mg, 0.3 mmol, 1.3 equiv) in DCM (0.5 mL) dropwise. The resulting mixture was stirred for 16 h at room temperature then diluted with water (10 mL). This was extracted with DCM (3 x 10 mL), and the combined organics washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (2: 1) to afford 5-chloro-N-[3-([[3-ethyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)-2,4-difluoropheny l]-2-methoxypyridine-3- sulfonamide (130 mg, 85% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 594.

Synthesis of Compound 40: 5-chloro-N-[3-[([3-ethyl-1H-pyrazolol3,4-b1pyridin-5- yl1oxy)methyl1-2,4-difluorophenyl1-2-methoxypyridine-3-sulfo namide

[0401] A mixture of 5-chloro-N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin -5- yl]oxy]methyl)-2,4-difluorophenyl]-2-methoxypyridine-3-sulfo namide (80 mg, 0.13 mmol, 1 equiv) in 4 M HC1 in MeOH (1.5 mL) was stirred for 2 h at room temperature then concentrated. The solid formed was washed with MeCN (2 x 3 mL) and dried to give 5- chloro-N-[3-[([3-ethyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)me thyl]-2,4-difluorophenyl]-2- methoxypyridine-3 -sulfonamide (31 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 510.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.38 (s, 1H), 8.48 (d, J= 2.6 Hz, 1H), 8.18 (d, J= 2.7 Hz, 1H), 8.03 (d, J= 2.6 Hz, 1H), 7.87 (d, J= 2.7 Hz, 1H), 7.38 (td, J= 8.9, 6.0 Hz, 1H), 7.23-7.11 (m, 1H), 5.15 (s, 2H), 3.92 (s, 3H), 2.90 (q, J= 7.6 Hz, 2H), 1.32 (t, J= 7.6 Hz, 3H).

Example 52: Synthesis of 5-cyano-N-[3-[([3-ethyl-1H-pyrazolo[3.,4-b]pyridin-5- yl] methyl]-2.,4-difluorophenyl]-2-methoxypyridine-3-sulfonamide (Compound 41)

Compound 41 Synthesis of 41-a: 5-cyano-N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin- 5- yl1oxy1methyl)-2,4- difluorophenyl1-2-methoxypyridine-3-sulfonamide

[0402] To a stirred solution of 3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]°xy]methyl)-2,4-difluoroaniline (100 mg, 0.26 mmol, 1 equiv) and pyridine (82 mg, 1.1 mmol, 4 equiv) in DCM (3 mL) was added 5-cyano-2-methoxypyridine-3-sulfonyl chloride (78 mg, 0.36 mmol, 1.3 equiv) in DCM (0.5 mL). The reaction was stirred for 16 h then diluted with water (10 mL). The resulting mixture was extracted with DCM (3 x 10mL), and the combined organics washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (2: 1) to afford 5-cyano-N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin- 5- yl]oxy]methyl)-2,4-difluorophenyl]-2-methoxypyridine-3-sulfo namide (140 mg, 93% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 585.

Synthesis of Compound 41 : 5-cyano-N-13-l([3-ethyl-1H-pyrazolo[3,4-b1pyridin-5- yl1oxy)methyl1-2,4-difluorophenyl1-2-methoxypyridine-3-sulfo namide

[0403] A mixture of 5-cyano-N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin- 5- yl]oxy]methyl)-2,4-difluorophenyl]-2-methoxypyridine-3-sulfo namide (130 mg, 0.22 mmol, 1 equiv) in 4 M HC1 in MeOH (2 mL) was stirred for 2 h then diluted with water (10 mL). The resulting mixture was extracted with EtOAc (3 x 10 mL), and the combined organics concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; Flowrate 20 mL/min, Mobile Phase: 14-36% MeCN / 0.05% aqueous ammonia; to afford 5-cyano-N-[3-[([3-ethyl-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy)methyl]-2,4-difluorophenyl]-2-methoxypyri dine-3-sulfonamide (45 mg, 41% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 501.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.08 (s, 1H), 8.86 (d, J= 2.2 Hz, 1H), 8.42 (d, J= 2.3 Hz, 1H), 8.19 (d, J= 2.7 Hz, 1H), 7.88 (d, J= 2.7 Hz, 1H), 7.42-7.28 (m, 1H), 7.08 (t, J= 8.9 Hz, 1H), 5.13 (s, 2H), 3.98 (s, 3H), 2.90 (q, J= 7.6 Hz, 2H), 1.32 (t, J= 7.6 Hz, 3H). Example 53: Synthesis of 5-chloro-N-[2.,4-difluoro-3-[([l-methylpyrazolo[4.,3-b]pyrid in-

6-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfonamide (Compound 42)

Compound 42

Synthesis of 42-a: 6-bromo-l-methylpyrazolo[4,3-b]pyridine

[0404] To a stirred solution of 6-bromo-lH-pyrazolo[4,3-b]pyridine (700 mg, 3.5 mmol, 1 equiv) and CS 2 CO 3 (2.3 g, 7 mmol, 2 equiv) in DMF (7 mL) was added methyl iodide (552 mg, 3.9 mmol, 1.1 equiv) dropwise at room temperature. The resulting solution was stirred overnight, then quenched with water (20 mL). The mixture was extracted with EA (3 x 10 mL), and the combined organics washed with brine (2 x 5 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (3: 1) to afford 6-bromo-l-methylpyrazolo[4,3-b]pyridine (200 mg, 26% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 212

Synthesis of 42-b/c: l-methylpyrazolo[4,3-b]pyridin-6-ol

[0405] To a stirred mixture of 6-bromo-l-methylpyrazolo[4,3-b]pyridine (324 mg, 1.5 mmol, 1 equiv) and bis(pinacolato)diboron (582 mg, 2.3 mmol, 1.5 equiv) in dioxane (3 mL) were added KOAc (300 mg, 3.1 mmol, 2 equiv) and Pd(dppf)Cl 2 CH 2 Cl 2 (62 mg, 0.076 mmol, 0.05 equiv) under N2 atmosphere. The resulting mixture was stirred for 4 h at 90 °C under N2 atmosphere, then cooled. THF (1 mL) was added, followed by NaOH (1 mL, 2%) and 30% H 2 O 2 (1.73 g, 15.3 mmol, 10 equiv) at room temperature. The resulting mixture was stirred for 0.5 h then concentrated. The residue was purified by silica gel chromatography, eluting with PE:EA (3: l) to give l-methylpyrazolo[4,3-b]pyridin-6-ol (189 mg, 82% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 150

Synthesis of 42-d: 2,4-difluoro-3-[([i-methylpyrazolo[4,3-b1pyridin-6-yl1oxy)me thyl1aniline [0406] To a stirred mixture of l-methylpyrazolo[4,3-b]pyridin-6-ol (189 mg, 1.3 mmol, 1 equiv), (3 -amino-2,6-difluorophenyl)m ethanol (302 mg, 1.9 mmol, 1.5 equiv) and PPhs (500 mg, 1.9 mmol, 1.5 equiv) in THF (2 mL) was added DEAD (330 mg, 1.9 mmol, 1.5 equiv) dropwise at 0 °C. The resulting mixture was stirred for 2 h then concentrated. The residue was purified by silica gel chromatography, eluting with PE:EA (1 : 1) to give 2,4-difluoro-3- [([l-methylpyrazolo[4,3-b]pyridin-6-yl]oxy)methyl]aniline (330 mg, 89% yield) as a white solid.

Synthesis of Compound 42: 5-chloro-N-(2,4-difluoro-3-(((l-methyl-1H-pyrazolol4,3- b1pyridin-6-yl)oxy)methyl)phenyl)-2-methoxypyridine-3-sulfon amide

[0407] To a stirred solution of 2,4-difluoro-3-[([l-methylpyrazolo[4,3-b]pyridin-6- yl]oxy)methyl]aniline (100 mg, 0.34 mmol, 1 equiv) in pyridine (2 mL) was added 5-chloro- 2-methoxypyridine-3 -sulfonyl chloride (91 mg, 0.38 mmol, 1.1 equiv) in portions at room temperature. The resulting mixture was stirred for Ih then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 15-60% MeCN / 0.1% aqueous TFA; to give 5-chloro-N- (2,4-difluoro-3-(((1-methyl-lH-pyrazolo[4,3-b]pyridin-6-yl)o xy)methyl)phenyl)-2- methoxypyridine-3 -sulfonamide (120 mg, 70% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 496

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.41 (s, IH), 8.48 (d, J= 2.6 Hz, IH), 8.18 (dd, J= 9.2, 1.7 Hz, 2H), 8.04 (d, J= 2.6 Hz, IH), 7.84 (d, J= 2.6 Hz, IH), 7.42 (td, J= 9.0, 6.0 Hz, IH), 7.20 (td, J= 9.0, 1.5 Hz, IH), 5.20 (s, 2H), 4.04 (s, 3H), 3.92 (s, 3H). Example 54: Synthesis of N-[2,4-difluoro-3-[([l-methylpyrazolo[4.,3-b]pyridin-6-

Synthesis of Compound 43: N-[2,4-difluoro-3-[([i-methylpyrazolol4,3-b1pyridin-6- yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0408] To a stirred solution of 2,4-difluoro-3-[([l-methylpyrazolo[4,3-b]pyridin-6- yl]oxy)methyl]aniline (100 mg, 0.35 mmol, 1 equiv) in pyridine (4 mL) was added 5-fluoro- 2-methoxypyridine-3 -sulfonyl chloride (86 mg, 0.38 mmol, 1.1 equiv) in portions at room temperature. The resulting mixture was stirred for 4 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-70% MeCN / 0.1% aqueous TFA; to give N-[2,4- difluoro-3-[([l-methylpyrazolo[4,3-b]pyridin-6-yl]oxy)methyl ]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (120 mg, 73% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 480

'H NMR (300 MHz, DMSO-d 6 ) δ 10.40 (s, 1H), 8.45 (d, J= 2.9 Hz, 1H), 8.18 (dd, J= 8.8, 1.7 Hz, 2H), 7.99 (dd, J= 7.3, 3.0 Hz, 1H), 7.84 (dd, J= 2.6, 1.0 Hz, 1H), 7.41 (td, J= 8.9, 5.9 Hz, 1H), 7.19 (td, J= 9.1, 1.6 Hz, 1H), 5.19 (s, 2H), 4.04 (s, 3H), 3.91 (s, 3H).

Example 55: Synthesis of N-[2,4-difluoro-3-([[3-(trifluoromethyl)-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (Compound 44)

Compound 44

Synthesis of 44-a: l-(5-bromo-2-fluoropyri din-3 -yl)-2,2,2-trifluoroethanone

[0409] To a stirred solution of 5-bromo-2-fluoropyridine (5 g, 28.4 mmol, 1 equiv) in THF (35 mL) were added 2 M LDA solution in THF (15.6 mL, 31 mmol, 1.1 equiv) dropwise at - 78 °C under N2 atmosphere. The resulting solution was stirred for 1.5 h at -78 °C, then trifluoroethyl acetate (5.6 g, 40 mmol, 1.4 equiv) was added dropwise at -78 °C. The cooling bath was removed and the mixture was stirred for 1 h at room temperature. The reaction mixture was quenched with 1 M aqueous HC1 (10 mL) and extracted with EA (3 x 50 mL). The combined organics were washed with brine (2 x 25 mL), dried over anhydrous Na 2 SO 4 and concentrated to afford l-(5-bromo-2-fluoropyridin-3-yl)-2,2,2-trifluoroethanone (8 g, crude) as a red oil which was used in the next step directly without further purification. LCMS (ES, m/z): [M+H] + : 272

Synthesis of 44-b: 5-bromo-3-(trifluoromethyl)-1H-pyrazolo[3,4-b1pyridine

[0410] To a stirred solution of l-(5-bromo-2-fluoropyridin-3-yl)-2,2,2-trifluoroethanone (8 g, 29 mmol, 1 equiv) in EtOH (80 mL) was added NH 2 NH 2 H 2 O (3 mL, 59 mmol, 2 equiv) at room temperature. The resulting mixture was stirred at reflux for 2 h, then cooled and concentrated. The residue was dissolved in EA (200 mL), washed with water (2 x 50 mL), brine (2 x 20 mL) and dried over anhydrous Na 2 SO 4 . Concentration afforded 5-bromo-3- (trifluoromethyl)-lH-pyrazolo[3,4-b]pyridine (7 g, crude) as a brown oil which was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 266

Synthesis of 44-c: 5-bromo-3-(trifluoromethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine [0411] To a stirred solution of 5-bromo-3-(trifluoromethyl)-lH-pyrazolo[3,4-b]pyridine (7 g, 26 mmol, 1 equiv) in THF (70 mL) was added 60% NaH in oil (2.1 g, 52.6 mmol, 2 equiv) in portions at 0 °C. The resulting solution was stirred for 0.5 h at 0 °C, then SEMC1 (5.3 g, 31.6 mmol, 1.2 equiv) was added dropwise at 0 °C. The reaction mixture was stirred for 2 h at room temperature, then quenched with water (50 mL). The resulting mixture was extracted with EA (3 x 50 mL), and the combined organics were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE: EA (1 : 1) to afford 5-bromo-3-(trifluoromethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (6.5 g, 62% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 396

Synthesis of 44-d/e: 3-(trifluoromethyl)-l-[[2-(trimethylsilyl)ethoxy1methyl1pyra zolo[3,4- blpyridin-5-ol

[0412] To a solution of 5-bromo-3-(trifluoromethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (6.5 g, 16 mmol, 1 equiv) and bis(pinacolato)diboron (6.3 g, 25 mmol, 1.5 equiv) in dioxane (70 mL) were added KO Ac (3.3 g, 33 mmol, 2 equiv) and Pd(dppf) Cl 2 CH 2 Cl 2 (670 mg, 0.82 mmol, 0.05 equiv). After stirring for 2 h at 90 °C under a nitrogen atmosphere, the reaction mixture was allowed to cool. THF (50 mL) was added, followed by NaOH (30 mL, 2% aqueous) at room temperature. To this was added 30% H 2 O 2 (18.5 g, 164 mmol, 10 equiv) dropwise at 0 °C. The resulting mixture was stirred for 0.5 h then concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / water; to afford 3-(trifluoromethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyra zolo[3,4-b]pyridin-5-ol (2.3 g, 42% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 334

Synthesis of 44-f: 2,4-difluoro-3-([[3-(trifluoromethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)aniline [0413] To a stirred mixture of 3 -(trifluoromethyl)- 1 -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (550 mg, 1.7 mmol, 1 equiv), (3- amino-2,6-difluorophenyl)methanol (315 mg, 2 mmol, 1.2 equiv) and PPI13 (649 mg, 2.5 mmol, 1.5 equiv) in THF (10 mL) was added DEAD (430 mg, 2.5 mmol, 1.5 equiv) dropwise at 0 °C under N2 atmosphere. The resulting solution was stirred for 2 h then quenched with water (20 mL). This was extracted with EA (3 x 30 mL), and the combined organics were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 10-55% MeCN / water; to afford 2,4- difluoro-3-([[3-(trifluoromethyl)-l-[[2-(trimethylsilyl)etho xy]methyl]pyrazolo[3,4-b]pyridin- 5-yl]oxy]methyl)aniline (480 mg, 61% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 475

Synthesis of 44-g: N-[2A-difluoro-3-([[3-(trifluoromethyl)-l-IT2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0414] To a stirred solution of 2,4-difluoro-3-([[3-(trifluoromethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)aniline (260 mg, 0.55 mmol, 1 equiv) in pyridine (5 mL) was added 5 -fluoro-2-methoxypyridine-3 -sulfonyl chloride (185 mg, 0.82 mmol, 1.5 equiv) in portions. The resulting solution was stirred for 1 h at room temperature, then quenched with water (20 mL). This mixture was extracted with EA (3 x 10 mL), and the combined organics were washed with brine (2 x 5 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 20-65% MeCN / water; to afford N-[2,4-difluoro-3-([[3-(trifluoromethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (250 mg, 68% yield) as a white solid.

Synthesis of Compound 44: N-[2,4-difluoro-3-([[3-(trifluoromethyl)-1H-pyrazolol3,4- b1pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2-methoxypyridine -3-sulfonamide [0415] A solution of N-[2,4-difluoro-3-([[3-(trifluoromethyl)- l -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (200 mg, 0.3 mmol, 1 equiv) in TFA (2 mL) and DCM (2 mL) was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-75% MeCN / 0.1% aqueous TFA; to afford N-[2,4-difluoro-3-([[3-(trifluoromethyl)-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (140 mg, 87% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 534

1 H NMR (300 MHz, DMSO-d 6 ) d 8.47-8.34 (m, 2H), 7.96 (dd, J= 13, 3.0 Hz, 1H), 7.88 (d, J= 2.7 Hz, 1H), 7.38 (q, J= 8.2, 7.5 Hz, 1H), 7.14 (t, J= 8.9 Hz, 1H), 5.23 (s, 2H), 3.88 (s, 3H).

Example 56: Synthesis of/V-[2.,4-difluoro-3-([3-methyl-1H-pyrazolo[3.,4-b]pyridin- 5- yl]methoxy)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 45)

Compound 45

Synthesis of 45-a: 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridine-5-carbaldehyde

[0416] Into a 500 mL 3-necked round-bottom flask was placed 5-bromo-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (15 g, 51 mmol, 1 equiv) and tetrahydrofuran (160 mL). This was followed by the addition of 2.5 M n-BuLi in hexane (24.4 mL, 61 mmol, 1.2 equiv) dropwise with stirring at -78°C. The solution was stirred for 1 h at -78 °C, when dimethylformamide (7.4 g, 101 mmol, 2 equiv) was added dropwise at -78 °C. The resulting solution was stirred for 1 h at -78 °C, then quenched by the addition of 200 mL of saturated NH 4 Cl/ice. This mixture was extracted with 2 x 200 mL of ethyl acetate and the organics were combined, dried over anhydrous sodium sulfate and concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/PE (2/1) to give 3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridine-5-carbaldehyde (11 g, 88% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :246

Synthesis of 45-b: [3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1methanol [0417] Into a 50 mL 3-necked round-bottom flask was placed 3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine-5-carbaldehyde (2 g, 8 mmol, 1 equiv) in MeOH (20 mL). NaBEL (0.37 g, 9.78 mmol, 1.2 equiv) was added in portions at 0 °C. The resulting solution was stirred for 1 hr in a water/ice bath. The reaction was quenched by the addition of 50 mL of water/ice, then extracted with 3 x 50 mL of ethyl acetate. The combined organics were dried over anhydrous sodium sulfate and concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (2/1) to give [3 -methyl- l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]methanol (1.6 g, 79% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :248

Synthesis 45-c: of 3-amino-2,6-difluorophenol

[0418] Into a 50 mL round-bottom flask was placed 2,4-difluoro-3-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)aniline (1.9 g, 7 mmol, 1 equiv), THF (10 mL), NaOH (10 mL) and 30% H 2 O 2 (2.5 g, 74 mmol, 10 equiv). The reaction was stirred for 2 hr then quenched by the addition of 20 mL of saturated Na 2 S 2 O 4 . The resulting solution was extracted with 3 x 20 mL of ethyl acetate, and the extracts washed with 20 ml of NaCl solution, then dried over anhydrous sodium sulfate and concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1/1) to give 3-amino-2,6-difluorophenol (0.9 g, 83% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 146

Synthesis of 45-d: 2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridi n-5- yllmethoxylaniline

[0419] Into a 50 mL round-bottom flask was placed [3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methanol (1.5 g, 6 mmol, 1 equiv), DCM (30 mL), PPh 3 (2.4 g, 9 mmol, 1.5 equiv), DEAD (1.6 g, 9 mmol, 1.5 equiv) and 3-amino-2,6-difluorophenol (0.88 g, 6 mmol, 1 equiv). The resulting solution was stirred for 3 hr then concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (2/1) to give 2,4-difluoro-3- [[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin- 5-yl]methoxy]aniline (1.6 g, 70% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :375 Synthesis of 45-e: N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyr idin-5- yl1methoxy1phenyl)-5-fluoro-2-methoxypyridine-3-sulfonamide

[0420] Into an 8 mL vial was placed 2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methoxy]aniline (200 mg, 0.5 mmol, 1 equiv), pyridine (3 mL) and 5-fluoro-2- methoxypyridine-3 -sulfonyl chloride (181 mg, 0.8 mmol, 1.5 equiv). The resulting solution was stirred overnight then was concentrated. The crude product was purified by Flash-Prep- HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 5-60% MeCN / 0.1% aqueous formic acid; Detector 220 nm; to give N- (2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyrid in-5-yl] methoxy]phenyl)-5- fluoro-2-methoxypyridine-3-sulfonamide (180 mg, 60% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :564

Synthesis of Compound 45: N-[2,4-difluoro-3-([3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1methoxy)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0421] Into an 8 mL vial was placed N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methoxy]phenyl)-5-fluoro-2-methoxypyridine-3- sulfonamide (170 mg, 0.3 mmol, 1 equiv) and 4 M HC1 in 1,4-di oxane (2 mL). The resulting solution was stirred for 12 hr then concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; mobile phase: 18-58% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give N-[2,4-difluoro-3-([3-methyl- lH-pyrazolo[3,4-b]pyridin-5-yl]methoxy)phenyl]-5-fluoro-2-me thoxypyridine-3-sulfonamide (67 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + 480

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.28 (s, 1H), 10.28 (s, 1H), 8.42 (d, J= 3.0 Hz, 2H), 8.16 (d, J = 2.0 Hz, 1H), 7.91 (dd, J = 7.3, 3.0 Hz, 1H), 7.13-7.01 (m, 1H), 6.94 (td, J= 8.7, 5.4 Hz, 1H), 5.22 (s, 2H), 3.90 (s, 3H), 2.51 (s, 3H).

Example 57: Synthesis of 5-chloro-N-[2,4-difluoro-3-([3-methyl-1H-pyrazolo [3,4- b]pyridin-5-yl]methoxy)phenyl]-2-methoxypyridine-3-sulfonami de (Compound 46)

Compound 46

Synthesis of 46-a: 5-chloro-N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolol 3,4- b1pyridin-5-yl1methoxy1phenyl)-2-methoxypyridine-3-sulfonami de

[0422] Into an 8 mL vial was placed 2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methoxy]aniline (200 mg, 0.5 mmol, 1 equiv), pyridine (3 mL) and 5-chloro- 2-methoxypyridine-3 -sulfonyl chloride (194 mg, 0.8 mmol, 1.5 equiv). The resulting solution was stirred overnight then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 5-60% MeCN / 0.1% aqueous formic acid; Detector UV 254 nm; to give 5- chloro-N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3, 4-b]pyridin-5- yl]methoxy]phenyl)-2-methoxypyridine-3-sulfonamide (170 mg, 55% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :580

Synthesis of Compound 46: 5-chloro-N-12,4-difluoro-3-([3-methyl-1H-pyrazolo 13,4- b1pyridin-5-yl1methoxy)phenyl1-2-methoxypyridine-3-sulfonami de

[0423] Into an 8 mL vial was placed 5-chloro-N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5- yl]methoxy]phenyl)-2-methoxypyridine-3-sulfonamide (165 mg, 0.3 mmol, 1 equiv) and 2 M HC1 in 1,4-di oxane (2 mL). The resulting solution was stirred for 12 h then concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; mobile phase: 25-60% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give 5-chloro-N-[2,4- difluoro-3-([3 -methyl- IH-pyrazolo [3,4-b]pyridin-5-yl]methoxy)phenyl]-2-methoxypyridine- 3-sulfonamide (57 mg, 47% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 496

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.28 (s, 1H), 10.30 (s, 1H), 8.45 (dd, J= 5.3, 2.3 Hz, 2H), 8.17 (d, J= 2.0 Hz, 1H), 7.98 (d, J= 2.6 Hz, 1H), 7.13-7.01 (m, 1H), 6.95 (td, J= 8.7, 5.5 Hz, 1H), 5.23 (s, 2H), 3.91 (s, 3H), 2.51 (s, 3H).

Example 58: Synthesis of N-[3-[([3-ethyl-1H-pyrazolo[3.,4-b]pyridin-5-yl]oxy)methyl]- 2,4-difluorophenyl]-5-fluoro-2-methylpyridine-3-sulfonamide (Compound 47)

Compound 47

Synthesis of 47-a: N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolol3,4-b1pyridin-5-yl1oxy 1methyl)-2,4- difluorophenyl1-5-fluoro-2-methylpyridine-3-sulfonamide

[0424] To a stirred solution of 3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]oxy]methyl)-2,4-difluoroaniline (100 mg, 0.26 mmol, 1 equiv) and pyridine(163 mg, 2 mmol, 8 equiv) in DCM (3 mL) was added 5-fluoro-2-methylpyridine-3 -sulfonyl chloride (162 mg, 0.77 mmol, 3 equiv) in DCM (0.5 mL) dropwise. The reaction mixture was stirred for 16 h then diluted with water (10 mL). This was extracted with DCM (3 x 10 mL), and the combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (2: 1) to afford N-[3-([[3-ethyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)-2,4-difluoropheny l]-5-fluoro-2 -methylpyridine- 3-sulfonamide (110 mg, 76% yield) as a white solid.

LCMS (ES, m/z): [M+H] + :562. Synthesis of Compound 47: N-[3-[([3-ethyl-1H-pyrazolo[3,4-b1pyridin-5-yl1oxy)methyl1-2 ,4- difluorophenyl1-5-fluoro-2-methylpyridine-3-sulfonamide

[0425] A mixture of N-[3-([[3-ethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy ]methyl)- 2,4-difluorophenyl]-5-fluoro-2-methylpyridine-3-sulfonamide (110 mg, 0.2 mmol, 1 equiv) in 1.25 M HC1 in MeOH (2 mL) was stirred for 2 h then diluted with water (10 mL). The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organics were dried (MgSO 4 ) and concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; mobile phase: 20 mL/min, 12-28% MeCN / 0.05% aqueous ammonia; to afford N-[3-[([3-ethyl-lH- pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]-2,4-difluorophenyl]- 5-fluoro-2-methylpyridine-3- sulfonamide (46 mg, 45% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 478.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.71 (s, 1H), 8.70 (d, J= 2.8 Hz, 1H), 8.16 (d, J= 2.8 Hz, 1H), 7.93 -7.82 (m, 2H), 7.37 (td, J= 8.9, 5.9 Hz, 1H), 7.17 (td, J= 9.0, 1.6 Hz, 1H), 5.12 (s, 2H), 2.90 (q, J= 7.6 Hz, 2H), 2.75 (d, J= 1.2 Hz, 3H), 1.31 (t, J= 7.6 Hz, 3H).

Example 59: Synthesis of : 5-cyano-N-(2.,4-difluoro-3-((3-methyl-1H-pyrazolo[3.,4- b]pyridin-5-yl)methoxy)phenyl)-2-methoxypyridine-3-sulfonami de (Compound 48)

Compound 48

Synthesis of 48-a: 5-cyano-N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3 ,4- b]pyridin-5-yl]methoxy]phenyl)-2-methoxypyridine-3-sulfonami de

[0426] Into an 8 mL vial was placed 2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methoxy]aniline (200 mg, 0.5 mmol, 1 equiv), pyridine (3 mL) and 5-cyano-2- methoxypyridine-3 -sulfonyl chloride (186 mg, 0.8 mmol, 1.5 equiv). The resulting solution was stirred overnight then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 5-60% MeCN / 0.5% aqueous formic acid; Detector 220 nm; to give 5-cyano- N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyr idin-5-yl]methoxy]phenyl)-2- methoxypyridine-3 -sulfonamide (185 mg, 61% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :571

Synthesis of Compound 48: 5-cyano-N-(2,4-difluoro-3-((3-methyl-1H-pyrazolol3,4- b1pyridin-5-yl)methoxy)phenyl)-2-methoxypyridine-3-sulfonami de

[0427] Into a 50 mL round-bottom flask was placed 5-cyano-N-(2,4-difluoro-3-[[3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methoxy]phenyl)-2-met hoxypyridine-3-sulfonamide (185 mg, 0.3 mmol, 1 equiv) and 4 M HC1 in 1,4-dioxane (2 mL). The resulting solution was stirred for 2 h then concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; mobile phase: 15-55% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give 5-cyano-N-(2,4- difluoro-3-((3 -methyl- lH-pyrazolo[3,4-b]pyridin-5-yl)methoxy)phenyl)-2-methoxypyri dine- 3-sulfonamide (72 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 487

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.28 (s, 1H), 10.40 (s, 1H), 8.88 (d, J= 2.3 Hz, 1H), 8.43 (dd, J= 8.7, 2.1 Hz, 2H), 8.25-8.10 (m, 1H), 7.18-6.84 (m, 2H), 5.22 (s, 2H), 3.99 (s, 3H), 2.49 (d, J= 1.2 Hz, 3H).

Example 60: Synthesis of 5-cyano-N-[2.,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]methyl)amino]phenyl]-2-methoxypyridine-3-sulf onamide (Compound

Compound 49

Synthesis of 49-a: tert-butyl (3-amino-2,4-difluorophenyl)carbamate

[0428] To a stirred mixture of 2, 6-difluoro-3 -nitroaniline (300 mg, 1.7 mmol, 1 equiv) and BOC2O (414 mg, 1.9 mmol, 1.1 equiv) in MeOH (10 mL) was added 10% Pd/C (30 mg, 0.28 mmol, 0.16 equiv). The reaction mixture was stirred for 5 h at room temperature under a hydrogen atmosphere. The resulting mixture was filtered; the filter cake was washed with MeOH (3 x 3 mL) and the filtrate concentrated. The residue was purified by silica gel column chromatography, eluting with PEZEA (3/1) to afford tert-butyl N-(3-amino-2,4- difluorophenyl)carbamate (212 mg, 50% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 245

Synthesis of 49-b: tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b1pyridin-5-yl1methyl1amino)phenyl1carbamate

[0429] Into a 40 mL vial were added tert-butyl N-(3-amino-2,4-difluorophenyl)carbamate (500 mg, 2 mmol, 1 equiv), 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-5-carbaldehyde (502 mg, 2 mmol, 1 equiv), MeOH (10 mL), HO Ac (2 mL) and NaBH 3 CN (386 mg, 6.1 mmol, 3 equiv). The reaction mixture was stirred overnight at 60 °C, then concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 10- 85% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give tert-butyl N-[2,4-difluoro- 3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methy l]amino)phenyl] carbamate (100 mg, 10% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 474

Synthesis of 49-c: 2,6-difluoro-Nl-([3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1methyl)benzene-L3-di amine

[0430] Into a 40 mL vial was placed tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]amino)phenyl]carbamate (100 mg, 0.2 mmol, 1 equiv), DCM (3 mL) and TFA (1 mL). The resulting solution was stirred for 3 h then concentrated. The pH was adjusted to 8 with ammonia solution and this was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 5-55% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give 2,6-difluoro-Nl-([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]me thyl)benzene- 1,3-diamine (40 mg, 65% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 290

Synthesis of Compound 49: 5-cyano-N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4- b1pyridin-5-yl1methyl)amino1phenyl1-2-methoxypyridine-3-sulf onamide

[0431] Into an 8 mL vial were added 2,6-difluoro-Nl-([3-methyl-lH-pyrazolo[3,4-b]pyridin- 5-yl]methyl)benzene- 1,3 -diamine (40 mg, 0.14 mmol, 1 equiv) and DCM (2 mL), followed by pyridine (110 mg, 1.4 mmol, 10 equiv) and 5-cyano-2-methoxypyridine-3-sulfonyl chloride (48 mg, 0.2 mmol, 1.5 equiv). The mixture was stirred for 1 h, then concentrated.

