Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
6-(6-MEMBERED HETEROARYL & ARYL)ISOQUINOLIN-3-YL CARBOXAMIDES AND PREPARATION AND USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2019/084497
Kind Code:
A1
Abstract:
Isoquinoline compounds for treating various diseases and pathologies are disclosed. More particularly, the present disclosure concerns the use of an isoquinoline compound or analogs thereof, in the treatment of disorders characterized by the activation of Wnt pathway signaling (e.g., cancer, abnormal cellular proliferation, angiogenesis, Alzheimer's disease, lung disease, inflammation, auto-immune diseases and osteoarthritis), the modulation of cellular events mediated by Wnt pathway signaling, as well as neurological conditions/disorders/diseases linked to overexpression of DYRK1A.

Inventors:
KC SUNIL KUMAR (US)
MAK CHI CHING (US)
HOFILENA BRIAN JOSEPH (US)
EASTMAN BRIAN WALTER (US)
CAO JIANGUO (US)
BOLLU VENKATAIAH (US)
CHIRUTA CHANDRAMOULI (US)
Application Number:
PCT/US2018/057854
Publication Date:
May 02, 2019
Filing Date:
October 26, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SAMUMED LLC (US)
KC SUNIL KUMAR (US)
MAK CHI CHING (US)
HOFILENA BRIAN JOSEPH (US)
MITTAPALLI GOPI KUMAR (US)
EASTMAN BRIAN WALTER (US)
CAO JIANGUO (US)
BOLLU VENKATAIAH (US)
CHIRUTA CHANDRAMOULI (US)
International Classes:
C07D401/14; A61K31/4725; A61P25/00; C07D401/04; C07D403/04; C07D403/14; C07D413/14; C07D471/04; C07D498/04
Domestic Patent References:
WO2016046530A12016-03-31
WO2013169793A22013-11-14
WO1987005297A11987-09-11
WO2001053268A22001-07-26
WO2005009997A12005-02-03
Foreign References:
US201213614296A2012-09-13
US201314019229A2013-09-05
US201514664517A2015-03-20
US201514664517A2015-03-20
US6440102B12002-08-27
US6648873B22003-11-18
US6120484A2000-09-19
US4164559A1979-08-14
US4474752A1984-10-02
US6911211B22005-06-28
US6377849B12002-04-23
US33781506A2006-01-24
Other References:
YUANJUN HE ET AL: "Synthesis and SAR of novel quinazolines as potent and brain-penetrant c-jun N-terminal kinase (JNK) Inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, AMSTERDAM, NL, vol. 21, no. 6, 19 January 2011 (2011-01-19), pages 1719 - 1723, XP028169562, ISSN: 0960-894X, [retrieved on 20110122], DOI: 10.1016/J.BMCL.2011.01.079
LIANG JUN ET AL: "Identification of an imidazopyridine scaffold to generate potent and selective TYK2 inhibitors that demonstrate activity in an in vivo psoriasis model", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, AMSTERDAM, NL, vol. 27, no. 18, 12 August 2017 (2017-08-12), pages 4370 - 4376, XP085186059, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2017.08.022
"Goodman and Gilman's: The Pharmacological Basis of Therapeutics", 2017, THE MCGRAW-HILL COMPANIES
CHOU; TALALAY, ADVANCES IN ENZYME REGULATION, vol. 22, 1984, pages 27 - 55
THE AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, vol. 184, no. 1, 2011, pages 92 - 99
BRITISH JOURNAL OF PHARMACOLOGY, vol. 163, no. 1, 2011, pages 141 - 172
"Remington: The Science and Practice of Pharmacy", 2012, PHARMACEUTICAL PRESS
LEYNS, L. ET AL., CELL, vol. 88, no. 6, 1997, pages 747 - 756
"March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2013, JOHN WILEY & SONS
CAREY; SUNDBERG: "Advanced Organic Chemistry", 2007, SPRINGER
"Comprehensive Organic Transformations: A Guide to Functional Group Transformations", 1999, JOHN WILEY & SONS
P. WUTS: "Greene's Protective Groups in Organic Synthesis", 2014, JOHN WILEY & SONS
XIAOQIU LIU,ET AL.: "Fibrotic Lung Fibroblasts Show Blunted Inhibition by cAMP Due to Deficient cAMP Response Element-Binding Protein Phosphorylation", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 315, no. 2, 2005, pages 678 - 687
WATTS, K. L.: "RhoA signaling modulates cyclin D1 expression in human lung fibroblasts; implications for idiopathic pulmonary fibrosis", RESPIRATORY RESEARCH, vol. 7, no. 1, 2006, pages 88, XP021013367, DOI: doi:10.1186/1465-9921-7-88
Attorney, Agent or Firm:
DORIGO, Andrea et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound, or a pharmaceutically acceptable salt thereof, of Formula I:

I

wherein:

R1, R2, R4, and R5 are independently selected from the group consisting of H, halide, unsubstituted -(C1-3 haloalkyl), and unsubstituted -(C1-3 alkyl);

R3 is selected from the group consisting of -aryl optionally substituted with 1-5 R7 and a 6-10-membered heteroaryl optionally substituted with 1-4 R8;

R6 is selected from the group consisting of -(C1-4 alkylene)pheteroaryl optionally substituted with 1-6 R9, -(C1-4 alkylene )paryl optionally substituted with 1-6 R25, -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R10, -(Ci-4 alkylene)pcarbocyclyl optionally substituted with 1-12 R11, -(C1-4 alkylene)N(R12)(R13), -N(R14)(R15), -CF(Ci-9 alkyl)2, -(C1-4 alkylene)pO(Ci-9 alkyl), and -(C2-9 alkynyl) optionally substituted with one or more halides; wherein each alkyl of -CF(Ci-9 alkyl)2 is, independently, optionally substituted with one or more halides; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

R7 is selected from the group consisting of halide and -N(R16)2;

R8 is selected from the group consisting of halide, unsubstituted -(C1-9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(Ci-9 haloalkyl), -CN, -N(R17)(R18), - (Ci-4 alkylene)pX1R19, and -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R21; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R9 is independently selected from the group consisting of halide, unsubstituted -(Ci- 9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C 1-9 haloalkyl), -X2R22, -C(=0)N(R23)2, -(Ci-4 alkylene)pN(R24)2, -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R25, and -(C1-4 alkylene )pcarbocyclyl optionally substituted with 1-12 R26; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl); each R10 is independently selected from the group consisting of halide, unsubstituted -(Ci- 9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(Ci-9 haloalkyl), -(Ci-4 alkylene)pOR27, -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R28, -(C1-4 alkylene)pcarbocyclyl optionally substituted with 1-12 R29, -C(=0)(R30), -(C1-4 alkylene)pC(=0)OR30, -N(R20)2, -(C1-4 alkylene)aryl optionally substituted with one or more halides, -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides, and -S02(R31); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

alternatively, if two R10 are attached to the same carbon atom can together represent =0 to form a carbonyl group;

each R11 is independently selected from the group consisting of halide, unsubstituted -(Ci- 9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(Ci-9 haloalkyl), -(CM alkylene)pOR27, -N(R20)2, -C(=0)(R30), -C(=0)OR30, -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R28, and -(C1-4 alkylene)pcarbocyclyl optionally substituted with 1-12 R29; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

R12 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C1-9 haloalkyl), -(C1-4 alkylene )pheterocyclyl optionally substituted with 1-10 R32, and -carbocyclyl optionally substituted with 1-12 R33; wherein -(C1-4 alkylene) is, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

R13 is attached to the nitrogen and is selected from the group consisting of unsubstituted - (Ci-9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C1-9 haloalkyl), -(C1-4 alkylene )pheterocyclyl optionally substituted with 1-10 R32, and -carbocyclyl optionally substituted with 1-12 R33; wherein -(C1-4 alkylene) is, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

R14 is attached to the nitrogen and selected from the group consisting of H, unsubstituted - (Ci-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

R15 is attached to the nitrogen and is selected from the group consisting of -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R32, and— (C1-4 alkylene )pcarbocyclyl optionally substituted with 1-12 R33; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl); R16 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(C1-4 alkylene)paryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

alternatively, two adjacent R16 are taken together to form a -heterocyclyl ring optionally substituted with 1-10 R32;

R17 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

R18 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), - (C=0)R34, and -(C1-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R19 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein -(C1-4 alkylene) is optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R20 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

each R21 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -N(R20)2, -CN, and -OH;

each R22 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(Ci-4 alkylene)N(R 4)2, -(C1-4 alkylene )paryl optionally substituted with 1-10 R35, -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-12 R36, and -(C1-4 alkylene )pcarbocyclyl optionally substituted with 1-12 R29; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl); each R23 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

each R24 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene)N(R 4)2; wherein -(C1-4 alkylene) is optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R25 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -CN, -C(=0)R37, -N(R 8)2, -(Ci-4 alkylene)pOR27, -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R32, and -(C1-4 alkylene )pcarbocyclyl optionally substituted with 1-12 R26; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R26 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -CN;

each R27 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene)pcarbocyclyl optionally substituted with 1-12 R29, and -(C1-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(Ci-5 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R28 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -CN, and -(C1-4 alkylene)pcarbocyclyl optionally substituted with 1-12 R29; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R29 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -CN;

R30 is selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted - (C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(C1-4 alkylene)paryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

R31 is selected from the group consisting of unsubstituted -(C1-5 alkyl), unsubstituted -(C2- 5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(C1-4 alkylene)paryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein -(C1-4 alkylene) is, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R32 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(C 1-4 alky lene)pcarbocyclyl optionally substituted with 1-12 R29; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted - (Ci-3 alkyl);

each R33 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(C 1-4 alky lene)pcarbocyclyl optionally substituted with 1-12 R29; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted - (Ci-3 alkyl);

R34 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

each R35 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -CN;

each R36 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -CN, -OH, -C(=0)R37, -N(R 8)2, and -(C1-4 alkylene)pcarbocyclyl optionally substituted with 1- 12 R29; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R37 is a heteroaryl optionally substituted with 1 -6 R39;

each R38 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene)N(R 4)2; wherein -(C1-4 alkylene) is optionally substituted with one or more halides or one or more unsubstituted -(C1-3 alkyl);

each R39 is a -heterocyclyl optionally substituted with 1-10 R29;

each X1 is selected from the group consisting of O and S;

each X2 is selected from the group consisting of O, S, and NR34;

each p is independently 0 or 1 ; and

rmula I is not a structure selected from the group consisting of:

2. The compound of claim 1, wherein R1, R2, R4, and R5 are H.

3. The compound according to any one of claims 1-2, wherein R3 is phenyl optionally substituted with 1-2 R7.

4. The compound according to any one of claims 1 -3, wherein R3 is pyridinyl optionally substituted with 1 R8.

5. The compound according to any one of claims 1-4, wherein R3 is pyrimidinyl optionally substituted with 1 R8.

6. The compound according to any one of claims 1-5, wherein R3 is pyridazinyl optionally substituted with 1 R8.

7. The compound according to any one of claims 1-6, wherein R6 is -heterocyclyl optionally substituted with 1-2 R10.

8. The compound according to any one of claims 1 -7, wherein R6 is a -CFbheterocyclyl optionally substituted with 1-2 R10.

9. The compound according to any one of claims 1-8, wherein R6 is either a piperidinyl or a pyrrolidinyl both optionally substituted with 1-2 R10.

10. The compound according to any one of claims 1-9, wherein R6 is a piperidinyl substituted with one - N(R20)2.

11. The compound according to any one of claims 1-10, wherein each R20 is independently selected from the group consisting of H, Me, and -Ci-4 haloalkyl.

12. The c mpound according to any one of claims 1-11, wherein R6 is selected from the

group consisting of , and q is 1-2.

13. The compound according to any one of claims 1-12, wherein R6 is -carbocyclyl substituted with 1-2 R11.

14. The compound according to any one of claims 1-13, wherein R6 is cyclohexyl substituted with 1 R11.

15. The compound according to any one of claims 1-14, wherein the compound of

196

16. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1-15, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

17. A method of treating a disorder or disease in a patient, wherein the disorder or disease is selected from the group consisting of: chronic inflammation, systemic inflammation, diabetes, cancer, pulmonary fibrosis, idiopathic pulmonary fibrosis (IPF), degenerative disc disease, bone/osteoporotic fractures, a bone or cartilage disease, a neurological condition/disorder/disease, osteoarthritis, lung disease, a fibrotic disorder, the method comprising administering to the patient a therapeutically effective amount of a compound according to any one of claims 1-15, or a pharmaceutically acceptable salt, or a pharmaceutical composition.

18. A method of claim 17, wherein the disorder or disease is cancer.

19. A method of claim 17, wherein the disorder or disease is pulmonary fibrosis.

20. A method of claim 17, wherein the disorder or disease is idiopathic pulmonary fibrosis (IPF).

21. The method of claim 17, wherein the disorder or disease is lung disease.

22. A method of claim 17, wherein the disorder or disease is degenerative disc disease.

23. A method of claim 17, wherein the disorder or disease is a bone/osteoporotic fracture .

24. A method of claim 17, wherein the disorder or disease is a bone or cartilage disease.

25. A method of claim 17, wherein the disorder or disease is osteoarthritis.

26. A method of claim 17, wherein the disorder or disease is a neurological condition/disorder/disease .

27. The method of claim 18, wherein the cancer is selected from the group consisting of: osteoma, hemangioma, granuloma, xanthoma, osteitis deformans, meningioma, meningiosarcoma, and gliomatosis, astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, oligodendrocytoma, schwannoma, retinoblastoma, and congenital tumors, neurofibroma, meningioma, glioma, and sarcoma.

28. The method of claim 26, wherein the disorder or disease is a neurological condition/disorder/disease, wherein the neurological condition/disorder/disease is selected from the group consisting of: frontotemporal dementias, dementia with lewy bodies, prion diseases, multiple system atrophy, inclusion body myositis, degenerative myopathies, diabetic neuropathy, other metabolic neuropathies, endocrine neuropathies, orthostatic hypotension, and Charcot-Marie- Tooth disease.

29. The method of claim 26, wherein the disorder or disease is a neurological condition/disorder/disease, wherein the neurological condition/disorder/disease is selected from the group consisting of: Alzheimer's disease, amyotrophic lateral sclerosis (ALS), down syndrome, frontotemporal dementia (FTD) including FTD with Parkinsonism- 17 (FTDP-17), behavioural variant frontotemporal dementia (bvFTD), FTD in patients with motor neuron disease (MND) (typically amyotrophic lateral sclerosis, also called FTD -ALS), corticobasal degeneration (CBD) (also called corticobasal ganglionic degeneration), progressive supranuclear palsy, primary progressive aphasia (PPA), Prion Diseases, globular glial tauopathy (GGT), myotonic dystrophy type 1 (DM1) (also called Steinert disease), myotonic dystrophy type 2 (DM2) (also called proximal myotonic myopathy), Guam complex, argyrophilic grain disease, dementia pugilistica, postencephalitic parkinsonism, lewy body dementia, Parkinson's disease, Pick's disease, and additional diseases with pronounced neurodegeneration such as autism, dementia, epilepsy, Huntington's disease, multiple sclerosis; diseases and disorders associated with acquired brain injury such as chronic traumatic encephalopathy, traumatic brain injury, tumor, and stroke.

30. A method of claim 29, wherein the disorder or disease is Alzheimer's disease.

31. The method of claim 26, wherein the disorder or disease is a neurological condition/disorder/disease associated with tau protein, amyloid, alpha-synuclein, Tar DNA-binding Protein of 43KDa (TDP-43), Prion protein PrP or fused in sarcoma (FUS) pathology.

32. The method of claim 17, wherein the disorder or disease is a fibrotic disorder, wherein the fibrotic disorder is selected from the group consisting of: skin fibrosis; scleroderma; progressive systemic fibrosis; lung fibrosis; muscle fibrosis; kidney fibrosis; glomerulosclerosis; glomerulonephritis; hypertrophic scar formation; uterine fibrosis; renal fibrosis; cirrhosis of the liver, liver fibrosis; adhesions; chronic obstructive pulmonary disease; fibrosis following myocardial infarction; pulmonary fibrosis; fibrosis and scarring associated with diffuse/interstitial lung disease; central nervous system fibrosis; fibrosis associated with proliferative vitreoretinopathy (PVR); restenosis; endometriosis; ischemic disease, and radiation fibrosis.

33. The method of claim 17, wherein the disorder or disease is systemic inflammation.

34. The method of claim 17, wherein the disorder or disease is chronic inflammation associated eye disorders, joint pain, arthritis (rheumatoid, osteo, psoriatic gout), cancers (colon, breast, lung, pancreas, and others), gastrointestinal disorders (ulcerative colitis and inflammatory bowel diseases), pulmonary disorders (chronic obstructive pulmonary disorder and asthma), allergies, skin disorders (atopic dermatitis and psoriasis), diabetes, pancreatitis, tendonitis, hepatitis, heart disease, myocarditis, stroke, lupus, and neurological disorders such as multiple sclerosis, Parkinson's and dementia including Alzheimer's disease.

35. The method of claim 17, wherein the patient is a human.

36. The method of claim 17, wherein the compound inhibits one or more proteins in the Wnt pathway.

37. The method of claim 17, wherein the compound inhibits signaling induced by one or more Wnt proteins.

38. The method of claim 17, wherein the compound inhibits DYRK1A.

39. The method of claim 17, wherein the compound inhibits GSK3 .

40. The method of claim 17, wherein the compound inhibits DYRKIA and GSK3 .

41. The method of claim 17, wherein the compound inhibits a kinase activity.

42. A method of preventing or reducing angiogenesis in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound according to any one of claims 1-15, or a pharmaceutically acceptable salt, or a pharmaceutical composition.

Description:
6-(6-MEMBERED HETEROARYL & ARYL)ISOQUINOLIN-3-YL

CARBOXAMIDES AND PREPARATION AND USE THEREOF

RELATED APPLICATION

[001] This application claims the benefit of U.S. Provisional Application No. 62/577,818, filed October 27, 2017, which is incorporated herein by reference in its entirety.

BACKGROUND

Technical Field

[002] This disclosure relates to inhibitors of one or more proteins in the Wnt pathway, including inhibitors of one or more Wnt proteins, and compositions comprising the same. More particularly, it concerns the use of an isoquinoline compound or salts or analogs thereof, in the treatment of disorders characterized by the activation of Wnt pathway signaling (e.g., cancer, abnormal cellular proliferation, angiogenesis, Alzheimer's disease, lung disease, inflammation, auto-immune diseases fibrotic disorders, cartilage (chondral) defects, and osteoarthritis), the modulation of cellular events mediated by Wnt pathway signaling, as well as genetic diseases and neurological conditions/disorders/diseases due to mutations or dysregulation of the Wnt pathway and/or of one or more of Wnt signaling components. Also provided are methods for treating Wnt- related disease states, as well as neurological conditions/disorders/diseases linked to overexpression of DYRK1A.

Background

[003] The Wnt growth factor family includes more than 10 genes identified in the mouse and at least 19 genes identified in the human. Members of the Wnt family of signaling molecules mediate many short-and long-range patterning processes during invertebrate and vertebrate development. The Wnt signaling pathway is known for its role in the inductive interactions that regulate growth and differentiation, and it also plays roles in the homeostatic maintenance of post-embryonic tissue integrity. Wnt stabilizes cytoplasmic β-catenin, which stimulates the expression of genes including c-myc, c jun, fra-1, and cyclin Dl. In addition, misregulation of Wnt signaling can cause developmental defects and is implicated in the genesis of several human cancers. The Wnt pathway has also been implicated in the maintenance of stem or progenitor cells in a growing list of adult tissues including skin, blood, gut, prostate, muscle, and the nervous system. [004] Dual specificity tyrosine-phosphorylation-regulated kinase 1A is an enzyme that in humans is encoded by the DYRKIA gene. DYRKIA is a member of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) family. DYRKIA contains a nuclear targeting signal sequence, a protein kinase domain, a leucine zipper motif, and a highly conservative 13 - consecutive-histidine repeat. It catalyzes its autophosphorylation on serine/threonine and tyrosine residues. It may play a significant role in a signaling pathway regulating cell proliferation and may be involved in brain development. DYRKIA is localized in the Down syndrome critical region of chromosome 21 , and is considered to be a candidate gene for learning defects associated with Down syndrome. DYRKIA is also expressed in adult brain neurons, indicating that DYRKIA may play a role in the mature central nervous system. Thus, several lines of evidence point to some synaptic functions of DYRKIA. For instance, it has been found that DYRKIA phosphorylates and modulates the interaction of several components of the endocytic protein complex machinery (Dynamin 1, Amphiphysin, and Synaptojanin), suggesting a role in synaptic vesicle recycling. In addition, a polymorphism (SNP) in DYRKIA was found to be associated with HIV-1 replication in monocyte-derived macrophages, as well as with progression to AIDS in two independent cohorts of HIV-1 -infected individuals .

SUMMARY

[005] The present disclosure provides methods and reagents, involving contacting a cell with an agent, such as an isoquinoline compound, in a sufficient amount to antagonize a Wnt activity, e.g., to reverse or control an aberrant growth state or correct a genetic disorder due to mutations in Wnt signaling components.

[006] The present disclosure also provides methods and reagents, involving contacting a cell with an agent, such as an isoquinoline compound, in a sufficient amount to antagonize DYRKIA activity, e.g., i) to normalize prenatal and early postnatal brain development; ii) to improve cognitive function in youth and adulthood; and/or iii) to attenuate Alzheimer' s-type neurodegeneration.

[007] Some embodiments disclosed herein include Wnt and/or DYRKIA inhibitors containing an isoquinoline core. Other embodiments disclosed herein include pharmaceutical compositions and methods of treatment using these compounds.

[008] One embodiment disclosed herein includes a compound having the structure of Formula I:

I

as well as prodrugs and pharmaceutically acceptable salts thereof.

[009] In some embodiments of Formula (I):

R 1 , R 2 , R 4 , and R 5 are independently selected from the group consisting of H, halide, unsubstituted -(C1-3 haloalkyl), and unsubstituted -(C1-3 alkyl);

R 3 is selected from the group consisting of -aryl optionally substituted with 1-5 R 7 and a 6-10-membered heteroaryl optionally substituted with 1-4 R 8 ;

R 6 is selected from the group consisting of -(C1-4 alkylene) p heteroaryl optionally substituted with 1-6 R 9 , -(C1-4 alkylene) p aryl optionally substituted with 1-6 R 25 , -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-10 R 10 , -(Ci-4 alkylene) p carbocyclyl optionally substituted with 1-12 R 11 , -(C1-4 alkylene)N(R 12 )(R 13 ), -N(R 14 )(R 15 ), -CF(Ci- 9 alkyl) 2 , -(C1-4 alkylene) p O(Ci-9 alkyl), and -(C2-9 alkynyl) optionally substituted with one or more halides; wherein each alkyl of -CF(Ci-9 alkyl) 2 is, independently, optionally substituted with one or more halides; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

R 7 is selected from the group consisting of halide and -N(R 16 )2;

R 8 is selected from the group consisting of halide, unsubstituted -(C1-9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(Ci -9 haloalkyl), -CN, -N(R 17 )(R 18 ), - (Ci-4 alkylene)pX 1 R 19 , and -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R 21 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 9 is independently selected from the group consisting of halide, unsubstituted -(Ci- 9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C 1-9 haloalkyl), -X 2 R 22 , -C(=0)N(R 23 ) 2 , -(Ci-4 alkylene) p N(R 24 ) 2 , -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-10 R 25 , and -(C1-4 alkylene ) p carbocyclyl optionally substituted with 1-12 R 26 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 10 is independently selected from the group consisting of halide, unsubstituted -(Ci- 9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C 1-9 haloalkyl), -(Ci-4 alkylene) p OR 27 , -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-10 R 28 , -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 R 29 , -C(=0)(R 30 ), -(C1-4 alkylene)pC(=0)OR 30 , -N(R 20 )2, -(C1-4 alkylene)aryl optionally substituted with one or more halides, -(Ci-4 alkylene)pheteroaryl optionally substituted with one or more halides, and -S02(R 31 ); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

alternatively, if two R 10 are attached to the same carbon atom can together represent =0 to form a carbonyl group;

each R 11 is independently selected from the group consisting of halide, unsubstituted -(Ci- 9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(Ci-9 haloalkyl), -(Ci-4 alkylene) p OR 27 , -N(R 20 ) 2 , -C(=0)(R 30 ), -C(=0)OR 30 , -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R 28 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

R 12 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C1-9 haloalkyl), -(C1-4 alkylene ) p heterocyclyl optionally substituted with 1-10 R 32 , and -carbocyclyl optionally substituted with 1-12 R 33 ; wherein -(C1-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein;

R 13 is attached to the nitrogen and is selected from the group consisting of unsubstituted - (Ci-9 alkyl), unsubstituted -(C2-9 alkenyl), unsubstituted -(C2-9 alkynyl), unsubstituted -(C1-9 haloalkyl), -(C1-4 alkylene ) p heterocyclyl optionally substituted with 1-10 R 32 , and -carbocyclyl optionally substituted with 1-12 R 33 ; wherein -(C1-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein;

R 14 is attached to the nitrogen and selected from the group consisting of H, unsubstituted - (Ci-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

R 15 is attached to the nitrogen and is selected from the group consisting of -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-10 R 32 , and— (C1-4 alkylene ) p carbocyclyl optionally substituted with 1-12 R 33 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

R 16 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(C1-4 alkylene) p aryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

alternatively, two adjacent R 16 are taken together to form a -heterocyclyl ring optionally substituted with 1-10 R 32 ;

R 17 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

R 18 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), - (C=0)R 34 , and -(C1-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 19 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein -(C1-4 alkylene) is optionally substituted with one or more substituents as defined anywhere herein;

each R 20 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

each R 21 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -N(R 20 ) 2 , -CN, and -OH;

each R 22 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(Ci-4 alkylene)N(R 4 )2, -(C1-4 alkylene ) p aryl optionally substituted with 1-10 R 35 , -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-12 R 36 , and -(C1-4 alkylene ) p carbocyclyl optionally substituted with 1-12 R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 23 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl); each R 24 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene)N(R 4 )2; wherein -(C1-4 alkylene) is optionally substituted with one or more substituents as defined anywhere herein;

each R 25 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -CN, -C(=0)R 37 , -N(R 38 ) 2 , -(CM alkylene) p OR 27 , -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 R 32 , and -(C1-4 alkylene ) p carbocyclyl optionally substituted with 1-12 R 26 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 26 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -CN;

each R 27 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene) p carbocyclyl optionally substituted with 1-12 R 29 , and -(C1-4 alkylene) p heterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(Ci-5 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 28 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -CN, and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 29 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -CN;

R 30 is selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted - (C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(C1-4 alkylene) p aryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein; R 31 is selected from the group consisting of unsubstituted -(C1-5 alkyl), unsubstituted -(C2- 5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -(C1-4 alkylene) p aryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl); wherein -(C1-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein;

each R 32 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(C 1-4 alky lene) p carbocyclyl optionally substituted with 1-12 R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein; each R 33 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(C 1-4 alky lene) p carbocyclyl optionally substituted with 1-12 R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

R 34 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), and unsubstituted -(C1-5 haloalkyl);

each R 35 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -CN;

each R 36 is independently selected from the group consisting of halide, unsubstituted -(Ci- 5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), -CN, -OH, -C(=0)R 37 , -N(R 8 ) 2 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1 - 12 R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

each R 37 is a heteroaryl optionally substituted with 1 -6 R 39 ;

each R 38 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl), unsubstituted -(C2-5 alkenyl), unsubstituted -(C2-5 alkynyl), unsubstituted -(C1-5 haloalkyl), and -(Ci-4 alkylene )N(R 4 )2; wherein -(C1-4 alkylene) is optionally substituted with one or more substituents as defined anywhere herein;

each R 39 is a -heterocyclyl optionally substituted with 1-10 R 29 ;

each X 1 is selected from the group consisting of O and S;

each X 2 is selected from the group consisting of O, S, and NR 34 ; each p is independently 0 or 1 ;

rmula I is not a structure selected from the group consisting

[010] Some embodiments include stereoisomers and pharmaceutically acceptable salts of a compound of Formula (I). Some embodiments include pharmaceutically acceptable salts of a compound of Formula (I).

[Oil] Some embodiments include pro-drugs of a compound of Formula (I).

[012] Some embodiments of the present disclosure include pharmaceutical compositions comprising a compound of Formula (I) and a pharmaceutically acceptable carrier, diluent, or excipient.

[013] Other embodiments disclosed herein include methods of inhibiting one or more members of the Wnt pathway, including one or more Wnt proteins by administering to a patient affected by a disorder or disease in which aberrant Wnt signaling is implicated, such as cancer and other diseases associated with abnormal angiogenesis, cellular proliferation, cell cycling and mutations in Wnt signaling components, a compound according to Formula (I). Accordingly, the compounds and compositions provided herein can be used to treat cancer, to reduce or inhibit angiogenesis, to reduce or inhibit cellular proliferation and correct a genetic disorder due to mutations in Wnt signaling components.

[014] Other embodiments disclosed herein include methods of inhibiting DYRK1A by administering to a patient affected by a disorder or disease in which DYRK1A overexpression is implicated, such as Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Down Syndrome, Frontotemporal Dementia with Parkinsonism- 17 (FTDP-17), Lewy body dementia, Parkinson's Disease, Pick's Disease, and additional diseases with pronounced neurodegeneration such as Autism, Dementia, Epilepsy, Huntington's Disease, Multiple Sclerosis; diseases and disorders associated with acquired brain injury such as Chronic Traumatic Encephalopathy, Traumatic Brain Injury, Tumor and Stroke.

[015] Non-limiting examples of diseases which can be treated with the compounds and compositions provided herein include a variety of cancers, diabetic retinopathy, pulmonary fibrosis, rheumatoid arthritis, sepsis, ankylosing spondylitis, psoriasis, scleroderma, mycotic and viral infections, osteochondrodysplasia, Alzheimer's disease, lung disease, bone/osteoporotic (wrist, spine, shoulder and hip) fractures, articular cartilage (chondral) defects, degenerative disc disease (or intervertebral disc degeneration), polyposis coli, osteoporosis-pseudoglioma syndrome, familial exudative vitreoretinopathy, retinal angiogenesis, early coronary disease, tetra-amelia syndrome, Miillerian-duct regression and virilization, SERKAL syndrome, diabetes mellitus type 2, Fuhrmann syndrome, Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndrome, odonto- onycho-dermal dysplasia, obesity, split-hand/foot malformation, caudal duplication syndrome, tooth agenesis, Wilms tumor, skeletal dysplasia, focal dermal hypoplasia, autosomal recessive anonychia, neural tube defects, alpha-thalassemia (ATRX) syndrome, fragile X syndrome, ICF syndrome, Angelman syndrome, Prader-Willi syndrome, Beckwith-Wiedemann Syndrome, Norrie disease, and Rett syndrome.

[016] Some embodiments of the present disclosure include methods to prepare compounds of Formula (I).

[017] It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the disclosure, as claimed.

DETAILED DESCRIPTION

[018] Provided herein are compositions and methods for inhibiting one or more members of the Wnt pathway, including one or more Wnt proteins. Other Wnt inhibitors and methods for using the same are disclosed in U.S. Application Ser. Nos. 13/614,296; 14/019,229; and 14/664,517, all of which are incorporated by reference in their entirety herein.

[019] Provided herein are compositions and methods for inhibiting DYRK1A. Other DYRK1A inhibitors and methods for using the same are disclosed in U.S. Application Ser. No. 14/664,517, which is incorporated by reference in its entirety herein.

[020] Some embodiments provided herein relate to a method for treating a disease including, but not limited to, neurological diseases or disorders, cancers, chronic inflammation, diabetic retinopathy, pulmonary fibrosis, rheumatoid arthritis, sepsis, ankylosing spondylitis, psoriasis, scleroderma, mycotic and viral infections, bone and cartilage diseases, lung disease, osteoarthritis, articular cartilage (chondral) defects, degenerative disc disease (or intervertebral disc degeneration), polyposis coli, bone density and vascular defects in the eye (Osteoporosis - pseudoglioma Syndrome, OPPG), familial exudative vitreoretinopathy, retinal angiogenesis, early coronary disease, tetra-amelia, Miillerian-duct regression and virilization, SERKAL syndrome, type II diabetes, Fuhrmann syndrome, Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndrome, odonto-onycho-dermal dysplasia, obesity, split-hand/foot malformation, caudal duplication, tooth agenesis, Wilms tumor, skeletal dysplasia, focal dermal hypoplasia, autosomal recessive anonychia, neural tube defects, alpha-thalassemia (ATRX) syndrome, fragile X syndrome, ICF syndrome, Angelman's syndrome, Prader-Willi syndrome, Beckwith-Wiedemann Syndrome, Nome disease, and Rett syndrome.

[021] In some embodiments, non-limiting examples of bone and cartilage diseases which can be treated with the compounds and compositions provided herein include bone spur (osteophytes), craniosynostosis, fibrodysplasia ossificans progressive, fibrous dysplasia, giant cell tumor of bone, hip labral tear, meniscal tears, osteoarthritis, articular cartilage (chondral) defects, degenerative disc disease (or intervertebral disc degeneration), osteochondritis dissecans, osteochondroma (bone tumor), osteopetrosis, relapsing polychondritis, and Salter-Harris fractures.

[022] In some embodiments, non-limiting examples of a neurological disease or disorder associated with tau protein, amyloid or alpha-synuclein pathology which can be treated with the compounds and compositions provided herein include, but are not limited to, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Down Syndrome, Frontotemporal Dementia with Parkinsonism- 17 (FTDP-17), Lewy body dementia, Parkinson's Disease, Pick's Disease, and additional diseases with pronounced neurodegeneration such as Autism, Dementia, Epilepsy, Huntington's Disease, Multiple Sclerosis; diseases and disorders associated with acquired brain injury such as Chronic Traumatic Encephalopathy, Traumatic Brain Injury, Tumor, and Stroke.

[023] In some embodiments, non-limiting examples of diseases in which chronic inflammation is involved which can be treated with the compounds and compositions provided herein include eye disorders, joint pain, arthritis (rheumatoid, osteo, psoriatic gout), cancers (colon, breast, lung, pancreas, and others), gastrointestinal disorders (ulcerative colitis and inflammatory bowel diseases), pulmonary disorders (chronic obstructive pulmonary disorder and asthma), allergies, skin disorders (atopic dermatitis and psoriasis), diabetes, pancreatitis, tendonitis, hepatitis, heart disease, myocarditis, stroke, lupus, and neurological disorders such as multiple sclerosis, Parkinson's and dementia including Alzheimer's disease.

[024] In some embodiments, non-limiting examples of cancers which can be treated with the compounds and compositions provided herein include colon, ovarian, pancreatic, breast, liver, prostate, and hematologic cancers.

[025] In some embodiments, pharmaceutical compositions are provided that are effective for treatment of a disease of an animal, e.g., a mammal, caused by either the pathological activation or mutations of the Wnt pathway or DYRK 1 A overexpression. The composition includes a pharmaceutically acceptable carrier and a compound as described herein. Definitions

[026] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents, applications, published applications, and other publications are incorporated by reference in their entirety. In the event that there is a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.

[027] As used herein, "alkyl" means a branched, or straight chain chemical group containing only carbon and hydrogen, such as methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso- butyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, sec-pentyl and neo-pentyl. Alkyl groups can either be unsubstituted or substituted with one or more substituents. In some embodiments, alkyl groups include 1 to 9 carbon atoms (for example, 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms).

[028] As used herein, "alkenyl" means a straight or branched chain chemical group containing only carbon and hydrogen and containing at least one carbon-carbon double bond, such as ethenyl, 1-propenyl, 2-propenyl, 2-methyl-l -propenyl, 1-butenyl, 2-butenyl, and the like. In various embodiments, alkenyl groups can either be unsubstituted or substituted with one or more substituents. Typically, alkenyl groups will comprise 2 to 9 carbon atoms (for example, 2 to 6 carbon atoms, 2 to 4 carbon atoms, or 2 carbon atoms).

[029] As used herein, "alkynyl" means a straight or branched chain chemical group containing only carbon and hydrogen and containing at least one carbon-carbon triple bond, such as ethynyl, 1-propynyl, 1 -butynyl, 2-butynyl, and the like. In various embodiments, alkynyl groups can either be unsubstituted or substituted with one or more substituents. Typically, alkynyl groups will comprise 2 to 9 carbon atoms (for example, 2 to 6 carbon atoms, 2 to 4 carbon atoms, or 2 carbon atoms).

[030] As used herein, "alkylene" means a bivalent branched, or straight chain chemical group containing only carbon and hydrogen, such as methylene, ethylene, n-propylene, iso-propylene, n-butylene, iso-butylene, sec-butylene, tert-butylene, n-pentylene, iso-pentylene, sec-pentylene and neo-pentylene. Alkylene groups can either be unsubstituted or substituted with one or more substituents. In some embodiments, alkylene groups include 1 to 9 carbon atoms (for example, 1 to 6 carbon atoms, 1 to 4 carbon atoms, or 1 to 2 carbon atoms).

[031] As used herein, "alkenylene" means a bivalent branched, or straight chain chemical group containing only carbon and hydrogen and containing at least one carbon-carbon double bond, such as ethenylene, 1-propenylene, 2-propenylene, 2-methyl-l -propenylene, 1- butenylene, 2-butenylene, and the like. In various embodiments, alkenylene groups can either be unsubstituted or substituted with one or more substituents. Typically, alkenylene groups will comprise 2 to 9 carbon atoms (for example, 2 to 6 carbon atoms, 2 to 4 carbon atoms, or 2 carbon atoms).

[032] As used herein, "alkynylene" means a bivalent branched, or straight chain chemical group containing only carbon and hydrogen and containing at least one carbon-carbon triple bond, such as ethynylene, 1-propynylene, 1-butynylene, 2-butynylene, and the like. In various embodiments, alkynylene groups can either be unsubstituted or substituted with one or more substituents. Typically, alkynylene groups will comprise 2 to 9 carbon atoms (for example, 2 to 6 carbon atoms, 2 to 4 carbon atoms, or 2 carbon atoms).

[033] As used herein, "alkoxy" means an alkyl-0— group in which the alkyl group is as described herein. Exemplary alkoxy groups include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, s-butoxy, t-butoxy, pentoxy, hexoxy and heptoxy, and also the linear or branched positional isomers thereof.

[034] As used herein, "haloalkoxy" means a haloalkyl-0— group in which the haloalkyl group is as described herein. Exemplary haloalkoxy groups include fluoromethoxy, difluoromethoxy, trifluoromethoxy, and also the linear or branched positional isomers thereof.

[035] As used herein, "carbocyclyl" means a cyclic ring system containing only carbon atoms in the ring system backbone, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclohexenyl. Carbocyclyls may include multiple fused rings. Carbocyclyls may have any degree of saturation provided that none of the rings in the ring system are aromatic. Carbocyclyl groups can either be unsubstituted or substituted with one or more substituents. In some embodiments, carbocyclyl groups include 3 to 10 carbon atoms, for example, 3 to 6 carbon atoms.

[036] As used herein, "aryl" means a mono-, bi-, tri- or polycyclic group with only carbon atoms present in the ring backbone having 5 to 14 ring atoms, alternatively 5, 6, 9, or 10 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic. Aryl groups can either be unsubstituted or substituted with one or more substituents. Examples of aryl include phenyl, naphthyl, tetrahydronaphthyl, 2,3-dihydro-lH- indenyl, and others. In some embodiments, the aryl is phenyl.

[037] As used herein, "arylalkylene" means an aryl-alkylene- group in which the aryl and alkylene moieties are as previously described. In some embodiments, arylalkylene groups contain a Ci-4alkylene moiety. Exemplary arylalkylene groups include benzyl and 2-phenethyl.

[038] As used herein, the term "heteroaryl" means a mono-, bi-, tri- or polycyclic group having 5 to 14 ring atoms, alternatively 5, 6, 9, or 10 ring atoms; and having 6, 10, or 14 pi electrons shared in a cyclic array; wherein at least one ring in the system is aromatic, and at least one ring in the system contains one or more heteroatoms independently selected from the group consisting of N, O, and S. Heteroaryl groups can either be unsubstituted or substituted with one or more substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-<f]pyrimidinyl, pyrrolo[2,3-6]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-6]pyridinyl, pyrazolo[3,4- c]pyridinyl, pyrazolo[4,3-c]pyridine, pyrazolo[4,3-6]pyridinyl, tetrazolyl, chromane, 2,3- dihydrobenzo[6] [l,4]dioxine, benzo[<f] [ l,3]dioxole, 2,3-dihydrobenzofuran, tetrahydroquinoline, 2,3-dihydrobenzo[6] [l,4]oxathiine, isoindoline, and others. In some embodiments, the heteroaryl is selected from thienyl, pyridinyl, furyl, pyrazolyl, imidazolyl, isoindolinyl, pyranyl, pyrazinyl, and pyrimidinyl.

[039] As used herein, "halo", "halide" or "halogen" is a chloro, bromo, fluoro, or iodo atom radical. In some embodiments, a halo is a chloro, bromo or fluoro. For example, a halide can be fluoro.

[040] As used herein, "haloalkyl" means a hydrocarbon substituent, which is a linear or branched, alkyl, alkenyl or alkynyl substituted with one or more chloro, bromo, fluoro, and/or iodo atom(s). In some embodiments, a haloalkyl is a fluoroalkyls, wherein one or more of the hydrogen atoms have been substituted by fluoro. In some embodiments, haloalkyls are of 1 to about 3 carbons in length (e.g., 1 to about 2 carbons in length or 1 carbon in length). The term "haloalkylene" means a diradical variant of haloalkyl, and such diradicals may act as spacers between radicals, other atoms, or between a ring and another functional group.

[041] As used herein, "heterocyclyl" means a nonaromatic cyclic ring system comprising at least one heteroatom in the ring system backbone. Heterocyclyls may include multiple fused rings. Heterocyclyls may be substituted or unsubstituted with one or more substituents. In some embodiments, heterocycles have 3-1 1 members. In six membered monocyclic heterocycles, the heteroatom(s) are selected from one to three of O, N or S, and wherein when the heterocycle is five membered, it can have one or two heteroatoms selected from O, N, or S. Examples of heterocyclyl include azirinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, 1,4,2- dithiazolyl, dihydropyridinyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-dioxolanyl, morpholinyl, thiomorpholinyl, piperazinyl, pyranyl, pyrrolidinyl, tetrahydrofuryl, tetrahydropyridinyl, oxazinyl, thiazinyl, thiinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, isoxazolidinyl, piperidinyl, pyrazolidinyl imidazolidinyl, thiomorpholinyl, and others. In some embodiments, the heterocyclyl is selected from azetidinyl, morpholinyl, piperazinyl, pyrrolidinyl, and tetrahydropyridinyl.

[042] As used herein, "monocyclic heterocyclyl" means a single nonaromatic cyclic ring comprising at least one heteroatom in the ring system backbone. Heterocyclyls may be substituted or unsubstituted with one or more substituents. In some embodiments, heterocycles have 3-7 members. In six membered monocyclic heterocycles, the heteroatom(s) are selected from one to three of O, N or S, and wherein when the heterocycle is five membered, it can have one or two heteroatoms selected from O, N, or S. Examples of heterocyclyls include azirinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, 1,4,2-dithiazolyl, dihydropyridinyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3- dioxolanyl, morpholinyl, thiomorpholinyl, piperazinyl, pyranyl, pyrrolidinyl, tetrahydrofuryl, tetrahydropyridinyl, oxazinyl, thiazinyl, thiinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, isoxazolidinyl, piperidinyl, pyrazolidinyl imidazolidinyl, thiomorpholinyl, and others.

[043] As used herein, "bicyclic heterocyclyl" means a nonaromatic bicyclic ring system comprising at least one heteroatom in the ring system backbone. Bicyclic heterocyclyls may be substituted or unsubstituted with one or more substituents. In some embodiments, bicyclic heterocycles have 4-1 1 members with the heteroatom(s) being selected from one to five of O, N or S. Examples of bicyclic heterocyclyls include 2 -azabicyclo[ 1.1.0] butane, 2- azabicyclo[2.1.0]pentane, 2-azabicyclo[l . l . l]pentane, 3-azabicyclo[3.1.0]hexane, 5- azabicyclo[2.1.1]hexane, 3-azabicyclo[3.2.0]heptane, octahydrocyclopenta[c]pyrrole, 3- azabicyclo[4.1.0]heptane, 7-azabicyclo[2.2.1]heptane, 6-azabicyclo[3.1.1]heptane, 7- azabicyclo[4.2.0]octane, 2-azabicyclo[2.2.2]octane, and the like.

[044] As used herein, "spirocyclic heterocyclyl" means a nonaromatic bicyclic ring system comprising at least one heteroatom in the ring system backbone and with the rings connected through just one atom. Spirocyclic heterocyclyls may be substituted or unsubstituted with one or more substituents. In some embodiments, spirocyclic heterocycles have 5-1 1 members with the heteroatom(s) being selected from one to five of O, N or S. Examples of spirocyclic heterocyclyls include 2-azaspiro[2.2]pentane, 4-azaspiro[2.5]octane, l-azaspiro[3.5]nonane, 2- azaspiro[3.5]nonane, 7-azaspiro[3.5]nonane, 2-azaspiro[4.4]nonane, 6-azaspiro[2.6]nonane, 1,7- diazaspiro[4.5]decane, 2,5-diazaspiro[3.6]decane, and the like.

[045] The term "substituted" refers to moieties having substituents replacing a hydrogen on one or more non -hydrogen atoms of the molecule. It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. Substituents can include, for example, -(C1-9 alkyl) optionally substituted with one or more of hydroxyl, -NH2, -NH(Ci-3 alkyl), and -N(Ci-3 alkyl)2; -(C1-9 haloalkyl); a halide; a hydroxyl; a carbonyl [such as -C(0)OR, and -C(0)R]; a thiocarbonyl [such as -C(S)OR, -C(0)SR, and -C(S)R]; -(C1-9 alkoxy) optionally substituted with one or more of halide, hydroxyl, -NH2, -NH(Ci-3 alkyl), and -N(Ci-3 alkyl)2; - OPO(OH) 2 ; a phosphonate [such as -PO(OH) 2 and -PO(OR') 2 ]; -OPO(OR')R"; -NRR'; - C(0)NRR'; -C(NR)NR'R"; -C(NR')R"; a cyano; a nitro; an azido; -SH; -S-R; -OS0 2 (OR); a sulfonate [such as -S02(OH) and -S02(OR)]; -S02NR'R"; and -SO2R; in which each occurrence of R, R' and R" are independently selected from H; -(C1-9 alkyl); Ce-io aryl optionally substituted with from 1-3R'"; 5-10 membered heteroaryl having from 1-4 heteroatoms independently selected from N, O, and S and optionally substituted with from 1-3 R'"; C3-7 carbocyclyl optionally substituted with from 1-3 R'"; and 3-8 membered heterocyclyl having from 1-4 heteroatoms independently selected from N, O, and S and optionally substituted with from 1-3 R'"; wherein each R'" is independently selected from -(Ci-6 alkyl), -(C1-5 haloalkyl), ahalide (e.g., F), a hydroxyl, -C(0)OR, -C(0)R, -(Ci-6 alkoxyl), -NRR', -C(0)NRR', and a cyano, in which each occurrence of R and R' is independently selected from H and -(Ci-6 alkyl). In some embodiments, the substituent is selected from -(Ci-6 alkyl), -(Ci-β haloalkyl), a halide (e.g., F), a hydroxyl, -C(0)OR, -C(0)R, -(Ci-e alkoxyl), -NRR', -C(0)NRR', and a cyano, in which each occurrence of R and R' is independently selected from H and -(C1-5 alkyl).

[046] As used herein, when two groups are indicated to be "linked" or "bonded" to form a "ring", it is to be understood that a bond is formed between the two groups and may involve replacement of a hydrogen atom on one or both groups with the bond, thereby forming a carbocyclyl, heterocyclyl, aryl, or heteroaryl ring. The skilled artisan will recognize that such rings can and are readily formed by routine chemical reactions. In some embodiments, such rings have from 3-7 members, for example, 5 or 6 members.

[047] The skilled artisan will recognize that some chemical structures described herein may be represented on paper by one or more other resonance forms; or may exist in one or more other tautomeric forms, even when kinetically, the artisan recognizes that such tautomeric forms represent only a very small portion of a sample of such compound(s). Such compounds are clearly contemplated within the scope of this disclosure, though such resonance forms or tautomers are not explicitly represented herein.

[048] The compounds provided herein may encompass various stereochemical forms. The compounds also encompass diastereomers as well as optical isomers, e.g., mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and diastereomers, which arise as a consequence of structural asymmetry in certain compounds. Separation of the individual isomers or selective synthesis of the individual isomers is accomplished by application of various methods which are well known to practitioners in the art. Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.

[049] The present disclosure includes all pharmaceutically acceptable isotopically labeled compounds of Formula I wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature. Examples of isotopes suitable for inclusion in the compounds of the disclosure include, but are not limited to, isotopes of hydrogen, such as 2 H (deuterium) and H (tritium), carbon, such as n C, 1 C and 14 C, chlorine, such as 6 C1, fluorine, such as 18 F, iodine, such as 12 I and 125 I, nitrogen, such as 1 N and 15 N, oxygen, such as 15 0, 17 0 and 18 0, phosphorus, such as 2 P, and sulfur, such as 5 S.

[050] The term "administration" or "administering" refers to a method of providing a dosage of a compound or pharmaceutical composition to a vertebrate or invertebrate, including a mammal, a bird, a fish, or an amphibian, where the method is, e.g., orally, subcutaneously, intravenously, intralymphatic, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, ontologically, neuro-otologically, intraocularly, subconjuctivally, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, via wound irrigation, intrabuccally, intra-abdominally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intrameningeally, via inhalation, via endotracheal or endobronchial instillation, via direct instillation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, epidurally, intratympanically, intracisternally, intravascularly, intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with a prosthetic device . The method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, the site of the disease, the disease involved, and the severity of the disease.

[051] A "diagnostic" as used herein is a compound, method, system, or device that assists in the identification or characterization of a health or disease state. The diagnostic can be used in standard assays as is known in the art.

[052] The term "mammal" is used in its usual biological sense. Thus, it specifically includes humans, cattle, horses, monkeys, dogs, cats, mice, rats, cows, sheep, pigs, goats, and non- human primates, but also includes many other species. [053] The term "pharmaceutically acceptable carrier", "pharmaceutically acceptable diluent" or "pharmaceutically acceptable excipient" includes any and all solvents, co-solvents, complexing agents, dispersion media, coatings, isotonic and absorption delaying agents and the like which are not biologically or otherwise undesirable. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. In addition, various adjuvants such as are commonly used in the art may be included. These and other such compounds are described in the literature, e.g., in the Merck Index, Merck & Company, Rahway, NJ. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g. , in Brunton et al. (Eds.) (2017); Goodman and Gilman's: The Pharmacological Basis of Therapeutics. 13th Ed.. The McGraw-Hill Companies.

[054] The term "pharmaceutically acceptable salt" refers to salts that retain the biological effectiveness and properties of the compounds provided herein and, which are not biologically or otherwise undesirable. In many cases, the compounds provided herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. Many such salts are known in the art, for example, as described in WO 87/05297. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, gly colic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethane sulfonic acid, p- toluene sulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like; particularly preferred are the ammonium, potassium, sodium, calcium, and magnesium salts. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.

[055] "Patient" as used herein, means a human or a non-human mammal, e.g., a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate, or a bird, e.g., a chicken, as well as any other vertebrate or invertebrate. In some embodiments, the patient is a human. [056] A "therapeutically effective amount" of a compound as provided herein is one which is sufficient to achieve the desired physiological effect and may vary according to the nature and severity of the disease condition, and the potency of the compound. "Therapeutically effective amount" is also intended to include one or more of the compounds of Formula I in combination with one or more other agents that are effective to treat the diseases and/or conditions described herein. The combination of compounds can be a synergistic combination. Synergy, as described, for example, by Chou and Talalay, Advances in Enzyme Regulation (1984), 22, 27-55, occurs when the effect of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at sub-optimal concentrations of the compounds. It will be appreciated that different concentrations may be employed for prophylaxis than for treatment of an active disease. This amount can further depend upon the patient's height, weight, sex, age and medical history.

