Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FUSED BICYCLIC RAF INHIBITORS AND METHODS FOR USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2022/023447
Kind Code:
A2
Abstract:
The present disclosure generally relates to improved synthesis of fused bicyclic Raf inhibitors of formula (I), (I-A), (I-B), (II), or (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. The disclosure also relates to method of using the compound of formula (I), (I-A), (I-B), (II), or (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, for treating diseases such as cancer, including colorectal cancer.

Inventors:
BELFIELD ANDREW (IE)
JONES CLIFFORD DAVID (IE)
MARGATHE JEAN-FRANÇOIS (IE)
COLLETTO CHIARA (IE)
Application Number:
PCT/EP2021/071212
Publication Date:
February 03, 2022
Filing Date:
July 28, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JAZZ PHARMACEUTICALS IRELAND LTD (IE)
International Classes:
A61P35/00; A61K31/4178; A61K31/4375; A61K31/4439; C07D405/14; C07D417/14; C07D471/04; C07D519/00
Foreign References:
US10183939B22019-01-22
US3932657A1976-01-13
US4311712A1982-01-19
US4743449A1988-05-10
US4452747A1984-06-05
US4830858A1989-05-16
US4921757A1990-05-01
US5013556A1991-05-07
US4235871A1980-11-25
US4744989A1988-05-17
US5616341A1997-04-01
US5736155A1998-04-07
US5785987A1998-07-28
US5939096A1999-08-17
US7238367B22007-07-03
US7744921B22010-06-29
US4756910A1988-07-12
US5077056A1991-12-31
US5225212A1993-07-06
US8518437B22013-08-27
US4726955A1988-02-23
US5766635A1998-06-16
US6117454A2000-09-12
US8137699B22012-03-20
US7850990B22010-12-14
Other References:
"Pharmaceuticals—The Science of Dosage Form Designs", 1988, M. E. AULTON
Attorney, Agent or Firm:
COOLEY (UK) LLP (GB)
Download PDF:
Claims:
What is Claimed: 1. A compound of formula (I), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: ring A is a 5-membered heterocycle or heteroaryl containing 1, 2, or 3, nitrogen atom as a ring member; one of R1 or R2 is a substituted C1-8 alkyl, an unsubstituted C5-8 alkyl, a substituted or unsubstituted C1-8 haloalkyl, a substituted or unsubstituted aryl, a substituted or unsubstituted heterocyclyl, or a substituted heteroaryl, and the other R1 or R2 is H; or alternatively, R1 and R2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted; and wherein when ring A is an imidazole, then the substituted aryl is not , ; and wherein when ring A is an imidazole, R1 and R2 together with the atoms to which they are attached do not form an unsubstituted phenyl ring. 2. The compound of claim 1, wherein ring A is an imidazole, a pyrazole, or a triazole. 3. The compound of claim 1 or 2, wherein ring A is , or 4. The compound of claim 1 having the structure of formula (I-A) or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: one of R1 or R2 is a substituted or unsubstituted C5-8 alkyl, a substituted or unsubstituted C1-8 haloalkyl, a substituted aryl, a substituted or unsubstituted heterocyclyl, or a substituted heteroaryl, and the other R1 or R2 is H; or alternatively, R1 and R2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted; and wherein the substituted aryl is not , or ; and wherein R1 and R2 together with the atoms to which they are attached do not form an unsubstituted phenyl.

5. The compound of any one of claims 1-4, wherein one or more substituent is selected from halogen, -ORA, -NRARB, -SO2RC, -SORC, -CN, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or - C(O)C1-6 alkyl; wherein: RA and RB are each independently, H, C1-6 alkyl or C1-6 haloalkyl; RC is C1-6 alkyl or C1-6 haloalkyl; and wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -ORA, -CN, -SORC, –NRARB, or –NRDRE; wherein RD and RE together with the N atom to which they are attached forms a 5- or 6- membered saturated or partially unsaturated ring containing 1 or 2 heteroatoms selected from N, O, or S; wherein the saturated or partially unsaturated ring is optionally substituted with C1-6 alkyl. 6. The compound of any one of claims 1-5, wherein R1 and R2 together with the atoms to which they are attached forms a 5- or 6-membered partially unsaturated or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted. 7. The compound of claim 6, wherein R1 and R2 together with the atoms to which they are attached forms a 6-membered partially unsaturated or unsaturated ring containing 0 or 1 nitrogen atom in the ring, wherein the ring is substituted or unsubstituted. 8. The compound of claim 6 or 7, wherein R1 and R2 together with the atoms to which they are attached forms a substituted or unsubstituted phenyl ring or a substituted or unsubstituted pyridyl ring. 9. The compound of claim 6 or 7, wherein R1 and R2 together with the atoms to which they are attached forms a substituted or unsubstituted tetrahydropyridyl ring. 10. The compound of claim 1, wherein the compound has the structure of formula (I-B), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein one of R1 or R2 is a substituted aryl, a substituted or unsubstituted heterocyclyl, or a substituted heteroaryl; and wherein the substituted aryl is not , or 11. The compound of any one of claims 1-10, wherein R1 is a substituted aryl, a substituted or unsubstituted heterocyclyl, or a substituted heteroaryl. 12. The compound of any one of claims 1-11, wherein R1 is a substituted phenyl or a substituted 5- or 6-membered N-heteroaryl. 13. The compound of any one of claims 1-12, wherein R1 is a substituted phenyl, a substituted pyridyl, a substituted pyrazole, a substituted pyrimidinyl, or a substituted thiophenyl. 14. The compound of claim 11, wherein R1 is a monocyclic or bicyclic substituted aryl or a monocyclic or bicyclic substituted heteroaryl. 15. The compound of claim 1, wherein R1 is a monocyclic or bicyclic, substituted or unsubstituted aryl.

16. The compound of claim 14 or 15, wherein the bicyclic aryl or the bicyclic heteroaryl is a fused bicyclic aryl or a fused bicyclic heteroaryl. 17. The compound of claim 11, wherein R1 is a substituted indazole or a substituted benzoimidazole. 18. The compound of any one of claims 1-10, wherein R1 is an unsubstituted or substituted heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. 19. The compound of any one of claims 1-10, wherein R1 is a substituted or unsubstituted tetrahydropyran. 20. The compound of any one of claims 1-10, wherein R1 is a substituted or unsubstituted C5-6 alkyl. 21. The compound of any one of claims 1-10, R1 is substituted with 1 or 2 substituents. 22. The compound of any one of claims 1-21, wherein the substituent is selected from halogen, -ORA, -NRARB, -SO2RC, -SORC, -CN, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or -C(O)C1-6 alkyl; wherein: RA and RB are each independently, H, C1-6 alkyl or C1-6 haloalkyl; RC is C1-6 alkyl or C1-6 haloalkyl; and wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -ORA, -CN, -SORC, or -NRARB. 23. The compound of any one of claims 1-22, wherein the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -C(O)CH3, -CN, -OH, -NH2, -NH(C1-3 alkyl), -N(C1-3 alkyl)2, -CH2NH2, -CH2NH(C1-3 alkyl), or -CH2N(C1-3 alkyl)2.

24. The compound of any one of claims 1-23, wherein the substituent on the substituted aryl or the substituted heteroaryl is selected from methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t- butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, - CH2CF3, -C(O)CH3, -CN, -OH, -NH2, -NH(C1-3 alkyl), -N(C1-3 alkyl)2, -CH2NH2, -CH2NH(C1-3 alkyl), or -CH2N(C1-3 alkyl)2. 25. The compound of any one of claims 1-24, wherein the compound has (R) or (S) stereochemistry at the carbon indicated by *. 26. The compound of claim 1, wherein the compound is selected from Table A-1, Table A-2, or Table A-3, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. 27. A compound of formula (II), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: X1 and X2 are each N or CH; R1 is a substituted C1-8 alkyl, an unsubstituted C5-8 alkyl, a substituted or unsubstituted C1-8 haloalkyl, a substituted or unsubstituted aryl, a substituted or unsubstituted heterocyclyl, or a substituted or unsubstituted heteroaryl; and R4 is –NRFC(O)R5, –NRFC(O)CH2R5, –NRFC(O)CH(CH3)R5, or –NRFR5; R5 is a substituted or unsubstituted group selected from alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl; and RF is H or C1-3 alkyl. 28. The compound of claim 27, wherein one of X1 and X2 is N. 29 The compound of claim 27 wherein X1 and X2 are both CH

30. The compound of any one of claims 27-29, wherein R1 is a substituted or unsubstituted aryl, a substituted or unsubstituted heterocyclyl, or a substituted or unsubstituted heteroaryl. 31. The compound of any one of claims 27-30, wherein R1 is a substituted or unsubstituted phenyl, a substituted or unsubstituted pyridyl, a substituted or unsubstituted pyrazole, a substituted or unsubstituted pyrimidinyl, or a substituted or unsubstituted thiophenyl. 32. The compound of any one of claims 27-31, wherien R4 is –NHC(O)R5, –NHC(O)CH2R5, –NHC(O)CH(CH3)R5, or –NHR5. 33. The compound of any one of claims 27-32, wherein R5 is a substituted or unsubstituted group selected from alkyl, 3-6 membered carbocyclyl, phenyl, 3-6 membered heterocyclyl, or 5-6 membered heteroaryl. 34. The compound any one of claims 27-33, wherein R5 is a substituted or unsubstituted group selected from methyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidine, pyrrolidine, piperidine, piperazine, morpholine, pyridine, thiazole, imidazole, pyrazole, or triazole. 35. The compound of any one of claims 27-34, wherein RF is H or methyl. 36. The compound of any one of claims 27-35, wherein the substitutent is selected from halogen, -ORA, -NRARB, -SO2RC, -SORC, -CN, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or - C(O)C1-6 alkyl; wherein RA and RB are each independently, H, C1-6 alkyl or C1-6 haloalkyl; and RC is C1-6 alkyl or C1-6 haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -ORA, -CN, -SORC, or - NRARB. 37. The compound of any one of claims 27-35, wherein the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -C(O)CH3, -CN, -OH, -NH2, -NH(C1-3 alkyl), -N(C1-3 alkyl)2, -CH2NH2, -CH2NH(C1-3 alkyl), or -CH2N(C1-3 alkyl)2. 38. The compound of any one of claims 27-37, wherein R5 is substituted with one or more substituents selected from halogen, methyl, ethyl, propyl, isopropyl, -CN, -OH, or -NH2. 39. The compound of any one of claims 27-38, wherein the compound has (R) or (S) stereochemistry at the carbon indicated by *. 40. The compound of claim 27, wherein the compound is selected from Table B, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. 41. A compound of formula (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, wherein: R6 is –C(O)NRFR5, –C(O)NRFCH2R5, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl; R5 is a substituted or unsubstituted group selected from carbocyclyl, aryl, heterocyclyl, or heteroaryl; and RF is H or C1-3 alkyl. 42. The compound of claim 41, wherein R6 is –C(O)NHR5 or –C(O)NHCH2R5. 43. The compound of claim 41 or 42, wherein R5 is a substituted or unsubstituted aryl.

44. The compound of claim 41, wherein R6 is a substituted or unsubstituted aryl or a substituted or unsubstituted heteroaryl. 45. The compound of claim 44, wherein R6 is a monocyclic or bicyclic substituted aryl or a monocyclic or bicyclic substituted heteroaryl. 46. The compound of claim 45, wherein the bicyclic aryl or the bicyclic heteroaryl is a fused bicyclic aryl or fused bicyclic heteroaryl. 47. The compound of claim any one of claims 44-46, wherein R6 is a substituted or unsubstituted indazole or a substituted or unsubstituted benzoimidazole. 48. The compound of any one of claims 41-47, wherein the substitutent is selected from halogen, -ORA, -NRARB, -SO2RC, -SORC, -CN, C1-6 alkyl, C1-6 haloalkyl, C3-6 cycloalkyl, or - C(O)C1-6 alkyl; wherein RA and RB are each independently, H, C1-6 alkyl or C1-6 haloalkyl; and RC is C1-6 alkyl or C1-6 haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -ORA, -CN, -SORC, or - NRARB. 49. The compound of any one of claims 41-47, wherein the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, -CH2CF3, -C(O)CH3, -CN, -OH, -NH2, -NH(C1-3 alkyl), -N(C1-3 alkyl)2, -CH2NH2, -CH2NH(C1-3 alkyl), or -CH2N(C1-3 alkyl)2. 50. The compound of any one of claims 41-49, wherein R6 is a substituted aryl, a substituted heterocyclyl, or a substituted heteroaryl; wherein the substitutent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH2F, -CHF2, -CF3, -CH2CH2F, - CH2CHF2, -CH2CF3, -C(O)CH3, -CN, -OH, -NH2, -NH(C1-3 alkyl), -N(C1-3 alkyl)2, -CH2NH2, - CH2NH(C1-3 alkyl), -CH2N(C1-3 alkyl)2, optionally substituted phenyl, or optionally substituted heteroaryl. 51. The compound of any one of claims 41-50, wherein R6 is a substituted aryl or a substituted heteroaryl, wherein the substituent is a phenyl or a pyrazole substituted with one or two C1-3 alkyl groups. 52. The compound of claim 41 selected from Table C, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. 53. A compound having the structure , or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. 54. A pharmaceutical composition comprising a compound of any one of claims 1-53 and a pharmaceutically acceptable excipient or carrier. 55. The pharmaceutical composition of claim 54, further comprising an additional therapeutic agent. 56. The pharmaceutical composition of claim 55, wherein the additional therapeutic agent is an antiproliferative or an antineoplastic drug, a cytostatic agent, an anti-invasion agent, an inhibitor of growth factor function, an antiangiogenic agent, a steroid, a targeted therapy agent, or an immunotherapeutic agent. 57. A method of treating a condition which is modulated by a RAF kinase, comprising administering an effective amount of the compound of any one of claims 1-53 to a subject in need thereof

58. The method of claim 57, wherein the condition is treatable by the inhibition of one or more Raf kinases. 59. The method of claim 57 or 58, wherein the condition is cancer, sarcoma, melanoma, skin cancer, haematological tumors, lymphoma, carcinoma or leukemia. 60. The method of claim 57 or 58, wherein the condition is Barret's adenocarcinoma; biliary tract carcinomas; breast cancer; cervical cancer; cholangiocarcinoma; central nervous system tumors; primary CNS tumors; glioblastomas, astrocytomas; glioblastoma multiforme; ependymomas; seconday CNS tumors (metastases to the central nervous system of tumors originating outside of the central nervous system); brain tumors; brain metastases; colorectal cancer; large intestinal colon carcinoma; gastric cancer; carcinoma of the head and neck; squamous cell carcinoma of the head and neck; acute lymphoblastic leukemia; acute myelogenous leukemia (AML); myelodysplastic syndromes; chronic myelogenous leukemia; Hodgkin's lymphoma; non-Hodgkin's lymphoma; megakaryoblastic leukemia; multiple myeloma; erythroleukemia; hepatocellular carcinoma; lung cancer; small cell lung cancer; non- small cell lung cancer; ovarian cancer; endometrial cancer; pancreatic cancer; pituitary adenoma; prostate cancer; renal cancer; metastatic melanoma or thyroid cancer. 61. A method of treating cancer, comprising administering an effective amount of the compound of any one of claims 1-53 to a subject in need thereof. 62. The method of claim 61, wherein the cancer comprises at least one mutation of the BRAF kinase. 63. The method of claim 62, wherein the cancer comprises a BRAFV600E mutation. 64. The method of claim 62, wherein the cancer is melanomas, thyroid cancer, Barret's adenocarcinoma, biliary tract carcinomas, breast cancer, cervical cancer, cholangiocarcinoma, central nervous system tumors, glioblastomas, astrocytomas, ependymomas, colorectal cancer, large intestine colon cancer, gastric cancer, carcinoma of the head and neck, hematologic cancers, leukaemia, acute lymphoblastic leukaemia, myelodysplastic syndromes, chronic myelogenous leukaemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, megakaryoblastic leukaemia, multiple myeloma, hepatocellular carcinoma, lung cancer, ovarian cancer, pancreatic cancer, pituitary adenoma, prostate cancer, renal cancer, sarcoma, uveal melanoma or skin cancer. 65. The method of claim 63, wherein the cancer is BRAFV600E melanoma, BRAFV600E colorectal cancer, BRAFV600E papillary thyroid cancers, BRAFV600E low grade serous ovarian cancers, BRAFV600E glioma, BRAFV600E hepatobiliary cancers, BRAFV600E hairy cell leukaemia, BRAFV600E non-small cell cancer, or BRAFV600E pilocytic astrocytoma. 66. The method of any one of claims 61-65, wherein the cancer is colorectal cancer.

Description:
FUSED BICYCLIC RAF INHIBITORS AND METHODS FOR USE THEREOF

CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of U.S. Provisional Application No. 63/057,536, filed July 28, 2020, the disclosures of which are incorporated by reference herein in its entirety.

FIELD OF THE INVENTION

[0002] The present disclosure generally relates to novel RAF inhibitors and method of use thereof for treatment of various diseases, including but not limited to, colorectal cancer and melanoma.

BACKGROUND OF THE INVENTION

[0003] Mutations leading to uncontrolled signaling via the RAS-RAF-MAPK pathway are seen in more than one third of all cancers. The RAF kinases (A-RAF, B-RAF and C-RAF) are an integral part of this pathway, with B-RAF mutations commonly seen in the clinic. Although most B-RAF V600E mutant skin cancers are sensitive to approved B-RAF selective drugs, B- RAF V600E mutant colorectal cancers are surprisingly insensitive to these agents as monotherapy due to the functions of other RAF family members and require combination therapy. B-RAF selective therapies fail to show clinical benefit against atypical B-RAF (non- V600E), other RAF and RAS driven tumors.

[0004] U.S. Patent No. 10,183,939 discloses racemic Raf inhibitors that demonstrated binding affinity for B-RAF V600E and C-RAF, the disclosures of which are hereby incorporated by reference in its entirety. These pan-RAF inhibitors are identified to be promising candidates in overcome resistance mechanisms associated with clinically approved B-RAF selective drugs.

SUMMARY OF THE INVENTION

[0005] The present disclosure relates to compounds that are Raf inhibitors. In embodiments, the compounds of the disclosure are pan-Raf inhibitors.

[0006] The present disclosure relates to a compound of formula (I), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, [0007] wherein:

[0008] ring A is a 5-membered heterocycle containing 1, 2, or 3, nitrogen atom as a ring member;

[0009] one of R 1 or R 2 is selected from substituted C 1-8 alkyl, unsubstituted C 5-8 alkyl, substituted or unsubstituted C 1-8 haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl, and the other R 1 or R 2 is H;

[0010] or alternatively, R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted; and

[0011] wherein when ring A is an imidazole, then the substituted aryl is not

[0012] wherein when ring A is an imidazole, R 1 and R 2 together with the atoms to which they are attached do not form an unsubstituted phenyl ring.

[0013] In embodiments of compound of formula (I), ring A is imidazole, pyrazole, or triazole.

In embodiments,

[0014] In embodiments, R 1 is a monocyclic or bicyclic, substituted or unsubstituted aryl. In embodiments, the bicyclic aryl is a fused bicyclic aryl.

[0015] The present disclosure also relates to a compound of formula (I-A), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0016] wherein:

[0017] one of R 1 or R 2 is selected from substituted or unsubstituted C 5-8 alkyl, substituted or unsubstituted C 1-8 haloalkyl, substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl, and the other R 1 or R 2 is H;

[0018] or alternatively, R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted; and

[0019] wherein substituted aryl is not , or ; and

[0020] wherein R 1 and R 2 together with the atoms to which they are attached do not form an unsubstituted phenyl.

[0021] In embodiments of compound of formula (I) or (I- A), R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered partially unsaturated or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a 6-membered partially unsaturated or unsaturated ring containing 0 or 1 nitrogen atom in the ring, wherein the ring is substituted or unsubstituted. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a substituted or unsubstituted phenyl ring or a substituted or unsubstituted pyridyl ring. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a substituted or unsubstituted tetrahydropyridyl ring.

[0022] The present disclosure also relates to a compound of formula (I-B), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, [0023] wherein:

[0024] one of R 1 or R 2 is selected from substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl; and

[0025] wherein substituted

[0026] In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl. In embodiments, R 1 is substituted phenyl or substituted 5- or 6-membered N-heteroaryl. In embodiments, R 1 is substituted phenyl, substituted pyridyl, substituted pyrazole, substituted pyrimidinyl, or substituted thiophenyl. In embodiments, R 1 is a monocyclic or bicyclic substituted aryl or a monocyclic or bicyclic substituted heteroaryl. In embodiments, the bicyclic aryl or the bicyclic heteroaryl is a fused bicyclic aryl or fused bicyclic heteroaryl. In embodiments, R 1 is substituted indazole or substituted benzoimidazole. In embodiments,

[0027] In embodiments of formula (I), (I-A), or (I-B), R 1 is unsubstituted or substituted heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R 1 is substituted or unsubstituted tetrahydropyran.

[0028] In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted or unsubstituted C 5-6 alkyl.

[0029] In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted with 1 or 2 substituents. [0030] In embodiments of formula (I), (I-A), or (I-B), the substituent is selected from halogen, -OR A , -NR A R B , -SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; wherein:

[0031] R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and

[0032] R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; and [0033] wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -OR A , -CN, -SOR c , -NR A R B , or-NR D R E ; [0034] R D and R E together with the N atom to which they are attached forms a 5- or 6- membered saturated or partially unsaturated ring containing 1 or 2 heteroatoms selected from N, O, or S; wherein the saturated or partially unsaturated ring is optionally substituted with C 1-6 alkyl.

[0035] In embodiments of formula (I), (I-A), or (I-B), the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, - NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 . [0036] In embodiments of formula (I), (I-A), or (I-B), the substituent on the substituted aryl or the substituted heteroaryl is selected from methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t- butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1- 3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 .

[0037] In embodiments of formula (I), (I-A), or (I-B), the compound has (R) or (S) stereochemistry at the carbon indicated by *.

[0038] The present disclosure also relates to a compound selected from Table A-1, Table A-2, or Table A-3, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0039] The present disclosure also relates to a compound of formula (II), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0040] wherein:

[0041] X 1 and X 2 are each N or CH;

[0042] R 1 is selected from substituted C 1-8 alkyl, unsubstituted C 5-8 alkyl, substituted or unsubstituted C 1-8 haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl; and

[0043] R 4 is -NR F C(O)R 5 , -NR F C(O)CH 2 R 5 , -NR F C(O)CH(CH 3 )R 5 , or -NR F R 5 ; [0044] R 5 is substituted or unsubstituted group selected from alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl; and [0045] R F is selected from H or C 1-3 alkyl.

[0046] In embodiments of formula (II), one of X 1 and X 2 is N. In embodiments, X 1 and X 2 are both CH.

[0047] In embodiments of formula (II), R 1 is substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl. In embodiments, R 1 is substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrimidinyl, or substituted or unsubstituted thiophenyl.

[0048] In embodiments of formula (II), R 4 is -NHC(O)R 5 , -NHC(O)CH 2 R 5 , - NHC(O)CH(CH 3 )R 5 , or -NHR 5 .

[0049] In embodiments of formula (II), R 5 is substituted or unsubstituted group selected from alkyl, 3-6 membered carbocyclyl, phenyl, 3-6 membered heterocyclyl, or 5-6 membered heteroaryl. In embodiments, R 5 is substituted or unsubstituted group selected from methyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, azetidine, pyrrolidine, piperidine, piperazine, morpholine, pyridine, thiazole, imidazole, pyrazole, or triazole.

[0050] In embodiments of formula (II), R F is H or methyl.

[0051] In embodiments of formula (II), the substitutent is selected from halogen, -OR A , - NR A R B , -SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; [0052] wherein R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and

[0053] R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -OR A , -CN, -SOR c , or -NR A R B .

[0054] In embodiments of formula (II), the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 .

[0055] In embodiments of formula (II), R 5 is substituted with one or more substituents selected from halogen, methyl, ethyl, propyl, isopropyl, -CN, -OH, or -NH 2 .

[0056] In embodiments of formula (II), the compound has (R) or (S) stereochemistry at the carbon indicated by *.

[0057] The present disclosure also relates to a compound selected from Table B, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof. [0058] The present disclosure also relates to a compound of formula (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0059] wherein:

[0060] R 6 is -C(O)NR F R 5 , -C(O)NR F CH 2 R 5 , substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl;

[0061] R 5 is substituted or unsubstituted group selected from carbocyclyl, aryl, heterocyclyl, or heteroaryl; and

[0062] R F is selected from H or C 1-3 alkyl.

[0063] In embodiments of formula (III), R 6 is -C(O)NHR 5 or -C(O)NHCH 2 R 5 .

[0064] In embodiments of formula (III), R 5 is substituted or unsubstituted aryl. In embodiments, R 5 is substituted or unsubstituted phenyl.

[0065] In embodiments of formula (III), R 6 is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. In embodiments, R 6 is a monocyclic or bicyclic substituted aryl or a monocyclic or bicyclic substituted heteroaryl. In embodiments, the bicyclic aryl or the bicyclic heteroaryl is a fused bicyclic aryl or fused bicyclic heteroaryl. In embodiments, R 6 is substituted or unsubstituted indazole or substituted or unsubstituted benzoimidazole.

[0066] In embodiments of formula (III), the substitutent is selected from halogen, -OR A , - NR A R B , -SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; [0067] wherein R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and

[0068] R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -OR A , -CN, -SOR c , or -NR A R B .

[0069] In embodiments of formula (III), the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 .

[0070] In embodiments of formula (III), R 6 is substituted aryl, substituted heterocyclyl, or substituted heteroaryl; wherein the substitutent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), -CH 2 N(C 1-3 alkyl) 2 , optionally substituted phenyl, or optionally substituted heteroaryl. In embodiments, R 6 is a substituted aryl or a substituted heteroaryl, wherein the substituent is a phenyl or a pyrazole substituted with one or two C 1-3 alkyl groups.

[0071] The present disclosure also relates to a compound selected from Table C, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0072] The present disclosure also relates to a compound selected from Table D, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0073] The present disclosure relates to a pharmaceutical composition comprising any one of the compounds as disclosed herein and a pharmaceutically acceptable excipient or carrier. [0074] In embodiments of the pharmaceutical composition, the composition further comprises an additional therapeutic agent. In embodiments, the additional therapeutic agent is selected from an antiproliferative or an antineoplastic drug, a cytostatic agent, an anti-invasion agent, an inhibitor of growth factor function, an anti angiogenic agent, a steroid, a targeted therapy agent, or an immunotherapeutic agent.

[0075] The present disclosure relates to a method of treating a condition which is modulated by a RAF kinase, comprising administering an effective amount of any one of the compounds disclosed herein.

[0076] In embodiments of the method of treatment, the condition treatable by the inhibition of one or more Raf kinases. In embodiments, the condition is selected from cancer, sarcoma, melanoma, skin cancer, haematological tumors, lymphoma, carcinoma or leukemia. In embodiments, the condition is selected from Barret's adenocarcinoma; biliary tract carcinomas; breast cancer; cervical cancer; cholangiocarcinoma; central nervous system tumors; primary CNS tumors; glioblastomas, astrocytomas; glioblastoma multiforme; ependymomas; seconday CNS tumors (metastases to the central nervous system of tumors originating outside of the central nervous system); brain tumors; brain metastases; colorectal cancer; large intestinal colon carcinoma; gastric cancer; carcinoma of the head and neck; squamous cell carcinoma of the head and neck; acute lymphoblastic leukemia; acute myelogenous leukemia (AML); myelodysplastic syndromes; chronic myelogenous leukemia; Hodgkin's lymphoma; non- Hodgkin's lymphoma; megakaryoblastic leukemia; multiple myeloma; erythroleukemia; hepatocellular carcinoma; lung cancer; small cell lung cancer; non-small cell lung cancer; ovarian cancer; endometrial cancer; pancreatic cancer; pituitary adenoma; prostate cancer; renal cancer; metastatic melanoma or thyroid cancers.

[0077] The present disclosure relates to a method of treating cancer, comprising administering an effective amount of any one of the compounds disclosed herein.

[0078] In embodiments of the method of treating cancer, the cancer comprises at least one mutation of the BRAF kinase. In embodiments, the cancer comprises a BRAF V600E mutation. [0079] In embodiments, the cancer is selected from melanomas, thyroid cancer, Barret's adenocarcinoma, biliary tract carcinomas, breast cancer, cervical cancer, cholangiocarcinoma, central nervous system tumors, glioblastomas, astrocytomas, ependymomas, colorectal cancer, large intestine colon cancer, gastric cancer, carcinoma of the head and neck, hematologic cancers, leukaemia, acute lymphoblastic leukaemia, myelodysplastic syndromes, chronic myelogenous leukaemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma, megakaryoblastic leukaemia, multiple myeloma, hepatocellular carcinoma, lung cancer, ovarian cancer, pancreatic cancer, pituitary adenoma, prostate cancer, renal cancer, sarcoma, uveal melanoma or skin cancer. In embodiments, the cancer is BRAF V600E melanoma, BRAF V600E colorectal cancer, BRAF V600E papillary thyroid cancers, BRAF V600E low grade serous ovarian cancers, BRAF V600E glioma, BRAF V600E hepatobiliary cancers, BRAF V600E hairy cell leukaemia, BRAF V600E non-small cell cancer, or BRAF V600E pilocytic astrocytoma. In embodiments, the cancer is colorectal cancer.

DETAILED DESCRIPTION

[0080] All publications, patents and patent applications, including any drawings and appendices therein are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent or patent application, drawing, or appendix was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

Definitions

[0081] While the following terms are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter.

[0082] Throughout the present specification, the terms “about” and/or “approximately” may be used in conjunction with numerical values and/or ranges. The term “about” is understood to mean those values near to a recited value. Furthermore, the phrases “less than about [a value]” or “greater than about [a value]” should be understood in view of the definition of the term “about” provided herein. The terms “about” and “approximately” may be used interchangeably. [0083] Throughout the present specification, numerical ranges are provided for certain quantities. It is to be understood that these ranges comprise all subranges therein. Thus, the range “from 50 to 80” includes all possible ranges therein (e.g., 51-79, 52-78, 53-77, 54-76, 55-75, 60-70, etc.). Furthermore, all values within a given range may be an endpoint for the range encompassed thereby (e.g., the range 50-80 includes the ranges with endpoints such as 55-80, 50-75, etc.).

[0084] The term “a” or “an” refers to one or more of that entity; for example, “a RAF inhibitor” refers to one or more RAF inhibitor or at least one RAF inhibitor. As such, the terms “a” (or “an”), “one or more” and “at least one” are used interchangeably herein. In addition, reference to “an inhibitor” by the indefinite article “a” or “an” does not exclude the possibility that more than one of the inhibitors is present, unless the context clearly requires that there is one and only one of the inhibitors.

[0085] As used herein, the verb “comprise” as is used in this description and in the claims and its conjugations are used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. The present invention may suitably “comprise”, “consist of’, or “consist essentially of’, the steps, elements, and/or reagents described in the claims.

[0086] It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as "solely", "only" and the like in connection with the recitation of claim elements, or the use of a "negative" limitation.

[0087] The term “pharmaceutically acceptable salts” includes both acid and base addition salts. Pharmaceutically acceptable salts include those obtained by reacting the active compound functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, camphorsulfonic acid, oxalic acid, maleic acid, succinic acid, citric acid, formic acid, hydrobromic acid, benzoic acid, tartaric acid, fumaric acid, salicylic acid, mandelic acid, carbonic acid, etc. Those skilled in the art will further recognize that acid addition salts may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods.

[0088] The term "treating" means one or more of relieving, alleviating, delaying, reducing, improving, or managing at least one symptom of a condition in a subject. The term "treating" may also mean one or more of arresting, delaying the onset (i.e., the period prior to clinical manifestation of the condition) or reducing the risk of developing or worsening a condition. [0089] The compounds of the invention, or their pharmaceutically acceptable salts contain at least one asymmetric center. The compounds of the invention with one asymmetric center give rise to enantiomers, where the absolute stereochemistry can be expressed as ( R )- and (5)-, or (+) and (-). When the compounds of the invention have more than two asymmetric centers, then the compounds can exist as diastereomers or other stereoisomeric forms. The present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms whether or not they are specifically depicted herein. Optically active (+) and (-) or ( R )- and (S)- isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.