The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-50% MeCN / 0.1% aqueous formic acid; detector 220 nm; to give 5-cyano-N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4- b]pyridin-5-yl]methyl)amino]phenyl]-2-methoxypyridine-3-sulf onamide (9 mg, 13% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 486

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.11 (s, 1H), 10.14 (s, 1H), 8.80 (d, J= 2.2 Hz, 1H), 8.36 (d, J= 2.1 Hz, 2H), 7.97 (d, J= 2.0 Hz, 1H), 6.82 (t, J= 10.3 Hz, 1H), 6.51-6.40 (m, 1H), 6.00-5.91 (m, 1H), 4.42 (s, 2H), 3.95 (s, 3H), 2.46 (s, 3H).

Example 61: Synthesis of 5-cyano-N-[2.,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (Compound 50)

Compound 50

Synthesis of 50-a: N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1py ridin-5- ylloxylmethyl) phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0432] To a stirred solution of 2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl) aniline (100 mg, 0.27 mmol, 1 equiv) and pyridine (84 mg, 1.1 mmol, 4 equiv) in DCM (2 mL) was added 5-cyano-2-methoxypyridine-3-sulfonyl chloride (93 mg, 0.4 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for 4 h then diluted with water (2 mL). This was extracted with DCM (3 x 10 mL), and the combined organics were washed with water (2 x 10 mL) and brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluted with EtOAc/PE (0-50%) to afford N-[2,4-difluoro-3-([[3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5- fluoro-2-methoxypyridine-3- sulfonamide (120 mg, 80% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 571.

Synthesis of Compound 50: 5-cyano-N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-2-methoxypyridine-3-sulfon amide

[0433] To a stirred mixture of N-[2, 4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (100 mg, 0.17 mmol, 1 equiv) was added 4 M HC1 in MeOH (1 mL, 4 mmol, 22 equiv) dropwise at 0-5 °C. The resulting mixture was stirred for 1 h at room temperature, then concentrated. The residue was neutralized to pH 7 with 5% ammonia solution and purified by Prep-HPLC with the following conditions: Column: welch Vltimate XB-C18, 50 x 250 mm, 10 μm; mobile phase: 90 mL/min, 20-50% MeCN / 0.1% aqueous formic acid; to afford 5-cyano-N-[2,4-difluoro-3- [([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phenyl ]-2-methoxypyridine-3- sulfonamide (34 mg, 40% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 487.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 8.89 (d, J= 2.3 Hz, 1H), 8.45 (d, J= 2.2 Hz, 1H), 8.19 (d, J= 2.7 Hz, 1H), 7.86 (d, J= 2.8 Hz, 1H), 7.38 (td, J= 9.0, 5.9 Hz, 1H), 7.14 (t, J= 8.9 Hz, 1H), 5.13 (s, 2H), 4.00 (s, 3H), 2.48 (s, 3H).

Example 62: Synthesis of N-[2,4-difluoro-3-([3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]methoxy)phenyl]-5-fluoro-2-methylpyridine-3-sulfonamide (Compound 51)

Compound 51

Synthesis of 51-a: N-(2N-difluoro-3-[[3-rnethyl-l-(oxan-2-yl)pyrazolol3,4-b]pyr idin-5- yl]methoxy]phenyl)-5-fluoro-2-methylpyridine-3-sulfonamide

[0434] Into an 8 mL vial was placed 2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methoxy]aniline (200 mg, 0.5 mmol, 1 equiv), pyridine (3 mL) and 5-fluoro-2- methylpyridine-3 -sulfonyl chloride (224 mg, 1 mmol, 2 equiv) and the solution stirred overnight. The reaction mixture was concentrated, and the crude product purified by Flash- Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40 μm, 120 g; mobile phase: 5-60% MeCN / 0.5% aqueous formic acid; Detector 220 nm; to give N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyr idin-5- yl]methoxy]phenyl)-5-fluoro-2-methylpyridine-3-sulfonamide (210 mg, 72% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 548

Synthesis of Compound 51 : N-[2,4difluoro-3-([3-methyl-lEl-pyrazolo[3,4-b]pyridin-5- yl1methoxy)phenyl1-5-fluoro-2-methylpyridine-3-sulfonamide

[0435] Into an 8 mL vial was placed N-(2,4-difluoro-3-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]methoxy]phenyl)-5-fluoro-2-methylpyridine-3-s ulfonamide (205 mg, 0.4 mmol, 1 equiv) and 4 M HC1 in 1,4-di oxane (2 mL). The resulting solution was stirred for 2 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20 MeCN / 0.1% aqueous formic acid; detector, 220 nm; to give N-[2,4-difluoro-3-([3-methyl-lH- pyrazolo[3,4-b]pyridin-5-yl]methoxy)phenyl]-5-fluoro-2-methy lpyridine-3-sulfonamide (100 mg, 58% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 464

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.28 (s, 1H), 10.61 (s, 1H), 8.69 (d, J= 2.8 Hz, 1H), 8.43 (d, J= 2.0 Hz, 1H), 8.16 (d, J= 2.0 Hz, 1H), 7.83 (dd, J= 8.2, 2.9 Hz, 1H), 7.07 (td, J= 10.5, 9.9, 1.9 Hz, 1H), 6.93 (td, J= 8.8, 5.5 Hz, 1H), 5.21 (s, 2H), 2.69 (d, J= 1.2 Hz, 3H), 2.49 (d, J= 1.2 Hz, 3H).

Example 63: Synthesis of N-[2,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3.,4-b]pyridin- 5- yl] methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 52)

Synthesis of 52-a: l-(5-bromo-2-fluoropyri din-3 -yl)-2-methylpropan-l-ol

[0436] To a stirred solution of 5-bromo-2-fluoropyridine (10 g, 57 mmol, 1 equiv) in THF (200 mL) was added 2M LDA in THF solution (37 mL, 74 mmol, 1.3 equiv) dropwise at - 78 °C under nitrogen atmosphere. The solution was stirred for 30 min at -60 °C, then isobutyraldehyde (4.9 g, 68 mmol, 1.2 equiv) was added dropwise over 5 min at -78 °C. The resulting mixture was stirred for 1 h at -30 °C, then quenched with sat. NH4CI (aq.) at low temperature. This was extracted with EtOAc (3 x 100 mL), and the combined organics were washed with brine (300 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (7: 1) to afford l-(5-bromo-2-fhioropyridin-3-yl)-2-methylpropan-l-ol (8 g, 57% yield) as a light yellow oil. LCMS (ES, m/z): [M+H] + : 248.

Synthesis of 52-b: l-(5-bromo-2-fluoropyridin-3-yl)-2-methylpropan-l-one

[0437] To a stirred solution of l-(5-bromo-2-fhioropyridin-3-yl)-2-methylpropan-l-ol (8 g, 32 mmol, 1 equiv) in DCM (150 mL) was added pyridinium chlorochromate (13.9 g, 65 mmol, 2 equiv). The resulting mixture was stirred for 3 h at 40 °C, then filtered, the filter cake being washed with DCM (3 x 20 mL). The filtrate was concentrated and the residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford l-(5- bromo-2-fluoropyridin-3-yl)-2-methylpropan-l-one (5 g, 63% yield) as a light yellow solid. LCMS (ES, m/z): [M+H] + : 246.

Synthesis of 52-c: 5-bromo-3-isopropyl-1H-pyrazolo[3,4-b]pyridine

[0438] To a stirred solution of l-(5-bromo-2-fhioropyridin-3-yl)-2-methylpropan-l-one (5 g, 20 mmol, 1 equiv) in EtOH (60 mL) was added hydrazine hydrate (3.05 g, 61 mmol, 3 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 80 °C, then concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (2: 1) to afford 5-bromo-3-isopropyl-lH-pyrazolo[3,4-b]pyridine (2.5 g, 51% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 240.

Synthesis of 52-d: 5-bromo-3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine

[0439] To a stirred solution of 5-bromo-3-isopropyl-lH-pyrazolo[3,4-b]pyridine (2.4 g, 10 mmol, 1 equiv) and dihydropyran (4.2 g, 50 mmol, 5 equiv) in THF (40 mL) was added DL- Camphorsulfonic acid (347 mg, 1.5 mmol, 0.15 equiv). The reaction was stirred for 2 h at 60 °C, then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford 5 -bromo-3 -isopropyl- 1- (oxan-2-yl)pyrazolo[3,4-b]pyridine (2.5 g, 77% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 324.

Synthesis of 52-e: 3-isopropyl-l-(tetrahydro-2H-pyran-2-yl)-5-(4,4,5,5-tetramet hyl-L3,2- dioxaborolan -2-yl)-1H-pyrazolo[3,4-b1pyridine

[0440] To a solution of 5-bromo-3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (2.6 g, 8 mmol, 1 equiv) and bis(pinacolato)diboron (3.05 g, 12 mmol, 1.5 equiv) in dioxane (50 mL) were added KO Ac (1.57 g, 16 mmol, 2 equiv) and Pd(dppf)C12 (587 mg, 0.8 mmol, 0.1 equiv). After stirring for 2 h at 100 °C under nitrogen atmosphere, desired product could be detected by LCMS. The resulting mixture was used in the next step directly without any workup.

LCMS (ES, m/z): [M+H] + : 372. Synthesis of 52-f: 3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-ol

[0441] To the solution from the previous step was added THF (50 mL) and NaOH (2% aq., 40 mL), followed by 30% H 2 O 2 (7.6 g, 67 mmol, 8 equiv) dropwise at room temperature. The mixture was stirred for 2 h at room temperature under air, then diluted with water (50 mL). This was extracted with EtOAc (3 x 30 mL), and the combined organics were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (1 : 1) to afford 3 -isopropyl- 1- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (1.5 g, 85% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + :262.

Synthesis of 52-g: 2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b1py ridin-5-yl1 oxy]methylaniline

[0442] To a stirred solution of 3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (750 mg, 2.8 mmol, 1 equiv), (3-amino-2,6-difluorophenyl)methanol (548 mg, 3.4 mmol, 1.2 equiv) and PPI13 (1.13 g, 4.3 mmol, 1.5 equiv) in DCM (15 mL) was added TMAD (741 mg, 4.3 mmol, 1.5 equiv) at room temperature. The reaction mixture was stirred for 1 h, then diluted with water (30 mL). The resulting mixture was extracted with DCM (2 x 30 mL), and the combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (3: 1) to afford 2,4-difluoro-3-([[3-isopropyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)aniline (600 mg, 52% yield) as off-white solid.

LCMS (ES, m/z): [M+H] + : 403.

Synthesis of 52-h: N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b 1pyridin-5- ylloxy] methyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0443] To a stirred solution of 2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (100 mg, 0.25 mmol, 1 equiv) and pyridine (78 mg, 1 mmol, 4 equiv) in DCM (3 mL) was added 5-fluoro-2-methoxypyridine-3 -sulfonyl chloride (73 mg, 0.32 mmol, 1.3 equiv) in DCM (0.5 mL) dropwise. The resulting mixture was stirred for 16 h then concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (4: 1) to afford N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (110 mg, 75% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 592. Synthesis of Compound 52: N-[2,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3,4-b1pyridin-5 - yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0444] A solution of N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b ]pyridin-5- yl]oxy]methyl) phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (110 mg, 0.2 mmol, 1 equiv) in 4 M HC1 in MeOH (2 mL) was stirred for 2 h then concentrated. The residue was purified by Prep-HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; mobile phase: 20 mL/min, 16-38% MeCN / 0.05% aqueous ammonia; to afford N-[2,4- difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy )methyl]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (50 mg, 49% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 508.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.06 (s, 1H), 10.35 (s, 1H), 8.43 (d, J= 3.0 Hz, 1H), 8.16 (s, 1H), 8.02-7.93 (m, 1H), 7.88 (s, 1H), 7.37 (d, J= 7.8 Hz, 1H), 7.15 (t, J= 8.9 Hz, 1H), 5.16 (s, 2H), 3.90 (s, 3H), 3.30-3.28 (m, 1H), 1.36 (d, J = 6.9 Hz, 6H).

Example 64: Synthesis of 5-chloro-N-[2.,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (Compound 53)

Synthesis of 53-a: 5-chloro-N-[2A-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazo lo[3,4- blpyridin-5-ylloxyl methyl)phenyl1-2-methoxypyridine-3-sulfonamide

[0445] To a stirred solution of 2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (100 mg, 0.25 mmol, 1 equiv) and pyridine (78 mg, 0.1 mmol, 4 equiv) in DCM (3 mL) was added 5-chloro-2-methoxypyridine-3-sulfonyl chloride (78 mg, 0.32 mmol, 1.3 equiv) in DCM (0.5 mL) dropwise, and the reaction was stirred for 16 h, before being concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford 5-chloro-N-[2,4-difluoro-3-([[3- isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methy l)phenyl]-2-methoxypyridine- 3-sulfonamide (120 mg, 79% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + :608.

Synthesis of Compound 53: 5-chloro-N-[2,4-difluoro-3-[([3-isopropyl-1H-pyrazolol3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-2-methoxypyridine-3-sulfon amide

[0446] A solution of 5-chloro-N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyraz olo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-2-methoxypyridine-3-sulfon amide (120 mg, 0.2 mmol, 1 equiv) in 4N HC1 in MeOH (2 mL) was stirred for 2 h then diluted with water (10 mL). The aqueous layer was extracted with EtOAc (2 x 10 mL), and the combined organics were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; mobile phase: 20 mL/min, 17-42% MeCN / 0.05% aqueous ammonia; to afford 5-chloro-N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (50 mg, 45% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 524.

1 H NMR (300 MHz, DMSO-d 6 ) 513.05 (s, 1H), 10.37 (s, 1H), 8.47 (d, J= 2.5 Hz, 1H), 8.17 (s, 1H), 8.03 (d, J = 2.5 Hz, 1H), 7.88 (s, 1H), 7.37 (d, J= 8.3 Hz, 1H), 7.16 (t, J= 9.1 Hz, 1H), 5.16 (s, 2H), 3.91 (s, 3H), 3.30-3.28 (m, 1H), 1.37 (d, J = 7.0 Hz, 6H).

Example 65: Synthesis of N-[2,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3.,4-b]pyridin- 5- yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine-3-sulfonamid e (Compound 54)

Compound 54 Synthesis of 54-a: N-[2A-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b] pyridin-5- ylloxylmethyl) phenyl1-5-fluoro-2-methylpyridine-3-sulfonamide

[0447] To a stirred solution of 2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (100 mg, 0.25 mmol, 1 equiv) and DIEA (129 mg, 1 mmol, 4 equiv) in DCM (3 mL) was added 5-fluoro-2-methylpyridine-3 -sulfonyl chloride (156 mg, 0.75 mmol, 3 equiv) in DCM (0.5 mL) dropwise. The reaction mixture was stirred for 20 h then diluted with water (10 mL). This was extracted with DCM (2 x 10 mL), and the combined organics washed with brine (2 x 10 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (4: 1) to afford N-[2,4-difluoro-3-([[3-isopropyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5- fluoro-2-methylpyridine-3- sulfonamide (100 mg, 70% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 576.

Synthesis of Compound 54: N-[2,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3,4-b1pyridin-5 - yl1oxy)methyl1phenyl1-5-fluoro-2-methylpyridine-3-sulfonamid e

[0448] A solution of N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4-b ]pyridin-5- yl] oxy]methyl)phenyl]-5-fluoro-2-methylpyridine-3-sulfonamide (100 mg, 0.19 mmol, 1 equiv) in 4 M HC1 in MeOH (1.5 mL) was stirred for 2 h then diluted with water (10 mL). The resulting mixture was extracted with EtOAc (2 x 10 mL). The combined organics were washed with brine (10 mL), dried over anhydrous ISfeSCL and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; mobile phase: 20 mL/min, 13-35% MeCN / 0.05% aqueous ammonia; to afford N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4-b]pyridin-5 - yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine-3-sulfonamid e (37 mg, 39% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 492.

1 H NMR (300 MHz, DMSO-d 6 ) 513.05 (s, 1H), 10.70 (s, 1H), 8.70 (d, J= 2.8 Hz, 1H), 8.15 (d, J= 2.7 Hz, 1H), 7.92-7.83 (m, 2H), 7.37 (td, J= 8.9, 5.9 Hz, 1H), 7.16 (td, J= 9.0, 1.7 Hz, 1H), 5.14 (s, 2H), 3.30-3.26 (m, 1H), 2.78-2.72 (m, 3H), 1.36 (d, J= 6.9 Hz, 6H). Example 66: Synthesis of 5-cyano-N-[2.,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfon amide (Compound 55)

Compound 55

Synthesis of 55-a: 5-cyano-N-[2A-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazol o[3,4- blpyridin-5-ylloxylmethyl) phenyl1-2-methoxypyridine-3-sulfonamide

[0449] To a stirred solution of 2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (100 mg, 0.25 mmol, 1 equiv) and pyridine (79 mg, 1 mmol, 4 equiv) in DCM (3 mL) was added 5-cyano-2-methoxypyridine-3-sulfonyl chloride (75 mg, 0.32 mmol, 1.3 equiv) in DCM (0.5 mL) dropwise. The resulting mixture was stirred for 16 h then concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (4: 1) to afford 5-cyano-N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-2-methoxyp yridine-3-sulfonamide (120 mg, 81% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :599.

Synthesis of Compound 55: 5-cyano-N-[2,4-difluoro-3-[([3-isopropyl-1H-pyrazolo[3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-2-methoxypyridine-3-sulfon amide

[0450] A solution of 5-cyano-N-[2,4-difluoro-3-([[3-isopropyl-l-(oxan-2-yl)pyrazo lo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-2-methoxypyridine-3-sulfon amide (120 mg, 0.2 mmol, 1 equiv) in 4 M HC1 in MeOH (2 mL) was stirred for 1 h at 40 °C. The resulting mixture was diluted with water (10 mL) and extracted with EtOAc (2 x 10 mL).The combined organics were washed with brine (10 mL), dried over anhydrous ISfeSCL and concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column: XbridgeC18, 19*150 mm, 5 μm; mobile phase: 20 mL/min, 17-47% MeCN / 0.05% aqueous ammonia; to afford 5-cyano-N-[2,4-difluoro-3-[([3-isopropyl-lH-pyrazolo[3,4-b]p yridin-5- yl]oxy)methyl]phenyl]-2-methoxypyridine-3-sulfonamide (55 mg, 53% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 515.

1 H NMR (300 MHz, DMSO-d 6 ) δ13.05 (s, 1H), 10.47 (s, 1H), 8.91 (d, J= 2.3 Hz, 1H), 8.45

(d, J= 2.3 Hz, 1H), 8.17 (d, J= 2.7 Hz, 1H), 7.88 (d, J= 2.8 Hz, 1H), 7.46-7.32 (m, 1H), 7.21-7.09 (m, 1H), 5.15 (s, 2H), 4.00 (s, 3H), 3.27, (m, 1H), 1.37 (d, J= 6.9 Hz, 6H).

Example 67: Synthesis of N-[3,5-difluoro-4-[([3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]pyridin-2-yl]-5-fluoro-2-methoxypyridine-3-sul fonamide (Compound 56)

Compound 56

Synthesis of 56-a: methyl 2-amino-3,5-difluoropyridine-4-carboxylate

[0451] To a solution of 3,5-difluoro-4-iodopyridin-2-amine (640 mg, 2.5 mmol, 1 equiv) and TEA (760 mg, 7.5 mmol, 3 equiv) in MeOH (30 mL) was added Pd(dppf)Cl 2 CH 2 Cl 2 (204 mg, 0.25 mmol, 0.1 equiv) in a pressure tank. The mixture was purged with nitrogen for 3 min, then pressurized to 15 atm with carbon monoxide at 70 °C for 3 hrs. The reaction mixture was cooled and filtered. The filtrate was concentrated and the residue purified by silica gel column chromatography, eluting with PE: EA (4: 1) to afford methyl 2-amino-3,5- difluoropyridine-4-carboxylate (260 mg, 55% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 189 Synthesis of 56-b: (2-amino-3,5-difluoropyridin-4-yl)methanol

[0452] To a stirred solution of methyl 2-amino-3,5-difluoropyridine-4-carboxylate (250 mg, 1.3 mmol, 1 equiv) in THF (3 mL) was added LAH (76 mg, 2 mmol, 1.5 equiv) in portions at 0 °C. The resulting solution was stirred for 1 h at 0 °C, then quenched with HOAc (0.5 mL) and concentrated. The residue was purified by Prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-30% MeCN / 0.05% aqueous ammonia; to afford (2-amino-3,5-difluoropyridin-4-yl)methanol (120 mg, 56% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 161

Synthesis of 56-c: 3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyrid in-5- ylloxylmethyl) pyridin-2-amine

[0453] To a stirred mixture of 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (110 mg, 0.47 mmol, 1 equiv) and (2-amino-3,5-difluoropyridin-4-yl)methanol (90 mg, 0.56 mmol, 1.2 equiv) in THF (3 mL) were added PPh 3 (148 mg, 0.56 mmol, 1.2 equiv) and TMAD (98 mg, 0.56 mmol, 1.2 equiv) at room temperature. The resulting solution was stirred for 2 h then diluted with water (10 mL). This mixture was extracted with EA (3 x 10 mL), and the combined organics were washed with brine (2 x 5 mL), dried over anhydrous ISfeSCL and concentrated. The residue was purified by Prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous TFA; to give 3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyrid in-5- yl]°xy]methyl)pyridin-2-amine (130 mg, 73% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 376

Synthesis of 56-d: N-[3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1py ridin-5- yl1oxy1methyl)pyridin-2-yl1-S-(5-fluoro-2-methoxypyridin-3-y l)methanesulfinamide [0454] To a stirred mixture of 3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)pyridin-2-amine (40 mg, 0.11 mmol, 1 equiv) in pyridine (2 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (36 mg, 0.16 mmol, 1.5 equiv) at room temperature. The reaction mixture was stirred for 3 day at 40 °C then concentrated. The residue was purified by Prep-HPLC with following conditions: Column, welch Vltimate XB- C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-80% / 0.1% aqueous formic acid; to afford N- [3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyri din-5-yl]oxy]methyl)pyridin-2- yl]-S-(5-fluoro-2-methoxypyridin-3-yl)methanesulfinamide (24 mg, 40% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 565 Synthesis of Compound 56: N-[3,5-difluoro-4-[([3-methyl-1H-pyrazolol3,4-b1pyridin-5- yl1oxy)methyl1pyridin-2-yl1-5-fluoro-2-methoxypyridine-3-sul fonamide

[0455] A mixture of N-[3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]py ridin-5- yl]°xy]methyl)pyridin-2-yl]-5-fluoro-2-methoxypyridine-3-su lfonamide (22 mg, 39 pmol, 1 equiv) in 4M HC1 in MeOH (1 mL) was stirred for 0.5 h then concentrated. The residue was purified by Prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-60% MeCN / 0.1% aqueous formic acid; to afford N-[3,5- difluoro-4-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)me thyl]pyridin-2-yl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (8.7 mg, 46% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 481

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.12 (s, 1H), 11.44 (s, 1H), 8.46 (d, J= 2.9 Hz, 1H), 8.25 (d, J = 2.7 Hz, 1H), 8.20 (s, 1H), 8.13 (dd, J= 7.5, 3.1 Hz, 1H), 7.90 (d, J= 2.8 Hz, 1H), 5.28 (s, 2H), 3.90 (s, 3H), 2.48 (s, 3H).

Example 68: Synthesis of N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]methyl)amino]phenyl]-5-fluoro-2-methoxypyridine-3-sulfona mide (Compound 57)

Compound 57

Synthesis of 57-a: 3-methyl-l-(oxan-2-yl)pyrazolol3,4-b1pyridine-5-carbaldehyde

[0456] To a solution of 5-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (3 g, 10 mmol, 1 equiv) in THF (60 mL) at -78°C was added 2.5 M n-BuLi in hexanes (5.3 mL, 13 mmol, 1.3 equiv) dropwise, and the resulting solution was stirred for 1 h at -78 °C. DMF (1.5 g, 20 mmol, 2 equiv) was added dropwise at the same temperature, and the mixture stirred to room temperature over 3 h. The reaction was quenched with 4 M HC1 in dioxane to pH 6, and water (100 mL) was added. The resulting solution was extracted with 3 x 50 mL of ethyl acetate. The organics were washed with 50 ml of water and 50 mL of brine, then dried (MgSOp and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 : 1) to afford 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine-5- carbaldehyde (1.7 g, 68% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 246

Synthesis of 57-b: [3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1methanol

[0457] Into a 100 mL 3-necked round-bottom flask in an ice bath was placed 3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridine-5-carbaldehyde (1.5 g, 6.1 mmol, 1 equiv), THF (30 mL), MeOH (6 mL) and NaBH 4 (0.28 g, 7.4 mmol, 1.2 equiv). The resulting solution was stirred for 5 h at 0 °C. The reaction was quenched by 20 mL of MeOH and concentrated. The residue was purified by silica gel column chromatography, eluting with PE: EA (1 : 1) to afford [3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methanol (1 g, 66% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 248

Synthesis of 57-c: tert-butyl N-(tert-butoxycarbonyl)-N-(2,6-difluoro-3- nitrophenyl carbamate

[0458] Into a 100 mL 3 -necked round-bottom flask was placed 2, 6-difluoro-3 -nitroaniline (2 g, 11.5 mmol, 1 equiv), DCM (40 mL), BOC2O (5.5 g, 25 mmol, 2.2 equiv), TEA (3.5 g, 35 mmol, 3.0 equiv) and DMAP (0.28 g, 2.3 mmol, 0.2 equiv). The resulting solution was stirred for 1 h then quenched by the addition of 100 mL of water. This was extracted with 3 x 30 mL of di chloromethane, and the organics were washed with 50 ml of water and 50 mL of brine. The organic solution was dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography, eluting with PE: EA (3:1) to afford tertbutyl N-(tert-butoxycarbonyl)-N-(2,6-difluoro-3-nitrophenyl)carbam ate (3.9 g, 91% yield) as a white solid.

Synthesis of 57-d: tert-butyl N-(2,6-difluoro-3-nitrophenyl)carbamate

[0459] Into a 250 mL 3 -necked round-bottom flask was placed tert-butyl N-(tert- butoxycarbonyl)-N-(2,6-difluoro-3-nitrophenyl)carbamate (4.6 g, 12 mmol, 1 equiv) and DCM (90 mL). This was followed by the addition of TFA (3.7 mL, 49 mmol, 4 equiv) dropwise with stirring at 0 °C. The reaction was stirred for 2 h in an ice bath, then was quenched by the addition of 150 mL of saturated aqueous NaHCO 3 . The resulting solution was extracted with 3 x 100 mL of di chloromethane, and the organics washed with 150 ml of water and 150 mL of brine. After drying over anhydrous sodium sulfate, it was concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (4: 1) to afford tert-butyl N-(2,6-difluoro-3-nitrophenyl)carbamate (2.8 g, 83% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 275

Synthesis of 57-e: tert-butyl N-(2,6-difluoro-3-nitrophenyl)-N-[[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b1pyridin-5-yl1methyl1carbamate

[0460] Into a 100 mL 3-necked round-bottom flask was placed tert-butyl N-(2,6-difluoro-3- nitrophenyl)carbamate (1.06 g, 3.9 mmol, 1.2 equiv), DCM (30 mL), [3 -methyl -l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]methanol (0.8 g, 3.2 mmol, 1 equiv), TMAD (0.84 g, 4.9 mmol, 1.5 equiv) and PPh 3 (1.27 g, 4.9 mmol, 1.5 equiv). The resulting solution was stirred overnight then concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford tert-butyl N-(2,6-difluoro-3-nitrophenyl)-N-[[3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]carbamate (1.5 g, 92% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 504

Synthesis of 57-f: tert-butyl N-(3-amino-2,6-difluorophenyl)-N-i[3-methyl-l-(oxan-2- yl)pyrazolol3,4-b1pyridin-5-yl1methyl1carbamate

[0461] Into a 250 mL round-bottom flask was placed tert-butyl N-(2,6-difluoro-3- nitrophenyl)-N-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridi n-5-yl]methyl]carbamate (1.9 g, 3.8 mmol, 1 equiv) and THF (50 mL). To the resulting solution was added 10% Pd/C (100 mg) and the mixture stirred for 2 h under 2 atm of hydrogen. The solids were removed by filtration and the filtrate concentrated to give tert-butyl N-(3-amino-2,6-difluorophenyl)-N- [[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]c arbamate (1.45 g, 80% yield) as an off-white solid.

LCMS (ES, m/z): [M+H] + : 474

Synthesis of 57-g: tert-butyl N-[2,6-difluoro-3-(5-fluoro-2-methoxypyridine-3- sulfonamido)phenyl1-N-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b 1pyridin-5- yllmethyllcarbamate

[0462] Into a 40 mL vial was placed tert-butyl N-(3-amino-2,6-difluorophenyl)-N-[[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]carba mate (150 mg, 0.32 mmol, 1 equiv), DCM (4 mL), pyridine (75 mg, 0.95 mmol, 3 equiv) and 5-fluoro-2-methoxypyridine- 3-sulfonyl chloride (100 mg, 0.44 mmol, 1.4 equiv). The resulting solution was stirred overnight then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 5-90% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give tert-butyl N-[2,6- difluoro-3-(5-fluoro-2-methoxypyridine-3-sulfonamido)phenyl] -N-[[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]carbamate (100 mg, 48% yield) as an off-white solid. LCMS (ES, m/z): [M+H] + : 663

Synthesis of Compound 57: N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1methyl)amino1phenyl1-5-fluoro-2-methoxypyridine-3-sulfona mide

[0463] Into a 40 mL vial was placed tert-butyl N-[2,6-difluoro-3-(5-fluoro-2- methoxypyridine-3-sulfonamido)phenyl]-N-[[3-methyl-l-(oxan-2 -yl)pyrazolo[3,4-b]pyridin- 5-yl]methyl]carbamate (90 mg, 0.14 mmol, 1 equiv), DCM (3 mL) and TFA (1 mL). The resulting solution was stirred for 3 h then concentrated. The residue was purified by Prep- HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-50% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give N- [2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]m ethyl)amino]phenyl]-5-fluoro- 2-methoxypyridine-3 -sulfonamide (60 mg, 92% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 479

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.12 (s, 1H), 10.01 (s, 1H), 8.35 (dd, J = 5.5, 2.5 Hz, 2H), 7.97 (d, J= 2.0 Hz, 1H), 7.85 (dd, J= 13, 3.0 Hz, 1H), 6.88-6.74 (m, 1H), 6.49-6.37 (m, 1H), 5.97 (s, 1H), 4.42 (s, 2H), 3.87 (s, 3H), 2.45 (s, 3H).