[057] A therapeutic effect relieves, to some extent, one or more of the symptoms of the disease.

[058] "Treat," "treatment," or "treating," as used herein refers to administering a compound or pharmaceutical composition as provided herein for therapeutic purposes. The term "therapeutic treatment" refers to administering treatment to a patient already suffering from a disease thus causing a therapeutically beneficial effect, such as ameliorating existing symptoms, ameliorating the underlying metabolic causes of symptoms, postponing or preventing the further development of a disorder, and/or reducing the severity of symptoms that will or are expected to develop.

[059] "Drug-eluting" and/or controlled release as used herein refers to any and all mechanisms, e.g., diffusion, migration, permeation, and/or desorption by which the drug(s) incorporated in the drug-eluting material pass therefrom over time into the surrounding body tissue.

[060] "Drug-eluting material" and/or controlled release material as used herein refers to any natural, synthetic or semi -synthetic material capable of acquiring and retaining a desired shape or configuration and into which one or more drugs can be incorporated and from which incorporated drug(s) are capable of eluting over time.

[061] "Elutable drug" as used herein refers to any drug or combination of drugs having the ability to pass over time from the drug-eluting material in which it is incorporated into the surrounding areas of the body. Compounds

[062] The compounds and compositions described herein can be used as antiproliferative agents, e.g., anti-cancer and anti-angiogenesis agents, and/or as inhibitors of the Wnt signaling pathway, e.g., for treating diseases or disorders associated with aberrant Wnt signaling. In addition, the compounds can be used as inhibitors of one or more kinases, kinase receptors, or kinase complexes. Such compounds and compositions are also useful for controlling cellular proliferation, differentiation, and/or apoptosis.

[063] The compounds and compositions described herein can be used to inhibit DYRKIA for treating a disorder or disease in which DYRKIA overexpression is implicated, such as Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Down Syndrome, Frontotemporal Dementia with Parkinsonism- 17 (FTDP-17), Lewy body dementia, Parkinson's Disease, Pick's Disease, and additional diseases with pronounced neurodegeneration such as Autism, Dementia, Epilepsy, Huntington's Disease, Multiple Sclerosis; diseases and disorders associated with acquired brain injury such as Chronic Traumatic Encephalopathy, Traumatic Brain Injury, Tumor, and Stroke.

[064] Some embodiments of the present disclosure include compounds of Formula

I:

I

or salts, pharmaceutically acceptable salts, or prodrugs thereof.

[065] R 1 , R 2 , R 4 , and R 5 are independently selected from the group consisting of H, halide (e.g., F, CI, Br, I), unsubstituted -(C1-3 haloalkyl) (e.g., C2-3, 1-2, lhaloalkyl), and unsubstituted -(Ci-3 alkyl) (e.g., C2-3, 1-2, 1 alkyl).

[066] In some embodiments, R 1 , R 2 , R 4 , and R 5 are independently selected from the group consisting of H and halide (e.g., F, CI, Br, I);

[067] In some embodiments, R 1 , R 2 , R 4 , and R 5 are independently selected from the group consisting of H and F.

[068] In some embodiments, R 1 , R 2 , R 4 , and R 5 are all H.

[069] In some embodiments, R 1 is F, and R 2 , R 4 , and R 5 are all H.

[070] In some embodiments, R 2 is F, and R 1 , R 4 , and R 5 are all H. [071] In some embodiments, R 4 is F, and R 1 , R 2 , and R 5 are all H.

[072] In some embodiments, R 5 is F, and R 1 , R 2 , and R 4 are all H.

[073] In some embodiments, R 3 is a aryl ring or a 6-10-membered heteroaryl ring, each optionally substituted as defined anywhere herein.

[074] In some embodiments, R 3 is selected from the group consisting of -aryl optionally substituted with 1-5 (e.g., 1-4, 1-3, 1-2, 1) R 7 and a 6-10-membered heteroaryl optionally substituted with 1-4 (e.g., 1-3, 1-2, 1) R 8 .

[075] In some embodiments, R 3 is selected from the group consisting of -phenyl optionally substituted with 1-2 (e.g., 1) R 7 and a 6-membered heteroaryl optionally substituted with 1-2 (e.g., 1) R 8 .

[076] In some embodiments, R 3 is selected from the group consisting of -phenyl optionally substituted with 1-2 (e.g., 1) R 7 , -pyridinyl optionally substituted with 1 R 8 , - pyrimidinyl optionally substituted with 1 R 8 , -pyridazinyl optionally substituted with 1 R 8 , and - pyrazinyl optionally substituted with 1 R 8 .

[077] In some embodiments, R 3 is -aryl optionally substituted with 1-2 (e.g., 1) R 7 .

[078] In some embodiments, R 3 is -phenyl optionally substituted with 1-2 (e.g., 1)

R 7 .

[079] In some embodiments, R 3 is a 6-10-membered heteroaryl optionally substituted with 1-2 (e.g., 1) R 8 .

[080] In some embodiments, R 3 1 is a 6-membered heteroaryl optionally substituted with 1-2 (e.g., 1) R 8 .

[081] In some embodiments, R 3 is pyridinyl optionally substituted with 1 R 8 .

[082] In some embodiments, R 3 is pyridin-3-yl optionally substituted with 1 R 8 .

[083] In some embodiments, R 3 is pyridin-4-yl optionally substituted with 1 R 8 .

[084] In some embodiments, R 3 is pyrazinyl optionally substituted with 1 R 8 .

[085] In some embodiments, R 3 is pyrazin-2-yl optionally substituted with 1 R 8 .

[086] In some embodiments, R 3 is pyrimidinyl optionally substituted with 1 R 8 .

[087] In some embodiments, R 3 is pyrimidin-5-yl optionally substituted with 1 R 8 .

[088] In some embodiments, R 3 is pyridazinyl optionally substituted with 1 R 8 .

[089] In some embodiments, R 3 is pyridazin-4-yl optionally substituted with 1 R 8 .

[090] In some embodiments, R 3 is pyridazin-3-yl optionally substituted with 1 R 8 .

[091] In some embodiments, R 3 1 is a 9-membered heteroaryl optionally substituted with 1-2 (e.g., 1) R 8 . [092] In some embodiments, R 3 is a 10-membered heteroaryl optionally substituted with 1-2 (e.g., 1) R 8 .

[093] In some embodiment, -(C1-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein.

[094] In some embodiment, -(C1-4 alkylene) is, optionally substituted with one or more halides (e.g., F, CI, Br, I) or one or more unsubstituted -(C1-3 alkyl) (e.g., C2-3, 1-2, 1 alkyl).

[095] In some embodiment, -(C1-4 alkylene) is, optionally substituted with one or more halides (e.g., F, CI, Br, I) and one or more unsubstituted -(C1-3 alkyl) (e.g., C2-3, 1-2, 1 alkyl).

[096] In some embodiment, -(C1-4 alkylene) is, optionally substituted with one or more F or one or more Me.

[097] In some embodiment, -(C1-4 alkylene) is, optionally substituted with one or more F and one or more Me.

[098] In some embodiment, -(C1-4 alkylene) is -CH2-.

[099] In some embodiment, -(C1-4 alkylene) is -CH2CH2-.

[0100] In some embodiment, -(C1-4 alkylene) is -CH2CH2CH2-.

[0101] In some embodiment, R 6 is selected from the group consisting of -(C1-4 alkylene)pheteroaryl optionally substituted with 1-6 (e.g., 1-5, 1-4, 1-3, 1-2, 1) R 9 , -(C1-4 alkylene) p aryl optionally substituted with 1-6 (e.g., 1-5, 1-4, 1-3, 1-2, 1) R 25 , -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 10 , -(Ci-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1- 7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 11 , -(C1-4 alkylene)N(R 12 )(R 13 ), -N(R 14 )(R 15 ), -CF(G -9 alkyl) 2 , -(C1-4 alkylene) p O(Ci-9 alkyl), and -(C2-9 alkynyl) optionally substituted with one or more halides; wherein each alkyl of -CF(Ci-9 alkyl)2 is, independently, optionally substituted with one or more halides; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0102] In some embodiment, R 6 is selected from the group consisting of -heteroaryl optionally substituted with 1-2 R 9 , -(Ctyheteroaryl optionally substituted with 1-2 R 9 , -phenyl optionally substituted with 1-2 R 25 , -heterocyclyl optionally substituted with 1-2 R 10 , - (Ctyheterocyclyl optionally substituted with 1-2 R 10 , and -carbocyclyl optionally substituted with 1-2 R 11 . [0103] In some embodiments, R 6 is selected from the group consisting of:

[0104] In some embodiments, R 6 is selected from the group consisting of:

[0105] In some embodiments, R 6 is -N(R 0 )(R 31 ).

[0106] In some embodiments, R 6 is -NH(R 31 ). V

6 is selected from the group consisting of -NH

[0108] In some embodiments, R 6 is -NMe(R 31 ). 6 is selected from the group consisting of

; wherein n is 1-3 (e.g., 1-2,

1), m is 0-2 (e.g., 2, 1, 0) and each Z is independently selected from the group consisting of CR 34 and N.

[0111] In some embodiments, R 6 is selected from the group consisting of

[0112] In some embodiments, R 6 is selected from the group consisting

[0113] In some embodiments, R 6 is selected from the group consisting

In some embodiments, R 6 is selected from the group consisting

[0115] In some embodiments, R 6 is selected from the group consisting of

6 is selected from the group consisting

and ^ 6 is selected from the group consisting

[0118] In some embodiments, R 6 is selected from the group consisting of -aryl substituted with 1-5 R 25 and a 6-membered heteroaryl optionally substituted with 1-6 R 9 .

[0119] In some embodiments, R 6 is selected from the group consisting of -phenyl substituted with 1-5 R 25 , and -pyridinyl optionally substituted with 1-4 R 9 .

[0120] In some embodiments, R 6 is a -phenyl substituted with one R 25 .

[0121] In some embodiments, R 6 is a -pyridinyl substituted with one R 9 .

[0122] In some embodiments, R 6 is a -pyridin-2-yl substituted with one R 9 .

[0123] In some embodiments, R 6 is a -pyridin-3-yl substituted with one R 9 .

[0124] In some embodiments, R 6 is a -pyridin-4-yl substituted with one R 9 .

[0125] In some embodiments, R 7 is selected from the group consisting of halide (e.g., F, CI, Br, I) and -N(R 16 ) 2 .

[0126] In some embodiments, R 7 is selected from the group consisting of F, -NH2, - NH(Ci-4 alkyl), -NH(Ci- 3 alkyl), -NH(Ci- 2 alkyl), and -NHMe.

[0127] In some embodiments, R 8 is selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-9 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-9 alkenyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-9 alkynyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-9 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, i haloalkyl), -CN, -N(R 17 )(R 18 ), -(Ci-4 alkylene^R 19 , - C(=O)N(R 20 ) 2 , and -(Ci- 4 alkylene) p heterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1- 7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 21 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0128] In some embodiments, R 8 is selected from the group consisting of F, Me, - CH 2 F, -CHF 2 , -CF 3 , -CN, -OMe, -CH 2 OH, -NH 2 , -NH(Ci -3 alkyl), -NMe 2 , -NHheterocyclyl, - -heterocyclyl optionally substituted with 1-2 R 21 , and -(Ctyheterocyclyl optionally substituted with 1-2 R 21 .

[0129] In some embodiments, each R 9 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-9 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1- 6, 1-5, 1-4, 1-3, 1-2, l alkyl), unsubstituted -(C2-9 alkenyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-9 alkynyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-9 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 haloalkyl), -X 2 R 22 , -C(=0)N(R 2 ) 2 , -(Ci- 4 alkylene) p N(R 24 )2, -(Ci-4 alkylene)pheterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1- 7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 25 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 26 ; wherein each -(Ci- 4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0130] In some embodiments, R 9 is selected from the group consisting of F, methyl,

odiments, R 9 is selected from the group consisting of

[0133] In some embodiments, each R 1 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(Ci- 9 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1- 6, 1-5, 1-4, 1-3, 1-2, l alkyl), unsubstituted -(C2-9 alkenyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-9 alkynyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-9 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene) p OR 27 , - N(R 20 ) 2 , -(Ci- 4 alkylene) p heterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 28 , -(Ci-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 29 , -C(=0)(R 30 ), -(C1-4 alkylene) p C(=0)OR 3 °, -(C1-4 alkylene)aryl optionally substituted with one or more halides, -(C1-4 alkylene) p heteroaryl optionally substituted with one or more halides, and -S02(R 31 ); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0134] In some embodiments, if two R 10 are attached to the same carbon atom can together represent =0 to form a carbonyl group.

[0135] In some embodiments, each R 10 is F.

[0136] In some embodiments, R 10 is -N(R 43 )(R 44 ).

[0137] In some embodiments, one R 10 is F and one R 10 is -N(R 4 )(R 44 ).

[0138] In some embodiments, R 10 is -NMe2; in some embodiments, R 10 is -NH(Me); in some embodiments, R 10 is -NH(Et); in some embodiments, R 10 is -NH(nPr); in some embodiments, R 10 is -NH(iPr); in some embodiments, R 10 is -NH(nBu); in some embodiments, R 10 is -NH(iBu); in some embodiments, R 10 is -NH(tBu).

[0139] In some embodiments, one R 10 is F and one R 10 is -NMe2; in some embodiments, one R 10 is F and one R 10 is -NH(Me); in some embodiments, one R 10 is F and one R 10 is -NH(Et); in some embodiments, one R 10 is F and one R 10 is -NH(nPr); in some embodiments, one R 10 is F and one R 10 is -NH(iPr); in some embodiments, one R 10 is F and one R 10 is -NH(nBu); in some embodiments, one R 10 is F and one R 10 is -NH(iBu); in some embodiments, one R 10 is F and one R 10 is -NH(tBu). [0140] In some embodiments, R 10 is -NH(Ci-5 haloalkyl); in some embodiments, R 10 is -NH(Ci-4 haloalkyl); in some embodiments, R 10 is -NH(Ci-3 haloalkyl); in some embodiments, R 10 is -NH(Ci-2 haloalkyl); in some embodiments, R 10 is -NH(CH2CH2F); in some embodiments, R 10 is -NH(CH2CHF2); in some embodiments, R 10 is -NH(CH2CF3); in some embodiments, R 10 is -NHiCFbCFbCFbF); in some embodiments, R 10 is -NHiCFbCFbCFff^); in some embodiments, R 10 is -NH(CH 2 CH 2 CF 3 ); in some embodiments, R 10 is -NH(CH(CH 2 F) 2 ); in some embodiments, R 10 is -NH(CH(CHF 2 ) 2 ); in some embodiments, R 10 is -NH(CH(CF 3 ) 2 ); in some embodiments, R 10 is -N(Ci-5 haloalkyl)2; in some embodiments, R 10 is -N(Ci-4 haloalkyl)2; in some embodiments, R 10 is -N(Ci-3 haloalkyl)2; in some embodiments, R 10 is -N(Ci-2 haloalkyl)2; in some embodiments, R 10 is -N(CH2CH2F)2; in some embodiments, R 10 is -N(CH2CHF2)2; in some embodiments, R 10 is - N(CH2CF3)2; in some embodiments, R 10 is -N(CH2CH2CH2F)2; in some embodiments, R 10 is - N(CH2CH2CHF2)2; in some embodiments, R 10 is -N(CH2CH2CF3)2; in some embodiments, R 10 is -N(CH(CH2F)2)2; in some embodiments, R 10 is -N(CH(CHF2)2)2; in some embodiments, R 10 is - N(CH(CF3)2)2; in some embodiments, R 10 is -NMe(Ci-5 haloalkyl); in some embodiments, R 10 is - NMe(C 1-4 haloalkyl); in some embodiments, R 10 is -NMe(Ci-3 haloalkyl); in some embodiments, R 10 is -NMe(Ci -2 haloalkyl); in some embodiments, R 10 is -NMe (CH 2 CH 2 F) ; in some embodiments, R 10 is -NMe(CH2CHF2); in some embodiments, R 10 is -NMe(CH2CF3); in some embodiments, R 10 is -NMe(CH 2 CH 2 CH 2 F); in some embodiments, R 10 is -NMe(CH 2 CH 2 CHF 2 ); in some embodiments, R 10 is -NMe(CH2CH2CF3); in some embodiments, R 10 is - NMe(CH(CH 2 F) 2 ); in some embodiments, R 10 is -NMe(CH(CHF 2 ) 2 ); in some embodiments, R 10 is -NMe(CH(CF 3 ) 2 ).

[0141] In some embodiments, R 10 is -NH(CF 3 ).

[0142] In some embodiments, one R 10 is F and one R 10 is -NH(Ci-5 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NH(Ci-4 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NH(Ci-3 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NH(Ci-2 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NHiCfbCfbF); in some embodiments, one R 10 is F and one R 10 is -NH(CH2CHF2); in some embodiments, one R 10 is F and one R 10 is -NH(CH 2 CF 3 ); in some embodiments, one R 10 is F and one R 10 is -NH(CH 2 CH 2 CH 2 F); in some embodiments, one R 10 is F and one R 10 is -NH(CH2CH2CHF2); in some embodiments, one R 10 is F and one R 10 is -NHiCfbCfbCFs); in some embodiments, one R 10 is F and one R 10 is - NH(CH(CH 2 F) 2 ); in some embodiments, one R 10 is F and one R 10 is -NH(CH(CHF 2 ) 2 ); in some embodiments, one R 10 is F and one R 10 is -NH(CH(CF3)2); in some embodiments, one R 10 is F and one R 10 is -N(Ci-5 haloalkyl)2; in some embodiments, one R 10 is F and one R 10 is -N(Ci-4 haloalkyl^; in some embodiments, one R 10 is F and one R 10 is -N(Ci-3 haloalkyl)2; in some embodiments, one R 10 is F and one R 10 is -N(Ci-2 haloalkyl)2; in some embodiments, one R 10 is F and one R 10 is - N(CH2CH2F)2; in some embodiments, one R 10 is F and one R 10 is -N(CH2CHF2)2; in some embodiments, one R 10 is F and one R 10 is -N(CH2CF3)2; in some embodiments, one R 10 is F and one R 10 is -NiCfbCfbCfbF^; in some embodiments, one R 10 is F and one R 10 is - N(CH2CH2CHF2)2; in some embodiments, one R 10 is F and one R 10 is -N(CH2CH2CF3)2; in some embodiments, one R 10 is F and one R 10 is -N(CH(CH2F)2)2; in some embodiments, one R 10 is F and one R 10 is -N(CH(CHF2)2)2; in some embodiments, one R 10 is F and one R 10 is -N(CH(CF3)2)2; in some embodiments, one R 10 is F and one R 10 is -NMe(Ci-5 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NMe(Ci-4 haloalkyl); in some embodiments, one R 10 is F and one R 10 is - NMe(Ci-3 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NMe(Ci-2 haloalkyl); in some embodiments, one R 10 is F and one R 10 is -NMe(CH2CH2F); in some embodiments, one R 10 is F and one R 10 is -NMe(CH2CHF2); in some embodiments, one R 10 is F and one R 10 is - NMe(CH2CF3); in some embodiments, one R 10 is F and one R 10 is -NMeiCFbCFbCFbF); in some embodiments, one R 10 is F and one R 10 is -NMe(CH2CH2CHF2); in some embodiments, one R 10 is F and one R 10 is -NMe(CH2CH2CF3); in some embodiments, one R 10 is F and one R 10 is - NMe(CH(CH 2 F) 2 ); in some embodiments, one R 10 is F and one R 10 is -NMe(CH(CHF 2 ) 2 ); in some embodiments, one R 10 is F and one R 10 is -NMe(CH(CF 3 ) 2 ).

[0143] In some embodiments, one R 10 is F and one R 10 is -NH(CFs).

[0144] In some embodiments, each R 11 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(Ci- 9 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1- 6, 1-5, 1-4, 1-3, 1-2, l alkyl), unsubstituted -(C2-9 alkenyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-9 alkynyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-9 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene) p OR 27 , - N(R 20 ) 2 , -C(=0)(R 30 ), -C(=0)OR 30 , -(Ci- 4 alkylene)pheterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 28 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 29 ; wherein each - (Ci-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0145] In some embodiments, each R 11 is independently selected from the group consisting of -NH 2 , -NH(Me), -NMe 2 , -CH 2 OH, -CH 2 OMe, -C(=0)(R 30 ), -heterocyclyl optionally substituted with 1-5 (e.g., 1-4, 1-3, 1-2, 1) R 28 , and -CFbheterocyclyl optionally substituted with 1-5 (e.g., 1-4, 1-3, 1-2, 1) R 28 . 11 is independently selected from the group

[0147] In some embodiments, each R 11 is -N(R 33 )(R 34 ).

[0148] In some embodiments, R 11 is -NH2.

[0149] In some embodiments, R 11 is -NMe2.

[0150] In some embodiments, R 11 is -NH(Me).

[0151] In some embodiments, R 11 is -NH(Ci-5 haloalkyl); in some embodiments, R 11 is -NH(unsubstituted (C1-4 haloalkyl)); in some embodiments, R 11 is -NH(C 1-3 haloalkyl); in some embodiments, R 11 is -NH(C 1-2 haloalkyl); in some embodiments, R 11 is -NH(CH2CH2F); in some embodiments, R 11 is -NH(CH2CHF2); in some embodiments, R 11 is -NH(CH2CF3); in some embodiments, R 11 is -NH(CH 2 CH 2 CH 2 F); in some embodiments, R 11 is -NH(CH 2 CH 2 CHF 2 ); in some embodiments, R 11 is -NHiCFfcCfbCFs); in some embodiments, R 11 is -NH(CH(CH2F)2); in some embodiments, R 11 is -NH(CH(CHF2)2); in some embodiments, R 11 is -NH(CH(CF3)2); in some embodiments, R 11 is -N(Ci-5 haloalkyl)2; in some embodiments, R 11 is -N(Ci-4 haloalkyl)2; in some embodiments, R 11 is -N(Ci-3 haloalkyl)2; in some embodiments, R 11 is -N(C 1-2 haloalkyl^; in some embodiments, R 11 is -NiCfbCfbF^; in some embodiments, R 11 is -N(CH2CHF2)2; in some embodiments, R 11 is -N(CH2CF3)2; in some embodiments, R 11 is -N(CH2CH2CH2F)2; in some embodiments, R 11 is -N(CH2CH2CHF2)2; in some embodiments, R 11 is in some embodiments, R 11 is -N(CH(CH2F)2)2; in some embodiments, R 11 is -N(CH(CHF2)2)2; in some embodiments, R 11 is -N(CH(CF3)2)2; in some embodiments, R 11 is -NMe(C 1-5 haloalkyl); in some embodiments, R 11 is -NMe(C 1-4 haloalkyl); in some embodiments, R 11 is -NMe(Ci-3 haloalkyl); in some embodiments, R 11 is -NMe(Ci-2 haloalkyl); in some embodiments, R 11 is -NMeiCfbCfbF); in some embodiments, R 11 is -NMe(CH2CHF2); in some embodiments, R 11 is -NMe(CH2CF3); in some embodiments, R 11 is -NMe(CH2CH2CH2F); in some embodiments, R 11 is - NMe(CH2CH2CHF2); in some embodiments, R 11 is -NMeiCFbCFbCFs); in some embodiments, R 11 is -NMe(CH(CH 2 F) 2 ); in some embodiments, R 11 is -NMe(CH(CHF 2 ) 2 ); in some embodiments, R 11 is -NMe(CH(CF 3 ) 2 ).

[0152] In some embodiments, R 11 is -NH(CF 3 ).

[0153] In some embodiments, R 12 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(Ci-9 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-9 alkenyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2- 9 alkynyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-9 haloalkyl) (e.g., C2-5, 3- 5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene) p heterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 32 , and -carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 33 ; wherein -(C1-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein.

[0154] In some embodiments, R 13 is attached to the nitrogen and is selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-9 alkyl) (e.g.,€2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1- 8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-9 alkenyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-9 alkynyl) (e.g., C3-5, 4-5, 2-8, 2-7, 2-6, 2-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-9 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 32 , and -carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 33 ; wherein -(C1-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein.

[0155] In some embodiments, R 14 is attached to the nitrogen and selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g.,

C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), and unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl).

[0156] In some embodiments, R 15 is attached to the nitrogen and is selected from the group consisting of -(Ci-4 alkylene)pheterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1- 7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 32 , and— (C1-4 alkylene ) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 33 ; wherein each -(Ci- 4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0157] In some embodiments, R 16 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene) p aryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), and -(C1-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0158] In some embodiments, , two adjacent R 16 are taken together to form a - heterocyclyl ring optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 32 .

[0159] In some embodiments, R 17 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), and unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl).

[0160] In some embodiments, R 18 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -(C=0)R 34 , and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl); wherein -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0161] In some embodiments, each R 19 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -(C1-4 alkylene) p heterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(Ci-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl); wherein -(C1-4 alkylene) is optionally substituted with one or more substituents as defined anywhere herein.

[0162] In some embodiments, each R 20 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), and unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl).

[0163] In some embodiments, each R 21 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, l alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -N(R 20 ) 2 , -CN, and -OH.

[0164] In some embodiments, each R 22 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene)N(R 4 )2, -(Ci-4 alkylene ) p aryl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1- 5, 1-4, 1-3, 1-2, 1) R 35 , -(Ci-4 alkylene)pheterocyclyl optionally substituted with 1-12 (e.g., 1-11, 1- 10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 36 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 29 ; wherein each - (Ci-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0165] In some embodiments, each R 23 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), and unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl).

[0166] In some embodiments, each R 24 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -(C1-4 alkylene)N(R 4 )2; wherein -(C1-4 alkylene) is optionally substituted with one or more substituents as defined anywhere herein.

[0167] In some embodiments, each R 25 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -CN, -OH, -C(=0)R 37 , -N(R 8 ) 2 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 26 ; wherein each - (Ci-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0168] In some embodiments, each R 25 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -CN, -C(=0)R 37 , -N(R 38 ) 2 , -(C1-4 alkylene) p OR 27 , -(C1-4 alkylene)pheterocyclyl optionally substituted with 1-10 (e.g., 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 32 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1- 4, 1-3, 1-2, 1) R 26 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein;

[0169] In some embodiments, R 25 is selected from the group consisting of F,

ethyl, n-propyl, isopropyl, -0(Ci-3 alkyl), -0(Ci-3 haloalkyl),

[0170] In some embodiments, each R 26 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -CN.

[0171] In some embodiments, each R 27 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1- 4, 1-3, 1-2, 1) R 29 , and -(Ci-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl); wherein each -(Ci-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0172] In some embodiments, each R 28 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -CN, and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-1 1, 1 - 10, 1-9, 1-8, 1 -7, 1 -6, 1 -5, 1-4, 1-3, 1 -2, 1) R 29 ; wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0173] In some embodiments, each R 29 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -CN.

[0174] In some embodiments, R 30 is selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene ) p aryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), -(C1-4 alkylene)pheteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), and -(C1-4 alkylene)pheterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl); wherein each -(C1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0175] In some embodiments, R 31 is selected from the group consisting of unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -(C1-4 alkylene ) p aryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3- 4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), -(C1-4 alkylene ) p heteroaryl optionally substituted with one or more halides or unsubstituted -(C1-5 alkyl), and -(C1-4 alkylene ) p heterocyclyl optionally substituted with one or more halides or one or more unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl); wherein -(Ci-4 alkylene) is, optionally substituted with one or more substituents as defined anywhere herein.

[0176] In some embodiments, each R 32 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1- 4, 1-3, 1-2, 1) R 29 ; wherein each -(C 1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0177] In some embodiments, each R 33 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1- 4, 1-3, 1-2, 1) R 29 ; wherein each -(C 1-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein.

[0178] In some embodiments, R 34 is attached to the nitrogen and is selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), and unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl).

[0179] In some embodiments, each R 35 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e .g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -CN.

[0180] In some embodiments, each R 36 is independently selected from the group consisting of halide (e.g., F, CI, Br, I), unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1- 2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), -CN, -OH, -C(=0)R 37 , -N(R 8 ) 2 , and -(C1-4 alkylene) p carbocyclyl optionally substituted with 1-12 (e.g., 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1) R 29 ; wherein each - (Ci-4 alkylene) is, independently, optionally substituted with one or more substituents as defined anywhere herein. [0181] In some embodiments, each R 37 is a heteroaryl optionally substituted with 1-6

R 39 .

[0182] In some embodiments, each R 38 is independently selected from the group consisting of H, unsubstituted -(C1-5 alkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 alkyl), unsubstituted -(C2-5 alkenyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkenyl), unsubstituted -(C2-5 alkynyl) (e.g., C3-5, 4-5, 2-4, 3-4, 2-3, 2 alkynyl), unsubstituted -(C1-5 haloalkyl) (e.g., C2-5, 3-5, 4-5, 2-4, 3-4, 2-3, 1-4, 1-3, 1-2, 1 haloalkyl), and -(C1-4 alkylene)N(R 4 )2; wherein -(C1-4 alkylene) is optionally substituted with one or more substituents as defined anywhere herein.

[0183] In some embodiments, each R 39 is a -heterocyclyl optionally substituted with 1-10 R 29 .

[0184] In some embodiments, each X 1 is selected from the group consisting of O and

S.

[0185] In some embodiments, each X 2 is selected from the group consisting of O, S, and NR 34 .

[0186] In some embodiments, each p is independently 0 or 1.

[0187] In some embodiments, there is the proviso that Formula I is not a structure consisting of:

[0188] Illustrative compounds of Formula (I) are shown in Table 1

Table 1.

Administration and Pharmaceutical Compositions

[0189] Some embodiments include pharmaceutical compositions comprising: (a) a therapeutically effective amount of a compound provided herein, or its corresponding enantiomer, diastereoisomer or tautomer, or pharmaceutically acceptable salt; and (b) a pharmaceutically acceptable carrier.

[0190] The compounds provided herein may also be useful in combination (administered together or sequentially) with other known agents.

[0191] Non-limiting examples of diseases which can be treated with a combination of a compound of Formula (I) and other another active agent are colorectal cancer, ovarian cancer, chronic inflammation, diabetic retinopathy, pulmonary fibrosis, and osteoarthritis. For example, a compound of Formula (I) can be combined with one or more chemotherapeutic compounds.

[0192] In some embodiments, colorectal cancer can be treated with a combination of a compound of Formula (I) and one or more of the following drugs: 5-Fluorouracil (5-FU), which can be administered with the vitamin-like drug leucovorin (also called folinic acid); capecitabine (XELODA ® ), irinotecan (CAMPOSTAR ® ), oxaliplatin (ELOXATIN ® ). Examples of combinations of these drugs which could be further combined with a compound of Formula (I) are FOLFOX (5- FU, leucovorin, and oxaliplatin), FOLFIRI (5-FU, leucovorin, and irinotecan), FOLFOXIRI (leucovorin, 5-FU, oxaliplatin, and irinotecan) and CapeOx (Capecitabine and oxaliplatin). For rectal cancer, chemo with 5-FU or capecitabine combined with radiation may be given before surgery (neoadjuvant treatment).

[0193] In some embodiments, ovarian cancer can be treated with a combination of a compound of Formula (I) and one or more of the following drugs: Topotecan, Liposomal doxorubicin (DOXIL ® ), Gemcitabine (GEMZAR ® ), Cyclophosphamide (CYTOXAN ® ), Vinorelbine (NAVELBINE ® ), Ifosfamide (IFEX ® ), Etoposide (VP-16), Altretamine (HEXALEN ® ), Capecitabine (XELODA ® ), Irinotecan (CPT-11, CAMPTOSAR ® ), Melphalan, Pemetrexed (ALIMTA ® ) and Albumin bound paclitaxel (nab-paclitaxel, ABRAXANE ® ). Examples of combinations of these drugs which could be further combined with a compound of Formula (I) are TIP (paclitaxel [Taxol], ifosfamide, and cisplatin), VelP (vinblastine, ifosfamide, and cisplatin) and VIP (etoposide [VP- 16], ifosfamide, and cisplatin).

[0194] In some embodiments, a compound of Formula (I) can be used to treat cancer in combination with any of the following methods: (a) Hormone therapy such as aromatase inhibitors, LHRH [luteinizing hormone-releasing hormone] analogs and inhibitors, and others; (b) Ablation or embolization procedures such as radiofrequency ablation (RFA), ethanol (alcohol) ablation, microwave thermotherapy and cryosurgery (cryotherapy); (c) Chemotherapy using alkylating agents such as cisplatin and carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil and ifosfamide; (d) Chemotherapy using anti-metabolites such as azathioprine and mercaptopurine; (e) Chemotherapy using plant alkaloids and terpenoids such as vinca alkaloids (i.e. Vincristine, Vinblastine, Vinorelbine and Vindesine) and taxanes; (f) Chemotherapy using podophyllotoxin, etoposide, teniposide and docetaxel; (g) Chemotherapy using topoisomerase inhibitors such as irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, and teniposide; (h) Chemotherapy using cytotoxic antibiotics such as actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin; (i) Chemotherapy using tyrosine-kinase inhibitors such as Imatinib mesylate (GLEEVEC ® , also known as STI-571), Gefitinib (Iressa, also known as ZD 1839), Erlotinib (marketed as TARCEVA ® ), Bortezomib (VELCADE ® ) , tamoxifen , tofacitinib, crizotinib, Bcl-2 inhibitors (e.g. obatoclax in clinical trials, ABT-263, and Gossypol), PARP inhibitors (e.g. Iniparib, Olaparib in clinical trials), PI3K inhibitors (e.g. perifosine in a phase III trial), VEGF Receptor 2 inhibitors (e.g. Apatinib), AN-152, (AEZS-108), Braf inhibitors (e.g. vemurafenib, dabrafenib and LGX818), MEK inhibitors (e.g. trametinib and MEK162), CDK inhibitors, (e.g. PD-0332991), salinomycin and Sorafenib; (j) Chemotherapy using monoclonal antibodies such as Rituximab (marketed as MABTHERA ® or RITUXAN ® ), Trastuzumab (Herceptin also known as ErbB2), Cetuximab (marketed as ERBITUX ® ), and Bevacizumab (marketed as AVASTIN ® ); and (k) radiation therapy.

[0195] In some embodiments, diabetic retinopathy can be treated with a combination of a compound of Formula (I) and one or more of the following natural supplements: Bilberry, Butcher's broom, Ginkgo, Grape seed extract, and Pycnogenol (Pine bark).

[0196] In some embodiments, idiopathic pulmonary fibrosis/pulmonary fibrosis can be treated with a combination of a compound of Formula (I) and one or more of the following drugs: pirfenidone (pirfenidone was approved for use in 201 1 in Europe under the brand name Esbriet ® ), prednisone, azathioprine, N-acetylcysteine, interferon-γ lb, bosentan (bosentan is currently being studied in patients with IPF, [The American Journal of Respiratory and Critical Care Medicine (2011), 184(1), 92-9]), Nintedanib (BIBF 1 120 and Vargatef), QAX576 [British Journal of Pharmacology (2011), 163(1), 141-172], and anti-inflammatory agents such as corticosteroids.

[0197] In some embodiments, a compound of Formula (I) can be used to treat idiopathic pulmonary fibrosis/pulmonary fibrosis in combination with any of the following methods: oxygen therapy, pulmonary rehabilitation and surgery.

[0198] In some embodiments, a compound of Formula (I) can be used to treat osteoarthritis in combination with any of the following methods: (a) Nonsteroidal antiinflammatory drugs (NSAIDs) such as ibuprofen, naproxen, aspirin and acetaminophen; (b) physical therapy; (c) injections of corticosteroid medications; (d) injections of hyaluronic acid derivatives (e.g. Hyalgan, Synvisc); (e) narcotics, like codeine; (f) in combination with braces and/or shoe inserts or any device that can immobilize or support your joint to help you keep pressure off it (e.g., splints, braces, shoe inserts or other medical devices); (g) realigning bones (osteotomy); (h) joint replacement (arthroplasty); and (i) in combination with a chronic pain class.

[0199] In some embodiments, macular degeneration can be treated with a combination of a compound of Formula (I) and one or more of the following drugs: Bevacizumab (Avastin ® ), Ranibizumab (Lucentis ® ), Pegaptanib (Macugen), Aflibercept (Eylea ® ), verteporfin (Visudyne ® ) in combination with photodynamic therapy (PDT) or with any of the following methods: (a) in combination with laser to destroy abnormal blood vessels (photocoagulation); and (b) in combination with increased vitamin intake of antioxidant vitamins and zinc.

[0200] In some embodiments, retinitis pigmentosa can be treated with a combination of a compound of Formula (I) and one or more of the following drugs: UF-021 (Ocuseva™), vitamin A palmitate and pikachurin or with any of the following methods: (a) with the Argus ® II retinal implant; and (b) with stem cell and/or gene therapy.

[0201] Administration of the compounds disclosed herein or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration, including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, ontologically, neuro- otologically, intraocularly, subconjuctivally, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, via wound irrigation, intrabuccally, intra-abdominally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intrameningeally, via inhalation, via endotracheal or endobronchial instillation, via direct instillation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, epidurally, intratympanically, intracisternally, intravascularly, intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with a prosthetic devices. In some embodiments, the administration method includes oral or parenteral administration.

[0202] Compounds provided herein intended for pharmaceutical use may be administered as crystalline or amorphous products. Pharmaceutically acceptable compositions may include solid, semi-solid, liquid, solutions, colloidal, liposomes, emulsions, suspensions, complexes, coacervates and aerosols. Dosage forms, such as, e.g., tablets, capsules, powders, liquids, suspensions, suppositories, aerosols, implants, controlled release or the like. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, milling, grinding, supercritical fluid processing, coacervation, complex coacervation, encapsulation, emulsification, complexation, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose. The compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills (tablets and or capsules), transdermal (including electrotransport) patches, implants and the like, for prolonged and/or timed, pulsed administration at a predetermined rate.

[0203] The compounds can be administered either alone or in combination with a conventional pharmaceutical carrier, excipient or the like. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self- emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose -based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene- poly oxypropylene -block polymers, and wool fat. Cyclodextrins such as α-, β, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl- -cyclodextrins, or other solubilized derivatives can also be used to enhance delivery of compounds described herein. Dosage forms or compositions containing a compound as described herein in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. The contemplated compositions may contain 0.001%-100% of a compound provided herein, in one embodiment 0.1 -95%, in another embodiment 75-85%, in a further embodiment 20-80%. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 22 nd Edition (Pharmaceutical Press, London, UK. 2012).

[0204] In one embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with a compound provided herein, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g. , in propylene carbonate, vegetable oils, PEG's, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which one or more compounds provided herein or additional active agents are physically separated are also contemplated; e.g. , capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two -compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.

[0205] Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. a compound provided herein and optional pharmaceutical adjuvants in a carrier (e.g. , water, saline, aqueous dextrose, glycerol, glycols, ethanol or the like) to form a solution, colloid, liposome, emulsion, complexes, coacervate or suspension. If desired, the pharmaceutical composition can also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, co-solvents, solubilizing agents, pH buffering agents and the like (e.g., sodium acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, and the like).

[0206] In some embodiments, the unit dosage of compounds of Formula (I) is about 0.25 mg/Kg to about 50 mg/Kg in humans.

[0207] In some embodiments, the unit dosage of compounds of Formula (I) is about 0.25 mg/Kg to about 20 mg/Kg in humans.

[0208] In some embodiments, the unit dosage of compounds of Formula (I) is about 0.50 mg/Kg to about 19 mg/Kg in humans.

[0209] In some embodiments, the unit dosage of compounds of Formula (I) is about 0.75 mg/Kg to about 18 mg/Kg in humans. [0210] In some embodiments, the unit dosage of compounds of Formula (I) is about 1.0 mg/Kg to about 17 mg/Kg in humans.

[0211] In some embodiments, the unit dosage of compounds of Formula (I) is about 1.25 mg/Kg to about 16 mg/Kg in humans.

[0212] In some embodiments, the unit dosage of compounds of Formula (I) is about 1.50 mg/Kg to about 15 mg/Kg in humans.

[0213] In some embodiments, the unit dosage of compounds of Formula (I) is about 1.75 mg/Kg to about 14 mg/Kg in humans.

[0214] In some embodiments, the unit dosage of compounds of Formula (I) is about 2.0 mg/Kg to about 13 mg/Kg in humans.

[0215] In some embodiments, the unit dosage of compounds of Formula (I) is about 3.0 mg/Kg to about 12 mg/Kg in humans.

[0216] In some embodiments, the unit dosage of compounds of Formula (I) is about 4.0 mg/Kg to about 1 1 mg/Kg in humans.

[0217] In some embodiments, the unit dosage of compounds of Formula (I) is about 5.0 mg/Kg to about 10 mg/Kg in humans.

[0218] In some embodiments, the compositions are provided in unit dosage forms suitable for single administration.

[0219] In some embodiments, the compositions are provided in unit dosage forms suitable for twice a day administration.

[0220] In some embodiments, the compositions are provided in unit dosage forms suitable for three times a day administration.

[0221] Injectables can be prepared in conventional forms, either as liquid solutions, colloid, liposomes, complexes, coacervate or suspensions, as emulsions, or in solid forms suitable for reconstitution in liquid prior to injection. The percentage of a compound provided herein contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the patient. However, percentages of active ingredient of 0.01% to 10% in solution are employable, and could be higher if the composition is a solid or suspension, which could be subsequently diluted to the above percentages.

[0222] In some embodiments, the composition will comprise about 0.1-10% of the active agent in solution.

[0223] In some embodiments, the composition will comprise about 0.1 -5% of the active agent in solution. [0224] In some embodiments, the composition will comprise about 0.1-4% of the active agent in solution.

[0225] In some embodiments, the composition will comprise about 0.15-3% of the active agent in solution.

[0226] In some embodiments, the composition will comprise about 0.2-2% of the active agent in solution.

[0227] In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of about 1-96 hours.

[0228] In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of about 1-72 hours.

[0229] In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of about 1-48 hours.

[0230] In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of about 1-24 hours.

[0231] In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of about 1-12 hours.

[0232] In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of about 1-6 hours.

[0233] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 5 mg/m 2 to about 300 mg/m 2 .

[0234] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 5 mg/m 2 to about 200 mg/m 2 .

[0235] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 5 mg/m 2 to about 100 mg/m 2 .

[0236] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 10 mg/m 2 to about 50 mg/m 2 .

[0237] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 50 mg/m 2 to about 200 mg/m 2 .

[0238] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 75 mg/m 2 to about 175 mg/m 2 .

[0239] In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of about 100 mg/m 2 to about 150 mg/m 2 .

[0240] It is to be noted that concentrations and dosage values may also vary depending on the specific compound and the severity of the condition to be alleviated. It is to be further understood that for any particular patient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.

[0241] In one embodiment, the compositions can be administered to the respiratory tract (including nasal and pulmonary) e.g., through a nebulizer, metered-dose inhalers, atomizer, mister, aerosol, dry powder inhaler, insufflator, liquid instillation or other suitable device or technique.

[0242] In some embodiments, aerosols intended for delivery to the nasal mucosa are provided for inhalation through the nose. For optimal delivery to the nasal cavities, inhaled particle sizes of about 5 to about 100 microns are useful, with particle sizes of about 10 to about 60 microns being preferred. For nasal delivery, a larger inhaled particle size may be desired to maximize impaction on the nasal mucosa and to minimize or prevent pulmonary deposition of the administered formulation. In some embodiments, aerosols intended for delivery to the lung are provided for inhalation through the nose or the mouth. For delivery to the lung, inhaled aerodynamic particle sizes of about less than 10 μπι are useful (e.g., about 1 to about 10 microns). Inhaled particles may be defined as liquid droplets containing dissolved drug, liquid droplets containing suspended drug particles (in cases where the drug is insoluble in the suspending medium), dry particles of pure drug substance, drug substance incorporated with excipients, liposomes, emulsions, colloidal systems, coacervates, aggregates of drug nanoparticles, or dry particles of a diluent which contain embedded drug nanoparticles.

[0243] In some embodiments, compounds of Formula (I) disclosed herein intended for respiratory delivery (either systemic or local) can be administered as aqueous formulations, as non-aqueous solutions or suspensions, as suspensions or solutions in halogenated hydrocarbon propellants with or without alcohol, as a colloidal system, as emulsions, coacervates, or as dry powders. Aqueous formulations may be aerosolized by liquid nebulizers employing either hydraulic or ultrasonic atomization or by modified micropump systems (like the soft mist inhalers, the Aerodose ® or the AERx ® systems). Propellant-based systems may use suitable pressurized metered-dose inhalers (pMDIs). Dry powders may use dry powder inhaler devices (DPIs), which are capable of dispersing the drug substance effectively. A desired particle size and distribution may be obtained by choosing an appropriate device. [0244] In some embodiments, the compositions of Formula (I) disclosed herein can be administered to the ear by various methods. For example, a round window catheter (e.g., U.S. Pat. Nos. 6,440,102 and 6,648,873) can be used.

[0245] Alternatively, formulations can be incorporated into a wick for use between the outer and middle ear (e.g., U.S. Pat. No. 6, 120,484) or absorbed to collagen sponge or other solid support (e.g., U.S. Pat. No. 4, 164,559).

[0246] If desired, formulations of the disclosure can be incorporated into a gel formulation (e.g., U.S. Pat. Nos. 4,474,752 and 6,911,211).

[0247] In some embodiments, compounds of Formula (I) disclosed herein intended for delivery to the ear can be administered via an implanted pump and delivery system through a needle directly into the middle or inner ear (cochlea) or through a cochlear implant stylet electrode channel or alternative prepared drug delivery channel such as but not limited to a needle through temporal bone into the cochlea.

[0248] Other options include delivery via a pump through a thin film coated onto a multichannel electrode or electrode with a specially imbedded drug delivery channel (pathways) carved into the thin film for this purpose. In other embodiments the acidic or basic solid compound of Formula (I) can be delivered from the reservoir of an external or internal implanted pumping system.

[0249] Formulations of the disclosure also can be administered to the ear by intratympanic injection into the middle ear, inner ear, or cochlea (e.g., U.S. Pat. No. 6,377,849 and Ser. No. 11/337,815).

[0250] Intratympanic injection of therapeutic agents is the technique of injecting a therapeutic agent behind the tympanic membrane into the middle and/or inner ear. In one embodiment, the formulations described herein are administered directly onto the round window membrane via transtympanic injection. In another embodiment, the ion channel modulating agent auris-acceptable formulations described herein are administered onto the round window membrane via a non-transtympanic approach to the inner ear. In additional embodiments, the formulation described herein is administered onto the round window membrane via a surgical approach to the round window membrane comprising modification of the crista fenestrae cochleae.

[0251] In some embodiments, the compounds of Formula (I) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), and the like. [0252] Suppositories for rectal administration of the drug (either as a solution, colloid, suspension or a complex) can be prepared by mixing a compound provided herein with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt or erode/dissolve in the rectum and release the compound. Such materials include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol. In suppository forms of the compositions, a low -melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter, is first melted.

[0253] Solid compositions can be provided in various different types of dosage forms, depending on the physicochemical properties of the compound provided herein, the desired dissolution rate, cost considerations, and other criteria. In one of the embodiments, the solid composition is a single unit. This implies that one unit dose of the compound is comprised in a single, physically shaped solid form or article. In other words, the solid composition is coherent, which is in contrast to a multiple unit dosage form, in which the units are incoherent.

[0254] Examples of single units which may be used as dosage forms for the solid composition include tablets, such as compressed tablets, film-like units, foil-like units, wafers, lyophilized matrix units, and the like. In one embodiment, the solid composition is a highly porous lyophilized form. Such lyophilizates, sometimes also called wafers or lyophilized tablets, are particularly useful for their rapid disintegration, which also enables the rapid dissolution of the compound.

[0255] On the other hand, for some applications the solid composition may also be formed as a multiple unit dosage form as defined above. Examples of multiple units are powders, granules, microparticles, pellets, mini-tablets, beads, lyophilized powders, and the like. In one embodiment, the solid composition is a lyophilized powder. Such a dispersed lyophilized system comprises a multitude of powder particles, and due to the lyophilization process used in the formation of the powder, each particle has an irregular, porous microstructure through which the powder is capable of absorbing water very rapidly, resulting in quick dissolution. Effervescent compositions are also contemplated to aid the quick dispersion and absorption of the compound.

[0256] Another type of multiparticulate system which is also capable of achieving rapid drug dissolution is that of powders, granules, or pellets from water-soluble excipients which are coated with a compound provided herein so that the compound is located at the outer surface of the individual particles. In this type of system, the water-soluble low molecular weight excipient may be useful for preparing the cores of such coated particles, which can be subsequently coated with a coating composition comprising the compound and, for example, one or more additional excipients, such as a binder, a pore former, a saccharide, a sugar alcohol, a film-forming polymer, a plasticizer, or other excipients used in pharmaceutical coating compositions.

[0257] Also provided herein are kits. Typically, a kit includes one or more compounds or compositions as described herein. In certain embodiments, a kit can include one or more delivery systems, e.g., for delivering or administering a compound as provided herein, and directions for use of the kit (e.g., instructions for treating a patient). In another embodiment, the kit can include a compound or composition as described herein and a label that indicates that the contents are to be administered to a patient with cancer. In another embodiment, the kit can include a compound or composition as described herein and a label that indicates that the contents are to be administered to a patient with one or more of hepatocellular carcinoma, colon cancer, leukemia, lymphoma, sarcoma, ovarian cancer, diabetic retinopathy, pulmonary fibrosis, rheumatoid arthritis, sepsis, ankylosing spondylitis, psoriasis, scleroderma, mycotic and viral infections, bone and cartilage diseases, Alzheimer's disease, lung disease, bone/osteoporotic (wrist, spine, shoulder and hip) fractures, articular cartilage (chondral) defects, degenerative disc disease (or intervertebral disc degeneration), polyposis coli, bone density and vascular defects in the eye (Osteoporosis - pseudoglioma Syndrome, OPPG), familial exudative vitreoretinopathy, retinal angiogenesis, early coronary disease, tetra-amelia, Mullerian-duct regression and virilization, SERKAL syndrome, type II diabetes, Fuhrmann syndrome, Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndrome, odonto-onycho-dermal dysplasia, obesity, split-hand/foot malformation, caudal duplication, tooth agenesis, Wilms tumor, skeletal dysplasia, focal dermal hypoplasia, autosomal recessive anonychia, neural tube defects, alpha-thalassemia (ATRX) syndrome, fragile X syndrome, ICF syndrome, Angelman syndrome, Prader-Willi syndrome, Beckwith-Wiedemann Syndrome, Norrie disease, and Rett syndrome.

Methods of Treatment

[0258] The compounds and compositions provided herein can be used as inhibitors and/or modulators of one or more components of the Wnt pathway, which may include one or more Wnt proteins, and thus can be used to treat a variety of disorders and diseases in which aberrant Wnt signaling is implicated, such as cancer and other diseases associated with abnormal angiogenesis, cellular proliferation, and cell cycling. Accordingly, the compounds and compositions provided herein can be used to treat cancer, to reduce or inhibit angiogenesis, to reduce or inhibit cellular proliferation, to correct a genetic disorder, and/or to treat a neurological condition/disorder/disease due to mutations or dysregulation of the Wnt pathway and/or of one or more of Wnt signaling components. Non-limiting examples of diseases which can be treated with the compounds and compositions provided herein include a variety of cancers, diabetic retinopathy, pulmonary fibrosis, rheumatoid arthritis, scleroderma, mycotic and viral infections, bone and cartilage diseases, neurological conditions/diseases such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS), motor neuron disease, multiple sclerosis or autism, lung disease, bone/osteoporotic (wrist, spine, shoulder and hip) fractures, polyposis coli, bone density and vascular defects in the eye (Osteoporosis-pseudoglioma Syndrome, OPPG), familial exudative vitreoretinopathy, retinal angiogenesis, early coronary disease, tetra-amelia, Mullerian-duct regression and virilization, SERKAL syndrome, type II diabetes, Fuhrmann syndrome, Al- Awadi/Raas-Rothschild/Schinzel phocomelia syndrome, odonto-onycho-dermal dysplasia, obesity, split-hand/foot malformation, caudal duplication, tooth agenesis, Wilms tumor, skeletal dysplasia, focal dermal hypoplasia, autosomal recessive anonychia, neural tube defects, alpha- thalassemia (ATRX) syndrome, fragile X syndrome, ICF syndrome, Angelman syndrome, Prader- Willi syndrome, Beckwith-Wiedemann Syndrome, Norrie disease and Rett syndrome.