[0090] A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposable mirror images of one another.

[0091] A “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present disclosure includes tautomers of any said compounds.

[0092] An "effective amount" means the amount of a formulation according to the invention that, when administered to a patient for treating a state, disorder or condition is sufficient to effect such treatment. The "effective amount" will vary depending on the active ingredient, the state, disorder, or condition to be treated and its severity, and the age, weight, physical condition and responsiveness of the mammal to be treated.

[0093] The term "therapeutically effective" applied to dose or amount refers to that quantity of a compound or pharmaceutical formulation that is sufficient to result in a desired clinical benefit after administration to a patient in need thereof. [0094] As used herein, a “subject” can be a human, non-human primate, mammal, rat, mouse, cow, horse, pig, sheep, goat, dog, cat and the like. The subject can be suspected of having or at risk for having a cancer, including but not limited to colorectal cancer and melanoma.

[0095] “Mammal” includes humans and both domestic animals such as laboratory animals (e.g., mice, rats, monkeys, dogs, etc.) and household pets ( e.g ., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.

[0096] All weight percentages (i.e., "% by weight" and "wt. %" and w/w) referenced herein, unless otherwise indicated, are measured relative to the total weight of the pharmaceutical composition.

[0097] As used herein, "substantially" or "substantial" refers to the complete or nearly complete extent or degree of an action, characteristic, property, state, structure, item, or result. For example, an object that is "substantially" enclosed would mean that the object is either completely enclosed or nearly completely enclosed. The exact allowable degree of deviation from absolute completeness may in some cases depend on the specific context. However, generally speaking, the nearness of completion will be so as to have the same overall result as if absolute and total completion were obtained. The use of "substantially" is equally applicable when used in a negative connotation to refer to the complete or near complete lack of action, characteristic, property, state, structure, item, or result. For example, a composition that is "substantially free of other active agents would either completely lack other active agents, or so nearly completely lack other active agents that the effect would be the same as if it completely lacked other active agents. In other words, a composition that is "substantially free of an ingredient or element or another active agent may still contain such an item as long as there is no measurable effect thereof.

[0098] The term “halo” refers to a halogen. In particular the term refers to fluorine, chlorine, bromine and iodine.

[0099] “Alkyl” or “alkyl group” refers to a fully saturated, straight or branched hydrocarbon chain group, and which is attached to the rest of the molecule by a single bond. Alkyls comprising any number of carbon atoms, including but not limited to from 1 to 12 are included. An alkyl comprising up to 12 carbon atoms is a C 1 -C 12 alkyl, an alkyl comprising up to 10 carbon atoms is a C 1 -C 10 alkyl, an alkyl comprising up to 6 carbon atoms is a C 1 -C 6 alkyl and an alkyl comprising up to 5 carbon atoms is a C 1 -C 5 alkyl. A C 1 -C 5 alkyl includes C 5 alkyls, C 4 alkyls, C 3 alkyls, C 2 alkyls and C 1 alkyl ( i.e ., methyl). A C 1 -C 6 alkyl includes all moieties described above for C 1 -C 5 alkyls but also includes C 6 alkyls. A C 1 -C 10 alkyl includes all moieties described above for C 1 -C 5 alkyls and C 1 -C 6 alkyls, but also includes C 7 , C 8 , C 9 and C 10 alkyls. Similarly, a C 1 -C 12 alkyl includes all the foregoing moieties, but also includes C 11 and C 12 alkyls. Non-limiting examples of C 1 -C 12 alkyl include methyl, ethyl, n-propyl, /-propyl, sec-propyl, n-butyl, i-butyl, sec-butyl, t-butyl, n-pentyl, t-amyl, n-hexyl, n-heptyl, n-octyl, n- Nonyl, n-decyl, n-undecyl, and n-dodecyl. Unless stated otherwise specifically in the specification, an alkyl group can be optionally substituted.

[0100] “Cycloalkyl” refers to a stable non-aromatic monocyclic or polycyclic fully saturated hydrocarbon group consisting solely of carbon and hydrogen atoms, which can include fused or bridged ring systems, having from three to twenty carbon atoms, preferably having from three to ten carbon atoms, and which is attached to the rest of the molecule by a single bond. Monocyclic cycloalkyl groups include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyl groups include, for example, adamantyl, norbornyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyl group can be optionally substituted.

[0101] “Haloalkyl” refers to an alkyl group, as defined above, that is substituted by one or more halo groups, as defined above, e.g ., trifluoromethyl, difluorom ethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group can be optionally substituted.

[0102] “Aryl” refers to a hydrocarbon ring system group comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl group can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems. Aryl groups include, but are not limited to, aryl groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term “aryl” is meant to include aryl groups that are optionally substituted. [0103] “Heterocyclyl,” “heterocyclic ring” or “heterocycle” refers to a stable 3- to 20-membered ring group which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Heterocyclycl or heterocyclic rings include heteroaryls as defined below. Unless stated otherwise specifically in the specification, the heterocyclyl group can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl group can be optionally oxidized; the nitrogen atom can be optionally quaternized; and the heterocyclyl group can be partially or fully saturated. Examples of such heterocyclyl groups include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocyclyl group can be optionally substituted. In embodiments, heterocyclyl, heterocyclic ring or heterocycle is a stable 3- to 20-membered non-aromatic ring group which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.

[0104] “N-heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the specification, a N-heterocyclyl group can be optionally substituted.

[0105] “Heteroaryl” refers to a 5- to 20-membered ring system group comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl group can be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which can include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl group can be optionally oxidized; the nitrogen atom can be optionally quaternized. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyridinyl, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl,

1 -phenyl- 1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group can be optionally substituted. [0106] “N-heteroaryl” refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an N-heteroaryl group can be optionally substituted. [0107] The term “substituted” used herein means any of the above groups (i.e., alkyl, alkylene, alkenyl, alkenylene, alkynyl, alkynylene, alkoxy, alkylamino, alkylcarbonyl, thioalkyl, aryl, aralkyl, carbocyclyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, “substituted” includes any of the above groups in which one or more hydrogen atoms are replaced with –NR g R h , –NR g C(=O)R h , –NR g C(=O)NR g R h , –NR g C(=O)OR h , –NR g SO 2 R h , -OC(=O)NR g R h , -OR g , -SR g , -SOR g , -SO 2 R g , -OSO 2 R g , -SO 2 OR g , =NSO 2 R g , and -SO 2 NR g R h . “Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=O)R g , -C(=O)OR g , -C(=O)NR g R h , –CH 2 SO 2 R g , – CH 2 SO 2 NR g R h . In the foregoing, R g and R h are the same or different and independently hydrogen, alkyl, alkenyl, alkynyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, haloalkenyl, haloalkynyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl. “Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkenyl, alkynyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, cycloalkylalkyl, haloalkyl, haloalkenyl, haloalkynyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing groups can also be optionally substituted with one or more of the above groups.

Compounds of the Invention

[0108] The present disclosure relates to pan-RAF inhibitors having the structure of formula (I), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0109] wherein ring A is a 5-membered heterocycle or heteroaryl containing 1, 2, or 3, nitrogen atom as a ring member;

[0110] wherein one of R 1 or R 2 is selected from substituted C 1-8 alkyl, unsubstituted C 5- 8 alkyl, substituted or unsubstituted C 1-8 haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl, and the other R 1 or R 2 is H;

[0111] or alternatively, R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted;

[0112] wherein when ring A is an imidazole, then the substituted aryl (e.g., R 1 or R 2 ) is

[0113] wherein when ring A is an imidazole, R 1 and R 2 together with the atoms to which they are attached do not form an unsubstituted phenyl ring.

[0114] In embodiments of formula (I), ring A is imidazole, pyrazole, or triazole. In

[0115] In embodiments, the present disclosure relates compounds of formula (I- A), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0116] wherein one of R 1 or R 2 is selected from substituted or unsubstituted C 5-8 alkyl, substituted or unsubstituted C 1-8 haloalkyl, substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl, and the other R 1 or R 2 is H;

[0117] or alternatively, R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted;

[0118] wherein the substituted aryl (e.g., R 1 or R 2 ) is not

[0119] wherein R 1 and R 2 together with the atoms to which they are attached do not form an unsubstituted phenyl.

[0120] In embodiments of formula (I) or (I-A), substituent is selected from halogen, -OR A , - NR A R B , -SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; wherein:

[0121] R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and

[0122] R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; and

[0123] wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -OR A , -CN, -SOR c , -NR A R B , or-NR D R E ; [0124] R D and R E together with the N atom to which they are attached forms a 5- or 6- membered saturated or partially unsaturated ring containing 1 or 2 heteroatoms selected from N, O, or S; wherein the saturated or partially unsaturated ring is optionally substituted with C 1-6 alkyl.

[0125] In embodiments, the present disclosure relates compounds of formula (I-B), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0126] wherein one of R 1 or R 2 is selected from substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl; and

[0127] wherein the substituted

[0128] In embodiments of formula (I), (I-A), or (I-B), substituent is selected from halogen, - OR A , -NR A R B , -SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; wherein R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -OR A , -CN, -SOR c , or -NR A R B

[0129] In embodiments of formula (I), (I-A), or (I-B), substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, - NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 . [0130] In embodiments of formula (I), (I-A), or (I-B), substituent on the substituted aryl or the substituted heteroaryl is selected from halogen, -OR A , -NR A R B , -SO 2 R C , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; wherein R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and R c is selected from C 1-6 alkyl and C 1-6 haloalkyl. In embodiments, substituent is selected from methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 .

[0131] In embodiments of formula (I), (I-A), or (I-B), substituent on the substituted aryl is selected from C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, -OR A , -NR A R B , -CN, -or -C(O)C 1-6 alkyl. [0132] In embodiments of formula (I) or (I-A), R 1 is substituted aryl, substituted or unsubstituted heterocyclyl, or substituted heteroaryl. In embodiments, substituted aryl is substituted phenyl. In embodiments, substituted heteroaryl is substituted N-heteroaryl. In embodiments, substituted heteroaryl is substituted 5- or 6-membered N-heteroaryl.

[0133] In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted phenyl, substituted pyridyl, substituted pyrazole, substituted pyrimidinyl, or substituted thiophenyl. In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted phenyl, substituted pyridyl, or substituted pyrazole.

[0134] In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted or unsubstituted heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R 1 is substituted or unsubstituted tetrahydropyran. In embodiments, R 1 is substituted or unsubstituted

[0135] In embodiments of formula (I-B), R 1 is substituted or unsubstituted heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R 1 is substituted or unsubstituted 5- or 6-membered heterocyclyl containing 0, 1, or 2 heteroatoms selected from

N, O, or S. In embodiments, R 1 is substituted or unsubstituted saturated heterocyclyl containing

O, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R 1 is substituted or unsubstituted saturated 5- or 6-membered heterocyclyl containing 0, 1, or 2 heteroatoms selected from N, O, or S. In embodiments, R 1 is substituted or unsubstituted tetrahydropyran. [0136] In embodiments of formula (I) or (I-A), R 1 is substituted or unsubstituted C 5-6 alkyl. In embodiments, C 5-6 alkyl is linear or branched.

[0137] In embodiments of formula (I), (I-A), or (I-B), R 1 is a monocyclic substituted aryl or a monocyclic substituted heteroaryl. In embodiments of formula (I), R 1 is a bicyclic substituted or unsubstitued aryl or a bicyclic substituted heteroaryl. In embodiments of formula (I), (I-A), or (I-B), R 1 is a bicyclic substituted aryl or a bicyclic substituted heteroaryl. In embodiments of formula (I), (I-A), or (I-B), R 1 is a fused bicyclic substituted aryl or a fused bicyclic substituted heteroaryl. In embodiments of formula (I), R 1 is substituted or unsubstituted indazole or substituted or unsubstituted benzoimidazole. In embodiments of formula (I), (I-A), or (I-B), R 1 is substituted indazole or substituted benzoimidazole.

[0138] In embodiments of formula (I) or (I-A), R 1 or R 2 is substituted with 1, 2, or 3 substituents. In embodiments of formula (I) or (I-A), R 1 or R 2 is substituted with 1 or 2 substituents. [0139] In embodiments of formula (I-B), R 1 is substituted with 1, 2, or 3 substituents. In embodiments of formula (I-B), R 1 is substituted with 1 or 2 substituents.

[0140] In embodiments of formula (I) or (I-A), R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered partially unsaturated or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted or unsubstituted. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a 6- membered partially unsaturated or unsaturated ring containing 0 or 1 nitrogen atom in the ring, wherein the ring is substituted or unsubstituted.

[0141] In embodiments of formula (I-A), R 1 and R 2 together with the atoms to which they are attached forms a phenyl ring, which is substituted or unsubstituted. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a phenyl ring thereby forming a benzoimidazole ring with the imidazole ring depicted in formula (I-A), which is substituted or unsubstituted.

[0142] In embodiments of formula (I-A), R 1 and R 2 together with the atoms to which they are attached forms a pyridyl ring, which is substituted or unsubstituted. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a pyridyl ring thereby forming a imidazopyridine ring with the imidazole ring depicted in formula (I-A), which is substituted or unsubstituted.

[0143] In embodiments of formula (I-A), R 1 and R 2 together with the atoms to which they are attached forms a tetrahydropyridyl ring, which is substituted or unsubstituted. In embodiments, R 1 and R 2 together with the atoms to which they are attached forms a tetrahydropyridyl ring thereby forming a tetrahydroimidazopyridine ring with the imidazole ring depicted in formula (I-A), which is substituted or unsubstituted.

[0144] In embodiments of formula (I) or (I-A), R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is unsubstituted. [0145] In embodiments of formula (I) or (I-A), R 1 and R 2 together with the atoms to which they are attached forms a 5- or 6-membered saturated, partially unsaturated, or unsaturated ring containing 0, 1, or 2 heteroatoms selected from N, O, or S, wherein the ring is substituted with 1, 2, 3, or 4 substituents. In embodiments, the ring is substituted with 1 or 2 substituents. [0146] In embodiments, any substituents listed in embodiments of formula (I), (I-A), or (I-B) can be applicable to embodiments of formula (II) or (III). [0147] In embodiments of formula (I), (I-A), or (I-B), the compound is a racemate. In embodiments, the compound is an (R) stereoisomer. In embodiments, the compound is an (S) stereoisomer.

[0148] In embodiments, the compound of formula (I), (I-A), or (I-B) is selected from Tables A-1, A-2, or A-3, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0149] Table A-1

[0150] Table A-2

[0151] Table A-3

[0152] In embodiments, the present disclosure relates compounds of formula (II), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0153] wherein: X 1 and X 2 are each N or CH;

[0154] R 1 is selected from substituted C 1-8 alkyl, unsubstituted C 5-8 alkyl, substituted or unsubstituted C 1-8 haloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl; and

[0155] R 4 is -NR F C(O)R 5 , -NR F C(O)CH 2 R 5 , -NR F C(O)CH(CH 3 )R 5 , or -NR F R 5 ;

[0156] R 5 is substituted or unsubstituted group selected from carbocyclyl, aryl, heterocyclyl, or heteroaryl; and

[0157] R F is selected from H or C 1-3 alkyl. [0158] In embodiments of the compounds of formula (II), one of X 1 and X 2 is N. In embodiments, X 1 is N and X 2 CH. In embodiments, X 2 is N and X 1 CH. In embodiments, X 1 and X 2 are both CH.

[0159] In embodiments of the compounds of formula (II), R 1 is substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl. In embodiments, R 1 is substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrazole, substituted or unsubstituted pyrimidinyl, or substituted or unsubstituted thiophenyl. In embodiments, R 1 is substituted or unsubstituted phenyl. In embodiments, R 1 is substituted phenyl.

[0160] In embodiments of the compounds of formula (II), R 4 is -NHC(O)R 5 , -NHC(O)CH 2 R 5 , -NHC(O)CH(CH 3 )R 5 , or -NHR 5 . In embodiments, R 4 is -NHC(O)R 5 , -NHC(O)CH 2 R 5 , or - NHR 5 .

[0161] In embodiments of the compounds of formula (II), R 5 is substituted or unsubstituted group selected from alkyl, 3-6 membered carbocyclyl, phenyl, 3-6 membered heterocyclyl, or 5-6 membered heteroaryl. In embodiments, R 5 is substituted or unsubstituted group selected from 3-6 membered carbocyclyl, phenyl, 3-6 membered heterocyclyl, or 5-6 membered heteroaryl. In embodiments, R 5 is substituted or unsubstituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In embodiments, R 5 is substituted or unsubstituted heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R 5 is substituted or unsubstituted 5- or 6-membered heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R 5 is substituted or unsubstituted saturated heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R 5 is substituted or unsubstituted saturated 5- or 6-membered heterocyclyl containing 1 or 2 heteroatoms selected from N, O, or S. In embodiments, R 5 is substituted or unsubstituted azetidine, pyrrolidine, piperidine, piperazine, or morpholine moiety. In embodiments, R 5 is substituted or unsubstituted group selected from methyl, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, azetidine, pyrrolidine, piperidine, piperazine, or morpholine, pyridine, thiazole, imidazole, pyrazole, or tri azole.

[0162] In embodiments of the compounds of formula (II), R 5 is substituted or unsubstituted 5- 6 membered heteroaryl. In embodiments, R 5 is substituted or unsubstituted 5-6 membered heteroaryl containing at least 1 nitrogen atom as a ring member. In embodiments, R 5 is substituted or unsubstituted pyridine, thiazole, imidazole, pyrazole, or triazole.

[0163] In embodiments of the compounds of formula (II), R F is H or methyl. In embodiments, R F is H. [0164] In embodiments of formula (II), substituent is selected from halogen, -OR A , -NR A R B , - SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, or -C(O)C 1-6 alkyl; wherein R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; wherein the alkyl, haloalkyl and cycloalkyl groups are optionally substituted with 1 to 3 groups independently selected from: -OR A , -CN, -SOR c , or

-NR A R B

[0165] In embodiments of formula (II), substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 .

[0166] In embodiments of formula (II), R 5 is substituted with a substituent selected from methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, - NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), or -CH 2 N(C 1-3 alkyl) 2 . In embodiments, R 5 is substituted with one or more substituent selected from halogen, methyl, ethyl, propyl, isopropyl, -CN, -OH, or -NH 2 .

[0167] In embodiments of formula (II), R 1 is substituted with 1, 2, or 3 substituents. In embodiments of formula (II), R 1 is substituted with 1 or 2 substituents. In embodiments of formula (II), R 1 is un substituted.

[0168] In embodiments of formula (II), R 5 is substituted with 1, 2, or 3 substituents. In embodiments of formula (II), R 5 is substituted with 1 or 2 substituents. In embodiments of formula (II), R 5 is un substituted.

[0169] In embodiments, any substituents listed in embodiments of formula (II) can be applicable to embodiments of formula (I), (I-A), (I-B) or (III).

[0170] In embodiments of formula (II), the compound is a racemate. In embodiments, the compound is an (R) stereoisomer. In embodiments, the compound is an (S) stereoisomer. [0171] In embodiments, the compound of formula (II) is selected from Table B, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0172] Table B

[0173] In embodiments, the present disclosure relates compounds of formula (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof,

[0174] wherein: R 6 is -C(O)NR F R 5 , -C(O)NR F CH 2 R 5 , substituted or unsubstituted aryl, substituted or unsubstituted heterocyclyl, or substituted or unsubstituted heteroaryl; [0175] R 5 is substituted or unsubstituted group selected from carbocyclyl, aryl, heterocyclyl, or heteroaryl; and [0176] R F is selected from H or C 1-3 alkyl.

[0177] In embodiments of the compounds of formula (III), R 6 is -C(O)NR F R 5 or - C(O)NR F CH 2 R 5 . In embodiments, R 6 is -C(O)NHR 5 or -C(O)NHCH 2 R 5 .

[0178] In embodiments of the compounds of formula (III), R 5 is substituted or unsubstituted aryl. In embodiments, R 5 is substituted or unsubstituted phenyl.

[0179] In embodiments of the compounds of formula (III), R 6 is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. In embodiments, a monocyclic substituted aryl or a monocyclic substituted heteroaryl. In embodiments, R 6 is a bicyclic substituted aryl or a monocyclic substituted heteroaryl. In embodiments, R 6 is a fused bicyclic substituted aryl or a fused bicyclic substituted heteroaryl. In embodiments, R 6 is substituted or unsubstituted indazole or substituted or unsubstituted benzoimidazole.

[0180] In embodiments of the compounds of formula (III), R 6 is substituted aryl or substituted heteroaryl, wherein the substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , - CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), -CH 2 N(C 1-3 alkyl) 2 , optionally substituted phenyl, optionally substituted heteroaryl. In embodiments of the compounds of formula (III), R 6 is substituted aryl or substituted heteroaryl, wherein the substituent is selected from optionally substituted phenyl or optionally substituted 5- or 6-membered heteroaryl. In embodiments, R 6 is substituted aryl or substituted heteroaryl, wherein the substituent is selected from phenyl or optionally substituted 5- or 6-membered N-heteroaryl. In embodiments, R 6 is substituted aryl or substituted heteroaryl, wherein the substituent is selected from phenyl or optionally substituted 5-membered N-heteroaryl. In embodiments, R 6 is substituted aryl or substituted heteroaryl, wherein the substituent is selected from phenyl or pyrazole substituted with one or two C 1-3 alkyl groups. [0181] In embodiments of formula (III), substituent is selected from halogen, -OR A , -NR A R B , -SO 2 R c , -SOR c , -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, -C(O)C 1-6 alkyl, aryl, or heteroaryl; wherein R A and R B are each independently selected from H, C 1-6 alkyl and C 1-6 haloalkyl; and R c is selected from C 1-6 alkyl and C 1-6 haloalkyl; wherein the alkyl, haloalkyl, cycloalkyl, aryl, and heteroaryl groups are optionally substituted with 1 to 3 groups independently selected from: C 1-6 alkyl, -OR A , -CN, -SOR c , or -NR A R B .

[0182] In embodiments of formula (II), substituent is selected from halogen, methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, t-butyl, cyclopropyl, methoxy, ethoxy, isopropoxy, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , -CH 2 CF 3 , -C(O)CH 3 , -CN, -OH, -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CH 2 NH 2 , -CH 2 NH(C 1-3 alkyl), -CH 2 N(C 1-3 alkyl) 2 , optionally substituted phenyl, optionally substituted heteroaryl.

[0183] In embodiments of formula (III), R 5 is substituted with 1, 2, or 3 substituents. In embodiments of formula (III), R 5 is substituted with 1 or 2 substituents. In embodiments of formula (III), R 5 is unsubstituted.

[0184] In embodiments, any substituents listed in embodiments of formula (III) can be applicable to embodiments of formula (I), (I-A), (I-B) or (II).

[0185] In embodiments, the compound of formula (III) is selected from Table C, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0186] Table C

[0187] In embodiments, the present disclosure relates a compound in Table D, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof.

[0188] Table D

[0189] In embodiments, the compounds of the present disclosure have pERK A375 (lhr) pIC 50 in the range of about 4 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK A375 (lhr) pIC 50 in the range of about 5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK A375 (lhr)p IC 50 value of about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about

6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about 6.7, about 6.8, about

6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about

7.8, about 7.9, or about 8.0, including any values therebetween. [0190] In embodiments, the compounds of the present disclosure have pERK HCT116 dimer (lhr) pIC 50 in the range of about 4 to about 9 including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 dimer (lhr) pIC 50 in the range of about 5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 dimer (lhr) pIC 50 of about 5.0, about 5.1, about 5.2, about 5.3, about 5.4, about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, about 6.0, about 6.1, about 6.2, about 6.3, about 6.4, about 6.5, about 6.6, about

6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2about 7.3, about 7.4, about 7.5, about

7.6, about 7.7, about 7.8, about 7.9, or about 8.0 including any values therebetween.

[0191] In embodiments, the compounds of the present disclosure have a pERK A375 monomer/ pERK HCT116 dimer ratio in the range of about 0.01 to about 2.5, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have a pERK A375 monomer/ pERK HCT116 dimer ratio in the range of about 0.03 to about 2.0, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have a pERK monomer/dimer ratio of about 0.1, about 0.2, about 0.3, about 0.4, about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1.0, about 1.1, about 1.2, about

1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2.0, including any values therebetween. In embodiments, the compounds of the present disclosure have a pERK A375 monomer/ pERK HCT116 dimer ratio of 2 or less. A pERK A375 monomer/ pERK HCT116 dimer ratio is calculated using the potency in nM.

[0192] In embodiments, the compounds of the present disclosure have pERK HCT116 (2hr) absolute pIC 50 in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 (2hr) absolute pIC 50 in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 (2hr) absolute pIC 50 of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pERK HCT116 (2hr) absolute pIC 50 of about 6.5 or greater.

[0193] In embodiments, the compounds of the present disclosure have pERK WiDr (2hr) absolute pIC 50 in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK WiDr (2hr) absolute pIC 50 in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pERK WiDr (2hr) absolute pIC 50 of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pERK WiDr (2hr) absolute pIC 50 of about 6.5 or greater.

[0194] In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 of about 6.5, about 6.6, about

6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D HCT116 of about 6.5 or greater.

[0195] In embodiments, the compounds of the present disclosure have pGI50 3D WiDr in the range of about 6 to about 9, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D WiDr in the range of about 6.5 to about 8, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have pGI503D WiDr of about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about

7.8, about 7.9, or about 8.0, including any values therebetween. In embodiments, the compounds of the present disclosure have pGI50 3D WiDr of about 6.5 or greater.

[0196] In embodiments, the compounds of the present disclosure have human liver microsome (HLM) intrinsic clearance (CLint) in the range of about 1 μL/min/mg to about 25μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint in the range of about 1 μL/min/mg to about 20 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint in the range of about 1 μL/min/mg to about 15 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint in the range of about 1 μL/min/mg to about 10 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have HLM CLint of about 1 μL/min/mg, about 2 μL/min/mg, about 3 μL/min/mg, about 4 μL/min/mg, about 5 μL/min/mg, about 6 μL/min/mg, about 7 μL/min/mg, about 8 μL/min/mg, about 9 μL/min/mg, about 10 μL/min/mg, about 11 μL/min/mg, about 12 μL/min/mg, about 13 μL/min/mg, about 14 μL/min/mg, or about 15 μL/min/mg, including any values therebetween. In embodiments, the compounds of the present disclosure have HLM CLint of less than about 15 μL/min/mg In embodiments, the compounds of the present disclosure have HLM CLint of less than about 20 μL/min/mg.

[0197] In embodiments, the compounds of the present disclosure have mouse liver microsome (MLM) intrinsic clearance (CLint) in the range of about 1 μL/min/mg to about 130 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint in the range of about 1 μL/min/mg to about 50 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint in the range of about 1 μL/min/mg to about 30 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint in the range of about 1 μL/min/mg to about 20 μL/min/mg, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have MLM CLint of about 1 μL/min/mg, about 2 μL/min/mg, about 3 μL/min/mg, about 4 μL/min/mg, about 5 μL/min/mg, about 6 μL/min/mg, about 7 μL/min/mg, about 8 μL/min/mg, about 9 μL/min/mg, about 10 μL/min/mg, about 11 μL/min/mg, about 12 μL/min/mg, about 13 μL/min/mg, about 14 μL/min/mg, about 15 μL/min/mg, about 16 μL/min/mg, about 17 μL/min/mg, about 18 μL/min/mg, about 19 μL/min/mg, or about 20 μL/min/mg, including any values therebetween. In embodiments, the compounds of the present disclosure have MLM CLint of less than about 30 μL/min/mg. In embodiments, the compounds of the present disclosure have MLM CLint of less than about 25 μL/min/mg. In embodiments, the compounds of the present disclosure have MLM CLint of less than about 20 μL/min/mg. [0198] In embodiments, the compounds of the present disclosure have human plasma protein binding (hPPB) percent free value in the range of about 0.01% to about 15%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have hPPB percent free value in the range of about 0.05% to about 10%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have hPPB percent free value in the range of about 0.1% to about 8%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have hPPB percent free value in the range of about 0.1% to about 5%, including any values and subranges therebetween.

[0199] In embodiments, the compounds of the present disclosure have mouse plasma protein binding (mPPB) percent free value in the range of about 0.01% to about 15%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mPPB percent free value in the range of about 0.05% to about 10%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mPPB percent free value in the range of about 0.1% to about 8%, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have mPPB percent free value in the range of about 0.1% to about 5%, including any values and subranges therebetween.

[0200] In embodiments, the compounds of the present disclosure have fed state simulated intestinal fluid (FESSIF) solubility in the range of about 10 mg/L to about 1000 mg/L, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have FESSIF solubility in the range of about 20 mg/L to about 750 mg/L, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have FESSIF solubility in the range of about 25 mg/L to about 600 mg/L, including any values and subranges therebetween. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 10 mg/L or greater. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 20 mg/L or greater. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 25 mg/L or greater. In embodiments, the compounds of the present disclosure have FESSIF solubility of about 30 mg/L or greater.

Therapeutic Use

[0201] The present disclosure also relates to method of using compounds of formula (I), (I- A), (I-B), (II), or (III), or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, for treating various diseases and conditions. In embodiments, compounds of formula (I), (I-A), (I-B), (II), or (III), or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition implicated by abnormal activity of one or more Raf kinase. In embodiments, compounds of formula (I), (I-A), (I-B), (II), or (III), or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition treatable by the inhibition of one or more Raf kinase. RAF kinase inhibition is relevant for the treatment of many different diseases associated with the abnormal activity of the MAPK pathway. In embodiments the condition treatable by the inhibition of RAF kinases, such as B-RAF or C-RAF.

[0202] The present disclosure also relates to method of using compounds of Tables A1-A3 and B-D, or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, for treating various diseases and conditions. In embodiments, compounds of Tables A1-A3 and B-D, or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition implicated by abnormal activity of one or more Raf kinase. In embodiments, compounds of Tables A1-A3 and B-D, or pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, are useful for treating a disease or a condition treatable by the inhibition of one or more Raf kinase. RAF kinase inhibition is relevant for the treatment of many different diseases associated with the abnormal activity of the MAPK pathway. In embodiments the condition treatable by the inhibition of RAF kinases, such as B-RAF or C- RAF.

[0203] In embodiments, the disease or the condition is cancer. In embodiments, the disease or the condition is selected from Barret's adenocarcinoma; biliary tract carcinomas; breast cancer; cervical cancer; cholangiocarcinoma; central nervous system tumors; primary CNS tumors; glioblastomas, astrocytomas; glioblastoma multiforme; ependymomas; seconday CNS tumors (metastases to the central nervous system of tumors originating outside of the central nervous system); brain tumors; brain metastases; colorectal cancer; large intestinal colon carcinoma; gastric cancer; carcinoma of the head and neck; squamous cell carcinoma of the head and neck; acute lymphoblastic leukemia; acute myelogenous leukemia (AML); myelodysplastic syndromes; chronic myelogenous leukemia; Hodgkin's lymphoma; non-Hodgkin's lymphoma; megakaryoblastic leukemia; multiple myeloma; erythroleukemia; hepatocellular carcinoma; lung cancer; small cell lung cancer; non-small cell lung cancer; ovarian cancer; endometrial cancer; pancreatic cancer; pituitary adenoma; prostate cancer; renal cancer; metastatic melanoma or thyroid cancers.

[0204] In embodiments, the disease or the condition is melanoma, non-small cell cancer, colorectal cancer, ovarian cancer, thyroid cancer, breast cancer or cholangiocarcinoma. In embodiments, the disease or the condition is colorectal cancer. In embodiments, the disease or the condition is melanoma.

[0205] In embodiments, the disease or the condition is cancer comprising a BRAF V600E mutation. In embodiments, the disease or the condition is modulated by BRAF V600E . In embodiments, the disease or the condition is BRAF V600E melanoma, BRAF V600E colorectal cancer, BRAF V600E papillary thyroid cancers, BRAF V600E low grade serous ovarian cancers, BRAF V600E glioma, BRAF V600E hepatobiliary cancers, BRAF V600E hairy cell leukaemia, BRAF V600E non-small cell cancer, or BRAF V600E pilocytic astrocytoma.

[0206] In embodiments, the disease or the condition is cardio-facio cutaneous syndrome and polycystic kidney disease.