Example 69: Synthesis of 5-chloro-N-[2.,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4- b]Dyridin-5-yl]methyl)amino]Dhenyl]-2-methoxyDyridine-3-sulf onamide (Compound 58)

Compound 58

Synthesis of 58-a: tert-butyl N-(5-chloro-2-methoxypyridin-3-ylsulfonyl)-N-[2,4-difluoro-3 -

(r[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1meth yl1amino)phenyl1carbamate

[0464] Into a 40 mL vial was placed tert-butyl N-(3-amino-2,6-difluorophenyl)-N-[[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]carba mate (150 mg, 0.32 mmol, 1 equiv), DCM (4 mL), pyridine (75 mg, 0.95 mmol, 3 equiv) and 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (107 mg, 0.44 mmol, 1.4 equiv). The resulting solution was stirred overnight then concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 20-90% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give tert-butyl N-(5-chloro-2-methoxypyridin-3-ylsulfonyl)-N-[2,4-difluoro-3 - ([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methyl] amino)phenyl]carbamate (120 mg, 56% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 679

Synthesis of Compound 58: 5-chloro-N-12,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4- b1pyridin-5-yl1methyl)amino1phenyl1-2-methoxypyridine-3-sulf onamide

[0465] Into an 8 mL vial was placed tert-butyl N-(5-chloro-2-methoxypyridin-3-ylsulfonyl)- N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]py ridin-5- yl]methyl]amino)phenyl]carbamate (90 mg, 0.13 mmol, 1 equiv), DCM (3 mL) and TFA (1 mL). The resulting solution was stirred for 3 h then concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-55% MeCN / 0.1% aqueous formic acid;

Detector, 220 nm; to give 5-chloro-N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyr idin- 5-yl]methyl)amino]phenyl]-2-methoxypyridine-3 -sulfonamide (64 mg, 98% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 495

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.11 (s, 1H), 10.04 (s, 1H), 8.37 (dd, J= 2.5, 1.4 Hz, 2H), 7.97 (d, J= 2.0 Hz, 1H), 7.91 (d, J= 2.6 Hz, 1H), 6.88-6.75 (m, 1H), 6.51-6.37 (m, 1H), 5.98 (s, 1H), 4.42 (d, J= 7.1 Hz, 2H), 3.87 (s, 3H), 2.45 (s, 3H).

Example 70: Synthesis of N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]methyl)amino]phenyl]-5-fluoro-2-methylpyridine-3-sulfonam ide (Compound 59)

Compound 59

Synthesis of 59-a: tert-butyl N-[2,6-difluoro-3-(5-fluoro-2-methylpyridine-3- sulfonamido)phenyl1-N-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b 1pyridin-yl1methyl1carbamate [0466] Into a 40 mL vial was placed tert-butyl N-(3-amino-2,6-difluorophenyl)-N-[[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]methyl]carba mate (150 mg, 0.32 mmol, 1 equiv), DCM (4 mL), pyridine (75 mg, 0.95 mmol, 3 equiv) and 5-fluoro-2-methylpyridine- 3-sulfonyl chloride (93 mg, 0.44 mmol, 1.4 equiv). The resulting solution was stirred overnight then concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 20-90% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give tertbutyl N-[2,6-difluoro-3-(5-fluoro-2-methylpyridine-3-sulfonamido)p henyl]-N-[[3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-yl]methyl]carbamate (75 mg, 37% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 647

Synthesis of Compound 59: N-[2,4-difluoro-3-[([3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1methyl)amino1phenyl1-5-fluoro-2-methylpyridine-3-sulfonam ide

[0467] Into a 40 mL vial was placed tert-butyl N-[2,6-difluoro-3-(5-fluoro-2-methylpyridine- 3-sulfonamido)phenyl]-N-[[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin-5- yl]methyl]carbamate (75 mg, 0.12 mmol, 1 equiv), DCM (3 mL) and TFA (1 mL). The resulting solution was stirred for 3 h, then concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-55% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to give N-[2,4-difluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5- yl]methyl)amino]phenyl]-5-fluoro-2-methylpyridine-3-sulfonam ide (40 mg, 75% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 463

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.11 (s, 1H), 10.04 (s, 1H), 8.37 (dd, J= 2.5, 1.4 Hz, 2H), 7.97 (d, J= 2.0 Hz, 1H), 7.91 (d, J= 2.6 Hz, 1H), 6.88-6.75 (m, 1H), 6.51-6.37 (m, 1H), 5.98 (s, 1H), 4.42 (d, J= 7.1 Hz, 2H), 3.87 (s, 3H), 2.45 (s, 3H).

Example 71: Synthesis of 5-chloro-/V-(2.,4-difluoro-3-((imidazo[l.,5-b]pyridazin-3- yloxy)methyl)phenyl)-2-methoxypyridine-3-sulfonamide (Compound 60)

Synthesis of 60-a: ethyl 5-methoxypyridazine-3-carboxylate

[0468] To a solution of 3-chloro-5-methoxypyridazine (16 g, 110 mmol, 1 equiv) in EtOH (190 mL) was added Pd(PPh 3 ) 2 Cl 2 (7.8 g, 11 mmol, 0.1 equiv) and TEA (16.8 g, 166 mmol, 1.5 equiv) in a pressure tank. The mixture was purged with nitrogen, then pressurized to 40 bar with carbon monoxide. The reaction was stirred at 120 °C overnight, then cooled and filtered. The filtrate was concentrated and the residue purified by silica gel column chromatography, eluting with PE:EA (1 : 1) to afford ethyl 5-methoxypyridazine-3-carboxylate (8 g, 40% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 183

Synthesis of 60-b: 5-methoxypyridazine-3 -carboxamide

[0469] Ethyl 5-methoxypyridazine-3-carboxylate (9 g, 49 mmol, 1 equiv) was dissolved in 7 M NHs/MeOH (45 mL) and stirred at room temperature for 2 h. The reaction solution was concentrated under vacuum directly and the residue was triturated with DCM (50 mL). The precipitated solids were collected by filtration, washed with DCM (50 mL) and dried to afford 5-methoxypyridazine-3-carboxamide (6 g, 79% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 154

Synthesis of 60-c: 5-methoxypyridazine-3 -carbonitrile

[0470] To a stirred solution of 5-methoxypyridazine-3-carboxamide (6 g, 39 mmol, 1 equiv) in THF (120 mL) was added TFAA (16 g, 78 mmol, 2 equiv) and TEA (10 g, 98 mmol, 2.5 equiv) dropwise at 0 °C. The reaction was stirred at room temperature for 3 h, then concentrated. The residue was diluted with H 2 O (10 mL) and saturated NaHCO 3 (15 mL), then extracted with EA (3 x 20 mL). The combined organics were washed with brine (3 x 10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford 5-methoxypyridazine-3- carbonitrile (5 g, 94% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 136

Synthesis of 60-d: l-(5-methoxypyridazin-3-yl)methanamine

[0471] To a solution of 5-methoxypyridazine-3-carbonitrile (5.4 g, 40 mmol, 1 equiv) in MeOH (500 mL) was added Pd/C (10%, 0.54 g) and 6 N HC1 (5 mL) in a pressure tank. The mixture was hydrogenated at room temperature under 10 bar of hydrogen pressure for 3 h. After completion, the reaction solution was filtered through a Celite pad and concentrated under reduced pressure to afford l-(5-methoxypyridazin-3-yl)methanamine (5 g, crude) as a red solid, which was used for next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 140

Synthesis of 60-e: 3-methoxyimidazo[L5-b1pyridazine

[0472] l-(5-Methoxypyridazin-3-yl)methanamine (4 g, 28.7 mmol, 1 equiv), (diethoxymethoxy)ethane (30 mL) and AcOH (7.5 mL) were stirred at 120 °C for 2 h in a s 6 aled tube. The reaction solution was cooled and concentrated, and the residue diluted with H 2 O (10 mL). The mixture was basified to pH 8 with saturated NaHCO 3 aqueous, then extracted with EA (3 x 10 mL). The combined organics were washed with brine (2 x 5 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 :2) to afford 3- methoxyimidazo[l,5-b]pyridazine (1 g, 23% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 150

Synthesis of 60-f: imidazo1,5-b1pyridazin-3-ol

[0473] To a stirred solution of 3-methoxyimidazo[l,5-b]pyridazine (1.1 g, 7.4 mmol, 1 equiv) in DCM (14 mL) was added IM BBr 3 (44 mL, 44 mmol, 6 equiv) dropwise at 0 °C, and the mixture stirred at 30 °C for 2 days. Stirring was stopped and the mixture allowed to stand for 30 mins. The supernatant liquid was removed by decanting, and the precipitate diluted with DCM (20 mL). MeOH (10 mL) was added dropwise at 0 °C until all the precipitate was dissolved. The resulting solution was concentrated and the residue triturated with MTBE (10 mL). The precipitated solids were collected by filtration, washed with MTBE (1 x 10 mL) and dried to give imidazo[l,5-b]pyridazin-3-ol (300 mg, 30% yield) as a grey solid.

LCMS (ES, m/z): [M+H] + : 136

Synthesis of 60-g: 2,4-difluoro-3-((imidazo[1,5-b]pyridazin-3-yloxy)methyl)anil ine [0474] To a stirred solution of imidazo[1,5-b]pyridazin-3-ol (180 mg, 1.3 mmol, 1 equiv) in DCM (18 mL) was added (3-amino-2,6-difluorophenyl)methanol (233 mg, 1.4 mmol, 1.1 equiv) and PPh 3 (384 mg, 1.4 mmol, 1.1 equiv). The solution was cooled to 0 °C and TMAD (252 mg, 1.4 mmol, 1.1 equiv) in DCM (5 mL) was added dropwise. The reaction was stirred at room temperature overnight. The resulting mixture was diluted with H 2 O (5 mL) and extracted with DCM (3 x 5 mL). The combined organics were washed with brine (2 x 5 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 : 1) to afford 2,4- difluoro-3-([imidazo[l,5-b]pyridazin-3-yloxy]methyl)aniline (160 mg, 44% yield) as a red solid.

LCMS (ES, m/z): [M+H] + : 277

Synthesis of Compound 60: 5-chloro-A-(2,4-difluoro-3-((imidazo[L5-b1pyridazin-3- yloxy)methyl)phenyl)-2-methoxypyridine-3-sulfonamide

[0475] To a stirred solution of 2,4-difluoro-3-([imidazo[l,5-b]pyridazin-3- yloxy]methyl)aniline (150 mg, 0.54 mmol, 1 equiv) in DCM (3 mL) was added 5-chloro-2- methoxypyridine-3 -sulfonyl chloride (144 mg, 0.6 mmol, 1.1 equiv) and pyridine (129 mg, 1.6 mmol, 3 equiv). The reaction was stirred at room temperature overnight. The resulting solution was concentrated and the residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 35-85% MeCN / 0.1% aqueous formic acid; Detector, 220 nm; to afford 5-chloro-N-[2,4-difluoro-3- ([imidazo[l,5-b]pyridazin-3-yloxy]methyl)phenyl]-2-methoxypy ridine-3-sulfonamide (50 mg, 19% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 482

1 H NMR (300 MHz, DMSO-d 6 ) δ 8.51 (s, 1H), 8.44 (d, J= 2.6 Hz, 1H), 8.11 (d, J= 2.8 Hz, 1H), 8.03 (d, J= 2.6 Hz, 1H), 7.58 (d, J= 2.9 Hz, 1H), 7.38 (td, J= 9.0, 6.0 Hz, 1H), 7.24 (s, 1H), 7.13 (t, J= 9.0 Hz, 1H), 5.11 (s, 2H), 3.90 (s, 3H).

Example 72: Synthesis of N-[2-chloro-4-fluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n- 5-yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfona mide (Compound 61)

Compound 61

Synthesis of 61-a: 2-chloro-6-fluoro-3 -nitrobenzoic acid

[0476] To a stirred solution of 2-chloro-6-fluorobenzoic acid (10 g, 57 mmol, 1 equiv) in H 2 SO4 (200 mL) was added HNO3 (4 g, 63 mmol, 1.1 equiv, 98%) dropwise at 0 °C. The resulting mixture was stirred for 2 h at low temperature, then poured into ice-water (I L) and extracted with EA (3 x 200 mL). The combined organics were washed with brine (3 x 100 mL), dried over anhydrous ISfeSCL and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with EA:MeOH (10: 1) to afford 2- chloro-6-fluoro-3 -nitrobenzoic acid (8.2 g, 65% yield ) as a light yellow solid.

Synthesis of 61-b: (2-chloro-6-fluoro-3-nitrophenyl)methanol

[0477] A solution of 2-chloro-6-fluoro-3-nitrobenzoic acid (5 g) in BH3-THF (100 mL, 1 mol/L in THF) was stirred for 12 h at 50 °C. The reaction was quenched by the addition of MeOH (100 mL) dropwise at room temperature. The resulting solution was concentrated under reduced pressure and the residue purified by silica gel column chromatography, eluting with PE:EA (4: 1) to afford (2-chloro-6-fluoro-3-nitrophenyl)methanol (3.7 g, 79% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 206

Synthesis of 61-c: (3-amino-2-chloro-6-fluorophenyl)methanol

[0478] To a solution of (2-chloro-6-fluoro-3-nitrophenyl)methanol (1.6 g, 7.7 mmol, 1 equiv) in EA (30 mL) was added Pd/C (10%, 80 mg) under nitrogen atmosphere. The mixture was hydrogenated at 4 atm for 4 h under hydrogen atmosphere, then filtered through a Celite pad and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford (3-amino-2-chloro-6- fluorophenyl)m ethanol (300 mg, 22% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 176

Synthesis of 61-d: 2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1 pyridin-5- ylloxylmethyDaniline

[0479] To a solution of (3-amino-2-chloro-6-fluorophenyl)methanol (500 mg, 2.8 mmol, 1 equiv) in DCM (10 mL) were added 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (664 mg, 2.8 mmol, 1 equiv), PPI13 (1.1 g, 4.2 mmol, 1.5 equiv) and TMAD (735. mg, 4.2 mmol, 1.5 equiv) at room temperature. The resulting solution was stirred at room temperature for 2 h, then water (300 mL) was added. The mixture was extracted with ethyl acetate (3 x 100 mL), and the combined organics washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The crude product was purified by column chromatography over silica gel (eluent: PE:EA =3: 1) to afford 2-chloro-4-fluoro-3-([[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)a niline (1.1 g, 98% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 391 Synthesis of 61-e: N-[2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b1pyridin- 5-yl1oxy1methyl)phenyl1-5-fluoro-2-methoxypyridine-3-sulfona mide

[0480] To a stirred solution of 2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (150 mg, 0.38 mmol, 1 equiv) and pyridine (91 mg, 1.1 mmol, 3 equiv) in DCM (5 mL) were added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (103 mg, 0.46 mmol, 1.2 equiv) in portions at room temperature. The reaction mixture was stirred for 2 h then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford N-[2-chloro-4-fluoro-3-([[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)p henyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (200 mg, 90% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 566

Synthesis of Compound 61 : N-12-chloro-4-fluoro-3-[(13-methyl-1H -pyrazolo[3,4-b1pyridin- 5-yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfona mide

[0481] A mixture of N-[2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 - b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (200 mg, 1 equiv) and 4M HC1 in MeOH (10 mL) was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-65% MeCN 0.1% aqueous formic acid; Detector, 220 nm; to afford N-[2-chloro-4-fluoro- 3-[([3 -methyl- lH-pyrazolo[3, 4-b]pyri din-5-yl]oxy )methyl]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (130 mg, 76% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 496

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.35 (s, 1H), 8.46 (d, J= 3.0 Hz, 1H), 8.19 (d, J= 2.7 Hz, 1H), 7.97 (dd, J= 13, 3.0 Hz, 1H), 7.89 (d, J= 2.7 Hz, 1H), 7.44 (dd, J= 9.0, 5.7 Hz, 1H), 7.33 (t, J= 8.9 Hz, 1H), 5.19 (d, J= 1.9 Hz, 2H), 3.86 (s, 3H), 2.48 (s, 3H).

Example 73: Synthesis of N-[2-cvano-4-fluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridin -

5-yl]

Synthesis of 62-a: 6-bromo-3-fluoro-2-methylbenzonitrile

[0482] To a stirred solution of 2-bromo-5-fluorobenzonitrile (1 g, 5 mmol, 1 equiv) in THF (10 mL) was added LDA (3.8 mL, 7.5 mmol, 1.5 equiv, 2 M solution in THF) dropwise at - 78 °C under nitrogen atmosphere. The resulting mixture was stirred for 1 h at low temperature, then CH 3 I (1.42 g, 10 mmol, 2 equiv) was added dropwise over 10 min at - 78 °C. The resulting mixture was stirred for additional 1 h at -78 °C, before being quenched with 1 M hydrochloric acid (10 mL). The mixture was extracted with EA (3 x 20 mL). The combined organics were washed with brine (1 x 20 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-70% MeCN / 0.1% aqueous formic acid to afford 6-bromo-3-fluoro-2- methylbenzonitrile (408 mg, 38% yield) as a brown solid.

Synthesis of 62-b: 6-bromo-2-(bromomethyl)-3-fluorobenzonitrile

[0483] To a stirred solution of 6-bromo-3-fluoro-2 -methylbenzonitrile (380 mg, 1.8 mmol, 1 equiv) and BPO (46 mg, 0.18 mmol, 0.1 equiv) in CCl 4 (10 mL) was added NBS (474 mg, 2.7 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for 6 h at 80 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase:30-70% MeCN / 0.1% aqueous formic acid; to afford 6-bromo-2-(bromomethyl)-3-fluorobenzonitrile (362 mg, 70% yield) as a light yellow solid.

Synthesis of 62-c: 6-bromo-3-fluoro-2-(((3-methyl-l-(tetrahydro-2H-pyran-2-yl)- 1H- pyrazolo[3,4-b1pyri din-5 -yl)oxy)methyl)benzonitrile

[0484] To a stirred solution of 6-bromo-2-(bromomethyl)-3-fluorobenzonitrile (340 mg, 1.16 mmol, 1 equiv) and 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (271 mg, 1.16 mmol, 1 equiv) in MeCN (10 mL) was added K 2 CO 3 (481 mg, 3.48 mmol, 3 equiv) in portions at room temperature. The mixture was stirred for 5 h at 50 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 6-bromo-3-fluoro-2-([[3-methyl- l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)benzonit rile (380 mg, 74% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 445

Synthesis of 62-d: N-(2-cyano-4-fluoro-3-(((3-methyl-l-(tetrahydro-2H-pyran-2-y l)-1H- pyrazolo[3,4-b1pyridin-5-yl)oxy)methyl)phenyl)-5-fluoro-2-me thoxypyridine-3-sulfonamide [0485] To a stirred solution of 6-bromo-3-fluoro-2-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)benzonitrile (350 mg, 0.79 mmol, 1 equiv) and 5-fluoro-2- methoxypyridine-3 -sulfonamide (178 mg, 0.86 mmol, 1.1 equiv) in dioxane (10 mL) were added BrettPhos Pd G3 (214 mg, 0.24 mmol, 0.3 equiv), t-BuONa (151 mg, 1.6 mmol, 2 equiv) and BrettPhos (127 mg, 0.24 mmol, 0.3 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 4 h at 80 °C then concentrated under reduced pressure to give N-[2-cyano-4-fluoro-3-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (500 mg, crude) as a brown solid which was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 571 Synthesis of Compound 62: N-[2-cyano-4-fluoro-3-[([3-methyl-1H-pyrazolo[3,4-b1pyridin- 5- yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0486] A solution of N-[2-cyano-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (500 mg, 0.88 mmol, 1 equiv) in TFA (1 mL) and DCM (3 mL) was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in EA (5 mL) and water (10 mL) added. The mixture was basified to pH 8 with saturated aqueous NaHCO 3 then extracted with EA (3 x 5 mL). The combined organics were washed with brine (1 x 10 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 50-100% MeCN / 0.1% aqueous formic acid to afford N-[2-cyano-4-fluoro-3-[([3-methyl-lH-pyrazolo[3,4-b]pyridin- 5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (130 mg, 31% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 487

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.13 (s, 1H), 10.89 (s, 1H), 8.50 (d, J= 2.9 Hz, 1H), 8.24 (d, J = 2.8 Hz, 1H), 8.01 (dd, J= 7.3, 3.0 Hz, 1H), 7.93 (d, J= 2.8 Hz, 1H), 7.67 (t, J = 9.1 Hz, 1H), 7.41 (dd, J= 9.1, 4.8 Hz, 1H), 5.28-5.22 (m, 2H), 3.87 (s, 3H), 2.49 (s, 3H).

Example 74: Synthesis of 5-fluoro-N-[3-fluoro-2-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n- 5-yl]oxy)methyl]pyridin-4-yl]-2-methoxypyridine-3-sulfonamid e (Compound 63)

Compound 63

Synthesis of 63-a: 5-fluoro-N-13-fluoro-2-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b1pyridin-5- yl1oxy1methyl)pyridin-4-yl1-2-methoxypyridine-3-sulfonamide

[0487] Into a 40 mL vial was placed 4-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (500 mg, 1.3 mmol, 1 equiv), dioxane (15 mL), 5-fluoro-2-methoxypyridine-3-sulfonamide (274 mg, 1.3 mmol, 1 equiv), Z-BuONa (255 mg, 2.6 mmol, 2 equiv) and BrettPhos Pd G3 (481 mg, 0.5 mmol, 0.4 equiv) at room temperature under nitrogen atmosphere. The reaction was stirred for 2 h at 80 °C then concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 20-70% MeCN / 0.1% aqueous formic acid; to afford 5-fluoro -N-[3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-4-yl]-2-me thoxypyridine-3-sulfonamide (400 mg, 55% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 547

Synthesis of Compound 63: 5-fluoro-N-[3-fluoro-2-[([3-methyl-1H-pyrazolo[3,4-b]pyridin - 5-yl]oxy)methyl]pyridin-4-yl]-2-methoxypyridine-3-sulfonamid e

[0488] Into a 40 mL vial was placed 5-fluoro -N-[3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-4-yl]-2-me thoxypyridine-3-sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 4 M HC1 solution in MeOH (5 mL) at room temperature. The resulting solution was stirred for 1 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-45% MeCN / 0.1% aqueous formic acid; to afford 5-fluoro-N-[3- fluoro-2-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)meth yl]pyridin-4-yl]-2- methoxypyridine-3 -sulfonamide (50 mg, 59%) as a white solid.

LCMS (ES, m/z): [M+H] + : 463 1 H NMR (300 MHz, DMSO-d 6 ) δ 13.12 (s, 1H), 8.39 (s, 1H), 8.31 (s, 1H), 8.13 (s, 2H), 7.91 (s, 1H), 7.56 (s, 1H), 5.32 (s, 2H), 3.81 (s, 3H), 2.47 (s, 3H).

Example 75: Synthesis of 5-chloro-N-[3-fluoro-2-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n-

5-yl]oxy)methyl]pyridin-4-yl]-2-methoxypyridine-3-sulfona mide (Compound 64)

Compound 64

Synthesis of 64-a: (4-chloro-3-fluoropyridin-2-yl)methanol

[0489] To a stirred solution of 4-chloro-3-fluoropyridine-2-carbaldehyde (10 g, 63 mmol, 1 equiv) in THF (150 mL) and MeOH (50 mL) was added NaBH 4 (6 g, 156 mmol, 2.5 equiv) in portions at 0 °C. The resulting solution was stirred for 12 h at room temperature, then was quenched with water (10 mL) and extracted with ethyl acetate (4 x 50 mL). The combined organics were washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (5: 1) to afford (4-chloro-3-fluoropyridin-2-yl)methanol (8 g, 79% yield) as light yellow oil.

LCMS (ES, m/z): [M+H] + : 162

Synthesis of 64-b: 4-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2-yl)pyrazolol3,4-b1 pyridin-5- ylloxylmethyl pyridine

[0490] Into a 40 mL vial was placed (4-chloro-3-fluoropyridin-2-yl)methanol (100 mg, 0.6 mmol, 1 equiv), DCM (6 mL), 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (144 mg, 0.6 mmol, 1 equiv), PPh3 (195 mg, 0.7 mmol, 1.2 equiv), and TMAD (128 mg, 0.7 mmol, 1.2 equiv) at room temperature. The reaction was stirred for 2 h, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 4-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b] pyridin-5- yl]°xy]methyl)pyridine (180 mg, 77% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 377

Synthesis of 64-c: 5-chloro-N-[3-fluoro-2-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b1pyridin- 5-yl1oxy1methyl)pyridin-4-yl1-2-methoxypyridine-3-sulfonamid e

[0491] Into a 40 mL vial was placed 4-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (500 mg, 1.3 mmol, 1 equiv), toluene (5 mL), 5-chloro-2-methoxypyridine-3-sulfonamide (591 mg, 2.6 mmol, 2 equiv), CS2CO3 (865 mg, 2.6 mmol, 2 equiv) and BrettPhos Pd G3 (481 mg, 0.5 mmol, 0.4 equiv). The resulting solution was stirred for 12 h at 100 °C under nitrogen atmosphere. The reaction mixture was concentrated and the residue purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[3-fluoro-2-([[3 -methyl- l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-4-yl]-2-me thoxypyridine-3-sulfonamide (300 mg, 40% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 563

Synthesis of Compound 64: 5-chloro-N-[3-fluoro-2-[([3-methyl-1H-pyrazolo[3,4-b1pyridin - 5-yl1oxy)methyl1pyridin-4-yl1-2-methoxypyridine-3-sulfonamid e

[0492] Into a 40 mL vial was placed 5-chloro-N-[3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-4-yl]-2-me thoxypyridine-3-sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 4 M HC1 solution in MeOH (5 mL) at room temperature. The resulting solution was stirred for 1 h, then was concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 10-45% MeCN / 0.1 aqueous formic acid; to afford 5-chloro-N-[3- fluoro-2-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)meth yl]pyridin-4-yl]-2- methoxypyridine-3 -sulfonamide (50 mg, 59% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 479 1 H NMR (300 MHz, DMSO-d6) δ 13.13 (s, 1H), 8.43 (s, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 8.14 (s, 1H), 7.91 (s, 1H), 7.57 (s, 1H), 5.33 (s, 2H), 3.82 (s, 3H), 2.47 (s, 3H) Example 76: Synthesis of 5-chloro-N-[3.,5-difluoro-4-[([3-methyl-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy)methyl]pyridin-2-yl]-2-methoxypyridine-3- sulfonamide (Compound 65)

Compound 65

Synthesis of 65-a: 5-chloro-N-[3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo [3,4- b1pyridin-5-yl1oxy1methyl)pyridin-2-yl1-2-methoxypyridine-3- sulfonamide

[0493] Into a 40 mL vial was placed 3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)pyridin-2-amine (200 mg, 0.5 mmol, 1 equiv), pyridine (8 mL) and 5-chloro-2-methoxypyridine-3-sulfonyl chloride (387 mg, 1.5 mmol, 3 equiv). The resulting solution was stirred for 24 h at 50 °C then diluted with H 2 O (50 mL) and extracted with ethyl acetate (3 x 20 mL). The combined organics were washed with brine (2 x 20 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by Flash- Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 5-chloro- N-[3,5-difluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]py ridin-5-yl]oxy]methyl)pyridin- 2-yl]-2-methoxypyridine-3-sulfonamide (157 mg, 51% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 581

Synthesis of Compound 65: 5-chloro-N-[3,5-difluoro-4-[([3-methyl-1H-pyrazolo[3,4- b1pyridin-5-yl1oxy)methyl1pyridin-2-yl1-2-methoxypyridine-3- sulfonamide

[0494] Into a 40 mL vial was placed 5-chloro-N-[3,5-difluoro-4-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-2-yl]-2-me thoxypyridine-3-sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 4 M HC1 solution in MeOH (5 mL). The resulting solution was stirred for 30 min then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 gm; Mobile Phase: 30-60% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[3,5-difluoro-4-[([3- methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]pyridin-2-y l]-2-methoxypyridine-3- sulfonamide (50 mg, 58% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 497

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.13 (s, 1H), 11.46 (s, 1H), 8.52 (d, J= 2.6 Hz, 1H), 8.29- 8.18 (m, 3H), 7.91 (d, J= 2.8 Hz, 1H), 5.29 (s, 2H), 3.92 (s, 3H), 2.48 (s, 3H).

Example 77: Synthesis of N-[3-chloro-5-fluoro-4-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n- 5-yl]oxy)methyl]pyridin-2-yl]-5-fluoro-2-methoxypyridine-3-s ulfonamide (Compound 66)

Synthesis of 66-a: 2,3-dichloro-5-fluoropyridine-4-carbaldehyde

[0495] To a stirred solution of 2,3-dichloro-5-fluoropyridine (1 g, 6 mmol, 1 equiv) in THF (20 mL) was added 2 M LDA in THF (4.5 mL, 9 mmol, 1.5 equiv) dropwise at -78°C. The resulting solution was stirred for 1 h at -78 °C, then DMF (881 mg, 12 mmol, 2 equiv) was added dropwise at -78 °C and the reaction stirred for 1 h at this temperature. The reaction was quenched with IM aq. HC1 (10 mL) at -5 °C and extracted with EA (3 x 50 mL). The combined organics were washed with brine (2 x 20 mL), dried over anhydrous sodium sulfate and concentrated to afford 2,3-dichloro-5-fluoropyridine-4-carbaldehyde (1 g, crude) as a yellow oil which was used in the next step directly without further purification. Synthesis of 66-b: (2,3-dichloro-5-fluoropyridin-4-yl)methanol

[0496] To a stirred solution of 2,3-dichloro-5-fluoropyridine-4-carbaldehyde (1 g, 5 mmol, 1 equiv) in MeOH (20 mL) was added NaBH 4 (0.6 g, 15 mmol, 3 equiv) in portions at 0 °C. The reaction was stirred for 30 min at room temperature, then was quenched with saturated aqueous NH4CI (50 mL). The resulting mixture was extracted with EA (3 x 100 mL), and the combined organic layers were washed with brine (3 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford (2,3-dichloro-5-fluoropyridin-4-yl)methanol (300 mg, 30% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 196

Synthesis of 66-c: 2,3-dichloro-5-fluoro-4-([[3-rnethyl-l-(oxan-2-yl)pyrazolol3 ,4-b1pyridin- 5-yl1oxy1methyl)pyridine

[0497] To a stirred solution of (2,3-dichloro-5-fluoropyridin-4-yl)methanol (300 mg, 1.5 mmol, 1 equiv) in DCM (10 mL) were added 3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-ol (393 mg, 1.7 mmol, 1.1 equiv), PPh 3 (602 mg, 2.3 mmol, 1.5 equiv), and TMAD (395 mg, 2.3 mmol, 1.5 equiv). The resulting mixture was stirred for 1 h, then concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 2,3-dichloro-5-fluoro-4-([[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)p yridine (490 mg, 78% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 411

Synthesis of 66-d: N-[3-chloro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b1pyridin- 5-yl1oxy1methyl)pyridin-2-yl1-5-fluoro-2-methoxypyridine-3-s ulfonamide

[0498] Into a 40 mL vial was placed 2,3-dichloro-5-fluoro-4-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (800 mg, 2 mmol, 1 equiv), dioxane (9 mL), 5-fluoro-2-methoxypyridine-3-sulfonamide (602 mg, 3 mmol, 1.5 equiv), CS2CO3 (1.3 g, 4 mmol, 2 equiv) and BrettPhos Pd G3 (705 mg, 0.8 mmol, 0.4 equiv) at room temperature under nitrogen atmosphere. The reaction was stirred for 12 h at 80 °C then diluted with H 2 O (20 mL) and extracted with ethyl acetate (3 x 10 mL). The combined organics were washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford N-[3-chloro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin-5- yl]oxy]methyl)pyridin-2-yl]-5-fluoro-2-methoxypyridine-3-sul fonamide (100 mg, 9% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 581

Synthesis of Compound 66: N-[3-chloro-5-fluoro-4-[([3-methyl-1H-pyrazolo[3,4-b1pyridin - 5-yl]oxy)methyl]pyridin-2-yl]-5-fluoro-2-methoxypyridine-3-s ulfonamide

[0499] Into a 40 mL vial was placed N-[3-chloro-5-fluoro-4-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-2-yl]-5-fl uoro-2-methoxypyridine-3- sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 4 M HC1 in MeOH (5 mL). The resulting solution was stirred for 30 min at room temperature, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous formic acid; to afford N-[3- chloro-5-fluoro-4-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl ]oxy)methyl]pyridin-2-yl]-5- fluoro-2-methoxypyridine-3-sulfonamide (50 mg, 58% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 497

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.13 (s, 1H), 11.17 (s, 1H), 8.48 (d, J= 2.9 Hz, 1H), 8.35- 8.23 (m, 2H), 8.12 (dd, J= 7.5, 3.0 Hz, 1H), 7.93 (d, J= 2.7 Hz, 1H), 5.28 (d, J= 1.7 Hz, 2H), 3.93 (s, 3H), 2.49 (s, 3H).