[0259] With respect to cancer, the Wnt pathway is known to be constitutively activated in a variety of cancers including, for example, colon cancer, hepatocellular carcinoma, lung cancer, ovarian cancer, prostate cancer , pancreatic cancer and leukemias such as CML, CLL and T-ALL. Accordingly, the compounds and compositions described herein may be used to treat these cancers in which the Wnt pathway is constitutively activated. In certain embodiments, the cancer is chosen from hepatocellular carcinoma, colon cancer, leukemia, lymphoma, sarcoma and ovarian cancer.

[0260] Other cancers can also be treated with the compounds and compositions described herein.

[0261] More particularly, cancers that may be treated by the compounds, compositions and methods described herein include, but are not limited to, the following:

[0262] 1) Breast cancers, including, for example ER + breast cancer, ER " breast cancer, her2 " breast cancer, her2 + breast cancer, stromal tumors such as fibroadenomas, phyllodes tumors, and sarcomas, and epithelial tumors such as large duct papillomas; carcinomas of the breast including in situ (noninvasive) carcinoma that includes ductal carcinoma in situ (including Paget's disease) and lobular carcinoma in situ, and invasive (infiltrating) carcinoma including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma, medullary carcinoma, colloid (mucinous) carcinoma, tubular carcinoma, and invasive papillary carcinoma; and miscellaneous malignant neoplasms. Further examples of breast cancers can include luminal A, luminal B, basal A, basal B, and triple negative breast cancer, which is estrogen receptor negative (ER ), progesterone receptor negative, and her2 negative (her2 ~ ). In some embodiments, the breast cancer may have a high risk Oncotype score.

[0263] 2) Cardiac cancers, including, for example sarcoma, e.g., angiosarcoma, fibrosarcoma, rhabdomyosarcoma, and liposarcoma; myxoma; rhabdomyoma; fibroma; lipoma and teratoma.

[0264] 3) Lung cancers, including, for example, bronchogenic carcinoma, e.g., squamous cell, undifferentiated small cell, undifferentiated large cell, and adenocarcinoma; alveolar and bronchiolar carcinoma; bronchial adenoma; sarcoma; lymphoma; chondromatous hamartoma; and mesothelioma.

[0265] 4) Gastrointestinal cancer, including, for example, cancers of the esophagus, e.g., squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, and lymphoma; cancers of the stomach, e.g., carcinoma, lymphoma, and leiomyosarcoma; cancers of the pancreas, e.g., ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, and vipoma; cancers of the small bowel, e.g., adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, and fibroma; cancers of the large bowel, e.g., adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, and leiomyoma.

[0266] 5) Genitourinary tract cancers, including, for example, cancers of the kidney, e.g., adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, and leukemia; cancers of the bladder and urethra, e.g., squamous cell carcinoma, transitional cell carcinoma, and adenocarcinoma; cancers of the prostate, e.g., adenocarcinoma, and sarcoma; cancer of the testis, e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, and lipoma.

[0267] 6) Liver cancers, including, for example, hepatoma, e.g., hepatocellular carcinoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hepatocellular adenoma; and hemangioma.

[0268] 7) Bone cancers, including, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochrondroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors.

[0269] 8) Nervous system cancers, including, for example, cancers of the skull, e.g., osteoma, hemangioma, granuloma, xanthoma, and osteitis deformans; cancers of the meninges, e.g., meningioma, meningiosarcoma, and gliomatosis; cancers of the brain, e.g., astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, oligodendrocytoma, schwannoma, retinoblastoma, and congenital tumors; and cancers of the spinal cord, e.g., neurofibroma, meningioma, glioma, and sarcoma.

[0270] 9) Gynecological cancers, including, for example, cancers of the uterus, e.g., endometrial carcinoma; cancers of the cervix, e.g., cervical carcinoma, and pre tumor cervical dysplasia; cancers of the ovaries, e.g., ovarian carcinoma, including serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma, granulosa theca cell tumors, Sertoli Leydig cell tumors, dysgerminoma, and malignant teratoma; cancers of the vulva, e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, and melanoma; cancers of the vagina, e.g., clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma, and embryonal rhabdomyosarcoma; and cancers of the fallopian tubes, e.g., carcinoma.

[0271] 10) Hematologic cancers, including, for example, cancers of the blood, e.g., acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, and myelodysplastic syndrome, Hodgkin's lymphoma, non-Hodgkin's lymphoma (malignant lymphoma) and Waldenstrom's macroglobulinemia.

[0272] 1 1) Skin cancers and skin disorders, including, for example, malignant melanoma and metastatic melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, and scleroderma.

[0273] 12) Adrenal gland cancers, including, for example, neuroblastoma.

[0274] More particularly, tumors of the central nervous system that may be treated by the compounds, compositions and methods described herein include:

[0275] 1) Astrocytic tumors, e.g., diffuse astrocytoma (fibrillary, protoplasmic, gemistocytic, mixed), anaplastic (malignant) astrocytoma, glioblastoma multiforme (giant cell glioblastoma and gliosarcoma), pilocytic astrocytoma (pilomyxoid astrocytoma), pleomorphic xanthoastrocytoma, subependymal giant cell astrocytoma, and gliomatosis cerebri.

[0276] 2) Oligodendroglial tumors, e.g., oligodendroglioma and anaplastic oligodendroglioma.

[0277] 3) Oligoastrocytic tumors, e.g., oligoastrocytoma and anaplastic oligoastrocytoma.

[0278] 4) Ependymal tumors, e.g., subependymoma, myxopapillary ependymoma, ependymoma, (cellular, papillary, clear cell, tanycytic), and anaplastic (malignant) ependymoma.

[0279] 5) Choroid plexus tumors, e.g., choroid plexus papilloma, atypical choroid plexus papilloma, and choroid plexus carcinoma. [0280] 6) Neuronal and mixed neuronal -glial tumors, e.g., gangliocytoma, ganglioglioma, dysembryoplastic neuroepithelial tumor (DNET), dysplastic gangliocytoma of the cerebellum (Lhermitte-Duclos), desmoplastic infantile astrocytoma/ganglioglioma, central neurocytoma, anaplastic ganglioglioma, extraventricular neurocytoma, cerebellar liponeurocytoma, Papillary glioneuronal tumor, Rosette-forming glioneuronal tumor of the fourth ventricle, and paraganglioma of the filum terminale.

[0281] 7) Pineal tumors, e.g., pineocytoma, pineoblastoma, papillary tumors of the pineal region, and pineal parenchymal tumor of intermediate differentiation.

[0282] 8) Embryonal tumors, e.g., medulloblastoma (medulloblastoma with extensive nodularity, anaplastic medulloblastoma, desmoplastic, large cell, melanotic, medullomyoblastoma), medulloepithelioma, supratentorial primitive neuroectodermal tumors, and primitive neuroectodermal tumors (PNETs) such as neuroblastoma, ganglioneuroblastoma, ependymoblastoma, and atypical teratoid/rhabdoid tumor.

[0283] 9) Neuroblastic tumors, e.g., olfactory (esthesioneuroblastoma), olfactory neuroepithelioma, and neuroblastomas of the adrenal gland and sympathetic nervous system.

[0284] 10) Glial tumors, e.g., astroblastoma, chordoid glioma of the third ventricle, and angiocentric glioma.

[0285] 1 1) Tumors of cranial and paraspinal nerves, e.g., schwannoma, neurofibroma Perineurioma, and malignant peripheral nerve sheath tumor.

[0286] 12) Tumors of the meninges such as tumors of meningothelial cells, e.g., meningioma (atypical meningioma and anaplastic meningioma); mesenchymal tumors, e.g., lipoma, angiolipoma, hibernoma, liposarcoma, solitary fibrous tumor, fibrosarcoma, malignant fibrous histiocytoma, leiomyoma, leiomyosarcoma, rhabdomyoma, rhabdomyosarcoma, chondroma, chondrosarcoma, osteoma, osteosarcoma, osteochondroma, haemangioma, epithelioid hemangioendothelioma, haemangiopericytoma, anaplastic haemangiopericytoma, angiosarcoma, Kaposi Sarcoma, and Ewing Sarcoma; primary melanocytic lesions, e.g., diffuse melanocytosis, melanocytoma, malignant melanoma, meningeal melanomatosis; and hemangioblastomas.

[0287] 13) Tumors of the hematopoietic system, e.g., malignant Lymphomas, plasmocytoma, and granulocytic sarcoma.

[0288] 14) Germ cell tumors, e.g., germinoma, embryonal carcinoma, yolk sac tumor, choriocarcinoma, teratoma, and mixed germ cell tumors.

[0289] 15) Tumors of the sellar region, e.g., craniopharyngioma, granular cell tumor, pituicytoma, and spindle cell oncocytoma of the adenohypophysis. [0290] Cancers may be solid tumors that may or may not be metastatic. Cancers may also occur, as in leukemia, as a diffuse tissue. Thus, the term "tumor cell," as provided herein, includes a cell afflicted by any one of the above identified disorders.

[0291] A method of treating cancer using a compound or composition as described herein may be combined with existing methods of treating cancers, for example by chemotherapy, irradiation, or surgery (e.g., oophorectomy). In some embodiments, a compound or composition can be administered before, during, or after another anticancer agent or treatment.

[0292] The compounds and compositions described herein can be used as anti- angiogenesis agents and as agents for modulating and/or inhibiting the activity of protein kinases, thus providing treatments for cancer and other diseases associated with cellular proliferation mediated by protein kinases. For example, the compounds described herein can inhibit the activity of one or more kinases. Accordingly, provided herein is a method of treating cancer or preventing or reducing angiogenesis through kinase inhibition.

[0293] In addition, and including treatment of cancer, the compounds and compositions described herein can function as cell-cycle control agents for treating proliferative disorders in a patient. Disorders associated with excessive proliferation include, for example, cancers, scleroderma, immunological disorders involving undesired proliferation of leukocytes, and restenosis and other smooth muscle disorders. Furthermore, such compounds may be used to prevent de -differentiation of post-mitotic tissue and/or cells.

[0294] Diseases or disorders associated with uncontrolled or abnormal cellular proliferation include, but are not limited to, the following:

• a variety of cancers, including, but not limited to, carcinoma, hematopoietic tumors of lymphoid lineage, hematopoietic tumors of myeloid lineage, tumors of mesenchymal origin, tumors of the central and peripheral nervous system and other tumors including melanoma, seminoma and Kaposi's sarcoma.

• a disease process which features abnormal cellular proliferation, e.g., benign prostatic hyperplasia, familial adenomatosis polyposis, neurofibromatosis, atherosclerosis, arthritis, glomerulonephritis, restenosis following angioplasty or vascular surgery, inflammatory bowel disease, transplantation rejection, endotoxic shock, and fungal infections. Fibrotic disorders such as skin fibrosis; scleroderma; progressive systemic fibrosis; lung fibrosis; muscle fibrosis; kidney fibrosis; glomerulosclerosis; glomerulonephritis; hypertrophic scar formation; uterine fibrosis; renal fibrosis; cirrhosis of the liver, liver fibrosis; fatty liver disease (FLD); adhesions, such as those occurring in the abdomen, pelvis, spine or tendons; chronic obstructive pulmonary disease; fibrosis following myocardial infarction; pulmonary fibrosis; fibrosis and scarring associated with diffuse/interstitial lung disease; central nervous system fibrosis, such as fibrosis following stroke; fibrosis associated with neuro -degenerative disorders such as Alzheimer's Disease or multiple sclerosis; fibrosis associated with proliferative vitreoretinopathy (PVR); restenosis; endometriosis; ischemic disease and radiation fibrosis.

• defective apoptosis-associated conditions, such as cancers (including but not limited to those types mentioned herein), viral infections (including but not limited to herpesvirus, poxvirus, Epstein-Barr virus, Sindbis virus and adenovirus), prevention of AIDS development in HIV-infected individuals, autoimmune diseases (including but not limited to systemic lupus erythematosus, rheumatoid arthritis, sepsis, ankylosing spondylitis, psoriasis, scleroderma, autoimmune mediated glomerulonephritis, inflammatory bowel disease and autoimmune diabetes mellitus), neuro -degenerative disorders (including but not limited to Alzheimer's disease, lung disease, amyotrophic lateral sclerosis, retinitis pigmentosa, Parkinson's disease, AIDS-related dementia, spinal muscular atrophy and cerebellar degeneration), myelodysplastic syndromes, aplastic anemia, ischemic injury associated with myocardial infarctions, stroke and reperfusion injury, arrhythmia, atherosclerosis, toxin-induced or alcohol related liver diseases, hematological diseases (including but not limited to chronic anemia and aplastic anemia), degenerative diseases of the musculoskeletal system (including but not limited to osteoporosis and arthritis), tendinopathies such as tendinitis and tendinosis, aspirin- sensitive rhinosinusitis, cystic fibrosis, multiple sclerosis, kidney diseases and cancer pain.

• genetic diseases due to mutations in Wnt signaling components, such as polyposis coli, bone density and vascular defects in the eye (Osteoporosis-pseudoglioma Syndrome, OPPG), familial exudative vitreoretinopathy, retinal angiogenesis, early coronary disease, tetra-amelia, Mullerian-duct regression and virilization, SERKAL syndrome, type II diabetes, Fuhrmann syndrome, Al-Awadi/Raas- Rothschild/Schinzel phocomelia syndrome, odonto-onycho-dermal dysplasia, obesity, split-hand/foot malformation, caudal duplication, tooth agenesis, Wilms tumor, skeletal dysplasia, focal dermal hypoplasia, autosomal recessive anonychia, neural tube defects, alpha-thalassemia (ATRX) syndrome, fragile X syndrome, ICF syndrome, Angelman syndrome, Prader-Willi syndrome, Beckwith-Wiedemann Syndrome, Norrie disease and Rett syndrome.

[0295] The compounds and compositions provided herein have been found to possess immunomodulatory activities and are expected to control the innate and adaptive immune system (e.g. macrophages, microglia, dendritic cells, B and T cells) and suppress pro-inflammatory cytokine release (e.g. TNF, IL-6, IL-1, IFNI ) which is well known to be involved in chronic inflammation in a wide variety of disease areas. Therefore compounds and compositions provided herein can used to treat chronic inflammation associated with disorders and diseases including but not limited to eye disorders, joint pain, arthritis (rheumatoid, osteo, psoriatic gout), cancers (colon, breast, lung, pancreas, and others), gastrointestinal disorders (ulcerative colitis and inflammatory bowel diseases), pulmonary disorders (chronic obstructive pulmonary disorder and asthma), allergies, skin disorders (atopic dermatitis and psoriasis), diabetes, pancreatitis, tendonitis, hepatitis, heart disease, myocarditis, stroke, lupus, and neurological disorders such as multiple sclerosis, Parkinson's and dementia including Alzheimer's disease.

[0296] The compounds and compositions provided herein can be used as inhibitors and/or modulators of the enzyme DYRK1 A, and thus can be used to treat a variety of disorders and diseases associated with tau protein, amyloid, alpha-synuclein, TDP-43 or FUS pathology including, but not limited to, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), down syndrome, frontotemporal dementia (FTD) including FTD with Parkinsonism-17 (FTDP-17), behavioural variant frontotemporal dementia (bvFTD), FTD in patients with motor neuron disease (MND) (typically amyotrophic lateral sclerosis, also called FTD-ALS), corticobasal degeneration (CBD) (also called corticobasal ganglionic degeneration), progressive supranuclear palsy, primary progressive aphasia (PPA), globular glial tauopathy (GGT), myotonic dystrophy type 1 (DM1) (also called Steinert disease), myotonic dystrophy type 2 (DM2) (also called proximal myotonic myopathy), Guam complex, argyrophilic grain disease, dementia pugilistica, post-encephalitic parkinsonism, Lewy body dementia, Parkinson's disease, Pick's disease, and additional diseases with pronounced neurodegeneration such as autism, dementia, epilepsy, Huntington's disease, multiple sclerosis; diseases and disorders associated with acquired brain injury such as chronic traumatic encephalopathy, traumatic brain injury, tumor, and stroke.

[0297] Non-limiting examples of neurological disorders (e.g., neurological conditions and neurological diseases) which can be treated with the compounds and compositions provided herein include Alzheimer's disease, aphasia, apraxia, arachnoiditis, ataxia telangiectasia, attention deficit hyperactivity disorder, auditory processing disorder, autism, alcoholism, Bell's palsy, bipolar disorder, brachial plexus injury, Canavan disease, carpal tunnel syndrome, causalgia, central pain syndrome, central pontine myelinolysis, centronuclear myopathy, cephalic disorder, cerebral aneurysm, cerebral arteriosclerosis, cerebral atrophy, cerebral gigantism, cerebral palsy, cerebral vasculitis, cervical spinal stenosis, Charcot-Marie-Tooth disease, Chiari malformation, chronic fatigue syndrome, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic pain, Coffin-Lowry syndrome, complex regional pain syndrome, compression neuropathy, congenital facial diplegia, corticobasal degeneration, cranial arteritis, craniosynostosis, Creutzfeldt-Jakob disease, cumulative trauma disorder, Cushing's syndrome, cytomegalic inclusion body disease (CIBD), Dandy-Walker syndrome, Dawson disease, De Morsier's syndrome, Dejerine-Klumpke palsy, Dejerine-Sottas disease, delayed sleep phase syndrome, dementia, dermatomyositis, developmental dyspraxia, diabetic neuropathy, diffuse sclerosis, Dravet syndrome, dysautonomia, dyscalculia, dysgraphia, dyslexia, dystonia, empty sella syndrome, encephalitis, encephalocele, encephalotrigeminal angiomatosis, encopresis, epilepsy, Erb's palsy, erythromelalgia, essential tremor, Fabry's disease, Fahr's syndrome, familial spastic paralysis, febrile seizure, Fisher syndrome, Friedreich's ataxia, fibromyalgia, Foville's syndrome, Gaucher's disease, Gerstmann's syndrome, giant cell arteritis, giant cell inclusion disease, globoid cell leukodystrophy, gray matter heterotopia, Guillain-Barre syndrome, HTLV-1 associated myelopathy, Hallervorden-Spatz disease, hemifacial spasm, hereditary spastic paraplegia, heredopathia atactica polyneuritiformis, herpes zoster oticus, herpes zoster, Hirayama syndrome, holoprosencephaly, Huntington's disease, hydranencephaly, hydrocephalus, hypercortisolism, hypoxia, immune-mediated encephalomyelitis, inclusion body myositis, incontinentia pigmenti, infantile phytanic acid storage disease, infantile Refsum disease, infantile spasms, inflammatory myopathy, intracranial cyst, intracranial hypertension, Joubert syndrome, Karak syndrome, Kearns- Sayre syndrome, Kennedy disease, Kinsbourne syndrome, Klippel Feil syndrome, Krabbe disease, Kugelberg-Welander disease, kuru, Lafora disease, Lambert-Eaton myasthenic syndrome, Landau- Kleffner syndrome, lateral medullary (Wallenberg) syndrome, Leigh's disease, Lennox -Gastaut syndrome, Lesch-Nyhan syndrome, leukodystrophy, Lewy body dementia, lissencephaly, locked- in syndrome, Lou Gehrig's disease, lumbar disc disease, lumbar spinal stenosis, Lyme disease, Machado-Joseph disease (Spinocerebellar ataxia type 3), macrencephaly, macropsia, megalencephaly, Melkersson-Rosenthal syndrome, Meniere's disease, meningitis, Menkes disease, metachromatic leukodystrophy, microcephaly, micropsia, Miller Fisher syndrome, misophonia, mitochondrial myopathy, Mobius syndrome, monomelic amyotrophy, motor neuron disease, motor skills disorder, Moyamoya disease, mucopolysaccharidoses, multi-infarct dementia, multifocal motor neuropathy, multiple sclerosis, multiple system atrophy, muscular dystrophy, myalgic encephalomyelitis, myasthenia gravis, myelinoclastic diffuse sclerosis, myoclonic Encephalopathy of infants, myoclonus, myopathy, myotubular myopathy, myotonia congenital, narcolepsy, neurofibromatosis, neuroleptic malignant syndrome, lupus erythematosus, neuromyotonia, neuronal ceroid lipofuscinosis, Niemann-Pick disease, O'Sullivan-McLeod syndrome, occipital Neuralgia, occult Spinal Dysraphism Sequence, Ohtahara syndrome, olivopontocerebellar atrophy, opsoclonus myoclonus syndrome, optic neuritis, orthostatic hypotension, palinopsia, paresthesia, Parkinson's disease, paramyotonia Congenita, paraneoplastic diseases, paroxysmal attacks, Parry- Romberg syndrome, Pelizaeus-Merzbacher disease, periodic paralyses, peripheral neuropathy, photic sneeze reflex, phytanic acid storage disease, Pick's disease, polymicrogyria (PMG), polymyositis, porencephaly, post-polio syndrome, postherpetic neuralgia (PHN), postural hypotension, Prader-Willi syndrome, primary lateral sclerosis, prion diseases, progressive hemifacial atrophy, progressive multifocal leukoencephalopathy, progressive supranuclear palsy, pseudotumor cerebri, Ramsay Hunt syndrome type I, Ramsay Hunt syndrome type II, Ramsay Hunt syndrome type III, Rasmussen's encephalitis, reflex neurovascular dystrophy, Refsum disease, restless legs syndrome, retrovirus-associated myelopathy, Rett syndrome, Reye's syndrome, rhythmic movement disorder, Romberg syndrome, Saint Vitus dance, Sandhoff disease, schizophrenia, Schilder's disease, schizencephaly, sensory integration dysfunction, septo -optic dysplasia, Shy-Drager syndrome, Sjogren's syndrome, snatiation, Sotos syndrome, spasticity, spina bifida, spinal cord tumors, spinal muscular atrophy, spinocerebellar ataxia, Steele-Richardson- Olszewski syndrome, Stiff-person syndrome, stroke, Sturge-Weber syndrome, subacute sclerosing panencephalitis, subcortical arteriosclerotic encephalopathy, superficial siderosis, Sydenham's chorea, syncope, synesthesia, syringomyelia, tarsal tunnel syndrome, tardive dyskinesia, tardive dysphrenia, Tarlov cyst, Tay-Sachs disease, temporal arteritis, tetanus, tethered spinal cord syndrome, Thomsen disease, thoracic outlet syndrome, tic douloureux, Todd's paralysis, Tourette syndrome, toxic encephalopathy, transient ischemic attack, transmissible spongiform encephalopathies, transverse myelitis, tremor, trigeminal neuralgia, tropical spastic paraparesis, trypanosomiasis, tuberous sclerosis, ubisiosis, Von Hippel-Lindau disease (VHL), Viliuisk Encephalomyelitis (VE), Wallenberg's syndrome, Werdnig, Hoffman disease, west syndrome, Williams syndrome, Wilson's disease, and Zellweger syndrome.

[0298] The compounds and compositions may also be useful in the inhibition of the development of invasive cancer, tumor angiogenesis and metastasis.

[0299] In some embodiments, the disclosure provides a method for treating a disease or disorder associated with aberrant cellular proliferation by administering to a patient in need of such treatment an effective amount of one or more of the compounds of Formula (I), in combination (simultaneously or sequentially) with at least one other agent. [0300] In some embodiments, the disclosure provides a method of treating or ameliorating in a patient a disorder or disease selected from the group consisting of: cancer, pulmonary fibrosis, idiopathic pulmonary fibrosis (IPF), degenerative disc disease, bone/osteoporotic fractures, bone or cartilage disease, and osteoarthritis, the method comprising administering to the patient a therapeutically effective amount of a compound according to Formula (I), or a pharmaceutically acceptable salt thereof.

[0301] In some embodiments, the disclosure provides a method of treating or ameliorating in a patient a disorder or disease selected from the group consisting of: chronic inflammation, systemic inflammation, diabetes, cancer, pulmonary fibrosis, idiopathic pulmonary fibrosis (IPF), degenerative disc disease, bone/osteoporotic fractures, a bone or cartilage disease, a neurological condition/disorder/disease, osteoarthritis, lung disease, a fibrotic disorder.

[0302] In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

[0303] In some embodiments, the method of treats a disorder or disease in which aberrant Wnt signaling is implicated in a patient, the method comprises administering to the patient a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

[0304] In some embodiments, the disorder or disease is the pain and inflammation associated with cancer.

[0305] In some embodiments, the disorder or disease is the pain and inflammation associated with a joint.

[0306] In some embodiments, the disorder or disease is the pain and inflammation associated with the knee.

[0307] In some embodiments, the disorder or disease is the pain and inflammation associated with the hip.

[0308] In some embodiments, the disorder or disease is the pain and inflammation associated with the shoulder.

[0309] In some embodiments, the disorder or disease is the pain and inflammation associated with arthritis.

[0310] In some embodiments, the disorder or disease is the pain and inflammation associated with gastrointestinal disorders.

[0311] In some embodiments, the disorder or disease is the pain and inflammation associated with pulmonary disorders. [0312] In some embodiments, the disorder or disease is the pain and inflammation associated with allergies.

[0313] In some embodiments, the disorder or disease is the pain and inflammation associated with skin disorders.

[0314] In some embodiments, the disorder or disease is the pain and inflammation associated with diabetes.

[0315] In some embodiments, the disorder or disease is the pain and inflammation associated with pancreatitis.

[0316] In some embodiments, the disorder or disease is the pain and inflammation associated with tendonitis.

[0317] In some embodiments, the disorder or disease is the pain and inflammation associated with heart disease.

[0318] In some embodiments, the disorder or disease is the pain and inflammation associated with lupus.

[0319] In some embodiments, the disorder or disease is the pain and inflammation associated with a neurological disorder.

[0320] In some embodiments, the disorder or disease is the pain and inflammation associated with multiple sclerosis.

[0321] In some embodiments, the disorder or disease is the pain and inflammation associated with Parkinson's.

[0322] In some embodiments, the disorder or disease is cancer,

[0323] In some embodiments, the disorder or disease is systemic inflammation,

[0324] In some embodiments, the disorder or disease is metastatic melanoma,

[0325] In some embodiments, the disorder or disease is fatty liver disease,

[0326] In some embodiments, the disorder or disease is liver fibrosis,

[0327] In some embodiments, the disorder or disease is tendon regeneration,

[0328] In some embodiments, the disorder or disease is diabetes,

[0329] In some embodiments, the disorder or disease is degenerative disc disease,

[0330] In some embodiments, the disorder or disease is osteoarthritis,

[0331] In some embodiments, the disorder or disease is diabetic retinopathy,

[0332] In some embodiments, the disorder or disease is pulmonary fibrosis,

[0333] In some embodiments, the disorder or disease is idiopathic pulmonary fibrosis

(IPF).

[0334] In some embodiments, the disorder or disease is degenerative disc disease. [0335] In some embodiments, the disorder or disease is rheumatoid arthritis.

[0336] In some embodiments, the disorder or disease is scleroderma.

[0337] In some embodiments, the disorder or disease is a mycotic or viral infection.

[0338] In some embodiments, the disorder or disease is a bone or cartilage disease.

[0339] In some embodiments, the disorder or disease is a neurological disorder.

[0340] In some embodiments, the disorder or disease is Alzheimer's disease.

[0341] In some embodiments, the disorder or disease is osteoarthritis.

[0342] In some embodiments, the disorder or disease is lung disease.

[0343] In some embodiments, the disorder or disease is a genetic disease caused by mutations in Wnt signaling components, wherein the genetic disease is selected from: polyposis coli, osteoporosis-pseudoglioma syndrome, familial exudative vitreoretinopathy, retinal angiogenesis, early coronary disease, tetra-amelia syndrome, Miillerian-duct regression and virilization, SERKAL syndrome, diabetes mellitus type 2, Fuhrmann syndrome, Al-Awadi/Raas- Rothschild/Schinzel phocomelia syndrome, odonto-onycho-dermal dysplasia, obesity, split- hand/foot malformation, caudal duplication syndrome, tooth agenesis, Wilms tumor, skeletal dysplasia, focal dermal hypoplasia, autosomal recessive anonychia, neural tube defects, alpha- thalassemia (ATRX) syndrome, fragile X syndrome, ICF syndrome, Angelman syndrome, Prader- Willi syndrome, Beckwith-Wiedemann Syndrome, Norrie disease and Rett syndrome.

[0344] In some embodiments, the patient is a human.

[0345] In some embodiments, the cancer is chosen from: hepatocellular carcinoma, colon cancer, breast cancer, pancreatic cancer, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia, chronic lymphocytic leukemia (CLL), acute myeloid leukemia, acute lymphocytic leukemia, Hodgkin lymphoma, lymphoma, sarcoma and ovarian cancer.

[0346] In some embodiments, the cancer is chosen from: lung cancer - non-small cell, lung cancer - small cell, multiple myeloma, nasopharyngeal cancer, neuroblastoma, osteosarcoma, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer - basal and squamous cell, skin cancer - melanoma, small intestine cancer, stomach (gastric) cancers, testicular cancer, thymus cancer, thyroid cancer, uterine sarcoma, vaginal cancer, vulvar cancer, laryngeal or hypopharyngeal cancer, kidney cancer, Kaposi sarcoma, gestational trophoblastic disease, gastrointestinal stromal tumor, gastrointestinal carcinoid tumor, gallbladder cancer, eye cancer (melanoma and lymphoma), Ewing tumor, esophagus cancer, endometrial cancer, colorectal cancer, cervical cancer, brain or spinal cord tumor, bone metastasis, bone cancer, bladder cancer, bile duct cancer, anal cancer and adrenal cortical cancer.

[0347] In some embodiments, the cancer is hepatocellular carcinoma. [0348] In some embodiments, the cancer is colon cancer.

[0349] In some embodiments, the cancer is colorectal cancer.

[0350] In some embodiments, the cancer is breast cancer.

[0351] In some embodiments, the cancer is pancreatic cancer.

[0352] In some embodiments, the cancer is chronic myeloid leukemia (CML).

[0353] In some embodiments, the cancer is chronic myelomonocytic leukemia.

[0354] In some embodiments, the cancer is chronic lymphocytic leukemia (CLL)

[0355] In some embodiments, the cancer is acute myeloid leukemia.

[0356] In some embodiments, the cancer is acute lymphocytic leukemia.

[0357] In some embodiments, the cancer is Hodgkin lymphoma.

[0358] In some embodiments, the cancer is lymphoma.

[0359] In some embodiments, the cancer is sarcoma.

[0360] In some embodiments, the cancer is ovarian cancer.

[0361] In some embodiments, the cancer is lung cancer - non-small cell.

[0362] In some embodiments, the cancer is lung cancer - small cell.

[0363] In some embodiments, the cancer is multiple myeloma.

[0364] In some embodiments, the cancer is nasopharyngeal cancer.

[0365] In some embodiments, the cancer is neuroblastoma.

[0366] In some embodiments, the cancer is osteosarcoma.

[0367] In some embodiments, the cancer is penile cancer.

[0368] In some embodiments, the cancer is pituitary tumors.

[0369] In some embodiments, the cancer is prostate cancer.

[0370] In some embodiments, the cancer is retinoblastoma.

[0371] In some embodiments, the cancer is rhabdomyosarcoma.

[0372] In some embodiments, the cancer is salivary gland cancer.

[0373] In some embodiments, the cancer is skin cancer - basal and squamous cell

[0374] In some embodiments, the cancer is skin cancer - melanoma.

[0375] In some embodiments, the cancer is small intestine cancer.

[0376] In some embodiments, the cancer is stomach (gastric) cancers.

[0377] In some embodiments, the cancer is testicular cancer.

[0378] In some embodiments, the cancer is thymus cancer.

[0379] In some embodiments, the cancer is thyroid cancer.

[0380] In some embodiments, the cancer is uterine sarcoma.

[0381] In some embodiments, the cancer is vaginal cancer. [0382] In some embodiments, the cancer is vulvar cancer.

[0383] In some embodiments, the cancer is Wilms tumor.

[0384] In some embodiments, the cancer is laryngeal or hypopharyngeal cancer.

[0385] In some embodiments, the cancer is kidney cancer.

[0386] In some embodiments, the cancer is Kaposi sarcoma.

[0387] In some embodiments, the cancer is gestational trophoblastic disease.

[0388] In some embodiments, the cancer is gastrointestinal stromal tumor.

[0389] In some embodiments, the cancer is gastrointestinal carcinoid tumor.

[0390] In some embodiments, the cancer is gallbladder cancer.

[0391] In some embodiments, the cancer is eye cancer (melanoma and lymphoma).

[0392] In some embodiments, the cancer is Ewing tumor.

[0393] In some embodiments, the cancer is esophagus cancer.

[0394] In some embodiments, the cancer is endometrial cancer.

[0395] In some embodiments, the cancer is colorectal cancer.

[0396] In some embodiments, the cancer is cervical cancer.

[0397] In some embodiments, the cancer is brain or spinal cord tumor.

[0398] In some embodiments, the cancer is bone metastasis.

[0399] In some embodiments, the cancer is bone cancer.

[0400] In some embodiments, the cancer is bladder cancer.

[0401] In some embodiments, the cancer is bile duct cancer.

[0402] In some embodiments, the cancer is anal cancer.

[0403] In some embodiments, the cancer is adrenal cortical cancer.

[0404] In some embodiments, the disorder or disease is a neurological condition/disorder/disease, wherein the neurological condition/disorder/disease is selected from: Alzheimer's disease, frontotemporal dementias, dementia with Lewy bodies, prion diseases, Parkinson's disease, Huntington's disease, progressive supranuclear palsy, corticobasal degeneration, multiple system atrophy, amyotrophic lateral sclerosis (ALS), inclusion body myositis, autism, degenerative myopathies, diabetic neuropathy, other metabolic neuropathies, endocrine neuropathies, orthostatic hypotension, multiple sclerosis and Charcot-Marie-Tooth disease.

[0405] In some embodiments, the disorder or disease is a neurological condition/disorder/disease associated with tau protein, amyloid, alpha-synuclein pathology, Tar DNA -binding Protein of 43KDa (TDP-43), Prion protein PrP or fused in sarcoma (FUS). [0406] In some embodiments, the disorder or disease is a neurological condition/disorder/disease, wherein the neurological condition/disorder/disease is selected from the group consisting of: Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Down Syndrome, Frontotemporal Dementia with Parkinsonism- 17 (FTDP-17), Lewy body dementia, Parkinson's Disease, Pick's Disease, and additional diseases with pronounced neurodegeneration such as Autism, Dementia, Epilepsy, Huntington's Disease, Multiple Sclerosis; diseases and disorders associated with acquired brain injury such as Chronic Traumatic Encephalopathy, Traumatic Brain Injury, Tumor, and Stroke.

[0407] In some embodiments, the disorder or disease is a fibrotic disorder, wherein the fibrotic disorder is selected from the group consisting of: skin fibrosis; scleroderma; progressive systemic fibrosis; lung fibrosis; muscle fibrosis; kidney fibrosis; glomerulosclerosis; glomerulonephritis; hypertrophic scar formation; uterine fibrosis; renal fibrosis; cirrhosis of the liver, liver fibrosis; adhesions; chronic obstructive pulmonary disease; fibrosis following myocardial infarction; pulmonary fibrosis; fibrosis and scarring associated with diffuse/interstitial lung disease; central nervous system fibrosis; fibrosis associated with proliferative vitreoretinopathy (PVR); restenosis; endometriosis; ischemic disease, and radiation fibrosis.

[0408] In some embodiments, the disorder or disease is chronic inflammation associated with eye disorders, joint pain, arthritis (rheumatoid, osteo, psoriatic gout), cancers (colon, breast, lung, pancreas, and others), gastrointestinal disorders (ulcerative colitis and inflammatory bowel diseases), pulmonary disorders (chronic obstructive pulmonary disorder and asthma), allergies, skin disorders (atopic dermatitis and psoriasis), diabetes, pancreatitis, tendonitis, hepatitis, heart disease, myocarditis, stroke, lupus, and neurological disorders such as multiple sclerosis, Parkinson's and dementia including Alzheimer's disease.

[0409] In some embodiments, a compound of Formula (I) inhibits DYRK1A.

[0410] In some embodiments, a compound of Formula (I) inhibits GSK3.

[0411] In some embodiments, a compound of Formula (I) inhibits GSK3 .

[0412] In some embodiments, a compound of Formula (I) inhibits DYRK1A and

GSK3 .

[0413] In some embodiments, the compound of Formula (I) inhibits one or more proteins in the Wnt pathway.

[0414] In some embodiments, the compound of Formula (I) inhibits signaling induced by one or more Wnt proteins. [0415] In some embodiments, the Wnt proteins are chosen from: WNT1, WNT2, WNT2B, WNT3, WNT3A, WNT4, WNT5A, WNT5B, WNT6, WNT7A, WNT7B, WNT8A, WNT8B, WNT9A, WNT9B, WNT10A, WNT10B, WNT11, and WNT16.

[0416] In some embodiments, the compound of Formula (I) inhibits a kinase activity.

[0417] In some embodiments, the method treats a disease or disorder mediated by the Wnt pathway in a patient, the method comprises administering to the patient a therapeutically effective amount of a compound (or compounds) of Formula (I), or a pharmaceutically acceptable salt thereof.

[0418] In some embodiments, the compound of Formula (I) inhibits one or more Wnt proteins.

[0419] In some embodiments, the method treats a disease or disorder mediated by kinase activity in a patient, the method comprises administering to the patient a therapeutically effective amount of a compound (or compounds) of Formula (I), or a pharmaceutically acceptable salt thereof.

[0420] In some embodiments, the disease or disorder comprises tumor growth, cell proliferation, or angiogenesis.

[0421] In some embodiments, the method inhibits the activity of a protein kinase receptor, the method comprises contacting the receptor with an effective amount of a compound (or compounds) of Formula (I), or a pharmaceutically acceptable salt thereof.

[0422] In some embodiments, the method treats a disease or disorder associated with aberrant cellular proliferation in a patient; the method comprises administering to the patient a therapeutically effective amount of a compound (or compounds) of Formula (I), or a pharmaceutically acceptable salt thereof.

[0423] In some embodiments, the method prevents or reduces angiogenesis in a patient; the method comprises administering to the patient a therapeutically effective amount of a compound (or compounds) of Formula (I), or a pharmaceutically acceptable salt thereof.

[0424] In some embodiments, the method prevents or reduces abnormal cellular proliferation in a patient; the method comprises administering to the patient a therapeutically effective amount of a compound (or compounds) of Formula (I), or a pharmaceutically acceptable salt thereof.

[0425] In some embodiments, the method treats a disease or disorder associated with aberrant cellular proliferation in a patient, the method comprises administering to the patient a pharmaceutical composition comprising one or more of the compounds of claim 1 in combination with a pharmaceutically acceptable carrier and one or more other agents. [0426] Moreover, the compounds and compositions, for example, as inhibitors of the cyclin-dependent kinases (CDKs), can modulate the level of cellular RNA and DNA synthesis and therefore are expected to be useful in the treatment of viral infections such as HIV, human papilloma virus, herpes virus, Epstein -Barr virus, adenovirus, Sindbis virus, pox virus and the like.

[0427] Compounds and compositions described herein can inhibit the kinase activity of, for example, CDK/cyclin complexes, such as those active in the Go or Gi stage of the cell cycle, e.g., CDK2, CDK4, and/or CDK6 complexes.

Evaluation of Biological Activity

[0428] The biological activity of the compounds described herein can be tested using any suitable assay known to those of skill in the art, see, e.g., WO 2001/053268 and WO 2005/009997. For example, the activity of a compound may be tested using one or more of the test methods outlined below.

[0429] In one example, tumor cells may be screened for Wnt independent growth. In such a method, tumor cells of interest are contacted with a compound (i.e. inhibitor) of interest, and the proliferation of the cells, e.g. by uptake of tritiated thymidine, is monitored. In some embodiments, tumor cells may be isolated from a candidate patient who has been screened for the presence of a cancer that is associated with a mutation in the Wnt signaling pathway. Candidate cancers include, without limitation, those listed above.

[0430] In another example, one may utilize in vitro assays for Wnt biological activity, e.g. stabilization of β-catenin and promoting growth of stem cells. Assays for biological activity of Wnt include stabilization of β-catenin, which can be measured, for example, by serial dilutions of a candidate inhibitor composition. An exemplary assay for Wnt biological activity contacts a candidate inhibitor with cells containing constitutively active Wnt/ -catenin signaling. The cells are cultured for a period of time sufficient to stabilize β-catenin, usually at least about 1 hour, and lysed. The cell lysate is resolved by SDS PAGE, then transferred to nitrocellulose and probed with antibodies specific for β-catenin.

[0431] In a further example, the activity of a candidate compound can be measured in a Xenopus secondary axis bioassay (Leyns, L. et al. Cell (1997), 88(6), 747-756).

[0432] In another example, in vitro assays for DYRK1A biological activity may be used, e.g. regulation of microtubule-associated protein tau (MAPT/Tau) phosphorylation in neuronal cell line such as the human SH-SY5Y neuroblastoma cell line. Assays for DYRK1A- regulated level of phosphorylation can include monitoring levels of basal pSer396 Tau, which can be measured, for example, by serial dilutions of a candidate inhibitor composition using a ten micromolar top concentration and detected by ELISA or Western Blotting. An exemplary assay for DYRK-1 A-regulated phosphorylation uses the SH-SY5Y cells cultured in a 96 well plate format for a period of time sufficient to stabilize microtubules and Tau phosphorylation, usually at least 2 days, then treated with a 1/3 serial dilution of compounds overnight and lysed. The cell lysate is resolved by SDS PAGE, then transferred to nitrocellulose and probed with an antibody specific for pSer396 Tau. The chemiluminescence signal for HRP -linked antibodies used in western blotting is detected using a Carestream Image Station and blot densitometry for pSer396 and beta-actin are analyzed using Image J (NIH).

[0433] In a further example, the activity of a candidate compound can be measured by ELISA by adding the lysate mentioned above onto total Tau-coated plates and detected with a specific pSer396 antibody. Colorimetric detection of ELISA signal is performed by Cytation3 plate reader (Biotek).

[0434] To further illustrate this disclosure, the following examples are included. The examples should not, of course, be construed as specifically limiting the disclosure. Variations of these examples within the scope of the claims are within the purview of one skilled in the art and are considered to fall within the scope of the disclosure as described, and claimed herein. The reader will recognize that the skilled artisan, armed with the present disclosure, and skill in the art is able to prepare and use the disclosure without exhaustive examples.

EXAMPLES

Compound preparation

[0435] The starting materials used in preparing the compounds of the disclosure are known, made by known methods, or are commercially available. It will be apparent to the skilled artisan that methods for preparing precursors and functionality related to the compounds claimed herein are generally described in the literature. The skilled artisan given the literature and this disclosure is well equipped to prepare any of the compounds.

[0436] It is recognized that the skilled artisan in the art of organic chemistry can readily carry out manipulations without further direction, that is, it is well within the scope and practice of the skilled artisan to carry out these manipulations. These include reduction of carbonyl compounds to their corresponding alcohols, oxidations, acylations, aromatic substitutions, both electrophilic and nucleophilic, etherifications, esterification and saponification and the like. These manipulations are discussed in standard texts such as March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure 7 th Ed., John Wiley & Sons (2013), Carey and Sundberg, Advanced Organic Chemistry 5 th Ed., Springer (2007), Comprehensive Organic Transformations: A Guide to Functional Group Transformations, 2 nd Ed., John Wiley & Sons (1999) (incorporated herein by reference in its entirety)and the like.

[0437] The skilled artisan will readily appreciate that certain reactions are best carried out when other functionality is masked or protected in the molecule, thus avoiding any undesirable side reactions and/or increasing the yield of the reaction. Often the skilled artisan utilizes protecting groups to accomplish such increased yields or to avoid the undesired reactions. These reactions are found in the literature and are also well within the scope of the skilled artisan. Examples of many of these manipulations can be found for example in P. Wuts Greene 's Protective Groups in Organic Synthesis, 5th Ed., John Wiley & Sons (2014), incorporated herein by reference in its entirety.

[0438] Trademarks used herein are examples only and reflect illustrative materials used at the time of the disclosure. The skilled artisan will recognize that variations in lot, manufacturing processes, and the like, are expected. Hence the examples, and the trademarks used in them are non-limiting, and they are not intended to be limiting, but are merely an illustration of how a skilled artisan may choose to perform one or more of the embodiments of the disclosure.

[0439] (¾) nuclear magnetic resonance spectra (NMR) were measured in the indicated solvents on a Bruker NMR spectrometer (Avance TM DRX300, 300 MHz for ¾ or Avance TM DRX500, 500 MHz for ¾) or Varian NMR spectrometer (Mercury 400BB, 400 MHz for ¾). Peak positions are expressed in parts per million (ppm) downfield from tetramethylsilane. The peak multiplicities are denoted as follows, s, singlet; d, doublet; t, triplet; q, quartet; ABq, AB quartet; quin, quintet; sex, sextet; sep, septet; non, nonet; dd, doublet of doublets; ddd, doublet of doublets of doublets; d/ABq, doublet of AB quartet; dt, doublet of triplets; td, triplet of doublets; dq, doublet of quartets; m, multiplet.

[0440] The following abbreviations have the indicated meanings:

brine = saturated aqueous sodium chloride

CDCh = deuterated chloroform

DCE = dichloroethane

DCM = dichloromethane

DEAD = diethyl azodicarboxylate

DIPEA = N,N-diisopropylethylamine

DMA = dimethylacetamide

DMAP = 4-dimethylaminopyridine

DMF = N,N-dimethylformamide

DMSO- e = deuterated dimethylsulfoxide

ESIMS = electron spray mass spectrometry EtOAc = ethyl acetate

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

HC1 = hydrochloric acid

HOAc = acetic acid

ISCO = Teledyne ISCO, Inc brand CombiFlash ® Rf 200

KOAc = potassium acetate

LC/MS = Liquid chromatography-mass spectrometry

MeCN = acetonitrile

Me OH = methanol

MgS04 = magnesium sulfate

MW = microwave irradiation

NaB¾CN = sodium cyanoborohydride

NaHCC = sodium bicarbonate

Na(OAc)3BH = Sodium triacetoxyborohydride

NazSaOs = sodium thiosulfate

NMR = nuclear magnetic resonance

ON = overnight

Pd(dppf)Cl2 = 1, r-bis(diphenylphosphino)ferrocene-palladium(II)dichloride

Pd(PPli3)4 = tetrakis(triphenylphosphine)palladium(0)

r.t. = room temperature

SPhos Pd G4 = Methanesulfonato(2-dicyclohexylphosphino-3,6-dimethox} -2',4',6'-tri-i- propyl- 1 , 1 '-biphenyl)(2'-methylamino- 1 , 1 L biphenyl-2-yl)palladium(II)

TBAF = Tetra-n4outylammonium fluoride,

TEA = triethylamine

TFA = trifiuoroacetic acid

THF = tetrahydrofuran

TLC = thin layer chromatography

[0441] The following example schemes are provided for the guidance of the reader, and collectively represent an example methods for making the compounds provided herein. Furthennore. other methods for preparing compounds of the disclosure will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. The skilled artisan is thoroughly equipped to prepare these compounds by those methods given the literature and this disclosure. The compound numberings used in the synthetic schemes depicted below are meant for those specific schemes only, and should not be construed as or confused with same numberings in other sections of the application. Unless otherwise indicated, all variables are as defined above.

General procedures

[001] Compounds of Formula I of the present disclosure can be prepared as depicted in Scheme 1.

Scheme 1

[002] Scheme 1 describes a method for preparation of isoquinoline-3-carboxamide derivatives (IX) by first coupling the amine with a variety of acids (III) to produce amide IV. The bromo derivative IV is then reacted with bis(pinacolato)diboron to give the pinacol ester (V). Suzuki coupling with a variety of 5-membered heteroaryl bromides (VIII) yields the desired R 3 substituted isoquinoline IX. Alternatively, the bromo derivative IV is Suzuki coupled with a variety of 5-membered heteroaryl pinacol esters (VI) or coupled to a variety of 5-membered heteroaryl stannanes (VII) to produce the final R 3 substituted isoquinoline IX.

[003] In some embodiments, compounds of Formula I of the present disclosure can be prepared as depicted in Scheme 2.

A = C, , O, or S,

wherein C or N may be substituted

as defined for R 6 herein

Scheme 2

[004] Scheme 2 describes a method for preparation of isoquinoline-3-carboxamide intermediate (IVa) by first coupling the amine 4-nitrophenyl carbonochloridate followed by coupling with a variety of R 6 NH heterocyclyls. Intermediate IVa could then be used in place of IV in Scheme 1 or 3.

[005] In some embodiments, compounds of Formula I of the present disclosure can

A = C, N, O, or S,

wherein C or N may be substituted

as defined for R 6 herein

Scheme 3

[006] Scheme 1 describes a method for preparation of isoquinoline-3-carboxamide derivatives (IXa) by first reacting with bis(pinacolato)diboron to give the pinacol ester (X). Suzuki coupling with a variety of 5-membered heteroaryl bromides (VIII) yields the desired R 3 substituted isoquinoline amine XI. The amine (XI) is then reacted with diphosgene to form the isocyanate (XII) which is then reacted with a variety of a variety of R 6 NH heterocyclyls produce the final R 3 and R 6 substituted isoquinoline IXa.

Illustrative Compound Examples

[007] Preparation of intermediate 6-bromoisoquinolin-l-d-3-amine (XIV) is depicted below in Scheme 4.

XIII XIV

(Oakwood Chemicals)

Scheme 4

Step 1

[008] To a mixture of l,6-dibromoisoquinolin-3-amine (XIII) (0.5 g, 1.66 mmol), ammonium formate- j (0.56 g, 8.28 mmol) and Pd(PPh 3 ) 4 (191.3 mg, 0.170 mmol) in DMF (5 mL) was heated to 50°C for 48 h. The solvents were concentrated and the residue was suspended in chloroform. The solid was collected by filtration and washed with water and EtOAc. The solid were dried under high vacuo to obtain 6-bromo-l-deuterio-isoquinolin-3-amine (XIV) (115 mg, 0.513 mmol, 31.0% yield) as a pale yellow solid. ¾ NMR (500 MHz, DMSO-t¾) δ ppm 6.11 (2 H, s), 6.55 (1 H, s), 7.22 (1 H, dd, J=8.78, 1.92 Hz), 7.73 (1 H, d, J=8.51 Hz), 7.79 (1 H, d, J=1.92 Hz); ESIMS found for C 9 H 6 DBrN 2 mlz 224.0 ( 79 BrM+H).

[009] Preparation of intermediate 6-bromo-4-chloroisoquinolin-3-amine (XVI) is depicted below in Scheme 5.

XV XVI

Scheme 5 Step 1

[010] To a stirred suspension of 6-bromoisoquinolin-3-amine (XV) (1.0 g, 4.48 mmol) in DMF (15 mL) at 0°C was added l-chloropyrrolidine-2,5-dione (598.6 mg, 4.48 mmol) portionwise. The mixture was stirred at 0°C for 6 h. The reaction mixture was added to water (150 mL), stirred for 1 h and the resulting solids were collected by filtration and air dried overnight to obtain 6-bromo-4-chloro-isoquinolin-3-amine (XVI) (922 mg, 3.58 mmol, 79.9% yield) as a beige solid which was used for next step without purification. ¾ NMR (499 MHz, DMSO- e) δ ppm 6.55 (2 H, s), 7.40 (1 H, dd, J=8.64, 1.78 Hz), 7.88 (1 H, d, J=8.51 Hz), 7.90 (1 H, d, J=1.10 Hz), 8.86 (1 H, s); ESIMS found for C 9 H 6 BrClN 2 mlz 256.9 ( 79 BrM+H).

[011] Preparation of intermediate 6-bromo-4-methylisoquinolin-3-amine (XVIII) is depicted below in Scheme 6.

XV XVII xvm

Scheme 6

Step 1

[012] To a stirred suspension of 6-bromoisoquinolin-3 -amine (XV) (2.g, 8.97mmol) in DMF (25.1 mL) at 0°C was added l -iodopyrrolidine-2,5-dione (2.02 g, 8.97 mmol) portionwise, The mixture was stirred at 0°C for 1 hr. LC/MS of the mixture showed completion of the reaction and the desired product. The solvent was removed under vacuum, the residue was purified by C 18 Silica gel (240g) [0→100% H 2 0/MeCN (0.1%Formic acid)] to produce 6-bromo-4-iodo-isoquinolin-3-amine (XVII) (1.95 g, 5.58 mmol, 62.2% yield) as a brown solid. ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 6.41 (2 H, br s), 7.40 (1 H, dd, J=8.64, 1.78 Hz), 7.76 - 7.81 (1 H, m), 7.82 ( 1 H, d, J=8.51 Hz), 8.81 (1 H, s); ESIMS found for C 9 H 6 BrIN 2 mlz 348.9 ( 79 BrM+H).