Pharmaceutical Compositions [0207] The present disclosure also relates to pharmaceutical compositions comprising the compounds of formula (I), (I-A), (I-B), (II), or (III), or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient. The present disclosure also relates to pharmaceutical compositions comprising the compounds of Tables A1-A3 and B-D, or a pharmaceutically acceptable salt, tautomer, or stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient.

[0208] In embodiments, the pharmaceutical composition may further comprise an additional pharmaceutically active agent. The additional pharmaceutically active agent may be an anti tumor agent.

[0209] In embodiments, the additional pharmaceutically active agent is an antiproliferative/antineoplastic drug. In embodiments, antiproliferative/antineoplastic drug is alkylating agent (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, bendamustin, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolite (for example gemcitabine and antifolates such as fluoropyrimidines like 5- fluorouracil and tegafur, raltitrexed, methotrexate, pemetrexed, cytosine arabinoside, and hydroxyurea); antibiotic (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agent (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like TAXOL® (paclitaxel) and taxotere and polokinase inhibitors); proteasome inhibitor, for example carfilzomib and bortezomib; interferon therapy; or topoisomerase inhibitor (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan, mitoxantrone and camptothecin).

[0210] In embodiments, the additional pharmaceutically active agent is a cytostatic agent. In embodiments, cytostatic agent is antiestrogen (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogen (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonist or LHRH agonist (for example goserelin, leuprorelin and buserelin), progestogen (for example megestrol acetate), aromatase inhibitor (for example as anastrozole, letrozole, vorazole and exemestane) or inhibitor of 5a- reductase such as finasteride.

[0211] In embodiments, the additional pharmaceutically active agent is an anti-invasion agent. In embodiments, the anti-invasion agent is dasatinib and bosutinib (SKI-606), metalloproteinase inhibitor, or inhibitor of urokinase plasminogen activator receptor function or antibody to Heparanase. [0212] In embodiments, the additional pharmaceutically active agent is an inhibitor of growth factor function. In embodiments, the inhibitor of growth factor function is growth factor antibody and growth factor receptor antibody, for example the anti-erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR antibody panitumumab, the anti-erbB1 antibody cetuximab, tyrosine kinase inhibitor, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitor such as gefitinib, erlotinib and 6- acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3-morpholinopropox y)-quinazolin-4-amine (Cl 1033), erbB2 tyrosine kinase inhibitor such as lapatinib); inhibitor of the hepatocyte growth factor family; inhibitor of the insulin growth factor family; modulator of protein regulators of cell apoptosis (for example Bcl-2 inhibitors); inhibitor of the platelet-derived growth factor family such as imatinib and/or nilotinib (AMN107); inhibitor of serine/threonine kinases (for example Ras/RAF signalling inhibitors such as farnesyl transferase inhibitor, for example sorafenib, tipifarnib and lonafamib), inhibitor of cell signalling through MEK and/or AKT kinase, c-kit inhibitor, abl kinase inhibitor, PI3 kinase inhibitor, Plt3 kinase inhibitor, CSF-1R kinase inhibitor, IGF receptor, kinase inhibitor; aurora kinase inhibitor or cyclin dependent kinase inhibitor such as CDK2 and/or CDK4 inhibitor.

[0213] In embodiments, the additional pharmaceutically active agent is an anti angiogenic agent. In embodiments, the anti angiogenic agent inhibits the effects of vascular endothelial growth factor, for example the anti-vascular endothelial cell growth factor antibody bevacizumab (Avastin™); thalidomide; lenalidomide; and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib, vatalanib, sunitinib, axitinib and pazopanib. [0214] In embodiments, the additional pharmaceutically active agent is a cln embodiments, the cytotoxic agent is fludaribine (fludara), cladribine, or pentostatin (Nipent™).

[0215] In embodiments, the additional pharmaceutically active agent is a steroid. In embodiments, the steroid is corticosteroid, including glucocorticoid and mineralocorticoid, for example aclometasone, aclometasone dipropionate, aldosterone, amcinonide, beclomethasone, beclomethasone dipropionate, betamethasone, betamethasone dipropionate, betamethasone sodium phosphate, betamethasone valerate, budesonide, clobetasone, clobetasone butyrate, clobetasol propionate, cloprednol, cortisone, cortisone acetate, cortivazol, deoxycortone, desonide, desoximetasone, dexamethasone, dexamethasone sodium phosphate, dexamethasone isonicotinate, difluorocortolone, fluclorolone, flumethasone, flunisolide, fluocinolone, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluorocortisone, fluorocortolone, fluocortolone caproate, fluocortolone pivalate, fluorometholone, fluprednidene, fluprednidene acetate, flurandrenolone, fluticasone, fluticasone propionate, halcinonide, hydrocortisone, hydrocortisone acetate, hydrocortisone butyrate, hydrocortisone aceponate, hydrocortisone buteprate, hydrocortisone valerate, icomethasone, icomethasone enbutate, meprednisone, methylprednisolone, mometasone paramethasone, mometasone furoate monohydrate, prednicarbate, prednisolone, prednisone, tixocortol, tixocortol pivalate, triamcinolone, triamcinolone acetonide, triamcinolone alcohol and their respective pharmaceutically acceptable derivatives. A combination of steroids may be used, for example a combination of two or more steroids as described herein.

[0216] In embodiments, the additional pharmaceutically active agent is a targeted therapy agent. In embodiments, the targeted therapy agent is a PI3Kd inhibitor, for example idelalisib and perifosine.

[0217] In embodiments, the additional pharmaceutically active agent is an immunotherapeutic agent. In embodiments, the immunotherapeutic agent is antibody therapy agent such as alemtuzumab, rituximab, ibritumomab tiuxetan (Zevalin®) and ofatumumab; interferon such as interferon a; interleukins such as IL-2 (aldesleukin); interleukin inhibitors for example IRAK4 inhibitors; cancer vaccine including prophylactic and treatment vaccines such as HPV vaccines, for example Gardasil, Cervarix, Oncophage and Sipuleucel-T (Provenge); toll -like receptor modulator for example TLR-7 or TLR-9 agonist; and PD-1 antagonist, PDL-1 antagonist, and IDO- 1 antagonist.

[0218] In embodiments, the pharmaceutical composition may be used in combination with another therapy. In embodiments, the other therapy is gene therapy, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2. [0219] In embodiments, the other therapy is immunotherapy approaches, including for example antibody therapy such as alemtuzumab, rituximab, ibritumomab tiuxetan (Zevalin®) and ofatumumab; interferons such as interferon a; interleukins such as IL-2 (aldesleukin); interleukin inhibitors for example IRAK4 inhibitors; cancer vaccines including prophylactic and treatment vaccines such as HPV vaccines, for example Gardasil, Cervarix, Oncophage and Sipuleucel-T (Provenge); toll-like receptor modulators for example TLR-7 or TLR-9 agonists; and PD-1 antagonists, PDL-1 antagonists, and IDO-1 antagonists.

[0220] Compounds of the invention may exist in a single crystal form or in a mixture of crystal forms or they may be amorphous. Thus, compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, or spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose. [0221] For the above-mentioned compounds of the invention the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. For example, if the compound of the invention is administered orally, then the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight (μg/kg) to 100 milligrams per kilogram body weight (mg/kg).

[0222] A compound of the invention, or pharmaceutically acceptable salt thereof, may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the compounds of the invention, or pharmaceutically acceptable salt thereof, is in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, “Pharmaceuticals — The Science of Dosage Form Designs”, M. E. Aulton, Churchill Livingstone, 1988.

[0223] Depending on the mode of administration of the compounds of the invention, the pharmaceutical composition which is used to administer the compounds of the invention will preferably comprise from 0.05 to 99% w (percent by weight) compounds of the invention, more preferably from 0.05 to 80% w compounds of the invention, still more preferably from 0.10 to 70% w compounds of the invention, and even more preferably from 0.10 to 50% w compounds of the invention, all percentages by weight being based on total composition.

[0224] The pharmaceutical compositions may be administered topically (e.g. to the skin) in the form, e.g., of creams, gels, lotions, solutions, suspensions, or systemically, e.g. by oral administration in the form of tablets, capsules, syrups, powders or granules; or by parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); by rectal administration in the form of suppositories; or by inhalation in the form of an aerosol.

[0225] For oral administration the compounds of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide. Alternatively, the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent. [0226] For the preparation of soft gelatine capsules, the compounds of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules. Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain colouring agents, flavouring agents, sweetening agents (such as saccharine), preservative agents and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art.

[0227] For intravenous (parenteral) administration the compounds of the invention may be administered as a sterile aqueous or oily solution.

[0228] Pharmaceutical compositions can be prepared as liposome and encapsulation therapeutic agents. For various methods of preparing liposomes and encapsulation of therapeutic agents: see, for example, U.S. Pat. Nos. 3,932,657, 4,311,712, 4,743,449, 4,452,747, 4,830,858, 4,921,757, and 5,013,556. Known methods include the reverse phase evaporation method as described in U.S. Pat. No. 4,235,871. Also, U.S. 4,744,989 covers use of, and methods of preparing, liposomes for improving the efficiency or delivery of therapeutic compounds, drugs and other agents.

[0229] Compounds of the invention can be passively or actively loaded into liposomes. Active loading is typically done using a pH (ion) gradient or using encapsulated metal ions, for example, pH gradient loading may be carried out according to methods described in U.S. Pat. Nos. 5,616,341, 5,736,155, 5,785,987, and 5,939,096. Also, liposome loading using metal ions may be carried out according to methods described in U.S. Pat. Nos. 7,238,367, and 7,744,921. [0230] Inclusion of cholesterol in liposomal membranes has been shown to reduce release of drug and/or increase stability after intravenous administration (for example, see: U.S. Pat. Nos. 4,756,910, 5,077,056, and 5,225,212). Inclusion of low cholesterol liposomal membranes continuing charged lipids has been shown to provide cryostability as well as increase circulation after intravenous administration (see: U.S. Pat. No. 8,518,437).

[0231] Pharmaceutical compositions can comprise nanoparticles. The formation of nanoparticles has been achieved by various methods. Nanoparticles can be made by precipitating a molecule in a water-miscible solvent, and then drying and pulverizing the precipitate to form nanoparticles. (U.S. Pat. No. 4,726,955). Similar techniques for preparing nanoparticles for pharmaceutical preparations include wet grinding or milling. Other methods include mixing low concentrations of polymers dissolved in a water-miscible solution with an aqueous phase to alter the local charge of the solvent and form a precipitate through conventional mixing techniques. (U.S. Pat. No. 5,766,635). Other methods include the mixing of copolymers in organic solution with an aqueous phase containing a colloid protective agent or a surfactant for reducing surface tension. Other methods of incorporating additive therapeutic agents into nanoparticles for drug delivery require that nanoparticles be treated with a liposome or surfactant before drug administration (U.S. Pat. No. 6,117,454). Nanoparticles can also be made by flash nanoprecipitation (U.S. Pat. No. 8,137,699).

[0232] U.S. Pat. No. 7,850,990 covers methods of screening combinations of agents and encapsulating the combinations in delivery vehicles such as liposomes or nanoparticles.

[0233] The size of the dose for therapeutic purposes of compounds of the invention will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well-known principles of medicine.

[0234] Dosage levels, dose frequency, and treatment durations of compounds of the invention are expected to differ depending on the formulation and clinical indication, age, and co-morbid medical conditions of the patient. The standard duration of treatment with compounds of the invention is expected to vary between one and seven days for most clinical indications. It may be necessary to extend the duration of treatment beyond seven days in instances of recurrent infections or infections associated with tissues or implanted materials to which there is poor blood supply including bones/joints, respiratory tract, endocardium, and dental tissues.

EXAMPLES

[0235] The disclosure now being generally described, it will be more readily understood by reference to the following examples which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.

[0236] As used herein the following terms have the meanings given: “Boc” refers to tert- butyloxycarbonyl; “Cbz” refers to carboxybenzyl; “dba” refers to dibenzylideneacetone; “DCM” refers to dichloromethane; “DIPEA” refers to N,N-diisopropylethylamine; “DMA” refers to dimethylacetamide; “DMF” refers to N,N-dimethylformamide; “DMSO” refers to dimethyl sulfoxide; “dppf’ refers to 1,1’ - bis(diphenylphosphino)ferrocene; “EtOAc” refers to ethyl acetate; “EtOH” refers to ethanol; “Et20” refers to diethyl ether; “IP A” refers to isopropyl alcohol; “LiHMDS” refers to lithium bis(trimethylsilyl)amide; “mCPBA” refers to meta-chloroperoxybenzoic acid; “MeCN” refers to acetonitrile; “MeOH” refers to methanol; “min” refers to minutes; “NMR” refers to nuclear magnetic resonance; “PhMe” refers to toluene; “pTsOH” refers to p-toluenesulfonic acid; “py” refers to pyridine; “r.t ” refers to room temperature; “SCX” refers to strong cation exchange; “T3P” refers to propylphosphonic anhydride; “Tf20” refers to trifluoromethanesulfonic anhydride; “THF” refers to tetrahydrofuran; “THP” refers to 2-tetrahydropyranyl; “(UP)LC-MS” refers to (ultra performance) liquid chromatography/mass spectrometry. Solvents, reagents and starting materials were purchased from commercial vendors and used as received unless otherwise described. All reactions were performed at room temperature unless otherwise stated. Compound identity and purity confirmations were performed by LC-MS UV using a Waters Acquity SQ Detector 2 (ACQ-SQD2#LCA081). The diode array detector wavelength was 254nM and the MS was in positive and negative electrospray mode (m/z: 150-800). A 2μL aliquot was injected onto a guard column (0.2μm x 2 mm filters) and UPLC column (Cl 8, 50 x 2.1 mm, < 2μm) in sequence maintained at 40 °C. The samples were eluted at a flow rate of 0.6 mL/min with a mobile phase system composed of A (0.1% (v/v) formic acid in water) and B (0.1% (v/v) formic acid in MeCN) according to the gradients outlined below. Retention times RT are reported in minutes. [0237] NMR was also used to characterise final compounds. NMR spectra were obtained on a Bruker AVIII 400 Nanobay with 5mm BBFO probe. Optionally, compound Rf values on silica thin layer chromatography (TLC) plates were measured.

[0238] Compound purification was performed by flash column chromatography on silica or by preparative LC-MS. LC-MS purification was performed using a Waters 3100 Mass detector in positive and negative electrospray mode ( m/z: 150-800) with a Waters 2489 UV/Vis detector. Samples were eluted at a flow rate of 20 mL/min on a Xbridge™ prep C 18 5 μM OBD 19x100mm column with a mobile phase system composed of A (0.1% (v/v) formic acid in water) and B (0.1% (v/v) formic acid in MeCN) according to the gradient outlined below:

[0239] Chemical names in this document were generated using mol2nam - Structure to Name Conversion by OpenEye Scientific Software. Starting materials were purchased from commercial sources or synthesised according to literature procedures.