Example 78: Synthesis of 5-fluoro-N-[5-fluoro-6-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n-

5-yl]oxy)methyl]pyridine-2-yl]-2-methoxypyridine-3-sulfon amide (Compound 67)

Compound 67

Synthesis of 67-a: 5-fluoro-2-methoxypyridine-3-sulfonamide

[0500] A mixture of 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (2.2 g, 9.8 mmol, 1 equiv) and 7 M NH 3 solution in MeOH (30 mL) was stirred for 5 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (2:1) to give 5-fluoro-2-methoxypyridine-3- sulfonamide (1.1 g, 55% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 207

Synthesis of 67-b: 2-(bromomethyl)-6 -chloro -3 -fluoropyridine

[0501] To a stirred solution of 6-chloro-3-fluoro-2-methylpyridine (5 g, 34 mmol, 1 equiv) and NBS (7.4 g, 41 mmol, 1.2 equiv) in CCI4 (50 mL) was added AIBN (1 g, 6.9 mmol, 0.2 equiv) in portions at room temperature. The reaction was stirred overnight at 90 °C, then cooled and diluted with water (300 mL). The resulting solution was extracted with EA (3 x 100 mL), and the combined organics were washed with brine (50 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (20: 1) to afford 2-(brom omethyl)-6-chl oro-3 - fluoropyridine (3.6 g, 47% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 224

Synthesis of 67-c: 6-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b] pyridin-5- yl]oxy]methyl)pyridine

[0502] To a stirred solution of 2-(bromomethyl)-6-chl oro-3 -fluoropyridine (100 mg, 0.5 mmol, 1 equiv) in MeCN (5 mL) were added 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin- 5-ol (114 mg, 0.5 mmol, 1 equiv) and K2CO3 (123 mg, 0.9 mmol, 1.2 equiv). The reaction was stirred overnight then diluted with water (20 mL) and extracted with EA (3 x 20 mL). The combined organics were washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 :1) to afford 6-chloro-3-fhioro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (100 mg, 60% yield) as a yellow solid. LCMS (ES, m/z): [M+H] + : 377

Synthesis of 67-d: 5-fluoro-N-[5-fluoro-6-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin-5- yl]oxy]methyl)pyridin-2-yl]-2-methoxypyridine-3-sulfonamide

[0503] Into a 40 mL vial was placed 6-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (500 mg, 1.3 mmol, 1 equiv), toluene (15 mL), 5-fluoro-2-methoxypyridine-3-sulfonamide (411 mg, 2 mmol, 1.5 equiv), CS2CO3 (865 mg, 2.7 mmol, 2 equiv) and BrettPhos Pd G3 (482 mg, 0.5 mmol, 0.4 equiv). The reaction was stirred for 12 h at 100 °C under nitrogen atmosphere, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 5-fluoro-N-[5-fluoro-6-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin-5- yl]°xy]methyl)pyridin-2-yl]-2-methoxypyridine-3 -sulfonamide (180 mg, 25% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 547

Synthesis of Compound 67: 5-fluoro-N-[5-fluoro-6-[([3-methyl-1H-pyrazolo[3,4-b1pyridin - 5-yl1oxy)methyl1pyridine-2-yl1-2-methoxypyridine-3-sulfonami de

[0504] Into a 40 mL vial was placed 5-fluoro-N-[5-fluoro-6-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-2-yl]-2-me thoxypyridine-3-sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 4 M HC1 solution in MeOH (5 mL). The resulting solution was stirred for 30 min then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-55% MeCN / 0.1% aqueous formic acid; to afford 5-fluoro-N-[5-fluoro-6-[([3-methyl- I H- pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]pyridine-2-yl]-2-meth oxypyridine-3-sulfonamide (50 mg, 59% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 463

^ NMR (300 MHz, DMSO-d 6 ) δ 13.07 (s, 1H), 11.44 (s, 1H), 8.37 (d, J= 3.0 Hz, 1H), 8.16 (dt, J= 5.2, 2.8 Hz, 2H), 7.80-7.68 (m, 2H), 7.10 (dd, J= 8.9, 3.2 Hz, 1H), 5.06 (d, J= 2.1 Hz, 2H), 3.84 (s, 3H), 2.46 (s, 3H).

Example 79: Synthesis of 5-chloro-N-[5-fluoro-6-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n- 5-yl]oxy)methyl]pyridin-2-yl]-2-methoxypyridine-3-sulfonamid e (Compound 68)

Compound 68

Synthesis of 68-a: 5-chloro-2-methoxypyridine-3 -sulfonamide

[0505] A solution of 5-chloro-2-methoxypyridine-3-sulfonyl chloride

(2 g, 8.3 mmol, 1 equiv) and 7 M NH 3 solution in MeOH (40 mL) was stirred for 5 h at room temperature. The reaction mixture was concentrated and the residue purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford 5-chloro-2-methoxypyridine-3- sulfonamide (1.7 g, 92% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 223 Synthesis of 68-b: 5-chloro-N-[5-fluoro-6-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b1pyridin- 5-yl1oxy1methyl)pyridin-2-yl1-2-methoxypyridine-3-sulfonamid e

[0506] Into a 40 mL vial was placed 6-chloro-3-fluoro-2-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (800 mg, 2 mmol, 1 equiv), toluene (24 mL), 5-chloro-2-methoxypyridine-3-sulfonamide (709 mg, 3 mmol, 1.5 equiv), CS2CO3 (1.4 mg, 4 mmol, 2 equiv) and BrettPhos Pd G3 (770 mg, 0.9 mmol, 0.4 equiv). The reaction was stirred for 12 h at 100 °C under nitrogen atmosphere, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[5-fhioro-6-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin-5- yl]°xy]methyl)pyridin-2-yl]-2-methoxypyridine-3 -sulfonamide (560 mg, 47% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 563

Synthesis of Compound 68: 5-chloro-N-[5-fluoro-6-[([3-methyl-1H-pyrazolo[3,4-b1pyridin - 5-yl1oxy)methyl1pyridin-2-yl1-2-methoxypyridine-3-sulfonamid e

[0507] Into a 40 mL round-bottom flask was placed 5-chloro-N-[5-fluoro-6-([[3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridin-2- yl]-2-methoxypyridine-3- sulfonamide (100 mg, 0.2 mmol, 1 equiv) and 4 M HC1 solution in MeOH (5 mL). The resulting solution was stirred for 30 min then concentrated. The residue was purified by prep- HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[5- fluoro-6-[([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)meth yl]pyridin-2-yl]-2- methoxypyridine-3 -sulfonamide (50 mg, 59% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 479

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.07 (s, 1H), 11.46 (s, 1H), 8.40 (d, J= 2.6 Hz, 1H), 8.26 (d, J= 2.7 Hz, 1H), 8.18 (d, J= 2.7 Hz, 1H), 7.80-7.68 (m, 2H), 7.08 (dd, J= 9.0, 3.2 Hz, 1H), 5.04 (d, J= 2.0 Hz, 2H), 3.85 (s, 3H), 2.46 (s, 3H). Example 80: Synthesis of N-[3-[([3-ethynyl-1H-pyrazolo[3.,4-b]pyridin-5-yl]oxy)methyl ]-

2,4-difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonam ide (Compound 69) Compound 69

Synthesis of 69-a: 5-bromo-l-[[2-(trimethylsilyl)ethoxy1methyl1-3-12- (trimethylsilyl)ethynyl1pyrazolol3,4-b1pyridine

[0508] To a stirred solution of 5-bromo-3-iodo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (800 mg, 1.7 mmol, 1 equiv) and trimethylsilylacetylene (190 mg, 1.9 mmol, 1.1 equiv) in DMF (10 mL) were added TEA (534 mg, 5.2 mmol, 3 equiv), Cui (33 mg, 0.17 mmol, 0.1 equiv) and Pd(PPh 3 ) 2 Cl 2 (123 mg, 0.17 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. The reaction was stirred for 2 h at 60 °C under nitrogen atmosphere, then cooled and quenched with water (50 mL). The resulting mixture was extracted with EA (3 x 100 mL), and the combined organics were washed with brine (3 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (20: 1) to afford 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]-3-[2- (trimethylsilyl)ethynyl]pyrazolo[3,4-b]pyridine (700 mg, 94% yield) as a colorless oil. LCMS (ES, m/z): [M+H] + : 424

Synthesis of 69-b/c: 1 -((2-(trimethylsilyl)ethoxy)methyl)-3 -((trimethyl silyl)ethynyl)-1H- pyrazolo[3, 4-blpyri din-5 -ol

[0509] To a solution of 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]-3-[2- (trimethylsilyl)ethynyl]pyrazolo[3,4-b]pyridine (700 mg, 1.6 mmol, 1 equiv) and bis(pinacolato)diboron (837 mg, 3.2 mmol, 2 equiv) in dioxane (20 mL) were added KO Ac (323 mg, 3.2 mmol, 2 equiv) and Pd(dppf)C12 (120 mg, 0.16 mmol, 0.1 equiv) at room temperature under nitrogen atmosphere. After stirring for 2 h at 90 °C, the resulting mixture was allowed to cool and was diluted with THF (10 mL). 2% aqueous NaOH (10 mL) was added, followed by 30% H 2 O 2 (1.92 g, 16.9 mmol, 10 equiv) dropwise at room temperature. The reaction was stirred for 1 h, then diluted with water (50 mL) and extracted with EA (3 x 20 mL). The combined organics were washed with brine (3 x 50 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford l-[[2-(trimethylsilyl)ethoxy]methyl]-3-[2- (trimethylsilyl)ethynyl]pyrazolo[3,4-b]pyridin-5-ol (500 mg, 84% yield) as a yellow oil. LCMS (ES, m/z): [M+H] + : 362

Synthesis of 69-d: 2,4-difluoro-3-[[(l-[[2-(trimethylsilyl)ethoxy1methyl1-3-[2- (trimethylsilyl)ethynyl1pyrazolol3,4-b1pyridin-5-yl)oxy1meth yl1aniline [0510] To a stirred solution of l-[[2-(trimethylsilyl)ethoxy]methyl]-3-[2- (trimethylsilyl)ethynyl]pyrazolo[3,4-b]pyridin-5-ol (500 mg, 1.4 mmol, 1 equiv) and (3- amino-2,6-difluorophenyl)methanol (220 mg, 1.4 mmol, 1 equiv) in DCM (10 mL) were added TMAD (357 mg, 2.1 mmol, 1.5 equiv) and PPI13 (544 mg, 2.1 mmol, 1.5 equiv) at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was diluted with water (50 mL) and extracted with EA (3 x 100 mL). The combined organics were washed with brine (3 x 50 mL), dried over anhydrous ISfeSCL and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford 2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy]methyl]-3-[2- (trimethylsilyl)ethynyl]pyrazolo[3,4-b]pyridin-5-yl)oxy]meth yl]aniline (300 mg, 43% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 503 Synthesis of 69-e : N-(2,4-difluoro-3-[[(l-[[2-(trimethylsilyl)ethoxy1methyl1-3- [2- (trimethylsilyl)ethynyl1pyrazolo[3,4-b1pyridin-5-yl)oxy1meth yl1phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0511] To a stirred solution of 2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy]methyl]-3-[2- (trimethylsilyl)ethynyl]pyrazolo[3,4-b]pyridin-5-yl)oxy]meth yl]aniline (280 mg, 0.55 mmol, 1 equiv) and pyridine (132 mg, 1.6 mmol, 3 equiv) in DCM (5 mL) was added 5-fluoro-2- methoxypyridine-3 -sulfonyl chloride (188 mg, 0.8 mmol, 1.5 equiv) in portions. The reaction was stirred for 1 h then concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (1 : 1) to afford N-(2,4-difluoro-3-[[(1-[[2- (trimethylsilyl)ethoxy]methyl]-3-[2-(trimethylsilyl)ethynyl] pyrazolo[3,4-b]pyridin-5- yl)oxy]methyl]phenyl)-5-fluoro-2-methoxypyridine-3-sulfonami de (200 mg, 52% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 692

Synthesis of 69-f: N-(3-[[(3-ethynyl-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazo lo[3,4- b1pyridin-5-yl)oxy1methyl1-2,4-difluorophenyl)-5-fluoro-2-me thoxypyridine-3-sulfonamide [0512] To a stirred solution of N-(2,4-difluoro-3-[[(1-[[2-(trimethylsilyl)ethoxy]methyl]-3- [2- (trimethylsilyl)ethynyl] pyrazolo[3,4-b]pyridin-5-yl)oxy]methyl]phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide (200 mg, 0.29 mmol, 1 equiv) in MeOH (5 mL) was added K 2 CO 3 (111 mg, 0.87 mmol, 3 equiv). The resulting mixture was stirred for 6 h at room temperature, then concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (2: 1) to afford N-(3-[[(3-ethynyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]-2,4-difluorophenyl)- 5-fluoro-2-methoxypyridine-3-sulfonamide (150 mg, 84% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 620

Synthesis of Compound 69: N-[3-[([3-ethynyl-lE[-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl ]- 2A-difluorophenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0513] To a stirred solution of N-(3-[[(3-ethynyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]-2,4-difluorophenyl)- 5-fluoro-2-methoxypyridine-3-sulfonamide (120 mg, 0.19 mmol, 1 equiv) in DCM (4 mL) was added TFA (1 mL). The resulting mixture was stirred for 2 h then concentrated. The residue was purified by prep-HPLC with following conditions: Column, welch Vltimate XB- C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-60% MeCN / 0.1% aqueous formic acid; to afford N-[3-[([3-ethynyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl] -2,4-difluorophenyl]-5- fluoro-2-methoxypyridine-3 -sulfonamide (60 mg, 42% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 490

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.97 (s, 1H), 10.37 (s, 1H), 8.44 (d, J= 3.0 Hz, 1H), 8.28 (d, J= 2.7 Hz, 1H), 7.98 (dd, J= 13, 3.0 Hz, 1H), 7.81 (d, J= 2.7 Hz, 1H), 7.39 (td, J= 8.9, 5.9 Hz, 1H), 7.17 (td, J= 9.0, 1.6 Hz, 1H), 5.20 (s, 2H), 4.57 (s, 1H), 3.90 (3H, s).

Example 81: Synthesis of N-(3-(((3-cvdopropyl-1H-pyrazolo[3.,4-b]pyridin-5- yl)oxy)methyl)-2.,4-difluorophenyl)-5-fluoro-2-methoxypyridi ne-3-sulfonamide Compound 70

Synthesis of 70-a: 5-bromo-3-cyclopropyl-l-((2-(trimethylsilyl)ethoxy)methyl)-1 H- pyrazolol3,4-b1pyridine

[0514] To a stirred solution of 5-bromo-3-iodo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (500 mg, 1.1 mmol, 1 equiv) and cyclopropylboronic acid (104 mg, 1.2 mmol, 1.1 equiv) in dioxane (10 mL) were added K 3 PO 4 (467 mg, 2.2 mmol, 2 equiv) and Pd(dppf)C12 (81 mg, 0.11 mmol, 0.1 equiv) at room temperature, under nitrogen atmosphere. The reaction was stirred for 3 h at 90 °C then concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 40-90% MeCN / 0.1% aqueous formic acid; to afford 5-bromo-3-cyclopropyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (230 mg, 57% yield) as a brown oil. LCMS (ES, m/z): [M+H] + : 368

Synthesis of 70-b/c: 3-cyclopropyl-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazo lo[3,4- blpyridin-5-ol

[0515] To a stirred solution of 5-bromo-3 -cyclopropyl- 1 -[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (220 mg, 0.6 mmol, 1 equiv) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (156 mg, 0.66 mmol, 1.1 equiv) in dioxane (10 mL) were added KO Ac (117 mg, 1.2 mmol, 2 equiv) and Pd(dppf)C12 (44 mg, 0.06 mmol, 0.1 equiv) at room temperature, under nitrogen atmosphere. The reaction was stirred for 2 h at 90 °C then cooled to room temperature. THF (4 mL) was added, followed by 2% aqueous NaOH (4 mL) and 30% H 2 O 2 (1.73 g, 15.3 mmol, 10 equiv) dropwise. The resulting mixture was stirred for 1 h at room temperature, then quenched with saturated aqueous Na 2 S 2 O 3 (10 mL) and extracted with EA (3 x 20 mL). The combined organics were washed with brine (10 mL), dried over anhydrous ISfeSCL and concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 3-cyclopropyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3 ,4-b]pyridin-5-ol (140 mg, 76% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 306

Synthesis of 70-d : 3-(((3-cyclopropyl-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-p yrazolo[3,4- b1pyridin-5-yl)oxy)methyl)-2,4-difluoroaniline

[0516] To a stirred solution of 3-cyclopropyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (130 mg, 0.43 mmol, 1 equiv) and (3-amino-2,6-difluorophenyl)methanol (75 mg, 0.47 mmol, 1.1 equiv) in DCM (4 mL) were added TMAD (110 mg, 0.64 mmol, 1.5 equiv) and PPh 3 (167 mg, 0.64 mmol, 1.5 equiv). The reaction was stirred for 3 h, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50-95% MeCN / 0.1% aqueous formic acid; to afford 3-[[(3-cyclopropyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]-2,4-difluoroaniline (64 mg, 34% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 447

Synthesis of 70-e : N-(3-(((3-cyclopropyl-l-((2-(trimethylsilyl)ethoxy)methyl)-1 H- pyrazolo[3,4-b1pyridin-5-yl)oxy)methyl)-2,4-difluorophenyl)- 5-fluoro-2-methoxypyridine-3- sulfonamide

[0517] To a stirred solution of 3-[[(3-cyclopropyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]-2,4-difluoroaniline (60 mg, 0.13 mmol, 1 equiv) in pyridine (2 mL) was added dropwise a solution of 5-fluoro-2- methoxypyridine-3 -sulfonyl chloride (45 mg, 0.2 mmol, 1.5 equiv) in DCM (0.25 mL). The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by Flash- Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40 μm, 120 g; mobile phase: 50-95% MeCN / 0.1% aqueous formic acid; to afford N-(3-[[(3- cyclopropyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4 -b]pyridin-5-yl)oxy]methyl]- 2,4-difluorophenyl)-5-fluoro-2-methoxypyridine-3 -sulfonamide (66 mg, 77% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 636

Synthesis of Compound 70: N-(3-(((3-cyclopropyl-1H-pyrazolo[3,4-b]pyridin-5- yl)oxy)methyl)-2,4-difluorophenyl)-5-fluoro-2-methoxypyridin e-3-sulfonamide [0518] A solution of N-(3-[[(3-cyclopropyl-l-[[2-(trimethylsilyl)ethoxy]methyl]py razolo[3,4- b]pyridin-5-yl)oxy]rnethyl]-2,4-difluorophenyl)-5-fluoro-2-m ethoxypyridine-3-sulfonamide (65 mg, 0.1 mmol, 1 equiv) in TFA (0.33 mL) and DCM (1 mL) was stirred for 1 h at room temperature, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-100% MeCN / 0.1% aqueous formic acid; to afford N-[3-[([3-cyclopropyl-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy)rnethyl]-2,4-difluorophenyl]-5-fluoro-2-m ethoxypyridine-3-sulfonamide (19 mg, 37% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 506 1 H NMR (300 MHz, DMSO-d 6 ) δ 13.04 (s, 1H), 10.37 (s, 1H), 8.44 (d, J= 3.0 Hz, 1H), 8.16 (d, J= 2.8 Hz, 1H), 7.98 (dd, J= 13, 3.0 Hz, 1H), 7.87 (d, J= 2.8 Hz, 1H), 7.38 (td, J= 8.8, 5.9 Hz, 1H), 7.22-7.10 (m, 1H), 5.15 (s, 2H), 3.90 (s, 3H), 2.25 (qd, J= 7.9, 5.3 Hz, 1H), 0.98 (dp, J= 7.8, 2.5 Hz, 4H).

Example 82: Synthesis of N-[2,4-difluoro-3-[([l-methyl-2-oxo-3H-imidazo[4.,5-b]pyridi n-

6-yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulf onamide (Compound 71)

Compound 71

Synthesis of 71-a: benzyl N-(2-amino-5-bromopyridin-3-yl)carbamate

[0519] To a stirred solution of 5-bromopyridine-2,3-diamine (5 g, 27 mmol, 1 equiv) and pyridine (7.5 mL, 94 mmol, 3.5 equiv) in THF (125 mL) was added benzyl chloroformate (4.54 g, 27 mmol, 1 equiv) dropwise at 0 °C. The reaction was stirred for 3 h then diluted with water (20 mL). The resulting mixture was extracted with EtOAc (3 x 50 mL), and the combined organics were washed with water, brine, dried over anhydrous Na 2 SO 4 , then concentrated. The residue was purified by silica gel column chromatography, eluting with EtOAc/PE (0-50%) to afford benzyl N-(2-amino-5-bromopyridin-3-yl)carbamate (3.7 g, 43% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 322.

Synthesis of 71-b: 5-bromo-N 3 -methylpyridine-2,3-diamine

[0520] To a stirred mixture of LiAlH4 (1.7 g, 45.9 mmol, 4 equiv) in Et20 (40 mL) was added benzyl N-(2-amino-5-bromopyridin-3-yl)carbamate (3.7 g, 11.5 mmol, 1 equiv) in portions at 0 °C. The reaction mixture was stirred overnight at room temperature, then cooled back to 0 °C. MeOH (5 mL) was added followed by Na 2 SO 4 . 10 H 2 O (5 g) in portions. The resulting mixture was filtered, and the filter cake was washed with EtOAc (3 x 100 mL). The combined filtrate was concentrated and the residue was purified by silica gel column chromatography, eluting with EtOAc/PE (0-50%) to afford 5-bromo-N 3 -methylpyridine-2,3- diamine (1.8 g, 78% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 202.

Synthesis of 71-c: 6-bromo-l-methyl-3H-imidazo[4,5-b1pyridin-2-one

[0521] A solution of 5-bromo-N 3 -methylpyridine-2,3-diamine (1.7 g, 8.4 mmol, 1 equiv), EtsN (1.7 g, 17 mmol, 2 equiv) and CDI (2.05 g, 13 mmol, 1.5 equiv) in THF (20 mL) was stirred for 30 min, then heated for 3 h at 60 °C. The mixture was allowed to cool to room temperature, then was diluted with water (50 mL). The resulting mixture was extracted with EtOAc (3 x 150 mL) and the combined organics were washed with water, brine, dried over anhydrous Na 2 SO 4 , then concentrated. The residue was purified by silica gel column chromatography, eluting with 0-70% EtOAc/PE to afford 6-bromo-l-methyl-3H-imidazo[4,5- b]pyridin-2-one(1.7 g, 89% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 228.

Synthesis of 71-d: tert-butyl 6-bromo-l-methyl-2-oxoimidazo[4,5-b1pyridine-3-carboxylate [0522] A solution of 6-bromo-l-methyl-3H-imidazo[4,5-b]pyridin-2-one (1.7 g, 7.5 mmol, 1 equiv), BOC2O (2.44 g, 11 mmol, 1.5 equiv) and DMAP (182 mg, 1.5 mmol, 0.2 equiv) in MeCN (20 mL) was stirred overnight. The resulting mixture was diluted with water (20 mL) and extracted with EtOAc (3 x 50 mL). The combined organics were washed with water, brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-30% EtOAc/PE to afford tert-butyl 6-bromo-l- methyl-2-oxoimidazo[4,5-b]pyridine-3-carboxylate (1.8 g, 74% yield) as a yellow solid. LCMS (ES, m/z): [M+H] + : 328.

Synthesis of 71-e: (tert-butyl l-methyl-2-oxo-6-(4,4,5,5-tetramethyl-L3,2-dioxaborolan-2- yDimidazo [4,5-b1pyridine-3-carboxylate

[0523] A mixture of tert-butyl 6-bromo-l-methyl-2-oxoimidazo[4,5-b]pyridine-3-carboxylate (1 g, 3 mmol, 1 equiv), bis(pinacolato)diboron (1.2 g, 4.5 mmol, 1.5 equiv), KO Ac (0.6 g, 6.1 mmol, 2 equiv) and Pd(dppf)C12 CH 2 C12 (0.25 g, 0.3 mmol, 0.1 equiv) in dioxane (10 mL) was stirred for 2 h at 100 °C under nitrogen atmosphere. Desired product could be detected by LCMS. The resulting mixture was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 376.

Synthesis of 71-f: tert-butyl 6-hydroxy-l-methyl-2-oxoimidazo[4,5-b1pyridine-3-carboxylate [0524] The solution of (tert-butyl-l-methyl-2-oxo-6-(4,4,5,5-tetramethyl-l,3,2-diox aborolan- 2-yl)imidazo[4,5-b]pyridine-3-carboxylate (2.9 mmol, 1 equiv) was diluted with THF (11 mL), and 2% aqueous NaOH (11 mL) was added, followed by 30% H 2 O 2 (0.68 g, 5.9 mmol, 2 equiv) dropwise at room temperature. The reaction was stirred for 2 h then quenched with aqueous Na 2 S 2 O 4 (2 mL). The resulting mixture was extracted with EtOAc (3 x30 mL). The combined organics were washed with water (2 x 30 mL) and brine (30 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-50% EtOAc/PE to afford tert-butyl 6-hy droxy-1 -methyl -2- oxoimidazo[4,5-b]pyridine-3-carboxylate (450 mg, 57% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 266.

Synthesis of 71-g: tert-butyl 6-l(3-amino-2,6-difluorophenyl)methoxy1-l-methyl-2- oxoimidazo[4,5-b1 pyridine-3 -carboxylate

[0525] A solution of tert-butyl 6-hydroxy-l-methyl-2-oxoimidazo[4,5-b]pyridine-3- carboxylate (450 mg, 1.7 mmol, 1 equiv), (3-amino-2,6-difluorophenyl)methanol (540 mg, 3.4 mmol, 2 equiv), TMAD (584 mg, 3.4 mmol, 2 equiv) and PPI13 (890 mg, 3.4 mmol, 2 equiv) in DCM (4.5 mL) was stirred for 2 h then diluted with water (20 mL). The resulting mixture was extracted with DCM (3 x 50 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-50% EtOAc/PE to afford tert-butyl 6-[(3- amino-2,6-difluorophenyl)methoxy]-l-methyl-2-oxoimidazo[4,5- b]pyridine-3-carboxylate (306 mg, 44% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 407.

Synthesis of 71-h: 6-[[2,6-difluoro-3-(5-fluoro-2-methoxypyridine-3- sulfonamido)phenyl1methoxy1-l- methyl -2-oxoimidazo[4,5-b]pyridine-3-carboxylate [0526] To a stirred solution of tert-butyl 6-[(3-amino-2,6-difluorophenyl)methoxy]-l-methyl- 2-oxoimidazo[4,5-b]pyridine-3-carboxylate (150 mg, 0.37 mmol, 1 equiv) in pyridine (1.5 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (166 mg, 0.74 mmol, 2 equiv). The reaction was stirred for 2 h, then diluted with water (5 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-50% EtOAc/PE to afford tert-butyl 6-[[2,6-difluoro- 3-(5-fluoro-2-methoxypyri dine-3-sulfonamido)phenyl]methoxy]-l -methyl -2-oxoimidazo[4, 5- b]pyridine-3 -carboxylate (130 mg, 59% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 596.

Synthesis of Compound 71 : N-[2,4-difluoro-3-[([1-methyl-2-oxo-3H-imidazol4,5-b1pyridin - 6-yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfona mide

[0527] To a stirred of tert-butyl 6-[[2,6-difluoro-3-(5-fluoro-2-methoxypyridine-3- sulfonamido) phenyl]methoxy]-l-methyl-2-oxoimidazo[4,5-b]pyridine-3-carbo xylate (130 mg, 0.22 mmol, 1 equiv) was added 4 M HC1 in 1,4-dioxane (1.3 mL) dropwise at 0-5 °C. The resulting mixture was stirred for 2 h at room temperature, then concentrated. The residue was neutralized to pH 7 with saturated aqueous NaHCO 3 solution, and extracted with EtOAc (3 x 10 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column, 50*250 mm 5 μm 10 nm; mobile phase: 15-60% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-[([l-methyl-2- oxo-3H-imidazo[4,5-b]pyridin-6-yl]oxy)methyl]phenyl]-5-fluor o-2-methoxypyridine-3- sulfonamide (41 mg, 38% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 496.

1 H NMR (300 MHz, DMSO-d 6 ) δ11.36 (s, 1H), 10.36 (s, 1H), 8.45 (d, J= 2.9 Hz, 1H), 7.97 (dd, J= 13, 3.0 Hz, 1H), 7.61 (d, J = 2.5 Hz, 1H), 7.43-7.29 (m, 1H), 7.29 (d, J= 2.5 Hz, 1H), 7.14 (td, J = 9.0, 1.6 Hz, 1H), 5.08 (s, 2H), 3.91 (s, 3H), 3.27 (s, 3H).

Example 83: Synthesis of N-[2,4-difluoro-3-([2-oxo-1H-spiro[cvclopropane-l.,3- pyrrolo [2,3-b] pyridin] -5-yloxy] methyl)phenyl] -5-fluoro-2-methoxypyridine-3- sulfonamide (Compound 72

Synthesis of 72-a: 5'-bromo-l'H-spirorcyclopropane-L3'-pyrrolo[2,3-b1pyridin1-2 '-one [0528] To a stirred solution of 5-bromo-lH,3H-pyrrolo[2,3-b]pyridin-2-one (5 g, 24 mmol, 1 equiv) and dibromoethane (13.2 g, 70 mmol, 3 equiv) in DMF (50 mL) was added NaH (5.63 g, 141 mmol, 6 equiv, 60% in oil) in portions at 0-5 °C. The reaction was stirred for 2 h at room temperature, then cooled again to 0-5 °C and quenched with water (50 mL). The resulting mixture was extracted with EtOAc (3 x 150 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-40% EtOAc/PE to afford 5'-bromo-l'H-spiro[cyclopropane-l,3'-pyrrolo[2,3-b]pyridin]- 2'-one (2.4 g, 43% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 239.