Step 2

[013] A stirred solution of 6-bromo-4-iodo-isoquinolin-3-amine (XVII) (1.0 g, 2.87 mmol), 2,4,6-trimethyl-l,3,5,2,4,6-trioxatriborinane (0.72 g, 2.87 mmol), Pd(dppf)Cl 2 (0.23 g, 0.29 mmol), and K3PO4 (5.73 mL, 5.73 mmol) in 1,4-dioxane ( 10 mL) was heated to 90°C for 3 days. The solvent was removed under high vacuum and the residue was purified by C 18 silica gel (240g) [0→20% H 2 0/MeCN (0.1%Formic acid)] to produce 6-bromo-4-methyl-isoquinolin-3-amine (XVIII) (74 mg, 0.312 mmol, 10.9% yield) as an off-white solid. ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 2.23 (3 H, br s), 5.91 (2 H, br s), 7.27 ( 1 H, br d, J=2.20 Hz), 7.71 - 7.82 ( 1 H, m), 7.92 (1 H, br s), 8.72 ( 1 H, br s); ESIMS found for Ci 0 H 9 BrN 2 ra/z 239.0 ( 81 BrM+H).

[014] Preparation of intermediate 6-bromo-7-fluoroisoquinolin-3-amine (XXI) is depicted below in Scheme 7.

Scheme 7 Step 1

[015] To a vial was added 2,2-diethoxyacetonitrile (XIX) (1.0 g, 7.74 mmol) dissolved MeOH (7.74 mL) followed by addition of MeONa/MeOH (0.18 mL, 0.77 mmol) dropwise. The reaction was stirred at room temperature for 20 h. HO Ac (44.3 μί, 0.77 mmol) was added until pH=7-8 (using pH strips). (4-Bromo-3-fluoro-phenyl)methanamine hydrochloride (XX) ( 1.86 g, 7.74 mmol) was added and stirred at 40°C for 4 h. The solvent was removed under vacuum. Sulfuric acid (12.6 mL, 232.3 mmol) was added and stirred at 40°C for 16 h. NH4OH (30.8 mL, 240.0 mmol) was added dropwise at 0°C. The solvent was removed under vacuum and the residue was purified by C18 silica gel (240g) [0→50% H 2 0/MeCN (0.1%Formic acid)] to produce 6-bromo-7-fluoro-isoquinolin-3-amine (XXI) (1.33 g, 5.50 mmol, 71.1% yield) as an off- white solid. 'H NMR (499 MHz, DMSO- 6 ) δ ppm 6.07 (2 H, s), 6.61 ( 1 H, s), 7.76 (1 H, d, J=9.33 Hz), 8.01 (1 H, d, J=6.86 Hz), 8.80 ( 1 H, s); ESIMS found for C 9 H 6 BrFN 2 mlz 242.9 ( 81 BrM+H).

[016] Preparation of intermediates 6-bromo-7-chloroisoquinolin-3-amine (XXIII) and 6-bromo-5-chloroisoquinolin-3-amine (XXIV) is depicted below in Scheme 8.

Scheme 8

Step 1

[017] To a stirred solution of 2,2-diethoxyacetonitrile (XIX) (0.59 g, 4.57 mmol) in a vial containing MeOH (4.57 mL) was added MeONa (0.1 mL, 0.46 mmol) dropwise. The reaction was stirred at 35°C for 20 h. HOAc was added (26.1 μί, 0.46 mmol) (checked that the pH is 7-8 using pH strips) followed by (4-bromo-3-chloro-phenyl)methanamine (XXII) ( 1.01 g, 4.57 mmol). The mixture was stirred at 35°C for 40 h. The solvent was removed under vacuum. Sulfuric Acid (7.43 mL, 137.0 mmol) was then added and stirred at 35°C for 16 h. NH 4 OH (60.6 mL, 141.6 mmol) was added at 0°C. The reaction was filtered through Celite and purified by CI 8 silica gel (240g) [0→30% H 2 0/MeCN (0.1%Formic acid)] to produce a 1 : 1 mixture (by NMR) of 6-bromo-7- chloro-isoquinolin-3-amine (XXIII) and 6-bromo-5-chloroisoquinolin-3-amine (XXIV) (633.7 mg, 2.46 mmol, 53.9% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 6.23 (2 H, s), 6.46 (2 H, s), 6.57 (1 H, s), 6.83 (1 H, s), 7.40 ( 1 H, d, J=8.51 Hz), 7.74 (1 H, d, J=8.51 Hz), 8.05 (1 H, s), 8.09 ( 1 H, s), 8.81 ( 1 H, s), 8.88 ( 1 H, s); ESIMS found for C 9 H 6 BrClN 2 mlz 256.9 ( 79 BrM+H).

[018] Preparation of intermediates 6-bromo-7-methylisoquinolin-3 -amine (XXVI) and -bromo-5-methylisoquinolin-3-amine (XXVII) is depicted below in Scheme 9.

XXV

Scheme 9

Step 1

[019] To a stirred solution of 2,2-diethoxyacetonitrile (XIX) (0.33 g, 2.52 mmol) in a vial containing MeOH (2.52 mL) was added MeONa (0.23 mL, 0.25 mmol) dropwise. The reaction was stirred at 22°C for 20 h. HO Ac was added (14.4 μί, 0.25 mmol) (checked that the pH is 7-8 using pH strips) followed by (4-bromo-3-methyl-phenyl)methanamine (XXV) (0.5 g, 2.52 mmol). The mixture was stirred at 40°C for 40 h. The solvent was removed under vacuum. Sulfuric Acid (4.09 mL, 75.49 mmol) was then added and stirred at 40°C for 16 h. NH 4 OH (33.4 mL, 78 mmol) was added at 0°C. The reaction was filtered through Celite and purified by CI 8 silica gel (240g) [0→30% H 2 0/MeCN (0.1% Formic acid)] to produce a 1: 1 mixture (by NMR) of 6-bromo- 7-methylisoquinolin-3-amine (XXVI) and 6-bromo-5-methylisoquinolin-3-amine (XXVII) (378 mg, 1.59 mmol, 63.4% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 2.40 (3 H, s), 2.52 (3 H, s), 5.96 (2 H, s), 6.12 (1 H, s), 6.54 (1 H, s), 6.71 (1 H, s), 7.27 (1 H, d, J=8.78 Hz), 7.58 (1 H, d, J=8.78 Hz), 7.73 (1 H, s), 7.86 (1 H, s), 8.74 (1 H, s), 8.79 (1 H, s); ESIMS found for CioH 9 BrN 2 mlz 237.0 ( 79 BrM+H).

[020] Preparation of intermediate fr «5-4-((teri-butoxycarbonyl)(methyl)amino) cyclohexane-l-carboxylic acid (XXX) is depicted below in Scheme 10.

Scheme 10

Step 1

[021] To a solution of methyl fr «5-4-((teri-butoxycarbonyl)amino)cyclohexane-l- carboxylate (XXVIII) (1.3 g, 5 mmol) in DMF (15 mL) and cooled to 0°C was added sodium hydride (60% in oil, 240 mg, 6 mmol) over 30 minutes. The mixture is stirred at room temperature for 1 h, then cooled to 0°C and treated with iodomethane (0.38 mL, 6 mmol). After stirring overnight at room temperature, the mixture is poured into a saturated aqueous ammonium chloride and extracted with EtOAc. The combined organic phase is washed with water and brine, dried over anhydrous Na2SC>4 and concentrated under reduced pressure. The obtained residue is purified by column chromatography on silica gel (eluent: n-hexane -EtOAc 10: 1) to obtain methyl fra«s-4- ((teri-butoxycarbonyl)(methyl)amino)cyclohexane-l-carboxylat e (XXIX) (1.3 g, 4.79 mmol, 94.8% yield).

Step 2

[022] To a stirred solution of methyl fr «5-4-((teri-butoxycarbonyl)(methyl)amino) cyclohexane-l-carboxylate (XXIX) (130 mg, 4.79 mmol) in a mixture of MeOH (10 mL) and THF (10 mL) was added 2 N aqueous NaOH (4.79 mL, 9.58 mmol) and the mixture was stirred for 4 h. The solvent was concentrated, the residue taken in water and acidified with IN HC1 and extracted with EtOAc. The organics were washed with 2 x water then 1 x brine. The organics were then separated and dried (MgSO.*) before concentration to dryness to obtain trans-4-((tert- butoxycarbonyl)(methyl)amino)cyclohexane-l-carboxylic acid (XXX) as a thick gum (1.198 g, 4.65 mmol, 97.2% yield) which was used for next step without purification.

[023] Preparation of intermediate 2-(4-(fert-butoxycarbonyl)piperazin-l- yl)isonicotinic acid (XXXIV) is depicted below in Scheme 11.

ΧΧΧΠ

Scheme 11 Step 1

[024] To a solution of ethyl 2-chloroisonicotinate (XXXI) ( 10 g, 53.88 mmol) was added fert-butyl piperazine-l-carboxylate (XXXII) (0.32 mL, 2.89 mmol) and DIPEA (18.8 mL, 107.75 mmol). The reaction was stirred at 1 10°C for 16 h. The mixture was poured into water, extracted with EtO Ac, and dried over Na2SC>4. The solvent was removed under high vacuum and the residue was purified on a silica gel column (120g) (0→100% hexane/EtOAc) to give fert-butyl 4-(4-(ethoxycarbonyl)pyridin-2-yl)piperazine-l -carboxylate (XXXIII) as a brown oil (10.84 g, 32.32 mmol, 60.0% yield). ESIMS found for mlz 336.15 (M+H).

Step 2

[025] To a solution of tert-buty\ 4-(4-(ethoxycarbonyl)pyridin-2-yl)piperazine-l- carboxylate (XXXIII) ( 10.7 g, 31.9 mmol) in Me OH (130 mL) and water (26 mL) was added 4 M aqueous lithium hydroxide (7.98 mL, 31.9 mmol). The reaction was stirred at room temperature for 16 h. The reaction was poured into water and neutralized with concentrated hydrogen chloride (31.9 mL, 31.9 mmol) and extracted with EtO Ac, The organic layer was dried over Na2S04 and evaporated under high vacuum to produce 2-(4-(fert-butoxycarbonyl)piperazin-l-yl)isonicotinic acid (XXXIV) as a white solid (8.79 g, 28.6 mmol, 89.7% yield) which was used without further purification. ESIMS found for Ci5H 2 iN 3 0 4 mlz 308.15 (M+H).

[026] Preparation of intermediate 4-((l-(fer/-butoxycarbonyl)piperidin-4-yl)oxy) benzoic acid (XXXVIII) is depicted below in Scheme 12.

XXXV XXXVII XXXVIII

Scheme 12

Step 1

[027] To a solution of DEAD ( 12.3 mL, 27.08 mmol) (40% in toluene) was added to a mixture of ethyl 4-hydroxybenzoate (XXXV) (3.0 g, 18.05 mmol), fert-butyl 4- hydroxypiperidine-l-carboxylate (XXXVI) (4.72 g, 23.47 mmol) and triphenylphosphane (6.16 g, 23.47 mmol) in THF (40 mL) at 0°C. The mixture was stirred from 0°C to room temperature over 1 day before concentrating in vacuo. The residue was diluted with EtO Ac, washed with 1 N NaOH and brine, and then evaporated under vacuum. The crude product was purified by chromatography (0→30% EtOAc/hexanes) to give fert-butyl 4-(4-ethoxycarbonylphenoxy)piperidine-l- carboxylate (XXXVII) (5.4 g, 15.45 mmol, 85.6% yield) as a colorless oil. ESIMS found for Ci 9 H 2 7N0 5 mlz 372.1 (M+Na).

Step 2

[028] To a solution of fert-butyl 4-(4-ethoxycarbonylphenoxy)piperidine-l- carboxylate (XXXVII) (5.4 g, 15.45 mmol) in Me OH (10 mL) and THF (10 mL) was added LiOH ( 15.5 mL, 61.82 mmol) and the mixture stirred at 60°C for 2h. The mixture was concentrated and the residue triturated with water. The resulting solution was acidified with 2 N HCl until a solid precipitated. The solid was filtered and washed with water to afford 4-[(l-fert-butoxycarbonyl-4- piperidyl)oxy]benzoic acid (XXXVIII) (4.7 g, 14.63 mmol, 94.6% yield) as a white solid. ¾ NMR (499 MHz, DMSO- e) δ ppm 1.40 (9 H, s), 1.47 - 1.57 (2 H, m), 1.89 - 1.97 (2 H, m), 3.12 - 3.23 (2 H, m), 3.63 - 3.70 (2 H, m), 4.63 - 4.71 (1 H, m), 7.04 (2 H, d, J=9.06 Hz), 7.87 (2 H, d, J=9.06 Hz); ESIMS found for Ci7H 23 N0 5 mlz 344.1 (M+Na).

[029] Preparation of intermediate 2-( 1 -(fert-butoxycarbonyl)piperidin-4-yl)oxazole- 4-carboxylic acid (XLIII) is depicted below in Scheme 13.

XLIII XLII

Scheme 13 Step 1

[030] To a suspension of methyl (2S)-2-amino-3 -hydroxy-propanoate hydrochloride (XL) (3.21 g, 20.63 mmol) (1.1 eq) in DCM (40 mL) was added l,4-diazabicyclo[2.2.2]octane (6.3 lg, 56.27mmol) (3.0 eq). The reaction mixture was stirred at room temperature for 20 min before adding fert-butyl 4-formylpiperidine-l-carboxylate (XXXIX) (4.0 g, 18.76 mmol) (1.0 eq). The reaction mixture was stirred at room temperature for 30 min. The reaction mixture was cooled to 0°C, and l-chloropyrrolidine-2,5-dione (2.75 g, 20.63 mmol) (1.1 eq) was added and stirred at the room temperature for 16 h. Saturated aqueous Na2S2C>3 was added to the reaction mixture and extracted with DCM. Organic layer was washed with saturated aqueous NaHCC and brine, dried over Na2SC>4, and evaporated in vacuo. The crude product was purified by silica gel chromatography (0→50% EtOAc/Hexanes) to afford methyl 2-(l-(fert-butoxycarbonyl)piperidin- 4-yl)-2,5-dihydrooxazole-4-carboxylate (XLI) as a light brown oil (4.9 g, 15.69 mmol, 83.6% yield). ESIMS found for Ci 5 H 2 4N 2 0 5 mlz 213.1 (M+H-Boc).

Step 2

[031] To a suspension of methyl 2-(l -(tert-butoxycarbonyl)piperidin-4-yl)-2,5- dihydrooxazole-4-carboxylate (XLI) (3.63 g, 20.39 mmol) (4.9 g, 15.69 mmol) and K 2 C0 3 (2.82 g, 20.39 mmol) in DCM (50 mL) was added l -bromopyrrolidine-2,5-dione (3.63 g, 20.39 mmol). The reaction mixture heated at reflux for 16 h. Water ( 100 mL) was then added and the mixture extracted with DCM. The organic layer was separated, washed with brine, dried over Na 2 SC>4, and evaporated in vacuo. The crude product was purified by silica gel chromatography (0→50% EtOAc/Hexanes) to give methyl 2-(l -(fer7-butoxycarbonyl)piperidin-4-yl)oxazole-4-carboxylate (XLII) as a light brown oil (3.7 g, 1 1.92 mmol, 76.0% yield). ESIMS found for Ci 5 H 2 2N 2 0 5 mlz 333.10 (M+Na).

Step 3

[032] To a solution of methyl 2-( l-(fert-butoxycarbonyl)piperidin-4-yl)-2,5- dihydrooxazole-4-carboxylate (XLII) in Me OH (20 mL) and THF (20 mL) was added 3 M aqueous LiOH (1 1.54 mL, 34.61 mmol). The mixture stirred at 60°C for 1 h. The mixture was then concentrated to remove the organic solvents. The residual water was acidified with 2 N HC1 and the mixture extracted with EtOAc. The organic layer was washed with brine, dried, filtered and concentrated to afford 2-(l -(fer7-butoxycarbonyl)piperidin-4-yl)oxazole-4-carboxylic acid (XLIII) as a brown foam (4.6 g, 15.52 mmol, 89.7% yield). ESIMS found for Ci4H 2 oN 2 0 5 mlz 319.10 (M+Na).

[033] Preparation of intermediate 2-( 1 -(fert-butoxycarbonyl)piperidin-4-yl)oxazole- 4-carboxylic acid XLVI) is depicted below in Scheme 14.

XLIV XLVI

Scheme 14

Step 1

[034] A mixture of 5-bromopyridin-3-ol (XLIV) (2 g, 1 1.49 mmol), fert-butyl 4- ((methylsulfonyl)oxy)piperidine-l -carboxylate (XLV) (3.53 g, 12.64 mmol) and Cs 2 CC>3 (4.87 g, 14.94 mmol) in DMF (20 mL) was stirred at 80°C for 16 h. The mixture was diluted with water and then extracted with EtOAc. The organic layer was washed with water, brine, and dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified on a silica gel column (0→35% EtOAc/hexane) to give fert-butyl 4-((5-bromopyridin-3-yl)oxy)piperidine-l- carboxylate (XL VI) as a white solid (2.88 g, 8.06 mmol, 70.1% yield). ESIMS found for Ci 5 H 2 iBrN 2 0 3 mlz 357.05 (M+H).

[035] Preparation of intermediate 2-( 1 -(fert-butoxycarbonyl)piperidin-4-yl)oxazole- 4-carboxylic acid XLIX) is depicted below in Scheme 15.

XLVII XLIX

Scheme 15

Step 1

[036] To a solution of fert-butyl 4-aminopiperidine-l-carboxylate (XLVIII) (2 g, 9.99 mmol) in dry DMF (9.99 ml) was added 2,6-dichloropyrazine (XLVII) (1.488 g, 9.99 mmol). To the mixture was added DIPEA (5.22 mL, 30.0 mmol) and the reaction was stirred at 95 °C or 20 h. The solution was poured into water (200 mL). The solution was allowed to stand for 16 h. The solid was filtered and dried under vacuum, to produce fert-butyl 4-((6-chloropyrazin-2- yl)amino)piperidine-l-carboxylate) (XLIX) as an off-white solid (1.0172 g, 3.25 mmol, 32.6% yield. ESIMS found for C14H21CIN4O2 mlz 335.1 (M+Na).

[037] The following intermediates were prepared in accordance with the procedure described in the above Scheme 15.

L [038] fert-Butyl 3-[[(6-chloropyrazin-2-yl)amino]methyl]-3-fluoro-azetidine-l - carboxylate (L): Off-white solid, (56.4% ield). ESIMS found Ci 3 Hi 8 ClFN 4 02 mlz 338.95 (M+Na).

LI

[039] fert-Butyl (3S,4S)-4-((6-chloropyrazin-2-yl)amino)-3-fluoropiperidine-l - carboxylate (LI): Off white solid (54.0% yield). ESIMS found C14H20CIFN4O2 mlz 352.90 (M+H).

Example 1.

[040] Preparation of fr «5-N-(6-(5-Fluoropyridin-3-yl)isoquinolin-3-yl)-3- mo holinocyclobutane-l-carboxamide (16) is depicted below in Scheme 16.

reflux, 24 h

LIV

16

Scheme 16 Step 1

[041] To a suspension of 6-bromoisoquinolin-3 -amine (XV) (0.89 g, 3.99 mmol), fr «5-3-((teri-butoxycarbonyl)amino)cyclobutane-l-carboxylic acid (LII) (1.03 g, 4.79 mmol), DMAP (0.24 g, 1.99 mmol) and HATU (1.82 g, 4.79 mmol) in MeCN (20 mL) was added DIPEA (2.08 mL, 11.97 mmol). The resulting mixture was heated at 50°C for 16 h. The reaction was concentrated and purified via column chromatography (40 g of silica gel) (CHCl3→l% MeOH/CHCh) to produce a crude product which was then and triturated with diethyl ether to give fert-butyl (fr «5-3-((6-bromoisoquinolin-3-yl)carbamoyl)cyclobutyl)carbama te (LIII) as a white solid (1.18 g, 2.81 mmol, 70.4% yield). ESIMS found for Ci9H 2 2BrN 3 0 3 mlz 420.1 (M+H).

Step 2

[042] A solution of fert-butyl (fra«s-3-((6-bromoisoquinolin-3-yl)carbamoyl) cyclobutyl)carbamate (LIII) (1.18 g, 2.81 mmol) and TFA (2.16 mL, 28.08 mmol) was stirred at room temperature for 2 h. The reaction was concentrated and neutralized with NH3/CHCI3, washed with EtOAc and hexanes to yield fra«s-3-amino-N-(6-bromoisoquinolin-3-yl)cyclobutane-l- carboxamide (LIV) as a beige solid (2.77 g). Used for the next step without further purification. ESIMS found for Ci 4 Hi 4 BrN 3 0 mlz 320.0 (M+H).

Step 3

[043] A mixture of fra«s-3-amino-N-(6-bromoisoquinolin-3-yl)cyclobutane-l- carboxamide (LIV) (699 mg, 2.18 mmol),l-bromo-2-(2-bromoethoxy)ethane (LV) (0.33 mL, 2.62 mmol) and K2CO3 (905.2 mg, 6.55 mmol) in MeCN (10 mL) was heated to reflux for 24 h. An additional 1 eq of K2CO3 and 0.5 eq of l-bromo-2-(2-bromoethoxy)ethane was added and heated at reflux for another 8 h. The reaction mixture was filtered, washed with a mixture of CHCh/MeOH, the filtrates were concentrated and the residue was purified column chromatography (0→20% CHC1 3 /10% 7N NH 3 MeOH in CHCI3) to afford fra«s-N-(6-bromoisoquinolin-3-yl)-3- (piperidin-l-yl)cyclobutane-l-carboxamide (LVI) as a beige solid (620 mg, 1.59 mmol, 72.8% yield). ESIMS found for CuH^BrNsO mlz 390.10 ( 81 BrM+H).

Step 4

[0442] To a stirred solution of afford fra«s-N-(6-bromoisoquinolin-3-yl)-3- (piperidin-l-yl)cyclobutane-l-carboxamide (LVI) (0.08 g, 0.19 mmol) in dioxane (2 mL) was added 1 M aqueous K 3 PO 4 (0.38 mL, 0.38 mmol), 3-fluoro-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyridine (LVII) (42.86 mg, 0.19 mmol), and Pd(dppf)Cl 2 (20 mg, 0.02 mmol). The reaction mixture was purged with argon and heated to 90°C for 16 h. The reaction was then concentrated under vacuum and purified via prep TLC (6% MeOH/CHCh) to produce trans-N-(6- (5-Fluoropyridin-3-yl)isoquinolin-3-yl)-3-mo holinocyclobutane-l -carboxamide 16 as a grey solid (16.0 mg, 0.039 mmol, 20.5% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 2.07 - 2.17 (2 H, m), 2.27 (6 H, br s), 2.91 (1 H, quin, J=7.00 Hz), 3.24 - 3.30 (1 H, m), 3.59 (4 H, br t, J=4.25 Hz), 7.94 (1 H, dd, J=8.78, 1.65 Hz), 8.17 (1 H, d, J=8.78 Hz), 8.28 (1 H, dt, J=10.29, 2.26 Hz), 8.35 (1 H, s), 8.63 (1 H, s), 8.66 (1 H, d, J=2.74 Hz), 8.99 (1 H, s), 9.18 ( 1 H, s), 10.54 (1 H, s); ESIMS found for C23H23FN4O2 mlz 407.2 (M+l).

Example 2.

[044] Preparation of N-(6-(5-(hydroxymethyl)pyridin-3-yl)isoquinolin-3-yl)-l- methylpiperidine-4-carboxamide (31) is depicted below in Scheme 17.

Scheme 17

Step 1

[045] A mixture of l -methylpiperidine-4-carboxylic acid (LVIII) ( 1.07 mL, 16.14 mmol), HATU (7.67 g, 20.17 mmol) and DIPEA (8.2 mL, 47.07 mmol) in MeCN (50 mL) was stirred for 10 min. 6-bromoisoquinolin-3-amine (XV) (3.0 g, 13.45 mmol) and DMAP (0.33 g, 2.69 mmol) was then added and the mixture was heated to 80°C overnight. The solvent was evaporated and the residue purified via column chromatography (80 g of silica gel) (CHC¾→3% MeOH/7 N NH3 in CHCI3) to afford N-(6-bromoisoquinolin-3-yl)-l -methylpiperidine-4-carboxamide (LIX) as a beige solid (3.2 g, 9.19 mmol, 68.3% yield). ESIMS found for Ci 6 Hi 8 BrN 3 0 mlz 347.90 (M+H).

Step 2

[046] To a stirred solution of N-(6-bromoisoquinolin-3-yl)-l-methylpiperidine-4- carboxamide (LIX) (0.08 g, 0.24 mmol) in dioxane (2 mL) was added 1 M aqueous K3PO4 (0.35 mL, 0.35 mmol) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)nicotinaldehy de (LX) (0.07 g, 0.28 mmol). The reaction mixture was purged with argon before adding Pd(PPh3)4 (0.01 g, 0.01 mmol) and then allowed to stir at 90°C for 16 h. The solvent was evaporated and the residue purified via column chromatography (12 g of silica gel) (CHCl3→8% MeOH/7 N NIL in CHCI3) to afford N-[6-(5-formyl-3-pyridyl)-3-isoquinolyl]-l -methyl-piperidine-4-carboxamide (LXI) as a beige solid (40 mg, 0.1 1 mmol, 45.4% yield). ESIMS found for C22H22N4O2 mlz 375.0 (M+H).

Step 3

[047] To a solution of N-[6-(5-formyl-3-pyridyl)-3-isoquinolyl]-l -methyl- piperidine-4-carboxamide (LXI) ( 190 mg, 0.51 mmol) in Me OH (2 mL) was added NaBH 4 ( 19.2 mg, 0.51 mmol) and let stir at room temperature for 2 h. The reaction was quenched with water, concentrated under vacuum and purified via column chromatography (4 g of silica gel) (CHC1 3 →10% MeOH/7 N NH 3 in CHCI 3 ) to afford N-(6-(5-(Hydroxymethyl)pyridin-3-yl) isoquinolin-3-yl)-l-methylpiperidine-4-carboxamide 31 as a white solid ( 10.0 mg, 0.027 mmol, 5.2% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.62 - 1.74 (2 H, m), 1.74 - 1.81 (2 H, m), 1.87 (2 H, td, J=1 1.53, 1.92 Hz), 2.16 (3 H, s), 2.51 - 2.56 ( 1 H, m), 2.81 (2 H, br d, J=1 1.25 Hz), 4.65 (2 H, d, J=5.49 Hz), 5.43 ( 1 H, t, J=5.63 Hz), 7.88 (1 H, dd, J=8.51, 1.65 Hz), 8.13 - 8.19 (2 H, m), 8.25 (1 H, s), 8.58 ( 1 H, s), 8.60 ( 1 H, d, J=1.92 Hz), 8.95 ( 1 H, d, J=2.20 Hz), 9.17 (1 H, s), 10.56 ( 1 H, s); ESIMS found for C22H24N4O2 mlz 377.0 (M+l).

Example 3.

[048] Preparation of N-(6-(5-aminopyridin-3-yl)isoquinolin-3-yl)-4-(piperidin-4- yloxy)benzamide (37) is depicted below in Scheme 18.

Scheme 18

Step 1

[049] A mixture of 4-[(l-fert-butoxycarbonyl-4-piperidyl)oxy]benzoic acid (XXXVIII) (1.56 g, 4.84 mmol), HATU (1.84 g, 4.84 mmol) and DIPEA (1.41 mL, 8.07 mmol) in DMF (12 mL) was stirred at room temperature for 10 min. To the above mixture was added DMAP (0.33 g, 2.69 mmol) and the mixture stirred at room temperature for another 5 min. To the mixture was added 6-bromoisoquinolin-3 -amine (XV) (0.6 g, 2.69 mmol) and the mixture stirred at 80°C for 21 h. The reaction mixture was diluted with EtOAc and washed with 2 N HC1 and saturated NaHCC . The organic layer was washed with brine, dried, filtered and concentrated. The crude product was purified by silica gel chromatography (0→30% EtOAc/hexanes). The fractions containing the product were concentrated and the residue triturated in MeOH. The resulting solid was filtered to afford fert-butyl 4-(4-((6-bromoisoquinolin-3-yl)carbamoyl)phenoxy)piperidine- l- carboxylate (LXII) as a white solid (340 mg, 0.65 mmol, 24.0% yield). ESIMS found for C 26 H28BrN 3 (Wz 526.1 (M+H).

Step 2

[050] A mixture of fert-butyl 4-(4-((6-bromoisoquinolin-3-yl)carbamoyl)phenoxy) piperidine-l-carboxylate (LXII) (100 mg, 0.19 mmol), 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)pyridin-3 -amine (LXIII) (46 mg, 0.21 mmol), Pd(dppf)Cl 2 (10.9 mg, 0.01 mmol) and 1 M aqueous K3PO4 (0.24 mL, 0.47 mmol) in dioxane (5 mL) was purged with nitrogen. The mixture was heated at 90°C under nitrogen for 16 h. The reaction was cooled to room temperature and concentrated. The crude product was purified by flash column chromatography (0→10% MeOH/CHCh) to afford fert-butyl 4-(4-((6-(5-aminopyridin-3-yl)isoquinolin-3-yl)carbamoyl) phenoxy)piperidine-l-carboxylate (LXIV) as a brown oil (27 mg, 0.05 mmol, 26.3% yield). ESIMS found for C 3 iH 33 N 5 0 4 ra/z 540.25 (M+H).

Step 3

[051] To a solution of tert-butyl 4-(4-((6-(5-aminopyridin-3-yl)isoquinolin-3-yl) carbamoyl)phenoxy)piperidine-l-carboxylate (LXIV) (27 mg, 0.05 mmol) in MeOH (2 mL) was added HQ (4 M in dioxane) (0.13 mL, 0.50 mmol). The resulting solution was stirred at 60°C for 2 h and then concentrated. The crude product was purified by silica gel chromatography (0→10% 7 N NH3-MeOH/CHCl3). The fractions containing the product were concentrated and the residue triturated in ether. The resulting solid was filtered to afford N-(6-(5-Aminopyridin-3-yl) isoquinolin-3-yl)-4-(piperidin-4-yloxy) benzamide 37 as a beige solid (10.0 mg, 0.022 mmol, 43.2% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.40 - 1.53 (2 H, m), 1.89 - 2.00 (2 H, m), 2.55 - 2.64 (2 H, m), 2.96 (2 H, dt, J=12.42, 3.81 Hz), 4.50 - 4.59 (1 H, m), 5.48 (2 H, s), 7.06 (2 H, d, J=9.06 Hz), 7.33 (1 H, t, J=2.20 Hz), 7.76 - 7.84 (1 H, m), 8.01 (1 H, d, J=2.47 Hz), 8.07 (2 H, d, J=8.78 Hz), 8.11 - 8.20 (2 H, m), 8.22 (1 H, d, J=1.92 Hz), 8.68 (1 H, s), 9.22 (1 H, s), 10.70 (1 H, s); ESIMS found for CzeHzsNsOz mlz 440.2 (M+l).

Example 4.

[052] Preparation of N-(6-(5-((4-hydroxypiperidin-l-yl)methyl)pyridin-3-yl) isoquinolin-3-yl)cyclopropanecarboxamide (53) is depicted below in Scheme 19.

53

Scheme 19

Step 1

[053] To a stirred suspension of 6-bromoisoquinolin-3 -amine (XV) (2.0 g, 8.97 mmol) and DMAP (0.22 g, 1.79 mmol) in pyridine (30 mL) was added cyclopropanecarbonyl chloride (LXV) (0.98 mL, 10.76 mmol) and the mixture was stirred at room temperature for 2 h. Another 0.5 equivalents of cyclopropanecarbonyl chloride was added and stirring continued at room temperature for another 1 h. The reaction went to completion, solvent was concentrated and the residue taken up in EtOAc and washed with water. The organic/aqueous layer was filtered, washed with EtOAc and the solids were dried under high vacuo to obtain 440 mg of the product. The filtrates were transferred to separating funnel, organic layer separated, washed with brine solution, dried over anhydrous Na2SC>4 and then concentrated to dryness to obtain N-(6- bromo-3-isoquinolyl)cyclopropanecarboxamide (LXVI) as a brown solid ( 1.74 g, 5.98 mmol, 66.7% yield). ESIMS found for Ci 3 HnBrN 2 0 mlz 291.0 (M+H).

Step 2

[054] To a mixture of N-(6-bromo-3-isoquinolyl)cyclopropanecarboxamide (LXVI) (300 mg, 1.03 mmol), 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine-3-ca rbaldehyde (LX) (264.3 mg, 1.13 mmol) and Pd(dppf)Cl2 (42 mg, 0.05 mmol) in dioxane (3 mL) was added 2 M solution of K3PO4 (1.03 mL, 2.06 mmol). N2 gas was bubbled into the mixture for 10 min and then the mixture was heated to 1 10°C under microwave irradiation for 30 min. The organic layer was separated carefully, absorbed on silica gel and purified by flash column chromatography (0→100% hexanes/EtOAc) to obtain N-[6-(5-formyl-3-pyridyl)-3-isoquinolyl] cyclopropanecarboxamide (LXVII) as a beige solid (180 mg, 0.57 mmol, 55.0% yield). ESIMS found for Ci 9 Hi 5 N 3 0 2 mlz 317.9 (M+H).

Step 3

[055] A suspension of N-[6-(5-formyl-3-pyridyl)-3-isoquinolyl] cyclopropanecarboxamide (LXVII) (60 mg, 0.19 mmol) and piperidin-4-ol (LXVIII) (0.07 mL, 0.57 mmol) in Me OH (1 mL) was stirred for 10 min. Then, NaCNBH 3 (17.8 mg, 0.28 mmol) was added and the mixture was stirred at room temperature overnight. The reaction was concentrated to dryness and the residue was taken up in DCM, washed with sat.NaHC0 3 , water and then with brine solution. The organics were then separated and dried (MgSC ) before concentration to dryness. The crude was then purified by preparative TLC (50% of CHC1 3 /10% 7N NH 3 /MeOH in CHC1 3 ). The purified product was suspended in diethyl ether, sonicated and the solids were collected by filtration and dried under high vacuo to obtain N-(6-(5-((4-Hydroxypiperidin-l -yl) methyl)pyridin-3-yl)isoquinolin-3-yl) cyclopropanecarboxamide 53 as an off-white solid (9.0 mg, 0.022 mmol, 1 1.8% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 0.84 (2 H, dt, J=7.89, 2.64 Hz), 0.85 - 0.90 (2 H, m), 1.36 - 1.46 (2 H, m), 1.68 - 1.76 (2 H, m), 2.05 - 2.16 (3 H, m), 2.67 - 2.75 (2 H, m), 3.42 - 3.51 (1 H, m), 3.58 (2 H, s), 4.53 ( 1 H, d, J=4.12 Hz), 7.88 (1 H, dd, J=8.51, 1.92 Hz), 8.12 ( 1 H, t, J=2.06 Hz), 8.16 (1 H, d, J=8.51 Hz), 8.24 (1 H, s), 8.54 (1 H, d, J=1.92 Hz), 8.55 (1 H, s), 8.94 ( 1 H, d, J=2.20 Hz), 9.18 ( 1 H, s), 10.91 (1 H, s); ESIMS found for C24H26N4O2 mlz 403.0 (M+l).

Example 5.

[056] Preparation of N-methyl-5-(3-(2-(4-methylpiperazin-l-yl)isonicotinamido) isoquinolin-6-yl)nicotinamide (59) is depicted below in Scheme 20.

Lxrx

59

Scheme 20

Step 1

[057] To a suspension of 6-bromoisoquinolin-3-amine (XV) (4.0 g, 17.93 mmol), 2- fluoropyridine-4-carboxylic acid (LXIX) (3.04 g, 21.52 mmol), DMAP (2.19 g, 17.93 mmol) and HATU (8.18 g, 21.52 mmol) in DMF (90 mL) was added DIPEA (12.5 mL, 71.73 mmol). The resulting mixture was heated at 80°C for 3 h followed by an additional 1.2 equiv. of HATU and continued heating at 80°C for an additional 15 h. The reaction mixture was poured into water and the resulting solid filtered and washed with water to produce N-(6-bromoisoquinolin-3-yl)-2- fluoroisonicotinamide (LXX) as a brown solid (5.9 g, 17.05 mmol, 95.1% yield). ESIMS found for Ci 5 H 9 BrFN 3 0 mlz 348.0 ( 81 BrM+H).

Step 2

[058] To a suspension of N-(6-bromoisoquinolin-3-yl)-2-fluoroisonicotinamide (LXX) (5.9 g, 17.04 mmol) in MeCN (100 mL) was added 1-methylpiperazine (LXXI) (0.8 mL, 7.22 mmol). The mixture was heated at reflux for 1 day and then cooled to room temperature. The reaction was concentrated and the residue poured into water. The resulting solid was filtered and then re-dissolved in a mixture of MeOH/EtOAc. The solution was concentrated and the residue triturated in ether. The solid was filtered to afford N-(6-bromoisoquinolin-3-yl)-2-(4- methylpiperazin-l-yl)isonicotinamide (LXXII) as a brown solid (6.3 g, 14.78 mmol, 86.7% yield). 'H NMR (499 MHz, DMSO- 6 ) δ ppm 2.23 (3 H, s), 2.42 (4 H, t, J=5.08 Hz), 3.56 - 3.63 (4 H, m), 7.15 (1 H, dd, J=4.94, 1.10 Hz), 7.46 (1 H, s), 7.70 (1 H, dd, J=8.78, 1.92 Hz), 8.06 (1 H, d, J=8.78 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.29 (1 H, d, J=1.37 Hz), 8.61 ( 1 H, s), 9.25 (1 H, s), 1 1.16 (1 H, br s). ESIMS found for C 2 oH 2 oBrN 5 0 mlz 426 A (M+H).

Step 3

[059] A mixture of N-(6-bromoisoquinolin-3-yl)-2-(4-methylpiperazin-l-yl) isonicotinamide (LXXII) (80 mg, 0.19 mmol), N-methyl-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)nicotinamide (LXXIII) (54.1 mg, 0.21 mmol), Pd(dppf)Cl 2 ( 10.7 mg, 0.01 mmol) and 2 M solution of K3PO4 (0.23 mL, 0.47 mmol) in dioxane (3 mL) was purged with nitrogen. The mixture was heated at 90°C under nitrogen for 3 h. The reaction was cooled to room temperature and concentrated. The crude product was purified by flash column chromatography (0→10% 7 N ML-MeOH/CHCh). The fractions containing the product were concentrated and the residue triturated in ether. The resulting solid was filtered and dried to afford N-methyl-5-(3-(2- (4-methylpiperazin-l-yl)isonicotinamido)isoquinolin-6-yl)nic otinamide 59 an off-white solid (73.0 mg, 0.152 mmol, 80.8% yield). 'H NMR (499 MHz, DMSO- 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 2.87 (3 H, d, J=4.39 Hz), 3.57 - 3.65 (4 H, m), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.47 (1 H, s), 8.03 (1 H, dd, J=8.65, 1.78 Hz), 8.26 (1 H, s), 8.27 ( 1 H, d, J=1.92 Hz), 8.45 (1 H, s), 8.64 (1 H, t, J=2.20 Hz), 8.72 - 8.81 (2 H, m), 9.04 (1 H, d, J=1.92 Hz), 9.23 (1 H, d, J=1.92 Hz), 9.30 (1 H, s), 1 1.14 (1 H, s); ESIMS found for C27H27N7O2 mlz 482.2 (M+l).

Example 6.

[060] Preparation of 2-morpholino-N-(6-(pyridazin-3 -yl)isoquinolin-3 -yl)acetamide (66) and 2-mo holino-N-(6-(pyridazin-4-yl)isoquinolin-3-yl)acetamide (67) are depicted below in Scheme 21.

66

Scheme 21

Steps 1

[061] To a suspension of 6-bromoisoquinolin-3-amine (XV) (10 g, 44.83 mmol) and

2- chloroacetic anhydride (9.2 g, 53.79 mmol) in dry THF (100 mL) was added TEA (15.62 mL, 1 12.07 mmol). The mixture was stirred at room temperature for 16 h then poured into a mixture of aqueous saturated NaHCC (200 mL) and water (300 mL). The mixture was stirred for an hour and the resulting solids were collected by filtration, air-dried and suspended in diethyl ether (400 mL), stirred for 30 min. The solid filtered and dried under high vacuo to obtain N-(6-bromoisoquinolin-

3- yl)-2-chloroacetamide (LXXIV) as a brown solid (9.36 g, 31.25 mmol, 69.7% yield). ESIMS found for CnH 8 BrClN 2 0 mlz 298.95 (M+l).

Step 2

[062] A mixture of N-(6-bromoisoquinolin-3-yl)-2-chloroacetamide (LXXIV) (9.28 g, 30.98 mmol), morpholine (LXXV) (8.04 mL, 92.94 mmol), and DIPEA ( 10.79 mL, 61.96 mmol) in MeCN ( 100 mL) was heated to 90°C for 16 h. The reaction mixture was filtered and the solid was dried under high vacuo to obtain 8.45 grams of the product. The filtrate was concentrated, absorbed on silica and purified by column chromatography (25→ 100% hexanes/EtOAc) to obtain another 1.75 g of N-(6-bromoisoquinolin-3-yl)-2-mo holinoacetamide (LXXVI) as a brown solid ( 10.2 g, 28.05 mmol, 90.5% yield). ESIMS found for Ci 5 Hi 6 BrN 3 0 2 mlz 350.1 (M+l). Step 3

[063] To a flask was added KOAc (0.01 mL, 0.22 mmol), N-(6-bromoisoquinolin- 3-yl)-2-morpholinoacetamide (LXXVI) (70 mg, 0.20 mmol), Pd(PPh 3 ) 4 (0.02 g, 0.02 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (0.07 g, 0.20 mmol), and dioxane (2 mL). Reaction mixture was allowed to stir at 90°C for 5 h. The reaction was filtered reaction through short pad of Celite and rinsed with minimal amount of dioxane (2 mL). To the filtrate was added 3-chloropyridazine hydrochloride (XLXXVII) (30.2 mg, 0.20 mmol), 2 M solution of K 3 P0 4 (0.02 mL, 0.20 mmol), and Pd(PPh 3 ) 4 (0.02 g, 0.02 mmol) and let stir at 90°C for 16 h. The reaction was evaporated and the residue purified via column chromatography (12 g of silica gel) (CHC1 3 →3% MeOH/7 N NH 3 in CHCI3) to yield 2-mo holino-N-(6-(pyridazin-3-yl)isoquinolin-3-yl)acetamide 66 as a white solid (5.0 mg, 0.014 mmol, 7.2% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 2.56 - 2.63 (4 H, m), 3.27 (2 H, s), 3.63 - 3.69 (4 H, m), 8.05 (1 H, dd, J=8.65, 1.78 Hz), 8.21 (1 H, dd, J=5.35, 2.61 Hz), 8.25 (1 H, d, J=8.78 Hz), 8.59 (1 H, s), 8.63 (1 H, s), 9.24 (1 H, s), 9.37 (1 H, dd, J=5.21, 1.10 Hz), 9.83 (1 H, dd, J=2.47, 1.10 Hz), 10.16 (1 H, s); ESIMS found for Ci9Hi 9 N 5 0 2 mlz 350.2 (M+l).

Step 4

[064] To a solution of N-(6-bromoisoquinolin-3-yl)-2-moφholinoacetamide (LXXVI) (60 mg, 0.17 mmol) in p-xylene (3 mL) was added 4-(tributylstannyl)pyridazine (LXXVIII) (189.7 mg, 0.51 mmol) and S-Phos Pd G4 (13.6 mg, 0.02 mmol). Reaction mixture was heated to 90°C for 16 h. The reaction was concentrated and the residue purified via column chromatography (12 g of silica gel). (100% CHC1 3 →3% MeOH/7 N NH 3 in CHC1 3 ). The product was further purified by prep TLC, eluting with (4% MeOH/7 N NH 3 in CHC1 3 ) to produce 2- mo holino-N-(6-(pyridazin-4-yl)isoquinolin-3-yl)acetamide 67 as a beige solid (4.0 mg, 0.011 mmol, 6.7% yield). ¾ NMR (499 MHz, CHLOROFORM- ) δ ppm 2.63 - 2.73 (4 H, m), 3.26 (2 H, s), 3.81 - 3.89 (4 H, m), 7.64 (1 H, dd, J=8.64, 4.80 Hz), 8.05 (1 H, dd, J=8.65, 1.23 Hz), 8.09 (1 H, d, J=8.51 Hz), 8.37 (1 H, dd, J=8.64, 1.51 Hz), 8.44 (1 H, s), 8.73 (1 H, s), 9.09 (1 H, s), 9.26 (1 H, dd, J=4.80, 1.23 Hz), 9.71 (1 H, br s); ESIMS found for Ci9Hi 9 N 5 0 2 mlz 350.1 (M+l).

Example 7.

[065] Preparation of fr «5-4-(dimethylamino)-N-(6-(pyrazin-2-yl)isoquinolin-3-yl) cyclohexane-l-carboxamide (70) is depicted below in Scheme 22.

70

Scheme 22

Step 1

[066] A mixture of fr «5-4-((teri-butoxycarbonyl)amino)cyclohexane-l-carboxylic acid (LXXIX) (1.15 mL, 11.25 mmol), HATU (4.28 g, 11.25 mmol), and DIPEA (4.7 mL, 27 mmol) in DMF (40 mL) was stirred for 10 min before adding 6-bromoisoquinolin-3 -amine (XV) (2.01 g, 9 mmol) and DMAP (220 mg, 1.8 mmol). The reaction was stirred at 80°C overnight. The solvents were concentrated, the residue was taken up in EtOAc and the organics washed with aqueous saturated NaHCC , water and then with brine solution. The organics were then separated and dried (MgSO- before concentrating to dryness to obtain fert-butyl (fr «5-4-((6- bromoisoquinolin-3-yl)carbamoyl)cyclohexyl)carbamate (LXXX) as a beige solid (2.75 g, 6.13 mmol, 68.2% yield) which was used for next step as it is. ESIMS found for C2iH2eBrN303 mlz 448.1 (M+H).

Step 2

[067] To a solution of fert-butyl (fra«s-4-((6-bromoisoquinolin-3-yl)carbamoyl) cyclohexyl)carbamate (LXXX) (5.0 g, 11.15 mmol) in DCM (20 mL) was added TFA (10 mL, 129.8 mmol) and the mixture was stirred at room temperature at 1 h. The solvent was concentrated and the residue treated with 7N NH 3 /MeOH. The crude product was purified by column chromatography (25→100% CHC1 3 /10% 7 N NH 3 /MeOH in CHC1 3 ). The pure fractions were combined, concentrated, the residue suspended in EtOAc, sonicated and the solids were collected by filtration, washed with diethyl ether and dried under high vacuo to obtain fra«s-4-amino-N-(6- bromoisoquinolin-3-yl)cyclohexane-l-carboxamide (LXXXI) as a beige solid (3 g, 8.61 mmol, 77.2% yield). ESIMS found for Ci 6 Hi 8 BrN 3 0 mlz 348.1 (M+H).

Step 3

[068] To a stirred solution of fra«s-4-amino-N-(6-bromoisoquinolin-3-yl) cyclohexane-l-carboxamide (LXXXI) (3.0 g, 8.61 mmol) in MeOH (50 mL) was added formaldehyde (8.64 mL, 42.93 mmol). After 15 min of stirring, Na(OAc) 3 BH (9.1 g, 42.93 mmol) was added and the mixture was stirred at room temperature for 2 h . The solvent was removed in vacuo and the residue taken in water, basified with 1 N NaOH solution and extracted with CHC1 3 . The organic layer was separated, washed with water and brine, dried over anhydrous Na2SC>4 and concentrated. The crude was suspended in diethyl ether, sonicated and the solid was collected by filtration and dried under high vacuo to obtain the desired product fra«s-N-(6-bromoisoquinolin-3- yl)-4-(dimethylamino)cyclohexane-l-carboxamide (LXXXII) as a beige solid (2.28 g, 6.06 mmol, 70.3% yield). ESIMS found for Ci 8 H 22 BrN 3 0 mlz 376.1 (M+H).

Step 4

[069] A mixture of 2-(tributylstannyl)pyrazine (LXXXIII) (215.8 mg, 0.58 mmol), Cul (10 mg, 0.05 mmol), Pd(PPh3) 4 (61.4 mg, 0.05 mmol) and trans-N-(6- bromoisoquinolin-3-yl)-4-(dimethylamino) cyclohexane-l-carboxamide (LXXXII) (200 mg, 0.53 mmol) was taken in dioxane (2 mL). N 2 was bubbled into the mixture for 10 min and then heated to 90°C overnight. The reaction mixture was absorbed on silica gel and the crude was then purified by flash column chromatography (0→80% CHC1 3 /10% 7 N NH 3 /MeOH in CHC1 3 ). The pure fractions were combined, concentrated, the product was suspended in EtOAc, sonicated and the solids were collected by filtration, dried under high vacuo to obtain fra«s-4-(dimethylamino)-N-(6- (pyrazin-2-yl)isoquinolin-3-yl)cyclohexane-l-carboxamide 70 as an off-white solid (98.0 mg, 0.248 mmol, 46.7% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.14 - 1.25 (2 H, m), 1.49 (2 H, qd, J=12.72, 3.02 Hz), 1.84 - 1.90 (2 H, m), 1.90 - 1.97 (2 H, m), 2.11 - 2.17 (1 H, m), 2.18 (6 H, s), 2.45 - 2.49 (1 H, m), 8.16 - 8.22 (1 H, m), 8.23 - 8.29 (1 H, m), 8.61 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.33, 1.51 Hz), 9.20 (1 H, s), 9.47 (1 H, d, J=1.37 Hz), 10.55 (1 H, s); ESIMS found for C22H 25 N 5 0 mlz 376.2 (M+l).

Example 8.

[070] Preparation of fr «5-4-((3-fluoroazetidin-l-yl)methyl)-N-(6-(pyrazin-2-yl) isoquinolin-3-yl)cyclohexane-l-carboxamide (72) is depicted below in Scheme 23.

LXXXIV

XV DMF, 70°C, ON

then THF/DMSO, TEA at r.t., 1 h

dioxane, 90°C, ON

LXXXIX 72

Scheme 23

Step 1

[071] A mixture of DIPEA (5.86 mL, 33.62 mmol), DMAP (0.27 g, 2.24 mmol) and HATU (5.11 g, 13.45 mmol) in DMF (30 mL) was stirred for lOmin before adding 6- bromoisoquinolin-3-amine (XV) (2.5 g, 11.21 mmol) followed by the addition of trans -4-(((tert- butyldimethylsilyl)oxy)methyl) cyclohexane-l-carboxylic acid (LXXXIV) (3.66 g, 13.45 mmol). The reaction was heated at 70°C overnight. An additional 0.5 eq. of HATU was added and the mixture was heated at 70°C for an additional 6 h. The solvent was concentrated and the residue taken up in EtOAc, washed with saturated aqueous NaHCC and brine solution. The organic layer was then concentrated and the crude was purified column chromatography (0→30% EtOAc/hexanes). The pure fractions were combine and concentrated to obtain trans- ~ N-(6- bromoisoquinolin-3 -yl)-4-(((teri-butyldimethylsilyl)oxy)methyl)cyclohexane- 1 -carboxamide (LXXXV) as a crystalline off-white solid (2.25 g,4.71 mmol, 42.0 % yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.87 (9 H, s), 0.98 (2 H, qd, J=12.72, 3.29 Hz), 1.37 - 1.50 (3 H, m), 1.74 - 1.82 (2 H, m), 1.85 - 1.92 (2 H, m), 2.46 - 2.54 ( 1 H, m), 3.41 (2 H, d, J=6.04 Hz), 7.62 ( 1 H, dd, J=8.64, 1.78 Hz), 7.99 (1 H, d, J=8.78 Hz), 8.16 ( 1 H, d, J=1.65 Hz), 8.44 (1 H, s), 9.13 (1 H, s), 10.53 ( 1 H, s); ESIMS found for C 2 3H 33 BrN 2 02Si mlz Ml 2 (M+H).