[0240] Synthesis of Common Intermediates

[0241] Example 1. Synthesis of 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one [0242] Step 1 - tert-butyl N-(4-fluoro-2-pyridyl)carbamate [0243] 2-Amino-4-fluoropyridine (400g, 3.57mol) was charged into a 10L fixed reactor vessel and then taken up in DCM (4L) under a nitrogen atmosphere. DMAP (43.6g, 357 mmol) was added and the reaction cooled to 10°C. Boc 2 O (934g, 4.2mol) was added as a solution in DCM (1L) over 1.5 hours. The reaction was stirred at rt for 2 hours. The reaction was then treated with N,N-dimethylethylenediamine (390mL, 3.57mmol) and heated to 40°C overnight. The reaction was cooled to rt, diluted with DCM (2L) and washed with water. The aqueous layer was extracted with further DCM (2L) and the organics washed with water, brine and dried over MgSO 4 . During evaporation of the solvent, a solid crashed out, which was filtered, washed with petroleum ether to give tert-butyl N-(4-fluoro-2-pyridyl)carbamate (505g, 2.38mol, 67% yield) as a cream solid product. UPLC-MS (ES+, Short acidic): 1.64 min, m/z 213.1 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 10.13 (1H, d, J = 1.7Hz), 8.26 (1H, dd, J = 9.4Hz, 5.7Hz), 7.60 (1H, dd, J = 12.3Hz, 2.4Hz), 6.94 (1H, ddd, J = 8.2Hz, 5.7Hz, 2.4Hz), 1.47 (9H, s). [0244] Step 2 – tert-butyl N-(4-fluoro-3-iodo-2-pyridyl)carbamate [0245] Tert-butyl N-(4-fluoro-2-pyridyl)carbamate (126g, 593.7mmol) and N,N,N,N- tetramethylethylenediamine (223mL, 1.48mol) were taken up in dry THF (1.7L) and then cooled to -78°C under a nitrogen atmosphere. To this solution was added n-BuLi (2.5M in hexane - 285mL, 712.5mmol) and then allowed to stir for a further 10 minutes. sec-BuLi (1.2M in cyclohexane - 509mL, 712.5mmol) was then added keeping the reaction temperature below -70°C and stirred for 1 hour. After this time, iodine (226g, 890.6mmol) in THF (300mL) was added dropwise over 30 minutes and the temperature kept below -65°C. The reaction was stirred at -70°C for another 10 minutes then quenched by the addition of sat. aq. NH 4 Cl (400mL) and then a solution of sodium thiosulphate (134.1g, 848.2mmol) dissolved in water (600mL) which raised the temperature to -25°C. The reaction was warmed to rt, transferred to a 5L separator and extracted with EtOAc (2x1.5L) and then washed with brine (500ml). The organic phase was dried over MgSO 4 and the solvent removed in vacuo. The residue was taken up in DCM (500mL) and passed through a 2Kg silica pad, which was washed with DCM (10x1L) and then the product was eluted from the column using as eluent a gradient 10-100% EtOAc in petroleum ether (1L at each 10% increase) tert-butyl N-(4-fluoro-3-iodo-2- pyridyl)carbamate (154.6g, 457.1mmol, 77% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.60 min, m/z 339.1 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 9.47 (1H, s), 8.33 (1H, dd, J = 8.7Hz, 5.5Hz), 7.19 (1H, dd, J = 7.3Hz, 5.5Hz), 1.46 (9H, s). [0246] Step 3 – 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one [0247] tert-butyl N-(4-fluoro-3-iodo-2-pyridyl)carbamate (263.3g, 778.72mmol) and 3,3- dimethoxyprop-1-ene (120mL, 1.01mol) and DIPEA (285mL, 1.64mol) were taken up in DMF (2.2L) and water (440mL) and degassed under a nitrogen atmosphere for 20 minutes. To this mixture, palladium (II) acetate (17.5g, 77.9mmol) was added, the reaction degassed for 15 minutes and then heated to 110°C for 18 hours. The reaction was cooled to rt and filtered through celite. The solvent was removed under reduce pressure, the residue was taken up in water and acidified to pH~1-2 with 2N HCl solution. It was then basified to pH~9 with solid NaHCO 3 solution followed by extraction with DCM (2x2L). The organic phases were combined, washed with water, brine and dried over MgSO 4 . EtOAc (2L) was added to the solution and the organics were passed through a silica plug eluting with 40% EtOAc in DCM. Fractions containing the product were combined and the solvent removed in vacuo to give a solid, which was slurried in cold Et 2 O (300mL) and filtered. The solid was washed with Et 2 O and then petroleum ether, pulled dry to give 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (5.7mg, 343.1mmol, 44% yield) as a pale yellow solid. UPLC-MS (ES+, Short acidic): 1.04 min, m/z 167.0 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 10.69 (1H, s), 8.29-7.90 (1H, m), 6.92 (1H, dd, J = 8.8Hz, 5.7Hz, 1H), 2.88 (2H, dd, J = 8.3Hz, 7.1Hz), 2.57-2.47 (2H, m). [0248] Example 2. Synthesis of 6-hydroxychromane-3-carboxylic acid [0249] Step 1 – 2-hydroxy-5-tetrahydropyran-2-yloxy-benzaldehyde [0250] To a solution of 2,5-dihydroxybenzaldehyde (400g, 2.90mol) and pyridinium p- toluenesulfonate (36.4g, 144.8mmol) in DCM (7.5L) was added 3,4-dihydro-2H-pyran (396.3mL, 4.34mol) dropwise over 10 minutes and the reaction stirred at 30°C overnight. The reaction was washed with water (1.5L), the organic layer separated and passed through a 1.5Kg silica pad, which was washed with DCM (2.5L), 25% EtOAc in petroleum ether (2.5L) and finally 50% EtOAc in petroleum ether (2.5L). Fractions containing the product were combined and the solvent removed in vacuo. The residue was slowly diluted with petroleum ether (1.75L) and cooled to 10°C to give a thick slurry. The product was filtered, washed with petroleum ether (2x150mL) and dried to give 2-hydroxy-5-tetrahydropyran-2-yloxy-benzaldehyde (570.7g, 2.57mol, 89% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 1.64 min, m/z 223.0 [M+H]+.1H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 10.35 (1H, s), 10.23 (1H, s), 7.32-7.19 (2H, m), 6.94 (1H, d, J = 8.9Hz), 5.36 (1H, t, J = 3.3Hz), 3.77 (1H, ddd, J = 11.2Hz, 8.8Hz, 3.6Hz), 3.59-3.49 (1H, m), 1.94-1.45 (6H, m). [0251] Step 2 – tert-butyl 6-tetrahydropyran-2-yloxy-2H-chromene-3-carboxylate [0252] 2-hydroxy-5-tetrahydropyran-2-yloxy-benzaldehyde (107g, 481.5mmol) was dissolved in diglyme (750mL) and K 2 CO 3 (133g, 962.9mmol) was added. The reaction was then heated to 140°C and tert-butyl acrylate (155mL, 1059.2mmol) in DMF (75mL) was added over 10 minutes at ~110°C; the reaction was stirred at 140°C for a further 5 hour. The reaction was cooled to rt overnight, filtered and the solvent removed in vacuo. The reaction was suspended in EtOAc (2.5L) and water (2.5L) and the phases separated. The aqueous phase was extracted with EtOAc (2.5L) and the combined organic layers were washed with brine (50%) and the solvent removed in vacuo. The crude was dissolved in DCM, loaded onto a 2Kg silica pad, which was flushed with a gradient from 10-25% EtOAc in petroleum ether to give tert-butyl 6- tetrahydropyran-2-yloxy-2H-chromene-3-carboxylate (122.5g, 368.5mmol, 77% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 2.18 min, m/z nd. 1 H NMR (400 MHz, DMSO- d 6 ) δ/ppm: 7.38 (1H, s), 7.05 (1H, d, J = 2.9 Hz), 6.94 (1H, dd, J = 8.8, 2.9 Hz), 6.79 (1H, dd, J = 8.8, 0.7 Hz), 5.35 (1H, t, J = 3.3 Hz), 4.82 (2H, d, J = 1.4 Hz), 3.78 (1H, ddd, J = 11.8, 8.6, 3.6 Hz), 3.58-3.49 (1H, m), 1.92-1.66 (3H, m), 1.66-1.52 (3H, m), 1.49 (s, 9H). [0253] Step 3 – tert-butyl 6-hydroxy-2H-chromene-3-carboxylate [0254] tert-Butyl 6-tetrahydropyran-2-yloxy-2H-chromene-3-carboxylate (110.4g, 332.1mmol) was suspended in MeOH (1.2L) at rt and pyridinium p-toluenesulfonate (8.35g, 33.2mmol) was added. The reaction was warmed to 34°C for 3 hours. The solvent was removed in vacuo, the crude dissolved in EtOAc (1L) and washed with water (750mL). The organic layer was dried over MgSO4, filtered and the solvent removed under reduce pressure to give a yellow solid. This solid was suspended in petroleum ether and the solvent removed in vacuo to give tert-butyl 6-hydroxy-2H-chromene-3-carboxylate (82g, 330.3mmol, 99% yield) as a yellow solid. UPLC-MS (ES-, Short acidic): 1.71 min, m/z 247.2 [M-H]-.1H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 9.17 (1H, s), 7.33 (1H, s), 6.76-6.64 (3H, m), 4.77 (2H, d, J = 1.4Hz), 1.49 (9H, s). [0255] Step 4 – tert-butyl 6-hydroxychromane-3-carboxylate [0256] tert-Butyl 6-hydroxy-2H-chromene-3-carboxylate (177.3g, 714.1mmol) was suspended in MeOH (2.5L) at rt and palladium, 10 wt. % on carbon powder, 50% wet (15.2g, 142.8mmol) added. The reaction was fitted with a H 2 balloon, extra H 2 added and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 5 hours. The crude was filtered over celite, washed with MeOH and the filtrate concentrated in vacuo to give tert-butyl 6-hydroxychromane-3-carboxylate (171g, 683.2mmol, 96% yield) as a pale cream solid. UPLC-MS (ES+, Short acidic): 1.64 min, m/z nd. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 8.81 (1H, s), 6.60-6.52 (1H, m), 6.47 (2H, d, J = 7.7Hz), 4.19 (1H, dd, J = 10.6Hz, 3.0Hz), 3.96 (1H, dd, J = 10.6Hz, 7.4Hz), 2.96-2.73 (3H, m), 1.40 (9H, s). [0257] Step 5 – 6-hydroxychromane-3-carboxylic acid [0258] tert-Butyl 6-hydroxychromane-3-carboxylate (23g, 91.9mmol) was dissolved in DCM (250mL) and TFA (48.3mL, 630.35mmol) added and the reaction stirred at rt overnight. Solvents were removed in vacuo and azeotroped with toluene (100mL); the residue was slurried with Et 2 O and filtered to give 6-hydroxychromane-3-carboxylic acid (16.8g, 86.5mmol, 94% yield) as a pink solid. UPLC-MS (ES+, Short acidic): 1.08 min, m/z 194.1 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 12.60 (1H, s), 8.81 (1H, s), 6.60-6.53 (1H, m), 6.52-6.45 (2H, m), 4.22 (1H, dd, J = 10.7Hz, 3.0Hz), 4.06-3.96 (1H, m), 2.97-2.77 (3H, m). [0259] Example 3. Synthesis of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4- yl)oxy]chromane-3-carboxylic acid [0260] 6-hydroxychromane-3-carboxylic acid (45.3g, 233.3mmol), 5-fluoro-3,4-dihydro-1H- 1,8-naphthyridin-2-one (38.8g, 233.3mmol) were suspended in DMSO (230mL) and K 2 CO 3 (119.7g, 865.8mmol) was added in portions. The reaction was heated to 98°C for 48h. It was then cooled to 60°C, diluted with water (2L) and extracted with EtOAc (750mL). The aqueous layer was separated and slowly added to a solution of citric acid (179.3g, 933.2mmol) in water (400mL) to give a cream solid, which was filtered, washed with water/acetone (1:1) and Et 2 O and dried to give 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3- carboxylic acid (74.7g, 219.5mmol, 94% yield) as a cream solid. UPLC-MS (ES+, Short acidic): 1.26 min, m/z 341.2 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 12.69 (1H, s), 10.46 (1H, s), 7.95 (1H, d, J = 5.8Hz), 6.97 (1H, d, J = 2.8Hz), 6.89 (1H, dd, J = 8.8Hz, 2.8Hz), 6.84 (1H, t, J = 8.8Hz), 6.24 (1H, d, J = 5.8Hz), 4.33 (1H, dd, J = 10.9Hz, 3.1Hz), 4.20-4.10 (1H, m), 3.06-2.95 (3H, m), 2.92 (2H, t, J = 8.4Hz, 7.0Hz), 2.59-2.51 (2H, m). [0261] Example 4. Synthesis of 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2- yl]chroman-6-ol and 5-[3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl] chroman- 6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [0262] Step 1 – methyl 6-hydroxy-2H-chromene-3-carboxylate [0263] To 6-hydroxy-2H-chromene-3-carboxylic acid (16g, 83.3mmol) in MeOH (200mL) was added sulfuric acid (0.44mL, 8.33mmol) and the reaction heated to 70°C until completion. The solvent was removed in vacuo and the residue dissolved in DCM and washed with water. The organic layer was separated and passed through a 100g silica pad eluting with DCM (500mL) and then with 50% EtOAc in petroleum ether (2x500mL). The solid obtained was slurried with petroleum ether, filtering and dried to give methyl 6-hydroxy-2H-chromene-3- carboxylate (14.1g, 68.4mmol, 82% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 1.38 min, m/z 207.1 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 9.19 (1H, s), 7.46 (1H, d, J = 1.4Hz), 6.79-6.65 (2H, m), 6.70 (1H, s), 4.82 (2H, d, J = 1.4Hz), 3.75 (3H, s). [0264] Step 2 – methyl 6-hydroxychromane-3-carboxylate [0265] methyl 6-hydroxy-2H-chromene-3-carboxylate (53.3g, 258.3mmol) was suspended in MeOH (600mL) at rt and palladium, 10 wt. % on carbon powder, 50% wet (2.75g, 25.8mmol) was added under a nitrogen atmosphere. The reaction was fitted with a H 2 balloon, extra H 2 added and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 3 hours. The crude was filtered over celite, washed with MeOH and the solvent removed in vacuo to give methyl 6-hydroxychromane-3-carboxylate (50.1g, 233.9mmol, 91% yield) as a cream solid. UPLC-MS (ES+, Short acidic): 1.27 min, m/z 209.1 [M+H]+. 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 8.83 (1H, s), 6.56 (1H, dd, J = 8.1, 1.0 Hz), 6.48 (2H, dd, J = 9.7Hz, 1.8 Hz), 4.22 (1H, dd, J = 10.8Hz, 3.2Hz), 4.04 (1H, dd, J = 10.7Hz, 7.6 Hz), 3.64 (3H, s), 3.10-2.98 (1H, m), 2.89 (2H, d, J = 6.9 Hz). [0266] Step 3 – methyl 6-benzyloxychromane-3-carboxylate [0267] Benzyl bromide (2.85mL, 23.97mmol) was slowly added to a stirred mixture of methyl 6-hydroxychromane-3-carboxylate (4.16g, 19.98mmol), K 2 CO 3 (8.28g, 59.93mmol) and DMF (100mL) at rt under a nitrogen atmosphere. The reaction was heated to 50°C and stirred for 1.5 hour, after which time it was cooled to rt and the solvent removed in vacuo. The residue was partitioned between water (300mL) and DCM (300mL). The organic layer was separated and the aqueous extracted with DCM (300mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-50% EtOAc in petroleum ether to give methyl 6-benzyloxychromane-3-carboxylate (5.9g, 19.78mmol, 99% yield) as a colourless oil which solidified to a white solid on standing. UPLC-MS (ES+, short acidic): 1.92 min, m/z 299.2 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.46-7.32 (5H, m), 6.79-6.77 (2H, m), 6.73 (1H, d, J = 7.2Hz), 5.02 (2H, s), 4.44-4.39 (1H, m), 4,14-4.08 (1H, m), 3.75 (3H, s), 3.14-2.97 (3H, m). [0268] Step 4 – 6-benzyloxychromane-3-carboxylic acid [0269] Methyl 6-benzyloxychromane-3-carboxylate (5.9g, 19.78mmol) was stirred in a solution of aq. NaOH (100mL, 100mmol) and THF (100mL) at rt for 18 hours. The organic solvent was removed in vacuo and the resulting mixture stirred, cooled in ice, followed by acidification to ~pH 5 using conc. HCl. The resulting solid was filtered off and dried to give 6-benzyloxychromane-3-carboxylic acid (5.1g, 17.94mmol, 91% yield) as a white solid. UPLC-MS (ES-, short acidic): 1.71 min, m/z 283.2 [M-H]-. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 12.64 (1H, br s), 7.46-7.37 (4H, m), 7.35-7.30 (1H, m), 6.80 (1H, d, J = 2.8Hz), 6.74 (1H, dd, J = 9.2Hz, 2.8Hz), 6.67 (1H, d, J = 9.2Hz), 5.02 (2H, s), 4.27-4.22 (1H, m), 4.09-4.04 (1H, m), 2.99-2.91 (3H, m). [0270] Step 5 – 6-benzyloxychroman-3-yl)methanol [0271] Di-methylsulfide borane (13.45mL, 26.91mmol) was added to a stirred solution of 6- benzyloxychromane-3-carboxylic acid (5.1g, 17.94mmol) and THF (200mL) at 0°C under a nitrogen atmosphere and stirred at rt for 3 hours. The reaction was cooled to 0°C and quenched carefully with water (500mL). This was extracted with EtOAc (2x500mL). the combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-10% EtOAc in petroleum ether to give 6-benzyloxychroman-3-yl)methanol (4.5g, 16.65mmol, 93% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.69 min, m/z 271.2 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.46-7.31 (5H, m), 6.78-6.75 (2H, m), 6.72-6.69 (1H, m), 5.01 (2H, s), 4.27 (1H, ddd, J = 10.8Hz, 2.8Hz, 1.2Hz), 3.99 (1H, dd, J = 10.8Hz, 2.8Hz), 3.77-3.65 (2H, m), 2.92-2.85 (1H, m), 2.59 (1H, dd, J = 16.8Hz, 7.6Hz), 2.33-2.23 (1H, m). Exchangeable proton not seen. [0272] Step 6 – 6-benzyloxychromane-3-carbaldehyde [0273] A solution of DMSO (1.77mL, 24.97mmol) in DCM (200mL) was stirred under a nitrogen atmosphere at -78°C. Oxalyl chloride (2.11mL, 24.97mmol) was then added and left stirring for 20 min; a solution of (6-benzyloxychroman-3-yl)methanol in DCM (50mL) was then added dropwise whilst maintaining the temperature below -70°C. Upon completion of addition, the reaction was stirred for 30 minutes and triethylamine (5.8mL, 41.62mmol) was added dropwise. The reaction was left stirring at -78°C for 20 min after which time it was allowed to warm to rt. The reaction was diluted with water (200mL) and the organic layer separated, passed through a phase separator and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-50% EtOAc in petroleum ether to give 6-benzyloxychromane-3-carbaldehyde (4.4g, 16.4mmol, 99% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.82 min, m/z 268.2 [M]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 9.86 (1H, s), 7.47-7.32 (5H, m), 6.81-6.74 (3H, m), 5.02 (2H, s), 4.42-4.32 (2H, m), 3.17-3.09 (1H, m), 3.04-2.93 (2H, m). [0274] Step 7 – 2-(6-benzyloxychroman-3-yl)-1H-imidazole [0275] Ammonium Hydroxide (25mL, 750mmol) was added to a stirred solution of 6- benzyloxychromane-3-carbaldehyde (3.9g, 14.54mmol), glyoxal (9.96mL, 87.21mmol) and MeOH (25mL) at rt. After 1 hour, the reaction was diluted with water (200mL) and extracted with DCM (2x200mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo to give 2-(6-benzyloxychroman-3-yl)-1H-imidazole (2.4g, 7.83mmol, 54% yield) as a yellow solid. UPLC-MS (ES+, short acidic), 1.28 min, m/z 307.3 [M+H]+. 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 7.46-7.30 (6H, m), 7.08 (1H, d, J = 9.2Hz), 6.83-6.80 (1H, d), 6.79-6.75 (1H, d), 6.71 (1H, d, J = 8.8Hz), 5.03 (2H, s), 4.40-4.35 (1H, m), 3.97 (1H, t, J = 10.0Hz), 3.31-3.26 (1H, m), 3.15-3.10 (1H, m), 3.05-2.97 (1H, m). Exchangeable proton not seen. [0276] Step 8 – 2-[[2-(6-benzyloxychroman-3-yl)imidazol-1-yl]methoxy]ethyl-t rimethyl- silane [0277] Sodium hydride (60% dispersed in mineral oil - 895.6mg, 22.39mmol) was added to a stirred solution of 2-(6-benzyloxychroman-3-yl)-1H-imidazole (3.43g, 11.2mmol) in DMF (5mL) at rt under a nitrogen atmosphere. After 20 minutes, 2-(trimethylsilyl)ethoxymethyl chloride (2.97mL, 16.79mmol) was added. The reaction was allowed to stir for 1 hour, then quenched with water (1mL) and solvent removed in vacuo. The residue was partitioned between water (20mL) and DCM (20mL). The organic layer was separated and the aqueous extracted with DCM (20mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-5% MeOH in DCM to give 2-[[2-(6-benzyloxychroman-3-yl)imidazol- 1-yl]methoxy]ethyl-trimethyl-silane (4.3g, 9.85mmol, 88% yield) as a yellow solid. UPLC- MS (ES+, short acidic): 1.67 min, m/z 437.7 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.45-7.29 (5H, m), 7.03-7.01 (1H, m), 6.99-6.97 (1H, m), 6.80-6.78 (2H, m), 6.73-6.71 (1H, m), 5.31-5.28 (2H, m), 5.01 (2H, s), 4.44-4.38 (1H, m), 4.12 (1H, t, J = 10.4Hz), 3.55-3.50 (2H, m), 3.48-3.33 (2H, m), 2.99-2.92 (1H, m), 0.96-0.87 (2H, m), 0.00 (9H, s). [0278] Step 9 – 3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-o l [0279] Palladium, 10 wt. % on carbon powder, dry (400mgl) was added to a stirred solution of 2-[[2-(6-benzyloxychroman-3-yl)imidazol-1-yl]methoxy]ethyl-t rimethyl-silane (4.3g, 9.85mmol) in MeOH (50mL) at rt. The reaction was fitted with a H2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 2 hours. The crude was filtered over celite, washed with MeOH and the filtrate concentrated in vacuo to give 3-[1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (3.32g, 9.58mmol, 97% yield) as a yellow solid. The compound was used directly in the next step without further purification. UPLC-MS (ES+, short acidic): 1.36 min, m/z 347.7 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 6.98 (1H, d, J = 1.6Hz), 6.99 (1H, d, J = 1.6Hz), 6.69 (1H, d, J = 8.8Hz), 6.63 (1H, dd, J = 8.8Hz, 3.2Hz), 6.56 (1H, d, J = 3.2Hz), 5.32 (1H, d, J = 10.8Hz), 5.28 (1H, d, J = 10.8Hz), 4.33 (1H, ddd, J = 10.8Hz, 3.2Hz, 2.4Hz), 4.10 (1H, t, J = 10.4Hz), 3.55-3.50 (3H, m), 3.33- 3.24 (1H, m), 2.94-2.87 (1H, m), 0.91-0.95 (2H, m), 0.10 (9H, s). Exchangeable proton not seen. [0280] Step 10 – [3-[1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6- yl] acetate [0281] Acetyl chloride (0.94mL, 13.29mmol) was added to a stirred solution of 2-(1- benzylimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-ol (3.07g, 8.86mmol), Et3N (1.85mL, 13.29mmol) and DCM (100mL) at rt under a nitrogen atmosphere. The reaction was stirred for 30 minutes, after which time water (100mL) was added. The mixture was extracted with DCM (2x100mL). The combined organic extracts were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-100% EtOAc in petroleum ether to give [3-[1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (2.93g, 7.53mmol, 85% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.48 min, m/z 389.5 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.07-6.99 (2H, m), 6.88-6.81 (3H, m), 5.33-5.27 (2H, m), 4.46-4.41 (1H, m), 4.25-4.18 (1H, m), 3.57-3.50 (2H, m), 3.48-3.38 (2H, m), 3.03-2.95 (1H, m), 2.25 (3H, s), 0.96-0.89 (2H, m), 0.00 (9H, s). [0282] Step 11 – [3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]ch roman-6-yl] acetate [0283] N-Bromosuccinimide (0.46g, 2.57mmol) was added to a stirred solution of [3-[1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (1g, 2.57mmol) and DCM (100mL) at 0°C under a nitrogen atmosphere. The reaction was stirred for 30 minutes and then diluted with water (100mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-50% EtOAc in petroleum ether to give [3-[4-bromo-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl] acetate (684mg, 1.46mmol, 57% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 2.07 min, m/z 467.2, 469.2 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 6.98 (1H, s), 6.85-6.80 (3H, m), 5.33 (2H, s), 4.46-4.41 (1H, m), 4.15-4.07 (2H, m), 3.62-3.56 (1H, m), 3.51-3.41 (1H, m), 3.38-3.30 (1H, m), 3.00-2.94 (1H, m), 2.27 (3H, s), 0.96-0.89 (2H, m), 0.00 (9H, s). [0284] Step 12 - 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chr oman-6-ol [0285] [3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]ch roman-6-yl] acetate (800mg, 1.71mmol) was stirred in a mixture of aq. 1M NaOH (10mL, 10mmol) and THF (10mL) at rt. After 30 minutes, the reaction was taken to ~pH 5 using aq.1M HCl and then it was diluted with water (50ml) and extracted with DCM (2x50mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo to give 3-[4-bromo-1- (2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (728mg, 1.71mmol, 100% yield) as a yellow oil. The compound was used in the next step without further purification. UPLC- MS (ES+, short acidi): 1.87 min, m/z 425.1, 427.1 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 6.99 (1H, s), 6.75-6.73 (1H, m), 6.64-6.60 (1H, m), 6.58-6.56 (1H, m), 5.34 (2H, s), 4.80-4.71 (1H, m), 4.42-4.36 (1H, m), 4.06 (1H, t, J = 10.4Hz), 3.61-3.57 (2H, m), 3.51-3.40 (1H, m), 3.37-3.28 (1H, m), 2.95-2.88 (1H, m), 0.96-0.89 (2H, m), 0.00 (9H, s). [0286] Step 13 – 5-[3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl] chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [0287] 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chr oman-6-ol (2g, 4.7mmol), 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (781.2mg, 4.7mmol), K 2 CO 3 (2.6g, 18.81mmol) and DMSO (5mL) were combined and stirred at 110°C under a nitrogen atmosphere for 1 hour. The reaction was cooled to rt and poured into a solution of water (100mL) and citric acid monohydrate (3.95g, 18.81mmol). The resultant mixture was extracted with DCM (2x100mL) and the combined organic layers dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-10% MeOH in DCM to give 5-[3-[4-bromo-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3, 4-dihydro-1H-1,8- naphthyridin-2-one (230mg, 0.40mmol, 9% yield) as a yellow solid. UPLC-MS (ES+, short acidic), 1.96 min, m/z 571.2, 573.2 [M+H]+. [0288] Example 5. Synthesis of 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid and 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-c arboxylic acid [0289] Step 1 – 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid [0290] A solution of 6-hydroxychromane-3-carboxylic acid hydrochloride (1.4g, 6.07mmol), 2-chloro-4-fluoropyridine (550µL, 6.09mmol) and K 2 CO 3 (3.36g, 24.28mmol) in DMSO (7.6mL) was heated to 110°C under a nitrogen atmosphere for 1.5 hours. The reaction mixture was cooled to rt and poured into a solution of citric acid (4.67g, 24.28mmol) in water (100mL). The resulting precipitate was filtered, washed with water and dried to give 6-[(2-chloro-4- pyridyl)oxy]chromane-3-carboxylic acid (1.74g, 5.69mmol, 94% yield) as a brown solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.55 min, m/z 306.1 [M+H]+. 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.69 (1H, br s), 8.27 (1H, d, J = 5.7Hz), 7.04 (1H, d, J = 2.9Hz), 6.97-6.89 (3H, m), 6.86 (1H, d, J = 8.8Hz), 4.34 (1H, dd, J = 10.9, 3.2Hz), 4.19-4.13 (1H, m), 3.05-2.95 (3H, m). [0291] Step 2 – 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-c arboxylic acid [0292] To a solution of 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid (500.mg, 1.64mmol), cyclopropanecarboxamide (278.4mg, 3.27mmol) and Cs 2 CO 3 (1.07g, 3.27mmol) in 1,4-dioxane (16mL) was added (+/-)-BINAP (203.7mg, 0.33mmol) and tris(dibenzylideneacetone)dipalladium (0) (149.7mg, 0.16mmol) under a nitrogen atmosphere. The mixture was heated at 100°C for 20 hrs. After cooling to rt, the mixture was filtered through celite and washed with methanol. The solvent was removed under reduce pressure and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM. Fractions containing the product were combined, solvent removed in vacuo, the residue suspended in DCM and filtered. The precipitate was washed with DCM and dried to give 6- [[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-car boxylic acid (272.3mg, 0.77mmol, 47% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.23 min, m/z 355.2 [M+H]+. 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.69 (1H, br s), 10.81 (1H, s), 8.15 (1H, d, J = 5.7Hz), 7.63 (1H, d, J = 2.4Hz), 6.97 (1H, d, J = 2.7Hz), 6.88 (1H, dd, J = 8.8Hz, 2.7Hz), 6.83 (1H, d, J = 8.8Hz), 6.59 (1H, dd, J = 5.7Hz, 2.4Hz), 4.34 (1H, dd, J = 10.7Hz, 2.8Hz), 4.14 (1H, dd, J = 10.7Hz, 6.7Hz), 3.04-2.92 (3H, m), 1.97 (1H, quint, J = 6.2Hz), 0.77 (4H, d, J = 6.2Hz). [0293] Example 6. Synthesis of 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline hydrochloride [0294] Step 1 - acetic acid; 1,2,3,4-tetrahydroisoquinolin-7-ol [0295] Platinum(IV) oxide (Adam’s Catalyst) (500mg, 2.2mmol) was added to 7- hydroxyisoquinoline (5.g, 34.45mmol) in acetic acid (20mL) at rt. The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 18 hours. The crude was filtered over celite and the filtrate concentrated in vacuo. The residue was stirred in an acetone/petroleum ether mixture (1:2, 15mL) for 1 hour, causing a solid to crash out which was filtered off and dried in vacuo to give acetic acid; 1,2,3,4- tetrahydroisoquinolin-7-ol (6.53g, 31.2mmol, 91% yield) as a light yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 0.36 min, m/z 150.2 [M+H]+. [0296] Step 2 - benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate [0297] Benzyl chloroformate (2.05mL, 14.34mmol) was added to a stirred solution of THF (15mL), acetic acid; 1,2,3,4-tetrahydroisoquinolin-7-ol (2.5g, 11.95mmol) and NaOH (35.84mL, 35.84mmol) at rt. The reaction was stirred for 1 hour and then diluted with water (200mL) and EtOAc (200mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give benzyl 7-hydroxy-3,4-dihydro-1H- isoquinoline-2-carboxylate (3.3g, 11.65mmol, 97% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.65 min, m/z 284.2 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.40-7.29 (5H, m), 6.98 (1H, d, J = 8.4Hz), 6.70-6.54 (2H, m), 5.18 (2H, s), 4.61-4.56 (2H, m), 3.75-3.66 (2H, m), 2.81-2.73 (2H, m). Exchangeable proton not seen. [0298] Step 3 – benzyl 7-benzyloxy-3,4-dihydro-1H-isoquinoline-2-carboxylate [0299] Benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (569mg, 1.45mmol) was dissolved in DMF (8mL) followed by addition of benzyl bromide (0.21mL, 1.74mmol) and K 2 CO 3 (600mg, 4.34mmol). The reaction was left stirring at 50°C for 1.5 hour and then at rt overnight. The solvent was removed under reduce pressure. Water was added, followed by extraction with EtOAc (2x). The organic layers were combined, washed with brine (2x), dried over Na 2 SO 4 and the solvent removed in vacuo. The crude was purified by column chromatography using as eluent a gradient 0-25% EtOAc in petroleum ether to give benzyl 7- benzyloxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (428mg, 1.15mmol, 79% yield) as a colourless oil. UPLC-MS (ES+, short acidic): 2.12 min, m/z 374.2 [M+H]+. 1 H-NMR (400MHz, CDCl 3 ) δ/ppm: 7.50-7.28 (10H, m), 7.04 (1H, d, J = 8.4Hz), 6.81 (1H, dd, J = 8.4Hz, 2.4Hz), 6.72 (1H, br d, J = 12.0Hz), 5.18 (2H, s), 5.04 (2H, s), 4.61 (2H, s), 3.71 (2H, br s), 2.78 (2H, br s). [0300] Step 4 – 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline hydrochloride [0301] To benzyl 7-benzyloxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (196mg, 0.52mmol) was added HCl (4N in 1,4-dioxane - 6mL, 24mmol) and the reaction heated to 80°C for 72 hours. The solvent was removed under reduce pressure to give 7-benzyloxy-1,2,3,4- tetrahydroisoquinoline hydrochloride (144.7mg, 0.52mmol, 100% yield) as a cream solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.27 min, m/z 240.2 [M+H]+. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 9.18 (1H, br s), 7.52-7.37 (4H, m), 7.36-7.27 (1H, m), 7.14 (1H, d, J = 8.4Hz), 6.96-6.89 (2H, m), 5.10 (2H, s), 4.21 (2H, br s), 3.35-3.28 (2H, t, (under water)), 2.92 (2H, t, J = 6.0Hz). [0302] Example 7. Synthesis of 3-methyl-5-(trifluoromethyl)benzene-1,2-diamine [0303] 3-Bromo-4,5-diaminobenzotrifluoride (500mg, 1.96mmol), K 2 CO 3 (542mg, 3.92mmol), tetrakis(triphenylphosphine)palladium(0) (227mg, 0.20mmol) were mixed under N 2 followed by the addition of 2,4,6-Trimethylboroxin (0.27mL, 1.96mmol). The reaction was left stirring for 72 hours at 110°C, after which time a second portion of 2,4,6-trimethylboroxin (0.14mL, 0.98mmol), K 2 CO 3 (271mg, 1.96mmol) and tetrakis(triphenylphosphine)palladium(0) (113mg, 0.10mmol) were added. The reaction was stirred at 110°C overnight, after which time water was added (20mL) followed by extraction with EtOAc (2x). The organic phases were combined, washed with brine, dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The crude was purified by column chromatography using as eluent a gradient 0-5% MeOH in DCM to give 3-methyl-5-(trifluoromethyl)benzene- 1,2-diamine (82mg, 0.43mmol, 22% yield) as a brown oil. UPLC-MS (ES+, short acidic): 1.37 min, m/z 191.1 [M+H]+. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 6.70-6.67 (1H, m), 6.63-6.59 (1H, m), 4.81 (2H, s), 4.79 (2H, s), 2.07 (3H, s). [0304] Example 8. Synthesis of 6-(trifluoromethyl)pyridine-3,4-diamine [0305] Step 1 - 5-nitro-2-(trifluoromethyl)pyridin-4-ol [0306] To a cooled solution of 2-(trifluoromethyl)pyridin-4-ol (1000mg, 6.13mmol) at 0°C in sulfuric acid (2.46mL, 46.15mmol) was added nitric acid, fuming, 90% (6.12mL, 144.04mmol) dropwise over 15 min, after which time the reaction was heated to 120°C overnight. The reaction mixture was cooled to rt and poured into ice water. The mixture was brought to neutral pH by addition of a solution 32% of aq. NaOH and extracted with EtOAc (3x) and n-BuOH. The combined organic phases were dried over Na 2 SO 4 , filtered and the solvent removed in vacuo to give 5-nitro-2-(trifluoromethyl)pyridin-4-ol (1.27g, 6.10mmol, 100% yield) as a yellow solid. The compound was used without further purification in the following step. UPLC- MS (ES-, short acidic): 1.17 min, m/z 207.1 [M-H]-. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 9.00 (1H, s), 7.30 (1H, s). [0307] Step 2 - 4-chloro-5-nitro-2-(trifluoromethyl)pyridine [0308] 5-nitro-2-(trifluoromethyl)pyridin-4-ol (1.28g, 6.13mmol), phosphorus pentachloride (1.91g, 9.19mmol) and phosphorus oxychloride (0.86mL, 9.19mmol) were stirred at 80°C overnight. The mixture was cooled to rt, diluted with DCM and washed with water, sat. aq. Na2CO 3 and brine. The organic phase was dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The compound was re-dissolved in DCMand poured into water-ice, followed by addition of aq. 1M NaOH. The organic phase was washed with brine, dried over Na 2 SO 4 , filtered and the solvent removed under reduce pressure to give 4-chloro-5-nitro-2- (trifluoromethyl)pyridine (804mg, 3.55mmol, 58% yield) as a yellow oil. UPLC-MS (ES-, short acidic): 1.68 min, m/z nd. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 9.43 (1H, s), 8.58 (1H, s). [0309] Step 3 - 5-nitro-2-(trifluoromethyl)pyridin-4-amine [0310] 4-chloro-5-nitro-2-(trifluoromethyl)pyridine (752mg, 3.32mmol) was dissolved with ammonia in MeOH (30.08mL, 69.19mmol) and left stirring at rt for 3 hours. The solvent was removed in vacuo to give 5-nitro-2-(trifluoromethyl)pyridin-4-amine (687mg, 3.32mmol, 100% yield) as a yellow solid. The product was used without further purification in the following step. UPLC-MS (ES+, short acidic): 1.36 min, m/z 208.1 [M+H]+. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 9.07 (1H, s), 7.44 (1H, s). [0311] Step 4 - 6-(trifluoromethyl)pyridine-3,4-diamine [0312] 5-nitro-2-(trifluoromethyl)pyridin-4-amine (734mg, 3.54mmol) was dissolved in EtOAc (15mL) and Ethanol (25mL) followed by the addition of palladium, 10 wt. % on carbon powder, (238mg) under a nitrogen atmosphere. The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 72 hours. A second portion of palladium, 10 wt. % on carbon powder, dry (238mg) was added and left stirring overnight. The crude was filtered over celite, washed with EtOAc and the solvent removed under reduce pressure to give 6-(trifluoromethyl)pyridine-3,4-diamine (571mg, 3.22mmol, 91% yield) as a colourless oil. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 7.69 (1H, s), 6.83 (1H, s), 5.79 (2H, br s), 5.21 (2H, br s). [0313] Example 9. Synthesis of 5-[(dimethylamino)methyl]-3-(trifluoromethyl)benzene-1,2- diamine [0314] Step 1 - 4-chloro-3-nitro-5-(trifluoromethyl)benzoic acid [0315] To 4-chloro-3-(trifluoromethyl)benzoic acid (1.5g, 6.68mmol) was added sulfuric acid (5.34mL, 100.20mmol) and nitric acid, fuming, 90% (0.85mL, 20.04mmol) and the reaction mixture was heated at 90°C for 1 hour. The crude was poured in ice-cold water (150mL), filtered and dried in vacuo to give 4-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (1.66g, 6.15mmol, 92% yield) as a white solid. UPLC-MS (ES-, short acidic): 1.66 min, m/z 268.1 [M- H]-. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 14.22 (1H, br s), 8.78 (1H, d, J = 2.0Hz), 8.44 (1H, d, J = 1.6Hz). [0316] Step 2 - [4-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol [0317] 4-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (1.66g, 6.15mmol) was dissolved in THF (20mL) and cooled to 0°C, followed by dropwise addition of di-methylsulfide borane (9.24mL, 18.46mmol). The reaction was stirred at rt for 72 hours, after which time it was quenched by slow addition of a sat. aq. NaHCO 3 and extracted with EtOAc (x2). The organic layers were combined, dried over Na 2 SO 4 , filtered and the solvent removed under reduce pressure to give 4-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.31g, 5.11mmol, 83% yield) as a pale yellow oil. 1 H-NMR (400MHz, CDCl 3 ) δ/ppm: 7.94 (1H, s), 7.91 (1H, s), 4.84 (2H, s). Exchangeable proton missing. [0318] Step 3 - 5-(bromomethyl)-2-chloro-1-nitro-3-(trifluoromethyl)benzene [0319] A solution of [4-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.31g, 5.11mmol) in DCM (15mL) was cooled to 0°C followed by the addition of triphenylphosphine (1.61g, 6.13mmol) and tetrabromomethane (1864.1mg, 5.62mmol). The reaction was left stirring at rt overnight, after which time it was washed with sat. aq. NaHCO 3 . The organic layer was separated, dried over Na 2 SO 4 and the solvent removed in vacuo to give 5-(bromomethyl)-2- chloro-1-nitro-3-(trifluoromethyl)benzene (3.94g, 5.07mmol, 99% yield) as a yellow oil. 1 H- NMR (400MHz, CDCl3) δ/ppm: 7.97 (1H, d, J = 2.0Hz), 7.95 (1H, d, J = 2.0Hz), 4.52 (2H, s). [0320] Step 4 - 1-[4-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl- methanamine [0321] To 5-(bromomethyl)-2-chloro-1-nitro-3-(trifluoromethyl)benzene (500.mg, 0.6300mmol) was added dimethylamine (2M in THF - 3.14mL, 62.8mmol) and the reaction was stirred at rt for 2 hours. The solvent was removed in vacuo and the crude purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether. Fractions containing the product were loaded into an SCX-2 column and flushed at first with MeOH (20mL) and then NH3 in MeOH (20mL) to give 1-[4-chloro-3-nitro-5- (trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (101mg, 0.36mmol, 57% yield) as a pale yellow oil. UPLC-MS (ES+, short acidic): 1.19 min, m/z 283.1 [M+H]+. 1 H-NMR (400MHz, CDCl 3 ) δ/ppm: 7.91-7.90 (1H, m), 7.89-7.87 (1H, m), 3.50 (2H, s), 2.27 (6H, s). [0322] Step 5 - 4-[(dimethylamino)methyl]-2-nitro-6-(trifluoromethyl)aniline [0323] 1-[4-Chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl- methanamine (101mg, 0.36mmol) was dissolved in 1,4-dioxane (1mL) in a seal tube followed by addition of NH 3 (28% in H 2 O - 0.99mL, 7.15mmol). The reaction was stirred at 120°C overnight, after which time a second portion of NH 3 (28% in H 2 O - 0.99mL, 7.15mmol) was added. The reaction was left stirring overnight at 120°C. The crude was filtered over a phase separator to give 4- [(dimethylamino)methyl]-2-nitro-6-(trifluoromethyl)aniline (94mg, 0.36mmol, 100% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 0.90 min, m/z 264.2 [M+H]+. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.36 (1H, s), 7.95 (1H, s), 7.42 (2H, s), 3.78 (2H, s), 2.40 (6H, s). [0324] Step 6 - 5-[(dimethylamino)methyl]-3-(trifluoromethyl)benzene-1,2-dia mine [0325] 4-[(Dimethylamino)methyl]-2-nitro-6-(trifluoromethyl)aniline (94mg, 0.36mmol) was dissolved in EtOAc (10mL) and EtOH (17mL) followed by the addition of palladium, 10 wt. % on carbon powder, (12.1mg) under a nitrogen atmosphere. The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere overnight. The crude was filtered over celite and the solvent removed under reduce pressure to give 5-[(dimethylamino)methyl]-3-(trifluoromethyl)benzene-1,2-dia mine (77mg, 0.33mmol, 92% yield) as a yellow solid. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 6.81 (1H, s), 6.77 (1H, s), 5.15-5.00 (4H, m), 3.83 (2H, br s), 2.50 (6H, s, (under DMSO)). UPLC-MS (ES+, short acidic): 0.76 min, m/z 234.2 [M+H]+. [0326] Intermediates in the table below was made in an analogous manner, using 1- methylpiperazine in place of dimethylamine in step 4. [0327] Example 10. Synthesis of 3-[(dimethylamino)methyl]-5-(trifluoromethyl)benzene-1,2- diamine [0328] Step 1 - 2-chloro-3-nitro-5-(trifluoromethyl)benzoic acid [0329] 2-(Trifluoromethyl)pyridin-4-ol (1g, 6.13mmol) was dissolved in sulfuric acid (10.68mL, 200.39mmol) following by the addition of nitric acid, fuming, 90% (1.7mL, 40.08mmol). The reaction mixture was then heated to 90°C for 45 min, after which time the reaction mixture was cooled to rt and poured into ice-cold water (250mL). The product was filtered, washed with ice-cold water (40 mL) and dried in vacuo to give 2-chloro-3-nitro-5- (trifluoromethyl)benzoic acid (3.36g, 12.48mmol, 93% yield) as a white solid in 93% yield. UPLC-MS (ES-, short acidic): 1.46 min, m/z 268.1 [M-H]-. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.73-8.70 (1H, m), 8.43-8.39 (1H, m). Exchangeable proton not seen. [0330] Step 2 - [2-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol [0331] Isobutyl chloroformate (3.41mL, 26.31mmol) was added to a stirred solution of 4- methylmorpholine (4.82mL, 43.85mmol), 2-chloro-3-nitro-5-(trifluoromethyl)benzoic acid (2.36g, 8.77mmol) in DCM (70mL) at 0°C under a nitrogen atmosphere. The reaction was stirred at rt overnight after which time the solvent was evaporated under reduce pressure. The crude was dissolved in THF (50mL) and slowly added to a solution of NaBH 4 (497.64mg, 13.16mmol) in H 2 O (50mL) at 0°C. The reaction was stirred for 1h at rt, after which time it was quenched by carefully adding to sat. aq. NH 4 Cl (100mL), followed by extraction with DCM (2x100mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The crude was purified by column chromatography using an eluent of 0- 50% EtOAc in petroleum ether to give [2-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.21g, 1.51mmol, 17%) yield as a yellow oil. UPLC-MS (ES+/-, short acidic): 1.63 min, m/z nd. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.13 (1H, s), 8.00 (1H, s), 4.93 (2H, s). [0332] Step 3 - 1-(bromomethyl)-2-chloro-3-nitro-5-(trifluoromethyl)benzene [0333] A solution of [2-chloro-3-nitro-5-(trifluoromethyl)phenyl]methanol (1.21g, 1.51mmol) in DCM (10mL) was cooled to 0°C followed by the addition of triphenylphosphine (475mg, 1.81mmol) and tetrabromomethane (551mg, 1.66mmol). The reaction was left stirring at rt overnight, after which time a second portion of triphenylphosphine (475mg, 1.81mmol) and tetrabromomethane (551mg, 1.66mmol) were added and left stirring for 4h. The reaction was washed with a sat. aq. NaHCO 3 ; the organic phase was separated, dried over Na 2 SO 4 and the solvent removed in vacuo to give 1-(bromomethyl)-2-chloro-3-nitro-5- (trifluoromethyl)benzene (2.71g, 1.49mmol, 99% yield) as a yellow oil. UPLC-MS (ES+/-, short acidic): 1.26 min, m/z nd. [0334] Step 4 - 1-[2-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl- methanamine [0335] To 1-(Bromomethyl)-2-chloro-3-nitro-5-(trifluoromethyl)benzene (1g, 0.55mmol) was added dimethylamine (2M in THF - 2.74mL, 54.95mmol). The reaction was left stirring at rt for 5 min, after which time the solvent was removed in vacuo and the crude purified by column chromatography using as eluent a gradient 0-20% EtOAc in petroleum ether. Fractions containing the product were loaded into an SCX-2 column and flushed at first with MeOH (10mL) and then NH3 in MeOH (10mL) to give 1-[2-chloro-3-nitro-5- (trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (82mg, 0.29mmol, 53% yield) as an orange oil. UPLC-MS (ES+, short acidic): 1.07 min, m/z 283.1 [M+H]+. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.08-8.05 (1H, m), 7.94-7.90 (1H, m), 3.65 (2H, s), 2.34 (6H, s). [0336] Step 5 - 2-[(dimethylamino)methyl]-6-nitro-4-(trifluoromethyl)aniline [0337] 1-[2-chloro-3-nitro-5-(trifluoromethyl)phenyl]-N,N-dimethyl- methanamine (82mg, 0.29mmol) was dissolved in 1,4-dioxane (0.8100mL) in a seal tube followed by addition of NH3 (28% in H 2 O - 0.81mL, 5.8mmol) and the reaction was left stirring at 120°C overnight. The crude was filtered over a phase separator to give 2-[(dimethylamino)methyl]-6-nitro-4- (trifluoromethyl)aniline (76mg, 0.29mmol, 100% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.11 min, m/z 264.2 [M+H]+. 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.24 (1H, s), 8.12 (2H, s), 7.70 (1H, s), 3.66 (2H, br s), 2.23 (6H, s). [0338] Step 6 - 3-[(dimethylamino)methyl]-5-(trifluoromethyl)benzene-1,2-dia mine [0339] 2-[(Dimethylamino)methyl]-6-nitro-4-(trifluoromethyl)aniline (75mg, 0.28mmol) was dissolved in EtOAc (5mL) and EtOH (9mL) followed by the addition of palladium, 10 wt. % on carbon powder, (10mg) under a nitrogen atmosphere. The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 4 hours. The crude was filtered over celite and the solvent removed under reduce pressure to give 3-[(dimethylamino)methyl]-5-(trifluoromethyl)benzene-1,2-dia mine (66mg, 0.28mmol, 99% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 0.91 min, m/z 234.2 [M+H]+. 1 H- NMR (400MHz, DMSO-d 6 ) δ/ppm: 6.78 (2H, s), 6.70 (2H, br s), 4.98 (4H, br s), 2.28 (6H, br s). [0340] Example 11. Synthesis of N-[[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5- (trifluoromethyl)phenyl]methyl]propan-2-amine [0341] Step 1 - N-[[3-bromo-5-(trifluoromethyl)phenyl]methyl]propan-2-amine [0342] 2-Aminopropane (0.34mL, 3.95mmol) was added to a stirred solution of 3-Bromo-5- (trifluoromethyl)benzaldehyde (0.6mL, 3.95mmol) and MeOH (10mL) at rt under a nitrogen atmosphere for 18 hours. NaBH 4 (224.3mg, 5.93mmol) was then added and the resulting mixture stirred for 10 minutes and quenched with water (5mL). Organic solvent was then removed in vacuo and the remaining mixture partitioned between water (50mL) and EtOAc (50mL). The aqueous layer was further extracted with EtOAc (50mL). The combined organic layers were dried over Na2SO4, filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give N-[[3- bromo-5-(trifluoromethyl)phenyl]methyl]propan-2-amine (665mg, 2.25mmol, 57% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.13 min, m/z 295.9, 297.9 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.72 (1H, s), 7.66 (1H, s), 7.56 (1H, s), 3.84 (2H, s), 2.86 (1H, septet, J = 6.4Hz), 1.12 (6H, d, J = 6.4Hz). Exchangeable proton not seen. [0343] Step 2 - N-[[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5- (trifluoromethyl)phenyl]methyl]propan-2-amine [0344] N-[[3-bromo-5-(trifluoromethyl)phenyl]methyl]propan-2-amine (300mg, 1.01mmol), bis(pinacolato)diboron (283mg, 1.11mmol), KOAc (298.3mg, 3.04mmol), [1,1'- bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (82.7mg, 0.10mmol) and 1,4-dioxane (10mL) were stirred at 90°C under a nitrogen atmosphere for 2 hours, after which time the reaction was cooled to rt and solvent removed in vacuo. The residue was dissolved in DCM (10mL), filtered over celite, which was washed with DCM (10mL). The filtrate was concentrated in vacuo and the residue purified by column chromatography using an eluent of 0-5% MeOH in DCM to give N-[[3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 5-(trifluoromethyl)phenyl]methyl]propan-2-amine (185mg, 0.54mmol, 53% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.02 min, m/z 262.1 [M-pinacol+H]+; 1.41 min, m/z 344.3 [M+H]+. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.96-7.93 (2H, m), 7.71 (1H, s), 3.85 (2H, s), 2.88 (1H, septet, J = 6.4Hz), 1.38 (12H, s), 1.13 (6H, d, J = 6.4Hz). Exchangeable proton not seen. [0345] Example 12. Synthesis of [3-[(dimethylamino)methyl]-5- (trifluoromethyl)phenyl]boronic acid [0346] Step 1 - [3-bromo-5-(trifluoromethyl)phenyl]methanol [0347] Sodium Borohydride (149.5mg, 3.95mmol) was added to a stirred solution of 3-bromo- 5-(trifluoromethyl)benzaldehyde (500mg, 1.98mmol) and MeOH (10mL) at rt for 1 hour. The reaction was poured into water (50mL) and the resulting mixture was extracted with EtOAc (2x50mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo to give [3-bromo-5-(trifluoromethyl)phenyl]methanol (444mg, 1.74mmol, 88% yield) as a colourless oil. The product was used in the next step without further purification. UPLC- MS (ES+, short acidic): 1.68 min, m/z nd. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.74 (1H, s), 7.71 (1H, s), 7.59 (1H, s), 4.79 (2H, s). Exchangeable proton not seen. [0348] Step 2 - 1-bromo-3-(bromomethyl)-5-(trifluoromethyl)benzene [0349] [3-Bromo-5-(trifluoromethyl)phenyl]methanol (444mg, 1.74mmol) was added to a stirred solution of triphenylphosphine (685mg, 2.61mmol), tetrabromomethane (866mg, 2.61mmol) and THF (20mL) at -10°C under a nitrogen atmosphere. The reaction was allowed to warm to rt and stir for 2 hours. It was then concentrated in vacuo and partitioned between water (100mL) and DCM (100mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-50% EtOAc in petroleum ether to give 1-bromo-3-(bromomethyl)-5- (trifluoromethyl)benzene (469mg, 1.48mmol, 85% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 2.06 min, m/z nd. 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.76-7.72 (2H, m), 7.60 (1H, s), 4.47 (2H, s). [0350] Step 3 - 1-[3-bromo-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanami ne [0351] 1-Bromo-3-(bromomethyl)-5-(trifluoromethyl)benzene (469mg, 1.48mmol) was added to a stirred solution of dimethylamine (2M in THF - 3mL, 6mmol) at rt under a nitrogen atmosphere and left stirring for 10 min. Solvent was removed in vacuo and the residue partitioned between water (20mL) and DCM (20mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 1-[3-bromo-5- (trifluoromethyl)phenyl]-N,N-dimethyl-methanamine (337mg, 1.19mmol, 81% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 1.13 min, m/z 281.9, 283.9 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.70-7.67 (2H, m), 7.54 (1H, s), 3.46 (2H, s), 2.27 (6H, s). [0352] Step 4 – [3-[(dimethylamino)methyl]-5-(trifluoromethyl)phenyl]boronic acid [0353] 1-[3-Bromo-5-(trifluoromethyl)phenyl]-N,N-dimethyl-methanami ne (337mg, 1.19mmol), bis(pinacolato)diboron (334mg, 1.31mmol), KOAc (352mg, 3.58mmol), [1,1'- bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (98mg, 0.1200mmol) and 1,4-dioxane (10mL) under a nitrogen atmosphere and stirred at 90°C for 2 hours. The reaction was cooled to rt and solvent removed in vacuo. The residue was suspended in DCM (10mL), filtered over celite, which was washed with DCM (10mL). The filtrate was concentrated in vacuo and the residue purified by column chromatography using an eluent of 0-5% MeOH in DCM to give [3-[(dimethylamino)methyl]-5-(trifluoromethyl)phenyl]boronic acid (109mg, 0.44mmol, 37% yield) as a yellow oil. UPLC-MS (ES+, short acidic): 0.93 min, m/z 248.1 [M+H] + . [0354] Example 13. Synthesis of 2-ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridine [0355] Step 1 - 2-ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrid ine [0356] [1,1'-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (0.09g, 0.11mmol) was added to a stirred solution of bis(pinacolato)diboron (0.3g, 1.18mmol), 4-bromo-2-ethylpyridine (0.2g, 1.07mmol), KOAc (0.16g, 1.61mmol) and 1,4- dioxane (20mL) under a nitrogen atmosphere and the reaction was heated to 100°C for 2 hours. The reaction was cooled to rt and solvent removed in vacuo. The residue was taken up in DCM (50mL), filtered over celite, which was washed with DCM (10mL). The filtrate was concentrated in vacuo to give 2-ethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrid ine (250mg, 1.0725mmol, 99.765% yield) as a black oil. The product was used in the next step without further purification. UPLCMS (ES+, short acidic): 0.83 min, m/z 151.9 [M-pinacol] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 8.57 (1H, d, J = 9.2Hz), 7.54 (1H, d, J = 6.4Hz), 7.47 (1H, d, J = 9.2Hz), 2.85 (2H, q, J = 8.0Hz), 1.39 (12H, s), 1.33 (3H, t, J = 8.0Hz). [0357] Intermediates in the table below was made in an analogous manner, using the appropriate (hetero)aryl bromide in place of 4-bromo-2-ethylpyridine:

[0358] Example 14. Synthesis of 1-ethyl-3-methyl-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazole [0359] n-Butyllithium solution (0.4mL, 1mmol) was added to a stirred solution of 1-ethyl-3- methyl-pyrazole (0.1mL, 0.91mmol) and THF (5mL) at 0°C under a nitrogen atmosphere. The mixture was allowed to warm to 25°C for 1 hour and then cooled back to -78°C.2-Isopropoxy- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (0.22mL, 1.09mmol) was added; after 10 mins at - 78°C the reaction was allowed to warm to room temperature and stir for 2 hours. The reaction was quenched with water and the resulting mixture reduced in vacuo. The residue was azeotroped with toluene to give 1-ethyl-3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2 - yl)pyrazole (214mg, 0.91mmol, 100% yield) as an off white solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 0.83 min, m/z 154.6 [M+H] + . [0360] Example 15. Synthesis of 1-cyclopropyl-3-ethyl-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazole [0361] Step 1 – 5-bromo-1-cyclopropyl-3-ethyl-pyrazole [0362] 2-cyclopropyl-5-ethyl-pyrazol-3-ol (730mg, 4.8mmol) was suspended in PBr3 (3.05mL, 16.79mmol) with MeCN (3mL) in a sealable vial. The vial was sealed and the reaction was heated thermally to 150°C for 1.5 hours. After cooling to rt, the reaction separates into two layers; the top layer (MeCN) was quenched into iced saturated aq. NaHCO 3 to pH ~7- 8. The aqueous layer was separated, extracted with EtOAc (3x); the organics were combined, dried through a phase separator and reduced in vacuo to give 5-bromo-1-cyclopropyl-3-ethyl- pyrazole (340mg, 1.58mmol, 33% yield) as an orange oil. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.75 min, m/z 214.9, 216.9 [M+H] + . [0363] Step 2 – 1-cyclopropyl-3-ethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2- yl)pyrazole [0364] [1,1'-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (129.1mg, 0.16mmol) was added to a stirred solution of bis(pinacolato)diboron (401.4mg, 1.58mmol), 5-bromo-1-cyclopropyl-3-ethyl-pyrazole (340mg, 1.58mmol), potassium acetate (310.3mg, 3.16mmol) and 1,4-dioxane (10mL) at room temperature under inert atmosphere. The reaction was heated to 85°C for 1 hour, cooled to room temperature and solvent removed in vacuo. The residue was taken up in DCM (20mL), filtered through celite and the filter cake washed with DCM (10mL). The filtrate was concentrated in vacuo to give 1-cyclopropyl-3-ethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2-yl)pyrazole (414mg, 1.58mmol, 100% yield) as a black oil. The compound was used in the next step without further purification. UPLCMS (ES+, short acidic): 1.05 min, m/z 180.7 [M-pinacol] + . [0365] Intermediates in the table below were made in an analogous manner, using the appropriate starting material in step 1: [0366] Synthesis of Compounds of the Disclosure [0367] Example 16. Synthesis of 5-[3-[4-[3-(trifluoromethyl)phenyl]-1H-imidazol-2- yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 12) [0368] Step 1 - [2-oxo-2-[3-(trifluoromethyl)phenyl]ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8- naphthyridin-4-yl)oxy]chromane-3-carboxylate [0369] 6-[(7-Oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3-carboxylic acid (200mg, 0.59mmol) and K 2 CO 3 (244mg, 1.76mmol) were dissolved in dry DMF (5mL) and treated with 3-(trifluoromethyl)phenacyl bromide (173mg, 0.65mmol) portionwise. The reaction was stirred for 2.5 hours at rt, after which time, a second portion of 3- (trifluoromethyl)phenacyl bromide was added (172mg, 0.65mmol) and the reaction was left stirring for 1h. Water (15 mL) was added and the product was extracted with DCM (x3). The combined organic phases were dried over Na 2 SO 4 , filtered and the solvent removed under reduce pressure. The product was purified by column chromatography using as eluent a gradient from 0-100% EtOAc in petroleum to give 2-oxo-2-[3-(trifluoromethyl)phenyl]ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3-carboxylate (55mg, 0.10mmol, 18% yield) as yellow solid. UPLC-MS (ES+, short acidic): 1.78 min, m/z 527.4 [M+H] + . [0370] Step 2 - 5-[3-[4-[3-(trifluoromethyl)phenyl]-1H-imidazol-2-yl]chroman -6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one [0371] [2-oxo-2-[3-(trifluoromethyl)phenyl]ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8- naphthyridin-4-yl)oxy]chromane-3-carboxylate (55mg, 0.10mmol) and ammonium acetate (1.21g, 15.67mmol) were mixed in a sealed vial and the reaction heated to 130°C for 8 hours. Water was added and the product extracted with EtOAc (x3). The organic phase was dried over Na 2 SO 4 , filtered and the solvent removed under reduce pressure. The product was purified by column chromatography using a gradient 0-100% EtOAc in petroleum ether followed by 0-5% MeOH in DCM to give 5-[3-[4-[3-(trifluoromethyl)phenyl]-1H-imidazol-2-yl]chroman -6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 12) as a white solid. UPLC-MS (ES+, final purity) 3.17 min, m/z 507.3 [M+H] + . 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.51 (0.10H, br s), 12.26 (0.90H, br s), 10.46 (1H, s), 8.10-7.99 (2H, m), 7.95 (1H, d, J = 6.0Hz), 7.82 (1H, s), 7.60-7.48 (2H, m), 7.02 (1H, s), 6.94-6.87 (2H, m), 6.26 (1H, d, J = 5.6Hz), 4.55- 4.45 (1H, m), 4.19-4.09 (1H, m), 3.46-3.38 (1H, m), 3.30-3.20 (1H, m), 3.17-3.08 (1H, m), 2.93 (2H, t, J = 7.6 Hz), 2.56-2.53 (m, 2H). [0372] Example 17. Synthesis of 5-[3-[4-(3-methoxyphenyl)-1H-imidazol-2-yl]chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 13) [0373] Step 1 - [2-(3-methoxyphenyl)-2-oxo-ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8- naphthyridin-4-yl)oxy]chromane-3-carboxylate [0374] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3-carboxylic acid (200mg, 0.59mmol) and 2-bromo-1-(3-methoxyphenyl)ethanone (135mg, 0.59mmol) were dissolved in dry MeCN (10mL) and cooled to 0°C followed by dropwise addition of DIPEA (0.15mL, 0.88mmol). The reaction was stirred at rt for 2 hours. After this time, iced water was added followed by extraction with EtOAc (x3) and DCM (x2) The combined organic phases were dried over Na 2 SO 4 , filtered and the solvent evaporated under reduce pressure. The product was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give [2-(3-methoxyphenyl)-2-oxo-ethyl] 6-[(7- oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3-carb oxylate (217mg, 0.44mmol, 76% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.69 min, m/z 489.3 [M+H] + . 1 H- NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.46 (1H, s), 7.95 (1H, d, J = 6.0Hz), 7.59-7.55 (1H, m), 7.51-7.43 (2H, m), 7.27 (1H, ddd, J = 8.2Hz, 2.6Hz, 0.8Hz), 7.01 (1H, d, J = 2.8Hz), 6.93-6.85 (2H, m), 6.25 (1H, d, J = 5.6Hz), 5.57 (2H, s), 4.44 (1H, dd, J = 10.8Hz, 3.2Hz), 4.24 (1H, dd, J = 10.8 Hz, 8.0Hz), 3.83 (3H, s), 3.31-3.25 (1H, m), 3.10-3.05 (2H, m), 2.92 (2H, t, J = 8.0Hz), 2.53 (2H, t, J = 8.0Hz, (partly under DMSO)). [0375] Step 2 - 5-[3-[4-(3-methoxyphenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy- 3,4-dihydro- 1H-1,8-naphthyridin-2-one [0376] [2-(3-methoxyphenyl)-2-oxo-ethyl] 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4- yl)oxy]chromane-3-carboxylate (98mg, 0.20mmol) and ammonium acetate (309mg, 4.01mmol) were mixed in a sealed vial followed by addition of xylenes (1mL) and heated to 150°C for 30 min. Water was added and the product extracted with EtOAc (x3). The organic phase was dried over Na 2 SO 4 , filtered and the solvent removed under reduce pressure. The residue was loaded into an SCX-2 column and flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. Fractions containing the product were combined and purified by column chromatography using as eluent a gradient 0-5% MeOH in DCM. Fractions containing the product were re-purified by reverse column chromatography using as eluent a gradient 5-100% of acetonitrile+0.1% formic acid in water+0.1% formic acid to give 5-[3-[4-(3-methoxyphenyl)-1H-imidazol-2-yl]chroman-6-yl]oxy- 3,4-dihydro-1H-1,8- naphthyridin-2-one (Compound 13) (8mg, 0.017mmol, 9% yield) as a white solid. UPLC-MS (ES+, final purity) 2.76 min, m/z 469.3 [M+H] + . 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.29 (br s, 0.2H), 12.09 (br s, 0.8H), 10.46 (s, 1H).7.95 (d, J = 5.6Hz, 1H), 7.61 (d, J = 2.0Hz, 0.8H), 7.35-7.28 (m, 2H), 7.25-7.19 (m, 1.2H), 7.01 (d, J = 2.4Hz, 1H), 6.94-6.85 (m, 2H), 6.84-6.78 (m, 0.2H), 6.74 (ddd, J = 8.4Hz, 2.8Hz, 1.2Hz, 0.8H), 6.27 (d, J = 5.6Hz, 1H), 4.53-4.47 (m, 1H), 4.15-4.09 (m, 1H), 3.80 (s, 0.6H), 3.77 (s, 2.4H), 3.45-3.35 (m, 1H), 3.30-3.19 (m, 1H), 3.16-3.06 (m, 1H), 2.93 (t, J = 7.2 Hz, 2H), 2.54-2.52 (m, 2H). [0377] The compounds in the table below were made in an analogous manner to Example 17, using the appropriate 2-bromo-1-arylethanone in place of 2-bromo-1-(3- methoxyphenyl)ethanone in step 1:

[0378] The compound in the table below was made in an analogous manner, using the appropriate 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3-c arboxylic acid (Example 5) in place of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3- carboxylic in step 1 and using 2-bromo-1-phenylethanone in place of 2-bromo-1-(3- methoxyphenyl)ethanone in step 2: [0379] Example 18. Synthesis of 5-[3-[4-(2-methoxy-4-pyridyl)-1H-imidazol-2-yl]chroman- 6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 19) [0380] Step 1 - 5-[3-[4-(2-methoxy-4-pyridyl)-1-(2-trimethylsilylethoxymethy l)imidazol-2- yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [0381] 5-[3-[4-Bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl] chroman-6-yl]oxy- 3,4-dihydro-1H-1,8-naphthyridin-2-one (100mg, 0.18mmol), 2-methoxypyridine-4-boronic acid (32.1mg, 0.21mmol), [1,1'-Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (14.3mg, 0.020mmol), K 2 CO 3 (72.6mg, 0.52mmol), 1,4-dioxane (5mL) and water (1mL) were combined under a nitrogen atmosphere and heated to 90°C for 1 hour. The reaction was cooled to rt and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 5-[3-[4-(2-methoxy- 4-pyridyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chr oman-6-yl]oxy-3,4-dihydro-1H- 1,8-naphthyridin-2-one (96mg, 0.16mmol, 91% yield) as a orange oil. UPLC-MS (ES+, short acidic): 1.82 min, m/z 600.5 [M+H] + . [0382] Step 2 - 5-[3-[4-(2-methoxy-4-pyridyl)-1H-imidazol-2-yl]chroman-6-yl] oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one [0383] Trifluoroacetic acid (1mL, 13.06mmol) was added to a stirred solution of 5-[3-[4-(2- methoxy-4-pyridyl)-1-(2-trimethylsilylethoxymethyl)imidazol- 2-yl]chroman-6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one (97mg, 0.16mmol) in DCM (4mL) and EtOH (0.1mL) at rt and stirred for 48 hours, after which time solvent and TFA were removed in vacuo. The residue was loaded into an SCX-2 column and flushed at first with MeOH (20mL) and then NH 3 in MeOH (20mL) to elute the product. Product containing fractions were combined and concentrated in vacuo. The residue was purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 5-[3-[4-(2-methoxy-4-pyridyl)-1H-imidazol-2-yl]chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (21mg, 0.045mmol, 28% yield) as a white solid. UPLC-MS (ES+, short acidic): 2.66 min, m/z 470.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.34 (1H, s), 10.47 (1H, s), 8.07 (1H, d, J = 5.2Hz), 7.96 (1H, d, J = 5.6Hz), 7.87 (1H, d, J = 6.4Hz), 7.34-7.31 (1H, m), 7.10 (1H, d, J = 8.0Hz), 7.03-7.00 (1H, m), 6.95- 6.87 (2H, m), 6.27 (1H, d, J = 6.0Hz), 4.54-4.48 (1H, m), 4.14 (1H, t, J = 10.0Hz), 3.88-3.83 (3H, m), 3.46-3.39 (1H, m), 3.28-3.09 (2H, m), 2.94 (2H, t, J = 7.6Hz), 2.62-2.57 (2H, m, (under DMSO peak). [0384] The compounds in the table below were made in an analogous manner to Example 18, using the appropriate boronic acids/pinacol esters in place of 2-methoxypyridine-4-boronic acid in step 1: [0385] Example 19. Synthesis of 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1H-imidazol-2- yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 106) [0386] Step 1 - 3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymet hyl)imidazol-2- yl]chroman-6-ol [0387] 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chr oman-6-ol (200mg, 0.47mmol), K 2 CO 3 (195mg, 1.41mmol), 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazole (104.4mg, 0.47mmol), 1,4-dioxane (4mL) and water (1mL) were combined and stirred at rt under a nitrogen atmosphere. [1,1'- Bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (38.4mg, 0.050mmol) was added and the reaction heated to 100°C for 2 hours. The reaction was cooled to rt and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 25-100% EtOAc in petroleum ether to give 3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (143mg, 0.32mmol, 69% yield) as an orange oil. UPLC-MS (ES+, short acidic): 1.74 min, m/z 441.8 [M+H] + . [0388] Step 2 - 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxy methyl)imidazol- 2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [0389] Potassium tert-Butoxide (40mg, 0.36mmol) was added to a stirred solution of 5-fluoro- 3,4-dihydro-1H-1,8-naphthyridin-2-one (53.9mg, 0.32mmol) and 3-[4-(2,5-dimethylpyrazol- 3-yl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman- 6-ol (143mg, 0.32mmol) in DMF (2mL) and the reaction heated at 80°C for 18 hours. The reaction was cooled to rt, solvent was reduced in vacuo and the residue partitioned between EtOAc and water. The organic layer was washed with sat. brine, separated, dried over a phase separator and the solvent reduced in vacuo. The residue was purified by column chromatography using as eluent a gradient 25- 100% EtOAc in petroleum ether to give 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-3, 4-dihydro-1H-1,8- naphthyridin-2-one (83mg, 0.14mmol, 44% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.83 min, m/z 587.5 [M+H] + . [0390] Step 3 – 5-[3-[4-(2,5-dimethylpyrazol-3-yl)-1H-imidazol-2-yl]chroman- 6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one [0391] Trifluoroacetic acid (0.54mL, 7.08mmol) was added to a stirred solution of 5-[3-[4- (2,5-dimethylpyrazol-3-yl)-1-(2-trimethylsilylethoxymethyl)i midazol-2-yl]chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (83mg, 0.14mmol) and DCM (2mL) in a sealed vial and heated to 40°C for 18 hours. The solvent was removed under reduce pressure and the residue was loaded into an SCX-2 column and flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. The residue was purified by column chromatography using as eluent a gradient 1-8% MeOH in DCM. Fractions containing the product were combined, the solvent removed in vacuo and the residue was triturated in MeCN/Et 2 O to give a white solid, which was filtered and dried to give 5-[3-[4-(2,5- dimethylpyrazol-3-yl)-1H-imidazol-2-yl]chroman-6-yl]oxy-3,4- dihydro-1H-1,8- naphthyridin-2-one (33.5mg, 0.073mmol, 52% yield). UPLC-MS (ES+, final purity): 2.62 min, m/z 457.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.30 (0.13H, s), 12.26 (0.87H, s), 10.46 (1H, s), 7.96 (1H, d, J = 6.0Hz), 7.42 (0.87H, d, J = 1.6Hz), 7.13 (0.13H, d, J = 1.6Hz), 7.01-6.98 (1H, m), 6.94-6.81 (2H, m), 6.26-6.25 (1.13H, m), 6.14 (0.87H, s), 4.53-4.44 (1H, m), 4.13 (1H, t, J = 10.4Hz), 3.91 (2.61H, s), 3.80 (0.39H, s), 3.45-3.35 (1H, m), 3.27-3.06 (2H, m), 2.93 (2H, t, J = 8.0Hz), 2.53 (2H, t, J = 8.0Hz, (party under DMSO), 2.15 (2.61H, s), 2.11 (0.39H, s). [0392] The compounds in the table below were made in an analogous manner to Example 19, using the appropriate boronic acids/pinacol esters in place of 1,3-dimethyl-5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole in step 1: [0393] Example 20. Synthesis of 5-[3-(4-tetrahydropyran-4-yl-1H-imidazol-2-yl)chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 132)

[0394] Step 1 - 3-[4-(3,6-dihydro-2H-pyran-4-yl)-1-(2-trimethylsilylethoxyme thyl)imidazol- 2-yl]chroman-6-ol [0395] 3,6-Dihydro-2H-pyran-4-boronic acid pinacol ester (108.7mg, 0.52mmol), 3-[4- bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman- 6-ol (220mg, 0.52mmol), [1,1'-bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (42.2mg, 0.050mmol), potassium carbonate (214.4mg, 1.55mmol), 1,4-dioxane (4mL) and water (1mL) were combined at room temperature under a nitrogen atmosphere in a sealable vial. The vial was sealed and the reaction was heated to 80°C and allowed to stir for 2 hours. It was then cooled to room temperature and solvent removed in vacuo. The residue was taken up in DCM (20mL) and filtered through celite and the filter cake washed with DCM (10mL). The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 3-[4-(3,6-dihydro-2H-pyran-4-yl)-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (149mg, 0.35mmol, 67% yield) as a yellow solid. UPLCMS (ES+, short acidic): 1.53 min, m/z 429.7 [M+H] + . [0396] Step 2 - 3-[4-tetrahydropyran-4-yl-1-(2-trimethylsilylethoxymethyl)im idazol-2- yl]chroman-6-ol [0397] 3-[4-(3,6-dihydro-2H-pyran-4-yl)-1-(2-trimethylsilylethoxyme thyl)imidazol-2- yl]chroman-6-ol (149.mg, 0.35mmol) was stirred in EtOAc (10mL) at room temperature under a nitrogen atmosphere. Palladium, 10 wt. % on carbon powder, dry (20 mg) was added and the reaction fitted with a hydrogen balloon and subjected to 3 x vacuum/hydrogen cycles and then allowed to stir under a hydrogen atmosphere for 18 hours. The reaction was then filtered through a celite plug and the plug washed with MeOH (10mL). The filtrate was concentrated in vacuo to give 3-[4-tetrahydropyran-4-yl-1-(2-trimethylsilylethoxymethyl)im idazol-2- yl]chroman-6-ol (95mg, 0.22mmol, 63% yield) as a yellow solid. The compound was used directly in the next step without further purification. UPLC-MS (ES+, short acidic): 1.49 min, m/z 431.7 [M+H] + . Step 3 - 5-[3-(4-tetrahydropyran-4-yl-1H-imidazol-2-yl)chroman-6-yl]o xy-3,4-dihydro-1H- 1,8-naphthyridin-2-one Potassium tert-butoxide (75.4mg, 0.67mmol) was added to a stirred solution of 5-fluoro-3,4- dihydro-1H-1,8-naphthyridin-2-one (36.7mg, 0.22mmol) and 3-[4-tetrahydropyran-4-yl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (95mg, 0.22mmol) in DMF (2mL) and the reaction was sealed in a tube and heated at 90°C for 60 hours.Reaction was cooled to room temperature and the reaction was reduced in vacuo and partitioned between EtOAc and water. The organics were washed with saturated brine, separated, dried over a phase separator and reduced in vacuo. The residue was purified by column chromatography using as eluent a gradient 1-10% MeOH/DCM. Fractions containing the product were combined and re-purified by preparative HPLC (early method). Fractions containing the product were combined, solvent removed under reduce pressure and loaded onto a SCX column, which was flushed at first with MeOH and then 1.0M MeOH/NH 3 to give 5-[3-(4-tetrahydropyran-4-yl-1H-imidazol-2- yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (5mg, 0.011mmol, 5% yield) as a white solid. UPLC-MS (ES+, Final purity): 2.36 min, m/z 447.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 +CF 3 CO 2 D) δ/ppm: 14.11 (1H, s), 10.65 (1H, s), 8.00 (1H, d, J = 6.0Hz), 7.46 (1H, s), 7.03 (1H, d, J = 2.8Hz), 7.00-6.91 (2H, m), 6.30 (1H, d, J = 6.0Hz), 4.51 (1H, dd, J = 10.9Hz, 3.1Hz), 4.34 (1H, dd, J = 11.0Hz, 8.1Hz, 1H), 3.93 (2H, ddd, J = 11.5Hz, 4.5Hz, 2.0Hz), 3.83-3.73 (1H, m), 3.44 (2H, td, J = 11.7Hz, 2.1Hz), 3.30-3.23 (2H, m), 3.01-2.89 (3H, m), 2.56 (2H, dd, J = 8.4Hz, 7.0Hz), 1.92-1.82 (2H, m), 1.63 (2H, qd, J = 12.1Hz, 4.4Hz). [0398] Example 21. Synthesis of 5-[3-[5-(3-methoxy-2-thienyl)-1H-imidazol-2-yl]chroman- 6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 122)