Synthesis of 72-b: tert-butyl 5'-bromo-2'-oxospirorcyclopropane-L3'-pyrrolo[2,3-b1pyridine 1- 1'- carboxylate

[0529] A solution of 5'-bromo-l'H-spiro[cyclopropane-l,3'-pyrrolo[2,3-b]pyridin]- 2'-one (2.3 g, 9.6 mmol, 1 equiv), di-tert-butyl dicarbonate (2.5 g, 12 mmol, 1.2 equiv) and DMAP (0.24 g, 1.9 mmol, 0.2 equiv) in MeCN (50 mL) was stirred overnight. The resulting mixture was diluted with water (20 mL) and extracted with EtOAc (3 x 50 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-30% EtOAc/PE to afford tert-butyl 5'-bromo-2'-oxospiro[cyclopropane-l,3'-pyrrolo[2,3-b]pyridin e]-l'- carboxylate (2.4 g, 74% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 339.

Synthesis of 72-c: (tert-butyl 2-oxo-5-(4A5,5-tetramethyl-L3,2-dioxaborolan-2- yDspirorcyclopropane- L3-pyrrolor2,3-b]pyridine]-l-carboxylate

[0530] A mixture of tert-butyl 5'-bromo-2'-oxospiro[cyclopropane-l,3'-pyrrolo[2,3- b]pyridine]-l'-carboxylate (2.4 g, 7.1 mmol, 1 equiv), bis(pinacolato)diboron (2.7 g, 11 mmol, 1.5 equiv), KO Ac (1.39 g, 14 mmol, 2 equiv) and Pd(dppf)C12.CH 2 C12 (289 mg, 0.35 mmol, 0.05 equiv) in dioxane (25 mL) was stirred for 2 h at 100 °C. Desired product could be detected by LCMS. The resulting mixture was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 387.

Synthesis of 72-d: tert-butyl 5-hydroxy-2-oxospiro[cyclopropane-L3-pyrrolor2,3- b]pyridine]-l- carboxylate

[0531] To the crude mixture (tert-butyl 2-oxo-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)spiro[cyclopropane-l,3-pyrrolo[2,3-b]pyridine]-l-carboxyl ate (5.85 mmol, 1 equiv) were added THF (22 mL), 2% aqueous NaOH (22 mL) and 30% H 2 O 2 (2.7 g, 23.4 mmol, 4 equiv) dropwise at room temperature. The reaction was stirred for 2 h then quenched with aqueous Na2S2C>4 (200 mL). The resulting mixture was extracted with EtOAc (3 x 150 mL). The combined organics were washed with water (2 x 150 mL) and brine (1 x 150 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-50% EtOAc/PE to afford tert-butyl 5-hydroxy-2- oxospiro[cyclopropane-l,3-pyrrolo[2,3-b]pyridine]-l-carboxyl ate (1 g, 62% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 277.

Synthesis of 72-e: tert-butyl 5'-[(3-amino-2,6-difluorophenyl)methoxy1-2'- oxospiro[cyclopropane-L3' -pyrrolo[2,3-b1pyridine1-l'-carboxylate

[0532] A solution of tert-butyl 5'-hydroxy-2'-oxospiro[cyclopropane-l,3'-pyrrolo[2,3- b]pyridine]-l'-carboxylate (1 g, 3.6 mmol, 1 equiv), (3-amino-2,6-difluorophenyl)methanol (0.63 g, 4 mmol, 1.1 equiv), TMAD (0.55 g, 5 mmol, 1.3 equiv) and PPI13 (1.23 g, 5 mmol, 1.3 equiv) in DCM (10 mL) was stirred for 2 h then diluted with water (20 mL). The resulting mixture was extracted with DCM (3 x 50 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-60% EtOAc/PE to afford tert-butyl 5'-[(3- amino-2,6-difluorophenyl)methoxy]-2'-oxospiro[cyclopropane-l ,3'-pyrrolo[2,3-b]pyridine]- l'-carboxylate (700 mg, 46% yield) as a yellow solid LCMS (ES, m/z): [M+H] + : 418.

Synthesis of 72-f: tert-butyl 5'-[[2,6-difluoro-3-(5-fluoro-2-methoxypyridine-3- sulfonamido)phenyl] methoxy]-2'-oxospirorcyclopropane-L3'-pyrrolor2,3-b]pyridine ]-l'- carb oxy late

[0533] To a stirred solution of tert-butyl 5'-[(3-amino-2,6-difluorophenyl)methoxy]-2'- oxospiro[cyclopropane-l,3'-pyrrolo[2,3-b]pyridine]-l'-carbox ylate (150 mg, 0.36 mmol, 1 equiv) in pyridine (1.5 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (162 mg, 0.72 mmol, 2 equiv). The reaction was stirred for 2 h then diluted with water (5 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organics were washed with water and brine, dried over anhydrous TsfeSCU and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-100% EtOAc/PE to afford tert-butyl 5'-[[2,6-difluoro-3-(5-fluoro-2-methoxypyridine-3-sulfonamid o)phenyl]methoxy]- 2'-oxospiro[cyclopropane-l,3'-pyrrolo[2,3-b]pyridine]-l'-car boxylate (145 mg, 67% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 607.

Synthesis of Compound 72: N-[2,4-difluoro-3-([2-oxo-1H-spiro[cyclopropane-L3- pyrrolo[2,3-b]pyridin]-5-yloxy]methyl)phenyl]-5-fluoro-2-met hoxypyridine-3-sulfonamide [0534] Tert-butyl 5 - [ [2, 6-difluoro-3 -(5 -fluoro-2-methoxy pyridine-3 - sulfonamido)phenyl]methoxy]-2-oxospiro[cyclopropane- 1 ,3 -pyrrolo[2,3 -b]pyridine]- 1 - carboxylate (100 mg) was treated with 4 N HC1 in 1,4-di oxane (1 mL) and stirred for 2 h at room temperature. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH F-Phenyl OBD Column, 19*150 mm, 5 um; mobile phase: 15-42% MeCN / 20 mmol/LNH 4 HCO 3 ; to afford N-[2,4-difluoro-3-([2-oxo-lELspiro[cyclopropane-l,3-pyrrolo[ 2,3-b]pyridin]-5- yloxy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamid e (12 mg, 14% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 507. 1 H NMR (300 MHz, DMSO-d 6 ) δ 11.00 (s, 1H), 8.30 (s, 1H), 7.90 (d, J= 5.7 Hz, 1H), 7.77 (d, J = 2.7 Hz, 1H), 7.31-7.19 (m, 1H), 6.94 (s, 1H), 5.01 (s, 2H), 3.83 (s, 3H), 1.63 (d, J = 3.9 Hz, 2H), 1.51 (q, J = 4.3, 3.7 Hz, 2H).

Example 84: Synthesis of N-(3-(((3-cyano-1H-pyrazolo[3.,4-b]pyridin-5-yl)oxy)methyl)-

2,4-difluorophenyl)-5-fluoro-2-methoxypyridine-3-sulfonam ide (Compound 73)

Synthesis of 73-a: 5-bromo-3-iodo-1H-pyrazolo[3,4-b]pyridine

[0535] A solution of 5-bromo-lH-pyrazolo[3,4-b]pyridine (10 g, 51 mmol, 1 equiv) and N- iodosuccinimide (11.9 g, 53 mmol, 1.05 equiv) in dry 1,2-di chloroethane (300 mL) was heated at reflux for 6 h. The reaction was cooled to room temperature and diluted with THF (300 mL). The resulting solution was washed with saturated aqueous Na 2 S 2 O 3 solution (2 x 100 mL), then brine (2 x 50 mL). The organic phase was dried over anhydrous Na 2 SO 4 and concentrated. The residue was triturated with a 1 : 1 mixture of DCM/ether (50 mL), and then washed with ether (100 mL) to afford 5-bromo-3-iodo-lH-pyrazolo[3,4-b]pyridine as a yellow solid (12.4 g, crude).

LCMS (ES, m/z): [M+H] + : 324

Synthesis of 73-b: 5-bromo-3-iodo-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyraz olo[3,4- blpyridine

[0536] To a stirred solution of 5-bromo-3-iodo-lH-pyrazolo[3,4-b]pyridine (5.2 g, 16 mmol, 1 equiv) in DMF (50 mL) was added 60% NaH in oil (1.93 g, 48 mmol, 3 equiv) in portions at 0 °C. The reaction was stirred for 0.5 h at 0 °C, then SEMC1 (4 g, 24 mmol, 1.5 equiv) was added dropwise over 30 min at 0 °C. The reaction was stirred for 1 h at room temperature, then quenched with water (300 mL). The resulting mixture was extracted with EA (3 x 100 mL). The combined organics were washed with brine (100 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (20: 1) to afford 5-bromo-3-iodo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (3.2 g, 44% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 454

Synthesis of 73-c: 5-bromo-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolo[3,4 -b1pyridine- 3 -carbonitrile

[0537] To a stirred solution of 5-bromo-3-iodo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (500 mg, 1.1 mmol, 1 equiv) and zinc cyanide (103 mg, 0.88 mmol, 0.8 equiv) in DMF (10 mL) were added dppf (122 mg, 0.22 mmol, 0.2 equiv) and Pd2(dba)3'CHC13 (114 mg, 0.11 mmol, 0.1 equiv) in portions at room temperature, under nitrogen atmosphere. The reaction was stirred for 3 h at 90 °C, then cooled, filtered and the filtrate concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50-90% MeCN / 0.1% aqueous formic acid; to afford 5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-3-carb onitrile (260 mg, 67% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 353

Synthesis of 73-d/e: 5-hydroxy-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolo[3 ,4- blpyridine-3 -carbonitrile

[0538] To a stirred solution of 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridine-3 -carbonitrile (255 mg, 0.72 mmol, 1 equiv) and bis(pinacolato)diboron (202 mg, 0.79 mmol, 1.1 equiv) in dioxane (8 mL) were added KO Ac (142 mg, 1.44 mmol, 2 equiv) and Pd(dppf)C12 (53 mg, 0.072 mmol, 0.1 equiv) in portions at room temperature, under nitrogen atmosphere. The reaction was stirred for 3 h at 90 °C, then cooled. THF (4 mL) and 2% aqueous NaOH (4 mL) were added, followed by 30% H 2 O 2 (246 mg, 7.24 mmol, 10 equiv) dropwise at room temperature. The reaction was stirred for 2 h, then quenched with saturated aqueous Na 2 S 2 O 3 (10 mL). The mixture was extracted with EA (3 x 20 mL). The combined organics were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 5-hydroxy-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-3-carb onitrile (195 mg, 93% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 291

Synthesis of 73-f: 5-((3-amino-2,6-difluorobenzyl)oxy)-l-((2-(trimethylsilyl)et hoxy)methyl)- lH-pyrazolo[3,4-b1pyridine-3-carbonitrile

[0539] To a stirred solution of 5-hydroxy-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridine-3 -carbonitrile (180 mg, 0.62 mmol, 1 equiv) and (3-amino-2,6- difluorophenyl)methanol (108 mg, 0.68 mmol, 1.1 equiv) in DCM (8 mL) were added TMAD (160 mg, 0.93 mmol, 1.5 equiv) and PPI13 (244 mg, 0.93 mmol, 1.5 equiv). The reaction was stirred for 3 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50-95% MeCN / 0.1% aqueous formic acid; to afford 5-[(3-amino-2,6- difluorophenyl)methoxy]-l-[[2-(trimethylsilyl)ethoxy]methyl] pyrazolo[3,4-b]pyridine-3- carbonitrile (148 mg, 55% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 432

Synthesis of 73-g: N-(3-(((3-cyano-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyra zolo[3,4- b1pyridin-5-yl)oxy)methyl)-2,4-difluorophenyl)-5-fluoro-2-me thoxypyridine-3-sulfonamide [0540] To a stirred solution of 5-[(3-amino-2,6-difluorophenyl)methoxy]-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-3-carb onitrile (140 mg, 0.32 mmol, 1 equiv) in pyridine (3 mL) was added 5 -fluoro-2-methoxypyridine-3 -sulfonyl chloride (110 mg, 0.49 mmol, 1.5 equiv). The reaction was stirred for 2 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50-95% MeCN / 0.1% aqueous formic acid; to afford N-(3-[[(3-cyano-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo [3,4-b]pyridin-5- yl)oxy]methyl]-2,4-difluorophenyl)-5-fluoro-2-methoxypyridin e-3-sulfonamide (190 mg, 94% yield) as a brown oil. LCMS (ES, m/z): [M+H] + : 621

Synthesis of Compound 73: N-(3-(((3-cyano-1H-pyrazolo[3,4-b1pyridin-5-yl)oxy)methyl)- 2,4-difluorophenyl)-5-fluoro-2-methoxypyridine-3 -sulfonamide

[0541] A solution of N-(3-[[(3-cyano-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo [3,4- b]pyridin-5-yl)oxy]methyl]-2,4-difluorophenyl)-5-fluoro-2-me thoxypyridine-3-sulfonamide (190 mg, 0.31 mmol, 1 equiv) in TFA (1 mL) and DCM (3 mL) was stirred for 1 h at room temperature. The resulting mixture was concentrated and the residue was purified by prep- HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 5-100% MeCN / 0.1% aqueous formic acid; to afford N-[3-[([3-cyano- lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]-2,4-difluoropheny l]-5-fluoro-2- methoxypyridine-3 -sulfonamide (72 mg, 48% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 491

1 H NMR (300 MHz, DMSO-d 6 ) δ 14.94 (s, 1H), 10.39 (s, 1H), 8.41 (dd, J= 8.3, 2.9 Hz, 2H), 8.08 (d, J= 2.7 Hz, 1H), 7.97 (dd, J= 7.4, 3.1 Hz, 1H), 7.40 (td, J= 8.9, 6.0 Hz, 1H), 7.18 (t, J= 8.7 Hz, 1H), 5.22 (s, 2H), 3.90 (s, 3H).

Example 85: Synthesis of N-[2-chloro-4-fluoro-3-[([3-methyl-1H-pyrazolo[3.,4-b]pyridi n-

5-yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine-3-sulfo namide (Compound 74)

Compound 74

Synthesis of 74-a: N-(2-chloro-4-fluoro-3-(((3-methyl-l-(tetrahydro-2H-pyran-2- yl)-1H- pyrazolo[3,4-b1pyridin-5-yl)oxy)methyl)phenyl)-5-fluoro-2-me thylpyridine-3-sulfonamide

[0523] To a stirred solution of 2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (300 mg, 0.7 mmol, 1 equiv) and pyridine (182 mg, 2.3 mmol, 3 equiv) in DCM (5 mL) was added 5-fluoro-2-methylpyridine-3 -sulfonyl chloride (241 mg, 1.1 mmol, 1.5 equiv). The resulting mixture was stirred for 2 h then concentrated. The residue was purified by silica gel column chromatography, eluting with PE: EA (4: 1) to afford N-[2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyri din-5- yl]°xy]methyl)phenyl]-5-fluoro-2-methylpyridine-3-sulfonami de (120 mg, 28% yield) as a colorless oil.

LCMS (ES, m/z): [M+H] + : 564.

Synthesis of Compound 74: N-[2-chloro-4-fluoro-3-[([3-methyl-1H-pyrazolo[3,4-b1pyridin - 5-yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine-3-sulfonam ide

[0524] To a stirred solution of N-[2-chloro-4-fluoro-3-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methylpyridine-3-sulfonamide (100 mg) in DCM (5 mL) was added TFA (1 mL). The resulting mixture was stirred for 2 h then concentrated. The crude product was purified by Prep-HPLC with following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10 μm; Mobile Phase: 20-60% MeCN / 0.1% aqueous formic acid; to afford N-[2-chloro-4-fluoro-3-[([3-methyl-lH-pyrazolo[3,4- b]pyridin-5-yl]oxy)methyl]phenyl]-5-fluoro-2-methylpyridine- 3-sulfonamide (50 mg, 59% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 480

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.70 (s, 1H), 8.71 (d, J= 2.8 Hz, 1H), 8.18 (d, J= 2.7 Hz, 1H), 7.93 -7.75 (m, 2H), 7.45-7.22 (m, 2H), 5.16 (s, 2H), 2.76 (s, 3H), 2.47 (s, 3H).

Example 86: Synthesis of N-[3-([[3-(2,2-difluoroethyl)-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy]methyl)-2.,4-difluorophenyl]-5-fluoro-2-methoxypyridi ne-3-sulfonamide

(Compound 75)

Synthesis of 75-a: 2-(5-bromo-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolol3,4- b1pyridin-3- yl acetaldehyde

[0525] Into a 40 mL vial was placed 2-(5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-3-yl)et hanol (600 mg, 1.6 mmol, 1 equiv), CH 3 CN (5 mL) and IBX (3.6 g, 13 mmol, 8 equiv). The resulting solution was stirred for 2 h at 80 °C then cooled and filtered. The filtrate was concentrated to give crude 2-(5- bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyr idin-3-yl)acetaldehyde (400 mg) as off-white oil which was used in next step directly without further purification. Synthesis of 75-b: 5-bromo-3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine [0526] Into a 40 mL vial was placed 2-(5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-3-yl)ac etaldehyde (400 mg, 1 mmol, 1 equiv), DCM (5 mL) and BAST (860 mg, 3.6 mmol, 3.6 equiv). The reaction was stirred for 2 h then diluted with NaHCO 3 , solution (5 mL). The resulting mixture was extracted with ethyl acetate (3 x 5 mL) and the combined organics were dried over anhydrous sodium sulfate, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 60- 95% MeCN / 0.1% aqueous formic acid; to afford 5-bromo-3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (300 mg, 71% yield) as a yellow oil. LCMS (ES, m/z): [M+H] + : 392

Synthesis of 75-c: 3-(2,2-difluoroethyl)-l-methyl-5-(4,4,5,5-tetramethyl-L3,2-d ioxaborolan- 2-yl)pyrazolo[3,4-b1pyridine

[0527] To a stirred solution of 5-bromo-3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (300 mg, 0.8 mmol, 1 equiv) and 4,4,5,5-tetramethyl-2-(tetramethyl-l,3,2-dioxaborolan-2-yl-l ,3,2-dioxaborolane (213 mg, 0.8 mmol, 1 equiv) in dioxane (4 mL) was added Pd(dppf)C12 (62 mg, 0.08 mmol, 0.1 equiv) and KO Ac (150 mg, 1.6 mmol, 2 equiv) in portions at room temperature under nitrogen atmosphere. The resulting solution was stirred for 2 h at 90 °C then concentrated under vacuum to give crude 3-(2,2-difluoroethyl)-l-methyl-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[3,4-b]pyridine (240 mg) as brown oil which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 440

Synthesis of 75-d: 3-(2,2-difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy1methyl1py razolo[3,4- blpyridin-5-ol

[0528] To crude 3-(2,2-difluoroethyl)-l-methyl-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2- yl)pyrazolo[3,4-b]pyridine (240 mg) was added THF (4 mL) and 2% aqueous NaOH (4 mL), followed by 30% H 2 O 2 (139 mg, 4 mmol, 10 equiv) dropwise at room temperature. The resulting mixture was stirred for 1 h then quenched with saturated aqueous Na 2 S 2 O 3 (10 mL) and extracted with EA (3 x 10 mL). The combined organics were washed with brine (5 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash-Prep- HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 3-(2,2- difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[ 3,4-b]pyridin-5-ol (200 mg, 95% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 330

Synthesis of 75-e: 3-(i[3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl1ox y1methyl)-2,4-difluoroaniline [0529] To a stirred solution of 3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (220 mg, 0.7 mmol, 1 equiv) in DCM (6 mL) was added (3-amino-2,6-difluorophenyl)methanol (117 mg, 0.7 mmol, 1 equiv), TMAD (138 mg, 0.8 mmol, 1.2 equiv) and PPh 3 (210 mg, 0.8 mmol, 1.2 equiv). The reaction was stirred for 1 h then diluted with H 2 O (5 mL). The resulting mixture was extracted with ethyl acetate (3x5 mL), and the combined organics were dried over anhydrous sodium sulfate, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30- 80% MeCN / 0.1% aqueous formic acid; to afford 3-([[3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)-2,4-difluoroaniline (200 mg, 64% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 471

Synthesis of 75-f: N-13-([[3-(2,2-difluoroethyl)-l-[i2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl1ox y1methyl)-2,4-difluorophenyl1- 5-fluoro-2-methoxypyridine-3-sulfonamide

[0530] Into an 8 mL vial was placed 3-([[3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)-2,4-difluoroaniline (190 mg, 0.4 mmol, 1 equiv), pyridine (2 mL) and 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (110 mg, 0.5 mmol, 1.2 equiv). The resulting solution was stirred for 1 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford N-[3-([[3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)-2,4-difluorophenyl]- 5-fluoro-2-methoxypyridine-3-sulfonamide (230 mg, 86% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 660

Synthesis of Compound 75: N-[3-([[3-(2,2-difluoroethyl)-1H-pyrazolo[3,4-b1pyridin-5- yl1oxy1methyl)-2,4-difluorophenyl1-5-fluoro-2-methoxypyridin e-3-sulfonamide [0531] Into a 40 mL vial was placed N-[3-([[3-(2,2-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)-2,4-difluorophenyl]- 5-fluoro-2-methoxypyridine-3-sulfonamide (230 mg, 0.3 mmol, 1 equiv), DCM (3 mL) and TFA (1 mL). The resulting solution was stirred for 1 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 25-65% MeCN / 0.1% aqueous formic acid; to afford N-[3- ([[3-(2,2-difluoroethyl)-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy] methyl)-2,4-difluorophenyl]-5- fluoro-2-methoxypyridine-3-sulfonamide (78 mg, 42% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 530

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.48 (s, 1H), 10.37 (s, 1H), 8.45 (d, J= 3.0 Hz, 1H), 8.23 (d, J= 2.8 Hz, 1H), 8.02-7.90 (m, 2H), 7.38 (td, J= 9.0, 6.0 Hz, 1H), 7.16 (t, J= 9.0 Hz, 1H), 6.44 (t, J= 4.5 Hz, 1H), 5.13 (s, 2H), 3.91 (s, 3H), 3.53 (td, J= 17.7, 4.5 Hz, 2H).

Example 87: Synthesis of N-[2,4-difluoro-3-([[3-(2-fluoroethyl)-1H-pyrazolo[3.,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (Compound 76)

76-f

76-e

Compound 76 Synthesis of 76-a: 5-bromo-3-ethenyl-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazo lo[3,4- blpyridine

[0532] To a stirred solution of 5-bromo-3-iodo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1 g, 2.2 mmol, 1 equiv) and 2- ethenyl-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (0.41 g, 2.64 mmol, 1.2 equiv) in dioxane (10 mL) were added CS 2 CO 3 (2.15 g, 6.6 mmol, 3 equiv) and Pd(dppf)C12 (0.16 g, 0.22 mmol, 0.1 equiv) under nitrogen atmosphere. The reaction was stirred for 5 h at 90 °C, before being cooled and quenched with H 2 O (30 mL), then extracted with EtOAc (3 x 10 mL). The combined organics were washed with brine (30 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (5: 1) to afford 5-bromo-3-ethenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (400 mg, 51% yield) as a white oil. Synthesis of 76-b: 2-(5-bromo-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4- b1pyridin-3- yDethanol

[0533] To a stirred solution of 5-bromo-3-ethenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1.5 g, 4.23 mmol, 1 equiv) in THF (15 mL) was added 9-borabicyclo[3.3.1]nonane (2.07 g, 16.93 mmol, 4 equiv) dropwise at 0 °C. The reaction was stirred for 18 h at room temperature, then cooled back to 0 °C and quenched with 2% aqueous NaOH (15 mL) dropwise over 1 min. 30% H 2 O 2 (1.44 g, 42.33 mmol, 10 equiv) was added dropwise over 2 min and the reaction stirred to room temperature over 30 mins. Saturated aqueous Na 2 S 2 O 3 (10 mL) was added, and the mixture extracted with EA (3 x 10 mL). The combined organics were washed with brine (5 mL), dried over anhydrous ISfeSCL and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (3 : 1) to afford 2-(5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-3-yl)et hanol (1.12 g, 71% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 372

Synthesis of 76-c: 5-bromo-3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine [0534] To a stirred solution of 2-(5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyri din-3 -yl)ethanol (500 mg, 1.3 mmol, 1 equiv) in DCM (10 mL) was added BAST (2.97 g, 13 mmol, 10 equiv) in portions at 0 °C. The reaction was stirred for 3 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50-95% MeCN / 0.1% aqueous formic acid; to afford 5-bromo-3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (380 mg, 76% yield) as a yellow oil. LCMS (ES, m/z): [M+H] + : 374

Synthesis of 76-d: 3-(2-fluoroethyl)-5-(4,4,5,5-tetramethyl-E3,2-dioxaborolan-2 -yl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine

[0535] To a stirred solution of 5-bromo-3-(2-fhioroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (39 mg, 0.1 mmol, 1 equiv) and bis(pinacolato)diboron (40 mg, 0.16 mmol, 1.5 equiv) in dioxane (2 mL) were added KO Ac (21 mg, 0.21 mmol, 2 equiv) and Pd(dppf)C12 (73 mg, 0.1 mmol, 0.1 equiv) at room temperature, under nitrogen atmosphere. The reaction was stirred for 3 h at 90 °C then cooled and quenched with H 2 O (20 mL). This was extracted with EtOAc (3 x 10 mL), and the combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 then concentrated to afford 3-(2-fluoroethyl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)-l- [[2-(trimethylsilyl)ethoxy] methyl]pyrazolo[3,4-b]pyridine (400 mg, 94% yield) as a brown oil, which was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 422

Synthesis of 76-e: 3-(2-fluoroethyl)-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazo lol3,4- blpyridin-5-ol

[0536] To 3-(2-fluoroethyl)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo [3,4-b]pyridine (400 mg, 0.95 mmol, 1 equiv) in tetrahydrofuran (4 mL) was added 2% aqueous NaOH solution (4 mL) and 30% H 2 O 2 (0.32 g, 9.49 mmol, 10 equiv) dropwise at room temperature. The reaction was stirred for 1 h at room temperature, then quenched with H 2 O (20 mL) and extracted with EtOAc (3 x 10 mL). The combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (290 mg, 98% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 312

Synthesis of 76-f: 2,4-difluoro-3-([[3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl1ox y1methyl)aniline [0537] A solution of 3-(2-fluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazo lo[3,4- b]pyridin-5-ol (290 mg, 0.93 mmol, 1 equiv), (3-amino-2,6-difluorophenyl)methanol (178 mg, 1.1 mmol, 1.2 equiv), PPh 3 (366 mg, 1.4 mmol, 1.5 equiv) and TMAD (241 mg, 1.4 mmol, 1.5 equiv) in DCM (10 mL) was stirred for 5 h at room temperature. The resulting mixture was quenched with H 2 O (20 mL) and extracted with EtOAc (3 x 10 mL). The combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 40-90% MeCN / 0.1% aqueous formic acid; to afford 2,4-difluoro-3-([[3-(2-fhioroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)aniline (343 mg, 81% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 453

Synthesis of 76-g: N-[2,4-difluoro-3-([[3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0538] To a stirred solution of 2,4-difluoro-3-([[3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)aniline (348 mg, 0.77 mmol, 1 equiv) in pyridine (5 mL) was added 5 -fluoro-2-methoxypyridine-3 -sulfonyl chloride (260 mg, 1.2 mmol, 1.5 equiv) in DCM (0.5 mL) dropwise. The reaction was stirred for 2 h then concentrated to afford crude N-[2,4-difluoro-3-([[3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (450 mg, 91% yield) as a brown oil which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 642

Synthesis of Compound 76: N-[2,4-difluoro-3-([[3-(2-fluoroethyl)-1H-pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide [0539] A solution of N-[2,4-difluoro-3-([[3-(2-fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (490 mg, 0.76 mmol, 1 equiv) in TFA (0.5 mL) and DCM (5 mL) was stirred for 1 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-70% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-([[3-(2-fluoroethyl)- lH-pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (82 mg, 21% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 512

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.30 (s, 1H), 10.37 (s, 1H), 8.45 (d, J= 2.9 Hz, 1H), 8.20 (d, J= 2.8 Hz, 1H), 8.02-7.89 (m, 2H), 7.38 (td, J= 8.9, 6.1 Hz, 1H), 7.16 (t, J= 8.3 Hz, 1H), 5.13 (s, 2H), 4.90 (t, J= 6.5 Hz, 1H), 4.74 (t, J= 6.5 Hz, 1H), 3.91 (s, 3H), 3.39-3.21 (m, 2H).

Example 88: Synthesis of N-(4-chloro-2-fluoro-3-(((3-methyl-1H-pyrazolo[3.,4-b]pyridi n- 5-yl)oxy)methyl)phenyl)-5-fluoro-2-methylpyridine-3-sulfonam ide (Compound 77)

77-d Compound 77

Synthesis of 77-a: ethyl 3-amino-6-chloro-2-fluorobenzoate

[0540] 4-Chloro-2-fluoroaniline (1 g, 6.9 mmol, 1 equiv) was dissolved in THF (2 mL) and cooled to -78 °C. 2.5 M n-Butyllithium in hexanes (3 mL, 7.6 mmol) was slowly added dropwise, keeping the temperature below -70 °C. The mixture was stirred at -70 °C for 30 minutes, then chloro[2-(chlorodimethylsilyl)ethyl]dimethylsilane (1.55 g, 7.2 mmol, 1.05 equiv) in THF (1 mL) was slowly added dropwise to the reaction mixture while keeping the temperature below -70 °C. Once addition was complete, the mixture was stirred at -78 °C for 1 hour, then further 2.5 M n-butyllithium (3 mL, 7.6 mmol) was slowly added dropwise. The mixture was stirred for 30 minutes at -78 °C, then allowed to warm to 15 °C over 1 hour. The reaction mixture was cooled to -78 °C again, and 2.5 M n-butyllithium (3 mL, 7.6 mmol) was slowly added dropwise. The mixture was stirred below -70 °C for 90 minutes, then ethyl chloroformate (970 mg, 8.9 mmol, 1.3 equiv) was added dropwise, again maintaining the temperature below -70 °C. The cooling bath was removed and the reaction mixture was allowed to reach room temperature over 16 hrs. The reaction was cooled in an ice/water bath and quenched by addition of 3 M hydrochloric acid (50 mL). The mixture was stirred at room temperature for 2 hours, then made basic by addition of potassium carbonate solution. The mixture was extracted with ethyl acetate (3 x 100 mL), and the combined organics were washed with brine (50 mL), dried over magnesium sulfate and concentrated. The residue was purified with silica gel column chromatography eluting with 0-100% ethyl acetate in hexane to provide ethyl 3-amino-6-chloro-2-fluorobenzoate (1.3 g, 87% yield) as a light yellow oil. LCMS (ES, m/z): [M+H] + : 218.