Step 2

[072] To a solution of fr «5-N-(6-bromoisoquinolin-3-yl)-4-(((tert- butyldimethylsilyl)oxy)methyl)cyclohexane-l -carboxamide (LXXXV) (2.24 g, 4.69 mmol) in THF (10 mL) was added 1 M solution (in THF) of TBAF (7.04 mL, 7.04 mmol) and stirred at room temperature overnight. Water was added to the reaction mixture and extracted with EtOAc (2x). The organic layer was separated, washed with brine, dried over anhydrous Na 2 S04, and concentrated. The crude product was suspended in EtOAc, sonicated and the solids were collected by filtration and dried under high vacuo to obtain fr «5-N-(6-bromoisoquinolin-3-yl)-4- (hydroxymethyl)cyclohexane-l -carboxamide (LXXXVI) as a light beige solid ( 1.44 g, 3.96 mmol, 84.3% yield). ESIMS found for Ci 7 Hi 9 BrN 2 0 2 mlz 363.1 (M+H).

Step 3

[073] To a solution of DMSO (0.59 mL, 8.26 mmol) in DCM (3 mL) at -78°C was added dropwise under Argon, oxalyl dichloride (0.36 mL, 4.13 mmol) in DCM (1 mL). After 15 min, fr «5-N-(6-bromoisoquinolin-3-yl)-4-(hydroxymethyl)cyclohexane -l-carboxamide (LXXXVI) ( 1.0 g, 2.7 5mmol) in a mixture of THF (12 mL) and DMSO (0.5 mL) (compound is not soluble in DCM or THF) was added and the mixture was stirred at -78°C for 1 h. Then, TEA ( 1.15 mL, 8.26 mmol) was added and the mixture was continued to stir for 1 h and at room temperature for 1 h. The reaction mixture was diluted with water and DCM, organic layer separated, washed with brine, dried over anhydrous Na2S04 and the solvents were concentrated in vacuo to obtain fr «5-N-(6-bromoisoquinolin-3-yl)-4-formylcyclohexane-l -carboxamide (LXXXVII) as a white solid (215 mg, 0.59 mmol, 108.1% yield) which was used for next step without purification. ESIMS found for Ci 7 Hi 7 BrN 2 0 2 mlz 361. (M+H). Step 4

[074] A mixture of 3-fluoroazetidine (LXXXVIII) (0.23 mL, 2.03 mmol), trans-Ή- (6-bromoisoquinolin-3-yl)-4-formylcyclohexane-l -carboxamide (LXXXVII) (490 mg, 1.36 mmol) in DCE (6 mL) was stirred for 20 min. Na(OAc) 3 BH (431 mg, 2.03 mmol) was then added and the mixture was stirred overnight at room temperature. The reaction mixture was diluted with DCM, washed with saturated NaHCC , water and brine. The organic layer was separated and dried (MgSO- before concentration to dryness to obtain fra«s-N-(6-bromoisoquinolin-3-yl)-4-((3- fluoroazetidin-l -yl)methyl)cyclohexane-l -carboxamide (LXXXIX) as an off-white solid (595 mg, 1.42 mmol, 104.4% yield) which was used for next step without purification. ESIMS found for C2oH 2 3BrFN 3 O ra/z 419.1 (M+H).

Step 5

[0443] To a mixture of 2-(tributylstannyl)pyrazine (LXXXIII) (96.6 mg, 0.26 mmol), Cul (4.3 mg, 0.02 mmol), Pd(PPli3)4 (27.5 mg, 0.02 mmol) and fr «5-N-(6-bromoisoquinolin-3- yl)-4-((3-fluoroazetidin-l -yl)methyl)cyclohexane-l -carboxamide (LXXXIX) ( 100 mg, 0.24 mmol) in dioxane (1 mL) was bubbled N2 for 10 min and then heated at 90°C overnight. The reaction mixture was absorbed on silica gel and the crude was then purified by flash column chromatography (0→50% CHC1 3 /10% 7 N NLL/MeOH in CHC1 3 ). The purified product was suspended in diethyl ether, sonicated and the solid was collected by filtration, dried under high vacuo to obtain fr «5-4-((3-Fluoroazetidin-l -yl)methyl)-N-(6-(pyrazin-2-yl)isoquinolin-3-yl) cyclohexane-l -carboxamide 72 as a white solid (50.0 mg, 0.1 13 mmol, 47.6% yield). ¾ NMR (499 MHz, DMSO- 6 ) 5 ppm 0.91 (2 H, qd, J=12.76, 3.16 Hz), 1.22 - 1.34 ( 1 H, m), 1.39 - 1.52 (2 H, m), 1.80 (2 H, br dd, J=12.90, 2.74 Hz), 1.87 (2 H, br d, J=10.70 Hz), 2.29 (2 H, d, J=6.59 Hz), 2.52 - 2.55 (1 H, m), 2.96 - 3.08 (2 H, m), 3.48 - 3.60 (2 H, m), 5.12 ( 1 H, dquin, J=58.00, 5.00 Hz), 8.15 - 8.22 (1 H, m), 8.24 - 8.30 ( 1 H, m), 8.61 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 ( 1 H, dd, J=2.33, 1.51 Hz), 9.19 (1 H, s), 9.47 ( 1 H, d, J=1.37 Hz), 10.53 ( 1 H, s); ESIMS found for C 24 H 26 FN 5 O mlz 420.2 (M+l).

Example 9.

[075] Preparation of N-(6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)-l- (piperidin-4-yl)-lH-l,2,3-triazole-4-carboxamide (94) and l-(l -(2-fluoroethyl)piperidin-4-yl)-N- (6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)-lH-l,2, 3-triazole-4-carboxamide (95) are depicted below in Scheme 24.

95

Scheme 24

Step 1-2

[076] A mixture of 6-bromoisoquinolin-3 -amine (XV) (1.0 g, 4.48 mmol), 4,4,5,5- tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- l,3,2-dioxaborolane (1.25 g, 4.93 mmol), Pd(dppf)Ci2 (0.26 g, 0.31 mmol) and KOAc (1.1 g, 11.21 mmol) in dioxane (30 mL) purged with N2 and heated at 90°C under N2 for 5 h. The reaction was cooled to room temperature before adding 6-chloro-N-isopropyl-pyrazin-2-amine (XCI) (0.8 5g, 4.9 3mmol) followed by Pd(dppf)Cl 2 (0.26 g, 0.31 mmol) and 2 M solution of K 3 P0 4 (5.6 mL, 11.21 mmol). The mixture was purged again with N2 and heated at 90°C under N2 for 18 h. The reaction was cooled to room temperature and concentrated. The crude product was purified by silica gel chromatography (0— > 15 % MeOH/CHCh) . The fractions containing the product were concentrated and the residue triturated in a mixture of ether/hexanes (1/3). The resulting solid was filtered and dried to afford 6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin-3-amine (XCII) as a lime green solid (978 mg, 3.50 mmol, 78.1% yield). ESIMS found for Ci 6 Hi 7 N 5 mlz 280.1 (M+H). Step 3

[077] To a suspension of 6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin-3-amine (XCII) (0.2 g, 0.72 mmol), l-(l-(tert-butoxycarbonyl)piperidin-4-yl)-lH-l,2,3-triazole- 4- carboxylic acid (XLIII) (0.23 g, 0.79 mmol), DMAP (0.04 g, 0.36 mmol) and HATU (0.33 g, 0.86 mmol) in DMF (4 mL) was added DIPEA (0.37 mL, 2.15 mmol). The resulting mixture was stirred 80°C for 1 h. An additional 1 equiv. of HATU was added and the reaction mixture stirred at 80°C for additional 1 h. The mixture was poured into water and the resulting solid filtered and dried to afford fert-butyl 4-[4-[[6-[6-(isopropylamino) pyrazin-2-yl]-3- isoquinolyl]carbamoyl]triazol-l-yl]piperidine-l-carboxylate (XCIII) as a brown solid (380 mg, 0.681 mmol, 95.2% yield). ESIMS found for C 2 9H 35 N 9 0 3 mlz 558.3 (M+H).

Step 4

[078] To a suspension of fert-butyl 4-[4-[[6-[6-(isopropylamino)pyrazin-2-yl]-3- isoquinolyl]carbamoyl]triazol-l-yl]piperidine-l-carboxylate (XCIII) (0.39 g, 0.70 mmol) in MeOH (5 mL) was added hydrogen chloride (4 M in dioxane) (0.87 mL, 3.5 mmol). The resulting solution was stirred at 60°C for 30 min and then the reaction mixture was concentrated under high vacuum. The crude product was purified by silica gel chromatography (0→10% 7 N N¾- MeOH/CHCh). The fractions containing the product were concentrated and triturated in ether. The resulting solid was filtered and dried to afford N-(6-(6-(Isopropylamino)pyrazin-2- yl)isoquinolin-3-yl)-l-(piperidin-4-yl)-lH-l,2,3-triazole-4- carboxamide 94 as a beige solid (176.0 mg, 0.385 mmol, 55.0% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.26 (6 H, d, J=6.31 Hz), 1.89 (2 H, qd, J=11.94, 3.98 Hz), 2.08 (2 H, br dd, J=11.80, 2.20 Hz), 2.58 - 2.72 (2 H, m), 3.02 - 3.12 (2 H, m), 4.21 (1 H, dq, J=13.34, 6.53 Hz), 4.66 (1 H, tt, J=11.66, 4.12 Hz), 7.13 (1 H, d, J=7.14 Hz), 7.94 (1 H, s), 8.14 - 8.21 (1 H, m), 8.21 - 8.30 (1 H, m), 8.48 (1 H, s), 8.62 (1 H, s), 8.65 (1 H, s), 8.96 (1 H, s), 9.23 (1 H, s), 10.12 (1 H, br s); ESIMS found for C 24 H 27 N 9 O mlz 458.3 (M+l).

Step 5

[079] A suspension of N-[6-[6-(isopropylamino)pyrazin-2-yl]-3-isoquinolyl]-l-(4- piperidyl)triazole-4-carboxamide 94 (70 mg, 0.15 mmol), DIPEA (0.08 mL, 0.46 mmol) and 1- fluoro-2-iodoethane (XCIV) (0.02 mL, 0.31 mmol) in MeCN (3 mL) was heated in a sealed tube at 110°C with microwave irradiation for 30 min. The mixture was concentrated and the crude product purified by column chromatography (0→5% 7 N NHs-MeOH/CHCh). The fractions containing the product were concentrated and the residue triturated in ether. The resulting solid was filtered and dried to afford l -( l-(2-Fluoroethyl)piperidin-4-yl)-N-(6-(6-(isopropylamino)pyr azin- 2-yl) isoquinolin-3-yl)-lH- l,2,3-triazole-4-carboxamide 95 as a beige solid (60.0 mg, 0.1 19 mmol, 77.9% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.26 (6 H, d, J=6.31 Hz), 2.02 - 2.19 (4 H, m), 2.25 - 2.33 (2 H, m), 2.69 (2 H, dt, J=28.60, 5.00 Hz), 3.02 (2 H, br d, J=l 1.80 Hz), 4.21 ( 1 H, dq, J=13.34, 6.53 Hz), 4.48 - 4.67 (3 H, m), 7.13 ( 1 H, d, J=7.14 Hz), 7.93 (1 H, s), 8.14 - 8.21 ( 1 H, m), 8.21 - 8.28 (1 H, m), 8.48 ( 1 H, s), 8.62 ( 1 H, s), 8.65 ( 1 H, s), 9.00 (1 H, s), 9.23 (1 H, s), 10.1 1 ( 1 H, s); ESIMS found for C 2 6H 3 oFN 9 0 mlz 504.3 (M+l).

Example 10.

[080] Preparation of N-(6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin-3-yl) piperidine-4-carboxamide (87), 1 -(2-fluoroethyl)-N-(6-(6-(isopropylamino)pyrazin-2-yl) isoquinolin-3-yl)piperidine-4-carboxamide (89), and l-isopropyl-N-(6-(6-(isopropylamino) pyrazin-2 heme 25.

90

Scheme 25 Step 1

[081] To a mixture of 6-[6-(isopropylamino)pyrazin-2-yl]isoquinolin-3-amine (XCII) (0.4 g, 1.43 mmol), l-fert-butoxycarbonylpiperidine-4-carboxylic acid (XCV) (0.39 g, 1.72 mmol), DMAP (0.09 g, 0.72 mmol) and HATU (0.65 g, 1.72 mmol) in DMF (8 mL) was added DIPEA (0.75 mL, 4.3 mmol). The resulting mixture was stirred at 80°C for 5 h. The reaction mixture was diluted with water and the resulting solid filtered, washed with water and dried to produce fert-butyl 4-[[6-[6-(isopropylamino) pyrazin-2-yl]-3-isoquinolyl]carbamoyl]piperidine-l- carboxylate (XCVI) as a brown solid (0.65 g, 1.32 mmol, 92.5% yield). ESIMS found for C 2 7H 3 4N 6 0 3 mlz 491.3 (M+H).

Step 2

[082] To a suspension of fert-butyl 4-[[6-[6-(isopropylamino)pyrazin-2-yl]-3- isoquinolyl]carbamoyl]piperidine-l -carboxylate (XCVI) (0.65 g, 1.32 mmol) in MeOH (8 mL) was added hydrogen chloride (4 M in dioxane) (1.66 mL, 6.62 mmol). The resulting solution was stirred at 60°C for 30 min and then concentrated under vacuum. The crude product was purified by silica gel chromatography (0→10% 7 N NLL-MeOH/CHCh). The fractions containing the product were concentrated and the residue triturated in ether. The resulting solid was filtered and dried to afford N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)piperi dine-4-carboxamide 87 as a beige solid (340.0 mg, 0.871 mmol, 65.7% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.25 (6 H, d, J=6.31 Hz), 1.54 (2 H, qd, J=12.12, 3.98 Hz), 1.67 - 1.76 (2 H, m), 2.06 (1 H, br s), 2.43 - 2.49 (2 H, m), 2.59 - 2.70 (1 H, m), 2.98 (2 H, br d, J=12.62 Hz), 4.20 (1 H, dq, J=13.28, 6.55 Hz), 7.11 (1 H, d, J=7.41 Hz), 7.92 (1 H, s), 8.07 - 8.14 (1 H, m), 8.14 - 8.20 (1 H, m), 8.43 (1 H, s), 8.52 (1 H, s), 8.56 (1 H, s), 9.14 (1 H, s), 10.48 (1 H, s); ESIMS found for C22H 2 6N 6 0 mlz 391.2 (M+l).

Step 3

[083] A suspension of N-[6-[6-(isopropylamino)pyrazin-2-yl]-3-isoquinolyl] piperidine-4-carboxamide 87 (100 mg, 0.26 mmol), DIPEA (0.1 3mL, 0.77 mmol) and l-fluoro-2- iodoethane (XCIV) (0.04 mL, 0.51 mmol) in MeCN (3 mL) was heated in a sealed tube at 110°C with microwave irradiation for 30 min. The mixture was concentrated and the crude product purified by column chromatography (0→8% 7 N ML-MeOH/CHCh). The fractions containing the product were concentrated and the residue triturated in ether. The resulting solid was filtered and dried to afford l-(2-Fluoroethyl)-N-(6-(6-(isopropylamino)pyrazin-2-yl)isoqu inolin-3-yl) piperidine-4-carboxamide 89 as a beige solid (50.0 mg, 0.115 mmol, 44.7% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.25 (6 H, d, J=6.31 Hz), 1.69 (2 H, qd, J=12.21, 3.98 Hz), 1.77 - 1.85 (2 H, m), 2.05 (2 H, td, J=11.66, 1.92 Hz), 2.52 - 2.57 ( 1 H, m), 2.62 (2 H, dt, J=28.40, 5.00 Hz), 2.91 - 3.00 (2 H, m), 4.20 ( 1 H, dq, J=13.28, 6.55 Hz), 4.54 (2 H, dt, J=48.10, 5.00 Hz), 7.1 1 (1 H, d, J=7.41 Hz), 7.92 ( 1 H, s), 8.08 - 8.14 ( 1 H, m), 8.14 - 8.21 ( 1 H, m), 8.43 (1 H, s), 8.52 (1 H, s), 8.57 ( 1 H, s), 9.15 (1 H, s), 10.55 ( 1 H, s); ESIMS found for C 24 H 29 FN 6 O mlz 437.2 (M+l).

Step 4

[084] A mixture of N-[6-[6-(isopropylamino)pyrazin-2-yl]-3-isoquinolyl] piperidine-4-carboxamide 87 ( 100 mg, 0.26 mmol), HOAc (0.04 mL, 0.77 mmol) and NaCNBH 3 (48.3 mg, 0.77 mmol) in a mixture of acetone (3 mL) and MeOH (3 mL) was stirred at 50°C for 1 h. The mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by chromatography (0→10% 7 N NHs-MeOH/CHCh). The fractions containing the product were concentrated and the residue triturated in ether. The resulting solid was filtered and dried to give l-Isopropyl-N-(6-(6-(isopropylamino) pyrazin-2-yl)isoquinolin-3-yl) piperidine-4- carboxamide 90 as a light yellow solid (94.0 mg, 0.217 mmol, 84.9% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.97 (6 H, d, J=6.59 Hz), 1.25 (6 H, d, J=6.31 Hz), 1.64 (2 H, qd, J=12.17, 3.29 Hz), 1.79 (2 H, br d, J=1 1.80 Hz), 2.05 - 2.17 (2 H, m), 2.51 - 2.57 (1 H, m), 2.61 - 2.74 ( 1 H, m), 2.80 - 2.88 (2 H, m), 4.15 - 4.26 (1 H, m), 7.1 1 (1 H, br d, J=7.14 Hz), 7.92 ( 1 H, s), 8.06 - 8.13 ( 1 H, m), 8.13 - 8.21 ( 1 H, m), 8.43 (1 H, s), 8.52 ( 1 H, s), 8.57 (1 H, s), 9.15 (1 H, s), 10.52 ( 1 H, s); ESIMS found for CzsH^NeO mlz 433.3 (M+l).

Example 11.

[085] Preparation of 4-fluoro-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl) isoquinolin-3-yl)benzamide (110) and 4-fluoro-N-(6-(6-((l -methylpiperidin-4-yl)amino)pyrazin- 2-yl)isoquinolin-3-yl)benzamide (115) are depicted below in Scheme 26.

xcvn

Scheme 26

Step 1

[086] To a suspension of 6-bromoisoquinolin-3 -amine (XV) (2.0 g, 8.97 mmol), 4- fluorobenzoic acid (XCVII) (1.51 g, 10.76 mmol), DMAP (1.1 g, 8.97 mmol) and HATU (4.09 g, 10.76 mmol) in DMF (30 mL) was added DIPEA (6.25 mL, 35.86 mmol). The resulting mixture was heated at 80°C for 2 h. An additional 1.2 eq. of HATU was added and the mixture stirred at 80°C for additional 15 h. The reaction mixture was poured into water (250 mL) and the resulting solid filtered, washed with water and dried to produce N-(6-bromo-3-isoquinolyl)-4-fluoro- benzamide (XCVIII) as a brown solid (3.0 g, 8.69 mmol, 96.9% yield). ESIMS found for Ci 6 HioBrFN 2 O ra/z 345.0 (M+H).

Step 2-3

[087] A mixture of N-(6-bromo-3-isoquinolyl)-4-fluoro-benzamide (XCVIII) (150 mg, 0.43 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (122 mg, 0.48 mmol), Pd(dppf)Cl2 (35.5 mg, 0.04 mmol) and KOAc (106.5 mg, 1.09 mmol) in dioxane (5 mL) was purged with N2 for 3 min. The reaction mixture was sealed and heated at 90°C for 3 h. The reaction was cooled to room temperature and to it was added fert-butyl 4-((6-chloropyrazin-2- yl)amino)piperidine-l-carboxylate (XLIX) (149.5 mg, 0.48 mmol) followed by 4,4,5,5- tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- l,3,2-dioxaborolane (121.4 mg, 0.48 mmol) and K3PO4 (0.54 mL, 1.09 mmol). The mixture was purged with N2 for a min, sealed and heated again at 90°C for 16 h. The mixture was concentrated and the crude product purified by silica gel chromatography (0→100% EtOAc/Hexanes) to afford fert-butyl 4-((6-(3-(4- fluorobenzamido)isoquinolin-6-yl)pyrazin-2-yl)amino)piperidi ne-l-carboxylate (C) as a brown solid (95 mg, 0.18 mmol, 40.3% yield). ESIMS found for C 3 oH 3 iFN 6 0 3 mlz 543.0 (M+H).

Step 4

[088] To a suspension of fert-butyl 4-((6-(3-(4-fluorobenzamido)isoquinolin-6-yl) pyrazin-2-yl)amino)piperidine-l-carboxylate (C) (95 mg, 0.18 mmol) in MeOH (3 mL) was added HCl (4 M in dioxane) (0.44 mL, 1.75 mmol). The mixture was stirred at 60°C for 1 h and concentrated under vacuum. The crude product was purified by silica gel chromatography (0→ 10% 7 N NLL-MeOH/CHCh). The fractions containing the product were concentrated to afford 4- fluoro-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl)isoquinolin -3-yl) benzamide 110 as an off-white solid (31.0 mg, 0.070 mmol, 40.0% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.33 - 1.49 (2 H, m), 1.92 - 2.03 (2 H, m), 2.64 - 2.75 (2 H, m), 3.00 - 3.09 (2 H, m), 3.93 - 4.05 (1 H, m), 7.20 (1 H, d, J=7.14 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.96 (1 H, s), 8.12 - 8.25 (4 H, m), 8.47 (1 H, s), 8.59 (1 H, s), 8.70 (1 H, s), 9.24 (1 H, s), 10.98 (1 H, s); ESIMS found for C25H23FN6O mlz 443.2 (M+l).

Step 5

[089] A solution of 4-fluoro-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl) isoquinolin-3-yl) benzamide 110 (45 mg, 0.10 mmol), formaldehyde (0.08 mL, 1.02 mmol) and HO Ac (0.02 mL, 0.31 mmol) in MeOH (2 mL) was stirred at room temperature for 5 min. To the mixture was added with NaBH(OAc)3 (64.7 mg, 0.31 mmol) and the mixture stirred at 65°C for 3 h. The solvent was removed and the residue was purified by chromatography (0→15% 7N NH3- MeOH/CHCh). The fractions containing the product were concentrated under vacuum. The residue was triturated in ether and the solid filtered and dried to give 4-fluoro-N-(6-(6-(( 1 -methylpiperidin- 4-yl)amino)pyrazin-2-yl)isoquinolin-3-yl)benzamide 115 as an off-white solid (33.0 mg, 0.069 mmol, 67.5% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 1.47 - 1.63 (2 H, m), 1.95 - 2.04 (2 H, m), 2.07 - 2.15 (2 H, m), 2.21 (3 H, s), 2.72 - 2.82 (2 H, m), 3.81 - 3.93 (1 H, m), 7.13 (1 H, br d, J=7.14 Hz), 7.36 (2 H, t, J=8.92 Hz), 7.97 (1 H, s), 8.11 - 8.24 (4 H, m), 8.47 (1 H, s), 8.58 (1 H, s), 8.69 (1 H, s), 9.24 (1 H, s), 10.93 (1 H, s); ESIMS found for CzeHzsFNeO mlz 457.0 (M+l).

Example 12.

[090] Preparation of N-(6-(6-(3-aminoazetidin-l-yl)pyrazin-2-yl)isoquinolin-3-yl) - 4-fluorobenzamide (122) is depicted below in Scheme 27.

122 cm

Scheme 27

Step 1-2

[091] To a stirred solution of N-(6-bromoisoquinolin-3-yl)-4-fluorobenzamide (XCVIII) (800 mg, 2.32 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (647.4 mg, 2.55 mmol) in dioxane (6 mL) was added KOAc (682.4 mg, 6.95 mmol). The reaction was purged with argon for 5 min before adding Pd(dppf)Cl2 (169.6 mg, 0.23 mmol). The reaction was purged with argon for 5 min before heating at 90°C for about 3 h. The reaction was cooled to room temperature and 2,6-dichloropyrazine (XLVII) (517.9 mg, 3.48 mmol) was added followed by water (3 mL) and K3P04 (0.58 mL, 6.95 mmol). The reaction was purged with argon and Pd(PPli3)4 (133.9 mg, 0.12 mmol) was added. The mixture was heated again at 90 °C for about 2 h. The reaction was extracted into EtOAc the organic layer was washed with water, brine and dried (MgSO- before concentration to dryness. The residue was purified on a silica gel column (0→80% hexanes/EtOAc) to produce N-(6-(6-chloropyrazin-2-yl)isoquinolin-3-yl)-4-fluorobenzami de (CI) as a white solid (130 mg, 0.343 mmol, 14.8% yield). ESIMS found for C20H12CIFN4O mlz 344.9 (M+H-Cl).

Step 3

[092] To a solution of fert-butyl azetidin-3-ylcarbamate (CII) (27.3 mg, 0.16 mmol) and N-(6-(6-chloropyrazin-2-yl)isoquinolin-3-yl)-4-fluorobenzami de (CI) (40 mg, 0.11 mmol) in DMA (1 mL) was added CS2CO3 (68.8 mg, 0.21 mmol). The reaction mixture was heated at 70°C overnight. Water was added and the precipitate was collected through filtration, purified on a silica gel column (20→80% hexane/EtOAc). The pure fractions were collected and concentrated to get the product fert-butyl (l -(6-(3-(4-fluorobenzamido)isoquinolin-6-yl)pyrazin-2-yl)azet idin-3- yl)carbamate (CIII) (42 mg, 0.08 mmol, 77.3% yield). ESIMS found for C28H 2 7FN 6 0 3 mlz 515.0 (M+H).

Step 4

[093] To a solution of fert-butyl (l-(6-(3-(4-fluorobenzamido)isoquinolin-6- yl)pyrazin-2-yl) azetidin-3-yl)carbamate (CIII) (42 mg, 0.08 mmol) in DCM (1 mL) was added TFA (1 mL, 0.08 mmol). The reaction mixture was stirred at room temperature overnight. The reaction was concentrated and the residue purified on a silica gel column (0→50% DCM/DCM (20% of 7 N ML/MeOH)). Pure fractions were combined and concentrated to give N-(6-(6-(3- Aminoazetidin-l-yl)pyrazin-2-yl)isoquinolin-3-yl)-4-fluorobe nzamide 122 as a white solid (19.0 mg, 0.046 mmol, 56.2% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 2.26 (2 H, br s), 3.76 (2 H, dd, J=8.23, 5.76 Hz), 3.92 ( 1 H, quin, J=6.45 Hz), 4.33 (2 H, t, J=7.82 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.91 ( 1 H, s), 8.15 - 8.19 (2 H, m), 8.20 (1 H, d, J=4.39 Hz), 8.22 - 8.27 ( 1 H, m), 8.63 (1 H, s), 8.65 (1 H, s), 8.72 (1 H, s), 9.25 (1 H, s), 10.98 (1 H, s); ESIMS found for CzsHigFNeO mlz 414.9 (M+l).

Example 13.

[094] Preparation of N-(6-(6-(azetidin-3-ylmethoxy)pyrazin-2-yl)isoquinolin- 3-yl)- -fluorobenzamide (124) is depicted below in Scheme 28.

Scheme 28

Step 1

[095] To a stirred solution of tert-buty\ 3-(hydroxymethyl)azetidine-l -carboxylate (CIV) (29.7 mg, 0.16 mmol) and N-(6-(6-chloropyrazin-2-yl)isoquinolin-3-yl)-4-fluorobenzami de (CI) (40 mg, 0.1 1 mmol) in DMA (1 mL) was added Cs 2 C0 3 (68.8 mg, 0.21 mmol). The reaction mixture was heated at 70°C overnight. Water was added and precipitate was collected through filtration and purified on silica column (20→80% hexane/EtOAc). Pure fractions were collected and concentrated to yield the product fert-butyl 3-(((6-(3-(4-fluorobenzamido)isoquinolin-6- yl)pyrazin-2-yl)oxy)methyl)azetidine-l-carboxylate (CV) as a white solid (38 mg, 0.07 mmol, 68.0% yield). ESIMS found for m/z 530.0 (M+H).

Step 2

[0444] To a stirred solution of fert-butyl 3-(((6-(3-(4-fluorobenzamido)isoquinolin-6- yl) pyrazin-2-yl)oxy)methyl)azetidine-l-carboxylate (CV) (38 mg, 0.07 mmol) in DCM (1 mL) was added TFA ( 1 mL, 0.07 mmol). The reaction mixture was stirred at room temperature overnight. The reaction was concentrated and the residue was purified on a silica gel column (0→50% DCM/DCM (20% of 7 N NH 3 /MeOH)). Pure fractions were combined and concentrated to get N-(6-(6-(azetidin-3-ylmethoxy) pyrazin-2-yl)isoquinolin-3-yl)-4-fluorobenzamide 124 as a white solid (17.0 mg, 0.040 mmol, 55.2% yield). ¾ NMR (499 MHz, DMSO- 6 ) δ ppm 3.1 1 (1 H, dt, J=13.86, 6.79 Hz), 3.42 (2 H, t, J=6.86 Hz), 3.61 (2 H, t, J=7.82 Hz), 4.66 (2 H, d, J=7.14 Hz), 7.37 (2 H, t, J=8.78 Hz), 8.15 - 8.21 (2 H, m), 8.21 - 8.25 ( 1 H, m), 8.28 - 8.33 (1 H, m), 8.34 (1 H, s), 8.76 (2 H, d, J=6.04 Hz), 9.05 (1 H, s), 9.28 (1 H, s), 1 1.00 (1 H, br s); ESIMS found for C24H20FN5O2 mlz 429.9 (M+l).

[0445] The following compounds were prepared in accordance with the procedures described in the above Examp

1

[0446] N-(6-(3 -Amino-5 -fluorophenyl)isoquinolin-3 -yl)-2-(4-methylpiperazin- 1 -yl) isonicotinamide 1.

[0447] Beige solid (248.0 mg, 0.543 mmol, 77.2% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.40 - 2.46 (4 H, m), 3.55 - 3.67 (4 H, m), 5.58 (2 H, s), 6.41 (1 H, dt, J=1 1.46, 2.09 Hz), 6.73 - 6.81 ( 1 H, m), 6.87 ( 1 H, t, J=1.78 Hz), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.47 ( 1 H, s), 7.79 (1 H, dd, J=8.51, 1.92 Hz), 8.16 (2 H, d, J=9.60 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.68 (1 H, s), 9.23 (1 H, s), 1 1.10 ( 1 H, s); ESIMS found for CzeHzsFNeO mlz 457.2 (M+l).

2

[0448] N-(6-(3-Fluoro-5-(isopropylamino)phenyl)isoquinolin-3-yl)-2- (4- methylpiperazin-1 -yl)isonicotinamide 2.

[0449] White solid (27.0 mg, 0.054 mmol, 23.0% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ρρηι 1.17 (6 H, d, J=6.31 Hz), 2.23 (3 H, s), 2.38 - 2.46 (4 H, m), 3.55 - 3.62 (4 H, m), 3.68 (1 H, dq, J=14.03, 6.39 Hz), 5.93 (1 H, d, J=7.96 Hz), 6.40 (1 H, dt, J=12.08, 2.06 Hz), 6.76 (1 H, dt, J=9.88, 1.92 Hz), 6.84 (1 H, t, J=1.78 Hz), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.47 (1 H, s), 7.82 (1 H, dd, J=8.51, 1.65 Hz), 8.16 (1 H, d, J=8.78 Hz), 8.18 (1 H, s), 8.26 (1 H, d, J=4.94 Hz), 8.69 (1 H, s), 9.23 (1 H, s), 11.10 (1 H, br s); ESIMS found for C 2 9H 3 iFN 6 0 mlz 499.3 (M+l).

[0450] l-Methyl-N-(6-(pyridin-2-yl)isoquinolin-3-yl)piperidine-4-ca rboxamide 3.

[0451] White solid (36.5.0 mg, 0.075 mmol, 48.9% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.63 - 1.73 (2 H, m), 1.75 - 1.82 (2 H, m), 1.87 (2 H, td, J=l 1.66, 2.20 Hz), 2.16 (3 H, s), 2.51 - 2.56 (1 H, m), 2.77 - 2.87 (2 H, m), 7.44 (1 H, ddd, J=7.48, 4.73, 0.96 Hz), 7.97 (1

H, td, J=7.75, 1.78 Hz), 8.14 (1 H, d, J=8.51 Hz), 8.19 (1 H, d, J=7.96 Hz), 8.26 (1 H, dd, J=8.51,

I .65 Hz), 8.56 (1 H, s), 8.59 (1 H, s), 8.72 - 8.79 (1 H, m), 9.17 (1 H, s), 10.56 (1 H, s); ESIMS found for C 21 H 22 N 4 O mlz 347.2 (M+l).

4

[0452] 2-(4-Methylpiperazin-l-yl)-N-(6-(pyridin-2-yl)isoquinolin-3- yl)

isonicotinamide 4.

[0453] Beige solid (27.0 mg, 0.062 mmol, 38.0% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.56 - 3.63 (4 H, m), 7.17 (1 H, dd, J=5.21, 1.10 Hz), 7.44 - 7.47 (1 H, m), 7.48 (1 H, s), 7.98 (1 H, td, J=7.75, 1.78 Hz), 8.22 (2 H, dd, J=10.84, 8.37 Hz), 8.27 (1 H, d, J=5.21 Hz), 8.34 (1 H, dd, J=8.51, 1.65 Hz), 8.66 (1 H, s), 8.75 (1 H, s), 8.76 - 8.80 (1 H, m), 9.27 (1 H, s), 11.12 (1 H, s); ESIMS found for C 25 H 24 N 6 O mlz 425.2 (M+l).

[0454] 2-(4-Methylpiperazin-l-yl)-N-(6-(pyridin-4-yl)isoquinolin-3- yl)

isonicotinamide 5.

[0455] White solid (53.0 mg, 0.125 mmol, 76.0% yield). 'H NMR (499 MHz,

DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.57 - 3.65 (4 H, m), 7.17 (1 H, dd, J=4.94, 1.10 Hz), 7.48 ( 1 H, s), 7.88 - 7.97 (2 H, m), 8.00 (1 H, dd, J=8.78, 1.65 Hz), 8.21 - 8.30 (2 H, m), 8.47 ( 1 H, s), 8.69 - 8.75 (2 H, m), 8.77 (1 H, s), 9.30 ( 1 H, s), 1 1.15 ( 1 H, s); ESIMS found for C25H24N6O mlz 425.2 (M+l).

6

[0456] N-(6-(Pyridin-3-yl)isoquinolin-3-yl)cyclopropanecarboxamide 6.

[0457] White solid (27.0 mg, 0.093 mmol, 27.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 0.78 - 0.91 (4 H, m), 2.04 - 2.14 (1 H, m), 7.55 ( 1 H, dd, J=8.10, 4.80 Hz), 7.89 ( 1 H, dd, J=8.51, 1.65 Hz), 8.17 (1 H, d, J=8.51 Hz), 8.24 (1 H, s), 8.26 (1 H, dt, J=7.96, 1.92 Hz), 8.55 (1 H, s), 8.64 (1 H, dd, J=4.67, 1.37 Hz), 9.07 (1 H, d, J=2.47 Hz), 9.18 ( 1 H, s), 10.93 (1 H, s); ESIMS found for Ci 8 Hi 5 N 3 0 mlz 290.1 (M+l).

[0458] N-(6-(Pyridin-3-yl)isoquinolin-3-yl)-2-(pyrrolidin-l -yl)acetamide 7.

[0459] Light brown solid (45.0 mg, 0.135 mmol, 45.2% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.78 (4 H, dt, J=6.52, 3.19 Hz), 2.67 (4 H, br s), 3.38 (2 H, s), 7.56 (1 H, dd, J=7.96, 4.94 Hz), 7.92 (1 H, dd, J=8.51, 1.65 Hz), 8.19 ( 1 H, d, J=8.78 Hz), 8.28 (1 H, dt, J=8.23, 1.78 Hz), 8.33 (1 H, s), 8.58 (1 H, s), 8.65 (1 H, dd, J=4.67, 1.37 Hz), 9.09 ( 1 H, d, J=2.20 Hz), 9.18 ( 1 H, s), 10.03 (1 H, s); ESIMS found for C20H20N4O mlz 333.2 (M+l).

8

[0460] l-Methyl-N-(6-(pyridin-3-yl)isoquinolin-3-yl)piperidine-4-ca rboxamide 8.

[0461] White solid (25.0 mg, 0.075 mmol, 33.5% yield). 'HNMR (499 MHz, DMSO- £¾) δ ρριη 1.62 - 1.74 (2 H, m), 1.74 - 1.82 (2 H, m), 1.87 (2 H, td, J=l 1.60, 2.06 Hz), 2.14 - 2.20 (3 H, m), 2.51 - 2.56 (1 H, m), 2.77 - 2.85 (2 H, m), 7.56 (1 H, dd, J=7.82, 4.80 Hz), 7.89 (1 H, dd, J=8.51, 1.65 Hz), 8.16 (1 H, d, J=8.51 Hz), 8.23 - 8.30 (2 H, m), 8.58 ( 1 H, s), 8.65 ( 1 H, dd, J=4.80, 1.51 Hz), 9.07 (1 H, d, J=2.20 Hz), 9.17 (1 H, s), 10.56 ( 1 H, s); ESIMS found for C21H22N4O mlz 347.2 (M+ l).

9

[0462] N-(6-(Pyridin-3-yl)isoquinolin-3-yl)quinuclidine-4-carboxami de 9.

[0463] Beige solid (25.0 mg, 0.070 mmol, 50.3% yield). ¾ NMR (499 MHz, DMSO- £¾) 5 ppm 1.73 - 1.83 (6 H, m), 2.75 - 2.84 (6 H, m), 7.56 ( 1 H, dd, J=7.82, 4.80 Hz), 7.91 ( 1 H, dd, J=8.51, 1.65 Hz), 8.18 (1 H, d, J=8.51 Hz), 8.24 - 8.32 (2 H, m), 8.56 ( 1 H, s), 8.65 ( 1 H, dd, J=4.67, 1.37 Hz), 9.07 (1 H, d, J=1.92 Hz), 9.20 ( 1 H, s), 9.71 ( 1 H, s); ESIMS found for C 22 H 22 N 4 O mlz 359.2 (M+l).

[0464] 2-Mo holino-N-(6-(pyridin-3-yl)isoquinolin-3-yl)acetamide 10.

[0465] Off-white solid (80.0 mg, 230.0 mmol, 45.9% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.55 - 2.62 (4 H, m), 3.26 (2 H, s), 3.62 - 3.70 (4 H, m), 7.53 - 7.61 ( 1 H, m), 7.93 (1 H, dd, J=8.51, 1.92 Hz), 8.19 ( 1 H, d, J=8.51 Hz), 8.28 (1 H, dt, J=7.96, 1.92 Hz), 8.33 (1 H, s), 8.58 (1 H, s), 8.65 (1 H, dd, J=4.80, 1.51 Hz), 9.05 - 9.12 ( 1 H, m), 9.20 (1 H, s), 10.10 ( 1 H, s); ESIMS found for C20H20N4O2

[0466] 2-(4-Methylpiperazin-l-yl)-N-(6-(pyridin-3-yl)isoquinolin-3- yl)acetamide

11.

[0467] Beige solid (60.0 mg, 0.166 mmol, 60.3% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.19 (3 H, s), 2.40 (4 H, br s), 2.59 (4 H, br s), 3.23 (2 H, s), 7.56 (1 H, dd, J=7.68, 4.67 Hz), 7.93 (1 H, dd, J=8.64, 1.23 Hz), 8.19 (1 H, d, J=8.51 Hz), 8.28 (1 H, br d, J=7.96 Hz), 8.33 (1 H, s), 8.58 ( 1 H, s), 8.62 - 8.69 ( 1 H, m), 9.09 ( 1 H, d, J=1.92 Hz), 9.19 (1 H, s), 10.02 (1 H, s); ESIMS found for C2iH 23 N 5 0 mlz 362.2 (M+l).

[0468] 2-(4-Methyl-l ,4-diazepan- 1 -yl)-N-(6-(pyridin-3 -yl)isoquinolin-3-yl) acetamide 12.

[0469] Beige solid (35.0 mg, 0.093 mmol, 42.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.78 (2 H, quin, J=5.90 Hz), 2.29 (3 H, s), 2.56 - 2.64 (4 H, m), 2.78 - 2.88 (4 H, m), 3.38 (2 H, s), 7.56 (1 H, dd, J=7.96, 4.67 Hz), 7.92 (1 H, dd, J=8.51, 1.65 Hz), 8.18 (1 H, d, J=8.51 Hz), 8.28 (1 H, dt, J=7.89, 1.96 Hz), 8.33 (1 H, s), 8.58 (1 H, s), 8.65 (1 H, dd, J=4.80, 1.51 Hz), 9.09 (1 H, d, J=2.20 Hz), 9.19 (1 H, s), 10.05 (1 H, s); ESIMS found for C 22 H 25 N 5 0 mlz 376.2 (M+l).

13

[0470] 2-(4-Methylpiperazin- 1 -yl)-N-(6-(pyridin-3 -yl)isoquinolin-3 -yl)

isonicotinamide 13.

[0471] White solid (51.0 mg, 0.120 mmol, 73.2% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.39 - 2.46 (4 H, m), 3.57 - 3.66 (4 H, m), 7.17 (1 H, dd, J=5.21, 1.10 Hz), 7.48 (1 H, s), 7.57 (1 H, ddd, J=7.96, 4.67, 0.82 Hz), 7.97 (1 H, dd, J=8.64, 1.78 Hz), 8.23 (1 H, d, J=8.51 Hz), 8.26 (1 H, d, J=5.49 Hz), 8.28 - 8.34 (1 H, m), 8.37 (1 H, d, J=0.82 Hz), 8.66 (1 H, dd, J=4.80, 1.51 Hz), 8.74 (1 H, s), 9.11 (1 H, dd, J=2.47, 0.82 Hz), 9.28 (1 H, s), 11.13 (1 H, s); ESIMS found for C25H24N6O mlz 425.2 (M+l).

14

[0472] 2-(2-Methyl-lH-imidazol-l-yl)-N-(6-(pyridin-3-yl)isoquinolin -3-yl) acetamide 14.

[0473] White solid (25.0 mg, 0.073 mmol, 21.9% yield). ¾NMR (499 MHz, DMSO- d 6 ) 5 ppm 2.27 (3 H, s), 4.94 (2 H, s), 6.74 (1 H, d, .7=1.37 Hz), 7.09 (1 H, d, J=1.37 Hz), 7.55 (1 H, dd, J=7.41, 4.67 Hz), 7.93 (1 H, dd, J=8.51, 1.65 Hz), 8.19 (1 H, d, J=8.51 Hz), 8.26 (1 H, dt, J=8.23, 1.78 Hz), 8.28 (1 H, s), 8.53 (1 H, s), 8.64 (1 H, dd, J=4.80, 1.51 Hz), 9.07 (1 H, d, J=1.65 Hz), 9.22 (1 H, s), 10.99 (1 H, s); ESIMS found for C 2 oHi 7 N 5 0 mlz 344.2 (M+l).

15

[0474] 2-( lH-Imidazol- 1 -yl)-N-(6-(pyridin-3 -yl)isoquinolin-3 -yl)acetamide 15.

[0475] Beige solid ( 15.0 mg, 0.046 mmol, 13.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ρρηι 5.03 (2 H, s), 6.92 (1 H, s), 7.20 (1 H, s), 7.51 - 7.59 (1 H, m), 7.67 (1 H, s), 7.93 (1 H, dd, J=8.51, 1.92 Hz), 8.19 ( 1 H, d, J=8.51 Hz), 8.22 - 8.32 (2 H, m), 8.52 (1 H, s), 8.64 (1 H, dd, J=4.80, 1.51 Hz), 9.07 (1 H, d, J=1.65 Hz), 9.22 (1 H, s), 10.99 (1 H, s); ESIMS found for Ci 9 Hi 5 N 5 0 mlz 330.1 (M+ l).

16

[0476] trans-N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-3 - morpholinocyclobutane - 1 -carboxamide 16.

[0477] Grey solid (16.0 mg, 0.039 mmol, 20.5% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.07 - 2.17 (2 H, m), 2.27 (6 H, br s), 2.91 (1 H, quin, J=7.00 Hz), 3.24 - 3.30 (1 H, m), 3.59 (4 H, br t, J=4.25 Hz), 7.94 (1 H, dd, J=8.78, 1.65 Hz), 8.17 ( 1 H, d, J=8.78 Hz), 8.28 (1 H, dt, J=10.29, 2.26 Hz), 8.35 (1 H, s), 8.63 (1 H, s), 8.66 ( 1 H, d, J=2.74 Hz), 8.99 (1 H, s), 9.18 (1 H, s), 10.54 (1 H, s); ESIMS found for C23H23FN4O2 mlz 407.2 (M+l).

[0478] trans-N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-4-((4-methylpiperazin- 1 - yl)methyl)cyclohexane- 1 -carboxamide 17.

[0479] Off-white solid (33.0 mg, 0.072 mmol, 31.8% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.82 - 0.95 (2 H, m), 1.41 - 1.54 (3 H, m), 1.85 (4 H, br t, J=15.64 Hz), 2.08 (2 H, d, J=7.14 Hz), 2.14 (3 H, s), 2.31 (8 H, br s), 2.51 - 2.57 (1 H, m), 7.93 ( 1 H, dd, J=8.51, 1.65 Hz), 8.17 (1 H, d, J=8.78 Hz), 8.23 - 8.30 (1 H, m), 8.34 (1 H, s), 8.59 ( 1 H, s), 8.65 ( 1 H, d, J=2.47 Hz), 8.99 (1 H, t, J=1.65 Hz), 9.18 ( 1 H, s), 10.51 ( 1 H, s); ESIMS found for C 2 7H 32 FN 5 0 mlz 462.2 (M+ l).

18

[0480] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-2-(pyrrolidin- 1 -yl)acetamide

18.

[0481] Light brown solid (55.0 mg, 0.157 mmol, 55.0% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.72 - 1.84 (4 H, m), 2.62 - 2.72 (4 H, m), 3.37 (2 H, s), 7.97 (1 H, dd, J=8.51,

1.65 Hz), 8.19 (1 H, d, J=8.51 Hz), 8.29 (1 H, dt, J=10.09, 2.37 Hz), 8.42 ( 1 H, s), 8.59 (1 H, s),

8.66 (1 H, d, J=2.74 Hz), 9.01 ( 1 H, t, J=1.78 Hz), 9.20 ( 1 H, s), 10.04 ( 1 H, s); ESIMS found for C20H19FN4O mlz 351.2 (M+l).

19

[0482] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)- 1 -methylpiperidine-4- carboxamide 19.

[0483] Brown solid (4.0 mg, 0.0 mmol, 5.1% yield). ¾ NMR (500 MHz, DMSO- 6 ) 5 ppm 1.62 - 1.74 (2 H, m), 1.74 - 1.83 (2 H, m), 1.87 (2 H, td, J=l 1.53, 1.92 Hz), 2.16 (3 H, s), 2.51 - 2.57 (1 H, m), 2.76 - 2.86 (2 H, m), 7.94 (1 H, dd, J=8.51, 1.65 Hz), 8.17 (1 H, d, J=8.51 Hz), 8.27 (1 H, dt, J=10.22, 2.30 Hz), 8.34 (1 H, s), 8.59 (1 H, s), 8.66 (1 H, d, J=2.47 Hz), 8.99 (1 H, t, J=1.65 Hz), 9.18 (1 H, s), 10.56 ( 1 H, s); ESIMS found for C 21 H 21 FN 4 O mlz 365.0 (M+l).

[0484] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-2-(4-methylpiperazin- 1 -yl) acetamide 20.

[0485] White solid (35.0 mg, 0.092 mmol, 32.3% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.19 (3 H, s), 2.40 (4 H, br s), 2.59 (4 H, br s), 3.24 (2 H, s), 7.97 (1 H, dd, J=8.51, 1.92 Hz), 8.20 (1 H, d, J=8.51 Hz), 8.29 (1 H, dt, J=10.22, 2.30 Hz), 8.42 (1 H, s), 8.59 ( 1 H, s), 8.66 (1 H, d, J=2.47 Hz), 9.01 ( 1 H, t, J=1.65 Hz), 9.20 (1 H, s), 10.04 (1 H, s); ESIMS found for C21H22FN5O mlz 380.2 (M+l).

21

[0486] N-(6-(6-Fluoropyridin-3-yl)isoquinolin-3-yl)-2-(4-methylpipe razin-l-yl) acetamide 21.

[0487] Beige solid (58.0 mg, 0.153 mmol, 55.5% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.19 (3 H, s), 2.40 (4 H, br s), 2.59 (4 H, br s), 3.23 (2 H, s), 7.36 (1 H, br dd, J=8.37, 2.06 Hz), 7.91 (1 H, br d, J=8.51 Hz), 8.18 ( 1 H, br d, J=8.51 Hz), 8.33 (1 H, s), 8.44 - 8.54 (1 H, m), 8.57 (1 H, s), 8.76 ( 1 H, br s), 9.19 ( 1 H, s), 10.02 ( 1 H, s); ESIMS found for C 2 iH 2 2FN 5 0 mlz 380.2 (M+l).

22

[0488] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-2-morpholinoacetamide 22.

[0489] Tan solid (32.0 mg, 0.087 mmol, 61.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.58 (4 H, br s), 3.26 (2 H, s), 3.66 (4 H, br s), 7.97 (1 H, br d, J=8.23 Hz), 8.20 (1 H, br d, J=8.51 Hz), 8.29 (1 H, br d, J=10.43 Hz), 8.42 (1 H, s), 8.59 ( 1 H, s), 8.66 (1 H, d, J=1.65 Hz), 9.01 ( 1 H, s), 9.21 (1 H, s), 10.12 ( 1 H, s); ESIMS found for C20H19FN4O2 mlz 367.1 (M+l).

[0490] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-3 -morpholinopropanamide 23.

[0491] Beige solid (25.0 mg, 0.066 mmol, 23.9% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.44 (4 H, br s), 2.58 - 2.65 (2 H, m), 2.65 - 2.72 (2 H, m), 3.59 (4 H, t, J=4.53 Hz), 7.95 ( 1 H, dd, J=8.51, 1.65 Hz), 8.17 ( 1 H, d, J=8.51 Hz), 8.24 - 8.33 (1 H, m), 8.38 (1 H, s), 8.59 ( 1 H, s), 8.65 (1 H, d, J=2.74 Hz), 9.00 ( 1 H, t, J=1.78 Hz), 9.19 (1 H, s), 10.85 (1 H, s); ESIMS found for C21H21FN4O2 mlz 381.2 (M+l).

24

[0492] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-2-(4-methylpiperazin- 1 -yl) isonicotinamide 24.

[0493] White solid (74.0 mg, 0.167 mmol, 71.3% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.37 - 2.46 (4 H, m), 3.49 - 3.67 (4 H, m), 7.10 - 7.22 (1 H, m), 7.47 (1 H, s), 8.01 (1 H, dd, J=8.51, 1.65 Hz), 8.24 (1 H, br d, J=8.51 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.29 (1 H, dt, J=10.15, 2.20 Hz), 8.45 (1 H, s), 8.66 (1 H, d, J=2.47 Hz), 8.74 (1 H, s), 8.97 - 9.06 (1 H, m), 9.29 (1 H, s), 11.10 (1 H, s); ESIMS found for C 25 H 23 FN 6 O mlz 442.9 (M+l).

[0494] N-(6-(5 -Chloropyridin-3 -yl)isoquinolin-3 -yl)-2-(4-methylpiperazin- 1 -yl) isonicotinamide 25.