[0399] Step 1 – tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate and tert-butyl N-[2-(3-methoxy-2-thienyl)-2-oxo-ethyl]carbamate [0400] n-Butyllithium solution (3.5mL, 8.76mmol) was added to a stirred solution of 3- methoxythiophene (0.87mL, 8.76mmol) and THF (20mL) at -78°C under a nitrogen atmosphere. The mixture was stirred for 20 minutes at -78°C, allowed to warm to 0°C for 20 minutes and then cooled back to -78°C and tert-butyl N-[2-[methoxy(methyl)amino]-2-oxo- ethyl]carbamate (477.9mg, 2.19mmol) was added. After 1 hour at -78°C the reaction was allowed to warm to room temperature and stir for 18 hours. The reaction was then poured into water (100mL) and the resulting mixture extracted with EtOAc (2x100mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate (93mg, 0.34mmol, 16% yield) and tert-butyl N-[2-(3-methoxy-2-thienyl)-2-oxo-ethyl]carbamate (170mg, 0.63mmol, 29% yield) as yellow oils. [0401] N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate: UPLCMS (ES+, short acidic): 1.62 min, m/z 294.0 [M+Na] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.37 (1H, d, J = 1.6Hz), 6.67 (1H, d, J = 1.6Hz), 5.39 (1H, br s), 4.53-4.52 (2H, m), 3.83 (3H, s), 1.45 (9H, s). [0402] N-[2-(3-methoxy-2-thienyl)-2-oxo-ethyl]carbamate: UPLCMS (ES+, short acidic): 1.60 min, m/z 272.0 [M+H]+, 294.0 [M+Na] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.59 (1H, d, J = 5.6Hz), 6.86 (1H, d, J = 5.6Hz), 5.56 (1H, br s), 4.52-4.50 (2H, m), 4.00 (3H, s), 1.47 (9H, s). [0403] Step 2 – 2-amino-1-(4-methoxy-2-thienyl)ethanone hydrochloride [0404] Hydrogen Chloride (0.26mL, 1.03mmol - 4M in dioxane) was added to a stirred solution of tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]carbamate (93mg, 0.34mmol) and DCM (5mL) at room temperature. The reaction was allowed to stir for 18 hours after which time the solvent was removed in vacuo to give 2-amino-1-(4-methoxy-2-thienyl)ethanone hydrochloride (71mg, 0.34mmol, 100% yield) as a yellow solid, which was used in the next step without further purification. UPLCMS (ES+, short acidic), 0.47 min, m/z 171.9 [M+H] + . [0405] Step 3 – N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]-6-[(7-oxo-6,8-dihydr o-5H-1,8- naphthyridin-4-yl)oxy]chromane-3-carboxamide [0406] Propylphosphonic anhydride (0.31mL, 0.51mmol) was added to a stirred solution of 6- [(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane-3- carboxylic acid (116.4mg, 0.34mmol), 2-amino-1-(4-methoxy-2-thienyl)ethanone hydrochloride (71mg, 0.34mmol), triethylamine (0.1mL, 0.68mmol) and THF (5mL). The reaction was heated to 65°C and stirred for 18 hours. The reaction was then cooled to room temperature and solvent removed in vacuo. The residue was partitioned between water (50mL) and DCM (50mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give N-[2-(4-methoxy- 2-thienyl)-2-oxo-ethyl]-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthy ridin-4-yl)oxy]chromane-3- carboxamide (52mg, 0.11mmol, 31% yield) as a white solid. UPLCMS (ES+, short acidic): 1.47 min, m/z 494.1 [M+H] + . [0407] Step 4 – 5-[3-[5-(3-methoxy-2-thienyl)-1H-imidazol-2-yl]chroman-6-yl] oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one [0408] N-[2-(4-methoxy-2-thienyl)-2-oxo-ethyl]-6-[(7-oxo-6,8-dihydr o-5H-1,8-naphthyridin- 4-yl)oxy]chromane-3-carboxamide (52mg, 0.11mmol) and ammonium acetate (243.7mg, 3.16mmol) were taken up in butyric acid (2mL, 21.88mmol) in a sealable vial, which was sealed and heated at 175°C for 2 hours. The reaction mixture was evaporated to dryness and then treated with aq. K 2 CO 3 solution and extracted with DCM (15ml). The organic layer was passed through a phase separator and then evaporated to dryness. The residue was chromatographed via preparative LCMS to give 5-[3-[5-(4-methoxy-2-thienyl)-1H-imidazol- 2-yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (8mg, 0.016mmol, 15% yield) as an off white solid product. UPLC-MS (ES+, final purity): 2.80 min, m/z 475.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 10.67 (1H, s), 8.05-7.91 (2H, m), 7.69 (1H, d, J = 1.5 Hz), 7.25 (1H, d, J = 1.7 Hz), 7.05 (1H, d, J = 2.5 Hz), 7.02-6.88 (2H, m), 6.76 (1H, d, J = 1.8 Hz), 6.32 (1H, d, J = 6.0 Hz), 4.56 (1H, dd, J = 10.9Hz, 3.2Hz), 4.35 (1H, dd, J = 11.0Hz, 8.6Hz), 3.79 (3H, s), 3.87-3.69 (1H, m), 3.35-3.23 (2H, m), 2.95 (2H, t, J = 7.7Hz), 2.60-2.53 (2H, m). [0409] The compounds in the table below were made in an analogous manner to Example 21, using the appropriate reagent in place of tert-butyl N-[2-(4-methoxy-2-thienyl)-2-oxo- ethyl]carbamate in step 2: [0410] Example 22. Synthesis of 5-[3-[4-(3,3-dimethylbutyl)-1H-imidazol-2-yl]chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 115) [0411] Step 1 - 3-[4-(3,3-dimethylbut-1-ynyl)-1-(2-trimethylsilylethoxymethy l)imidazol-2- yl]chroman-6-ol [0412] Tetrakis(triphenylphosphine)palladium(0) (54.3mg, 0.05mmol) was added to a stirred mixture of 3-[4-bromo-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chr oman-6-ol (200mg, 0.47mmol), 3,3-dimethyl-1-butyne (0.17mL, 1.41mmol), copper(I) iodide (9.0mg, 0.05mmol), K 2 CO 3 (130mg, 0.94mmol), monoglyme (2mL) and water (0.5mL) in a sealable vial at room temperature under a nitrogen atmosphere. The vial was sealed and the reaction heated to 90°C and stirred for 18 hours. After this time, the reaction mixture was cooled to room temperature and the solvent removed in vacuo. The residue was taken up in DCM, filtered through a celite plug and the filter cake washed with DCM (10mL). The filtrate was diluted with DCM (50mL) and the solution washed with water sat aq. NaHCO 3 (10mL), brine (50mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-10% EtOAc in petroleum ether to give 3-[4-(3,3-dimethylbut- 1-ynyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroma n-6-ol (156.5mg, 0.37mmol, 78% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 2.04 min, m/z 427.6 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.13 (1H, s), 6.73 (1H, d, J = 9.2Hz), 6.64-6.60 (1H, m), 6.59- 6.56 (1H, m), 5.37 (2H, s), 4.69 (1H, s), 4.43-4.37 (1H, m), 4.07 (1H, t, J = 10.0Hz), 3.60-3.55 (2H, m), 3.46-3.27 (2H, m), 2.95-2.88 (1H, m), 1.32 (9H, s), 0.95-0.88 (2H, m), 0.00 (9H, s). [0413] Step 2 – 3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imi dazol-2- yl]chroman-6-ol [0414] 3-[4-(3,3-dimethylbut-1-ynyl)-1-(2-trimethylsilylethoxymethy l)imidazol-2- yl]chroman-6-ol (233.mg, 0.55mmol) was stirred in MeOH (10mL) at room temperature under a nitrogen atmosphere followed by the addition of palladium, 10 wt. % on carbon powder, dry (30mg). The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 18 hours. The crude was filtered over celite, which was washed with MeOH (10mL) and the filtrate concentrated in vacuo to give 3-[4-(3,3- dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl ]chroman-6-ol (170mg, 0.39mmol, 72% yield) as a yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.76 min, m/z 431.9 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 6.77 (1H, s), 6.67 (1H, d, J = 8.8Hz), 6.61 (1H, dd, J = 8.8Hz, 2.4Hz), 6.56 (1H, d, J = 2.4Hz), 5.24-5.18 (2H, m), 4.33-4.27 (1H, m), 4.05 (1H, t, J = 10.8Hz), 3.57- 3.52 (2H, m), 3.43-3.36 (1H, m), 3.30-3.21 (1H, m), 2.86-2.79 (1H, m), 2.57-2.51 (2H, m), 1.59-1.53 (2H, m), 0.97 (9H, s), 0.96-0.91 (2H, m), 0.00 (9H, s). Exchangeable proton not seen. [0415] Step 3 – 5-[3-[4-(3,3-dimethylbutyl)-1-(2-trimethylsilylethoxymethyl) imidazol-2- yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [0416] Potassium tert-butoxide (132.9mg, 1.18mmol) was added to a stirred solution of 5- fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (65.6mg, 0.39mmol), 3-[4-(3,3- dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl ]chroman-6-ol (170mg, 0.39mmol) and DMF (2mL) at room temperature under a nitrogen atmosphere in a sealable vial. The vial was sealed and the reaction was heated to 80°C and stirred for 72 hours. The reaction was cooled to room temperature and solvent removed in vacuo. The residue was partitioned between water (20mL) and DCM (20mL). The organic layer was separated, passed through a phase separator and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient of 0-5% MeOH in DCM to give 5-[3-[4-(3,3- dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl ]chroman-6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one (116mg, 0.20mmol, 51% yield) as a yellow solid. UPLCMS (ES+, short acidic): 1.76 min, m/z 577.5 [M+H]+. [0417] Step 4 – 5-[3-[4-(3,3-dimethylbutyl)-1H-imidazol-2-yl]chroman-6-yl]ox y-3,4- dihydro-1H-1,8-naphthyridin-2-one [0418] Trifluoroacetic acid (0.5mL, 6.53mmol) was added to a stirred solution of 5-[3-[4-(3,3- dimethylbutyl)-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl ]chroman-6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one (116mg, 0.20mmol) and DCM (2mL) in a sealable vial at room temperature under a nitrogen atmosphere. The vial was sealed and the reaction was heated to 40°C and stirred for 18 hours. After this time the reaction was cooled to room temperature and solvent removed in vacuo and the residue loaded on to an SCX-2 column using the minimum amount of MeOH. MeOH (10mL) was passed through the column followed by NH 3 in MeOH (10 mL) to elute the product. Product containing fractions were concentrated in vacuo and the residue purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[4-(3,3-dimethylbutyl)-1H-imidazol-2-yl]chroman-6-yl]ox y-3,4-dihydro-1H-1,8- naphthyridin-2-one (10mg, 0.022mmol, 11% yield) as a white solid. UPLC-MS (ES+, final purity): 2.94 min, m/z 447.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 11.67 (0.5H, s), 11.58 (0.5H, s), 10.45 (1H, s), 7.95 (1H, d, J = 5.6Hz), 6.99-6.97 (1H, m), 6.93-6.85 (2H, m), 6.76 (0.5H, s), 6.49 (0.5H, s), 6.26 (1H, d, J = 5.6Hz), 4.46-4.40 (1H, m), 4.02 (1H, t, J = 10.8Hz), 3.29-3.23 (1H, m), 3.18-3.10 (1H, m), 3.06-2.99 (1H, m), 2.93 (2H, t, J = 7.2Hz), 2.55-2.50 (2H, m, (under DMSO)), 2.44-2.36 (2H, m), 1.50-1.43 (2H, m), 0.93-0.91 (9H, m). [0419] Example 23. Synthesis of 5-[3-[3-[2-(trifluoromethyl)phenyl]-1H-pyrazol-5- yl]chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 35) [0420] Step 1 - N-methoxy-N-methyl-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin -4- yl)oxy]chromane-3-carboxamide [0421] Propylphosphonic anhydride (50% in EtOAc - 1.31mL, 2.2mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3-carboxylic acid (500mg, 1.47mmol), N,O-dimethylhydroxylamine hydrochloride (158mg, 1.62mmol), triethylamine (0.31mL, 2.2mmol) and THF (100mL) at rt under a nitrogen atmosphere. The reaction was heated to 65°C and stirred for 18 hours, then cooled to rt and solvent removed in vacuo. The residue was partitioned between water (100mL) and DCM (100mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The crude was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give N-methoxy-N-methyl-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin -4-yl)oxy]chromane- 3-carboxamide (337mg, 0.88mmol, 60% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.39 min, m/z 384.3 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 8.13 (1H, s), 7.99 (1H, d, J = 6.0Hz), 6.92-6.89 (1H, m), 6.87-6.83 (2H, m), 6.32 (1H, d, J = 6.0Hz), 4.48-4.43 (1H, m), 4.06 (1H, t, J = 10.4Hz), 3.79 (3H, s), 3.45-3.35 (1H, m), 3.26 (3H, s), 3.23-3.14 (1H, m), 3.08 (2H, t, J = 7.2Hz), 2.93-2.86 (1H, m), 2.72 (2H, t, J = 7.2Hz). [0422] Step 2 - 5-[3-[3-[2-(trifluoromethyl)phenyl]prop-2-ynoyl]chroman-6-yl ]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one [0423] n-Butyllithium solution (2.5M in hexane - 0.35mL, 0.88mmol) was added dropwise to a stirred solution of 2-ethynyl-α,α,α-trifluorotoluene (0.12mL, 0.88mmol) in THF (20mL) at - 78°C under a nitrogen atmosphere. After stirring for 30 minutes a solution of N-methoxy-N- methyl-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]ch romane-3-carboxamide (168mg, 0.44mmol) in THF (10mL) was slowly added and the resultant reaction allowed to stir at -78°C for 1 hour, after which time it was warmed to rt and quenched with sat. aq. NH 4 Cl (100mL). This mixture was extracted with EtOAc (2x50mL), the combined organic layers dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[3-[2- (trifluoromethyl)phenyl]prop-2-ynoyl]chroman-6-yl]oxy-3,4-di hydro-1H-1,8-naphthyridin-2- one (163mg, 0.33mmol, 76% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.85 min, m/z 493.3 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 8.00 (1H, s), 7.97 (1H, d, J = 6.0Hz), 7.81-7.77 (2H, m), 7.65-7.62 (2H, m), 6.92-6.83 (3H, m), 6.32 (1H, d, J = 5.6Hz), 4.62-4.57 (1H, m), 4.46-4.40 (1H, m), 3.33-3.24 (2H, m), 3.16-3.11 (1H, m), 3.08 (2H, t, J = 8.0Hz), 2.72 (2H, t, J = 8.0Hz). [0424] Step 3 - 5-[3-[3-[2-(trifluoromethyl)phenyl]-1H-pyrazol-5-yl]chroman- 6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one [0425] Hydrazine Hydrate (0.03mL, 0.66mmol) was added to a stirred solution of 5-[3-[3-[2- (trifluoromethyl)phenyl]prop-2-ynoyl]chroman-6-yl]oxy-3,4-di hydro-1H-1,8-naphthyridin-2- one (163.mg, 0.3300mmol) and t-BuOH (2mL) at rt. The reaction was heated to 85°C for 1 hour, after which time it was poured into sat. aq. NH 4 Cl (100mL) and extracted with DCM (2x50mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[3-[2-(trifluoromethyl)phenyl]-1H-pyrazol-5-yl]chroman- 6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one (112mg, 0.22mmol, 67% yield) as a white solid. UPLC- MS (ES+, final purity): 3.94 min, m/z 507.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 13.05 (0.6H, s), 13.00 (0.4H, s), 10.46 (1H, s), 7.95 (1H, d, J = 5.6Hz), 7.90-7.54 (4H, br m), 7.01-6.98 (1H, m), 6.95-6.86 (2H, m), 6.39-6.33 (1H, m), 6.28-6.23 (1H, m), 4.50-4.41 (1H, m), 4.19-4.04 (1H, m), 3.52-3.42 (1H, m), 3.21-3.01 (2H, m), 2.93 (2H, t, J = 7.6Hz), 2.54 (2H, t, J = 7.6Hz). [0426] The compounds in the table below were made in an analogous manner to Example 23, using the appropriate arylacetylene in place of 2-ethynyl-α,α,α-trifluorotoluene in step 2: [0427] Example 24. Synthesis of 5-[3-[6-(trifluoromethyl)-1H-benzimidazol-2-yl]chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 22) [0428] 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (246mg, 0.65mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H-1,8- naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (200mg, 0.59mmol), 3,4- diaminobenzotrifluoride (114mg, 0.65mmol), DIPEA (0.31mL, 1.76mmol) and DMF (3mL) at rt under a nitrogen atmosphere and stirred for 1 hour. Solvent was removed in vacuo and the residue partitioned between water (50mL) and DCM (50mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was then stirred at 80°C in acetic acid (3mL) for 18 hours, after which time the reaction was cooled to rt and solvent removed in vacuo. The residue was partitioned between sat. aq. NaHCO 3 (100mL) and DCM (100mL) and the organic layer separated, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by column chromatography using an eluent of 0-5% MeOH in DCM to give 5-[3-[6-(trifluoromethyl)-1H-benzimidazol-2-yl]chroman-6-yl] oxy-3,4-dihydro- 1H-1,8-naphthyridin-2-one (75mg, 0.16mmol, 26% yield) as a white solid. UPLC-MS (ES+, final purity): 3.41 min, m/z 481.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.92 (1H, br s), 10.48 (1H, s), 7.95 (1H, d, J = 9.2Hz), 7.89-7.76 (1H, m), 7.76-7.70 (1H, m), 7.50 (1H, d, J = 8.8Hz), 7.06-7.03 (1H, m), 6.93 (1H, dd, J = 9.2Hz, 2.8Hz), 6.89 (1H, d, 8.8Hz), 6.27 (1H, d, J = 6.0Hz), 4.65 (1H, m), 4.33 (1H, t, J = 9.2Hz), 3.71-3.62 (1H, m), 3.38-3.22 (2H, m), 2.93 (2H, t, J = 8.4Hz), 2.53 (2H, t, J = 8.4Hz, (partly under DMSO)). [0429] The compounds in the table below were made in an analogous manner, using the appropriate diamine (either commercially available or synthesised) in place of 3,4- diaminobenzotrifluoride: Comp. Structure and Name Data No [0430] Example 25. Synthesis of 5-[3-(3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4- dihydro-1H-1,8-naphthyridin-2-one (Compound 134)

[0431] Step 1 – N-(4-amino-3-pyridyl)-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyri din-4- yl)oxy]chromane-3-carboxamide [0432] 2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) (307.2mg, 0.81mmol) was added to a stirred solution of 6-[(7-oxo-6,8-dihydro-5H- 1,8-naphthyridin-4-yl)oxy]chromane-3-carboxylic acid (250mg, 0.73mmol), DIPEA (0.38mL, 2.2mmol), 3,4-diaminopyridine (88.2mg, 0.81mmol) and DMF (10mL) under a nitrogen atmosphere and stirred at rt for 1h. Solvent was removed in vacuo and the residue partitioned between water (20mL) and DCM+few drops of MeOH (20mL), followed by extraction of the aqueous phase with EtOAc (20mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The crude was therefore purified by column chromatography using as eluent a gradient 0-5% MeOH in DCM to give N-(4-amino-3- pyridyl)-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy] chromane-3-carboxamide (195mg, 0.45mmol, 62% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.13 min, m/z 432.2 [M+H] + . [0433] Step 2 – 5-[3-(3H-imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dih ydro-1H-1,8- naphthyridin-2-one [0434] To N-(4-amino-3-pyridyl)-6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyri din-4- yl)oxy]chromane-3-carboxamide (144mg, 0.33mmol) in 1,4-dioxane (3mL) was added HCl (4M in dioxane - 0.67mL, 2.68mmol) in a sealed vial. The reaction was irradiated at 100°C for 2 hours, after which time the solvent was removed under reduce pressure. The reaction was quenched with sat. aq. NaHCO 3 and extracted with DCM+MeOH. The organic phase was dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The crude was purified by automated column chromatography using as eluent a gradient 0-12% MeOH in DCM to give 5-[3-(3H- imidazo[4,5-c]pyridin-2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1 ,8-naphthyridin-2-one (42.5mg, 0.10mmol, 31% yield) as a white solid. UPLC-MS (ES+, final purity): 2.30 min, m/z 414.2 [M+H] + . 1 H-NMR (400MHz, DMSO-d 6 ) δ/ppm: 13.00-12.79 (1H, m), 10.47 (1H, s), 8.87 (1H, s), 8.28 (1H, d, J = 5.2Hz), 7.96 (1H, d, J = 5.6Hz), 7.61-7.50 (1H, m), 7.03 (1H, d, J = 2.4Hz), 6.95-6.87 (2H, m), 6.27 (1H, d, J = 5.6Hz), 4.64-4.58 (1H, m), 4.31 (1H, t, J = 10.0Hz), 3.70-3.60 (1H, m), 3.36-3.19 (2H, m, (partly under water)), 2.93 (2H, t, J = 7.6Hz), 2.53 (2H, t, 8.0Hz, (partly under DMSO)). [0435] Example 26. Synthesis of 5-[[2-(3-phenyl-1H-1,2,4-triazol-5-yl)-3,4-dihydro-1H- isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one hydrochloride (Compound 44) [0436] Step 1 - 2-(5-bromo-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-di hydro-1H- isoquinolin-7-ol [0437] 3,5-dibromo-1-tetrahydropyran-2-yl-1,2,4-triazole (1.042g, 3.35mmol), 1,2,3,4- tetrahydroisoquinolin-7-ol (500mg, 3.35mmol), L-proline (38.6mg, 0.34mmol), copper(I) iodide (63.8mg, 0.34mmol), K 2 CO 3 (1.39g, 10.05mmol) and DMF (3mL) were combined in a sealed and heated to 120°C for 18 hours. The reaction was allowed to cool to rt, solvent removed in vacuo and the residue partitioned between water (100mL) and DCM (100mL). The organic layer was separated, dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 2-(5-bromo-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-di hydro- 1H-isoquinolin-7-ol (508mg, 1.34mmol, 40% yield) as a brown solid. UPLC-MS (ES+, short acidic): 1.64 min, m/z 379.1, 381.1 [M+H] + . [0438] Step 2 – 2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-d ihydro-1H- isoquinolin-7-ol [0439] [1,1'-Bis(diphenylphosphino)ferrocene]Palladium(II) chloride dichloromethane complex (173.6mg, 0.21mmol) was added to a stirred mixture of 2-(5-bromo-2- tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoq uinolin-7-ol (806mg, 2.13mmol), phenylboronic acid (311mg, 2.55mmol), K 2 CO 3 (881mg, 6.38mmol), 1,4-dioxane (9mL) and water (1mL) at rt under an inert atmosphere. The reaction was heated to 80°C for 3 hours, after which time it was cooled to rt and solvent removed in vacuo. The residue was suspended in DCM (10mL), filtered over celite, which was washed with DCM (10mL). The residue was concentrated in vacuo and purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4- dihydro-1H-isoquinolin-7-ol (510mg, 1.35mmol, 64% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.85min, m/z 399.3 [M+Na] + , 293.2 [M-THP] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 9.23 (1H, s), 7.98-7.93 (2H, m), 7.48-7.37 (3H, m), 6.98 (1H, d, 8.4Hz), 6.61 (1H, dd, J = 8.4Hz, 2.8Hz), 6.57 (1H, d, J = 2.8Hz), 5.31 (1H, dd, J = 10.0Hz, 2.0Hz), 4.50-4.46 (1H, m), 4.35-4.31 (1H, m), 4.10-4.03 (1H, m), 3.72-3.64 (2H, m), 3.45-3.35 (1H, m), 3.10-3.01 (1H, m), 2.85-2.76 (1H, m), 2.06-1.95 (2H, m), 1.90-1.53 (4H, m). [0440] Step 3 – 5-[[2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3 ,4-dihydro-1H- isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one [0441] 2-(5-Phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-d ihydro-1H-isoquinolin-7- ol (510mg, 1.35mmol), 5-fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (225.1mg, 1.35mmol), K 2 CO 3 (936mg, 6.77mmol) and DMSO (10mL) were combined in a sealed vial and stirred at 110°C for 18 hours. The reaction was cooled to rt and the solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 5-[[2-(5-phenyl-2-tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3 ,4- dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyr idin-2-one (192mg, 0.37mmol, 27% yield) as a yellow solid. UPLC-MS (ES+, short acidic): 1.92 min, m/z 523.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.56 (1H, s), 8.00-7.93 (2H, m), 7.48-7.39 (3H, m), 7.31-7.28 (1H, m), 7.03 (1H, d, J = 2.8Hz), 6.99 (1H, dd, J = 8.4Hz, 2.8Hz), 6.34 (1H, d, = 5.6Hz), 5.34 (1H, dd, J = 10.0Hz, 2.4Hz), 4.56 (1H, d, J = 16.0Hz), 4.44 (1H, d, J = 16.0Hz), 4.08-4.00 (1H, m), 3.81-3.65 (2H, m), 3.50-3.42 (1H, m), 3.25-3.14 (1H, m), 3.00- 2.88 (3H, m), 2.55-2.50 (3H, m), 1.90-1.54 (6H, m). [0442] Step 4 – 5-[[2-(3-phenyl-1H-1,2,4-triazol-5-yl)-3,4-dihydro-1H-isoqui nolin-7-yl]oxy]- 3,4-dihydro-1H-1,8-naphthyridin-2-one hydrochloride [0443] HCl (4M in 1,4-dioxane - 0.37mL, 1.47mmol) was added to 5-[[2-(5-phenyl-2- tetrahydropyran-2-yl-1,2,4-triazol-3-yl)-3,4-dihydro-1H-isoq uinolin-7-yl]oxy]-3,4-dihydro- 1H-1,8-naphthyridin-2-one (192.mg, 0.37mmol) in DCM (10mL) at rt under a nitrogen atmosphere and the reaction was stirred for 18 hours. Solvent was removed in vacuo and the residue purified by column chromatography using an eluent of 0-10% MeOH in DCM to give 5-[[2-(3-phenyl-1H-1,2,4-triazol-5-yl)-3,4-dihydro-1H-isoqui nolin-7-yl]oxy]-3,4-dihydro- 1H-1,8-naphthyridin-2-one hydrochloride (50mg, 0.11mmol, 29% yield) as a yellow solid. UPLC-MS (ES+, final purity): 3.39 min, m/z 439.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.63 (1H, s), 8.04-7.95 (3H, m), 7.52-7.44 (3H, m), 7.30 (1H, d, J = 7.6Hz), 7.05-6.99 (2H, m), 6.39 (1H, d, J = 6.0Hz), 4.72-4.67 (2H, m), 3.83-3.77 (2H, m), 2.99-2.91 (3H, m), 2.59-2.54 (3H, m). Broad peak at 5.4ppm will include 1 exchangeable and HCl salt. [0444] Example 27. Synthesis of 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-N- phenyl-3,4-dihydro-1H-isoquinoline-2-carboxamide (Compound 46) [0445] Step 1 - benzyl 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-3,4-dihy dro- 1H-isoquinoline-2-carboxylate [0446] Benzyl 7-hydroxy-3,4-dihydro-1H-isoquinoline-2-carboxylate (500mg, 1.76mmol), 5- fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (0.29g, 1.76mmol) and K 2 CO 3 (0.91g, 6.55mmol) were suspended in DMSO (50mL) and heated to 110°C 18 hours. The reaction was cooled to rt, carefully poured an aq. solution of citric acid (1.36g, 7.06mmol in 50mL of H 2 O), which caused fizzing, and left stirring for 1 hour. Water (200mL) was added and the resulting mixture extracted with DCM (2x100mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-75% EtOAc in petroleum ether to give benzyl 7-[(7-oxo- 6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-3,4-dihydro-1H-iso quinoline-2-carboxylate (510mg, 1.19mmol, 67% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.69 min, m/z 430.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.50 (1H, s), 7.97 (1H, d, J = 5.6Hz), 7.41-7.35 (5H, m), 7.25 (1H, d, J = 8.4Hz), 7.03 (1H, d, J = 6.8Hz), 6.96 (1H, dd, J = 8.0Hz, 2.4Hz), 6.31 (1H, d, J = 5.5Hz), 5.13 (2H, s), 4.66-4.55 (2H, m), 3.70-3.62 (2H, m), 2.90 (2H, t, J = 7.2Hz), 2.82 (2H, J = 5.6Hz), 2.55-2.53 (2H, m (partly under DMSO)). [0447] Step 2 - 5-(1,2,3,4-tetrahydroisoquinolin-7-yloxy)-3,4-dihydro-1H-1,8 -naphthyridin- 2-one [0448] Palladium, 10 wt. % on carbon powder, dry (50mg) was added to a stirred solution of benzyl 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-3,4-dihy dro-1H-isoquinoline-2- carboxylate (510.mg, 1.19mmol) in EtOAc (20mL) and MeOH (5mL) at rt under a nitrogen atmosphere. The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 24 hours. The crude was filtered over celite, which was washed with MeOH (10mL) and the filtrate concentrated in vacuo to give 5-(1,2,3,4- tetrahydroisoquinolin-7-yloxy)-3,4-dihydro-1H-1,8-naphthyrid in-2-one (222mg, 0.75mmol, 63% yield) as a yellow solid. The product was used in the next step without further purification. UPLC-MS (ES+, short acidic): 0.91 min, m/z 296.2 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 8.17 (1H, s), 7.99 (1H, d, J = 6.0Hz), 7.15 (1H, d, J = 8.4Hz), 6.86 (1H, dd, J = 8.4Hz, 2.8Hz), 6.76 (1H, d, J = 2.8Hz), 6.34 (1H, d, J = 6.0Hz), 4.03 (2H, s), 3.18 (2H, t, J = 6.0Hz), 3.08 (2H, t, J = 8.0Hz), 2.83 (2H, t, J = 6.0Hz), 2.72 (2H, t, J = 8.0Hz). Exchangeable proton not seen. [0449] Step 3 – 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-N-phenyl -3,4-dihydro- 1H-isoquinoline-2-carboxamide [0450] Phenyl isocyanate (0.02mL, 0.19mmol) was added to a stirred solution of 5-(1,2,3,4- tetrahydroisoquinolin-7-yloxy)-3,4-dihydro-1H-1,8-naphthyrid in-2-one (50mg, 0.17mmol) and DIPEA (0.06mL, 0.34mmol) in DCM (5mL) at rt under a nitrogen atmosphere. The reaction stirred for 1 hour, after which time it was diluted with water (20mL) and DCM (15mL). The organic layer was separated, passed through a phase separator and solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0-100% EtOAc in petroleum ether to give 7-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]-N-phenyl - 3,4-dihydro-1H-isoquinoline-2-carboxamide (29mg, 0.07mmol, 41% yield) as a white solid. UPLC-MS (ES+, final purity): 3.35 min, m/z 415.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.51 (1H, s), 8.58 (1H, s), 7.98 (1H, d, J = 5.6Hz), 7.50-7.46 (2H, m), 7.30-7.22 (3H, m), 7.00-6.92 (3H, m), 6.34 (1H, d, J = 5.6Hz), 4.64 (2H, s), 3.73 (2H, t, J = 6.0Hz), 2.94-2.85 (4H, m), 2.55-2.53 (2H, m, (partly under DMSO peak)). [0451] The compounds in the table below were made in an analogous manner, using the appropriate aryl isocyanate in place of phenyl isocyanate in step 3:

[0452] Example 28. Synthesis of 5-[[2-(1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin- 7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 159) [0453] Step 1 - 2-(1H-benzimidazol-2-yl)-7-benzyloxy-3,4-dihydro-1H-isoquino line [0454] 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline (158mg, 0.66mmol), 2- chlorobenzimidazole (101mg, 0.66mmol) and 1,4-dioxane (1.2mL) were mixed in a sealed vial and irradiated to 180°C for 1 hour. The residue was dissolved in DCM, washed with sat. aq. NaHCO 3 and then dried through a phase separator. The solvent was removed in vacuo and the residue was purified by column chromatography using as eluent a gradient 0-50% EtOAc in petroleum ether to give 2-(1H-benzimidazol-2-yl)-7-benzyloxy-3,4-dihydro-1H-isoquino line (175mg, 0.49mmol, 75% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.50 min, m/z 356.5 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 11.45 (1H, s), 7.49-7.27 (5H, m), 7.20 (2H, dd, J = 14.1Hz, 7.6Hz), 7.09 (1H, d, J = 8.3Hz), 6.99-6.81 (4H, m), 5.10 (2H, s), 4.68 (2H, s), 3.78 (2H, t, J = 5.9Hz), 2.84 (2H, t, J = 5.9Hz). [0455] Step 2 - 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)benzimida zol-1- yl]methoxy]ethyl-trimethyl-silane [0456] Sodium hydride (60% dispersed in mineral oil - 38.3mg, 0.96mmol) was added to a stirred solution of 2-(1H-benzimidazol-2-yl)-7-benzyloxy-3,4-dihydro-1H-isoquino line (170mg, 0.48mmol) and DMF (100mL) at rt under a nitrogen atmosphere. After 20 minutes, 2-(trimethylsilyl)ethoxymethyl chloride (0.13mL, 0.72mmol) was added and the mixture was stirred for 18 hours. The reaction was quenched with water (1mL) and solvent removed in vacuo. Water (20mL) was added followed by extraction with DCM (2x20mL). The combined organic layers were dried over Na 2 SO 4 , filtered and solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-40% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)benzimida zol-1- yl]methoxy]ethyl-trimethyl-silane (200mg, 0.41mmol, 86% yield) as a colourless gum. UPLC- MS (ES+, Short acidic): 1.93 min, m/z 486.9 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 7.64-7.55 (1H, m), 7.45-7.28 (6H, m), 7.24-7.15 (2H, m), 7.10 (1H, d, J = 8.4Hz), 6.85 (1H, dd, J = 8.4Hz, 2.7Hz), 6.76 (1H, d, J = 2.6 Hz), 5.34 (2H, s), 5.05 (2H, s), 4.60 (2H, s), 3.78- 3.66 (4H, m), 3.02 (2H, t, J = 5.9 Hz), 1.01-0.96 (2H, m), 0.00 (9H, s). [0457] Step 3 – 2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-di hydro-1H- isoquinolin-7-ol [0458] 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)benzimida zol-1- yl]methoxy]ethyl-trimethyl-silane (195mg, 0.40mmol) was dissolved in MeOH (4mL) and Pd(OH)2 (56mg, 0.40mmol) added under inert atmosphere. The reaction was fitted with a H 2 balloon and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere overnight. The crude was filtered over celite, flushed with MeOH and the filtrate concentrated in vacuo. The residue was purified by column chromatography using as eluent a gradient 0- 50% EtOAc in petroleum ether to give 2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2- yl]-3,4-dihydro-1H-isoquinolin-7-ol (120mg, 0.30mmol, 76% yield) as a colourless gum which crystallised on standing. UPLC-MS (ES+, short acidic): 1.58min, m/z 396.6 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 8.45 (1H, s), 7.61-7.55 (1H, m), 7.33-7.28 (1H, m), 7.21-7.14 (2H, m), 6.93 (1H, d, J = 8.3Hz), 6.68 (1H, dd, J = 8.3Hz, 2.6Hz), 6.63 (1H, d, J = 2.5Hz), 5.35 (2H, s), 4.57 (2H, s), 3.81-3.69 (4H, m), 2.92 (2H, t, J = 5.8 Hz), 1.03-0.96 (2H, m), 0.00 (9H, s). [0459] Step 4 – 5-[[2-[1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3, 4-dihydro-1H- isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one [0460] Potassium tert-butoxide (37.4mg, 0.33mmol) was added to a stirred solution of 5- fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (50.4mg, 0.30mmol) and 2-[1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-ol (120mg, 0.30mmol) in DMF (2mL) and the reaction was heated at 80°C for 18 hours. The reaction was cooled to rt and the solvent removed in vacuo. The residue was partitioned between EtOAc and water; the organics were washed with saturated brine, dried over a phase separator and the solvent removed in vacuo. The residue was purified by column chromatography eluting with 25-100% EtOAc in petroleum ether to give 5-[[2-[1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-yl]oxy]-3,4- dihydro-1H-1,8-naphthyridin-2-one (94mg, 0.17mmol, 57% yield) as a colourless gum. UPLC- MS (ES+, Short acidic): 1.65 min, m/z 542.5 [M+H] + . 1 H NMR (400MHz, CDCl 3 ) δ/ppm: 8.69 (1H, s), 8.01 (1H, d, J = 5.9Hz), 7.63-7.58 (1H, m), 7.34-7.30 (1H, m), 7.25-7.16 (3H, m), 6.93- 6.85 (2H, m), 6.35 (1H, d, J = 5.9Hz), 5.36 (2H, s), 4.65 (2H, s), 3.82-3.72 (4H, m), 3.13-3.02 (4H, m), 2.70 (2H, dd, J = 8.4Hz, 7.0 Hz), 1.03-0.96 (2H, m), 0.00 (9H, s). [0461] Step 5 – 5-[[2-(1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl ]oxy]-3,4- dihydro-1H-1,8-naphthyridin-2-one [0462] Trifluoroacetic acid (0.64mL, 8.31mmol) was added to a stirred solution of 5-[[2-[1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-yl]oxy]-3,4- dihydro-1H-1,8-naphthyridin-2-one (90mg, 0.17mmol) in DCM (2mL) in a sealed vial and heated to 40°C for 18 hours, after which time the solvent was removed in vacuo. The residue was loaded into an SCX-2 column and flushed at first with MeOH (2x10mL) and then NH 3 in MeOH (10mL) to elute the product. The reside was then purified by column chromatography using as eluent a gradient 1-8% MeOH in DCM to give a colourless gum, which was triturated in MeCN/Et 2 O to give a white solid. The solid was filtered, washed with Et 2 O and dried to give 5-[[2-(1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl ]oxy]-3,4-dihydro-1H-1,8- naphthyridin-2-one (29mg, 0.070mmol, 42% yield) as a white solid. UPLC-MS (ES+, final purity): 2.52 min, m/z 412.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 12.21 (1H, s), 10.52 (1H, s), 7.99 (1H, d, J = 5.8 Hz), 7.33-7.253H, m), 7.07-6.97 (4H, m), 6.35 (1H, d, J = 5.8 Hz), 4.76 (2H, s), 3.85 (2H, t, J = 5.9 Hz), 2.98 (2H, t, J = 5.9Hz), 2.92 (2H, t, J = 7.9Hz), 2.57-2.52 (2H, t, J = 7.9Hz, (partly under DMSO)). [0463] The compounds in the table below were made in an analogous manner, using the appropriate heteroaryl chloride in place of 2-chlorobenzimidazole in step 1:

[0464] Example 29. Synthesis of 5-[[2-(5-phenyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H- isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 162) [0465] Step 1 - 5-bromo-1,3-dihydrobenzimidazol-2-one [0466] 1,1'-Carbonyldiimidazole (1.73g, 10.69mmol) was added to a stirred solution of 4- bromo-1,2-diaminobenzene (1g, 5.35mmol) in DMF (10mL) at 25°C under a nitrogen atmosphere and the reaction was stirred for 18 hours. The solvent was reduced in vacuo and the residue was diluted with water and EtOAc, causing a solid to crash out. The solid was filtered, slurried in water, sonicated, filtered, washed with further water and dried to give 5- bromo-1,3-dihydrobenzimidazol-2-one (912mg, 4.28mmol, 80% yield) as a brown solid. UPLC-MS (ES+, Short acidic): 1.28 min, m/z 212.9, 214.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 10.79-10.73 (2H, m), 7.08 (1H, dd, J = 8.2Hz, 2.0Hz), 7.05 (1H, d, J = 1.9Hz), 6.87 (1H, d, J = 8.2Hz). [0467] Step 2 - 6-bromo-2-chloro-1H-benzimidazole [0468] 5-bromo-1,3-dihydrobenzimidazol-2-one (912mg, 4.28mmol) was dissolved in POCl 3 (7.98mL, 85.62mmol) and the reaction was heated to 95°C for 4 hours, after which time the reaction was reduced in vacuo and the residue was azeotroped with toluene. The residue was quenched with saturated NaHCO 3 solution causing a solid to crash out, which was sonicated, filtered, washed with water and dried to give 6-bromo-2-chloro-1H-benzimidazole (855mg, 3.69mmol, 86% yield) as a pale brown solid. The compound was used without further purification in the following step. UPLC-MS (ES+, Short acidic): 1.44 min, m/z 230.8, 232.8, 234.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 7.74 (1H, d, J = 1.9Hz), 7.49 (1H, d, J = 8.6Hz), 7.38 (1H, dd, J = 8.6Hz, 2.0Hz). Exchangeable proton missing. [0469] Step 3 - 7-benzyloxy-2-(5-bromo-1H-benzimidazol-2-yl)-3,4-dihydro-1H- isoquinoline [0470] Eight separate microwave vials each with 7-benzyloxy-1,2,3,4-tetrahydroisoquinoline hydrochloride (50 mg, 0.18mmol) and 6-bromo-2-chloro-1H-benzimidazole (46mg, 0.2mmol), and 1,4-Dioxane (2.5mL) were sealed and heated to 180 °C in the microwave for 60 min. Each vial precipitated a solid, all the reaction mixtures were filtered and the combined solid washed with further dioxane and dried to give 7-benzyloxy-2-(5-bromo-1H-benzimidazol-2-yl)-3,4- dihydro-1H-isoquinoline (626 mg, 1.4mmol, 99% yield) as a green/brown solid. UPLC-MS (ES+, Short acidic): 1.59 min, m/z 434.1;436.0 [M+H]+ (67%) [0471] Step 4 - 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo- benzimidazol- 1-yl]methoxy]ethyl-trimethyl-silane [0472] Sodium hydride (60% dispersed in mineral oil - 168.9mg, 4.22mmol) was added to a stirred solution of 7-benzyloxy-2-(5-bromo-1H-benzimidazol-2-yl)-3,4-dihydro-1H- isoquinoline (692mg, 1.12mmol) and DMF (10mL) at room temperature under a nitrogen atmosphere. After 20 minutes 2-(trimethylsilyl)ethoxymethyl chloride (0.3mL, 1.67mmol) was added and the reaction was allowed to stir for 2 hours. The reaction was quenched with water (1mL) and solvent removed in vacuo. The residue was then partitioned between water (20mL) and EtOAc (20mL). The organic layer was separated and the aqueous extracted with DCM (20mL). The combined organic layers were dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using an eluent of 0- 40% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)- 5-bromo-benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (442mg, 0.78mmol, 70% yield) as a pale yellow gum. UPLC-MS (ES+, Short acidic): 2.27 and 2.29 min, m/z 564.3, 566.2 [M+H] + . Presence of two SEM isomers. [0473] Step 5 (Method A) - 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-phenyl - benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane [0474] Potassium carbonate (151.3mg, 1.09mmol), 2-[[2-(7-benzyloxy-3,4-dihydro-1H- isoquinolin-2-yl)-5-bromo-benzimidazol-1-yl]methoxy]ethyl-tr imethyl-silane (206mg, 0.36mmol), phenylboronic acid (48.9mg, 0.40mmol), 1,4-dioxane (4mL) and water (1mL) were combined and stirred at room temperature under a nitrogen atmosphere, followed by addition of [1,1'-bis(diphenylphosphino)ferrocene]palladium(II) chloride dichloromethane complex (29.8mg, 0.04mmol). The reaction was heated to 90°C for 18 hours, after which time the reaction was cooled to room temperature and the solvent removed under reduce pressure. The residue was purified by column chromatography using an eluent of 0-35% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-phenyl - benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (150mg, 0.27mmol, 73% yield) as a pale yellow oil. UPLC-MS (ES+, short acidic): 2.11 min, m/z 562.8 [M+H]+. [0475] Step 6 - 2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-y l]-3,4-dihydro- 1H-isoquinolin-7-ol [0476] 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-phenyl -benzimidazol-1- yl]methoxy]ethyl-trimethyl-silane (150mg, 0.27mmol) was dissolved in MeOH (4mL) and palladium hydroxide (3.04mg, 0.02mmol) was added under an atmosphere of nitrogen. The reaction was fitted with a H 2 balloon, subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere overnight. The crude was filtered over celite, washed with MeOH and the solvent removed in vacuo to give 2-[5-phenyl-1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-ol (117mg, 0.25mmol, 93% yield) as a colourless gum. UPLC-MS (ES+, short acidic):1.81 min, m/z 472.6 [M+H] + . [0477] Step 7 - 5-[[2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol -2-yl]-3,4- dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyr idin-2-one ol [0478] Potassium tert-butoxide (83.5mg, 0.74mmol) was added to a stirred solution of 5- fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (41.2mg, 0.25mmol) in DMF (2mL) in a sealable vial. The vial was sealed and and heated at 80°C for 18 hours. The reaction was cooled to room temperature and the solvent reduced in vacuo. The crude was partitioned between EtOAc and water. The organic phase was separated, washed with saturated brine, dried over a phase separator and the solvent removed under reduce pressure. The residue was purified by column chromatography using as eluent a gradient 25-100% EtOAc in petroleum ether to give 5-[[2-[5-phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol -2-yl]-3,4-dihydro-1H- isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (90mg, 0.15mmol, 59% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.87 min, m/z 618.5 [M+H] + . [0479] Step 8 – 5-[[2-(5-phenyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquin olin-7- yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one [0480] Trifluoroacetic acid (0.56mL, 7.29mmol) was added to a stirred solution of 5-[[2-[5- phenyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3, 4-dihydro-1H-isoquinolin-7- yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (90mg, 0.15mmol) in DCM (2mL) in a sealable vial. The vial was sealed and and heated to 40°C for 18 hours. The reaction was reduced in vacuo and loaded onto an SCX-2 column, which was flushed at first with methanol (2x), then NH 3 in MeOH followed by 20% DCM in MeOH/NH 3 to elute the product. The crude was purified by column chromatography using as eluent a gradient 1-8% MeOH in DCM to give a colourless gum, which was triturated in MeCN/Et 2 O to give a white solid. The solid was filtered, washed with Et 2 O, followed by sonication in MeOH to precipitate a white solid. The whole suspension was evaporated and dried to give 5-[[2-(5-phenyl-1H-benzimidazol-2-yl)- 3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naph thyridin-2-one (27mg, 0.053mmol, 36% yield) as an off white solid. UPLC-MS (ES+, final purity): 3.04 min, m/z 488.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ/ppm: 11.57 (1H, s), 10.51 (1H, s), 7.99 (1H, dd, J = 5.7Hz, 0.7Hz), 7.66-7.59 (2H, m), 7.52-7.38 (3H, m), 7.33-7.18 (3H, m), 7.05 (1H, d, J = 2.5Hz), 6.98 (1H, dd, J = 8.3Hz, 2.6Hz), 6.35 (1H, d, J = 5.8Hz), 4.76 (2H, s), 3.85 (2H, t, J = 5.9Hz), 3.00-2.89 (4H, m), 2.58-2.52 (3H, m, (partly under DMSO)). [0481] As alternative, Step 5 could have been performed using Method B: [0482] 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo- benzimidazol-1- yl]methoxy]ethyl-trimethyl-silane (255mg, 0.45mmol), potassium phosphate tribasic (0.29g, 1.35mmol) and 1,3-dimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) -1H-pyrazole (150.5mg, 0.68mmol) were suspended in toluene (2mL) and water (1mL) and degassed under a nitrogen atmosphere in a sealable vial. Tricyclohexylphosphine (0.03g, 0.09mmol) and palladium (II) acetate (0.01g, 0.03mmol) were then added followed by further degassing, the vial was sealed and heated at 90°C for 18 hours. The reaction was cooled and the water removed. The organics were reduced in vacuo onto silica and the product was purified by silica column chromatography using as eluent a gradient 0-80% EtOAc in petroleum ether to give 2- [[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-(2,5-dim ethylpyrazol-3- yl)benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (110mg, 0.19mmol, 42% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 2.02 and 2.05 min, m/z 580.5 [M+H] + (2 SEM- protected isomers). [0483] The compounds in the table below were made in an analogous manner using the appropriate boronic acid/ester and applying the appropriate method: [0484] Example 30. Synthesis of 5-[[2-(5-propyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H- isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 161) [0485] Step 1 - 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-cyclop ropyl- benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane [0486] 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-bromo- benzimidazol-1- yl]methoxy]ethyl-trimethyl-silane (205mg, 0.36mmol), cyclopropylboronic acid (0.08g, 0.91mmol) and potassium phosphate tribasic (0.23g, 1.09mmol) were suspended in toluene (2mL) and water (1mL) at room temperature and degassed under nitrogen atmosphere in a sealable vial. Tricyclohexylphosphine (0.02g, 0.07mmol) and palladium (II) acetate (0.01g, 0.03mmol) were then added, the vial sealed and heated 100°C for 2 hours. The reaction was cooled and the water removed. The organics were reduced in vacuo onto silica and the product was purified by column chromatography using as eluent a gradient 0-35% EtOAc in petroleum ether to give 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-cyclop ropyl- benzimidazol-1-yl]methoxy]ethyl-trimethyl-silane (155mg, 0.29mmol, 81% yield) as a colourless gum. UPLC-MS (ES+, Short acidic): 1.98 min, m/z 526.9 [M+H] + . [0487] Step 2 - 2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-y l]-3,4-dihydro- 1H-isoquinolin-7-ol [0488] 2-[[2-(7-benzyloxy-3,4-dihydro-1H-isoquinolin-2-yl)-5-cyclop ropyl-benzimidazol-1- yl]methoxy]ethyl-trimethyl-silane (155mg, 0.29mmol) was dissolved in methanol (4mL) and palladium hydroxide (3.36mg, 0.02mmol) was added under an atmosphere of nitrogen. The reaction was fitted with a H 2 balloon, subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere overnight. The crude was filtered over celite, washed with MeOH and the solvent removed in vacuo to give 2-[5-propyl-1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-ol (107mg, 0.24mmol, 83% yield) as a colourless gum. UPLC-MS (ES+, short acidic): 1.78 min, m/z 438.6 [M+H] + . [0489] Step 3 - 5-[[2-[5-propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol -2-yl]-3,4- dihydro-1H-isoquinolin-7-yl]oxy]-3,4-dihydro-1H-1,8-naphthyr idin-2-one [0490] Potassium tert-butoxide (83.5mg, 0.74mmol) was added to a stirred solution of 5- fluoro-3,4-dihydro-1H-1,8-naphthyridin-2-one (40.6mg, 0.24mmol) and 2-[5-propyl-1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-ol (107mg, 0.24mmol) in DMF (2mL) in a sealable vial, which was sealed and heated at 100°C for 72 hours. Reaction was cooled to room temperature and the solvent reduced in vacuo. The crude was partitioned between EtOAc and water. The organic layer was separated, washed with saturated brine, dried over a phase separator and the solvent removed under reduce pressure. The residue was purified by column chromatography using as eluent a gradient 25-100% EtOAc in petroleum ether to give 5-[[2-[5-propyl-1-(2- trimethylsilylethoxymethyl)benzimidazol-2-yl]-3,4-dihydro-1H -isoquinolin-7-yl]oxy]-3,4- dihydro-1H-1,8-naphthyridin-2-one (41mg, 0.07mmol, 29% yield) as a colourless gum. UPLC- MS (ES+, Short acidic): 1.81 min, m/z 584.5 [M+H] + . [0491] Step 3 - 5-[[2-(5-propyl-1H-benzimidazol-2-yl)-3,4-dihydro-1H-isoquin olin-7- yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one [0492] Trifluoroacetic acid (0.27mL, 3.51mmol) was added to a stirred solution of 5-[[2-[5- propyl-1-(2-trimethylsilylethoxymethyl)benzimidazol-2-yl]-3, 4-dihydro-1H-isoquinolin-7- yl]oxy]-3,4-dihydro-1H-1,8-naphthyridin-2-one (41.mg, 0.07mmol) in DCM (2mL) in a sealable vial. The reaction was sealed and heated to 40°C for 18 hours. The reaction was reduced in vacuo and loaded onto an SCX cartridge, which was flushed at first with MeOH and then MeOH/NH 3 followed by 20% DCM:MeOH/NH 3 to elute the product. The product was then purified by silica column chromatography using as eluent a gradient 1-8% MeOH/DCM to give a colourless gum. The compound was re-purified using reverse column chromatography using as eluent a gradient 0-30% (acetonitrile+0.1% formic acid) in (water+0.1% formic acid). Fractions containing the product were re-purified using preparative LCMS (early method). Fractions containing compound were loaded onto an SCX column which was flushed at first with MeOH and then MeOH/NH 3 to elute the product giving 5-[[2-(5-propyl-1H- benzimidazol-2-yl)-3,4-dihydro-1H-isoquinolin-7-yl]oxy]-3,4- dihydro-1H-1,8-naphthyridin- 2-one (8mg, 0.018mmol, 25% yield) as an off white solid. UPLC-MS (ES+, final purity): 3.03min, m/z 454.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 11.80 (1H, s), 10.50 (1H, s), 7.98 (1H, d, J = 5.8Hz), 7.27 (1H, d, J = 8.3Hz), 7.13 (1H, d, J = 8.0Hz), 7.07-6.94 (3H, m), 6.81 (1H, dd, J = 8.0Hz, 1.6Hz), 6.34 (1H, d, J = 5.8Hz), 4.72 (2H, s), 3.81 (2H, t, J = 5.9 Hz), 2.98-2.87 (4H, m), 2.62-2.51 (4H, m), 1.65-1.51 (2H, m), 0.88 (3H, t, J = 7.3Hz). [0493] Example 31. Synthesis of N-[4-[3-[4-(4-fluorophenyl)-1H-imidazol-2-yl]chroman-6- yl]oxy-2-pyridyl]cyclopropanecarboxamide (Compound 155) [0494] Step 1 – 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]-N-[2-(4-flu orophenyl)-2- oxo-ethyl]chromane-3-carboxamide [0495] To a solution of 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]chromane-3- carboxylic acid (65mg, 0.18mmol) and 2-amino-1-(4-fluorophenyl)ethanone hydrochloride (54.7mg, 0.20mmol) in THF (1.8mL) were added T3P (164µL, 0.28mmol) and DIPEA (99µL, 0.57mmol) and the mixture was heated at 65°C for 1 hour. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (25mL). The organic layer was washed with water (2x10mL) and brine (10mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo to afford 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]-N-[2- (4-fluorophenyl)-2-oxo-ethyl]chromane-3-carboxamide (86mg, 0.17mmol, 95% yield) as a yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.46 min, m/z 490.6 [M+H] + . [0496] Step 2 – N-[4-[3-[4-(4-fluorophenyl)-1H-imidazol-2-yl]chroman-6-yl]ox y-2- pyridyl]cyclopropanecarboxamide [0497] To a suspension of 6-[[2-(cyclopropanecarbonylamino)-4-pyridyl]oxy]-N-[2-(4- fluorophenyl)-2-oxo-ethyl]chromane-3-carboxamide (85.5mg, 0.17mmol) in n-BuOH (1mL) were added NH4OAc (134.6mg, 1.75mmol) and Et3N (26µL, 0.18mmol) and the mixture was irradiated at 150°C for 1 hour. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (25mL). The organic layer was washed with sat. aq. NaHCO 3 (10mL), water (2x10mL) and brine (10mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by flash chromatography using as eluent a gradient 0-10% MeOH in DCM, followed by purification by preparative LC-MS. Fractions containing the product were dried and loaded into an SCX-2 column, which was flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. The filtrate was concentrated to give N-[4-[3-[4-(4-fluorophenyl)-1H-imidazol-2-yl]chroman-6-yl]ox y-2- pyridyl]cyclopropanecarboxamide (10mg, 0.022mmol, 13% yield) as a white solid. UPLC-MS (ES+, final purity): 2.73 min, m/z 471.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.33 & 12.10 (0.2 & 0.8H, 2 s, mixture of tautomers), 10.80 (1H, s), 8.16 (1H, d, J = 5.7Hz), 7.80- 7.42 (2H, m), 7.65 (1H, d, J = 2.5Hz), 7.57 (1H, br s), 7.25-7.10 (2H, m), 7.02 (1H, d, J = 2.5Hz), 6.94-6.86 (2H, m*), 6.62 (1H, dd, J = 5.7Hz, 2.4Hz), 4.54-4.48 (1H, m), 4.16-4.08 (1H, m), 3.44-3.34 (1H, m), 3.28-3.18 (1H, m), 3.15-3.07 (1H, m), 1.97 (1H, quint, J = 6.2Hz), 0.77 (4H, d, J = 6.2Hz). [0498] Example 32. Synthesis of 4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-N-(2- pyridyl)pyridin-2-amine (Compound 140)