Synthesis of 77-b: (3-amino-6-chloro-2-fluorophenyl)methanol

[0541] To a solution of ethyl 3-amino-6-chloro-2-fluorobenzoate (1.1 g, 5.1 mmol, 1 equiv) in THF (20 mL) was added LiA1H4 (0.38 g, 0.12 mmol, 2 equiv) and the solution stirred for 1 h at room temperature. The reaction was cooled in an ice bath then quenched by the addition of MeOH (5 mL). Na 2 SO 4 .10H 2 O (1 g) was added and the resulting mixture was stirred for 4 h. The mixture was filtered, and the filter cake washed with EA (3 x 20 mL). The combined filtrate was concentrated and the residue purified by silica gel column chromatography, eluting with PEZEA (1/1) to afford (3-amino-6-chloro-2-fluorophenyl)methanol (918 mg, 94% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 176.

Synthesis of 77-c: 4-chloro-2-fluoro-3-(((3-methyl-l-(tetrahydro-2H-pyran-2-yl) -1H- pyrazolo [3,4-b1pyridin-5-yl)oxy)methyl)aniline

[0542] A solution of (3-amino-6-chloro-2-fluorophenyl)methanol (818 mg, 4.7 mmol, 1 equiv), 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (1.6 g, 7 mmol, 1.5 equiv) and PPh 3 (1.8 g, 7 mmol, 1.5 equiv) in DCM (10 mL) was treated with TMAD (1.2 g, 7 mmol, 1.5 equiv), then stirred for 1 h. The resulting mixture was diluted with EA (50 mL), and the organics were washed with water and brine, dried over anhydrous ISfeSCL, then concentrated. The residue was purified by silica gel column chromatography, eluting with PEZEA (1/1) to afford 4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b] pyridin-5- yl]°xy]methyl)aniline (916 mg, 50% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 391.

Synthesis of 77-d: N-(4-chloro-2-fluoro-3-(((3-methyl-l-(tetrahydro-2H-pyran-2- yl)-1H- pyrazolo [3,4-b1pyridin-5-yl)oxy)methyl)phenyl)-5-fluoro-2-methylpyri dine-3-sulfonamide [0543] To a stirred solution of 4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (250 mg, 0.64 mmol, 1 equiv) in pyridine (2.5 mL) was added 5-fluoro-2-methylpyridine-3-sulfonyl chloride (268 mg, 1.3 mmol, 2 equiv). The reaction was stirred for 1 h then diluted with water (2 mL). The resulting mixture was extracted with DCM (3 x 10 mL). The combined organics were washed with water (2 x 10 mL) and brine (10 mL), dried over anhydrous Na 2 SO 4 then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with 0-50% EtOAc/PE to afford N-[4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin- 5-yl]oxy]methyl)phenyl]-5-fluoro-2-methylpyridine-3-sulfonam ide (163 mg, 45% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + :564 .

Synthesis of Compound 77: N-(4-chloro-2-fluoro-3-(((3-methyl-1H-pyrazolol3,4-b1pyridin - 5-yl)oxy)methyl)phenyl)-5-fluoro-2-methylpyridine-3-sulfonam ide

[0544] N-[4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 -b]pyridin-5- yl]oxy]methyl)phenyl]-5-fluoro-2-methylpyridine-3-sulfonamid e (153 mg, 0.27 mmol, 1 equiv) was treated with 4 N HC1 in MeOH (4 mL) and stirred for 1 h at room temperature. The resulting mixture was concentrated and the residue purified by Prep-HPLC with the following conditions: Column: Sunfire Prep C18 OBD Column, 50*250 mm 5 μm 10 nm; Mobile Phase: 5-45% MeCN / 0.05% aqueous ammonia; to afford N-[4-chloro-2-fluoro-3- [([3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phenyl ]-5-fluoro-2-methylpyridine- 3-sulfonamide (66 mg, 51% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 480

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 10.93 (s, 1H), 8.67 (d, J= 2.8 Hz, 1H), 8.19 (d, J= 2.7 Hz, 1H), 7.94 (dd, J= 8.3, 2.9 Hz, 1H), 7.87 (d, J= 2.7 Hz, 1H), 7.34 (d, J= 6.0 Hz, 2H), 5.14 (d, J= 2.1 Hz, 2H), 2.76 (s, 3H), 2.48 (s, 3H).

Example 89: Synthesis of N-(4-chloro-2-fluoro-3-(((3-methyl-1H-pyrazolo[3.,4-b]pyridi n-

5-yl)oxy)methyl)phenyl) -5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 78)

Compound 78

Synthesis of 78-a: ethyl N-(4-chloro-2-fluoro-3 -(((3 -methyl- l-(tetrahy dro-2H-pyran-2-yl)-

IH-pyrazolo [3,4-b1pyridin-5-yl)oxy)methyl)phenyl)-5-fluoro-2-methoxypyr idine-3- sulfonamide

[0545] To a stirred solution of 4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (250 mg, 0.64 mmol, 1 equiv) in pyridine (2.5 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (288 mg, 1.28 mmol, 2 equiv) in portions at room temperature. The resulting mixture was stirred 1 h at room temperature. The resulting mixture was diluted with water (2 mL). The resulting mixture was extracted with DCM (3 x 10 mL). The combined organic layers were washed with water (2x 10 mL) and brine (10 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EtOAc/PE (0-50%) to afford N-[4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (186 mg, 50%) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 580.

Synthesis of Compound 78: N-(4-chloro-2-fluoro-3-(((3-methyl-1H-pyrazolo[3,4-b1pyridin - 5-yl)oxy)methyl)phenyl) -5-fluoro-2-methoxypyridine-3-sulfonamide

[0546] A solution of N-[4-chloro-2-fluoro-3-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4 - b]pyridin-5-yl] oxy]methyl) phenyl] -5-fluoro-2-methoxypyridine-3-sulfonamide (176 mg, 0.3 mmol, 1 equiv) in 4M HC1 in MeOH (4 mL) was stirred for Ih at room temperature. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep- HPLC with the following conditions (Column: Sunfire Prep C 18 OBD Column, 50*250 mm, 5 μm, 10 nm; Mobile Phase A:Water(0.05% NH3H 2 O), Mobile Phase B:ACN; Flow rate: 90 mL/min; Gradient: 5 B to 45 B in 15 min; ) to afford N-[4-chloro-2-fluoro-3-[([3-methyl-lH- pyrazolo[3,4-b]pyri din-5 -yl]oxy)methyl]phenyl] -5-fluoro-2-methoxypyridine-3-sulfonamide (78 mg, 52%) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 496.

'H NMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, IH), 10.09 (br, IH) δ.42 (s, IH), 8.20 (s, IH), 8.02-7.99 (m, IH), 7.89-7.88 (m, IH), 7.04-7.31 (m, 2H), 5.16 (s, 2H), 3.88 (s, 3H), 2.46 (s, 3H).

Example 90: Synthesis of 5-chloro-N-[3-chloro-5-fluoro-4-[([3-methyl-1H-pyrazolo[3.,4 - b]pyridin-5-yl]oxy)methyl]pyridin-2-yl]-2-methoxypyridine-3- sulfonamide (Compound 79)

79-c Compound 79

Synthesis of 79-a: 3-chloro-N-l(2,4-dimethoxyphenyl)methyl1-5-fluoro-4-([[ methyl-l-

(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)pyridin -2-amine

[0547] To a stirred solution of 2,3-dichloro-5-fluoro-4-([[3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)pyridine (500 mg, 1.2 mmol, 1 equiv) in THF (30 mL) was added 1 -(3, 4-dimethylphenyl)m ethanamine (246 mg, 1.8 mmol, 1.5 equiv), Pd(0Ac)2 (55 mg, 0.2 mmol, 0.2 equiv), BINAP (152 mg, 0.2 mmol, 0.2 equiv) and Z-BuONa (234 mg, 2.4 mmol, 2 equiv). The reaction was stirred for 2 h at 85 °C under nitrogen atmosphere, then cooled and diluted with H 2 O (10 mL). The resulting mixture was extracted with ethyl acetate (4 x 10 mL). The combined organics were washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by Flash- Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40 μm, 120 g; mobile phase: 40-80% MeCN / 0.1% aqueous formic acid; to afford 3-chloro- N-[(2,4-dimethoxyphenyl)methyl]-5-fluoro-4-([[3 -methyl- l-(oxan-2-yl)pyrazolo[3, 4- b]pyridin-5-yl]oxy]methyl)pyridin-2-amine (362 mg, 55% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 542

Synthesis of 79-b: 3-chloro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1 pyridin-5- yl1oxy1methyl)pyridin-2-amine

[0548] To a stirred solution of 3-chloro-N-[(2,4-dimethoxyphenyl)methyl]-5-fluoro-4-([[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)p yridin-2-amine (100 mg, 0.2 mmol, 1 equiv) in DCM (4 mL) was added DDQ (84 mg, 0.4 mmol, 2 equiv). The resulting solution was stirred for 2 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 3-chloro-5-fluoro-4-([[3- methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)p yridin-2-amine (50 mg, 69% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 392

Synthesis of 79-c: 5-chloro-N-[3-chloro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyr azolo[3,4- b1pyridin-5-yl1oxy1methyl)pyridin-2-yl1-2-methoxypyridine-3- sulfonamide

[0549] 3-Chloro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b] pyridin-5- yl]°xy]methyl)pyridin-2-amine (40 mg, 0.1 mmol, 1 equiv) in pyridine (2 mL) was treated with 5-chloro-2-methoxypyridine-3-sulfonyl chloride (74 mg, 0.3 mmol, 3 equiv). The resulting solution was stirred for 36 h at 80 °C then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 50-80% MeCN / 0.1% aqueous formic acid; to afford 5- chloro-N-[3-chloro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyraz olo[3,4-b]pyri din-5- yl]°xy]methyl)pyridin-2-yl]-2-methoxypyridine-3-sulfonamide (6 mg, 10% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 597 Synthesis of Compound 79: 5-chloro-N-[3-chloro-5-fluoro-4-[([3-methyl-1H-pyrazolol3,4- b1pyridin-5-yl1oxy)methyl1pyridin-2-yl1-2-methoxypyridine-3- sulfonamide

[0550] 5-Chloro-N-[3 -chi oro-5-fluoro-4-([[3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyri din-5- yl]oxy]methyl)pyridin-2-yl]-2-methoxypyridine-3-sulfonamide (6 mg, 0.01 mmol, 1 equiv) was treated with 4 M HC1 in MeOH (1 mL) at room temperature. The resulting solution was stirred for 30 min then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 38-58% MeCN / 0.1% aqueous formic acid; to afford 5-chloro-N-[3-chloro-5-fluoro-4-[([3- methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]pyridin-2-y l]-2-methoxypyridine-3- sulfonamide (2 mg, 39% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 513

1 H NMR (300 MHz, Methanol-^) δ 8.29 (m, 3H), 8.03 (s, 1H), 7.87 (d, J= 2.7 Hz, 1H), 5.33 (d, J= 1.7 Hz, 2H), 3.99 (s, 3H), 2.57 (s, 3H).

Example 91: Synthesis of N-[2,4-difluoro-3-([[3-(2.,2.,2-trifluoroethyl)-1H-pyrazolo[ 3.,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (Compound 80)

Synthesis of 80-a: methyl 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolol3,4- blpyridine-3 -carboxylate

[0551] To a stirred solution of 5-bromo-3-iodo-l-[[2-

(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine 5-bromo-3-iodo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (3 g, 6.6 mmol, 1 equiv) and TEA (2 g, 20 mmol, 3 equiv) in MeOH (30 mL) was added Pd(dppf)C12 (0.48 g, 0.66 mmol, 0.1 equiv) in portions. The reaction was stirred for 2.5 h at 70 °C under 20 atm CO pressure, then concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (20: l) to afford methyl 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridine-3 -carboxylate (2.4 g, 94% yield) as an oil.

LCMS (ES, m/z): [M+H] + : 386 Synthesis of 80-b: 5-bromo-l-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazolo[3,4 - blpyridine-3 -carboxylic acid

[0552] To a stirred solution of methyl 5-bromo-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-3-carb oxylate (2.2 g, 5.8 mmol, 1 equiv) in MeOH (20 mL) was added LiOH.H 2 O (1.2 g, 29 mmol, 5 equiv) in H 2 O (2 mL) dropwise at 0 °C. The resulting mixture was stirred for 2 h then diluted with H 2 O (40 mL) and EA (20 mL). The mixture was acidified to pH 5 with AcOH, and extracted with EA (3 x 20 mL). The combined organics were washed with brine (40 mL), dried over anhydrous Na 2 SO 4 and concentrated to afford crude 5 -bromo- 1 -((2 -trimethyl silyl)ethoxy)m ethyl)- 1H- pyrazolo[3,4-b]pyridine-3 -carboxylic acid (2.1 g, 97% yield) as a white solid which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 372

Synthesis of 80-c: 5-bromo-N-methoxy-N-methyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine-3-carb oxamide [0553] To a stirred solution of 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridine-3 -carboxylic acid (2.2 g, 5.9 mmol, 1 equiv) and N,O-dimethylhydroxylamine (0.39 g, 6.4 mmol, 1.1 equiv) in DCM (40 mL) were added HATU (2.7 g, 7 mmol, 1.2 equiv) and DIEA (2.3 g, 17.6 mmol, 3 equiv). The resulting mixture was stirred for 2 h then diluted with H 2 O (50 mL) and extracted with DCM (3 x 20 mL). The combined organics were washed with brine (50 mL), dried over anhydrous ISfeSCL and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (10: 1) to afford 5-bromo- N-methoxy-N-methyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyraz olo[3,4-b]pyridine-3- carboxamide (2.24 g, 92% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 415

Synthesis of 80-d: 5-bromo-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1p yridine-3- carbaldehyde

[0554] To a stirred solution of 5-bromo-N-methoxy-N-methyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine-3-carb oxamide (1 g, 2.4 mmol, 1 equiv) in THF (10 mL) was added LAH (0.11 g, 2.9 mmol, 1.2 equiv) in portions at 0 °C. The resulting mixture was stirred for 1 h at 0 °C. The reaction was quenched by the addition of sat. NaHSCL (aq.) (30 mL) at 0 °C, then extracted with EtOAc (3 x 20 mL). The combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EtOAc (20: 1) to afford 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]p yridine-3- carbaldehyde (703 mg, 82% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 356

Synthesis of 80-e: 5-bromo-3-(2,2,2-trifluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine

[0555] To a stirred solution of 5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridine-3 -carbaldehyde (1.3 g, 3.7 mmol, 1 equiv) in DMF (10 mL) was added difluoro(triphenylphosphaniumyl)acetate (2.6 g, 7.3 mmol, 2 equiv) in portions at room temperature. The resulting mixture was stirred for 80 min at 60 °C. The mixture was allowed to cool to room temperature and TBAF (5.6 mL, 11 mmol, 3 equiv) was added dropwise over 1 min at room temperature. The resulting mixture was stirred for additional 45 min at 60 °C, then cooled and diluted with H 2 O (50 mL), then extracted with EtOAc (3 x 10 mL). The combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50-95% MeCN / 0.1% aqueous formic acid; to afford 5-bromo-3-(2,2,2-trifluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (520 mg, 35% yield) as a yellow oil. LCMS (ES, m/z): [M+H] + : 410

Synthesis of 80-f: 5-(4A5,5-tetramethyl-L3,2-dioxaborolan-2-yl)-3-(2,2,2-triflu oroethyl)-l- [[2-(trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine

[0556] To a stirred solution of 5-bromo-3-(2,2,2-trifluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (495 mg, 1.2 mmol, 1 equiv) and bis(pinacolato)diboron (337 mg, 1.3 mmol, 1.1 equiv) in dioxane (6 mL) were added KO Ac (237 mg, 2.4 mmol, 2 equiv) and Pd(dppf)C12 (88 mg, 0.12 mmol, 0.1 equiv) in portions, at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 90 °C then concentrated to afford crude 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3- (2,2,2-trifluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]p yrazolo[3,4-b]pyridine (550 mg) as a brown oil which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 458

Synthesis of 80-g: 3-(2,2,2-trifluoroethyl)-l-[[2-(trimethylsilyl)ethoxy1methyl 1pyrazolo[3,4- blpyridin-5-ol

[0557] To 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3-(2,2,2-tri fluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl] pyrazolo[3,4-b]pyridine (495 mg, 1.1 mmol, 1 equiv) in THF (5 mL) was added 2% aqueous NaOH solution (5 mL) and 30% H 2 O 2 (368 mg, 10.82 mmol, 10 equiv). The reaction was stirred for 1 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40 μm, 120 g; mobile phase: 30-75% MeCN / 0.1% aqueous formic acid; to afford 3- (2,2,2-trifluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]p yrazolo[3,4-b]pyridin-5-ol (207 mg, 55% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 348

Synthesis of 80-h: 2,4-difluoro-3-([[3-(2,2,2-trifluoroethyl)-l-[[2- (trimethylsilyl)ethoxylmethyllpyrazolo [3,4-b1pyridin-5-yl1oxy1methyl)aniline [0558] To a stirred solution of 3-(2,2,2-trifhioroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (200 mg, 0.58 mmol, 1 equiv) and (3-amino-2,6-difluorophenyl)methanol (110 mg, 0.69 mmol, 1.2 equiv) in DCM (2 mL) were added TMAD (149 mg, 0.86 mmol, 1.5 equiv) and PPh 3 (227 mg, 0.86 mmol, 1.5 equiv). The resulting mixture was stirred for 5 h then concentrated. The residue was purified by Flash- Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40 μm, 120 g; mobile phase: 40-90% MeCN / 0.1% aqueous formic acid; to afford 2,4- difluoro-3-([[3-(2,2,2-trifluoroethyl)-l-[[2-(trimethylsilyl )ethoxy]methyl]pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)aniline (192 mg, 68% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 489

Synthesis of 80-i: N-[2,4-difluoro-3-(ri3-(2,2,2-trifluoroethyl)-l-[[2- (trimethylsilyl)ethoxylmethyllpyrazolo [3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0559] To a stirred solution of 2,4-difluoro-3-([[3-(2,2,2-trifhioroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)aniline (187 mg, 0.38 mmol, 1 equiv) in pyridine (4 mL) was added 5 -fluoro-2-methoxypyridine-3 -sulfonyl chloride (130 mg, 0.57 mmol, 1.5 equiv) in DCM (0.5 mL). The resulting mixture was stirred for 2 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 50- 95% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-([[3-(2,2,2- trifluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo [3,4-b]pyridin-5- yl]°xy]methyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonam ide (249 mg, 96% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 678

Synthesis of Compound 80: N-[2.4-difluoro-3-([[ 3-(2.2.2-trifluoroethyl )- 1 H-pyrazolo[2,4- b]pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2-methoxypyridine -3-sulfonamide [0560] A solution of N-[2,4-difluoro-3-([[3-(2,2,2-trifluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (256 mg, 0.38 mmol, 1 equiv) in TFA (1 mL) and DCM (3 mL) was stirred for 1 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 30-60% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-([[3-(2,2,2- trifluoroethyl)-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)ph enyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (92 mg, 44% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 548

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.66 (s, 1H), 10.37 (s, 1H), 8.45 (d, J= 3.0 Hz, 1H), 8.26 (d, J= 2.8 Hz, 1H), 8.02 -7.90 (m, 2H), 7.38 (td, J= 8.9, 5.9 Hz, 1H), 7.22-7.10 (m, 1H), 5.13 (s, 2H), 4.05 (d, J= 11.3 Hz, 1H), 3.98 (d, J= 11.6 Hz, 1H), 3.91 (s, 3H).

Example 92: Synthesis of N-[3-([[3-(l,l-difluoroethyl)-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy]methyl)-2.,4-difluorophenyl]-5-fluoro-2-methoxypyridi ne-3-sulfonamide (Compound 81)

81 -f Compound 81 Synthesis of 81-a: 5-bromo-3-(l-ethoxyethenyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine [0561] To 5-bromo-3-iodo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[ 3,4-b]pyridine (400 mg, 0.88 mmol, 1 equiv) in toluene (10 mL) was added tributyl(1-ethoxyethenyl)stannane (318 mg, 0.88 mmol, 1 equiv) and Pd(PPh3)4 (1 mg, 0.044 mmol, 0.05 equiv). The resulting solution was stirred for 12 h at 100 °C then concentrated. The residue was applied onto a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 10) to give 5-bromo-3-(1- ethoxyethenyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[ 3,4-b]pyridine (300 mg, 77% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 398

Synthesis of 81-b: l-(5-bromo-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazolo[3,4- b1pyridin-3- yl ethanone

[0562] 5 -Bromo-3-(l -ethoxy ethenyl)- l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3, 4- b]pyridine (300 mg, 0.75 mmol, 1 equiv) in THF (5 mL) was treated with 2 M HC1 (2 mL). The resulting solution was stirred for 1 h, then the pH was adjusted to 7~8 with NaHCO 3 /H 2 O. H 2 O (20 ml) and the mixture extracted with dichloromethane (2x20 mL). The combined organics were washed with brine (30 ml), dried over anhydrous sodium sulfate and concentrated to give l-(5-bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridin- 3-yl)ethanone (260 mg, 84% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 370

Synthesis of 81-c: 5-bromo-3-(1,1-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridine [0563] l-(5-Bromo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyri din-3- yl)ethanone (500 mg, 1.4 mmol, 1 equiv) was treated with DAST (3 mL) and the solution was stirred for 4 days at room temperature. The reaction was quenched with water/ice (50 mL), and the pH adjusted to 7~8 with NaHCO 3 /H 2 O. The resulting solution was extracted with dichloromethane (3 x 20 mL), and the combined organic layers were dried over anhydrous sodium sulfate then concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 10). 5-Bromo-3-(l,l-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (120 mg, 22% yield) was obtained as a colorless oil.

LCMS (ES, m/z): [M+H] + : 392. Synthesis of 81-d: 3-(Ll-difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy1methyl1pyr azolo[3,4- blpyridin-5-ol

[0564] 5 -Bromo-3-(l,l-difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]me thyl]pyrazolo[3,4- b]pyridine (120 mg, 0.31 mmol, 1 equiv) in dioxane (5 mL) was treated with Pd(dppf)C12 (45 mg, 0.062 mmol, 0.2 equiv), KOAc (90 mg, 0.92 mmol, 3 equiv) and fhPim (117 mg, 0.46 mmol, 1.5 equiv). The solution was stirred for 2 h at 85 °C then cooled to room temperature. 2% aqueous NaOH solution (10 mL) was added, followed by 30% H 2 O 2 (1 mL). The resulting solution was stirred for 10 min at room temperature, then diluted with H 2 O (30 mL). The mixture was extracted with dichloromethane (3x20 mL) and the combined organics were dried over anhydrous sodium sulfate, then concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/PE (1:4) to give 3-(l,l-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (80 mg) as a colorless oil.

LCMS (ES, m/z): [M+H] + : 330

Synthesis of 81-e: 3-([[3-(1,1-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)-2,4-difluoroaniline [0565] To 3-(l,l-difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]py razolo[3,4-b]pyridin- 5-ol (80 mg, 0.24 mmol, 1 equiv) in DCM (5 mL) was added (3-amino-2,6- difluorophenyl)methanol (58 mg, 0.36 mmol, 1.5 equiv), TMAD (63 mg, 0.36 mmol, 1.5 equiv) and PPI13 (96 mg, 0.36 mmol, 1.5 equiv). The reaction was stirred for 5 h then concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, C18 silica gel; mobile phase: 20% MeCN / H 2 O; to afford 3 -([[3 -(1 , 1 - difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[ 3,4-b]pyridin-5-yl]oxy]methyl)- 2,4-difluoroaniline (80 mg, 63% yield) as a colorless oil.

LCMS (ES, m/z): [M+H] + : 471

Synthesis of 81-f: N-[3-([[3-(1,1-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl1ox y1methyl)-2,4-difluorophenyl1- 5-fluoro-2-methoxypyridine-3-sulfonamide

[0566] 3 -([[3 -(1,1 -Difluoroethyl)- 1 -[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3 ,4- b]pyridin-5-yl]oxy]methyl)-2,4-difluoroaniline (80 mg, 0.17 mmol, 1 equiv) in DCM (3 mL) was treated with pyridine (0.5 mL) and 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (57.5 mg, 0.26 mmol, 1.5 equiv). The resulting solution was stirred for 4 h at 45 °C then concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1:4) to give N-[3-([[3-(l,l-difluoroethyl)-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl]ox y]methyl)-2,4-difluorophenyl]- 5-fluoro-2-methoxypyridine-3-sulfonamide (90 mg, 72% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 660

Synthesis of Compound 81 : N-13-(113-(1,1-difluoroethyl)-lEl-pyrazolo[3,4-b1pyridin-5- yl1oxy1methyl)-2,4-difluorophenyl1-5-fluoro-2-methoxypyridin e-3-sulfonamide

[0567] N-[3-([[3-(l,l-Difluoroethyl)-l-[[2-(trimethylsilyl)ethoxy]m ethyl]pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)-2,4-difluorophenyl]-5-fluoro-2-me thoxypyridine-3-sulfonamide (90 mg, 0.14 mmol, 1 equiv) in DCM (2 mL) was treated with trifluoroacetic acid (2 mL). The resulting solution was stirred for 30 min at 50 °C then concentrated. The residue was applied to a silica gel column, eluting with ethyl acetate/petroleum ether (1 : 1) to afford N-[3- ([[3-(l,l-difluoroethyl)-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy] methyl)-2,4-difluorophenyl]-5- fluoro-2-methoxypyridine-3 -sulfonamide (21 mg, 29% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 530

1 H NMR (300 MHz, DMSO-d 6 ) δ 14.03 (s, 1H), 10.37 (s, 1H), 8.43 (d, J= 3.0 Hz, 1H), 8.32 (d, J = 2.7 Hz, 1H), 7.97 (dd, J = 7.3, 3.0 Hz, 1H), 7.82 (d, J= 2.8 Hz, 1H), 7.45-7.31 (m, 1H), 7.17 (t, J= 9.2 Hz, 1H), 5.20 (s, 2H), 3.90 (s, 3H), 2.17 (t, J= 18.9 Hz, 3H).

Example 93: Synthesis of N-[2,4-difluoro-3-[([5-methylimidazo[l.,5-b]pyridazin-3- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (Compound 82)

Synthesis of 82-a: 3-(l-ethoxyvinyl)-5-methoxypyridazine

[0568] To a solution of 3-chloro-5-methoxypyridazine (5 g, 35 mmol, 1 equiv) and ethyl tributyl stannanecarboxyl ate (18.8 g, 52 mmol, 1.5 equiv) in toluene (50 mb) was added Pd(PPh 3 ) 4 (4 g, 3.5 mmol, 0.1 equiv) under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 110 °C under nitrogen atmosphere, then concentrated. The residue was purified by silica gel column chromatography, eluting with EA/PE (3/2) to afford 3-(1- ethoxyvinyl)-5-methoxypyridazine (3.6 g, 57% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 181. Synthesis of 82-b: l-(5-methoxypyridazin-3-yl)ethanone

[0569] To a solution of 3-(1-ethoxyethenyl)-5-methoxypyridazine (2.4 g, 13 mmol, 1 equiv) in acetone (20 mL) was added 3 M aqueous HC1 (5 mL). The mixture was stirred for 4 h at room temperature, then neutralized to pH 7 with NaHCO, solution. The resulting mixture was extracted with EA (3 x 30 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PEZEA (1/1) to afford l-(5-methoxypyridazin-3- yl)ethanone (1.48 g, 73% yield) as a light yellow oil.

LCMS (ES, m/z): [M+H] + : 153.

Synthesis of 82-c: l-(5-methoxypyridazin-3-yl)ethan-l-amine

[0570] A mixture of l-(5-methoxypyridazin-3-yl)ethanone (1.3 g, 8.5 mmol, 1 equiv), NH4OAC (6.6 g, 85 mmol, 10 equiv) and NaBHsCN (0.54 g, 8.5 mmol, 1 equiv) in MeOH (13 mL) was stirred for 16 h at room temperature. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 10 mL). The filtrate was concentrated to give crude l-(5-methoxypyridazin-3-yl)ethanamine (1.3 g).

LCMS (ES, m/z): [M+H] + : 154.

Synthesis of 82-d: N-(l-(5-methoxypyridazin-3-yl)ethyl)formamide

[0571] A solution of l-(5-methoxypyridazin-3-yl)ethanamine (1.3 g, crude) in HCOOH (13 mL) was stirred for 4 h at 100 °C. The mixture was allowed to cool and was concentrated to give crude N-[l-(5-methoxypyridazin-3-yl)ethyl]formamide (1.4 g).

LCMS (ES, m/z): [M+H] + : 182.

Synthesis of 82-e: 3-methoxy-5-methylimidazo[L5-b1pyridazine

[0572] To a solution of N-[l-(5-methoxypyridazin-3-yl)ethyl]formamide (1.4 g, crude) in toluene (5 mL) was added POCL (1.2 g, 7.7 mmol, 1 equiv). After stirring for 4 h at 100 °C under nitrogen atmosphere, the resulting mixture was concentrated. The residue was purified by silica gel column chromatography, eluting with PEZEA (1/1) to afford 3-methoxy-5- methylimidazo[l,5-b]pyridazine (231 mg, 16% yield over 3 steps) as a light yellow solid. LCMS (ES, m/z): [M+H] + : 164.

Synthesis of 82-f: 5-methylimidazo[L5-b1pyridazin-3-ol

[0573] A solution of 3-methoxy-5-methylimidazo[l,5-b]pyridazine (231 mg, 1.4 mmol, 1 equiv) in DCE (2 mL) was treated with 1 M BBr 3 in DCM (8.4 mL, 6 equiv) and the solution stirred for 16 h at 60 °C. The reaction was concentrated, and the residue basified to pH 8 with aq. NaHCCL. The resulting mixture was extracted with EA (3 x 10 mL). The combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with EAto afford 5- methylimidazo[l,5-b]pyridazin-3-ol (110 mg, 52% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 150.

Synthesis of 82-g: 2-(bromomethyl)-E3-difluoro-4-nitrobenzene

[0574] A solution of (2, 6-difluoro-3-nitrophenyl)m ethanol (100 mg, 0.53 mmol, 1 equiv) and PPh 3 (152 mg, 0.58 mmol, 1.1 equiv) in THF (1 mL) was treated with CBn (263 mg, 0.794 mmol, 1.5 equiv) in portions at 0 °C, then stirred for 1 h at room temperature. The resulting mixture was diluted with EA (20 mL), and the organics were washed with water and brine, dried over anhydrous ISfeSCE and concentrated. The residue was purified by silica gel column chromatography, eluting with PE/EA(5/1) to afford 2-(bromom ethyl)- 1,3 -difluoro-4- nitrobenzene (193 mg, 72% yield) as a light yellow oil.