[0495] Beige solid (74.0 mg, 0.161 mmol, 68.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.38 - 2.47 (4 H, m), 3.54 - 3.69 (4 H, m), 7.16 (1 H, dd, J=4.94, 1.10 Hz), 7.46 (1 H, s), 8.00 (1 H, dd, J=8.64, 1.78 Hz), 8.23 (1 H, d, J=8.51 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.46 (1 H, s), 8.48 (1 H, t, J=2.06 Hz), 8.71 (1 H, d, J=2.20 Hz), 8.74 (1 H, s), 9.08 (1 H, d, J=1.92 Hz), 9.28 (1 H, s), 11.09 (1 H, s); ESIMS found for C 25 H 23 CIN 6 O mlz 458.95 (M+l).

26

[0496] N-(6-(5-(Difluoromethyl)pyridin-3-yl)isoquinolin-3-yl)-3-(pi peridin-4- yl)benzamide 26.

[0497] White solid (53.0 mg, 0.116 mmol, 64.6% yield). 'HNMR (499 MHz, DMSO- 6) 5 ppm 1.61 (2 H, qd, J=12.26, 3.84 Hz), 1.75 (2 H, br d, J=11.53 Hz), 2.61 (2 H, td, J=l 1.87, 1.78 Hz), 2.64 - 2.72 (1 H, m), 3.05 (2 H, br d, J=l 1.80 Hz), 7.25 (1 H, t, J=55.25 Hz), 7.42 - 7.51 (2 H, m), 7.85 - 7.93 (1 H, m), 7.97 - 8.06 (2 H, m), 8.25 (1 H, d, J=8.51 Hz), 8.47 (1 H, d, J=1.10 Hz), 8.51 ( 1 H, s), 8.77 (1 H, s), 8.87 (1 H, d, J=1.65 Hz), 9.24 - 9.35 (2 H, m), 10.97 ( 1 H, s); ESIMS found for C27H24F2N4O

27

[0498] N-(6-(5 -(Difluoromethyl)pyridin-3 -yl)isoquinolin-3 -yl)-3 -( 1 - methylpiperidin-4-yl)benzamide 27.

[0499] White solid (18.0 mg, 0.038 mmol, 43.7% yield). ¾ NMR (500 MHz, DMSO- d 6 ) δ ppm 1.67 - 1.85 (4 H, m), 1.99 (2 H, td, J=10.91, 3.98 Hz), 2.21 (3 H, s), 2.53 - 2.62 (1 H, m), 2.90 (2 H, br d, J=10.98 Hz), 7.26 ( 1 H, t, J=55.20 Hz), 7.42 - 7.53 (2 H, m), 7.90 (1 H, dt, J=7.27, 1.58 Hz), 8.01 (1 H, dd, J=8.51, 1.92 Hz), 8.03 (1 H, s), 8.25 (1 H, d, J=8.51 Hz), 8.48 ( 1 H, s), 8.52 (1 H, s), 8.77 (1 H, s), 8.87 (1 H, d, J=1.37 Hz), 9.22 - 9.35 (2 H, m), 10.97 ( 1 H, s); ESIMS found for C28H26F2N4O mlz 473.2 (M+l).

[0500] N-(6-(5-(Difluoromethyl)pyridin-3-yl)isoquinolin-3-yl)-2-(4- methylpiperazin-1 -yl)isonicotinamide 28.

[0501] Beige solid (65.0 mg, 0.137 mmol, 58.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=5.08 Hz), 3.57 - 3.66 (4 H, m), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.25 (1 H, t, J=55.25 Hz), 7.47 ( 1 H, s), 8.02 (1 H, dd, J=8.51, 1.65 Hz), 8.25 (1 H, d, J=3.84 Hz), 8.27 ( 1 H, s), 8.48 ( 1 H, s), 8.51 ( 1 H, s), 8.77 (1 H, s), 8.87 (1 H, d, J=1.37 Hz), 9.26 - 9.29 ( 1 H, m), 9.30 ( 1 H, s), 1 1.15 ( 1 H, s); ESIMS found for C26H24F2N6O mlz 475.2 (M+l).

[0502] 2-(4-Methylpiperazin- 1 -yl)-N-(6-(5 -(trifluoromethyl)pyridin-3 -yl) isoquinolin-3-yl)acetamide 29.

[0503] Brown solid (25.0 mg, 0.058 mmol, 42.3% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.19 (3 H, s), 2.40 (4 H, br s), 2.59 (4 H, br s), 3.24 (2 H, s), 8.04 (1 H, dd, J=8.51, 1.65 Hz), 8.22 (1 H, d, J=8.51 Hz), 8.53 (1 H, s), 8.62 (1 H, s), 8.70 (1 H, s), 9.04 ( 1 H, d, J=1.10 Hz), 9.22 ( 1 H, s), 9.41 ( 1 H, d, J=1.92 Hz), 10.04 (1 H, s); ESIMS found for C 22 H 22 F 3 N 5 0 mlz 430.2 (M+l).

[0504] 2-(4-Methylpiperazin-l-yl)-N-(6-(6-(trifluoromethyl)pyridin- 3-yl) isoquinolin-3-yl)acetamide 30.

[0505] Beige solid (75.0 mg, 0.175 mmol, 63.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.19 (3 H, br s), 2.40 (4 H, br s), 2.59 (4 H, br s), 3.24 (2 H, br s), 7.98 (1 H, br d, J=7.96 Hz), 8.06 (1 H, br d, J=7.96 Hz), 8.23 ( 1 H, br d, J=8.23 Hz), 8.45 ( 1 H, br s), 8.54 - 8.59 (1 H, m), 8.59 - 8.64 (1 H, m), 9.22 (1 H, br s), 9.28 (1 H, br s), 10.05 ( 1 H, br s) ; ESIMS found for C 2 2H 2 2F 3 N 5 0 mlz 430.2 (M+l).

31

[0506] N-(6-(5-(Hydroxymethyl)pyridin-3-yl)isoquinolin-3-yl)-l -methylpiperidine- 4-carboxamide 31.

[0507] White solid ( 10.0 mg, 0.027 mmol, 5.2% yield). ¾ NMR (499 MHz, DMSO- 6) 5 ppm 1.62 - 1.74 (2 H, m), 1.74 - 1.81 (2 H, m), 1.87 (2 H, td, J=l 1.53, 1.92 Hz), 2.16 (3 H, s), 2.51 - 2.56 (1 H, m), 2.81 (2 H, br d, J=l 1.25 Hz), 4.65 (2 H, d, J=5.49 Hz), 5.43 (1 H, t, J=5.63 Hz), 7.88 ( 1 H, dd, J=8.51, 1.65 Hz), 8.13 - 8.19 (2 H, m), 8.25 ( 1 H, s), 8.58 (1 H, s), 8.60 (1 H, d, .7=1.92 Hz), 8.95 (1 H, d, J=2.20 Hz), 9.17 ( 1 H, s), 10.56 (1 H, s); ESIMS found for C22H24N4O2 mlz 377.0 (M+l).

32

[0508] N-(6-(5-Cyanopyridin-3-yl)isoquinolin-3-yl)-l -methylpiperidine-4- carboxamide 32. [0509] Beige solid (37.0 mg, 0.100 mmol, 34.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.62 - 1.75 (2 H, m), 1.75 - 1.82 (2 H, m), 1.85 - 1.95 (2 H, m), 2.17 (3 H, s), 2.52 - 2.58 (1 H, m), 2.78 - 2.86 (2 H, m), 7.96 (1 H, dd, J=8.37, 1.51 Hz), 8.19 (1 H, d, J=8.51 Hz), 8.39 (1 H, s), 8.59 (1 H, s), 8.83 (1 H, t, J=1.92 Hz), 9.08 (1 H, d, J=1.92 Hz), 9.19 (1 H, s), 9.37 (1 H, d, J=2.20 Hz), 10.53 (1 H, s); ESIMS found for C 2 2H 2 iN 5 0 mlz 372.0 (M+l).

33

[0510] N-(6-(5-Methoxypyridin-3-yl)isoquinolin-3-yl)-l-methylpiperi dine-4- carboxamide 33.

[0511] White solid (48.6 mg, 0.081 mmol, 59.9% yield). ¾NMR (499 MHz, DMSO- d 6 ) 5 ppm 1.69 (2 H, qd, J=12.12, 3.43 Hz), 1.74 - 1.82 (2 H, m), 1.83 - 1.93 (2 H, m), 2.14 - 2.20 (3 H, m), 2.77 - 2.86 (2 H, m), 3.95 (3 H, s), 7.80 - 7.85 (1 H, m), 7.92 (1 H, dd, J=8.51, 1.65 Hz), 8.15 (1 H, d, J=8.51 Hz), 8.31 (1 H, s), 8.36 (1 H, d, J=2.74 Hz), 8.59 (1 H, s), 8.66 (1 H, d, J=1.65 Hz), 9.17 (1 H, s), 10.56 (1 H, s); ESIMS found for C22H24N4O2 mlz 377.2 (M+l).

[0512] N-(6-(5 -Methoxypyridin-3 -yl)isoquinolin-3-yl)-2-(4-methylpiperazin- 1 -yl) isonicotinamide 34.

[0513] Beige solid (51.0 mg, 0.112 mmol, 68.3% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.57 - 3.66 (4 H, m), 3.96 (3 H, s), 7.16 (1 H, dd, J=5.21, 1.10 Hz), 7.47 (1 H, s), 7.80 - 7.90 (1 H, m), 7.99 (1 H, dd, J=8.51, 1.65 Hz), 8.22 (1 H, d, J=8.51 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.37 (1 H, d, J=2.74 Hz), 8.41 (1 H, s), 8.69 (1 H, d, J=1.65 Hz), 8.75 (1 H, s), 9.28 (1 H, s), 11.13 (1 H, s); ESIMS found for CiMiWi mlz 455.2 (M+l).

[0514] 4-Fluoro-N-(6-(5-(piperidin-4-yloxy)pyridin-3-yl)isoquinolin -3-yl) benzamide 35.

[0515] White solid (51.0 mg, 0.1 15 mmol, 65.8% yield). ¾ NMR (499 MHz, DMSO- £¾) 5 ppm 1.44 - 1.58 (2 H, m), 1.93 - 2.03 (2 H, m), 2.57 - 2.67 (2 H, m), 2.97 (2 H, dt, J=12.69, 4.08 Hz), 4.67 - 4.76 ( 1 H, m), 7.37 (2 H, t, J=8.78 Hz), 7.86 ( 1 H, t, J=2.20 Hz), 7.96 (1 H, dd, J=8.51, 1.65 Hz), 8.14 - 8.24 (3 H, m), 8.35 (1 H, d, J=2.74 Hz), 8.38 ( 1 H, s), 8.65 ( 1 H, d, J=1.65 Hz), 8.74 (1 H, s), 9.26 ( 1 H, s), 10.97 (1 H, s); ESIMS found for C26H23FN4O2 mlz 442.95 (M+1).

[0516] N-(6-(5-Aminopyridin-3-yl)isoquinolin-3-yl)-4-fluorobenzamid e 36.

[0517] Beige solid (52.0 mg, 0.145 mmol, 44.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 5.48 (2 H, s), 7.33 (1 H, t, J=2.20 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.81 ( 1 H, dd, J=8.64, 1.51 Hz), 8.01 (1 H, d, J=2.47 Hz), 8.1 1 - 8.21 (4 H, m), 8.22 (1 H, d, J=1.92 Hz), 8.69 (1 H, s), 9.24 ( 1 H, s), 10.96 (1 H, s); ESIMS found for C 2 iHi 5 FN 4 0 mlz 359.1 (M+1).

37

[0518] N-(6-(5-Aminopyridin-3-yl)isoquinolin-3-yl)-4-(piperidin-4-y loxy) benzamide 37.

[0519] Beige solid ( 10.0 mg, 0.022 mmol, 43.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) 5 ppm 1.40 - 1.53 (2 H, m), 1.89 - 2.00 (2 H, m), 2.55 - 2.64 (2 H, m), 2.96 (2 H, dt, J=12.42, 3.81 Hz), 4.50 - 4.59 (1 H, m), 5.48 (2 H, s), 7.06 (2 H, d, J=9.06 Hz), 7.33 (1 H, t, J=2.20 Hz), 7.76 - 7.84 (1 H, m), 8.01 (1 H, d, J=2.47 Hz), 8.07 (2 H, d, J=8.78 Hz), 8.1 1 - 8.20 (2 H, m), 8.22 ( 1 H, d, J=1.92 Hz), 8.68 (1 H, s), 9.22 (1 H, s), 10.70 ( 1 H, s); ESIMS found for CzeHzsNsOz mlz 440.2 (M+1).

38 [0520] N-(6-(5 -Aminopyridin-3 -yl)isoquinolin-3 -yl)-2-(4-methylpiperazin- 1 -yl) isonicotinamide 38.

[0521] Beige solid (7.0 mg, 0.016 mmol, 6.8% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ρρηι 2.24 (3 H, s), 2.40 - 2.45 (4 H, m), 3.56 - 3.64 (4 H, m), 5.47 (2 H, s), 7.17 ( 1 H, d, J=5.21 Hz), 7.34 ( 1 H, d, J=1.92 Hz), 7.47 (1 H, s), 7.82 (1 H, dd, J=8.64, 1.24 Hz), 8.02 (1 H, d, J=2.47 Hz), 8.14 - 8.21 (2 H, m), 8.22 (1 H, d, J=1.65 Hz), 8.26 ( 1 H, d, J=4.94 Hz), 8.70 ( 1 H, s), 9.25 (1 H, s), 1 1.10 (1 H, s); ESIMS (M+l).

[0522] N-(6-(6-Aminopyridin-3-yl)isoquinolin-3-yl)-2-(4-methylpiper azin-l-yl) isonicotinamide 39.

[0523] White solid (75.0 mg, 0.171 mmol, 72.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=5.08 Hz), 3.55 - 3.66 (4 H, m), 6.23 (2 H, s), 6.59 ( 1 H, d, J=8.78 Hz), 7.16 (1 H, dd, J=5.21, 1.10 Hz), 7.47 (1 H, s), 7.85 ( 1 H, dd, J=8.51, 1.65 Hz), 7.93 (1 H, dd, J=8.51, 2.47 Hz), 8.1 1 (1 H, d, J=8.78 Hz), 8.13 ( 1 H, s), 8.26 ( 1 H, d, J=5.21 Hz), 8.49 ( 1 H, d, J=2.20 Hz), 8.64 (1 H, s), 9.17 ( 1 H, s), 1 1.04 ( 1 H, s); ESIMS found for C 25 H 25 N 7 O mlz 440.0 (M+ l).

40

[0524] N-(6-(6-(Methylamino)pyridin-3-yl)isoquinolin-3-yl)-2-(4-met hylpiperazin- l-yl)isonicotinamide 40.

[0525] White solid (70.0 mg, 0.154 mmol, 65.8% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 2.85 (3 H, d, J=4.67 Hz), 3.57 - 3.64 (4 H, m), 6.59 (1 H, d, J=8.78 Hz), 6.80 (1 H, q, J=4.57 Hz), 7.16 ( 1 H, dd, J=5.08, 1.24 Hz), 7.47 ( 1 H, s), 7.86 ( 1 H, dd, J=8.64, 1.78 Hz), 7.95 ( 1 H, dd, J=8.78, 2.47 Hz), 8.1 1 (1 H, d, J=8.78 Hz), 8.13 ( 1 H, s), 8.26 ( 1 H, d, J=4.94 Hz), 8.57 ( 1 H, d, J=2.20 Hz), 8.64 (1 H, s), 9.17 (1 H, s), 1 1.04 (1 H, s); ESIMS found for C26H27N7O mlz 454.0 (M+l).

41

[0526] N-(6-(5-(Isopropylamino)pyridin-3-yl)isoquinolin-3-yl)-3-(pi peridin-4-yl) benzamide 41.

[0527] White solid (46.0 mg, 0.099 mmol, 45.8% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.20 (6 H, d, J=6.31 Hz), 1.61 (2 H, qd, J=12.17, 3.84 Hz), 1.74 (2 H, br d, J=l 1.53 Hz), 2.61 (2 H, td, J=12.08, 2.20 Hz), 2.64 - 2.73 ( 1 H, m), 3.05 (2 H, br d, J=l 1.80 Hz), 3.76 (1 H, dq, J=14.24, 6.32 Hz), 5.85 ( 1 H, d, J=8.23 Hz), 7.29 (1 H, t, J=2.33 Hz), 7.42 - 7.50 (2 H, m), 7.86 (1 H, dd, J=8.51, 1.65 Hz), 7.88 - 7.94 (1 H, m), 8.00 (1 H, s), 8.02 ( 1 H, d, J=2.47 Hz), 8.18 (1 H, d, J=8.78 Hz), 8.20 (1 H, d, J=1.92 Hz), 8.25 (1 H, s), 8.72 (1 H, s), 9.24 (1 H, s), 10.93 ( 1 H, s); ESIMS found for C 2 9H 3 iN 5 0

42

[0528] N-(6-(5 -(Isopropylamino)pyridin-3 -yl)isoquinolin-3 -yl)-3 -( 1 - methylpiperidin-4-yl)benzamide 42.

[0529] White solid (13.0 mg, 0.027 mmol, 31.5% yield). ¾ NMR (499 MHz, DMSO- d 6 ) 5 ppm 1.20 (6 H, d, J=6.31 Hz), 1.73 - 1.84 (4 H, m), 1.99 (2 H, td, J=l 1.1 1, 3.57 Hz), 2.21 (3 H, s), 2.53 - 2.61 (1 H, m), 2.90 (2 H, br d, J=1 1.53 Hz), 3.76 (1 H, dq, J=14.10, 6.37 Hz), 5.85 ( 1 H, d, J=7.96 Hz), 7.29 (1 H, t, J=2.20 Hz), 7.41 - 7.51 (2 H, m), 7.86 (1 H, dd, J=8.51, 1.65 Hz), 7.88 - 7.93 ( 1 H, m), 8.02 (2 H, d, J=2.74 Hz), 8.18 ( 1 H, d, J=8.51 Hz), 8.20 ( 1 H, d, J=1.92 Hz), 8.25 (1 H, s), 8.72 ( 1 H, s), 9.24 ( 1 H, s), 10.92 (1 H, s); ESIMS found for C 3 oH 33 N 5 0 mlz 480.25 (M+ l).

43 [0530] N-(6-(5-(Isopropylamino)pyridin-3-yl)isoquinolin-3-yl)-2-(4- methylpiperazin-1 -yl)isonicotinamide 43.

[0531] White solid (10.0 mg, 0.021 mmol, 9.1% yield). 'H NMR (499 MHz, DMSO- d 6 ) δ ppm 1.19 (6 H, d, J=6.31 Hz), 2.24 (3 H, s), 2.43 (4 H, br t, J=4.80 Hz), 3.58 - 3.65 (4 H, m), 3.76 (1 H, dq, J=14.13, 6.36 Hz), 5.85 (1 H, d, J=8.23 Hz), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.28 (1 H, t, J=2.20 Hz), 7.48 (1 H, s), 7.87 (1 H, dd, J=8.51, 1.65 Hz), 8.02 (1 H, d, J=2.74 Hz), 8.17 - 8.22 (2 H, m), 8.26 (2 H, br d, J=4.39 Hz), 8.72 (1 H, s), 9.25 (1 H, s), 11.11 (1 H, s); ESIMS found for C 2 8H 3 iN 7 0 mlz 482.3 (M+l

[0532] N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl)isoquinolin-3-yl)

cyclohexanecarboxamide 44.

[0533] White solid (70.0 mg, 0.163 mmol, 78.4% yield). ¾NMR (499 MHz, DMSO- d 6 ) 5 ppm 1.15 - 1.33 (3 H, m), 1.33 - 1.40 (2 H, m), 1.40 - 1.51 (2 H, m), 1.66 (1 H, br d, J=11.25 Hz), 1.72 - 1.80 (2 H, m), 1.84 (2 H, br d, J=12.08 Hz), 1.91 - 1.98 (2 H, m), 2.54 - 2.66 (3 H, m), 2.95 - 3.03 (2 H, m), 3.88 - 3.98 (1 H, m), 7.15 (1 H, d, J=7.41 Hz), 7.95 (1 H, s), 8.09 - 8.13 (1 H, m), 8.13 - 8.17 (1 H, m), 8.43 (1 H, s), 8.49 (1 H, s), 8.55 (1 H, s), 9.14 (1 H, s), 10.47 (1 H, s); ESIMS found for C 2 5H 3 oN 6 0 mlz 431.2 (M+l).

[0534] 4-fluoro-N-(6-(5-(piperidin-4-ylamino)pyridin-3-yl)isoquinol in-3-yl) benzamide 45.

[0535] White solid (25.0 mg, 0.057 mmol, 65.3% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 1.25 - 1.35 (2 H, m), 1.87 - 1.96 (2 H, m), 2.56 - 2.66 (2 H, m), 2.92 - 3.02 (3 H, m), 3.43 - 3.54 (1 H, m), 5.85 (1 H, d, J=8.51 Hz), 7.30 (1 H, t, J=2.20 Hz), 7.32 - 7.40 (2 H, m), 7.85 (1 H, dd, J=8.51, 1.65 Hz), 8.05 (1 H, d, J=2.74 Hz), 8.13 - 8.21 (4 H, m), 8.23 (1 H, s), 8.70 (1 H, s), 9.24 (1 H, s), 10.91 (1 H, br s); ESIMS found for C 26 H 24 FN 5 0 mlz 441.95 (M+l).

[0536] 4-Fluoro-N-(6-(5-(( l-methylpiperidin-4-yl)amino)pyridin-3-yl)isoquinolin-3- yl)benzamide 46.

[0537] Beige solid (25.0 mg, 0.055 mmol, 9.5% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.39 - 1.50 (2 H, m), 1.88 - 1.97 (2 H, m), 2.03 - 2.12 (2 H, m), 2.18 (3 H, s), 2.73 (2 H, br d, J=l 1.80 Hz), 3.37 - 3.46 (1 H, m), 5.90 (1 H, d, J=8.23 Hz), 7.31 (1 H, t, J=2.20 Hz), 7.37 (2 H, t, J=8.78 Hz), 7.86 (1 H, dd, J=8.51, 1.65 Hz), 8.04 ( 1 H, d, J=2.47 Hz), 8.14 - 8.22 (4 H, m), 8.25 (1 H, s), 8.71 (1 H, s), 9.24 (1 H, s), 10.96 ( 1 H, s); ESIMS found for C 27 H 26 FN 5 0 mlz 456.2 (M+ l).

47

[0538] N-(6-(5-Acetamidopyridin-3-yl)isoquinolin-3-yl)-l-methylpipe ridine-4- carboxamide 47.

[0539] White solid (3.0 mg, 0.007 mmol, 3.5% yield). ¾ NMR (500 MHz, DMSO- d 6 ) 5 ppm 1.58 - 1.73 (2 H, m), 1.75 - 1.82 (2 H, m), 1.87 (2 H, td, J= 1 1.66, 2.20 Hz), 2.13 (3 H, s), 2.16 (3 H, s), 2.52 - 2.56 ( 1 H, m), 2.77 - 2.87 (2 H, m), 7.73 - 7.85 ( 1 H, m), 8.16 ( 1 H, s), 8.17 ( 1 H, d, J=5.76 Hz), 8.45 ( 1 H, t, J=2.20 Hz), 8.57 (1 H, s), 8.72 (1 H, d, J=2.20 Hz), 8.76 (1 H, d, J=2.20 Hz), 9.18 ( 1 H, s), 10.30 (1 H, s), 10.55 ( 1 H, s); ESIMS found for C 2 3H25N 5 0 2 mlz 404.0 (M+ l).

[0540] N-(6-(5-(Dimethylamino)pyridin-3-yl)isoquinolin-3-yl)-2-(4- methylpiperazin-1 -yl)isonicotinamide 48. [0541] Beige solid (58.0 mg, 0.124 mmol, 52.9% yield). ¾NMR(499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.40 - 2.47 (4 H, m), 3.05 (6 H, s), 3.55 - 3.65 (4 H, m), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.43 -7.52 (2 H, m), 7.95 (1 H, dd,J=8.51, 1.65 Hz), 8.15 - 8.23 (2 H, m), 8.26 (1 H, d,J=5.21 Hz), 8.32 - 8.39 (2 H, m), 8.74 (1 H, s), 9.26 (1 H, s), 11.11 (1 H, s); ESIMS found for C 27 H 29 N 7 O mlz 468.0 (M+l).

[0542] N-(6-(6-(Dimethylamino)pyridin-3-yl)isoquinolin-3-yl)-2-(4- methylpiperazin-1 -yl)isonicotinamide 49.

[0543] Beige solid (70.0 mg, 0.150 mmol, 63.8% yield). ¾NMR(499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, br t, J=4.94 Hz), 3.11 (6 H, s), 3.55 - 3.66 (4 H, m), 6.79 (1 H, d,J=8.78Hz), 7.16 (1 H, dd,J=4.94, 0.82 Hz), 7.47 (1 H, s), 7.89 (1 H, dd,J=8.51, 1.37 Hz), 8.06 (1 H, dd,J=8.92, 2.61 Hz), 8.12 (1 H, d,J=8.51 Hz), 8.17 (1 H, s), 8.26 (1 H, d,J=5.21 Hz), 8.65 (1 H, s), 8.66 (1 H, d, J=2.47 Hz), 9.18 (1 H, s), 11.04 (1 H, s); ESIMS found for C 27 H 29 N 7 O mlz 468.0 (M+l).

50

[0544] 1 -Methyl -N-(6-(5 -(pyrrolidin- 1 -yl)pyridin-3 -yl)isoquinolin-3 -yl)piperidine- 4-carboxamide 50.

[0545] White solid (3.0 mg, 0.007 mmol, 5.0% yield). ¾ NMR (499 MHz, DMSO- d 6 ) 5ppm 1.63 - 1.74 (2 H, m), 1.75 - 1.82 (2 H, m), 1.88 (2 H, td, J= 11.66, 2.20 Hz), 1.98 -2.04 (4 H, m), 2.16 (3 H, s), 2.52 - 2.57 (1 H, m), 2.77 - 2.86 (2 H, m), 3.37 (4 H, br t, J=6.45 Hz), 7.24 (1 H,t,J=2.20Hz), 7.86 (1 H, dd,J=8.51, 1.65 Hz), 8.00 (1 H, d,J=2.47Hz), 8.12 (1 H, d,J=8.51 Hz), 8.22 (1 H, s), 8.27 (1 H, d, J=1.65 Hz), 8.57 (1 H, s), 9.14 (1 H, s), 10.47 (1 H, s); ESIMS found for C 25 H29N 5 0 mlz 416.0 (M+l).

[0546] N-(6-(5 -(Pyrrolidin- 1 -ylmethyl)pyridin-3 -yl)isoquinolin-3 -yl)

cyclopropanecarboxamide 51.

[0547] Off-white solid (34.0 mg, 0.091 mmol, 48.3% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.80 - 0.85 (2 H, m), 0.85 - 0.90 (2 H, m), 1.72 (4 H, dt, J=6.66, 3.12 Hz), 2.05 - 2.13 (1 H, m), 3.26 - 3.31 (4 H, m), 3.71 (2 H, s), 7.88 (1 H, dd, J=8.51, 1.65 Hz), 8.14 ( 1 H, t, J=1.92 Hz), 8.16 ( 1 H, d, J=8.78 Hz), 8.25 (1 H, s), 8.52 - 8.58 (2 H, m), 8.94 (1 H, d, J=2.20 Hz), 9.18 ( 1 H, s), 10.91 (1 H, s); ESIMS found for C23H24N4O mlz 373.0 (M+l).

52

[0548] 4-Fluoro-N-(6-(5-(pyrrolidin-l -ylmethyl)pyridin-3-yl)isoquinolin-3-yl) benzamide 52.

[0549] White solid (40.0 mg, 0.089 mmol, 66.2% yield). 'HNMR (500 MHz, DMSO- d 6 ) δ ppm 1.73 (4 H, dt, J=6.72, 3.22 Hz), 3.72 (2 H, s), 7.30 - 7.42 (2 H, m), 7.95 (1 H, dd, J=8.51, 1.92 Hz), 8.13 - 8.20 (3 H, m), 8.22 ( 1 H, d, J=8.51 Hz), 8.37 (1 H, s), 8.58 ( 1 H, d, J=1.92 Hz), 8.74 ( 1 H, s), 8.98 (1 H, d, J=2.20 Hz), 9.26 ( 1 H, s), 10.96 (1 H, s); ESIMS found for C 26 H 23 FN 4 O mlz 426.95 (M+l).

53

[0550] N-(6-(5 -((4-Hydroxypiperidin- 1 -yl)methyl)pyridin-3 -yl)isoquinolin-3 -yl) cyclopropanecarboxamide 53.

[0551] Off-white solid (9.0 mg, 0.022 mmol, 1 1.8% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.84 (2 H, dt, J=7.89, 2.64 Hz), 0.85 - 0.90 (2 H, m), 1.36 - 1.46 (2 H, m), 1.68 - 1.76 (2 H, m), 2.05 -2.16(3 H, m), 2.67 -2.75 (2 H, m), 3.42-3.51 (1 H, m), 3.58 (2 H, s), 4.53 (1 H, d,J=4.12Hz), 7.88 (1 H, dd,J=8.51, 1.92 Hz), 8.12 (1 H, t, J=2.06 Hz), 8.16 (1 H, d,J=8.51 Hz), 8.24 (1 H, s), 8.54 (1 H, d, J=1.92 Hz), 8.55 (1 H, s), 8.94 (1 H, d, J=2.20 Hz), 9.18 (1 H, s), 10.91 (1 H, s); ESIMS found for C24H26N4O2 mlz 403.0 (M+l).

[0552] 4-Fluoro-N-(6-(5-((4-hydroxypiperidin-l-yl)methyl)pyridin-3- yl)isoquinolin- 3-yl)benzamide 54.

[0553] White solid (10.0 mg, 0.021 mmol, 61.5%yield). ¾NMR(499 MHz, DMSO- d 6 ) δ ppm 1.36 - 1.47 (2 H, m), 1.69 - 1.78 (2 H, m), 2.12 (2 H, br t, J=9.88 Hz), 2.68 - 2.78 (2 H, m), 3.47 (1 H, dt, J=8.23, 4.12 Hz), 3.60 (2 H, s), 4.52 (1 H, d, J=4.12 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.95 (1 H, dd,J=8.51, 1.65 Hz), 8.13 - 8.20 (3 H, m), 8.22 (1 H, d,J=8.51 Hz), 8.37 (1 H, s), 8.56 (1 H, d, J=1.65 Hz), 8.74 (1 H, s), 8.98 (1 H, d, J=1.92 Hz), 9.26 (1 H, s), 10.96 (1 H, s); ESIMS found for C27H25FN4O2

55

[0554] N-(6-(5 -(Piperidin-4-yloxy)pyridin-3 -yl)isoquinolin-3 -yl)

cyclopropanecarboxamide 55.

[0555] White solid (70.0 mg, 0.180 mmol, 45.1% yield). 'HNMR (499 MHz, DMSO- d 6 ) δ ppm 0.80 - 0.85 (2 H, m), 0.85 - 0.91 (2 H, m), 1.44 - 1.55 (2 H, m), 1.93 - 2.01 (2 H, m), 2.05 - 2.13 (1 H, m), 2.57 - 2.65 (2 H, m), 2.96 (2 H, dt, J=12.69, 3.95 Hz), 4.63 - 4.73 (1 H, m), 7.81 (1 H,t,J=2.33 Hz), 7.88 (1 H, dd,J=8.51, 1.65 Hz), 8.13 (1 H, d,J=8.51 Hz), 8.25 (1 H, s), 8.33 (1 H, d, J=2.74 Hz), 8.56 (1 H, s), 8.61 (1 H, d, J=1.65 Hz), 9.16 (1 H, s), 10.91 (1 H, s); ESIMS found for C23H24N4O2 mlz 389.0 (M+l).

56

[0556] N-(6-(5 -((4-Methylpiperazin- 1 -yl)methyl)pyridin-3 -yl)isoquinolin-3 -yl) cyclopropanecarboxamide 56.

[0557] Off-white solid (36.0 mg, 0.090 mmol, 28.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.77 - 0.85 (2 H, m), 0.85 - 0.91 (2 H, m), 2.04 - 2.12 (1 H, m), 2.15 (3 H, s), 2.33 (4 H, br s), 2.43 (4 H, br s), 3.60 (2 H, s), 7.88 ( 1 H, dd, J=8.51, 1.65 Hz), 8.12 ( 1 H, t, J=2.06 Hz), 8.16 (1 H, d, J=8.78 Hz), 8.24 (1 H, s), 8.51 - 8.58 (2 H, m), 8.95 (1 H, d, J=2.20 Hz), 9.18 ( 1 H, s), 10.91 (1 H, s); ESIMS found for Cz^vNsO mlz 402.0 (M+1).

57

[0558] N-(6-(5 -(Morpholinomethyl)pyridin-3 -yl)isoquinolin-3 -yl)

cyclopropanecarboxamide 57.

[0559] Off-white solid (54.0 mg, 0.139 mmol, 44.1% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.79 - 0.85 (2 H, m), 0.85 - 0.91 (2 H, m), 2.05 - 2.13 ( 1 H, m), 2.39 - 2.46 (4 H, m), 3.55 - 3.61 (4 H, m), 3.61 (2 H, s), 7.89 ( 1 H, dd, J=8.51, 1.65 Hz), 8.12 - 8.19 (2 H, m), 8.25 ( 1 H, s), 8.56 (1 H, s), 8.57 (1 H, d, J=1.92 Hz), 8.96 (1 H, d, J=2.20 Hz), 9.18 ( 1 H, s), 10.91 (1 H, s); ESIMS found for C23H24

[0560] N-Methyl-5-(3-( l-methylpiperidine-4-carboxamido)isoquinolin-6-yl) nicotinamide 58. [0561] Beige solid (3.0 mg, 0.007 mmol, 3.5% yield). ¾ NMR (500 MHz, DMSO- 6) 5 ppm 1.62 - 1.74 (2 H, m), 1.75 - 1.82 (2 H, m), 1.84 - 1.93 (2 H, m), 2.16 (3 H, s), 2.51 - 2.57 ( 1 H, m), 2.78 - 2.83 (2 H, m), 2.86 (3 H, d, J=4.39 Hz), 7.95 ( 1 H, dd, J=8.51, 1.65 Hz), 8.19 (1 H, d, J=8.51 Hz), 8.34 (1 H, s), 8.57 - 8.62 (2 H, m), 8.76 ( 1 H, br d, J=3.84 Hz), 9.03 (1 H, d, J=2.20 Hz), 9.17 - 9.23 (2 H, m), 10.56 ( 1 H, s); ESIMS found for C 2 3H25N 5 0 2 mlz 404.0 (M+l).

59

[0562] N-Methyl-5-(3-(2-(4-methylpiperazin-l-yl)isonicotinamido)iso quinolin-6- yl)nicotinamide 59.

[0563] Off-white solid (73.0 mg, 0.152 mmol, 80.8% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 2.87 (3 H, d, J=4.39 Hz), 3.57 - 3.65 (4 H, m), 7.16 (1 H, dd, J=5.08, 1.23 Hz), 7.47 ( 1 H, s), 8.03 (1 H, dd, J=8.65, 1.78 Hz), 8.26 (1 H, s), 8.27 (1 H, d, J=1.92 Hz), 8.45 ( 1 H, s), 8.64 (1 H, t, J=2.20 Hz), 8.72 - 8.81 (2 H, m), 9.04 (1 H, d, J=1.92 Hz), 9.23 (1 H, d, J=1.92 Hz), 9.30 (1 H, s), 1 1.14 (1 H, s); ESIMS found for C27H27N7O2 mlz 482.2 (M+l).

[0564] N-(6-(pyridin-3-yl-£/4)isoquinolin-3-yl)-2-(pyrrolidin-l-yl )acetamide 60.

[0565] Beige solid (44.0 mg, 0.131 mmol, 37.0% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.78 (4 H, dt, J=6.59, 3.29 Hz), 2.61 - 2.70 (4 H, m), 3.37 (2 H, s), 7.93 (1 H, dd, J=8.51, 1.65 Hz), 8.19 ( 1 H, d, J=8.51 Hz), 8.34 (1 H, s), 8.58 (1 H, s), 9.18 ( 1 H, s), 10.02 ( 1 H, s); ESIMS found for C 2 oHi6[ 2 H4]N 4 0 mlz 337.2 (M+l).

[0566] 2-(4-Methylpiperazin-l-yl)-N-(6-(pyridin-3-yl-£/ 4 ()isoquinolin-3-yl)acetamide

61. [0567] White solid (15.0 mg, 0.041 mmol, 14.9% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.19 (3 H, s), 2.40 (4 H, br s), 2.59 (4 H, br s), 3.23 (2 H, s), 7.93 (1 H, dd, J=8.64, 1.78 Hz), 8.19 ( 1 H, d, J=8.51 Hz), 8.34 ( 1 H, s), 8.58 ( 1 H, s), 9.19 ( 1 H, s), 10.02 ( 1 H, s); ESIMS found for C 2 iHi9[ 2 H 4 ]N 5 0 mlz 366.2 (M+l).

62

[0568] 2-Mo holino-N-(6-(pyridin-3-yl-ί 4)isoquinolin-3-yl)acetamide 62.

[0569] Brown solid (25.0 mg, 0.071 mmol, 41.4% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.55 - 2.63 (4 H, m), 3.26 (2 H, s), 3.63 - 3.70 (4 H, m), 7.93 ( 1 H, dd, J=8.51, 1.37 Hz), 8.19 ( 1 H, d, J=8.51 Hz), 8.33 (1 H, s), 8.58 (1 H, s), 9.19 ( 1 H, s), 10.10 ( 1 H, s); ESIMS found for C 2 oHi6[ 2 H4]N 4 02 mlz 353.2 (M+l).

63

[0570] l-Methyl-N-(6-(2-methylpyrimidin-5-yl)isoquinolin-3-yl)piper idine-4- carboxamide 63.

[0571] White solid (44.8 mg, 0.078 mmol, 57.6% yield). ¾ NMR (499 MHz, DMSO- 6) 5 ppm 1.62 - 1.73 (2 H, m), 1.75 - 1.81 (2 H, m), 1.87 (2 H, td, J=1 1.66, 2.20 Hz), 2.16 (3 H, s), 2.51 - 2.56 (1 H, m), 2.70 (3 H, s), 2.78 - 2.85 (2 H, m), 7.92 (1 H, dd, J=8.51, 1.65 Hz), 8.17 (1 H, d, J=8.51 Hz), 8.32 (1 H, s), 8.58 (1 H, s), 9.18 (1 H, s), 9.19 (2 H, s), 10.58 (1 H, s); ESIMS found for C 2 iH 23 N 5 0 mlz 362.2 (M+l).

64

[0572] 2-(4-Methylpiperazin-l-yl)-N-(6-(2-methylpyrimidin-5-yl)isoq uinolin-3- yl)isonicotinamide 64.

[0573] Beige solid (55.0 mg, 0.125 mmol, 76.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, br t, J=4.80 Hz), 2.71 (3 H, s), 3.53 - 3.68 (4 H, m), 7.16 ( 1 H, dd, J=5.08, 0.96 Hz), 7.47 ( 1 H, s), 8.00 (1 H, dd, J=8.51, 1.65 Hz), 8.24 (1 H, d, J=8.51 Hz), 8.26 ( 1 H, d, .7=5.21 Hz), 8.43 (1 H, s), 8.74 (1 H, s), 9.22 (2 H, s), 9.28 (1 H, s), 1 1.14 (1 H, s); ESIMS found for C25H25N7O mlz 440.2 (M+l).

[0574] N-(6-(2-Aminopyrimidin-5-yl)isoquinolin-3-yl)-l -methylpiperidine-4- carboxamide 65.

[0575] Beige solid ( 1 1.0 mg, 0.030 mmol, 14.1% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.62 - 1.73 (2 H, m), 1.74 - 1.83 (2 H, m), 1.85 - 1.93 (2 H, m), 2.17 (3 H, s), 2.52 - 2.56 ( 1 H, m), 2.81 (2 H, br d, J=1 1.53 Hz), 6.87 (2 H, br s), 7.81 ( 1 H, dd, J=8.78, 1.37 Hz), 8.07 (1 H, d, J=8.51 Hz), 8.12 (1 H, s), 8.49 (1 H, d, J=12.62 Hz), 8.77 (2 H, s), 9.09 ( 1 H, s), 10.43 (1 H, br s); ESIMS found for C 20 H 22 N 6 O

[0576] 2-Mo holino-N-(6-(pyridazin-3-yl)isoquinolin-3-yl)acetamide 66.

[0577] White solid (5.0 mg, 0.014 mmol, 7.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.56 - 2.63 (4 H, m), 3.27 (2 H, s), 3.63 - 3.69 (4 H, m), 8.05 (1 H, dd, J=8.65, 1.78 Hz), 8.21 (1 H, dd, J=5.35, 2.61 Hz), 8.25 (1 H, d, J=8.78 Hz), 8.59 (1 H, s), 8.63 (1 H, s), 9.24 (1 H, s), 9.37 ( 1 H, dd, J=5.21, 1.10 Hz), 9.83 ( 1 H, dd, J=2.47, 1.10 Hz), 10.16 (1 H, s); ESIMS found for Ci9Hi 9 N 5 0 2 mlz 350.2 (M+ l).

[0578] 2-Mo holino-N-(6-(pyridazin-4-yl)isoquinolin-3-yl)acetamide 67.

[0579] Beige solid (4.0 mg, 0.01 1 mmol, 6.7% yield). ¾ NMR (499 MHz,

CHLOROFORM- ) δ ppm 2.63 - 2.73 (4 H, m), 3.26 (2 H, s), 3.81 - 3.89 (4 H, m), 7.64 ( 1 H, dd, J=8.64, 4.80 Hz), 8.05 ( 1 H, dd, J=8.65, 1.23 Hz), 8.09 ( 1 H, d, J=8.51 Hz), 8.37 (1 H, dd, J=8.64, 1.51 Hz), 8.44 ( 1 H, s), 8.73 (1 H, s), 9.09 (1 H, s), 9.26 (1 H, dd, J=4.80, 1.23 Hz), 9.71 ( 1 H, br s); ESIMS found for Ci9Hi 9 N 5 0 2 mlz 350.1 (M+l).

[0580] N-(6-(Pyridazin-4-yl)isoquinolin-3-yl)-2-(pyrrolidin-l-yl)ac etamide 68.

[0581] White solid (25.0 mg, 0.075 mmol, 28.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ρρηι 1.78 (4 H, dt, J=6.86, 3.16 Hz), 2.63 - 2.71 (4 H, m), 3.38 (2 H, s), 8.05 (1 H, dd, J=8.51, 1.65 Hz), 8.20 (1 H, dd, J=5.49, 2.47 Hz), 8.25 (1 H, d, J=8.51 Hz), 8.59 ( 1 H, d, J=0.82 Hz), 8.62 ( 1 H, s), 9.23 (1 H, s), 9.36 (1 H, dd, J=5.35, 1.24 Hz), 9.83 ( 1 H, dd, J=2.61, 1.23 Hz), 10.08 (1 H, s); ESIMS found for Ci 9 Hi

69

[0582] fr «5-3-Mo holino-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)cyclobutane-l - carboxamide 69.

[0583] White solid (16.0 mg, 0.041 mmol, 21.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.08 - 2.17 (2 H, m), 2.23 - 2.32 (6 H, m), 2.91 (1 H, quin, J=7.27 Hz), 3.25 - 3.30 (1 H, m), 3.59 (4 H, t, J=4.53 Hz), 8.16 - 8.23 ( 1 H, m), 8.25 - 8.30 (1 H, m), 8.65 ( 1 H, s), 8.68 (1 H, s), 8.71 (1 H, d, J=2.47 Hz), 8.82 (1 H, dd, J=2.47, 1.65 Hz), 9.19 (1 H, s), 9.48 (1 H, d, J=1.37 Hz), 10.55 ( 1 H, s); ESIMS found for C 2 2H23N 5 0 2 mlz 390.2 (M+l).

70

[0584] fr «5-4-(Dimethylamino)-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)cy clohexane- 1 -carboxamide 70.

[0585] Off-white solid (98.0 mg, 0.248 mmol, 46.7% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.14 - 1.25 (2 H, m), 1.49 (2 H, qd, J=12.72, 3.02 Hz), 1.84 - 1.90 (2 H, m), 1.90

- 1.97 (2 H, m), 2.1 1 - 2.17 ( 1 H, m), 2.18 (6 H, s), 2.45 - 2.49 (1 H, m), 8.16 - 8.22 (1 H, m), 8.23

- 8.29 (1 H, m), 8.61 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 ( 1 H, dd, J=2.33, 1.51 Hz), 9.20 (1 H, s), 9.47 ( 1 H, d, J=1.37 Hz), 10.55 ( 1 H, s); ESIMS found for C 22 H 25 N 5 0 mlz 376.2 (M+ l).

71

[0586] ίΓ «5-4-Μο 1ιο1ίηο-Ν-(6-(ργΓαζίη-2-γ1)ί8ο ηίηο1ίη-3 -yl)cyclohexane- 1 - carboxamide 71.

[0587] White solid (55.0 mg, 0.125 mmol, 52.4% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 1.17 - 1.29 (2 H, m), 1.44 - 1.57 (2 H, m), 1.92 (4 H, br t, J=13.58 Hz), 2.16 - 2.27 (1 H, m), 2.45 - 2.49 (4 H, m), 2.53 (1 H, br s), 3.52 - 3.60 (4 H, m), 8.16 - 8.21 (1 H, m), 8.23 - 8.29 (1 H, m), 8.61 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.47, 1.65 Hz), 9.20 (1 H, s), 9.47 (1 H, d, J=1.65 for C24¾ 7 N 5 02 m > z 418 2 ( Μ+ 1

72

[0588] trans-4-((3 -Fluoroazetidin- 1 -yl)methyl)-N-(6-(pyrazin-2-yl)isoquinolin-3 - yl)cyclohexane-l -carboxamide 72.

[0589] White solid (50.0 mg, 0.113 mmol, 47.6% yield). 'HNMR (499 MHz, DMSO- d 6 ) 5 ppm 0.91 (2 H, qd, J=12.76, 3.16 Hz), 1.22 - 1.34 (1 H, m), 1.39 - 1.52 (2 H, m), 1.80 (2 H, br dd, J=12.90, 2.74 Hz), 1.87 (2 H, br d, J=10.70 Hz), 2.29 (2 H, d, J=6.59 Hz), 2.52 - 2.55 (1 H, m), 2.96 - 3.08 (2 H, m), 3.48 - 3.60 (2 H, m), 5.12 (1 H, dquin, J=58.00, 5.00 Hz), 8.15 - 8.22 (1 H, m), 8.24 - 8.30 (1 H, m), 8.61 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.33, 1.51 Hz), 9.19 (1 H, s), 9.47 (1 H, d, J=1.37 Hz), 10.53 (1 H, s); ESIMS found for C 24 H 26 FN 5 O mlz 420.2 (M+l).

73

[0590] fr «5-4-((4-Methylpiperazin-l-yl)methyl)-N-(6-(pyrazin-2-yl)is oquinolin-3- yl)cyclohexane-l -carboxamide 73.

[0591] White solid (48.0 mg, 0.103 mmol, 45.7% yield). ¾NMR (499 MHz, DMSO- d 6 ) 5 ppm 0.83 - 0.96 (2 H, m), 1.40 - 1.55 (3 H, m), 1.79 - 1.92 (4 H, m), 2.08 (2 H, d, J=7.14 Hz), 2.14 (3 H, s), 2.31 (8 H, br s), 2.52 - 2.58 (1 H, m), 8.15 - 8.21 (1 H, m), 8.23 - 8.31 (1 H, m), 8.61 ( 1 H, s), 8.66 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 ( 1 H, dd, J=2.47, 1.37 Hz), 9.19 ( 1 H, s), 9.47 ( 1 H, d, .7=1.37 Hz), 10.53 (1 H, s); ESIMS found for CzeH^NeO mlz 445.3 (M+l).

[0592] N-(6-(Pyrazin-2-yl)isoquinolin-3-yl)-2-(pyrrolidin-l -yl)acetamide 74.

[0593] White solid (16.0 mg, 0.046 mmol, 15.2% yield). 'HNMR (499 MHz, DMSO- d 6 ) δ ppm 1.79 (4 H, dt, J=6.72, 3.22 Hz), 2.61 - 2.71 (4 H, m), 3.38 (2 H, s), 8.22 (1 H, d, J=8.51 Hz), 8.27 - 8.33 (1 H, m), 8.62 ( 1 H, s), 8.70 ( 1 H, d, J=2.47 Hz), 8.74 (1 H, s), 8.81 ( 1 H, dd, J=2.33, 1.51 Hz), 9.21 (1 H, s), 9.49 ( 1 H, d, J=1.37 Hz), 10.05 ( 1 H, s); ESIMS found for CigHisNsO mlz 334.2 (M+ l).

75

[0594] 1 -Methyl -N-(6 -(pyrazin-2 -yl)isoquinolin-3 -yl)piperidine -4 -carboxamide 75.

[0595] White solid (820.0 mg, 2.36 mmol, 41.1% yield) . ¾ NMR (499 MHz, DMSO- 6) 5 ppm 1.63 - 1.75 (2 H, m), 1.76 - 1.82 (2 H, m), 1.84 - 1.93 (2 H, m), 2.16 (3 H, s), 2.51 - 2.57 ( 1 H, m), 2.82 (2 H, br d, J=1 1.25 Hz), 8.15 - 8.22 (1 H, m), 8.24 - 8.30 (1 H, m), 8.62 ( 1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.33, 1.51 Hz), 9.20 (1 H, s), 9.47 (1 H, d, J=1.37 Hz), 10.59 (1 H, s); ESIM mlz 348.1 (M+l).

[0596] 4-Fluoro-l -methyl -N-(6-(pyrazin-2 -yl)isoquinolin-3-yl)piperidine-4- carboxamide 76.

[0597] Off-white solid (56.0 mg, 0.146 mmol, 36.9% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.93 - 2.02 (2 H, m), 2.07 - 2.21 (4 H, m), 2.22 (3 H, s), 2.72 - 2.78 (2 H, m), 8.25 ( 1 H, d, J=8.78 Hz), 8.31 - 8.35 ( 1 H, m), 8.58 (1 H, s), 8.71 (1 H, d, J=2.47 Hz), 8.75 (1 H, s), 8.80 - 8.84 (1 H, m), 9.26 ( 1 H, s), 9.48 (1 H, d, J=1.37 Hz), 10.03 (1 H, d, J=3.84 Hz); ESIMS found for C 20 H 20 FN 5 O mlz 366.15 (M+l).

77

[0598] l-(2-Fluoroethyl)-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)piperi dine-4- carboxamide 77.

[0599] White solid (55.0 mg, 0.138 mmol, 54.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ρρηι 1.69 (2 H, qd, J=12.30, 3.70 Hz), 1.77 - 1.85 (2 H, m), 2.01 - 2.10 (2 H, m), 2.52 - 2.58 ( 1 H, m), 2.62 (2 H, dt, J=28.30, 5.00 Hz), 2.95 (2 H, br d, J=11.53 Hz), 4.54 (2 H, dt, J=47.80, 5.00 Hz), 8.16 - 8.22 (1 H, m), 8.27 (1 H, dd, J=8.51, 1.65 Hz), 8.63 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.33, 1.51 Hz), 9.20 (1 H, s), 9.47 (1 H, d, J=1.65 Hz), 10.58 ( 1 H, s); ESIMS found for C 2 iH 2 2

78

[0600] l-(Oxetan-3-yl)-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)piperidi ne-4- carboxamide 78.

[0601] White solid (37.0 mg, 0.090 mmol, 39.1% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.64 - 1.75 (2 H, m), 1.75 - 1.88 (4 H, m), 2.54 - 2.63 (1 H, m), 2.72 - 2.80 (2 H, m), 3.39 ( 1 H, quin, J=6.38 Hz), 4.43 (2 H, t, J=6.17 Hz), 4.53 (2 H, t, J=6.60 Hz), 8.16 - 8.22 ( 1 H, m), 8.24 - 8.30 (1 H, m), 8.63 (1 H, s), 8.68 (1 H, s), 8.70 ( 1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.47, 1.65 Hz), 9.20 (1 H, s), 9.47 (1 H, d, J=1.37 Hz), 10.60 ( 1 H, s); ESIMS found for C 2 2H23N 5 0 2 mlz 390.2 (M+l).

[0602] 2-Mo holino-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)acetamide 79.

[0603] Light yellow solid (52.0 mg, 0.141 mmol, 49.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.54 - 2.61 (4 H, m), 3.27 (2 H, s), 3.61 - 3.70 (4 H, m), 8.18 - 8.25 ( 1 H, m), 8.30 (1 H, dd, J=8.51, 1.65 Hz), 8.62 (1 H, s), 8.71 (1 H, d, J=2.47 Hz), 8.74 ( 1 H, s), 8.81 ( 1 H, dd, J=2.47, 1.65 Hz), 9.22 ( 1 H, s), 9.49 (1 H, d, J=1.37 Hz), 10.14 ( 1 H, s); ESIMS found for Ci9Hi 9 N 5 0 2 mlz 350.15 (M+l).

80

[0604] 1 -Methyl -N-(6-(pyrazin-2-yl)isoquinolin -3 -yl)azepane-4-carboxamide 80.

[0605] White solid (20.0 mg, 0.055 mmol, 38.4% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 1.56 - 1.67 (1 H, m), 1.72 - 1.83 (2 H, m), 1.83 - 1.94 (3 H, m), 2.29 (3 H, s), 2.52 - 2.60 (3 H, m), 2.70 (1 H, td, J=6.24, 3.43 Hz), 2.84 - 2.94 (1 H, m), 8.16 - 8.22 (1 H, m), 8.23 - 8.29 (1 H, m), 8.61 (1 H, s), 8.67 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.47, 1.37 Hz), 9.19 (1 H, s), 9.47 (1 H, d, J=1.37 found for C2iH 23 N 5 0 mlz 362.2 (M+l).

[0606] l-Isobutyl-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)azepane-4-car boxamide 81.

[0607] Yellow-white solid (20.0 mg, 0.050 mmol, 34.4% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.87 (3 H, d, J=2.47 Hz), 0.88 (3 H, d, J=2.47 Hz), 1.52 - 1.64 (1 H, m), 1.65 - 1.72 (1 H, m), 1.72 - 1.83 (3 H, m), 1.83 - 1.95 (2 H, m), 2.15 - 2.24 (2 H, m), 2.55 - 2.62 (3 H, m), 2.70 (1 H, qd, J=6.50, 3.84 Hz), 2.83 - 2.92 (1 H, m), 8.16 - 8.21 (1 H, m), 8.24 - 8.29 (1 H, m), 8.61 (1 H, s), 8.66 (1 H, s), 8.70 (1 H, d, J=2.47 Hz), 8.81 (1 H, dd, J=2.47, 1.65 Hz), 9.19 (1 H, s), 9.47 (1 H, d, J=1.65 Hz), 10.56 (1 H, s); ESIMS found for mlz 404.2 (M+l).

82

[0608] l-(2-Hydroxy-2-methylpropyl)-N-(6-(pyrazin-2-yl)isoquinolin- 3-yl)azepane- 4-carboxamide 82.

[0609] Beige solid (10.0 mg, 0.024 mmol, 16.6% yield). ¾ NMR (499 MHz, METHANOLS) δ ppm 1.28 (4 H, s), 1.30 (3 H, s), 1.82 - 1.90 (1 H, m), 1.93 - 2.03 (2 H, m), 2.04 - 2.22 (3 H, m), 2.81 - 2.92 (2 H, m), 2.92 - 2.98 (1 H, m), 3.17 (2 H, br s), 8.13 (1 H, d, J=8.51 Hz), 8.23 (1 H, dd, J=8.51, 1.65 Hz), 8.54 (1 H, s), 8.59 (1 H, s), 8.61 (1 H, d, J=2.74 Hz), 8.75 (1 H, dd, J=2.33, 1.51 Hz), 9.09 (1 H, s), 9.29 (1 H, d, J=1.37 Hz); ESIMS found for CuR KsOi mlz 420.2 (M+l).

[0610] 4-(Moφholinomethyl)-N-(6-(pyrazin-2-yl)isoquinolin-3-yl)ben zamide 83.

[0611] Light yellow solid (35.0 mg, 0.082 mmol, 18.3% yield). ¾ NMR (500 MHz, DMSO- e) δ ppm 2.39 (4 H, br s), 3.56 (2 H, s), 3.60 (4 H, t, J=4.53 Hz), 7.47 (2 H, d, J=8.51 Hz), 8.07 (2 H, d, J=8.23 Hz), 8.21 - 8.29 (1 H, m), 8.29 - 8.37 (1 H, m), 8.68 - 8.80 (2 H, m), 8.74 - 8.75 (1 H, m), 8.82 (1 H, dd, J=2.47, 1.65 Hz), 9.29 (1 H, s), 9.51 (1 H, d, J=1.65 Hz), 10.90 (1 H, s); ESIMS found for C 2 5H23N 5

84

[0612] 2-(4-Methylpiperazin-l-yl)-N-(6-(pyrazin-2-yl)isoquinolin-3- yl)isonicotinamide 84.

[0613] White solid (45.0 mg, 0.104 mmol, 63.0% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.56 - 3.66 (4 H, m), 7.17 (1 H, dd, J=5.08, 1.24 Hz), 7.48 (1 H, s), 8.22 - 8.27 (1 H, m), 8.27 (1 H, s), 8.31 - 8.39 (1 H, m), 8.72 (1 H, d, J=2.47 Hz), 8.78 (2 H, s), 8.83 (1 H, dd, J=2.47, 1.65 Hz), 9.30 (1 H, s), 9.51 (1 H, d, J=1.37 Hz), 11.16 (1 H, s); ESIMS found for C24

85

[0614] N-(6-(6-(Methylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(4-met hylpiperazin- l-yl)isonicotinamide 85.

[0615] Brown solid (40.0 mg, 0.088 mmol, 30.1% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.37 - 2.47 (4 H, m), 2.96 (3 H, d, J=4.67 Hz), 3.52 - 3.66 (4 H, m), 7.17 (1 H, dd, J=5.08, 1.24 Hz), 7.19 - 7.24 (1 H, m), 7.48 (1 H, s), 7.97 (1 H, s), 8.19 (1 H, d, J=8.51 Hz), 8.24 - 8.32 (2 H, m), 8.50 (1 H, s), 8.65 (1 H, s), 8.73 (1 H, s), 9.26 (1 H, s), 11.12 (1 H, s); ESIMS found for CzsHzeNgO mlz 455.2 (M+l).

86

[0616] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)azetid ine-3- carboxamide 86.

[0617] Yellow solid (150.0 mg, 0.414 mmol, 59.8% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.26 (6 H, d, J=6.59 Hz), 3.35 - 3.43 ( 1 H, m), 3.46 - 3.58 (2 H, m), 3.72 - 3.84 (2 H, m), 4.15 - 4.27 (1 H, m), 7.1 1 ( 1 H, d, J=7.14 Hz), 7.93 (1 H, s), 8.07 - 8.15 (1 H, m), 8.15 - 8.22 (1 H, m), 8.45 ( 1 H, s), 8.54 ( 1 H, s), 8.59 (1 H, s), 9.15 (1 H, s), 10.53 ( 1 H, br s); ESIMS found for C 2 oH 2 2N 6 0 mlz 363.2

87

[0618] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)piperi dine-4- carboxamide 87.

[0619] Beige solid (340.0 mg, 0.871 mmol, 65.7% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.25 (6 H, d, J=6.31 Hz), 1.54 (2 H, qd, J=12.12, 3.98 Hz), 1.67 - 1.76 (2 H, m), 2.06 (1 H, br s), 2.43 - 2.49 (2 H, m), 2.59 - 2.70 ( 1 H, m), 2.98 (2 H, br d, J=12.62 Hz), 4.20 ( 1 H, dq, J=13.28, 6.55 Hz), 7.1 1 ( 1 H, d, J=7.41 Hz), 7.92 (1 H, s), 8.07 - 8.14 (1 H, m), 8.14 - 8.20 ( 1 H, m), 8.43 (1 H, s), 8.52 ( 1 H, s), 8.56 (1 H, s), 9.14 ( 1 H, s), 10.48 (1 H, s); ESIMS found for C22H26N6O mlz 391.2 (M+ l).

88

[0620] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3 -yl)- 1 -methylpiperidine-

4-carboxamide 88. [0621] Yellow solid (40.0 mg, 0.099 mmol, 22.1% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.26 (6 H, d, J=6.31 Hz), 1.62 - 1.75 (2 H, m), 1.75 - 1.83 (2 H, m), 1.88 (2 H, td, J=l 1.53, 2.20 Hz), 2.17 (3 H, s), 2.51 - 2.58 ( 1 H, m), 2.76 - 2.87 (2 H, m), 4.15 - 4.26 (1 H, m), 7.07 ( 1 H, br d, J=7.14 Hz), 7.93 (1 H, s), 8.11 (1 H, s), 8.14 - 8.21 ( 1 H, m), 8.42 (1 H, s), 8.51 (1 H, s), 8.56 (1 H, s), 9.14 ( 1 H, s), 10.50 ( 1 H, s); ESIMS found for CzsHzsNeO mlz 405.0 (M+1).

89

[0622] l-(2-Fluoroethyl)-N-(6-(6-(isopropylamino)pyrazin-2-yl)isoqu inolin-3-yl) piperidine-4-carboxamide 89.

[0623] Beige solid (50.0 mg, 0.1 15 mmol, 44.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.25 (6 H, d, J=6.31 Hz), 1.69 (2 H, qd, J=12.21, 3.98 Hz), 1.77 - 1.85 (2 H, m), 2.05 (2 H, td, J=1 1.66, 1.92 Hz), 2.52 - 2.57 ( 1 H, m), 2.62 (2 H, dt, J=28.40, 5.00 Hz), 2.91 - 3.00 (2 H, m), 4.20 ( 1 H, dq, J=13.28, 6.55 Hz), 4.54 (2 H, dt, J=48.10, 5.00 Hz), 7.1 1 (1 H, d, J=7.41 Hz), 7.92 (1 H, s), 8.08 - 8.14 (1 H, m), 8.14 - 8.21 (1 H, m), 8.43 ( 1 H, s), 8.52 (1 H, s), 8.57 ( 1 H, s), 9.15 ( 1 H, s), 10.55 (1 H, s); ESIMS found for C 24 H 29 FN 6 O mlz 437.2 (M+1).

90

[0624] l-Isopropyl-N-(6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin -3-yl) piperidine-4-carboxamide 90.

[0625] Light yellow solid (94.0 mg, 0.217 mmol, 84.9% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 0.97 (6 H, d, J=6.59 Hz), 1.25 (6 H, d, J=6.31 Hz), 1.64 (2 H, qd, J=12.17, 3.29 Hz), 1.79 (2 H, br d, J=1 1.80 Hz), 2.05 - 2.17 (2 H, m), 2.51 - 2.57 (1 H, m), 2.61 - 2.74 ( 1 H, m), 2.80 - 2.88 (2 H, m), 4.15 - 4.26 (1 H, m), 7.1 1 (1 H, br d, J=7.14 Hz), 7.92 ( 1 H, s), 8.06 - 8.13 ( 1 H, m), 8.13 - 8.21 ( 1 H, m), 8.43 (1 H, s), 8.52 (1 H, s), 8.57 (1 H, s), 9.15 ( 1 H, s), 10.52 (1 H, s); ESIMS found for CzsH^NeO mlz 433.3 (M+1).

91

[0626] l-Isopentyl-N-(6-(6-(isopropylamino)pyrazin-2-yl)isoquinolin -3-yl) piperidine-4-carboxamide 91.

[0627] Beige solid (74.0 mg, 0.161 mmol, 57.0% yield). ¾NMR(499 MHz, DMSO- d 6 ) δρρηι 0.88 (6 H, d,J=6.86 Hz), 1.25 (6 H, d,J=6.31 Hz), 1.29 - 1.38 (2 H, m), 1.51 - 1.62 (1 H, m), 1.63 - 1.73 (2 H, m), 1.78 (2 H, br d, J=10.98 Hz), 1.83 - 1.93 (2 H, m), 2.21 - 2.32 (2 H, m), 2.52 - 2.60 (1 H, m), 2.90 (2 H, br d, J=11.53 Hz), 4.14 - 4.26 (1 H, m), 7.11 (1 H, d, J=7.41 Hz), 7.92 (1 H, s), 8.11 (1 H, s), 8.14 - 8.21 (1 H, m), 8.43 (1 H, s), 8.52 (1 H, s), 8.56 (1 H, s), 9.14 (1 H, s), 10.53 (1 H, s); ESIM 461.3 (M+1).

92

[0628] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)-3-(pi peridin-4-yl) benzamide 92.

[0629] Beige solid (70.0 mg, 0.150 mmol, 85.0% yield). ¾NMR(499 MHz, DMSO- d 6 ) δ ppm 1.26 (6 H, d, J=6.31 Hz), 1.61 (2 H, qd, J=12.21, 3.70 Hz), 1.75 (2 H, br d, J=l 1.53 Hz), 2.55 - 2.64 (2 H, m), 2.64 - 2.73 (1 H, m), 3.05 (2 H, br d, J=11.80 Hz), 4.22 (1 H, dq, J=13.21, 6.67 Hz), 7.13 (1 H, d, J=7.41 Hz), 7.40 - 7.50 (2 H, m), 7.86 - 7.92 (1 H, m), 7.94 (1 H, s), 8.01 (1 H, s), 8.13 - 8.20 (1 H, m), 8.20 - 8.28 (1 H, m), 8.47 (1 H, s), 8.61 (1 H, s), 8.73 (1 H, s), 9.24 (1 H, s), 10.95 (1 H, s); ESIM 467.25 (M+1).

93

[0630] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)-3-(l- methylpiperidin-4-yl)benzamide 93. [0631] Yellow solid (29.0 mg, 0.060 mmol, 48.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.26 (6 H, d, J=6.59 Hz), 1.71 - 1.85 (4 H, m), 1.99 (2 H, td, J=l 1.05, 3.43 Hz), 2.21 (3 H, s), 2.53 - 2.61 ( 1 H, m), 2.90 (2 H, br d, J=11.53 Hz), 4.22 (1 H, dq, J=13.34, 6.53 Hz),

7.13 (1 H, d, J=7.14 Hz), 7.39 - 7.51 (2 H, m), 7.85 - 7.92 (1 H, m), 7.94 (1 H, s), 8.03 ( 1 H, s),

8.14 - 8.21 (1 H, m), 8.21 - 8.24 ( 1 H, m), 8.47 (1 H, s), 8.61 ( 1 H, s), 8.73 ( 1 H, s), 9.24 ( 1 H, s), 10.94 ( 1 H, s); ESIMS found for mlz 481.3 (M+l).

[0632] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3 -yl)- 1 -(piperidin-4-yl)- lH-l,2,3-triazole-4-carboxamide 94.

[0633] Beige solid (176.0 mg, 0.385 mmol, 55.0% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.26 (6 H, d, J=6.31 Hz), 1.89 (2 H, qd, J=1 1.94, 3.98 Hz), 2.08 (2 H, br dd, J=1 1.80, 2.20 Hz), 2.58 - 2.72 (2 H, m), 3.02 - 3.12 (2 H, m), 4.21 ( 1 H, dq, J=13.34, 6.53 Hz), 4.66 ( 1 H, tt, J=1 1.66, 4.12 Hz), 7.13 (1 H, d, J=7.14 Hz), 7.94 (1 H, s), 8.14 - 8.21 ( 1 H, m), 8.21 - 8.30 ( 1 H, m), 8.48 ( 1 H, s), 8.62 ( 1 H, s), 8.65 ( 1 H, s), 8.96 ( 1 H, s), 9.23 (1 H, s), 10.12 (1 H, br s); ESIMS found for C24H27N9O mlz 458.3 (M+l).

95

[0634] 1 -( 1 -(2-Fluoroethyl)piperidin-4-yl)-N-(6-(6-(isopropylamino)pyra zin-2-yl) isoquinolin-3-yl)-lH-l,2,3-triazole-4-carboxamide 95.

[0635] Beige solid (60.0 mg, 0.1 19 mmol, 77.9% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.26 (6 H, d, J=6.31 Hz), 2.02 - 2.19 (4 H, m), 2.25 - 2.33 (2 H, m), 2.69 (2 H, dt, J=28.60, 5.00 Hz), 3.02 (2 H, br d, J=l 1.80 Hz), 4.21 (1 H, dq, J=13.34, 6.53 Hz), 4.48 - 4.67 (3 H, m), 7.13 ( 1 H, d, J=7.14 Hz), 7.93 (1 H, s), 8.14 - 8.21 (1 H, m), 8.21 - 8.28 ( 1 H, m), 8.48 (1 H, s), 8.62 (1 H, s), 8.65 ( 1 H, s), 9.00 (1 H, s), 9.23 (1 H, s), 10.1 1 ( 1 H, s); ESIMS found for C 2 6H 3 oFN 9 0 mlz 504.3 (M+l).

96

[0636] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3 -yl)- 1 -( 1 - isopropylpiperidin-4-yl)-lH-l,2,3-triazole-4-carboxamide 96.

[0637] Beige solid (66.0 mg, 0.126 mmol, 70.0% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.26 (6 H, d, J=6.59 Hz), 1.92 - 2.07 (2 H, m), 2.12 - 2.20 (2 H, m), 2.27 - 2.37 (2 H, m), 2.77 ( 1 H, quin, J=6.59 Hz), 2.92 (2 H, br d, J=12.08 Hz), 4.21 ( 1 H, dq, J=13.28, 6.55 Hz), 4.56 ( 1 H, tt, J=1 1.53, 4.25 Hz), 7.13 ( 1 H, d, J=7.41 Hz), 7.93 ( 1 H, s), 8.15 - 8.21 (1 H, m), 8.21 - 8.28 ( 1 H, m), 8.47 (1 H, s), 8.62 (1 H, s), 8.65 (1 H, s), 8.98 (1 H, s), 9.23 ( 1 H, s), 10.10 (1 H, s); ESIMS found for C 2 7H 33 N 9 0

[0638] N-(6-(6-(Isopropylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(4- methylpiperazin-1 -yl)isonicotinamide 97.

[0639] Beige solid (96.0 mg, 0.199 mmol, 56.4% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.26 (6 H, d, J=6.59 Hz), 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.56 - 3.65 (4 H, m), 4.21 (1 H, dq, J=13.38, 6.61 Hz), 7.14 ( 1 H, d, J=7.14 Hz), 7.17 (1 H, dd, J=5.21, 1.10 Hz), 7.48 ( 1 H, s), 7.94 (1 H, s), 8.16 - 8.22 (1 H, m), 8.22 - 8.26 ( 1 H, m), 8.26 (1 H, d, J=5.21 Hz), 8.47 ( 1 H, s), 8.62 (1 H, s), 8.72 (1 H, s), 9.25 (1 H, s), 1 1.14 ( 1 H, s); ESIMS found for C 27 H 3 oN 8 0 mlz 483.3 (M+l).

[0640] N-(6-(6-(tert-Butylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(3 - (dimethylamino)azetidin-l -yl)isonicotinamide 98.

[0641] Yellow solid (73.8 mg, 0.149 mmol, 40.2% yield). ¾ NMR (499 MHz, DMSO- e) δ ρρηι 1.52 (9 H, s), 2.13 (6 H, s), 3.17 - 3.25 (1 H, m), 3.81 (2 H, dd, J=8.37, 5.35 Hz), 4.07 (2 H, t, J=7.55 Hz), 6.99 (1 H, s), 7.05 (1 H, s), 7.16 (1 H, dd, J=5.21, 1.37 Hz), 7.97 (1 H, s), 8.19 - 8.24 (2 H, m), 8.47 (1 H, s), 8.58 (1 H, s), 8.70 (1 H, s), 9.25 (1 H, s), 11.07 (1 H, s); ESIMS found for C 2 8H 3 2N 8 0 mlz 497.3 (M+l).

[0642] N-(6-(6-(tert-Butylamino)pyrazin-2-yl)isoquinolin-3-yl)-r-me thyl-r,2',3',6'- tetrahydro-[2,4'-bipyridine]-4-carboxamide 99.

[0643] Yellow solid (41.5 mg, 0.089 mmol, 20.0% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.52 (9 H, s), 2.31 (3 H, s), 2.57 - 2.63 (2 H, m), 2.63 - 2.70 (2 H, m), 3.11 (2 H, br d, J=3.02 Hz), 6.89 (1 H, t, J=3.43 Hz), 7.00 (1 H, s), 7.79 (1 H, dd, J=4.94, 1.37 Hz), 7.97 (1 H, s), 8.18 (1 H, s), 8.20 - 8.28 (2 H, m), 8.47 (1 H, s), 8.59 (1 H, s), 8.72 (1 H, d, J=5.21 Hz), 8.73 (1 H, s), 9.26 (1 H, s), 11.34 mlz 494.3 (M+l).

100

[0644] N-(6-(6-(teri-Butylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(4 - methylpiperazin-1 -yl)isonicotinamide 100.

[0645] White solid (40.0 mg, 0.081 mmol, 18.7% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 1.52 (9 H, s), 2.24 (3 H, s), 2.38 - 2.49 (4 H, m), 3.53 - 3.70 (4 H, m), 6.99 (1 H, s), 7.17 (1 H, dd, J=5.08, 1.23 Hz), 7.48 (1 H, s), 7.97 (1 H, s), 8.14 - 8.25 (2 H, m), 8.26 (1 H, d, J=5.21 Hz), 8.47 (1 H, s), 8.58 (1 H, s), 8.71 ( 1 H, s), 9.25 (1 H, s), 11.13 (1 H, s); ESIMS found for C 2 8H 3 2N 8 0 mlz 497.3 (M+l).

101

[0646] N-(6-(6-(teri-Butylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(4 - isopropylpiperazin- 1 -yl)isonicotinamide 101.

[0647] Yellow solid (7.7 mg, 0.015 mmol, 4.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.02 (6 H, d, J=6.59 Hz), 1.52 (9 H, s), 2.52 - 2.60 (4 H, m), 2.70 ( 1 H, dt, J=13.10, 6.48 Hz), 3.53 - 3.63 (4 H, m), 6.99 ( 1 H, s), 7.16 (1 H, dd, J=5.21, 1.37 Hz), 7.46 (1 H, s), 7.97 ( 1 H, s), 8.17 - 8.25 (2 H, m), 8.26 ( 1 H, d, J=4.94 Hz), 8.47 (1 H, s), 8.58 (1 H, s), 8.71 ( 1 H, s), 9.25 (1 H, s), 1 1.13 (1 H, s); ESIMS (M+l).

102

[0648] N-(6-(6-(teri-Butylamino)pyrazin-2-yl)isoquinolin-3-yl)-2- mo holinoisonicotinamide 102.

[0649] Yellow solid (2.6 mg, 0.005 mmol, 2.1% yield). ¾ NMR (500 MHz, DMSO- d 6 ) δ ppm 1.52 (9 H, s), 3.52 - 3.63 (4 H, m), 3.69 - 3.82 (4 H, m), 7.01 ( 1 H, s), 7.22 ( 1 H, dd, J=5.08, 1.23 Hz), 7.49 (1 H, s), 7.97 (1 H, s), 8.18 - 8.25 (2 H, m), 8.29 (1 H, d, J=5.21 Hz), 8.47 ( 1 H, s), 8.59 (1 H, s), 8.71 ( 1 H, s), 9.26 (1 H, s), 1 1.16 (1 H, s); ESIMS found for C27H29N7O2 mlz 484.3 (M+l).

103

[0650] N-(6-(6-(((3-Fluoroazetidin-3-yl)methyl)amino)pyrazin-2-yl)i soquinolin-3- yl)cyclopropanecarboxamide 103. [0651] White solid (25.0 mg, 0.064 mmol, 62.8% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 0.79 - 0.86 (2 H, m), 0.86 - 0.91 (2 H, m), 2.03 - 2.13 (1 H, m), 3.54 (2 H, br dd, J=14.13, 10.02 Hz), 3.57 - 3.68 (2 H, m), 3.97 (2 H, dd, J=24.00, 6.10 Hz), 7.44 (1 H, br t, J=5.76 Hz), 8.06 ( 1 H, s), 8.10 - 8.16 (1 H, m), 8.20 ( 1 H, dd, J=8.51, 1.65 Hz), 8.50 ( 1 H, s), 8.54 (2 H, s), 9.16 ( 1 H, s), 10.87 (1 H, s); ESIMS found for C 2 iH 2 iFN 6 0 mlz 393.0 (M+l).

104

[0652] 4-fluoro-N-(6-(6-(((3-Fluoroazetidin-3-yl)methyl)amino)pyraz in-2-yl) isoquinolin-3-yl)benzamide 104.

[0653] White solid (28.0 mg, 0.063 mmol, 62.3% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 3.54 - 3.74 (4 H, m), 3.99 (2 H, dd, J=23.60, 5.80 Hz), 7.31 - 7.40 (2 H, m), 7.46 ( 1 H, br t, J=5.76 Hz), 8.08 (1 H, s), 8.13 - 8.19 (2 H, m), 8.19 (1 H, s), 8.26 (1 H, dd, J=8.78, 1.65 Hz), 8.54 ( 1 H, s), 8.65 ( 1 H, s), 8.72 ( 1 H, s), 9.24 (1 H, s), 10.92 (1 H, s); ESIMS found for C 24 H 20 F 2 N 6 O mlz 446.9 (M+l).

[0654] N-(6-(6-(Piperidin-4-ylamino)pyrazin-2-yl)isoquinolin-3-yl)

cyclobutanecarboxamide 105.

[0655] Beige solid (100.0 mg, 0.249 mmol, 59.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.30 - 1.43 (2 H, m), 1.80 - 1.89 ( 1 H, m), 1.91 - 2.01 (3 H, m), 2.09 - 2.19 (2 H, m), 2.21 - 2.33 (2 H, m), 2.57 - 2.67 (2 H, m), 2.95 - 3.05 (2 H, m), 3.40 - 3.48 ( 1 H, m), 3.87 - 3.99 ( 1 H, m), 7.16 (1 H, br d, J=7.41 Hz), 7.95 (1 H, s), 8.08 - 8.14 (1 H, m), 8.14 - 8.20 ( 1 H, m), 8.44 ( 1 H, s), 8.50 ( 1 H, s), 8.58 ( 1 H, s), 9.14 (1 H, s), 10.43 (1 H, s); ESIMS found for CzsHzeNeO mlz 403.2 (M+l).

106

[0656] 3,3-Difluoro-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl)isoqu inolin-3-yl) cyclobutane-l-carboxamide 106.

[0657] Beige solid (78.0 mg, 0.178 mmol, 42.6% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.31 - 1.44 (2 H, m), 1.90 - 2.00 (2 H, m), 2.58 - 2.67 (2 H, m), 2.76 - 2.92 (4 H, m), 2.96 - 3.04 (2 H, m), 3.87 - 4.01 (1 H, m), 7.16 ( 1 H, br d, J=7.14 Hz), 7.95 (1 H, s), 8.1 1 - 8.16 ( 1 H, m), 8.16 - 8.21 ( 1 H, m), 8.44 ( 1 H, s), 8.53 ( 1 H, s), 8.58 (1 H, s), 9.17 (1 H, s), 10.82 (1 H, s); ESIMS found for C23H24F2N6O mlz 439.2 (M+1).

[0658] N-(6-(6-(Piperidin-4-ylamino)pyrazin-2-yl)isoquinolin-3-yl)

cyclopentanecarboxamide 107.

[0659] Light yellow solid (78.0 mg, 0.187 mmol, 55.3% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.36 (2 H, qd, J=l 1.25, 3.57 Hz), 1.51 - 1.62 (2 H, m), 1.65 - 1.82 (4 H, m), 1.84 - 1.91 (2 H, m), 1.91 - 1.99 (2 H, m), 2.56 - 2.66 (2 H, m), 2.93 - 3.04 (3 H, m), 3.88 - 3.99 (1 H, m), 7.15 (1 H, br d, J=7.41 Hz), 7.95 (1 H, s), 8.07 - 8.18 (2 H, m), 8.43 (1 H, s), 8.49 (1 H, s), 8.56 ( 1 H, s), 9.15 ( 1 H, s), 10.55 (1 H, s); ESIMS found for Cz^sNeO mlz 417.2 (M+1).

108

[0660] 4,4-Difluoro-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl)isoqu inolin-3-yl) cyclohexane - 1 -carboxamide 108. [0661] White solid (97.0 mg, 0.208 mmol, 76.0% yield). ¾NMR(499 MHz, DMSO- £¾)5ppm 1.32- 1.44 (2H,m), 1.66- 1.92 (4H, m), 1.92-2.01 (4 H, m), 2.07 -2.20 (2H, m), 2.59 - 2.67 (2 H, m), 2.69 - 2.77 (1 H, m), 2.97 - 3.06 (2 H, m), 3.88 - 4.01 (1 H, m), 7.16 (1 H, br d, J=7.41 Hz), 7.95 (1 H, s), 8.09 - 8.14 (1 H, m), 8.14 - 8.20 (1 H, m), 8.43 (1 H, s), 8.50 (1 H, s), 8.55 (1 H, s), 9.16 (1 H, s), 10.64 for CzsHzsFzNeO mlz 467.0 (M+1).

109

[0662] N-(6-(6-(Piperidin-4-ylamino)pyrazin-2-yl)isoquinolin-3-yl)t etrahyd] pyran-4-carboxamide 109.

[0663] Beige solid (20.0 mg, 0.046 mmol, 82.1% yield). ¾NMR(500 MHz, DMSO- 6)5ppm 1.36(2 H, qd, J=l 1.43, 3.84 Hz), 1.65 - 1.80 (4 H, m), 1.95 (2 H, br dd, J=12.21, 2.61 Hz), 2.57 - 2.67 (2 H, m), 2.78 - 2.88 (1 H, m), 2.94 - 3.04 (2 H, m), 3.33 - 3.42 (2 H, m), 3.86 - 3.97 (3 H, m), 7.15 (1 H, br d, J=7.41 Hz), 7.95 (1 H, s), 8.09 - 8.19 (2 H, m), 8.43 (1 H, s), 8.50 (1 H, s), 8.56 (1 H, s), 9.15 (1 H, s), 10.58 (1 H, s); ESIMS found for C24¾8N 6 0 2 m > z 4 0 ( Μ+1

110

[0664] 4-Fluoro-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl)isoquinol in-3-yl) benzamide 110.

[0665] Off-white solid (31.0 mg, 0.070 mmol, 40.0% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.33 - 1.49 (2 H, m), 1.92-2.03 (2 H, m), 2.64 -2.75 (2 H, m), 3.00-3.09 (2 H, m), 3.93 - 4.05 (1 H, m), 7.20 (1 H, d, J=7.14 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.96 (1 H, s), 8.12 - 8.25 (4 H, m), 8.47 (1 H, s), 8.59 (1 H, s), 8.70 (1 H, s), 9.24 (1 H, s), 10.98 (1 H, s); ESIMS found for C 25 H 23 FN 6 O mlz 443.2 (M+1).

111

[0666] N-(6-(6-(((3S,4S)-3-Fluoropiperidin-4-yl)amino)pyrazin-2-yl) isoquinolin-3- yl)cyclopropanecarboxamide 111.

[0667] White solid (39.0 mg, 0.096 mmol, 74.8% yield). 'HNMR (499 MHz, DMSO- d 6 ) δ ppm 0.79 - 0.86 (2 H, m), 0.86 - 0.92 (2 H, m), 1.38 - 1.51 (1 H, m), 2.03 - 2.14 (2 H, m), 2.55 - 2.73 (3 H, m), 2.89 - 2.97 ( 1 H, m), 4.18 - 4.28 (1 H, m), 4.40 - 4.56 (1 H, m), 7.37 ( 1 H, br d, J=7.68 Hz), 8.00 ( 1 H, s), 8.09 - 8.18 (2 H, m), 8.47 (1 H, s), 8.48 (1 H, s), 8.53 ( 1 H, s), 9.16 ( 1 H, s), 10.87 (1 H, s); ESIMS found for C 22 H 23 FN 6 O mlz 406.95 (M+l).

112

[0668] 4-Fluoro-N-(6-(6-(((3S,4S)-3-fluoropiperidin-4-yl)amino)pyra zin-2-yl) isoquinolin-3-yl)benzamide 112.

[0669] White solid (26.0 mg, 0.057 mmol, 63.3% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.38 - 1.50 ( 1 H, m), 2.02 - 2.15 ( 1 H, m), 2.55 - 2.72 (3 H, m), 2.85 - 2.95 ( 1 H, m), 4.18 - 4.28 ( 1 H, m), 4.38 - 4.56 ( 1 H, m), 7.31 - 7.42 (3 H, m), 8.01 (1 H, s), 8.13 - 8.27 (4 H, m), 8.50 ( 1 H, s), 8.59 ( 1 H, s), 8.71 ( 1 H, s), 9.24 (1 H, s), 10.92 (1 H, s); ESIMS found for C25H22F2N6O mlz 460.95 (M+l).

113

[0670] N-(6-(6-(( 1 -Methylpiperidin-4-yl)amino)pyrazin-2-yl)isoquinolin-3 -yl) cyclobutanecarboxamide 113. [0671] Yellow solid (61.0 mg, 0.147 mmol, 84.2% yield). ¾ NMR (500 MHz, DMSO- e) δ ppm 1.46 - 1.59 (2 H, m), 1.78 - 1.88 ( 1 H, m), 1.91 - 2.03 (3 H, m), 2.06 - 2.18 (4 H, m), 2.20 (3 H, s), 2.22 - 2.32 (2 H, m), 2.76 (2 H, br d, J=1 1.53 Hz), 3.40 - 3.49 (1 H, m), 3.80 - 3.91 ( 1 H, m), 7.17 ( 1 H, d, J=7.41 Hz), 7.95 (1 H, s), 8.09 - 8.18 (2 H, m), 8.45 (1 H, s), 8.50 (1 H, s), 8.58 (1 H, s), 9.14 ( 1 H, s), 10.44 ( 1 H, s); ESIMS found for Cz^sNeO mlz 417.2 (M+1).

114

[0672] N-(6-(6-(( 1 -Methylpiperidin-4-yl)amino)pyrazin-2-yl)isoquinolin-3 -yl) cyclopentanecarboxamide 114.

[0673] White solid (34.0 mg, 0.079 mmol, 73.1% yield). ¾ NMR (500 MHz, DMSO- d 6 ) δ ppm 1.46 - 1.61 (4 H, m), 1.65 - 1.82 (4 H, m), 1.85 - 1.94 (2 H, m), 1.95 - 2.03 (2 H, m), 2.06 - 2.15 (2 H, m), 2.20 (3 H, s), 2.70 - 2.82 (2 H, m), 3.01 (1 H, quin, J=7.96 Hz), 3.80 - 3.91 ( 1 H, m), 7.17 ( 1 H, d, J=7.14 Hz), 7.95 (1 H, s), 8.09 - 8.18 (2 H, m), 8.44 (1 H, s), 8.48 (1 H, s), 8.56 ( 1 H, s), 9.15 ( 1 H, s), 10.57 (1 H, s); ESIMS found for C 2 5H 3 oN 6 0 mlz 431.2 (M+1).

115

[0674] 4-Fluoro-N-(6-(6-(( 1 -methylpiperidin-4-yl)amino)pyrazin-2-yl)isoquinolin- 3-yl)benzamide 115.

[0675] Off-white solid (33.0 mg, 0.069 mmol, 67.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.47 - 1.63 (2 H, m), 1.95 - 2.04 (2 H, m), 2.07 - 2.15 (2 H, m), 2.21 (3 H, s), 2.72 - 2.82 (2 H, m), 3.81 - 3.93 ( 1 H, m), 7.13 (1 H, br d, J=7.14 Hz), 7.36 (2 H, t, J=8.92 Hz), 7.97 (1 H, s), 8.11 - 8.24 (4 H, m), 8.47 (1 H, s), 8.58 (1 H, s), 8.69 ( 1 H, s), 9.24 ( 1 H, s), 10.93 (1 H, s); ESIMS found for mlz 457.0 (M+1).

[0676] N-(6-(6-((l-Isopropylpiperidin-4-yl)amino)pyrazin-2-yl)isoqu inolin-3-yl) cyclopropanecarboxamide 116.

[0677] Beige solid (55.0 mg, 0.121 mmol, 55.5% yield). ¾ NMR (499 MHz, DMSO- d 6 ) 5 ppm 0.78 - 0.85 (2 H, m), 0.85 - 0.90 (2 H, m), 0.99 (6 H, d, J=6.59 Hz), 1.40 - 1.54 (2 H, m), 2.00 (2 H, br d, J=10.70 Hz), 2.04 - 2.12 (1 H, m), 2.28 (2 H, td, J=l 1.18, 2.06 Hz), 2.71 ( 1 H, dt, J=13.10, 6.48 Hz), 2.77 - 2.84 (2 H, m), 3.79 - 3.89 ( 1 H, m), 7.14 ( 1 H, d, J=7.14 Hz), 7.95 (1 H, s), 8.09 - 8.18 (2 H, m), 8.43 ( 1 H, s), 8.46 (1 H, s), 8.52 (1 H, s), 9.16 ( 1 H, s), 10.92 ( 1 H, s); ESIMS found for C 2 5H 3 oN 6 0 mlz 431.2 (M+1).

117

[0678] 3,3-Difluoro-N-(6-(6-(( l-isopropylpiperidin-4-yl)amino)pyrazin-2-yl) isoquinolin-3 -yl)cyclobutane - 1 -carboxamide 117.

[0679] Off-white solid (39.0 mg, 0.081 mmol, 59.3% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.00 (6 H, d, J=6.31 Hz), 1.41 - 1.56 (2 H, m), 1.96 - 2.05 (2 H, m), 2.25 - 2.34 (2 H, m), 2.72 ( 1 H, dt, J=13.04, 6.66 Hz), 2.77 - 2.91 (5 H, m), 3.31 - 3.37 (2 H, m), 3.80 - 3.90 ( 1 H, m), 7.14 (1 H, d, J=7.14 Hz), 7.95 (1 H, s), 8.1 1 - 8.21 (2 H, m), 8.44 (1 H, s), 8.51 (1 H, s), 8.58 ( 1 H, s), 9.17 (1 H, s), 10.82 ( 1 H, s); ESIMS found for CzeH^FzNeO mlz 481.3 (M+1).

[0680] N-(6-(6-((l-Isopropylpiperidin-4-yl)amino)pyrazin-2-yl)isoqu inolin-3-yl) cyclopentanecarboxamide 118.

[0681] White solid (62.0 mg, 0.135 mmol, 75.1% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 1.00 (6 H, d, J=6.59 Hz), 1.41 - 1.52 (2 H, m), 1.52 - 1.63 (2 H, m), 1.65 - 1.81 (4 H, m), 1.84 - 1.94 (2 H, m), 1.96 - 2.05 (2 H, m), 2.25 - 2.34 (2 H, m), 2.72 (1 H, dt, J=13.17, 6.59 Hz), 2.76 - 2.85 (2 H, m), 2.95 - 3.04 (1 H, m), 3.79 - 3.90 (1 H, m), 7.13 (1 H, br d, J=7.14 Hz), 7.95 (2 H, s), 8.09 - 8.18 (2 H, m), 8.43 (1 H, s), 8.48 (1 H, s), 8.56 (1 H, s), 9.15 (1 H, s), 10.55 (2 H, s); ESIMS found for CZVHMNSO

119

[0682] 4-Fluoro-N-(6-(6-((l-isopropylpiperidin-4-yl)amino)pyrazin-2 -yl) isoquinolin-3-yl)benzamide 119.

[0683] White solid (56.0 mg, 0.116 mmol, 63.9% yield). 'HNMR (500 MHz, DMSO- d 6 ) δ ppm 1.00 (6 H, d, J=6.59 Hz), 1.42 - 1.54 (2 H, m), 1.96 - 2.07 (2 H, m), 2.30 (2 H, td, J=l 1.25, 2.20 Hz), 2.72 (1 H, dt, J=13.17, 6.59 Hz), 2.77 - 2.85 (2 H, m), 3.79 - 3.92 (1 H, m), 7.16 (1 H, d, J=7.41 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.96 (1 H, s), 8.14 - 8.25 (4 H, m), 8.47 (1 H, s), 8.58 (1 H, s), 8.70 (1 H, s), 9.24 (1 H, s), 10.99 (1 H, s); ESIMS found for CzsHzgFNeO mlz 485.25 (M+1).

120

[0684] N-(6-(6-(Dimethylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(4- methylpiperazin-1 -yl)isonicotinamide 120.

[0685] White solid (54.7 mg, 0.117 mmol, 36.8% yield). 'HNMR (499 MHz, DMSO- d 6 ) δ ppm 2.24 (3 H, s), 2.39 - 2.47 (4 H, m), 3.21 (6 H, s), 3.56 - 3.65 (4 H, m), 7.17 (1 H, dd, J=5.08, 1.24 Hz), 7.48 (1 H, s), 8.17 - 8.23 (2 H, m), 8.26 (1 H, d, J=5.21 Hz), 8.29 (1 H, dd, J=8.51, 1.65 Hz), 8.62 (1 H, s), 8.68 (1 H, s), 8.74 (1 H, s), 9.26 (1 H, s), 11.12 (1 H, s); ESIMS found for C 2 6H28N 8 0 mlz 469.2 (M+1).

121

[0686] N-(6-(6-(Diethylamino)pyrazin-2-yl)isoquinolin-3-yl)-2-(4-me thylpiperazin- l-yl)isonicotinamide 121.

[0687] Yellow solid (80.8 mg, 0.163 mmol, 43.2% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.22 (6 H, t, J=7.00 Hz), 2.23 (3 H, s), 2.43 (4 H, br t, J=4.94 Hz), 3.56 - 3.62 (4 H, m), 3.65 (4 H, q, J=7.04 Hz), 7.17 (1 H, dd, J=5.21, 1.10 Hz), 7.48 (1 H, s), 8.15 (1 H, s), 8.17 - 8.22 (1 H, m), 8.22 - 8.29 (2 H, m), 8.57 (1 H, s), 8.63 (1 H, s), 8.73 (1 H, s), 9.25 (1 H, s), 11.13 (1 H, s); ESIMS found for

122

[0688] N-(6-(6-(3-Aminoazetidin-l-yl)pyrazin-2-yl)isoquinolin-3-yl) -4- fluorobenzamide 122.

[0689] White solid (19.0 mg, 0.046 mmol, 56.2% yield). 'HNMR (499 MHz, DMSO- d 6 ) δ ppm 2.26 (2 H, br s), 3.76 (2 H, dd, J=8.23, 5.76 Hz), 3.92 (1 H, quin, J=6.45 Hz), 4.33 (2 H, t, J=7.82 Hz), 7.37 (2 H, t, J=8.92 Hz), 7.91 (1 H, s), 8.15 - 8.19 (2 H, m), 8.20 (1 H, d, J=4.39 Hz), 8.22 - 8.27 (1 H, m), 8.63 (1 H, s), 8.65 (1 H, s), 8.72 (1 H, s), 9.25 (1 H, s), 10.98 (1 H, s); ESIMS found for C23H19FN6O

123

[0690] 2-(4-Methylpiperazin-l-yl)-N-(6-(6-(pyrrolidin-l-yl)pyrazin- 2-yl) isoquinolin-3 -yl)isonicotinamide 123.

[0691] Beige solid (140.0 mg, 0.283 mmol, 60.3% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.95 - 2.07 (4 H, m), 2.24 (3 H, s), 2.36 - 2.46 (4 H, m), 3.49 - 3.71 (8 H, m), 7.13 - 7.20 (1 H, m), 7.48 (1 H, s), 8.02 ( 1 H, s), 8.20 ( 1 H, d, J=8.51 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.29 (1 H, dd, J=8.51, 1.65 Hz), 8.60 ( 1 H, s), 8.67 (1 H, s), 8.73 ( 1 H, s), 9.26 (1 H, s), 1 1.12 ( 1 H, s); ESIMS found for C 2 8H 3 oN 8 0 mlz 495.3 (M+l).

124

[0692] N-(6-(6-(Azetidin-3-ylmethoxy)pyrazin-2-yl)isoquinolin-3-yl) -4- fluorobenzamide 124.

[0693] White solid (17.0 mg, 0.040 mmol, 55.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 3.11 ( 1 H, dt, J=13.86, 6.79 Hz), 3.42 (2 H, t, J=6.86 Hz), 3.61 (2 H, t, J=7.82 Hz), 4.66 (2 H, d, J=7.14 Hz), 7.37 (2 H, t, J=8.78 Hz), 8.15 - 8.21 (2 H, m), 8.21 - 8.25 ( 1 H, m), 8.28 - 8.33 ( 1 H, m), 8.34 (1 H, s), 8.76 (2 H, d, J=6.04 Hz), 9.05 (1 H, s), 9.28 (1 H, s), 11.00 (1 H, br s); ESIMS found for C24H 2 oFN 5 0 +l).

125

[0694] N-(6-(6-(Azetidin-3-yloxy)pyrazin-2-yl)isoquinolin-3-yl)-4-f luorobenzamide

125.

[0695] White solid (15.0 mg, 0.036 mmol, 53.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 3.60 - 3.70 (2 H, m), 3.84 - 3.94 (2 H, m), 5.55 - 5.65 (1 H, m), 7.32 - 7.43 (2 H, m), 8.12 - 8.21 (2 H, m), 8.21 - 8.30 (2 H, m), 8.37 ( 1 H, s), 8.72 (1 H, s), 8.76 (1 H, s), 9.08 (1 H, s), 9.28 ( 1 H, s), 10.99 ( 1 H, br s); ESIMS found for C 2 3Hi 8 FN 5 02 mlz 415.9 (M+l).

126 [0696] N-(6-( lH-Pyrrolo[2,3-b]pyridin-5 -yl)isoquinolin-3-yl)- 1 -( 1 -methylpiperidin- 4-yl)-lH-pyrazole-4-carboxamide 126.

[0697] Off-white solid (25.0 mg, 0.053 mmol, 64.1% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.89 - 2.00 (2 H, m), 2.00 - 2.11 (4 H, m), 2.21 (3 H, s), 2.86 (2 H, br d, J=l 1.53 Hz), 4.17 (1 H, tt,J=11.15, 4.22 Hz), 6.56 (1 H, dd, J=3.29, 1.92 Hz), 7.53 - 7.59 (1 H, m), 7.94 (1 H, dd,J=8.51, 1.65 Hz), 8.16 (1 H, d,J=8.51 Hz), 8.19 (1 H, s), 8.25 (1 H, s), 8.44 (1 H, d,J=2.20 Hz), 8.63 (1 H, s), 8.66 (1 H, s), 8.73 (1 H, d, J=2.20 Hz), 9.20 (1 H, s), 10.55 (1 H, s), 11.80 (1 H, br s); ESIMS found for C26H2 +l).

[0698] N-(6-(lH-Pyrrolo[3,2-b]pyridin-6-yl)isoquinolin-3-yl)-l-(l-m ethylpiperidin- 4-yl)-lH-pyrazole-4-carboxamide 127.

[0699] Off-white solid (35.0 mg, 0.074 mmol, 89.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.91 -2.01 (2 H, m), 2.02 - 2.10 (4 H, m),2.21 (3 H, s), 2.86 (2 H, brd,J=11.25 Hz), 4.17 (1 H, tt, J=l 1.08, 4.15 Hz), 6.63 (1 H, d, J=1.92 Hz), 7.74 (1 H, t, J=2.88 Hz), 7.95 (1 H, dd, J=8.51, 1.65 Hz), 8.14 - 8.22 (3 H, m), 8.27 (1 H, s), 8.63 (1 H, s), 8.67 (1 H, s), 8.83 (1 H, d, J=1.92Hz),9.21 (1H, s), 10.56(1H, s), 11.51 (1 H, br s); ESIMS found for C 26 H 25 N 7 O mlz 452.2 (M+l).

[0700] N-(6-(lH-Pyrrolo[2,3-c]pyridin-4-yl)isoquinolin-3-yl)-l-(l-m ethylpiperidin- 4-yl)-lH-pyrazole-4-carboxamide 128.

[0701] Off-white solid (6.0 mg, 0.013 mmol, 15.3% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 1.89 - 2.00 (2 H, m), 2.01 - 2.12 (4 H, m), 2.21 (3 H, s), 2.86 (2 H, br d, J=l 1.53 Hz), 4.18 (1 H, tt, J=11.08, 4.29 Hz), 6.76 (1 H, d, J=2.74 Hz), 7.75 (1 H, d, J=3.02 Hz), 7.91 (1 H, dd,J=8.51, 1.65 Hz), 8.19 (1 H, s), 8.19 - 8.25 (2 H, m), 8.38 (1 H, s), 8.63 (1 H, s), 8.68 (1 H, s), 8.83 (1 H, s), 9.24 (1 H, s), 10.58 (1 H, s), 11.88 (1 H, br s); ESIMS found for C 26 H 25 N 7 O mlz 452.2 (M+l).

[0702] N-(6-(lH-Pyrrolo[2,3-b]pyridin-5-yl)isoquinolin-3-yl)-2-(4-m ethylpiperazin- l-yl)isonicotinamide 129.

[0703] Off-white solid (53.0 mg, 0.1 14 mmol, 48.7% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.56 - 3.67 (4 H, m), 6.56 (1 H, dd, J=3.57, 1.92 Hz), 7.12 - 7.23 ( 1 H, m), 7.48 ( 1 H, s), 7.53 - 7.59 (1 H, m), 7.99 (1 H, dd, J=8.64, 1.78 Hz), 8.20 ( 1 H, d, J=8.78 Hz), 8.27 (1 H, d, J=4.94 Hz), 8.31 ( 1 H, s), 8.45 (1 H, d, J=1.92 Hz), 8.71 (1 H, s), 8.74 (1 H, d, J=2.20 Hz), 9.24 ( 1 H, s), 1 1.10 ( 1 H, s), 1 1.81 ( 1 H, br s); ESIMS found for C 27 H 25 N 7 O mlz 464.2 (M+ l).

[0704] N-(6-(lH-Pyrrolo[3,2-b]pyridin-6-yl)isoquinolin-3-yl)-2-(4-m ethylpiperazin- l-yl)isonicotinamide 130.

[0705] Off-white solid (33.0 mg, 0.071 mmol, 30.4% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, br t, J=4.80 Hz), 3.56 - 3.63 (4 H, m), 6.64 ( 1 H, d, J=1.92 Hz), 7.17 ( 1 H, d, J=5.21 Hz), 7.48 ( 1 H, s), 7.74 ( 1 H, t, J=2.88 Hz), 8.00 ( 1 H, dd, J=8.51, 1.37 Hz), 8.18 - 8.24 (2 H, m), 8.27 (1 H, d, J=4.94 Hz), 8.34 (1 H, s), 8.73 (1 H, s), 8.84 (1 H, d, J=1.92 Hz), 9.25 (1 H, s), 1 1.10 ( 1 H, s), 1 1.52 (1 H, br s); ESIMS found for C 27 H 25 N 7 O mlz 464.2 (M+ l).

131

[0706] N-(6-(lH-Pyrrolo[2,3-c]pyridin-4-yl)isoquinolin-3-yl)-2-(4-m ethylpiperazin- l-yl)isonicotinamide 131.

[0707] Off-white solid (41.0 mg, 0.089 mmol, 37.7% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.56 - 3.66 (4 H, m), 6.77 (1 H, br s), 7.17 ( 1 H, dd, J=5.21, 1.10 Hz), 7.49 (1 H, s), 7.76 ( 1 H, t, J=2.74 Hz), 7.96 ( 1 H, dd, J=8.51, 1.65 Hz), 8.22 - 8.30 (3 H, m), 8.40 (1 H, s), 8.74 (1 H, s), 8.83 (1 H, s), 9.29 (1 H, s), 11.12 (1 H, s), 11.88 (1 H, br s); ESIMS found for C27H25N7O mlz 464.2 (M+l).

132

[0708] 2-(4-Methylpiperazin-l-yl)-N-(6-(oxazolo[5,4-b]pyridin-6-yl) isoquinolin-3- yl)isonicotinamide 132.

[0709] Off-white solid (21.6 mg, 0.046 mmol, 23.8% yield). ¾ NMR (500 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, br t, J=4.94 Hz), 3.56 - 3.65 (4 H, m), 7.17 (1 H, dd, J=4.94, 1.10 Hz), 7.48 (1 H, s), 8.04 (1 H, dd, J=8.51, 1.65 Hz), 8.23 - 8.30 (2 H, m), 8.44 (1 H, s), 8.75 (1 H, s), 8.82 (1 H, d, J=1.92 Hz), 8.95 (1 H, d, J=2.20 Hz), 9.00 (1 H, s), 9.29 (1 H, s), 11.15 (1 H, s); ESIMS found for C26H23N7O2 mlz 466.2 (M+l).