Synthesis of 82-h: 3-((2,6-difluoro-3-nitrobenzyl)oxy)-5-methylimidazo[L5-b]pyr idazine [0575] To a mixture of 5-methylimidazo[l,5-b]pyridazin-3-ol (50 mg, 0.3 mmol, 1 equiv) and K 2 CO 3 (46 mg, 0.3 mmol, 1 equiv) in DMF (1 mL) was added 2-(brom om ethyl)- 1,3- difluoro-4-nitrobenzene (84 mg, 0.3 mmol, 1 equiv) in DMF (1 ml) dropwise. The reaction was stirred for 1 h at 40°C, then diluted with EA (30 mL). The organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with EA/PE (3/2) to afford 3-[(2,6-difluoro-3- nitrophenyl)methoxy]-5-methylimidazo[l,5-b]pyridazine (68 mg, 63% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + :321.

Synthesis of 82-i: 2,4-difluoro-3-[([5-methylimidazo[1,5-b1pyridazin-3-yl1oxy)m ethyl1aniline [0576] To a solution of 3-[(2,6-difluoro-3-nitrophenyl)methoxy]-5-methylimidazo[l,5- b]pyridazine (63 mg, 0.2 mmol, 1 equiv) in EA (1 mL) was added 10% Pd/C (6 mg) under nitrogen atmosphere. The mixture was hydrogenated for 16 h under a hydrogen balloon, then filtered through a Celite pad. The filtrate was concentrated to give 2,4-difluoro-3-[([5- methylimidazo[l,5-b] pyridazin-3-yl]oxy)methyl]aniline (33 mg, 58% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + :291.

Synthesis of Compound 82: N-[2,4-difluoro-3-[([5-methylimidazo[l,5-b1pyridazin-3- yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0577] To 2,4-difluoro-3-[([5-methylimidazo[l,5-b]pyridazin-3-yl]oxy)m ethyl]aniline (30 mg, 0.1 mmol, 1 equiv) in pyridine (1 mL) was added 5-fluoro-2-methoxypyridine-3- sulfonyl chloride (117 mg, 0.5 mmol, 5 equiv). This was stirred for 0.5 h at room temperature, then diluted with EA (20 mL). The organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The crude product was purified by Prep- HPLC with the following conditions: Column: XBridge Prep C 18 OBD Column, 5 μm ,19*150 mm; Mobile Phase: 20-36% MeCN / 10 mM aqueous NH4HCO3; to afford N- [2,4-difluoro-3-[([5-methylimidazo[l,5-b]pyridazin-3-yl]oxy) methyl]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (18 mg, 36% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 515.

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.40 (s, 1H), 8.47-8.36 (m, 2H), 8.04-7.94 (m, 2H), 7.58 (d, J= 2.9 Hz, 1H), 7.47-7.33 (m, 1H), 7.17 (t, J= 9.0 Hz, 1H), 5.11 (s, 2H), 3.90 (s, 3H), 2.37 (s, 3H).

Example 94: Synthesis of N-[2,4-difluoro-3-([5-(trifluoromethyl)imidazo[1,5- b]pyridazin-3-yl]oxy]methyl) phenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (Compound 83) Synthesis of 83-a: 5-methoxypyridazine-3-carbonitrile

[0578] A mixture of 3-chloro-5-methoxypyridazine (2 g, 14 mmol, 1 equiv), Zn(CN)2 (3.3 g, 28 mmol, 2 equiv), Pd2(dba) 3 (1.2 g, 1.4 mmol, 0.1 equiv) and dppf (1.5 g, 2.8 mmol, 0.2 equiv) in DMF (20 mL) was stirred overnight at 100 °C under nitrogen atmosphere. The mixture was cooled and diluted with water (50 mL). This was extracted with EtOAc (3 x 200 mL), and the combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-50% EtOAc/PE to afford 5 -methoxypyridazine-3 -carbonitrile (1.1 g, 59% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 136.

Synthesis of 83-b: l-(5-methoxypyridazin-3-yl)methanamine

[0579] To a solution of 5-methoxypyridazine-3-carbonitrile (1.1 g, 8.1 mmol, 1 equiv) in MeOH (11 mL) and 4 M HC1 in MeOH (0.3 mL, 1.2 mmol, 0.15 equiv) was added 10% Pd/C (110 mg) under nitrogen atmosphere. The mixture was hydrogenated for 2 h under a hydrogen balloon, then was filtered through a Celite pad. The filtrate was concentrated to give crude l-(5-methoxypyridazin-3-yl)methanamine (1.1 g).

LCMS (ES, m/z): [M+H] + : 140.

Synthesis of 83-c: N-[(5-methoxypyridazin-3-yl)methyl1formamide

[0580] A solution of l-(5-methoxypyridazin-3-yl)methanamine (1.1 g, 7.9 mmol, 1 equiv) in HCOOH (11 mL) was stirred overnight at 80 °C. The resulting mixture was concentrated. The 1.3 g N-[(5-methoxypyridazin-3-yl)methyl]formamide crude product was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 168.

Synthesis of 83-d: 3-methoxyimidazo[1,5-b]pyridazine

[0581] To a stirred solution of N-[(5-methoxypyridazin-3-yl)methyl]formamide (1.3 g, 7.7 mmol, 1 equiv) in toluene (13 mL) was added phosphorus oxychloride (3.6 g, 23 mmol, 3 equiv) dropwise at 0-5 °C. The reaction was stirred for 2 h at 100 °C then was cooled and concentrated. The reaction was quenched by the addition of water/ice (10 mL) and extracted with EtOAc (3 x 50 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-100% EtOAc/PE to afford 3-methoxyimidazo[l,5- b]pyridazine (300 mg, 26% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 150. Synthesis of 83-e: 3-methoxy-5-(trifluoromethyl)imidazo[1,5-b]pyridazine

[0582] To a stirred solution of 3-methoxyimidazo[l,5-b]pyridazine (1 g, 6.7 mmol, 1 equiv) and trifluoromethanesulfmyl zinc (3.7 g, 20 mmol, 3 equiv) in DCM (40 mL) and H 2 O (20 mL) was added 70% TBHP in H 2 O (4.3 g, 34 mmol, 5 equiv) dropwise at 0-5 °C under nitrogen atmosphere. The reaction was stirred for 40 h at room temperature then diluted with water (20 mL). The resulting mixture was extracted with DCM (3 x 50 mL). The combined organics were washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with 0-100% EtOAc/PE to afford 3-methoxy-5-(trifluoromethyl)imidazo[l,5-b]pyridazine (230 mg, 16% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 218.

Synthesis of 83-f: 5-(trifluoromethyl)imidazo[1,5-b]pyridazin-3-ol

[0583] 3-Methoxy-5-(trifluoromethyl)imidazo[l,5-b]pyridazine (150 mg, 0.69 mmol, 1 equiv) was treated with 1 M BBr 3 in DCM (4 mL, 4 mmol, 5.8 equiv) and stirred for 40 h at room temperature. The resulting mixture was concentrated. The 5- (trifluoromethyl)imidazo[l,5-b]pyridazin-3-ol (100 mg) crude product was used in the next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 204.

Synthesis of 83-g: 3-((2,6-difluoro-3-nitrobenzyl)oxy)-5-(trifluoromethyl)imida zo[L5- blpyridazine

[0584] To a stirred mixture of 5-(trifluoromethyl)imidazo[l,5-b]pyridazin-3-ol (50 mg, 0.1 mmol, 1 equiv) and 2-(bromomethyl)-l,3-difluoro-4-nitrobenzene (32 mg, 0.12 mmol, 1.2 equiv) in DMF (1 mL) was added K2CO3 (29 mg, 0.2 mmol, 2 equiv). The reaction was stirred for 4 h at 40 °C, then cooled and diluted with EA (20mL). The combined organics were washed with water and brine, dried over anhydrous NajSCL and concentrated. The residue was purified by silica gel column chromatography, eluting with PEZEA (3/1) to afford 3-[(2,6-difluoro-3-nitrophenyl)methoxy]-5-(trifluoromethyl)i midazo[l,5-b]pyridazine (13 mg, 33% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + :375

Synthesis of 83-h: 3-((2,6-difluoro-3-nitrobenzyl)oxy)-5-methylimidazo[L5-b]pyr idazine [0585] To a stirred mixture of 3-[(2,6-difluoro-3-nitrophenyl)methoxy]-5- (trifhioromethyl)imidazo[l,5-b]pyridazine (26 mg, 0.07 mmol, 1 equiv) and Fe (12 mg, 0.21 mmol, 3 equiv) in EtOH (1 mL) and H 2 O (0.25 mL) was added NH4CI (11 mg, 0.21 mmol, 3 equiv) at room temperature. The resulting mixture was stirred for 2 h at 60 °C, then cooled and filtered. The filter cake was washed with EA (2 x 20 mL), and the combined filtrates washed with water and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with EAto afford 2,4-difluoro-3- ([[5-(trifluoromethyl)imidazo[l,5-b]pyridazin-3-yl]oxy]methy l)aniline (26 mg, 79% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + :345.

Synthesis of Compound 83: N-[2,4-difluoro-3-([[5-(trifluoromethyl)imidazo[1,5-b]pyrida zin- 3-ylloxylmethyl) phenyl1-5-fluoro-2-methoxypyridine-3-sulfonamide

[0586] To a solution of 2,4-difluoro-3-([[5-(trifluoromethyl)imidazo[l,5-b]pyridazin -3- yl]°xy]methyl)aniline (20 mg, 0.058 mmol, 1 equiv) in pyridine (0.2 mL) was added 5- fluoro-2-methoxypyridine-3-sulfonyl chloride (52 mg, 0.23 mmol, 4 equiv) and the solution was stirred for 0.5 h at room temperature. The resulting mixture was diluted with EA (20mL), and washed with aq NH4CI, water and brine. The organics were dried over anhydrous Na 2 SO 4 and concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column: XBridge Prep C 18 OBD Column, 5 μm, 19*150 mm; Mobile Phase: 28- 49% MeCN / 0.05% aqueous ammonia; to afford N-[2,4-difluoro-3-([[5- (trifluoromethyl)imidazo[l,5-b]pyridazin-3-yl]oxy]methyl) phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (15 mg, 49% yield) as a white solid .

LCMS (ES, m/z): [M+H] + : 534.

1 H NMR (300 MHz, DMSO-d 6 ) δ 10.41 (s, 1H), 8.75 (s, 1H), 8.41 (d, J = 3.0 Hz, 1H), 8.35 (d, J= 2.7 Hz, 1H), 7.98 (dd, J= 7.4, 3.0 Hz, 1H), 7.56 (d, J= 2.7 Hz, 1H), 7.40 (td, J= 9.0, 6.0 Hz, 1H), 7.16 (t, J= 9.1 Hz, 1H), 5.26 (s, 2H), 3.89 (s, 3H).

Example 95: Synthesis of N-[2,4-difluoro-3-[([3-phenyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (Compound 84)

Synthesis of 84-a: 5-bromo-3-phenyl-l-[[2(trimethylsilyl)ethoxy1methyl1pyrazolo [3,4- blpyridine

[0587] To a solution of 5-bromo-3-iodo-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[ 3,4- b]pyridine (800 mg, 1.8 mmol, 1 equiv) and phenyl boronic acid (215 mg, 1.8 mmol, 1 equiv) in dioxane (15 mL) and H 2 O (3 mL) were added Na 2 CO 3 (373 mg, 3.5 mmol, 2 equiv) and Pd(dppf)C12 (129 mg, 0.17 mmol, 0.1 equiv). After stirring for 3 h at 80 °C under nitrogen atmosphere, the resulting mixture was concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (10: 1) to afford 5-bromo-3-phenyl-l-[[2 (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (420 mg, 59% yield) as a yellow oil. LCMS (ES, m/z): [M+H] + : 404

Synthesis of 84 b/c: 3-phenyl-l-112-(trimethylsilyl) ethoxy1methyl1pyrazolo[3,4-b1pyridin-5- ol

[0588] To a solution of 5-bromo-3-phenyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazol o[3,4- b]pyridine (400 mg, 1 mmol, 1 equiv) and bis(pinacolato)diboron (502 mg, 2 mmol, 2 equiv) in dioxane (10 mL) were added KO Ac (194 mg, 2 mmol, 2 equiv) and Pd(dppf)C12 (72 mg, 0.1 mmol, 0.1 equiv). The reaction was stirred for 3 h at 100 °C under a nitrogen atmosphere, then cooled and diluted with THF (10 mL) and 2% aqueous NaOH solution (10 mL). 30% H 2 O 2 (151 mg, 4.4 mmol, 5 equiv) was added dropwise, and the reaction stirred at room temperature for 1 hr. The mixture was quenched with saturated aqueous Na2S2O3 solution (10 mL) and extracted with EA (3 x 20 mL). The combined organics were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (4: 1) to afford 3-phenyl-l-[[2-(trimethylsilyl) ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (260 mg, 86% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 342

Synthesis of 84-d: 2,4-difluoro-3-[[(3-phenyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5-yl)ox y1methyl1aniline [0589] To a stirred solution of 3-phenyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4- b]pyridin-5-ol (400 mg, 1.2 mmol, 1 equiv) and (3-amino-2,6-difluorophenyl)methanol (186 mg, 1.2 mmol, 1 equiv) in DCM (20 mL) were added TMAD (303 mg, 1.8 mmol, 1.5 equiv) and PPh 3 (461 mg, 1.7 mmol, 1.5 equiv). The reaction was stirred for 1 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30- 80% MeCN / 0.1% aqueous formic acid; to afford 2,4-difluoro-3-[[(3-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (380 mg, 67% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 483

Synthesis of 84-e: N-(2,4-difluoro-3-[[(3-phenyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl)ox y1methyl1phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0590] To a stirred solution of 2,4-difluoro-3-[[(3-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (200 mg, 0.4 mmol, 1 equiv) in pyridine (4 mL) was added 5 -fluoro-2-methoxypyridine-3 -sulfonyl chloride (140 mg, 0.6 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for 1 h then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-70% MeCN / 0.1% aqueous formic acid; to afford N-(2,4-difluoro-3-[[(3- phenyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]py ridin-5-yl)oxy]methyl]phenyl)- 5-fluoro-2-methoxypyridine-3-sulfonamide (200 mg, 72% yield) as a brown oil.

LCMS (ES, m/z): [M+H] + : 672

Synthesis of Compound 84: N-[2,4-difluoro-3-[([3-phenyl-1H-pyrazolol3,4-b1pyridin-5- yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine-3-sulfonami de

[0591] A solution of N-(2,4-difluoro-3-[[(3-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide (200 mg, 0.3 mmol, 1 equiv) in TFA (2 mL) and DCM (4 mL) was stirred for 1 h at room temperature, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250 mm, 10 μm; Mobile Phase: 35-70% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro- 3-[([3-phenyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]phen yl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (20 mg, 12% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 542

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.74 (s, 1H), 10.37 (s, 1H), 8.42 (d, J= 2.9 Hz, 1H), 8.27 (d, J= 2.6 Hz, 1H), 8.14 (d, J= 2.7 Hz, 1H), 8.09-8.00 (m, 2H), 7.96 (dd, J= 7.2, 2.9 Hz, 1H), 7.53 (dd, J= 8.3, 6.7 Hz, 2H), 7.48-7.32 (m, 2H), 7.17 (t, J= 8.9 Hz, 1H), 5.25 (s, 2H), 3.88 (s, 3H).

Example 96: N-[2,4-difluoro-3-[([3-methyl-4-phenyl-1H-pyrazolo[3.,4-b]py ridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (Compound 85)

Synthesis of 85-a: 5-bromo-2-hydrazinylpyrimidine

[0592] To a solution of 5-bromo-2-chloropyrimidine (5 g, 26 mmol, 1 equiv) in EtOH (17 mL) was added NH 2 NH 2 .H 2 O (2.6 g, 52 mmol, 2 equiv). The resulting solution was stirred for 3 h at 60 °C, then cooled and diluted with H 2 O (10 mL). The resulting solution was extracted with dichloromethane (8x10 mL). The combined organics were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. 5-Bromo-2-hydrazinylpyrimidine (4 g) was isolated as a white solid which was used in next step directly without further purification.

LCMS (ES, m/z): [M+H] + : 189

Synthesis of 85-c: 5-bromo-3-methyl-4-phenyl-1H-pyrazolo[3,4-b1pyridine

[0593] To a solution of 5-bromo-2-hydrazinylpyrimidine (2 g, 11 mmol, 1 equiv) and 4- phenyl-3-butyn-2-one (1.5 g, 11 mmol, 1 equiv) in THF (30 mL) was added TFA (60 mg, 0.5 mmol, 0.05 equiv). The reaction was stirred for 20 min at 60 °C, then concentrated. To the residue was added THF (20 mL), TFAA (3.56 g, 17 mmol, 3 equiv) and pentan-3-one (1.46 g, 17 mmol, 3 equiv). The resulting solution was heated with microwave irradiation for 1 h at 60 °C, then diluted with EA (10 mL) and washed with Na 2 CO 3 (2x10 mL). The organics were dried over anhydrous sodium sulfate and concentrated. The residue was purified by prep- HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 5-bromo-3- methyl-4-phenyl-lH-pyrazolo[3,4-b]pyridine (1.2 g, 74% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 288

Synthesis of 85-d: 5-bromo-3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridine

[0594] To a solution of 5-bromo-3-methyl-4-phenyl-lH-pyrazolo[3,4-b]pyridine (1 g, 3.5 mmol, 1 equiv) in THF (20 mL) cooled in an ice bath was added NaH (166 mg, 4 mmol, 1.2 equiv, 60% in oil) in portions. After stirring for 30 min, the ice bath was removed and SEM- C1 (694 mg, 4 mmol, 1.2 equiv) was added dropwise at room temperature. The resulting solution was stirred for 1 h then diluted with H 2 O (10 mL). The mixture was extracted with ethyl acetate (3x10 mL), and the combined organics were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (9: 1) to afford 5-bromo-3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1.4 g, 96% yield) as a red oil.

LCMS (ES, m/z): [M+H] + : 418

Synthesis of 85-f: 3-methyl-4-phenyl-l-[[2-(trimethylsilyl)ethoxy1methyl1pyrazo lo[3,4- blpyridin-5-ol

[0595] To a solution of 5-bromo-3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridine (1.4 g, 3 mmol, 1 equiv) and bis(pinacolato)diboron (1.7 g, 6 mmol, 2 equiv) in dioxane (33 mL) were added Pd(OAc)2 (75 mg, 0.34 mmol, 0.1 equiv), PCy3 (188 mg, 0.67 mmol, 0.2 equiv) and KO Ac (657 mg, 6.7 mmol, 2 equiv). The resulting mixture was stirred for 12 h at 80 °C under nitrogen atmosphere. The mixture was allowed to cool to room temperature and THF (15 mL) and aq. NaOH (15 mL, 2% aqueous solution) were added. 30% H 2 O 2 (1 g, 30 mmol, 10 equiv) was then added dropwise at room temperature. The resulting mixture was stirred for 1 h, then quenched with saturated aqueous Na2S2O3 (10 mL). The resulting mixture was extracted with EA (3 x 20 mL). The combined organics were washed with brine (10 mL), dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by prep-HPLC Flash-Prep- HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 3-methyl-4- phenyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]py ridin-5-ol (788 mg, 74% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 356

Synthesis of 85-g: 2,4-difluoro-3-[[(3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolol3,4-b1pyridin-5-yl)ox y1methyl1aniline

[0596] To a stirred solution of 3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-ol (780 mg, 2 mmol, 1 equiv) and (3- amino-2,6-difluorophenyl)methanol (699 mg, 4 mmol, 2 equiv) in DCM (20 mL) were added TMAD (567 mg, 3 mmol, 1.5 equiv) and PPh 3 (690 mg, 2.6 mmol, 1.2 equiv) in portions at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford 2,4-difluoro-3-[[(3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (300 mg, 28% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 497

Synthesis of 85-h: N-(2,4-difluoro-3-[[(3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy1methyl1pyrazolo[3,4-b1pyridin-5-yl)ox y1methyl1phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide

[0597] To a stirred solution of 2,4-difluoro-3-[[(3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]aniline (116 mg, 0.2 mmol, 1 equiv) in pyridine (2 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (79 mg, 0.3 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40 μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid; to afford N-(2,4-difluoro-3-[[(3-methyl-4- phenyl-l-[[2-(trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]py ridin-5-yl)oxy]methyl]phenyl)- 5-fluoro-2-methoxypyridine-3-sulfonamide (120 mg, 75% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 686

Synthesis of Compound 85: N-12,4-difluoro-3-[([3-methyl-4-phenyl-1H-pyrazolo[3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine -3-sulfonamide

[0598] A solution of N-(2,4-difluoro-3-[[(3-methyl-4-phenyl-l-[[2- (trimethylsilyl)ethoxy]methyl]pyrazolo[3,4-b]pyridin-5-yl)ox y]methyl]phenyl)-5-fluoro-2- methoxypyridine-3 -sulfonamide (120 mg, 0.17 mmol, 1 equiv) in TFA (1 mL) and DCM (2 mL) was stirred for 2 h at room temperature. The resulting mixture was concentrated and the residue purified by prep-HPLC with the following conditions: Column, welch Vltimate XB- C18, 50x 250 mm, 10 μm; Mobile Phase: 5-40% MeCN / 0.1% aqueous formic acid; to afford N-[2,4-difluoro-3-[([3-methyl-4-phenyl-lH-pyrazolo[3,4-b]pyr idin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (30 mg, 31% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 556

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.25 (s, 1H), 10.27 (s, 1H), 8.38 (t, J= 1.5 Hz, 2H), 7.93 (dd, J= 13, 3.0 Hz, 1H), 7.45-7.28 (m, 3H), 7.32-7.15 (m, 1H), 7.21-7.12 (m, 2H), 6.97 (t, J = 8.7 Hz, 1H), 4.93 (s, 2H), 3.79 (s, 3H), 1.89 (s, 3H).

Example 97: Synthesis of N-[3-[([4-cvclopropyl-3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]-2.,4-difluorophenyl]-5-fluoro-2-methoxypyridi ne-3-sulfonamide

(Compound 86) Synthesis of 86-a: 4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin-5-ol

[0599] To 3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (18 g, 77 mmol, 1 equiv) in DMF (450 mL) was added a solution of NBS (14.8 g, 83 mmol, 1.1 equiv) in DMF (50 mL) dropwise. The resulting solution was stirred for 5 h, then diluted with ice water (1.2 L). The solids were collected by filtration, washed with water (3 x 200 mL) and dried under reduced pressure to give 4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (20 g, 83% yield) as a grey solid.

LCMS (ES, m/z): [M+H] + : 312

Synthesis of 86-b: (2,6-difluoro-3-nitrophenyl)methanol

[0600] BH3-THF (62 mL, 1 M, 62 mmol, 2.5 equiv) was added dropwise to a stirred solution of 2,6-difluoro-3-nitrobenzoic acid (5.0 g, 25 mmol, 1 equiv) in THF (50 mL) at 0 °C. The reaction mixture was stirred 25 °C for 16 h and then quenched by the addition of MeOH (5 mL). The solution was diluted with water (100 mL) and extracted with EA (3 x 100 mL). The combined organic extracts were washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with PE:EA (5: 1) to afford (2,6-difluoro-3- nitrophenyl)methanol (3.0 g, 64% yield) as a light brown solid.

LCMS (ES, m/z): [M+H] + : 190

Synthesis of 86-c: (3-amino-2,6-difluorophenyl)methanol

[0601] Pd/C (0.5 g, 10% w/w) was added to a solution of (2,6-difluoro-3- nitrophenyl)methanol (1.5 g, 8 mmol, 1 equiv) in EA (30 mL) and the resulting suspension stirred at room temperature for 2 h under H 2 (2 atm). The reaction mixture was filtered and then concentrated under reduced pressure to afford (3 -amino-2,6-difluorophenyl)m ethanol (1.0 g, 79% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 160.

Synthesis of 86-d: N-[2,4-difluoro-3-(hydroxymethyl)phenyl1-5-fluoro-2-methoxyp yridine-3- sulfonamide

[0602] To (3 -amino-2,6-difluorophenyl)m ethanol (5 g, 31 mmol, 1 equiv) in DCM (100 mL) was added 5-fluoro-2-methoxypyridine-3-sulfonyl chloride (8.5 g, 38 mmol, 1.2 equiv) and pyridine (7.5 g, 94 mmol, 3 equiv). The reaction was stirred for 1 h, then quenched with water (100 mL). The resulting solution was extracted with ethyl acetate (3 x 100 mL), and the combined organics washed with water (100 mL) and brine (100 mL), then dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel chromatography, eluting with ethyl acetate/petroleum ether (1 :3) to give N-[2,4-difluoro-3- (hydroxymethyl)phenyl]-5-fluoro-2-methoxypyridine-3-sulfonam ide (6 g, 55% yield) as a solid.

LCMS (ES, m/z): [M+H] + : 349

Synthesis of 86-e: tert-butyl N-[2,4-difluoro-3-(hydroxymethyl)phenyl1-N-(5-fluoro-2- methoxypyridin-3-ylsulfonyl)carbamate

[0603] To N-[2,4-difluoro-3-(hydroxymethyl)phenyl]-5-fluoro-2-methoxyp yridine-3- sulfonamide (6.2 g, 18 mmol, 1 equiv) in THF (120 mL) was added TEA (3.6 g, 36 mmol, 2 equiv), DMAP (0.43 g, 3.5 mmol, 0.2 equiv) and BOC2O (5.3 g, 27 mmol, 1.5 equiv). The reaction was stirred overnight, then concentrated. The residue was purified by silica gel column chromatography, eluting with ethyl acetate/petroleum ether (1 :4) to give tert-butyl N- [2,4-difluoro-3-(hydroxymethyl)phenyl]-N-(5-fluoro-2-methoxy pyri din-3- ylsulfonyl)carbamate (6 g, 75% yield) as a light yellow solid.

LCMS (ES, m/z): [M+H] + : 449

Synthesis of 86-f: tert-butyl N-[3-([[4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b1pyridin - 5-yl1oxy1methyl)-2,4-difluorophenyl1-N-(5-fluoro-2-methoxypy ridin-3-ylsulfonyl)carbamate

[0604] To tert-butyl N-[2,4-difluoro-3-(hydroxymethyl)phenyl]-N-(5-fluoro-2- methoxypyridin-3-ylsulfonyl)carbamate (4 g, 8.9 mmol, 1 equiv) in DCM (100 mL) was added 4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (2.8 g, 8.9 mmol, 1 equiv), PPI13 (3.5 g, 13 mmol, 1.5 equiv) and TMAD (2.3 g, 13.38 mmol, 1.5 equiv). The reaction was stirred overnight, then concentrated. The residue was purified by silica gel column chromatography, eluting with ethyl acetate/petroleum ether (1 :3) to give tert-butyl N- [3-([[4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5 -yl]oxy]methyl)-2,4- difluorophenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfonyl)ca rbamate (1.9 g, 29% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 742 Synthesis of 86-g: tert-butyl N-[3-([[4-cyclopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b1pyridin-5-yl1oxy1methyl)-2,4-difluorophenyl1-N-(5-fluoro-2 -methoxypyridin-3- yl sulfonyl (carbamate

[0605] To tert-butyl N-[3-([[4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin -5- yl]oxy]methyl)-2,4-difluorophenyl]-N-(5-fluoro-2-methoxypyri din-3-ylsulfonyl)carbamate (200 mg, 0.27 mmol, 1 equiv) in dioxane (6 mL) was added cyclopropylboronic acid (81 mg, 0.94 mmol, 3.5 equiv), Pd(dppf)C12 (39 mg, 0.054 mmol, 0.2 equiv), K2CO3 (112 mg, 0.81 mmol, 3 equiv) and H 2 O (1 mL). The reaction was stirred overnight at 90 °C, then cooled and concentrated. The residue was purified by Flash -Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase, 10-95% MeCN / 0.1% aqueous formic acid. Concentration of the appropriate fractions gave tert-butyl N-[3- ([[4-cyclopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridi n-5-yl]oxy]methyl)-2,4- difluorophenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfonyl)ca rbamate (120 mg, 63% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 704

Synthesis of Compound 86: N-[3-[([4-cyclopropyl-3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1oxy)methyl1-2,4-difluorophenyl1-5-fluoro-2-methoxypyridin e-3-sulfonamide

[0606] To tert-butyl N-[3-([[4-cyclopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]p yridin-5- yl]°xy]methyl)-2,4-difluorophenyl]-N-(5-fluoro-2-methoxypyr idin-3-ylsulfonyl)carbamate (120 mg, 0.17 mmol, 1 equiv) in DCM (3 mL) was added TFA (1 mL). The resulting solution was stirred for 3 h, then concentrated. The pH of the solution was adjusted to 8 with NH3 (7 mol/L in MeOH, 3 mL) before being concentrated. The residue was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10 μm mobile phase; Mobile Phase: 20-70% MeCN / 0.05% aqueous ammonia. Concentration of the relevant fractions gave N-[3-[([4-cyclopropyl-3-methyl-lH-pyrazolo[3,4-b]pyridin-5- yl]oxy)methyl]-2,4-difluorophenyl]-5-fluoro-2-methoxypyridin e-3-sulfonamide (35 mg, 40% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 520

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.01 (s, 1H), 10.36 (s, 1H), 8.44 (d, 1H), 8.15 (s, 1H), 7.94 (dd, 1H), 7.36 (dt, 1H), 7.12 (dt, 1H), 5.09 (s, 2H), 3.93 (s, 3H), 2.64 (s, 3H), 2.11-1.94 (m, 1H), 1.00-0.78 (m, 4H). Example 98: Synthesis of N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-4-yl)-1H- pyrazolo [3,4-b] Dyridin-5-yl] oxy] methyl)phenyl] -5-fluoro-2-methoxypyridine-3- sulfonamide (Compound 87)

87-b Compound 87

Synthesis of 87-a: 3-methyl-l-(oxan-2-yl)-4-(pyridin-4-yl)pyrazolo[3,4-b]pyridi n-5-ol

[0607] 4 -Bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (200 mg, 0.64 mmol), pyridin-4-ylboronic acid (158 mg, 1.3 mmol, 2 equiv), Pd(dppf)Cl 2 (94 mg, 0.13 mmol, 0.2 equiv) and K 2 CO 3 (266 mg, 1.9 mmol, 3 equiv) in dioxane (10 mL) and H 2 O (2 mL) were heated for 6 h at 100 °C, then cooled and concentrated. The residue was purified by Prep- HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40μm, 120 g; mobile phase: 10-70% MeCN / 0.1% aqueous formic acid. The appropriate fractions were concentrated to give 3-methyl-l-(oxan-2-yl)-4-(pyridin-4-yl)pyrazolo[3,4-b]pyridi n-5- ol (130 mg, 65% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 311

Synthesis of 87-b: tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-4- yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl carbamate

[0608] 3-Methyl-l-(oxan-2-yl)-4-(pyridin-4-yl)pyrazolo[3,4-b]pyridi n-5-ol (150 mg, 0.48 mmol) and tert-butyl N-[2,4-difluoro-3-(hydroxymethyl)phenyl]-N-(5-fluoro-2- methoxypyridin-3-ylsulfonyl)carbamate (217 mg, 0.48 mmol, 1 equiv) in DCM (10 mL) were treated with PPI13 (254 mg, 0.97 mmol, 2 equiv), then TMAD (167 mg, 0.97 mmol, 2 equiv). The resulting solution was stirred overnight, then concentrated. The residue was purified by Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40μm, 120 g; mobile phase: 20-85% MeCN / 0.1% aqueous formic acid. Concentration of the appropriate fractions gave tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4- (pyridin-4-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl] -N-(5-fluoro-2- methoxypyridin-3-ylsulfonyl)carbamate (110 mg, 31% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 741

Synthesis of Compound 87: N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-4-yl)-1H-pyrazolo[3 ,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide

[0609] Tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-4-yl) pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl) phenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfonyl)carbamate (140 mg, 0.19 mmol) in DCM (5 mL) was treated with TFA (1.5 mL). The resulting solution was stirred for 5 h, then concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase; Mobile Phase: 20-70% MeCN / 0.1% aqueous ammonia. The relevant fractions were concentrated to give N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-4-yl)-lH-pyrazolo[3 ,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (60 mg, 57% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 557

'H NMR (300 MHz, DMSO-d 6 ) δ 13.37 (s, 1H), 10.32 (br s, 1H), 8.62-8.50 (m, 3H), 8.36 (d, 1H), 7.92 (dd, 1H), 7.26 (td, 1H), 7.20-7.13 (m, 2H), 6.94 (td, 1H), 5.02 (s, 2H), 3.79 (s, 3H), 1.92 (s, 3H).