133

[0710] N-(6-(2-Methyl-lH-pyrrolo[2,3-b]pyridin-5-yl)isoquinolin-3-y l)-2-(4- methylpiperazin-1 -yl)isonicotinamide 133.

[0711] Off-white solid (38.0 mg, 0.080 mmol, 17.0% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.41 - 2.45 (7 H, m), 3.55 - 3.66 (4 H, m), 6.25 (1 H, s), 7.17 (1 H, dd, J=4.94, 1.10 Hz), 7.48 (1 H, s), 7.97 (1 H, dd, J=8.51, 1.65 Hz), 8.18 (1 H, d, J=8.51 Hz), 8.26 (1 H, d, J=5.21 Hz), 8.28 (2 H, s), 8.61 (1 H, d, J=2.20 Hz), 8.70 (1 H, s), 9.23 (1 H, s), 11.08 (1 H, s), 11.62 (1 H, s); ESIMS (M+l).

134

[0712] N-(6-(3-Methyl-lH-pyrrolo[2,3-b]pyridin-5-yl)isoquinolin-3-y l)-2-(4- methylpiperazin-1 -yl)isonicotinamide 134.

[0713] Off-white solid (25.0 mg, 0.052 mmol, 11.2% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.35 (3 H, d, J=1.10 Hz), 2.43 (4 H, t, J=4.94 Hz), 3.58 - 3.67 (4 H, m), 7.17 (1 H, dd, J=5.08, 1.23 Hz), 7.31 (1 H, d, J=0.82 Hz), 7.48 (1 H, s), 8.03 (1 H, dd, J=8.78, 1.65 Hz), 8.20 (1 H, d, J=8.51 Hz), 8.26 (1 H, d, J=4.94 Hz), 8.35 ( 1 H, s), 8.43 ( 1 H, d, J=2.20 Hz), 8.68 - 8.76 (2 H, m), 9.24 ( 1 H, s), 1 1.09 (1 H, s), 1 1.44 (1 H, s); ESIMS found for C28H27N7O mlz 478.2 (M+ l).

135

[0714] N-(6-(2-Methyl-lH-pyrrolo[3,2-b]pyridin-6-yl)isoquinolin-3-y l)-2-(4- methylpiperazin-1 -yl)isonicotinamide 135.

[0715] Off-white solid (21.8 mg, 0.046 mmol, 1 1.6% yield). ¾ NMR (500 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, br t, J=4.94 Hz), 2.49 (3 H, br s), 3.56 - 3.64 (4 H, m), 6.35 ( 1 H, d, J=0.82 Hz), 7.17 (1 H, dd, J=5.21, 1.10 Hz), 7.48 (1 H, s), 7.98 (1 H, dd, J=8.64, 1.78 Hz), 8.05 ( 1 H, d, J=1.37 Hz), 8.19 ( 1 H, d, J=8.78 Hz), 8.27 ( 1 H, d, J=4.94 Hz), 8.31 (1 H, s), 8.72 (1 H, s), 8.75 (1 H, d, J=1.92 Hz), 9.24 (1 H, s), 1 1.10 (1 H, s), 1 1.38 (1 H, d, J=1.37 Hz); ESIMS found for C28H27N7O

136

[0716] N-(6-(5H-Pyrrolo[2,3-b]pyrazin-2-yl)isoquinolin-3-yl)-2-(4-m ethylpiperazin- l-yl)isonicotinamide 136.

[0717] Off-white solid (31.0 mg, 0.067 mmol, 28.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=4.94 Hz), 3.57 - 3.68 (4 H, m), 6.75 (1 H, dd, J=3.43, 1.51 Hz), 7.18 ( 1 H, dd, J=5.08, 0.96 Hz), 7.48 (1 H, s), 7.97 ( 1 H, t, J=3.02 Hz), 8.23 (1 H, d, J=8.78 Hz), 8.27 (1 H, d, J=4.94 Hz), 8.40 (1 H, dd, J=8.64, 1.51 Hz), 8.72 (1 H, s), 8.75 (1 H, s), 9.12 (1 H, s), 9.27 (1 H, s), 1 1.12 ( 1 H, s), 12.20 (1 H, br s); ESIMS found for C 2 6H24N 8 0 mlz 465.2 (M+ l).

137 [0718] N-(6-(5H-Pyrrolo[2,3-b]pyrazin-3-yl)isoquinolin-3-yl)-2-(4-m ethylpiperazin- l-yl)isonicotinamide 137.

[0719] Off-white solid (25.0 mg, 0.054 mmol, 23.4% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.24 (3 H, s), 2.43 (4 H, t, J=5.08 Hz), 3.58 - 3.67 (4 H, m), 6.72 ( 1 H, d, J=3.57 Hz), 7.18 ( 1 H, dd, J=5.21, 1.10 Hz), 7.48 (1 H, s), 7.98 ( 1 H, d, J=3.57 Hz), 8.24 (1 H, d, J=8.78 Hz), 8.27 (1 H, d, J=5.21 Hz), 8.38 (1 H, dd, J=8.51, 1.65 Hz), 8.74 (1 H, s), 8.75 (1 H, s), 9.27 (1 H, s), 9.28 (1 H, s), 11.13 (1 H, s), 12.22 (1 H, s); ESIMS found for C 2 6H24N 8 0 mlz 465.2 (M+1).

138

[0720] l-Isopropyl-N-(6-(6-(piperidin-4-ylamino)pyrazin-2-yl)isoqui nolin-3-yl)-lH- pyrazole-4-carboxamide 138.

[0721] Beige solid (62.0 mg, 0.136 mmol, 52.1% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.32 - 1.43 (2 H, m), 1.46 (6 H, d, J=6.59 Hz), 1.90 - 2.01 (2 H, m), 2.62 (2 H, br t, J=10.70 Hz), 2.95 - 3.05 (2 H, m), 3.88 - 3.99 (1 H, m), 4.55 ( 1 H, spt, J=6.68 Hz), 7.17 (1 H, br d, J=7.14 Hz), 7.95 (1 H, s), 8.12 - 8.22 (3 H, m), 8.46 (1 H, s), 8.54 (1 H, s), 8.62 ( 1 H, s), 8.66 (1 H, s), 9.21 (1 H, s), 10.58 (1 H, s); ESIMS found for CzsHzsNgO mlz 457.2 (M+1).

[0722] 1 -Isopropyl-N-(6-(6-(( 1 -methylpiperidin-4-yl)amino)pyrazin-2-yl) isoquinolin-3 -yl) - 1 H-pyrazole -4 -carboxamide 139.

[0723] Beige solid (32.0 mg, 0.068 mmol, 53.5% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.46 (6 H, d, J=6.86 Hz), 1.48 - 1.62 (2 H, m), 1.94 - 2.04 (2 H, m), 2.10 (2 H, br t, J=10.57 Hz), 2.21 (3 H, s), 2.77 (2 H, br d, J=l 1.53 Hz), 3.79 - 3.91 ( 1 H, m), 4.55 ( 1 H, spt, J=6.63 Hz), 7.17 (1 H, d, J=7.14 Hz), 7.96 (1 H, s), 8.12 - 8.21 (3 H, m), 8.47 (1 H, s), 8.54 ( 1 H, s), 8.62 ( 1 H, s), 8.66 ( 1 H, s), 9.21 (1 H, s), 10.58 (1 H, s) ESIMS found for C 2 6H 3 oN 8 0 mlz 471.25 (M+1).

140

[0724] 2-(Piperidin-4-yl)-N-(6-(pyridin-3-yl)isoquinolin-3-yl)oxazo le-4- carboxamide 140.

[0725] White solid (95.0 mg, 0.238 mmol, 70.7% yield). ¾ NMR (500 MHz, DMSO- 6) 5 ppm 1.58 - 1.70 (2 H, m), 1.95 (2 H, br dd, J=12.90, 2.47 Hz), 2.13 (1 H, br s), 2.59 (2 H, td, J=1 1.94, 2.47 Hz), 2.99 (3 H, dt, J=12.14, 3.53 Hz), 7.57 (1 H, ddd, J=7.96, 4.67, 0.82 Hz), 7.97 ( 1 H, dd, J=8.51, 1.65 Hz), 8.23 ( 1 H, d, J=8.51 Hz), 8.28 - 8.34 (1 H, m), 8.39 (1 H, d, J=0.82 Hz), 8.61 - 8.69 (2 H, m), 8.86 (1 H, s), 9.1 1 (1 H, dd, J=2.47, 0.82 Hz), 9.25 ( 1 H, s), 9.80 (1 H, br s); ESIMS found for C 23 H2iN 5 02 mlz 400.2 (M+l).

141

[0726] N-(6-(5-Fluoropyridin-3-yl)isoquinolin-3-yl)-2-(piperidin-4- yl)oxazole-4- carboxamide 141.

[0727] White solid (100.0 mg, 0.240 mmol, 62.6% yield). ¾ NMR (500 MHz, DMSO- e) δ ppm 1.61 - 1.73 (2 H, m), 1.98 (2 H, br dd, J=12.90, 2.74 Hz), 2.64 (2 H, td, J=l 1.94, 2.47 Hz), 2.99 - 3.08 (3 H, m), 8.01 (1 H, dd, J=8.51, 1.65 Hz), 8.24 ( 1 H, d, J=8.51 Hz), 8.29 - 8.36 (1 H, m), 8.47 ( 1 H, d, J=1.10 Hz), 8.66 (1 H, s), 8.67 (1 H, d, J=2.74 Hz), 8.87 ( 1 H, s), 9.02 ( 1 H, t, J=1.65 Hz), 9.26 ( 1 H, s), 9.82 ( 1 H, br s); ESIMS found for C 23 H2oFN 5 02 mlz 418.1 (M+l).

142

[0728] 2-(l -Methylpiperidin-4-yl)-N-(6-(pyridin-3-yl)isoquinolin-3-yl)o xazole-4- carboxamide 142.

[0729] White solid (32.0 mg, 0.077 mmol, 77.3% yield). 'HNMR (500 MHz, DMSO- d 6 ) 5 ppm 1.72 - 1.85 (2 H, m), 1.98 - 2.08 (4 H, m), 2.19 (3 H, s), 2.79 (2 H, br d, J=1 1.25 Hz), 2.88 ( 1 H, tt, J=1 1.25, 3.84 Hz), 7.52 - 7.61 (1 H, m), 7.97 ( 1 H, dd, J=8.51, 1.65 Hz), 8.23 ( 1 H, d, J=8.78 Hz), 8.27 - 8.34 ( 1 H, m), 8.39 (1 H, s), 8.61 - 8.68 (2 H, m), 8.87 (1 H, s), 9.07 - 9.14 (1 H, m), 9.24 (1 H, s), 9.80 (1 H, s); ESIMS found for Cz^NsOz mlz 414.2 (M+l).

143

[0730] N-(6-(5 -Fluoropyridin-3 -yl)isoquinolin-3 -yl)-2-( 1 -methylpiperidin-4-yl) oxazole-4-carboxamide 143.

[0731] White solid (36.0 mg, 0.083 mmol, 87.1% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 1.72 - 1.85 (2 H, m), 1.99 - 2.08 (4 H, m), 2.19 (3 H, s), 2.75 - 2.83 (2 H, m), 2.89 ( 1 H, tt, J=1 1.29, 3.67 Hz), 8.01 ( 1 H, dd, J=8.51, 1.65 Hz), 8.24 ( 1 H, d, J=8.51 Hz), 8.32 (1 H, dt, J=10.22, 2.30 Hz), 8.48 (1 H, s), 8.66 (1 H, s), 8.67 ( 1 H, d, J=2.74 Hz), 8.87 (1 H, s), 9.03 ( 1 H, t, J=1.51 Hz), 9.26 (1 H, s), 9.82 (1 H, s); ESIMS found for Cz^FNsOz mlz 432.2 (M+l).

144

[0732] N-(7-Fluoro-6-(pyridin-3-yl)isoquinolin-3-yl)-2-moφholinoac etamide 144.

[0733] White solid (40.0 mg, 0.105 mmol, 33.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.54 - 2.62 (4 H, m), 3.26 (2 H, s), 3.60 - 3.69 (4 H, m), 7.58 (1 H, ddd, J=7.96, 4.94, 0.82 Hz), 8.05 (1 H, d, J=10.98 Hz), 8.13 (1 H, dq, J=7.96, 1.92 Hz), 8.25 (1 H, d, J=7.41 Hz), 8.59 ( 1 H, s), 8.68 ( 1 H, dd, J=4.67, 1.65 Hz), 8.89 (1 H, t, J=1.65 Hz), 9.19 (1 H, s), 10.13 (1 H, s); ESIMS found for C20H19FN4O2 mlz 367.2 (M+l).

145

[0734] N-(7-Fluoro-6-(pyridin-3-yl)isoquinolin-3-yl)-2-(4-methylpip erazi acetamide 145.

[0735] Beige solid (35.0 mg, 0.089 mmol, 28.0% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.19 (3 H, s), 2.40 (4 H, br s), 2.58 (4 H, br s), 3.23 (2 H, s), 7.58 (1 H, ddd, J=7.96, 4.94, 0.82 Hz), 8.05 (1 H, d, J=10.98 Hz), 8.13 (1 H, dq, J=7.96, 1.92 Hz), 8.26 (1 H, d, J=7.68 Hz), 8.59 ( 1 H, s), 8.68 (1 H, dd, J=4.80, 1.51 Hz), 8.90 (1 H, d, J=1.65 Hz), 9.18 ( 1 H, s), 10.04 (1 H, s); ESIMS found for C 2 iH 2 2FN 5 0 mlz 380.2 (M+l).

146

[0736] N-(6-(5-Methylpyridin-3-yl)isoquinolin-3-yl)-2-moφholinoace tamide 146.

[0737] White solid (80.0 mg, 0.221 mmol, 38.7% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 2.41 (3 H, s), 2.55 - 2.62 (4 H, m), 3.26 (2 H, s), 3.62 - 3.70 (4 H, m), 7.92 ( 1 H, dd, J=8.51, 1.65 Hz), 8.09 - 8.15 ( 1 H, m), 8.18 (1 H, d, J=8.51 Hz), 8.32 ( 1 H, s), 8.49 ( 1 H, d, J=1.92 Hz), 8.57 (1 H, s), 8.89 (1 H, d, J=1.92 Hz), 9.19 (1 H, s), 10.10 ( 1 H, s); ESIMS found for C21H22N4O2 mlz 363.2 (M+l).

147

[0738] N-(6-(5-Methoxypyridin-3-yl)isoquinolin-3-yl)-2-mo holinoacetamide 147.

[0739] White solid ( 135.0 mg, 0.357 mmol, 62.5% yield). ¾ NMR (499 MHz, DMSO- e) δ ppm 2.54 - 2.61 (4 H, m), 3.26 (2 H, s), 3.62 - 3.69 (4 H, m), 3.95 (3 H, s), 7.84 ( 1 H, dd, J=2.74, 1.92 Hz), 7.95 (1 H, dd, J=8.64, 1.78 Hz), 8.18 ( 1 H, d, J=8.78 Hz), 8.36 (1 H, d, J=2.74 Hz), 8.38 (1 H, d, J=0.82 Hz), 8.59 (1 H, s), 8.68 (1 H, d, J=1.92 Hz), 9.19 ( 1 H, s), 10.10 (1 H, s); ESIMS found for C2iH 2 2N 4 0 3 mlz 379.2 (M+l).

148

[0740] N-(6-(5-Aminopyridin-3-yl)isoquinolin-3-yl)-2-(piperidin-4-y l)oxazole-4- carboxamide 148.

[0741] Beige solid (30.0 mg, 0.072 mmol, 15.5% yield). ¾ NMR (500 MHz, DMSO- 6) 5 ppm 1.58 - 1.69 (2 H, m), 1.91 - 2.01 (2 H, m), 2.59 (2 H, br t, J=10.98 Hz), 2.95 - 3.06 (3 H, m), 5.47 (2 H, br s), 7.34 ( 1 H, t, J=2.20 Hz), 7.82 ( 1 H, dd, J=8.51, 1.65 Hz), 8.01 (1 H, d, J=2.20 Hz), 8.18 (1 H, d, J=8.78 Hz), 8.20 (1 H, br s), 8.23 (1 H, d, J=1.65 Hz), 8.61 (1 H, s), 8.84 ( 1 H, s), 9.22 (1 H, s), 9.76 (1 H, br s); ESIMS found for C23H22N6O2 mlz 415.2 (M+l).

149

[0742] 2-(l -Isopropylpiperidin-4-yl)-N-(6-(pyridin-3-yl)isoquinolin-3-y l)oxazole-4- carboxamide 149.

[0743] White solid (47.0 mg, 0.107 mmol, 80.2% yield). ¾ NMR (499 MHz, DMSO- d 6 ) δ ppm 0.98 (6 H, d, J=6.59 Hz), 1.67 - 1.80 (2 H, m), 2.00 - 2.10 (2 H, m), 2.26 (2 H, td, J=l 1.39, 2.20 Hz), 2.72 (1 H, spt, J=6.63 Hz), 2.79 - 2.85 (2 H, m), 2.85 - 2.93 ( 1 H, m), 7.52 - 7.60 ( 1 H, m), 7.97 ( 1 H, dd, J=8.51, 1.65 Hz), 8.23 ( 1 H, d, J=8.51 Hz), 8.28 - 8.34 (1 H, m), 8.38 (1 H, d, J=0.82 Hz), 8.62 - 8.69 (2 H, m), 8.85 ( 1 H, s), 9.08 - 9.14 (1 H, m), 9.24 (1 H, s), 9.78 ( 1 H, s); ESIMS found for CzeHzvNsOz mlz 442.2 (M+l).

150

[0744] N-(6-(5-Fluoropyridin-3-yl)isoquinolin-3-yl)-2-(l -isopropylpiperidin-4-yl) oxazole-4-carboxamide 150.

[0745] White solid (52.0 mg, 0.1 13 mmol, 84.4% yield). 'HNMR (499 MHz, DMSO- d 6 ) δ ppm 0.98 (6 H, d, J=6.59 Hz), 1.65 - 1.81 (2 H, m), 2.00 - 2.10 (2 H, m), 2.26 (2 H, td, J=l 1.39, 2.20 Hz), 2.72 ( 1 H, spt, J=6.54 Hz), 2.79 - 2.85 (2 H, m), 2.85 - 2.93 (1 H, m), 8.01 (1 H, dd, J=8.51, 1.65 Hz), 8.24 (1 H, d, J=8.51 Hz), 8.28 - 8.36 (1 H, m), 8.47 (1 H, d, J=0.82 Hz), 8.66 (1 H, s), 8.66 ( 1 H, d, J=2.74 Hz), 8.86 ( 1 H, s), 9.02 ( 1 H, t, J=1.78 Hz), 9.26 (1 H, s), 9.81 (1 H, s); ESIMS found for CzeHzeFNsOz mlz 460.2 (M+l).

151

[0746] 2-(Moφholino-ί s)- -(6-(pyridin-3-yl)isoquinolin-3-yl)acetamide 151. [0747] White solid (65.0 mg, 0.182 mmol, 65.3% yield). ¾NMR (499 MHz, DMSO- d 6 ) δ ppm 3.26 (2 H, s), 7.56 (1 H, ddd, J=7.96, 4.67, 0.82 Hz), 7.92 (1 H, dd, J=8.51, 1.65 Hz), 8.19 (1 H, d, J=8.78 Hz), 8.25 - 8.30 (1 H, m), 8.33 (1 H, s), 8.58 (1 H, s), 8.65 (1 H, dd, J=4.67, 1.65 Hz), 9.06 - 9.13 (1 H, m), 9.19 (1 H, s), 10.10 (1 H, s); ESIMS found for mlz 357.2 (M+l).

Example 14.

[0748] The screening assay for Wnt activity is described as follows. Reporter cell lines can be generated by stably transducing cancer cell lines (e.g., colon cancer) or primary cells (e.g., IEC-6 intestinal cells) with a lentiviral construct that includes a Wnt-responsive promoter driving expression of the firefly luciferase gene.

[0749] SW480 colon carcinoma cells were transduced with a lentiviral vector expressing luciferase with a human Sp5 promoter consisting of a sequence of eight TCF/LEF binding sites. SW480 cells stably expressing the Sp5-Luc reporter gene and a hygromycin resistance gene were selected by treatment with 150 μg/mL of hygromycin for 7 days. These stably transduced SW480 cells were expanded in cell culture and used for all further screening activities. Each compound was dissolved in DMSO as a 10 mM stock and used to prepare compound source plates. Serial dilution (1 :3, 10-point dose-response curves starting from 10 μΜ) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, CA) into 384-well white solid bottom assay plates (Greiner Bio-One) with appropriate DMSO backfill for a final DMSO concentration of 0.1%. For Sp5-Luc reporter gene assays, the cells were plated at 4,000 cells/well in 384-well plates with a DMEM medium containing 1% fetal bovine serum, and 1% Penicillin- Streptomycin and incubated for 36 to 48 hours at 37°C and 5% CO2. Following incubation, 15 μΐ of BriteLite Plus luminescence reagent (Perkin Elmer) was added to each well of the 384-well assay plates. The plates were placed on an orbital shaker for 2 min and then luminescence was quantified using the Envision (Perkin Elmer) plate reader. Readings were normalized to DMSO only treated cells, and normalized activities were utilized for EC50 calculations using the dose- response log (inhibitor) vs. response -variable slope (four parameters) nonlinear regression feature available in GraphPad Prism 5.0 (or Dotmatics). For EC50 of >10 μΜ, the percent inhibition at 10 μΜ is provided.

[0750] Table 2 shows the measured activity for representative compounds of Formula I as described herein. Table 2.

Example 15.

[0751] Representative compounds were screened using the assay procedure for DYRK1A kinase activity as described below.

[0752] Each compound was dissolved in DMSO as a 10 mM stock and used to prepare compound source plates. Serial dilution (1 :3, 11 -point dose-response curves from 10 μΜΐο 0.00016 μΜ) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, CA) into 1536-well black-walled round bottom plates (Corning).

[0753] The DYRK1A kinase assay was run using the Ser/Thr 18 peptide Z-lyte assay kit according to manufacturer's instructions (Life Technologies- a Division of Thermo-Fisher). This is a non-radioactive assay using fluorescence resonance energy transfer (FRET) between coumarin and fluorescein to detect kinase activity which is represented as a ratio of coumarin emission/fluorescein emission. [0754] Briefly, recombinant DYRK1A kinase, ATP and Ser/Thr peptide 18 were prepared in IX Kinase buffer to final concentrations of 0.19 μg/mL, 30 μΜ, and 4 μΜ respectively. The mixture was allowed to incubate with the representative compounds for one hour at room temperature. All reactions were performed in duplicate. Unphosphorylated ("0% Control") and phosphorylated ("100% control") forms of Ser/Thr 18 served as control reactions. Additionally, an 11 -point dose-response curve of Staurosporine (luM top) was run to serve as a positive compound control.

[0755] After incubation, Development Reagent A was diluted in Development Buffer then added to the reaction and allowed to further incubate for one hour at room temperature. The plate was read at Ex 400 Em 455 to detect the coumarin signal and Ex 400 Em 520 to measure the signal (EnVision Multilabel Plate Reader, PerkinElmer).

[0756] The Emission ratio (Em) was calculated as a ratio of the coumarin (C) emission signal (at 445 nm)/Fluorescein (F) emission signal (at 520 nm). The percent phosphorylation was then calculated using the following formula: [1 - ((Em ratio X F100%)-C100%)/ ((C0%-C100%) + (Em ratio X (F100% - F0%)))]. Dose-response curves were generated and inhibitory concentration (IC50) values were calculated using non-linear regression curve fit in the Dotmatics' Studies Software (Bishops Stortford, UK).

[0757] Table 3 shows the measured activity for representative compounds of Formula I as described herein.

Table 3.

27 0.029 65 0.048 103 0.001 141 0.306

28 0.011 66 0.123 104 0.001 142 0.689

29 0.608 67 0.029 105 0.001 143 6.805

30 >10 68 0.328 106 0.001 144 0.009

31 0.003 69 0.020 107 0.001 145 0.013

32 0.057 70 0.017 108 0.002 146 0.009

33 0.004 71 0.011 109 0.002 147 0.020

34 0.004 72 0.007 110 0.002 148 0.012

35 0.010 73 0.004 111 0.001 149 0.102

36 0.002 74 0.094 112 0.008 150 1 .634

37 0.001 75 0.010 113 0.001 151 0.013

38 0.002 76 0.258 114 0.001

Example 16.

[0758] Representative compounds were screened using the assay procedure for GSK3 kinase activity as described below.

[0759] Each compound is dissolved in DMSO as a 10 mM stock and used to prepare compound source plates. Serial dilution (1 :3, 11-point dose-response curves from 10 μΜ to 0.0003 μΜ) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, CA) into 1536-well black-walled round bottom plates (Corning).

[0760] The GSK3 kinase assay is run using the Ser/Thr 09 peptide Z-lyte assay kit according to manufacturer's instructions (Life Technologies- a Division of Thermo-Fisher). This is a non-radioactive assay using fluorescence resonance energy transfer (FRET) between coumarin and fluorescein to detect kinase activity which is represented as ratio of coumarin emission/fluorescein emission.

[0761] Briefly, recombinant GSK3 kinase, ATP and Ser/Thr peptide 09 are prepared in IX Kinase buffer to final concentrations of 0.04 μg/mL, 46 μΜ, and 4 μΜ respectively. The mixture is allowed to incubate with the representative compounds for one hour at room temperature. All reactions were performed in duplicate. Unphosphorylated ("0% Control") and phosphorylated ("100% control") forms of Ser/Thr 18 serve as control reactions.

[0762] After incubation, diluted Development Buffer is added to the reaction and allowed to further incubate for one hour at room temperature. The plate is read at Ex 400 Em 455 to detect the coumarin signal and Ex 400 Em 520 to measure the signal (En Vision Multilabel Plate Reader, PerkinElmer).

[0763] The Emission ratio (Em) is calculated as a ratio of the coumarin (C) emission signal (at 445 nm)/Fluorescein (F) emission signal (at 520 nm). The percent phosphorylation is then calculated using the following formula: [1 - ((Em ratio X F100%)-C100%)/ ((C0%-C100%) + (Em ratio X (F100% - F0%)))] . [0764] Dose-response curves are generated and inhibitory concentration (IC50) values are calculated using non -linear regression curve fit in the Dotmatics' Studies Software (Bishops Stortford, UK).

[0765] Table 4 shows the activity of representative compounds of Formula I as provided herein.

Table 4.

Example 17.

[0766] Representative compounds were screened using the assay procedure for tau phosphorylation activity described below.

[0767] SH-SY5Y cells (human neuroblastoma) were cultured in DMEM/F-12 medium supplemented with 15% FBS, Non-essential Amino Acid and Penicillin/Streptomycin. Two days before treatment, cells were seeded onto 96 well plates at 5 x 10 4 cells/well. [0768] The above synthesized compounds were screened using the cell assay procedure to assess decrease Tau phosphorylation at Ser396 (pSer396) described below.

[0769] DMSO-resuspended compounds were dispensed to 8 wells as a serial titration from 10 μΜ to 4.6 nM final in medium and cells were exposed overnight (16-18 h) in a humidified incubator at 36.6c before harvest. Wells were visually checked for cell death or change in morphology and supernatants were tested for cytotoxicity by measurement of lactate dehydrogenase release (LDH, CytoToxOne kit, Promega) if necessary. As controls, commercially available DYRK1A inhibitors, Harmine and Indy which were shown to have good DYRK1A inhibition in the kinase assay with no CDK1 activity (EC50 18 and 53 nM respectively, 6 μΜ for CDK1) but weak EC50 in the Tau assay >10 μΜ.

[0770] Cells were lysed with RIPA buffer complemented with phosphatase and protease inhibitors then lysates were spun down at 12,000g for lO minto remove any cellular debris. Lysates are then either directly tested for pSer396 by ELISA (Life Technology, Kit KHB7031) or loaded on NuPage Bis-Tris gels for western blot analysis. Colorimetric detection of ELISA signal is performed by Cytation3 plate reader (Biotek) and the chemiluminescence signal for HRP -linked antibodies used in western blotting is detected using a Carestream Image Station. The same pSer396 antibody is used for detection of pTau in both assays.

[0771] Blot densitometry for pSer396 and β-actin were analyzed using ImageJ (NUT) and pSer396 Tau ELISA signal was used to plot, draw the curve fitting, and determine each compounds EC50 in Prism (GraphPad).

[0772] Table 5 shows the activity of representative compounds as provided herein.

Table 5.

Example 18.

[0773] Representative compounds were screened using the cell-based assay procedure for the ratio of signal of Cyclin D 1 phosphorylation at Threonine 286 (pCcnD 1) over the signal of total Cyclin Dl (CcnDl) (abbreviated: phosphoCcnD 1 to total CcnDl ratio, or PTT) as described below.

[0774] SH-SY5Y cells (human neuroblastoma, Sigma, Saint-Louis, MO) were cultured in 1 : 1 DMEM/F-12 medium supplemented with 15% FBS, 1% non-essential amino acid, and 1% penicillin/streptomycin.

[0775] SH-SY5Y cells were seeded at l .OxlO 5 cells in lmL perwell in a 24-well plate. After cell adherence was confirmed, DMSO-resuspended Samumed compounds were dispensed to eight wells as a serial dilution from 10 μΜ to 4.6 nM final concentration in medium, or at 1.1 μΜ to 4.6 nM with particularly potent compounds. Cells were exposed to Samumed compounds for 48 hours in a 37°C incubator.

[0776] Wells of compound-treated cells were visually checked for cell death before being washed with 500 per well of IX DPBS supplemented with phosphatase inhibitor diluted to IX. Cells were then lysed with 90 IX RIPA buffer supplemented with phosphatase and protease inhibitors (each diluted to a final IX). After scraping, 60 μί of cell lysate from each sample was used to dilute 4X LDS (ThermoFisher, NP0007) and 20X XT reducing agent (Bio-Rad, 1610792), each diluted to 1 X final . All samples were then denatured via boiling at 95 °C . Denatured cell sample lysates were directly tested for pCcnDl and total CcnDl signal by Western blot techniques and analysis. Specifically, denatured lysates were directly transferred to NuPAGE™ 4- 12% Bis-Tris Midi Protein Gels, 26-well gels (ThermoFisher, WG1403BOX). Gels were transferred to PVDF nitrocellulose membranes via iBlot™ Transfer Stack, nitrocellulose, regular size (ThermoFisher, IB301031). Membranes were blocked with IX TBS, 5% milk (Bio-Rad, 1706404XTU) before being probed for overnight with either anti-pCcnDl primary antibody (Cell Signaling Technology, 2978S) or anti-total CcnDl primary antibody (Cell Signaling Technology, 3300S) diluted 1:500 in IX TBS-0.05% Tween-20, 3% milk. After overnight incubation in primary antibodies, membranes were washed profusely with IX TBS-0.05% Tween-20 and probed for 1 hour with secondary anti-rabbit/HRP conjugate (Cell Signaling Technology, 7074S) diluted 1 : 1000 in IX TBS-0.05% Tween-20, 3% milk. After incubation in secondary antibody, membranes were washed with IX TBS-0.05% Tween-20.

[0777] Probed and washed membranes were developed for pCcnD 1 or total CcnD 1 signal with SuperSignal™ West Femto Maximum Sensitivity Substrate (ThermoFisher, 34096) using the ChemoDoc-It 2 (UVP) and related GelDocIt imaging software. Blots were then stripped, blocked, stained with anti- -actin/HRP conjugate antibody (Cell Signaling Technology, 5125S), developed, and imaged for housekeeping β-actin protein control as previously described. Densitometry was measured with Image J (NUT) and signal was used to plot, draw the curves fitting, and determine the EC50 values in GraphPad Prism for tested Samumed compounds. Table 6 shows the activity of selected compounds as provided herein.

Table 6.

Example 19.

[0778] Representative compounds were screened using the cell-based assay procedure for signal of Tau phosphorylation at Threonine 212 (pT212Tau) in a transiently double transfected (Dyrkla- and MAPT-overexpressing) cell type as described below.

[0779] HEK293T cells (transfectable human embryonic kidney cells) were cultured in DMEM medium supplemented with 10% FBS and 1% penicillin/streptomycin.

[0780] HEK293T cells were transiently transfected to overexpress Dyrkla and microtubule-associated protein Tau (MAPT) genes. Specifically, Dyrkla and MAPT expression vectors were obtained from OriGene (10 μg of each, catalog numbers SC314641 and RC216166, respectively). A MaxiPrep for each vector was ordered and received from GeneWiz, yielding 874.5 μg (resuspended at 1.749 μg/μL) of the Dyrkla expression vector, and 898 μg (resuspended at 1.796 μg/μL) of MAPT. HEK293T cells were seeded at l .OxlO 7 cells in 10 mL per T-75 flask. After overnight incubation, HEK293T cells in the T-75 flasks were transfected by creating a master mix of 10 μg of each expression vector per flask, with Lipofectamine™ 3000 Transfection Reagent (Invitrogen, L3000015) diluted in Opti-MEM medium according to the manufacturer's suggested protocol. One T-75 flask was designated as a no-vector negative control for pT212Tau signal.

[0781] After 4-6 hours of incubation in transfection reagents, transiently double transfected HEK293T cells were dissociated by treatment with trypsin EDTA and seeded at 1.OxlO 5 cells in 100 μί ^ per well in 96-well plates. No-vector negative controls were seeded in separate 96- well plates to avoid the risk of the negative control cells picking up the overexpression during incubation. At the time of seeding, DMSO-resuspended Samumed compounds were dispensed to eight wells as a serial dilution from 10 μΜ to 4.6nM final concentration in medium, or at 0.12 μΜ to 0.05 nM with particularly potent compounds. Cells were exposed to Samumed compounds overnight (16-18 hours) in a 37°C incubator.

[0782] 96-well flat-bottom plates were coated with 100 per well anti-HT7 capture antibody (ThermoFisher, MN1000) diluted 1 :300 in IX PBS at 4°C overnight, with shaking at 500 rpm. After overnight capture antibody incubation, coated plates were washed four times with 200 μΐ. per well of IX PBS-0.05% Tween-20, and blocked for 1 hour with 200 μΐ. per well of IX PBS with 2% BSA. After 1 hour of blocking, plates were washed four times with 200 per well of IX PBS-0.05% Tween-20, prior to sample loading.

[0783] Wells of compound-treated cells were visually checked for cell death before being washed with 200 μί per well of IX DPBS supplemented with phosphatase inhibitor diluted to IX. Cells were then lysed with 100 μί IX RIPA buffer supplemented with phosphatase and protease inhibitors (each diluted to a final IX). Cells were shaken at 500 rpm, 4°C for 20 minutes prior to further lysis (via manual scraping) and transfer to 96-well V-bottom collection plates (Corning, 3894). V-bottom plates were centrifuged at 4000 rpm, 4°C, for 15 minutes, and 100 μί of lysate supernatant from each well was directly tested for pT212Tau signal by sandwich ELISA.

[0784] Specifically, lysates were directly transferred to the coated and blocked ELISA plates for 2 hours before plates were washed four times with 200 μΐ ^ per well of IX PBS-0.05% Tween-20 and probed with 100 μΐ ^ per well of anti-pT212Tau antibody (ThermoFisher, 44-740G) diluted 1 :200 in IX PBS for 2 hours. Plates were washed four times with 200 μΐ ^ per well of IX PBS-0.05% Tween-20 and probed with 100 μΐ ^ per well of anti-rabbit/HRP conjugate (Cell Signaling Technology, 7074S) diluted 1 :600 in IX PBS for 1 hour. Plates were washed four times with 200 μΐ, per well of IX PBS-0.05% Tween-20 before 100 μΐ, per well of TMB substrate solution (ThermoFisher, N301) was added. When colour development was observed, 100 μΐ ^ per well of stop solution (ThermoFisher, N600) was added, and colorimetric detection of pT212Tau signal was read at 450nm with the Cytation 3 Cell Imaging Multi-Mode Reader (BioTek). The signal was used to plot, draw the curves fitting, and determine the EC50 values in GraphPad Prism for tested Samumed compounds. Table 7 shows the activity of selected compounds as provided herein.

Table 7.

14 0.043 33 0.033 72 0.628 145 0.810

15 0.200 44 <0.003 73 0.024 146 0.054

16 0.046 50 0.1 13 74 2.100 147 0.147

17 0.004 51 0.047 75 0.089 151 0.009

18 0.053 53 0.003 77 0.062

19 0.205 56 0.045 79 0.959

20 0.252 57 0.050 80 1 .400

Example 20.

[0785] Representative compounds were screened using the assay procedure to assess the effect on cell viability as described below.

[0786] SW480 colon carcinoma cells were transduced with a lentiviral vector expressing luciferase with a human Sp5 promoter consisting of a sequence of eight TCF/LEF binding sites. SW480 cells stably expressing the Sp5-Luc reporter gene and a hygromycin resistance gene were selected by treatment with 150 μg/mL of hygromycin for 7 days. These stably transduced SW480 cells were expanded in cell culture and used for all further screening activities. Each compound was dissolved in DMSO as a 10 mM stock and used to prepare compound source plates. Serial dilution (1 :3, 8-point dose-response curves from 10 μΜ to 0.0045 μΜ) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, CA) into 384-well white solid bottom assay plates (Greiner Bio-One) with appropriate DMSO backfill for a final DMSO concentration of 0.1%.

[0787] For the Cell Viability Assays, the cells were plated at 2,000 cells/well in 384- well plates with a DMEM medium containing 1% fetal bovine serum, and 1% Penicillin- Streptomycin and incubated for four days hours at 37°C and 5% CO2. Eight replicates of DMSO- treated cells served as controls and cells treated with compound were performed in duplicate.

[0788] After incubation, 10 μΐ ^ οΐ CellTiter-Glo (Promega) was added to each well allowed to incubate for approximately 12 minutes. This reagent "results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly proportional to the number of cells present in culture, in agreement with previous reports. The CellTiter-Glo® Assay generates a "glow-type" luminescent signal, produced by the luciferase reaction (Promega.com)".

[0789] After incubation, the plates were read at Ex 560 nm Em 590 nm (Cytation 3, BioTek). Dose-response curves were generated and EC50 concentration values were calculated using non-linear regression curve fit in the GraphPad Prism (San Diego, CA) or Dotmatics' Studies Software (Bishops Stortford, UK). For EC50 of >10 μΜ, the percent inhibition at 10 μΜ is provided.

[0790] Table 8 shows the activity of representative compounds of Formula I as provided herein. Table 8.

Example 21.

[0791] Representative compounds were screened using primary human fibroblasts (derived from IPF patients) treated with TGF-βΙ to determine their ability to inhibit the fibrotic process.

[0792] Human Fibroblast Cell Culture: Primary human fibroblasts derived from IPF patients (LL29 cells) [ 1 Xiaoqiu Liu, et.al., "Fibrotic Lung Fibroblasts Show Blunted Inhibition by cAMP Due to Deficient cAMP Response Element-Binding Protein Phosphorylation", Journal of Pharmacology and Experimental Therapeutics (2005), 315(2), 678-687; ^atts, K. L., et.al., "RhoA signaling modulates cyclin Dl expression in human lung fibroblasts; implications for idiopathic pulmonary fibrosis", Respiratory Research (2006), 7(1), 88] were obtained from American Type Culture Collection (ATCC) and expanded in F12 medium supplemented with 15% Fetal Bovine Serum and 1% Penicillin/Streptomycin. [0793] Compound Screening: Each compound was dissolved in DMSO as a 10 mM stock and used to prepare compound source plates. Serial dilution (1 :2, 1 1 -point dose-response curves from 10 μΜ to 0.94 nM) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, CA) into 384-well clear bottom assay plates (Greiner Bio-One) with appropriate DMSO backfill for a final DMSO concentration of 0.1%. LL29 cells were plated at 1,500 cells/well in 70 μίΛνεΙΙ F 12 medium supplemented with 1% Fetal Bovine Serum. TGF-βΙ (Peprotech; 20 ng/mL) was added to the plates to induce fibrosis (ref. 1 and 2 above). Wells treated with TGF-βΙ and containing DMSO were used as positive control, and cells with only DMSO were negative control. Cells were incubated at 37°C and 5% CO2 for 4 days. Following incubation for 4 days, SYTOX green nucleic acid stain (Life Technologies [Thermo Fisher Scientific]) was added to the wells at a final concentration of 1 μΜ and incubated at room temperature for 30 min. Cells were then fixed using 4% formaldehyde (Electron Microscopy Sciences), washed 3 times with PBS followed by blocking and permeabilization using 3% Bovine Serum Albumin (BSA; Sigma) and 0.3% Triton X-100 (Sigma) in PBS. Cells were then stained with antibody specific to a-smooth muscle actin (aSMA; Abeam) (ref. 1 and 2 above) in 3% Bovine Serum Albumin (BSA; Sigma) and 0.3% Triton X-100 (Sigma) in PBS, and incubated overnight at 4°C. Cells were then washed 3 times with PBS, followed by incubation with Alexa Flor-647 conjugated secondary antibody (Life Technologies [Thermo Fisher Scientific]) and DAPI in 3% Bovine Serum Albumin (BSA; Sigma) and 0.3% Triton X-100 (Sigma) in PBS at room temperature for 1 hour. Cells were then washed 3 times with PBS and plates were sealed for imaging. aSMA staining was imaged by excitation at 630 nm and emission at 665 nm and quantified using the Compartmental Analysis program on the Celllnsight CX5 (Thermo Scientific). Dead or apoptotic cells were excluded from analysis based on positive SYTOX green staining. % of total cells positive for aSMA were counted in each well and normalized to the average of 1 1 wells treated with TGF-βΙ on the same plate using Dotmatics' Studies Software. The normalized averages (fold change over untreated) of 3 replicate wells for each compound concentration were used to create dose-responses curves and EC50 values were calculated using non-linear regression curve fit in the Dotmatics' Studies Software. For EC50 of >10 μΜ, the percent inhibition at 10 μΜ is provided.

[0794] Table 9 shows the activity of representative compounds of Formula I as provided herein.

Table 9. 5 2.450 43 0.217 81 5.459 119 >10 (7.7%)

6 0.197 44 0.401 82 3.154 120 0.620

7 4.349 45 0.622 83 >10 (29.6%) 121 0.710

8 0.398 46 0.682 84 >10 (35.4%) 122 >10 (4.8%)

9 2.266 47 0.556 85 1 .849 123 1 .040

10 >10 (6.2%) 48 4.880 86 0.204 124 >10 (5.2%)

11 3.576 49 >10 (33.5%) 87 1 .924 125 >10 (43.6%)

12 1 .187 50 >10 (31 .1 %) 88 >10 (31 .9%) 126 1 .454

13 0.805 51 4.232 89 3.910 127 0.272

14 2.235 52 >10 (37.1 %) 90 0.037 128 0.102

15 1 .525 53 3.068 91 0.019 129 0.699

16 >10 (6.7%) 54 >10 (11 .2%) 92 3.216 130 0.941

17 0.471 55 2.058 93 2.241 131 0.604

18 5.970 56 5.132 94 0.103 132 1 .351

19 1 .210 57 2.158 95 0.091 133 4.029

20 3.136 58 >10 (44.6%) 96 0.152 134 2.753

21 3.405 59 0.705 97 0.311 135 2.481

22 2.063 60 2.604 98 0.032 136 0.933

23 1 .630 61 >10 (4.6%) 99 0.031 137 >10 (34.2%)

24 2.767 62 >10 (13.2%) 100 0.045 138 0.078

25 >10 (44.8%) 63 2.386 101 0.046 139 0.148

26 1 .326 64 1 .997 102 0.094 140 1 .675

27 0.979 65 2.440 103 0.547 141 1 .514

28 1 .517 66 >10 (10.6%) 104 0.284 142 >10 (8.8%)

29 4.016 67 1 .034 105 0.226 143 2.442

30 >10 (24.9%) 68 3.385 106 0.027 144 4.595

31 0.450 69 6.729 107 1 .274 145 6.522

32 8.403 70 0.634 108 3.386 146 >10 (23.8%)

33 0.481 71 >10 (43.4%) 109 0.027 147 >10 (24.4%)

34 0.507 72 1 .828 110 0.345 148 1 .189

35 2.803 73 >10 (43.1 %) 111 1 .525 149 3.396

36 0.292 74 >10 (18.7%) 112 0.975 150 >10 (42.0%)

37 >10 (39.5%) 75 1 .368 113 >10 (27.3%) 151 >10 (21 .2%)

38 0.603 76 >10 (13.1 %) 114 0.175

Example 22.

[0795] Representative compounds were screened using the cell-based assay procedure for secreted cytokines in a Lipopolysaccharide-stimulated mouse glial cell line described below.

[0796] BV-2 cells (mouse microglial cells) were cultured in 1 : 1 DMEM medium supplemented with 10% FBS, and 1% penicillin/streptomycin.

[0797] BV-2 cells are plated at 35,000 cells/well in a volume of 100 μΐ for at least 4 hours before compounds are added. DMSO-resuspended compounds were first dispensed in a 96- well plate and serial diluted from 10 μΜ to 4.6 nM final concentration in medium. Compounds were added to cells overnight. 250 ng/mL of lipopolysaccharide (Escherichia coli 0111:B4, SIGMA) was added for 5 hours. Supernatant is removed and saved for further cytokine detection. The original plates with seeded cells were tested for cytotoxicity by measure of adenosine triphosphate (ATP) release by adding CellTiter-Glo ® diluted 1 :4 in distilled water (G7573, Promega) and transferring lysed cells to a completely black 96 -well plate to be read with the Cytation3. Supernatant was then diluted 1:2 with a diluent from V-PLEX cytokine Kit and directly tested for the secreted cytokines TNFa, IL-6 and KC-GRO using electrochemiluminescence (Meso Scale Discovery). The standard curve for each cytokine was used to convert the electrochemiluminescent signal into pg of protein per mL. The signal was used to plot, draw the curve fitting, and determine each compounds EC50 in Prism (GraphPad).

[0798] Table 10 shows the activity of representative compounds of Formula I as provided herein.

Table 10.

Example 23.

[0799] Representative compounds were screened using the following assay procedure to determine their ability to inhibit IL-6 and therefore demonstrate their anti -inflammatory properties.

[0800] Human Peripheral Blood Mononuclear Cells: Fresh Normal PB MNC (Catalog # PB001, AllCells, Alameda, CA) were shipped overnight at 4°C and resuspended in Roswell Park Memorial Institute (RPMI) 1640 Medium, with GlutaMAX Supplement (Catalog #61870127, ThermoFisher Scientific, Waltham, MA) supplemented with 1% Penicillin- Streptomycin (Catalog# 15140163, ThermoFisher Scientific, Waltham, MA) and 1% fetal bovine serum (FBS) ( Catalog # 16140089, ThermoFisher Scientific, Waltham, MA) assay media.

[0801] Compound Screening: Fresh normal human peripheral blood mononuclear cells (huPBMCs) were resuspended in 1% FBS-RPMI assay media with 1% Penicillin- Streptomycin 1% to a cell concentration of 1 x 10e6 cells/mL. Each compound was dissolved in DMSO (Catalog # D8418- 100ml, Sigma-Aldrich, St. Louis, MO) as a 10 mM stock and used to prepare compound source plates. Serial dilution (1 :3, 10-point dose-response curves starting from 10 μΜ) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, CA) into 384-well white Proxiplate-Plus assay plates (Catalog #6008289, PerkinElmer, Shelton, CT) with appropriate DMSO backfill for a final DMSO concentration of 0.25%. huPBMCs were plated at 5000 cells/well in the 384-well Proxiplate-Plus assay plates and incubated at 37°C-5% CO2 for 2 hours. 50 ng/mL of Lipopolysaccharides from Escherichia coli 0111 :B4 (Catalog #L5293-2ML, Sigma-Aldrich, St. Louis, MO) was added after 2 hours and cells were incubated for another 22 hours at 37°C-5% CO 2 . After 22 hour incubation, a mixture of anti-IL6 XL665 and anti-IL-6 Cryptate diluted in reconstitution buffer (Catalog #62IL6PEC, Cisbio Inc., Bedford, MA) was added to each well. Following incubation for 3 hours at room temperature, Homogeneous Time- Resolved Fluorescence (HTRF) was measured using the Envision (Perkin Elmer, Shelton, CT) at 665 nm and 620 nM. The ratio of fluorescence at 665 nm to 620 nm was used as a readout for IL- 6 quantification. All samples were processed in duplicate. Readings were normalized to DMSO treated cells and normalized activities were utilized for EC50 calculations. EC50 was determined using software generated by Dotmatics Limited (Windhill Bishops Stortford Herts, UK) using the Levenberg-Marquardt 4 parameter fitting procedure with finite different gradients. For EC50 of >10 μΜ, the percent inhibition at 10 μΜ is provided.

[0802] Table 11 shows the activity of representative compounds of Formula I as provided herein.

Table 11.

Compound ECso (μΜ) Compound ECso (μΜ) Compound ECso (μΜ) Compound ECso (μΜ)

1 7.155 39 3.390 77 >10 (9.6%) 115 3.193

2 1 .326 40 >10 (3.7%) 78 >10 (8.9%) 116 0.358

3 >10 (5.1 %) 41 1 .1 14 79 >10 (3.9%) 117 1 .358

4 6.754 42 1 .100 80 >10 (4.9%) 118 0.932

5 >10 (4.1 %) 43 2.055 81 >10 (1.6%) 119 >10 (6.1 %)

6 1 .028 44 0.375 82 >10 (9.9%) 120 8.887

7 9.261 45 1 .131 83 >10 (5.0%) 121 1 .913

8 1 .971 46 1 .384 84 >10 (5.7%) 122 3.149

9 >10 (3.9%) 47 5.994 85 6.707 123 >10 (20.9%)

10 >10 (3.7%) 48 9.678 86 1 .127 124 2.091

11 >10 (8.5%) 49 >10 (0%) 87 0.377 125 >10 (4.5%)

12 9.862 50 8.952 88 0.400 126 2.721

13 2.823 51 2.995 89 0.390 127 1 .558

14 9.028 52 >10 (17.6%) 90 0.463 128 0.678

15 4.821 53 3.254 91 0.446 129 3.359

16 >10 (43.1 %) 54 >10 (17.9%) 92 0.374 130 3.925

17 9.351 55 3.226 93 0.811 131 2.901

18 >10 (5.8%) 56 9.503 94 0.137 132 >10 (8.5%)

19 9.147 57 3.800 95 0.211 133 3.281

20 >10 (3.6%) 58 9.664 96 0.312 134 8.812

21 >10 (6.3%) 59 >10 (4.7%) 97 0.184 135 2.1 11

22 >10 (22.9%) 60 9.091 98 0.087 136 9.057

23 >10 (8.6%) 61 >10 (26.4%) 99 0.125 137 >10 (3.9%)

24 >10 (2.8%) 62 >10 (8.0%) 100 0.223 138 2.764

25 >10 (4.6%) 63 >10 (7.9%) 101 0.260 139 0.361

26 1 .321 64 >10 (5.7%) 102 0.401 140 3.399

27 9.157 65 >10 (.0%) 103 0.146 141 8.995

28 7.429 66 >10 (8.1 %) 104 1 .167 142 >10 (7.8%)

29 >10 (4.3%) 67 >10 (6.3%) 105 0.207 143 >10 (8.2%)

30 >10 (5.2%) 68 8.765 106 0.369 144 >10 (9.4%)

31 8.938 69 >10 (4.4%) 107 0.1 19 145 >10 (5.9%)

32 >10 (5.1 %) 70 9.333 108 0.385 146 >10 (.0%)

33 3.651 71 >10 (11 .0%) 109 0.674 147 >10 (14.7%)

34 2.145 72 >10 (10.0%) 110 1 .178 148 1 .782

35 1 .012 73 0.320 111 0.106 149 3.785

36 1 .216 74 >10 (4.9%) 112 1 .055 150 >10 (10.7%)

37 0.662 75 >10 (27.5%) 113 0.383 151 >10 (6.0%)

38 0.765 76 >10 (1 .6%) 114 0.334