Example 99: N-[2,4-difluoro-3-[([4-isopropyl-3-methyl-1H-pyrazolo[3.,4-b ]pyridin-5- yl]oxy)methyl]phenyl]-5-fluoro-2-methoxypyridine-3-sulfonami de (Compound 88)

88-d Compound 88

Synthesis of 88-a: 4-bromo-5-l(tert-butyldimethylsilyl)oxy1-3-methyl-l-(oxan-2- yl)pyrazolol3,4-b1pyridine

[0610] To a solution of 4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-ol (8 g, 26 mmol, 1 equiv) and imidazole (5.2 g, 77 mmol, 3 equiv) in DCM (130 mL) was added TBSC1 (5.79 g, 38 mmol, 1.5 equiv) at room temperature. The reaction was stirred for 1 h, then diluted with H 2 O (50 mL). The resulting mixture was extracted with DCM (3 x 50 mL), and the combined organics were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , before being concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (9: 1) to afford 4-bromo-5-[(tert-butyldimethylsilyl)oxy]-3-methyl-l- (oxan-2-yl)pyrazolo[3,4-b]pyridine (10 g, 92% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 426 Synthesis of 88-b: 5-[(tert-butyldimethylsilyl)oxy1-4-isopropyl-3-methyl-l-(oxa n-2- yl)pyrazolo[3,4-b1pyridine

[0611] To a solution of 4-bromo-5-[(tert-butyldimethylsilyl)oxy]-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (2 g, 4.7 mmol, 1 equiv) and CuCN (1.3 g, 14 mmol, 3 equiv) in THF (10 mL) was added isopropyl magnesium chloride solution (2 M in THF, 14 mL, 28 mmol, 5 equiv) dropwise at 0 °C. The reaction was stirred for 2 h at 0 °C, then quenched with H 2 O (20 mL). The resulting mixture was extracted with EA (3 x 20 mL), and the combined organics washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 40-90% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave 5-[(tert-butyldimethylsilyl)oxy]-4- isopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridine (740 mg, 41% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 391

Synthesis of 88-c: 4-isopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-o l

[0612] To a solution of 5-[(tert-butyldimethylsilyl)oxy]-4-isopropyl-3-methyl-l-(oxa n-2- yl)pyrazolo[3,4-b]pyridine (700 mg, 1.8 mmol, 1 equiv) in THF (10 mL) was added TBAF (704 mg, 2.7 mmol, 1.5 equiv) in portions at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 20-60% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave 4-isopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-o l (260 mg, 53% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 276

Synthesis of 88-d: tert-butyl N-[2,4-difluoro-3-([[4-isopropyl-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl carbamate

[0613] To a solution of 4-isopropyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-o l (250 mg, 0.91 mmol, 1 equiv), tert-butyl N-[2,4-difluoro-3-(hydroxymethyl)phenyl]-N-(5-fluoro- 2-methoxypyridin-3-ylsulfonyl)carbamate (448 mg, 1 mmol, 1.1 equiv) and PPhs (286 mg, 1.1 mmol, 1.2 equiv) in DCM (9 mL) was added TMAD (188 mg, 1.1 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for 30 min, then concentrated. The residue was purified by silica gel column chromatography, eluting with PE:EA (4: 1) to afford tert-butyl N-[2,4-difluoro-3-([[4-isopropyl-3-methyl-l-(oxan-2-yl)pyraz olo[3,4-b]pyridin-5- yl]oxy]methyl)phenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfo nyl)carbamate (600 mg, 94% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 622

Synthesis of Compound 88: N-[2,4-difluoro-3-[([4-isopropyl-3-methyl-1H-pyrazolol3,4- b1pyridin-5-yl1oxy)methyl1phenyl1-5-fluoro-2-methoxypyridine -3- sulfonamide

[0614] To a stirred solution of tert-butyl N-[2,4-difluoro-3-([[4-isopropyl-3-methyl-l-(oxan- 2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-N-(5-flu oro-2-methoxypyridin-3- ylsulfonyl) carbamate (300 mg, 0.425 mmol, 1 equiv) in DCM (4 mL) was added TFA (2 mL) dropwise at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase; Mobile Phase 30-65% MeCN / 0.1% aqueous ammonia. The relevant fractions were concentrated to give N-[2,4-difluoro-3-[([4- isopropyl-3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl ]phenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (100 mg, 45% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 522

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.02 (s, 1H), 10.39 (s, 1H), 8.41 (d, 2H), 7.94 (dd, 1H), 7.48-7.34 (m, 1H), 7.18 (t, 1H), 5.15 (s, 2H), 3.92 (s, 3H), 3.58 (p, 1H), 2.57 (s, 3H), 1.16 (d, 6H).

Example 100: Synthesis of N-[3-[([4-benzyl-3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]-2.,4-difluorophenyl]-5-fluoro-2-methoxypyridi ne-3-sulfonamide

(Compound 89)

Synthesis of 89-a: tert-butyl N-[3-([[4-benzyl-3-rnethyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyrid in- 5-yl1oxy1methyl)-2,4-difluorophenyl1-N-(5-fluoro-2-methoxypy ridin-3-ylsulfonyl)carbamate

[0615] Tert-butyl N-[3-([[4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin -5- yl]oxy]methyl)-2,4-difluorophenyl]-N-(5-fluoro-2-methoxypyri din-3-ylsulfonyl)carbamate (200 mg, 0.27 mmol), 2-benzyl-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (206 mg, 0.94 mmol, 3.5 equiv), Pd(dppf)Cl 2 (59 mg, 0.08 mmol, 0.3 equiv) and K 2 CO 3 (112 mg, 0.81 mmol, 3 equiv) in dioxane (6 mL) and H 2 O (1 mL) were stirred for 5 h at 100 °C. The mixture was concentrated in vacuo, and the residue purified by Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 20-90% MeCN / 0.1% aqueous formic acid. Concentration of the appropriate fractions gave tert-butyl N-[3- ([[4-benzyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-y l]oxy]methyl)-2,4- difluorophenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfonyl)ca rbamate(90 mg, 44% yield) as a light brown semi-solid.

LCMS (ES, m/z): [M+H] + : 754

Synthesis of Compound 89: N-[3-[([4-benzyl-3-methyl-1H-pyrazolo[3,4-b1pyridin-5- yl1oxy)methyl1-2,4-difluorophenyl1-5-fluoro-2-methoxypyridin e-3-sulfonamide

[0616] Tert-butyl N-[3-([[4-benzyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridi n-5- yl]°xy]methyl)-2,4-difluorophenyl]-N-(5-fluoro-2-methoxypyr idin-3-ylsulfonyl)carbamate (90 mg, 0.12 mmol) in DCM (3 mL) was treated with TFA (1 mL). The resulting solution was stirred for 3 h, then concentrated. The pH of the residue was adjusted to 8 with 7M NH3 in MeOH, and concentrated again. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase; Mobile Phase: 30-75% MeCN / 0.1% aqueous ammonia. Concentration of the appropriate fractions gave N-[3-[([4-benzyl-3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy )methyl]-2,4- difluorophenyl]-5-fluoro-2-methoxypyridine-3-sulfonamide (15 mg 22% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 570 1 H NMR (300 MHz, DMSO-d 6 ) δ 13.12 (s, 1H), 10.35 (s, 1H), 8.45 (d, 1H), 8.38 (d, 1H), 7.93 (dd, 1H), 7.33 (dt, 1H), 7.25-7.01 (m, 4H), 6.90-6.99 (m, 2H), 5.13 (s, 2H), 4.19 (s, 2H), 3.87 (s, 3H), 2.35 (s, 3H).

Example 101: Synthesis of N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-2-yl)-1H- pyrazolo [3,4-b] pyridin-5-yl] oxy] methyl)phenyl] -5-fluoro-2-methoxypyridine-3- sulfonamide (Compound 90)

90-a Compound 90

Synthesis of 90-a: tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-2- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl carbamate

[0617] Tert-butyl N-[3-([[4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin -5- yl]°xy] m ethyl)-2,4-difluorophenyl]-N-(5-fluoro-2-methoxypyridi n-3-ylsulfonyl)carbamate (200 mg, 0.27 mmol) in toluene (10 mL) was treated with 2-(tributylstannyl)pyridine (198 mg, 0.54 mmol, 2 equiv) and Pd(PPh3)4 (63 mg, 0.054 mmol, 0.2 equiv). The resulting solution was stirred for overnight at 100 °C, then concentrated. The residue was purified by Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40μm, 120 g; mobile phase: 10-82% MeCN / 0.1% aqueous formic acid. The appropriate fractions were concentrated to give tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4- (pyridin-2-yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl] -N-(5-fluoro-2- methoxypyridin-3-ylsulfonyl)carbamate (110 mg, 55% yield) as a light brown semi-solid.

LCMS (ES, m/z): [M+H] + : 741

Synthesis of Compound 90: N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-2-yl)-1H-pyrazolol3 ,4- b1pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2-methoxypyridine -3-sulfonamide

[0618] Tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-2-yl) pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl) phenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfonyl)carbamate (115 mg, 0.16 mmol) in DCM (3 mL) was treated with TFA (1 mL). The resulting solution was stirred for 3 h, then concentrated. The pH of the residue was adjusted to 8 with 7M NH3 in MeOH, then concentrated again. The residue was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase, Mobile Phase: 20-65% MeCN / 0.05% aqueous ammonia. The relevant fractions were combined and concentrated to give N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-2-yl)-lH- pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-me thoxypyridine-3-sulfonamide (35 mg, 41% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 557

'H NMR (300 MHz, DMSO-d 6 ) δ 13.27 (s, 1H), 10.42 (br s, 1H), 8.65 (d, 1H), 8.48 (d, 1H), 8.37 (d, 1H), 7.93 (dd, 1H), 7.82 (dt, 1H), 7.44 (dd, 1H), 7.35 (dd, 1H), 7.25 (dt, 1H), 6.95 (t, 1H), 5.00 (s, 2H), 3.80 (s, 3H), 1.88 (s, 3H).

Example 102: Synthesis of N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-3-yl)-1H- pyrazolo [3,4-b] pyridin-5-yl] oxy] methyl)phenyl] -5-fluoro-2-methoxypyridine-3- sulfonamide (Compound 91)

91 -a Compound 91

Synthesis of 91-a: tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-3- yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl carbamate

[0619] Tert-butyl N-[3-([[4-bromo-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin -5- yl]oxy]inethyl)-2,4-difluorophenyl]-N-(5-fluoro-2-methoxypyr idin-3-ylsulfonyl)carbamate (200 mg, 0.27 mmol), pyri din-3 -ylboronic acid (66 mg, 0.54 mmol, 2 equiv), Pd(dppf)C12 (60 mg, 0.08 mmol, 0.3 equiv) and K 2 CO 3 (112 mg, 0.81 mmol, 3 equiv) in dioxane (6 mL) and H 2 O (1 mL) were heated for 5 h at 90 °C. After concentration, the residue was purified by Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20- 40μm, 120 g; mobile phase: 5-85% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-3- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl)carbamate (130 mg, 65% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 741

Synthesis of Compound 91 : N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-3-yl)-1H-pyrazolo[3 ,4- b1pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2-methoxypyridine -3-sulfonamide

[0620] Tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(pyridin-3-yl) pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl) phenyl]-N-(5-fluoro-2-methoxypyridin-3-ylsulfonyl)carbamate (140 mg, 0.19 mmol) in DCM (3 mL) was treated with TFA (1 mL). The resulting solution was stirred for 5 h, then concentrated. The pH of the residue was adjusted to 8 with 7M NH3 in MeOH, then was concentrated again. The crude product was purified by Prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase; Mobile Phase: 25-80% MeCN / 0.05% aqueous ammonia. The relevant fractions were concentrated to give N-[2,4-difluoro-3-([[3-methyl-4-(pyridin-3-yl)-lH-pyrazolo[3 ,4- b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-methoxypyridine -3-sulfonamide (60 mg, 57% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 557

'H NMR (300 MHz, DMSO-d 6 ) δ 13.34 (s, 1H), 10.35 (br s, 1H), 8.60 (d, 1H), 8.52 (s, 1H), 8.37 (dd, 2H), 7.92 (dd, 1H), 7.61 (dt, 1H), 7.41 (dd, 1H), 7.28-7.17 (m, 1H), 6.93 (t, 1H), 5.00 (s, 2H), 3.78 (s, 2H), 1.93 (s, 3H).

Example 103: Synthesis of N-[3-[([4-cvdopentyl-3-methyl-1H-pyrazolo[3.,4-b]pyridin-5- yl]oxy)methyl]-2.,4-difluorophenyl]-5-fluoro-2-methoxypyridi ne-3-sulfonamide (Compound 92)

Synthesis of 92-a: 5-l(tert-butyldimethylsilyl)oxy1-4-cyclopentyl-3-methyl-l-(o xan-2- yl)pyrazolol3,4-b1pyridine

[0621] To a solution of 4-bromo-5-[(tert-butyldimethylsilyl)oxy]-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (2 g, 4.7 mmol, 1 equiv) and CuCN (1.26 g, 14 mmol, 3 equiv) in THF (20 mL) was added cyclopentyl magnesium chloride (2M in THF, 12 mL, 24 mmol, 5 equiv) dropwise at 0 °C. The reaction was stirred for 2 h at 0 °C, then quenched by the addition of H 2 O (20 mL). The resulting mixture was extracted with EA (3 x 50 mL), and the combined organics washed with brine (50 mL), dried over anhydrous Na2 SO4. before being concentrated. The residue was purified by prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 40-90% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave 5-[(tert- butyldimethylsilyl)oxy]-4-cyclopentyl-3-methyl-l-(oxan-2-yl) pyrazolo[3,4-b]pyridine (480 mg, 25% yield) as a yellow oil.

LCMS (ES, m/z): [M+H] + : 416

Synthesis of 92-b: 4-cyclopentyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5 -ol

[0622] To a solution of 5-[(tert-butyldimethylsilyl)oxy]-4-cyclopentyl-3-methyl-l-(o xan-2- yl)pyrazolo[3,4-b]pyridine (480 mg, 1.2 mmol, 1 equiv) in THF (10 mL) was added TBAF (603 mg, 2.3 mmol, 2 equiv) in portions at room temperature. The resulting mixture was stirred for 1 h then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 20- 60% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave 4- cyclopentyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5-o l (308 mg, 88% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 302

Synthesis of 92-c: tert-butyl N-[3-([[4-cyclopentyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b1pyridin-5-yl1oxy1methyl)-2,4-difluorophenyl1-N-(5-fluoro-2 -methoxypyridin-3- ylsulfonyl)carbamate

[0623] To a solution of 4-cyclopentyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5 -ol (300 mg, 1 mmol, 1 equiv), tert-butyl N-[2,4-difluoro-3-(hydroxymethyl)phenyl]-N-(5- fluoro-2-methoxypyridin-3-ylsulfonyl)carbamate (446 mg, 1 mmol, 1 equiv) and PPI13 (313 mg, 1.2 mmol, 1.2 equiv) in DCM (10 mL) was added TMAD (205 mg, 1.2 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by silica gel column chromatography, eluting with 4: 1 PE:EAto afford tert-butyl N-[3-([[4-cyclopentyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b]p yridin-5-yl]oxy]methyl)-2,4- difluorophenyl]-N-(5-fluoro-2-methoxypyridin-3ylsulfonyl)car bamate (600 mg, 82% yield) as a white solid. LCMS (ES, m/z): [M+H] + : 732

Synthesis of Compound 92: N-[3-[([4-cyclopentyl-3-methyl-1H-pyrazolol3,4-b1pyridin-5- yl1oxy)methyl1-2,4-difluorophenyl1-5-fluoro-2-methoxypyridin e-3-sulfonamide

[0624] To a solution of tert-butyl N-[3-([[4-cyclopentyl-3-methyl-l-(oxan-2-yl)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)-2,4-difluorophenyl]-N-(5-fluoro-2 -methoxypyri din-3- ylsulfonyl)carbamate (300 mg, 0.41 mmol, 1 equiv) in DCM (4 mL) was added TFA (2 mL) dropwise at room temperature. The resulting mixture was stirred for 1 h, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase; Mobile Phase: 40-60% MeCN / 0.05% aqueous ammonia. Concentration of the relevant fractions gave N-[3-[([4-cyclopentyl-3- methyl-lH-pyrazolo[3,4-b]pyridin-5-yl]oxy)methyl]-2,4-difluo rophenyl]-5-fluoro-2- methoxypyridine-3 -sulfonamide (50 mg, 22% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 548

'H NMR (300 MHz, DMSO-d 6 ) δ 12.98 (s, 1H), 10.38 (s, 1H), 8.44-8.37 (m, 2H), 7.93 (dd, 1H), 7.39 (dt, 1H), 7.16 (t, 1H), 5.15 (s, 2H), 3.92 (s, 3H), 3.63 (p, 1H), 2.56 (s, 3H), 1.77 -1.67 (m, 4H), 1.49-1.46 (m, 4H).

Example 104: Synthesis of N-[2,4-difluoro-3-([[3-methyl-4-(l-methylpiperidin-4-yl)-1H- pyrazolo [3,4-b] Dyridin-5-yl] oxy] methyl)phenyl] -5-fluoro-2-methoxypyridine-3- sulfonamide (Compound 93)

Synthesis of 93-a: 4-[5-[(Tert-butyldirnethylsilyl)oxy]-3-rnethyl-l-(oxan-2-yl) pyrazolol3,4- b]pyridin-4-yl]-l-methyl-3,6-dihydro-2H-pyridine

[0625] To a solution of 4-bromo-5-[(tert-butyldimethylsilyl)oxy]-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridine (2 g, 4.7 mmol, 1 equiv), CS 2 CO 3 (4.3 g, 9.4 mmol, 2 equiv) and l-methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,6 -dihydro-2H-pyridine (1.3 g, 5.6 mmol, 1.2 equiv) in dioxane (20 mL) and H 2 O (2 mL) was added Pd(dppf)Cl 2 (0.34 g, 0.47 mmol, 0.1 equiv) in portions, at room temperature under a nitrogen atmosphere. The reaction was stirred for 3 h at 100 °C, then cooled and quenched with H 2 O (20 mL). The mixture was extracted with EtOAc (3 x 50 mL), and the combined organics washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , and concentrated. The residue was purified by silica gel column chromatography, eluting with 3: 1 PE/EtOAc to afford 4-[5-[(tert- butyldimethylsilyl)oxy]-3-methyl- l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-4-yl]-l -methyl -3,6- dihydro-2H-pyridine (1.3 g, 63% yield) as a brown solid.

LCMS (ES, m/z): [M+H] + : 443

Synthesis of 93-b: 3-Methyl-4-(l-methylpiperidin-4-yl)-l-(oxan-2-yl)pyrazolo[3, 4-b1pyridin- 5-ol

[0626] To a solution of 4-[5-[(tert-butyldimethylsilyl)oxy]-3-methyl-l-(oxan-2- yl)pyrazolo[3,4-b]pyridin-4-yl]-l-methyl-3,6-dihydro-2H-pyri dine (240 mg, 0.54 mmol, 1 equiv) in MeOH (5 mL) was added and Pd/C (240 mg). The resulting mixture was stirred for 12 h at 50 °C under hydrogen (20 atm) atmosphere. After filtration, the filtrate was concentrated and the residue purified by Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 10-50% MeCN / 0.1% aqueous formic acid. Concentration of the appropriate fractions gave 3-methyl-4-(1- methylpiperidin-4-yl)-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- ol (100 mg, 56% yield) as a light brown solid.

LCMS (ES, m/z): [M+H] + : 331

Synthesis of 93-c: Tert-butyl N-[2,4-difluoro-3-([[3-methyl-4-(l-methylpiperidin-4-yl)-l- (oxan-2-yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-N- (5-fluoro-2-methoxypyridin- 3-ylsulfonyl)carbamate

[0627] To a solution of 3-methyl-4-(1-methylpiperidin-4-yl)-l-(oxan-2-yl)pyrazolo[3, 4- b]pyridin-5-ol (71 mg, 0.22 mmol), tert-butyl (2,4-difluoro-3-(hydroxymethyl)phenyl)((5- fluoro-2-methoxypyridin-3-yl)sulfonyl)carbamate (96 mg, 0.22 mmol, 1 equiv) in DCM (5 mL) were added PPh 3 (113 mg, 0.43 mmol, 2 equiv) and TMAD (74 mg, 0.43 mmol, 2 equiv) in portions at room temperature. The resulting mixture was stirred for 4 h, then quenched with H 2 O (10 mL) and extracted with CH 2 Cl 2 (3 x 20 mL). The combined organics were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 , and concentrated. The residue was purified by Prep-HPLC with the following conditions: Column, WelFlash TM C18-1, Spherical C18 20-40μm, 120 g; mobile phase: 20-70% MeCN / 0.1% aqueous formic acid. Concentration of the appropriate fractions gave tert-butyl N-[2,4-difluoro-3-([[3-methyl-4-(1- methylpiperidin-4-yl)-l-(oxan-2-yl)pyrazolo[3,4-b]pyridin-5- yl]oxy]methyl)phenyl]-N-(5- fluoro-2-methoxypyridin-3-ylsulfonyl)carbamate (85 mg, 52% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 761 Synthesis of Compound 93: N-12,4-Difluoro-3-([[3-methyl-4-(l-methylpiperidin-4-yl)-1H- pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2-me thoxypyridine-3-sulfonamide [0628] A solution of tert-butyl N-[2,4-difluoro-3-([[3-methyl-4-(1-methylpiperidin-4-yl)-l- (oxan-2-yl)pyrazolo[3,4b]pyridin-5-yl]oxy]methyl)phenyl]-N-( 5-fluoro-2-methoxypyridin-3- ylsulfonyl)carbamate (90 mg, 0.12 mmol) in DCM (1 mL) was treated with TFA (0.3 mL), then stirred for 2 h. The resulting mixture was concentrated and the residue purified by prep- HPLC with the following conditions: Column, welch Vltimate XB-C18, 50x250mm, 10μm mobile phase, Mobile Phase: 20-70% MeCN / 0.1% aqueous formic acid. Concentration of the appropriate fractions gave N-[2,4-difluoro-3-([[3-methyl-4-(1-methylpiperidin-4-yl)-lH- pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2-me thoxypyridine-3-sulfonamide (25 mg, 37% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 577

1 HNMR (300 MHz, DMSO-d 6 ) δ 13.09 (s, 1H), 8.40 (s, 1H), 8.31 (d, 1H), 7.93 (dd, 1H), 7.30 (dt, 1H), 7.03 (t, 1H), 5.16 (s, 2H), 3.84 (s, 3H), 3.27 (t, 1H), 2.92-3.04 (m, 2H), 2.62 (s, 3H), 2.32 (s, 3H), 2.31-2.03 (m, 4H), 1.48-1.62 (m, 2H).

Example 105: Synthesis of N-[2,4-difluoro-3-([[3-methyl-4-(piperidin-l-yl)-1H- pyrazolo [3,4-b] pyridin-5-yl] oxy] methyl)phenyl] -5-fluoro-2-methoxypyridine-3- sulfonamide (Compound 94)

Compound 94

Synthesis of 94-a: l-[5-[(tert-butyldirnethylsilyl)oxy]-3-rnethyl-l-(oxan-2-yl) pyrazolol3,4- blpyridin-4-yllpiperidine

[0629] To a solution of 4-bromo-5-[(tert-butyldimethylsilyl)oxy]-3-methyl-l-(oxan-2- yl)pyrazolo[3,4- b]pyridine (200 mg, 0.47 mmol, 1 equiv), piperidine (545 mg, 4.7 mmol, 10 equiv) and 2-acetylcyclohexan-l- one (66 mg, 0.47 mmol, 1 equiv) in DMF (4 mL) were added Cs 2 CO 3 (458 mg, 1.4 mmol, 3 equiv) and copper (I) iodide (18 mg, 0.09 mmol, 0.2 equiv). After stirring for 12 h at 70 °C under nitrogen, the resulting mixture was concentrated. The residue was purified by Flash-Prep-HPLC with the following conditions: Column, WelFlash TM C18-I, Spherical C18 20-40μm, 120 g; mobile phase: 30-80% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave l-[5-[(tert- butyldimethylsilyl)oxy]-3-methyl-l-(oxan-2-yl)pyrazolo[3,4-b ]pyridin-4-yl]piperidine (80 mg, 40% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 317

Synthesis of 94-b: tert-butyl N-12,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(piperidin-l- yl)pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl)carbamate

[0630] To a solution of 3-methyl-l-(oxan-2-yl)-4-(piperidin-l-yl)pyrazolo[3,4-b]pyri din-5-ol (200 mg, 0.63 mmol, 1 equiv) and tert-butyl N-[2,4-difluoro-3-(hydroxymethyl)phenyl]-N- (5-fluoro-2-methoxypyridin-3-ylsulfonyl)carbamate (311 mg, 0.7 mmol, 1.1 equiv) in DCM (5 mL) were added PPh 3 (200 mg, 0.76 mmol, 1.2 equiv) and TMAD (130 mg, 0.76 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for 2 h, then concentrated. The residue was purified by silica gel column chromatography, eluting with 1:1 PE: EAto afford tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(piperidin-l-y l)pyrazolo[3,4- b]pyridin-5-yl]oxy]methyl)phenyl]-N-(5-fluoro-2-methoxypyrid in-3-ylsulfonyl)carbamate (160 mg, 34% yield) as a yellow solid.

LCMS (ES, m/z): [M+H] + : 747

Synthesis of Compound 94: N-12,4-difluoro-3-([[3-methyl-4-(piperidin-l-yl)-1H- pyrazolo[3,4-b1pyridin-5-yl1oxy1methyl)phenyl1-5-fluoro-2-me thoxypyridine-3-sulfonamide

[0631] To a solution of tert-butyl N-[2,4-difluoro-3-([[3-methyl-l-(oxan-2-yl)-4-(piperidin-l- yl)pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-N-(5-fluor o-2-methoxypyridin-3- ylsulfonyl)carbamate (150 mg, 0.2 mmol, 1 equiv) in DCM (5 mL) was added TFA (1 mL) dropwise. The resulting mixture was stirred for 2 h, then concentrated. The residue was purified by prep-HPLC with the following conditions: Column, welch Vltimate XB-C18, 50 x 250mm, lOum mobile phase; Mobile Phase 25-55% MeCN / 0.1% aqueous formic acid. Concentration of the relevant fractions gave N-[2,4-difluoro-3-([[3-methyl-4-(piperidin-l-yl)- lH-pyrazolo[3,4-b]pyridin-5-yl]oxy]methyl)phenyl]-5-fluoro-2 -methoxypyridine-3- sulfonamide (70 mg, 62% yield) as a white solid.

LCMS (ES, m/z): [M+H] + : 563

1 HNMR (300 MHz, DMSO-d 6 ) δ 12.93 (s, 1H), 10.40 (s, 1H), 8.41 (d, 1H), 8.10 (s, 1H), 7.95 (dd, 1H), 7.35 (q, 1H), 7.13 (dt, 1H), 5.00 (s, 2H), 3.89 (s, 3H), 3.0-3.12 (m, 4H), 2.55 (s, 3H), 1.45-1.66 (m, 6H). Table 1.

Example 97 - Biochemical Assay for Modulation (Inhibition/Activation) of GCN2

[0598] Exemplary compounds from the above Examples were tested for ability to inhibit GCN2 activity using a time resolved fluorescence energy transfer (TR-FRET) assay. Assay procedures and results are described below for Examples 12-105.

Part I - Procedures for TR-FRET Assay

[0599] GCN2 protein was obtained from Cama Biosciences (cat# 05-153). The protein was diluted in assay buffer (ThermoFisher Scientific, #PV6135), 2 mM dithiothreitol (DTT), to obtain a final concentration of 2nM and 5 pL was plated in a 384-well white assay plate. Test compounds were serially diluted to 11 concentrations by 3-fold dilution in DMSO and 10 nL of stock was plated into 384 well white assay plate. DMSO was used as a vehicle control. GFP-eIF2a protein was obtained from ThermoFisher (cat# PV4809). The protein was diluted in assay buffer to a 2x concentration of 200 nM along with 300 μM ATP (final concentration of 100 nM GFP-eIF2a and 150 μM ATP) in the presence of 2 mM DTT and a 5 pL aliquot was added to each well containing the GCN2 protein and test compound. The plate was incubated in the dark at 25 °C for 1.5 hours, shaking at 1250 rμm. Tb-anti P-eIF2a (ThermoFisher cat# PV4815) was diluted to a to a concentration of 1 nM in TR-FRET Dilution Buffer (ThermoFisher cat# PV3574). 10 pL of the Tb-anti P-eIF2α solution was added to the TR-FRET reaction. The plate was incubated in the dark for 2 h at 25 °C shaking at 600 rpm. The FRET signal from the plate was read on a Envision (PerkinElmer) plate reader:

[0600] Label 1 : Excitation: 340 nm, bandwidth 30 nm; Emission: 495 nm, bandwidth 10 nm. Lag time: 100 psec. Integration time: 400 psec. Flashes: 30.

[0601] Label 2: Excitation: 340 nm, bandwidth 30 nm; Emission: 520 nm, bandwidth 25 nm. Lag time: 100 psec. Integration time: 400 psec. Flashes: 30

[0602] The data were analyzed using GraphPad Prism employing a 4-parameter sigmoidal curve fit.

Part II - Results

[0603] Experimental results are provided in Table 2 below. The symbol "++++" indicates an IC 50 less than 0.0500 μM. The symbol "+++" indicates an IC 50 in the range of 0.0500 μM to 0.5000 μM. The symbol "++" indicates an IC 50 in the range of greater than 0.5000 μM to 1.0000 μM. The symbol "+" indicates an IC 50 greater than 1.0000 μM. The symbol "N/A" indicates that no data was available.

Table 2.

INCORPORATION BY REFERENCE

[0604] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.

EQUIVALENTS

[0605] The disclosure may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the disclosure described herein.

Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.