[0499] Step 1 – 6-[(2-chloro-4-pyridyl)oxy]-N-phenacyl-chromane-3-carboxamid e [0500] To a solution of 6-[(2-chloro-4-pyridyl)oxy]chromane-3-carboxylic acid (437mg, 1.43mmol) and 2-aminoacetophenone hydrochloride (270mg, 1.57mmol) in THF (14mL) were added T3P (1.28mL, 2.14mmol) and DIPEA (0.8mL, 4.58mmol) and the mixture was heated at 65°C for 1.5 hours. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (60mL). The organic layer was washed with water (2x20mL) and brine (20mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo to give 6-[(2- chloro-4-pyridyl)oxy]-N-phenacyl-chromane-3-carboxamide (604.5mg, 1.43mmol, 100% yield) as a light beige solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.72 min, m/z 423.3 [M+H] + . [0501] Step 2 - 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-py ridine [0502] To a suspension of 6-[(2-chloro-4-pyridyl)oxy]-N-phenacyl-chromane-3-carboxamid e (513mg, 1.21mmol) in n-BuOH (10mL) were added NH 4 OAc (935.2mg, 12.13mmol) and Et 3 N (169µL, 1.21mmol). The vial was sealed and the mixture irradiated at 150°C for 1 hr. After cooling to rt, the solvent was removed under reduce pressure and the residue dissolved in EtOAc (60mL). The organic layer was washed with sat. aq. NaHCO 3 (20mL), water (2x20mL) and brine (20mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-py ridine (314.9mg, 0.78mmol, 64% yield) as a beige solid. UPLC-MS (ES+, short acidic): 1.43 min, m/z 404.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.27 (1H, dd, J = 5.6, 0.3Hz), 7.77- 7.73 (2H, m), 7.69 (1H, br s), 7.43-7.36 (2H, m), 7.28-7.22 (1H, m), 7.10 (1H, d, J = 2.8Hz), 7.00 (1H, dd, J = 8.8, 2.8Hz), 6.96-6.92 (3H, m), 4.57-4.51 (1H, m), 4.25-4.18 (1H, m), 3.53 (1H, br s), 3.32-3.25 (2H, m), 3.18 (1H, dd, J = 16.6, 5.3Hz), 0.89-0.79 (1H, m). [0503] Step 3 – 4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-N-(2-pyridy l)pyridin-2- amine [0504] A solution of 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-py ridine (50mg, 0.12mmol), 2-aminopyridine (17.5mg, 0.19mmol), XPhos Pd G2 (9.8mg, 0.01mmol), XPhos (5.9mg, 0.01mmol) and K 2 CO 3 (51.3mg, 0.3700mmol) in tert-butanol under a nitrogen atmosphere (0.6mL) was irradiated at 130°C for 1 hour. After this time, additional 2- aminopyridine (17.5mg, 0.1900mmol), K 2 CO 3 (51.3mg, 0.37mmol), XPhos Pd G2 (9.8mg, 0.01mmol) and XPhos (5.9mg, 0.01mmol) were added and the mixture was irradiated at 160°C for 1 hour, after which time it was through a plug of celite and the filter cake washed with EtOAc. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM, followed by purification by preparative LC-MS. Fractions containing the product were dried and loaded into an SCX-2 column, which was flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. The filtrate was concentrated to give 4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-N-(2- pyridyl)pyridin-2-amine (11.5mg, 0.025mmol, 20% yield) as a white solid. UPLC-MS (ES+, final purity): 2.47 min, m/z 462.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.37 (1H, br s), 9.66 (1H, br s), 8.17-8.14 (1H, m), 8.08 (1H, d, J = 5.8Hz), 7.76-7.70 (2H, m), 7.69-7.60 (2H, m), 7.55 (1H, br s), 7.40-7.31 (3H, m), 7.21-7.15 (1H, m), 7.04 (1H, d, J = 2.7Hz), 6.94 (1H, dd, J = 8.8Hz, 2.7Hz), 6.90 (1H, d, J = 8.8Hz), 6.87-6.82 (1H, m), 6.41 (1H, dd, J = 5.8Hz, 2.1Hz), 4.56-4.49 (1H, m), 4.17-4.10 (1H, m), 3.45-3.36 (1H, m), 3.26 (1H, dd, J = 16.7Hz, 10.6Hz), 3.18-3.09 (1H, m). [0505] The compound in the table below was made in an analogous manner, using the appropriate amine in step 3: [0506] Example 33. Synthesis of 2,2-difluoro-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman- 6-yl]oxy-2-pyridyl]cyclopropanecarboxamide (Compound 137) [0507] Step 1 – 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imi dazol-1- yl]methoxy]ethyl-trimethyl-silane and - 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3- yl]-5-phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane [0508] To a solution of 2-chloro-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-py ridine (553mg, 1.37mmol) in anhydrous THF (13.7mL) at 0°C under a nitrogen atmosphere was added NaH (60% in mineral oil - 82.1mg, 2.05mmol) and the mixture was stirred at 0°C for 45 minutes.2-(trimethylsilyl)ethoxymethyl chloride (300µL, 1.7mmol) was added dropwise and the mixture was stirred for 2 hours at 0°C. The reaction mixture was quenched with water (15mL) and extracted with EtOAc (3x10mL). The combined organic layers were dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-50% EtOAc in petroleum ether to give 2-[[2-[6- [(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imidazol-1-y l]methoxy]ethyl-trimethyl- silane (444mg, 0.83mmol, 61% yield) and 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5- phenyl-imidazol-1-yl]methoxy]ethyl-trimethyl-silane (120.4mg, 0.23mmol, 16% yield), both as yellow gums. [0509] 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imi dazol-1- yl]methoxy]ethyl-trimethyl-silane: UPLC-MS (ES+, short acidic): 2.23 min, m/z 535.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.29-8.26 (1H, m), 7.77 (1H, s), 7.76-7.72 (2H, m), 7.37-7.32 (2H, m), 7.22-7.16 (1H, m), 7.08 (1H, d, J = 2.8Hz), 7.00 (1H, dd, J = 8.8Hz, 2.8Hz), 6.97-6.92 (3H, m), 5.48 (1H, d, J = 11.1Hz), 5.42 (1H, d, J = 11.1Hz), 4.50-4.44 (1H, m), 4.11-4.04 (1H, m), 3.61-3.52 (3H, m), 3.32-3.25 (1H, m), 3.08-2.99 (1H, m), 0.93-0.86 (2H, m), -0.02 (9H, s). [0510] 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5-phenyl-imi dazol-1- yl]methoxy]ethyl- trimethyl-silane: UPLC-MS (ES+, short acidic): 2.00 min, m/z 535.5 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.29-8.26 (1H, m), 7.51-7.38 (5H, m), 7.07 (1H, d, J = 2.8Hz), 7.02 (1H, s), 7.00 (1H, dd, J = 8.8Hz, 2.8Hz), 6.97-6.92 (3H, m), 5.39 (1H, d, J = 11.1Hz), 5.33 (1H, d, J = 11.1Hz), 4.51-4.45 (1H, m), 4.12-4.05 (1H, m), 3.65-3.55 (1H, m), 3.44-3.38 (2H, m), 3.30-3.20 (1H, m), 3.09-3.01 (1H, m), 0.88-0.77 (2H, m), -0.08 (9H, s). [0511] Step 2 – 1,1-diphenyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymeth yl)imidazol-2- yl]chroman-6-yl]oxy-2-pyridyl]methanimine [0512] To a solution of 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-4-phenyl-imi dazol- 1-yl]methoxy]ethyl-trimethyl-silane (444mg, 0.83mmol), benzophenone imine (210µL, 1.25mmol) and Cs 2 CO 3 (677.1mg, 2.08mmol) in dry 1,4-dioxane (4mL) under a nitrogen atmosphere were added (+/-)-BINAP (103.5mg, 0.17mmol) and tris(dibenzylideneacetone)dipalladium (0) (76.1mg, 0.08mmol) and the mixture was heated at 110°C for 18 hours. After cooling to rt, the mixture was filtered through celite and the filter cake washed with EtOAc. The solvent was removed in vacuo and the residue purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give 1,1-diphenyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymeth yl)imidazol-2-yl]chroman-6- yl]oxy-2-pyridyl]methanimine (564.3mg, 0.83mmol, 100% yield) as an orange solid. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 2.12 min, m/z 679.5 [M+H] + . [0513] Step 3 – acetic acid; 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2- yl]chroman-6-yl]oxypyridin-2-amine [0514] To a solution of 1,1-diphenyl-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]methanimine (564.3mg, 0.83mmol) in MeOH (8.3mL) were added NaOH (204.6mg, 2.49mmol) and hydroxylamine hydrochloride (127.1mg, 1.83mmol) and the mixture was stirred at rt for 1 hour. The solvent was removed in vacuo and the residue purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give acetic acid; 4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyr idin-2-amine (366.4mg, 0.64mmol, 77% yield) as an orange solid. UPLC-MS (ES+, short acidic): 1.70 min, m/z 515.4 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 7.79-7.72 (4H, m), 7.38-7.32 (2H, m), 7.22- 7.17 (1H, m), 7.00-6.97 (1H, m), 6.92-6.89 (2H, m), 6.13 (1H, dd, J = 5.9Hz, 2.3Hz), 5.96 (2H, br s), 5.82 (1H, d, J = 2.3Hz), 5.48 (1H, d, J = 11.1Hz), 5.42 (1H, d, J = 11.1Hz), 4.49-4.43 (1H, m), 4.07-4.00 (1H, m), 3.60-3.48 (3H, m), 3.35-3.28 (1H, m, (partly under water peak)), 3.07-2.99 (1H, m), 1.91 (3H, s, AcOH), 0.94-0.83 (2H, m), -0.02 (9H, s). [0515] Step 4 – 2,2-difluoro-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymeth yl)imidazol-2- yl]chroman-6-yl]oxy-2-pyridyl]cyclopropanecarboxamide [0516] To a solution of 2,2-difluorocyclopropanecarboxylic acid (63.7mg, 0.52mmol) in anhydrous THF (0.7mL) at 0°C under a nitrogen atmosphere were added DMF (2µL, 0.0300mmol) and oxalyl chloride (44.6µL, 0.53mmol) and the reaction was stirred at 0°C for 30 minutes. The mixture was then added to a solution of acetic acid; 4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyr idin-2-amine (60mg, 0.10mmol) in anhydrous Pyridine (0.70mL) and stirred at rt for 1 hr. The mixture was diluted with EtOAc (30mL) and washed with sat. aq. NaHCO 3 (10mL), water (10mL) and brine (10mL). The organic layer was dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give 2,2-difluoro-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]cyclopropanecarboxamide (47.3mg, 0.076mmol, 73% yield) as a glassy solid. UPLC- MS (ES+, short acidic): 2.06 min, m/z 619.5 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 11.03 (1H, s), 8.20 (1H, d, J = 5.7Hz), 7.77 (1H, s), 7.76-7.72 (2H, m), 7.62 (1H, br s), 7.37- 7.32 (2H, m), 7.22-7.16 (1H, m), 7.05 (1H, d, J = 2.6Hz), 6.96 (1H, dd, J = 8.8Hz, 2.6Hz), 6.92 (1H, d, J = 8.8Hz), 6.67 (1H, ddd, J = 5.7Hz, 4.1Hz, 2.3Hz), 5.48 (1H, d, J = 11.1Hz), 5.42 (1H, d, J = 11.1Hz), 5.50-5.44 (1H, m), 4.05 (1H, td, J = 10.7Hz, 1.7Hz), 3.60-3.54 (3H, m), 3.32-3.25 (1H, m), 3.07-2.90 (2H, m), 2.04-1.93 (2H, m), 0.93-0.85 (2H, m), -0.03 (9H, s). [0517] Step 5 – 2,2-difluoro-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl ]oxy-2- pyridyl]cyclopropanecarboxamide [0518] To a solution of 2,2-difluoro-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]cyclopropanecarboxamide (47.3mg, 0.08mmol) in DCM (1mL) was added trifluoroacetic acid (0.5mL, 6.53mmol) and the mixture was stirred at rt for 72h. The solvent was removed in vacuo and the residue loaded into loaded into an SCX-2 column, which was flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. The residue was purified by column chromatography using as eluent a gradient 0-6% MeOH in DCM to give 2,2-difluoro-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl ]oxy-2- pyridyl]cyclopropanecarboxamide (28.8mg, 0.059mmol, 77% yield) as a white solid. UPLC- MS (ES+, final purity): 2.94 min, m/z 489.3 [M+H]+. 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.32 (0.2H, br s), 12.10 (0.8H, br s), 11.03 (1H, s), 8.19 (1H, d, J = 5.7Hz), 7.79-7.56 (3.78H, m), 7.44-7.12 (3.22, m), 7.06-7.03 (1H, m), 6.94 (1H, dd, J = 8.8Hz, 2.8Hz), 6.90 (1H, d, J = 8.8Hz), 6.67 (1H, ddd, J = 5.7Hz, 2.3Hz, 0.7Hz), 4.55-4.49 (1H, m), 4.17-4.09 (1H, m), 3.45- 3.36 (1H, m), 3.29-3.19 (1H, m), 3.18-3.07 (1H, m), 3.01-2.90 (1H, m), 2.05-1.91 (2H, m). [0519] The compounds in the table below were made in an analogous manner, using the appropriate acid in step 4: [0520] Example 34. Synthesis of N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2- pyridyl]acetamide (Compound 156) [0521] Step 1 – N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2 -yl]chroman- 6-yl]oxy-2-pyridyl]acetamide [0522] To a solution of acetic acid; 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol- 2-yl]chroman-6-yl]oxypyridin-2-amine (69.5mg, 0.12mmol) in pyridine (0.6mL) was slowly added a solution of acetic anhydride (23µL, 0.24mmol) in THF (0.6mL) and the mixture was stirred at rt for 72 hrs. The mixture was diluted with EtOAc (30mL) and washed with sat. aq. NaHCO 3 (10mL), water (10mL) and brine (10mL). The organic layer was dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]acetamide (47.5mg, 0.085mmol, 71% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.90 min, m/z 557.5 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.50 (1H, s), 8.15 (1H, d, J = 5.7Hz), 7.77 (1H, s), 7.76-7.72 (2H, m), 7.66 (1H, d, J = 2.1Hz), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.02 (1H, d, J = 2.4Hz), 6.97-6.90 (2H, m), 6.60 (1H, dd, J = 5.7Hz, 2.4Hz), 5.49 (1H, d, J = 11.1Hz), 5.42 (1H, d, J = 11.1Hz), 4.50-4.43 (1H, m), 4.09-4.01 (1H, m), 3.61-3.50 (3H, m), 3.31-3.25 (1H, m), 3.07-2.98 (1H, m), 2.05 (3H, s), 0.93-0.86 (2H, m), -0.02 (9H, s). [0523] Step 2 – N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridy l]acetamide [0524] To a solution of N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2 - yl]chroman-6-yl]oxy-2-pyridyl]acetamide (39.4mg, 0.07mmol) in DCM (1mL) was added TFA (0.5mL, 6.53mmol) and the mixture was stirred at rt for 22 hrs. The solvent was removed under reduce pressure and the residue was loaded into an SCX-2 column and flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. The solvent was removed in vacuo and the residue was purified by column chromatography using as eluent a gradient 0-8% MeOH in DCM to give N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6- yl]oxy-2-pyridyl]acetamide (20.2mg, 0.047mmol, 70% yield) as a white solid. UPLC-MS (ES+, final purity): 2.45 min, m/z 427.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.32 (0.2, br s), 12.10 (0.8H, br s), 10.50 (1H, s), 8.15 (1H, d, J = 5.7Hz), 7.78-7.73 (1.6H, m), 7.68- 7.57 (2.2H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.18-7.13 (0.8H, m), 7.05-7.01 (1H, m), 6.93 (1H, dd, J = 8.8Hz, 2.7Hz), 6.89 (1H, d, J = 8.8Hz), 6.60 (1H, dd, J = 5.7Hz, 2.4Hz), 4.55-4.49 (1H, m), 4.16-4.08 (1H, m), 3.45-3.35 (1H, m), 3.29- 3.19 (1H, m), 3.16-3.07 (1H, m), 2.05 (3H, s). [0525] Example 35. Synthesis of N-cyclopropyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman- 6-yl]oxy-pyridine-2-carboxamide (Compound 158) [0526] Step 1 - 4-chloro-N-cyclopropyl-pyridine-2-carboxamide [0527] To a suspension of 4-chloro-2-pyridinecarboxylic acid (500mg, 3.17mmol) and cyclopropylamine (242µL, 3.49mmol) in THF (32mL) were added T3P (2.8mL, 4.7mmol) and DIPEA (1.1mL, 6.32mmol) and the mixture was heated at 65°C for 2.5 hours. The solvent was removed under reduce pressure and the residue dissolved in EtOAc (100mL). The organic layer was washed with sat. aq. NaHCO 3 (25mL), water (2 x 25mL) and brine (25mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo to give 4-chloro-N-cyclopropyl-pyridine-2- carboxamide (499.8mg, 2.54mmol, 80% yield) as an off-white solid. The compound used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.35 min, m/z 197.0 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.82 (1H, d, J = 4.4Hz), 8.59 (1H, dd, J = 5.3, 0.6Hz), 8.01 (1H, dd, J = 2.2Hz, 0.6Hz), 7.75 (1H, dd, J = 5.3Hz, 2.2Hz), 2.95-2.87 (1H, m), 0.72-0.65 (4H, m). [0528] Step 2 - 6-[[2-(cyclopropylcarbamoyl)-4-pyridyl]oxy]chromane-3-carbox ylic acid [0529] To a solution of 4-chloro-N-cyclopropyl-pyridine-2-carboxamide (200mg, 1.02mmol) in DMF (10mL) under a nitrogen atmosphere were added Cs 2 CO 3 (994.3mg, 3.05mmol) and 6-hydroxychromane-3-carboxylic acid (197.8mg, 1.02mmol) and the mixture was heated at 100°C overnight. Additional 6-hydroxychromane-3-carboxylic acid (49.4mg, 0.25mmol) and Cs 2 CO 3 (165.7mg, 0.51mmol) were added and the mixture was heated at 100°C for 72 hours. After cooling to rt, the mixture was poured into cold water and the aqueous was acidified to pH 2 with 1N aq. HCl followed by with EtOAc (4x50mL). The combined organic layers were dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give 6-[[2- (cyclopropylcarbamoyl)-4-pyridyl]oxy]chromane-3-carboxylic acid (192.9mg, 0.54mmol, 54% yield) as a beige solid. UPLC-MS (ES+, short acidic): 1.47 min, m/z 355.4 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.69 (1H, s), 8.71 (1H, d, J = 5.0Hz), 8.47 (1H, dd, J = 5.6Hz, 0.4Hz), 7.34 (1H, dd, J = 2.7Hz, 0.4Hz), 7.12 (1H, dd, J = 5.6Hz, 2.7Hz), 7.04 (1H, d, J = 2.8Hz), 6.94 (1H, dd, J = 8.8Hz, 2.8Hz), 6.88 (1H, d, J = 8.8Hz), 4.36 (1H, dd, J = 10.8Hz, 3.1Hz), 4.17 (1H, dd, J = 10.8Hz, 7.7Hz), 3.06-2.95 (3H, m), 2.89-2.82 (1H, m), 0.69-0.63 (4H, m). [0530] Step 3 - N-cyclopropyl-4-[3-(phenacylcarbamoyl)chroman-6-yl]oxy-pyrid ine-2- carboxamide [0531] To a solution of 6-[[2-(cyclopropylcarbamoyl)-4-pyridyl]oxy]chromane-3-carbox ylic acid (190.6mg, 0.54mmol) and 2-aminoacetophenone hydrochloride (101.6mg, 0.59mmol) in THF (5.3mL) were added T3P (480µL, 0.81mmol) and DIPEA (290µL, 1.66mmol) and the mixture was heated at 65°C for 2 hours. After cooling to rt, the solvent was removed in vacuo and the residue dissolved in EtOAc (50mL). The organic layer was washed with water (2x15mL) and brine (15mL), dried over Na 2 SO 4 , filtered and the solvent removed under reduce pressure. The residue was suspended in petroleum ether, filtered and dried to give N- cyclopropyl-4-[3-(phenacylcarbamoyl)chroman-6-yl]oxy-pyridin e-2-carboxamide (228mg, 0.48mmol, 90% yield) as a cream solid. UPLC-MS (ES+, short acidic): 1.64 min, m/z 472.5 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.72 (1H, d, J = 5.0Hz), 8.58-8.53 (1H, m), 8.47 (1H, dd, J = 5.6Hz, 0.3Hz), 8.02-7.97 (2H, m), 7.70-7.65 (1H, m), 7.58-7.52 (2H, m), 7.35 (1H, dd, J = 2.6Hz, 0.3Hz), 7.13 (1H, dd, J = 5.6Hz, 2.6Hz), 7.06 (1H, d, J = 2.8Hz), 6.96 (1H, dd, J = 8.8Hz, 2.8Hz), 6.90 (1H, d, J = 8.8Hz), 4.67 (2H, d, J = 5.6Hz), 4.43-4.37 (1H, m), 4.03-3.95 (1H, m), 3.07-2.82 (4H, m), 0.69-0.62 (4H, m). [0532] Step 4 - N-cyclopropyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]o xy-pyridine- 2-carboxamide [0533] To a suspension of N-cyclopropyl-4-[3-(phenacylcarbamoyl)chroman-6-yl]oxy- pyridine-2-carboxamide (227.6mg, 0.48mmol) in 1-butanol (4.8mL) were added ammonium acetate (372.1mg, 4.82mmol) and triethylamine (67µL, 0.48mmol) and the mixture was irradiated at 150°C for 45 minutes. The solvent was removed under reduce pressure and the residue purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM, followed by purification by reverse column chromatography using as eluent a gradient 0-50% (MeCN+0.1% formic acid) in (water+0.1% formic acid). Fractions containing product were combined, the solvent removed in vacuo and the residue loaded into a SCX-2 which was flushed at first with MeOH and then MeOH+NH 3 to elute the product. The solvent was removed in vacuo to give N-cyclopropyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]o xy- pyridine-2-carboxamide (75.7mg, 0.17mmol, 67% yield) as a white solid. UPLC-MS (ES+, final purity): 3.12 min, m/z 453.4 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.24 (1H, br s), 8.72 (1H, d, J = 5.0Hz), 8.47 (1H, dd, J = 5.6Hz, 0.3Hz), 7.76-7.70 (2H, m), 7.55 (1H, br s), 7.39-7.32 (3H, m), 7.22-7.16 (1H, m), 7.15 (1H, dd, J = 5.6Hz, 2.6Hz), 7.09 (1H, d, J = 2.8Hz), 6.99 (1H, dd, J = 8.8Hz, 2.8Hz), 6.94 (1H, d, J = 8.8Hz), 4.57-4.51 (1H, m), 4.20- 4.13 (1H, m), 3.49-3.39 (1H, m), 3.30-3.22 (1H, m), 3.19-3.10 (1H, m), 2.91-2.82 (1H, m), 0.71-0.62 (4H, m). [0534] Example 36. Synthesis of N-cyclobutyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6- yl]oxy-pyridin-2-amine (Compound 142) [0535] Step 1 – N-cyclobutyl-4-[3-[5-phenyl-1-(2-trimethylsilylethoxymethyl) imidazol-2- yl]chroman-6-yl]oxy-pyridin-2-amine [0536] A solution of 2-[[2-[6-[(2-chloro-4-pyridyl)oxy]chroman-3-yl]-5-phenyl-imi dazol-1- yl]methoxy]ethyl-trimethyl-silane (125mg, 0.23mmol; Example 33, Step 1), tris(dibenzylideneacetone)dipalladium (0) (21.5mg, 0.02mmol), (+/-)-BINAP (29.2mg, 0.05mmol) and Cs2CO 3 (152.5mg, 0.47mmol) in 1,4-Dioxane (2.2mL) was degassed under a nitrogen atmosphere for 10 minutes then cyclobutanamine (40µL, 0.47mmol) was added. The vial was sealed and the mixture heated at 100°C for 20 hours. After cooling to rt, the mixture was filtered through diatomaceous earth and the filter cake was washed with EtOAc. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give N-cyclobutyl-4-[3-[5-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-py ridin-2-amine (133.1mg, 0.23mmol, 100% yield) as an orange foam. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.83 min, m/z 569.3 [M+H] + . [0537] Step 2 – N-cyclobutyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]ox y-pyridin-2- amine [0538] To a solution of N-cyclobutyl-4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-py ridin-2-amine (133.1mg, 0.23mmol) in DCM (3.6mL) was added TFA (1.8mL, 23.51mmol) and the mixture was stirred at 25°C for 18 hours. The solvent was removed under reduce pressure, the residue loaded into an SCX-2 column and flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to elute the product. The residue was purified by column chromatography using as eluent a gradient 0-6% MeOH in DCM, followed by reverse column chromatography using as eluent a gradient 0-50% (MeCN+0.1% formic acid) in (water+0.1% formic acid). Fractions containing the product were loaded onto a SCX-2 column and flushed at first with MeOH (10mL) and then NH 3 in MeOH (10mL) to give N-cyclobutyl-4-[3-(4-phenyl-1H-imidazol-2-yl)chroman- 6-yl]oxy-pyridin-2-amine (30mg, 0.068mmol, 29% yield) as a white solid. UPLC-MS (ES+, final purity): 2.45 min, m/z 439.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.12 (1H, br s), 7.82 (1H, d, J = 5.8Hz), 7.73 (2H, m), 7.55 (1H, br s), 7.38-7.30 (2H, m), 7.21-7.14 (1H, m), 6.99-6.96 (1H, m), 6.91-6.85 (2H, m), 6.77 (1H, d, J = 6.6Hz, broad), 6.10 (1H, dd, J = 5.8, 2.2Hz), 5.76 (1H, d, J = 2.2Hz), 4.54-4.48 (1H, m), 4.27-4.15 (1H, m), 4.15-4.06 (1H, m), 3.43- 3.34 (1H, m), 3.28-3.19 (1H, m), 3.15-3.07 (1H, m), 2.25-2.16 (2H, m), 1.85-1.73 (2H, m), 1.69-1.53 (2H, m). [0539] Example 37. Synthesis of 1-methyl-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6- yl]oxy-2-pyridyl]piperidine-4-carboxamide (Compound 147) [0540] Step 1 - 1-methyl-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)i midazol-2- yl]chroman-6-yl]oxy-2-pyridyl]piperidine-4-carboxamide [0541] A solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl ]chroman- 6-yl]oxypyridin-2-amine (60mg, 0.12mmol), 1-methylpiperidine-4-carboxylic acid (18.4mg, 0.13mmol), propylphosphonic anhydride (104µL, 0.17mmol) and N,N-diisopropylethylamine (41µL, 0.23mmol) in THF (1.2mL) was heated at 65°C, in a sealed tube, overnight. Additional 1-methylpiperidine-4-carboxylic acid (18.4mg, 0.13mmol), propylphosphonic anhydride (104µL, 0.17mmol) and N,N-diisopropylethylamine (41µL, 0.23mmol) were added in a sealable vial. The vial was sealed and the mixture heated at 65°C for 2 hours. After cooling to room temperature, the mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM, followed by 20% NH 3 2N in MeOH in DCM to give 1-methyl-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]piperidine-4- carboxamide (38.4mg, 0.060mmol, 51% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.71 min, m/z 640.3 [M+H] + . [0542] Step 2 - 1-methyl-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy -2- pyridyl]piperidine-4-carboxamide [0543] To a solution of 1-methyl-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]piperidine-4- carboxamide (38.4mg, 0.060mmol) in DCM (1mL) was added trifluoroacetic acid (0.5mL, 6.53mmol) in a sealable vial. The vial was sealed and the mixture stirred at 25°C for 18 hours. The mixture was concentrated and the residue purified by reverse column chromatography using as eluent a gradient 0-50% of acetonitrile+0.1% formic acid in water+0.1% formic acid. Fraction containing the product were combined, the solvent removed in vacuo and loaded into an SCX-2 column. The column was washed at first with MeOH and then MeOH+NH 3 to give 1-methyl-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy -2-pyridyl]piperidine-4- carboxamide (16mg, 0.031mmol, 52% yield) as a white solid. UPLC-MS (ES+, final purity): 2.26 min, m/z 510.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.31 (0.2H, br s), 12.09 (0.8H, br s), 10.45 (1H, s), 8.15 (1H, d, J = 5.7Hz), 7.77-7.73 (1.6H, m), 7.67 (1H, d, J = 2.4Hz), 7.66-7.61 (0.4H, m), 7.61-7.57 (0.8H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.19-7.13 (0.8H, m), 7.04-7.01 (1H, m), 6.93 (1H, dd, J = 8.8, 2.6Hz), 6.90 (1H, d, J = 8.8Hz), 6.62 (1H, dd, J = 5.7, 2.4Hz), 4.56-4.49 (1H, m), 4.17-4.09 (1H, m), 3.46-3.36 (1H, m), 3.29-3.20 (1H, m), 3.17-3.07 (1H, m), 2.85-2.75 (2H, m), 2.43-2.36 (1H, m), 2.16 (3H, s), 1.92-1.69 (2H, m), 1.74-1.65 (2H, m), 1.64-1.51 (2H, m). [0544] The compounds in the table below were made in an analogous manner, using the appropriate acid in step 1:

[0545] Example 38. Synthesis of 2-(4-methylpiperazin-1-yl)-N-[4-[3-(4-phenyl-1H-imidazol- 2-yl)chroman-6-yl]oxy-2-pyridyl]acetamide (Compound 146) [0546] Step 1 - 2-(4-methylpiperazin-1-yl)-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]acetamide [0547] To a solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2- yl]chroman-6-yl]oxypyridin-2-amine (100mg, 0.19mmol) and Et3N (68µL, 0.49mmol) in anhydrous THF (2mL) at 0°C under a nitrogen atmosphere was slowly added a solution of chloroacetyl chloride (16µL, 0.20mmol) in THF (0.5mL) and the mixture was stirred at 25°C for 1 hour. A solution of 1-methylpiperazine (26µL, 0.23mmol) in anhydrous THF (0.5mL) was then added dropwise and the mixture was stirred at 25°C for 18hrs. Additional 1- methylpiperazine (26µL, 0.23mmol) was added and the mixture heated at 65°C for 4 hours. After cooling to room temperature, the solvent was removed under reduce pressure and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM to give 2-(4-methylpiperazin-1-yl)-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]acetamide (50.5mg, 0.077mmol, 40% yield) as a yellow solid. UPLC-MS (ES+, Short acidic): 1.68 min, m/z 655.4 [M+H] + . [0548] Step 2 - 2-(4-methylpiperazin-1-yl)-N-[4-[3-(4-phenyl-1H-imidazol-2-y l)chroman-6- yl]oxy-2-pyridyl]acetamide [0549] To a solution of 2-(4-methylpiperazin-1-yl)-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]acetamide (49.3mg, 0.0800mmol) in DCM (1.2mL) was added trifluoroacetic acid (0.6mL, 7.84mmol) and the mixture was stirred at 25°C, in a sealed tube, for 22 hrs. The mixture was concentrated and the residue purified by flash chromatography (C18, 12g column, gradient 0-50% MeCN in H2O + 0.1% HCO2H). Pure fractions were loaded onto a SCX-2 column pre-equilibrated with methanol. The column was washed with methanol and the filtrate was discarded. The product was then eluted with NH32N in methanol and the filtrate was concentrated to afford 2-(4- methylpiperazin-1-yl)-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chr oman-6-yl]oxy-2- pyridyl]acetamide (26mg, 0.050mmol, 66% yield) as a white solid. UPLC-MS (ES+, final purity): 2.29 min, m/z 525.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.31 (0.2H, br s), 12.09 (0.8H, br s), 9.89 (1H, s), 8.17 (1H, d, J = 5.8Hz), 7.77-7.73 (1.6H, m), 7.66 (1H, d, J = 2.4Hz), 7.65-7.61 (0.4H, m), 7.61-7.57 (0.8H, m), 7.43-7.37 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.19-7.13 (0.8H, m), 7.06-7.02 (1H, m), 6.94 (1H, dd, J = 8.8, 2.7Hz), 6.90 (1H, d, J = 8.8Hz), 6.66 (1H, dd, J = 5.8, 2.4Hz), 4.55-4.49 (1H, m), 4.17-4.09 (1H, m), 3.45-3.35 (1H, m), 3.26-3.19 (1H, m), 3.16-3.07 (3H, m), 2.37 (4H, m, seen as a br s), 2.18 (3H, s).4H missing (underneath DMSO / water peaks). [0550] The compounds in the table below were made in an analogous manner, using the appropriate amine in step 1: [0551] Example 39. Synthesis of 2-morpholino-N-[4-[3-(4-phenyl-1H-imidazol-2- yl)chroman-6-yl]oxy-2-pyridyl]propenamide (Compound 150) [0552] Step 1 – 2-morpholino-N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymeth yl)imidazol- 2-yl]chroman-6-yl]oxy-2-pyridyl]propanamide [0553] To a solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2- yl]chroman-6-yl]oxypyridin-2-amine (68mg, 0.13mmol) and triethylamine (55µL, 0.40mmol) in anhydrous THF (1.3mL) at 0°C under a nitrogen atmosphere slowly added solution of chloropropionyl chloride (16µL, 0.16mmol) in anhydrous THF (0.5mL) and the mixture was stirred at room temperature for 2 hrs. To this mixture was added morpholine (23µL, 0.26mmol) and sodium iodide (40mg, 0.26mmol) and the reaction was heated at 60°C for 72 hours. After cooling to room temperature, the mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM to give 2-morpholino-N-[4- [3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]c hroman-6-yl]oxy-2- pyridyl]propanamide (43mg, 0.065mmol, 49% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.82 min, m/z 656.5 [M+H] + . [0554] Step 2 – 2-morpholino-N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl ]oxy-2- pyridyl]propenamide [0555] To a solution of 2-morpholino-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-2- pyridyl]propanamide (42mg, 0.06mmol) in DCM (1mL) was added trifluoroacetic acid (490µL, 6.4mmol) and the mixture was stirred at 25°C in a sealed tube for 22 hrs. The mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-50% (acetonitrile+0.1% formic acid):(water+0.1%formic acid). Fractions containing the product were loaded onto a SCX-2 column, which was flushed with MeOH and then MeOH+NH 3 to give 2-morpholino- N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxy-2-pyridy l]propanamide (23mg, 0.043mmol, 68% yield) as a white solid. UPLC-MS (ES+, final purity): 2.46 min, m/z 526.3 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.16 (1H, br s), 10.13 (1H, s), 8.17 (1H, d, J = 5.7Hz), 7.76-7.70 (2H, m), 7.68 (1H, d, J = 2.3Hz), 7.54 (1H, br s), 7.38-7.31 (2H, m), 7.21-7.15 (1H, m), 7.04 (1H, d, J = 2.7Hz), 6.94 (1H, dd, J = 8.8Hz, 2.7Hz), 6.90 (1H, d, J = 8.8Hz), 6.66 (1H, dd, J = 5.7Hz, 2.3Hz), 4.56-4.50 (1H, m), 4.18-4.10 (1H, m), 3.65-3.56 (4H, m), 3.46-3.35 (2H, m), 3.30-3.08 (2H, m), 1.15 (3H, d, J = 6.7Hz).4H missing (underneath DMSO or water peak). [0556] Example 40. Synthesis of N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6- yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (Compound 148) [0557] Step 1 – 4-chloro-6-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imid azol-2- yl]chroman-6-yl]oxy-pyrimidin-2-amine [0558] A mixture of 4,6-dichloropyrimidin-2-amine (51.2mg, 0.31mmol), 3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-ol (132mg, 0.31mmol) and Cs2CO 3 (152.6mg, 0.47mmol) in DMF (1.5mL) was heated at 60°C overnight. After cooling to rt, the mixture was poured into water (30mL) and the aqueous was extracted with EtOAc (5x20mL) then EtOAc:MeOH (10:1, 5x20mL). The combined organic extracts were dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM to give 4-chloro-6-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxy-py rimidin-2-amine (120mg, 0.22mmol, 70% yield) as a white foam. UPLC-MS (ES+, short acidic): 2.10 min, m/z 550.5 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 7.77 (1H, s), 7.76-7.72 (2H, m), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.13 (2H, br s), 7.01 (1H, d, J = 2.8Hz), 6.95 (1H, dd, J = 8.8, 2.8Hz), 6.87 (1H, d, J = 8.8Hz), 6.13 (1H, s), 5.45 (2H, m), 4.49-4.42 (1H, m), 4.08-4.00 (1H, m), 3.63-3.46 (3H, m), 3.31-3.23 (1H, m), 3.07-2.97 (1H, m), 0.94-0.85 (2H, m), -0.02 (9H, s). [0559] Step 2 – 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl ]chroman-6- yl]oxypyrimidin-2-amine [0560] To a mixture of 4-chloro-6-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imid azol-2- yl]chroman-6-yl]oxy-pyrimidin-2-amine (119mg, 0.22mmol) and triethylamine (33µL, 0.24mmol) in THF (7mL) was added palladium, 10 wt. % on carbon powder, wet (57.5mg, 0.05mmol). The reaction was fitted with a H 2 balloon, extra H 2 added and subjected to 3 x vacuum/H 2 cycles and then left to stir under a H 2 atmosphere for 72 hours. The mixture was filtered through a plug of diatomaceous earth and the filter cake was washed with THF and MeOH. The filtrate was concentrated and the residue partitioned between EtOAc (10mL) and water (10mL). The organic layer was separated and the aqueous extracted with EtOAc (3x10mL). The combined organic extracts were washed with brine (10mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo to give 4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyr imidin-2-amine (111.5mg, 0.22mmol, 100% yield) as a white foam. The compound was used in the next step without further purification. UPLC-MS (ES+, short acidic): 1.75 min, m/z 516.5 [M+H] + . [0561] Step 3 – N-(cyclopropanecarbonyl)-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyr imidin-2- yl]cyclopropanecarboxamide [0562] To a solution of 4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2- yl]chroman-6-yl]oxypyrimidin-2-amine (111.5mg, 0.22mmol) in pyridine (1.6mL) and DCM (1.1mL) was added cyclopropanecarbonyl chloride (25µL, 0.27mmol) in DCM (0.1mL) dropwise and the mixture was stirred at 25°C under a nitrogen atmosphere for 18 hours. A solution of cyclopropanecarbonyl chloride (98µL, 1.08mmol) in DCM (0.1mL) was slowly added and the mixture was stirred at 25°C for 5 hours. The crude was diluted with DCM (30mL), washed with water (15mL) and brine (15mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-40% EtOAc in petroleum ether to give N-(cyclopropanecarbonyl)-N-[4-[3-[4- phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-yl]chroman -6-yl]oxypyrimidin-2- yl]cyclopropanecarboxamide (101mg, 0.16mmol, 72% yield) as colourless oil, which solidified upon standing. UPLC-MS (ES+, short acidic): 2.14 min, m/z 652.9 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 8.76 (1H, d, J = 5.8Hz), 7.77 (1H, s), 7.77-7.72 (2H, m), 7.38- 7.32 (2H, m), 7.23-7.16 (1H, m), 7.13 (1H, d, J = 5.8Hz), 7.07 (1H, d, J = 2.8Hz), 7.00 (1H, dd, J = 8.8, 2.8Hz), 6.91 (1H, d, J = 8.8Hz), 5.47 (1H, d, J = 11.1Hz), 5.43 (1H, d, J = 11.1Hz), 4.50-4.44 (1H, m), 4.09-4.02 (1H, m), 3.61-3.51 (3H, m), 3.31-3.25 (1H, m), 3.06-2.97 (1H, m), 1.95-1.87 (2H, m), 0.97-0.88 (10H, m), -0.02 (9H, s). [0563] Step 4 – N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2 -yl]chroman- 6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide [0564] N-(cyclopropanecarbonyl)-N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyr imidin-2- yl]cyclopropanecarboxamide (96mg, 0.15mmol) was dissolved in NH 3 (7M in MeOH - 3mL, 21mmol) and the mixture was stirred at 25°C for 1 hour. The solvent was removed under reduce pressure and the residue was purified by column chromatography using as eluent a gradient 0- 80% EtOAc in petroleum ether to give N-[4-[3-[4-phenyl-1-(2- trimethylsilylethoxymethyl)imidazol-2-yl]chroman-6-yl]oxypyr imidin-2- yl]cyclopropanecarboxamide (70mg, 0.12mmol, 82% yield) as a white solid. UPLC-MS (ES+, short acidic): 1.97 min, m/z 584.4 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.62 (1H, s), 8.47 (1H, d, J = 5.7Hz), 7.77 (1H, s), 7.77-7.72 (2H, m), 7.38-7.32 (2H, m), 7.22-7.17 (1H, m), 7.10 (1H, d, J = 2.8Hz), 7.02 (1H, dd, J = 8.8, 2.8Hz), 6.89 (1H, d, J = 8.8Hz), 6.63 (1H, d, J = 5.7Hz), 5.47 (1H, d, J = 11.1Hz), 5.42 (1H, d, J = 11.1Hz), 4.50-4.43 (1H, m), 4.09-4.02 (1H, m), 3.61-3.54 (2H, m), 3.54-3.47 (1H, m), 3.31-3.24 (1H, m), 3.07-2.98 (1H, m), 2.24- 2.16 (1H, m), 0.93-0.86 (2H, m), 0.80-0.68 (4H, m), -0.02 (9H, s). [0565] Step 5 – N-[4-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6-yl]oxypyrimidin -2- yl]cyclopropanecarboxamide [0566] To a solution of N-[4-[3-[4-phenyl-1-(2-trimethylsilylethoxymethyl)imidazol-2 - yl]chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (69mg, 0.12mmol) in DCM (1.8mL) was added trifluoroacetic acid (0.9mL, 11.75mmol) and the mixture was stirred at 25°C for 18 hours. The mixture was concentrated and the residue loaded onto a SCX-2 column, which was washed at first with MeOH and then NH 3 in MeOH to elute the product. The residue was purified by column chromatography using as eluent a gradient 0-8% MeOH in DCM. Like fractions were pooled and concentrated to afford N-[4-[3-(4-phenyl-1H-imidazol-2- yl)chroman-6-yl]oxypyrimidin-2-yl]cyclopropanecarboxamide (44mg, 0.096mmol, 82% yield) as a white solid. UPLC-MS (ES+, final purity): 2.65 min, m/z 454.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.32 (0.2H, br s), 12.10 (0.8H, br s), 10.62 (1H, s), 8.46 (1H, d, J = 5.7Hz), 7.79-7.72 (1.6H, m), 7.67-7.56 (1.2H, m), 7.45-7.36 (0.4H, m), 7.36-7.29 (1.6H, m), 7.29-7.20 (0.4H, m), 7.20-7.12 (0.8H, m), 7.10 (1H, d, J = 2.8Hz), 7.00 (1H, dd, J = 8.8, 2.8Hz), 6.86 (1H, d, J = 8.8Hz), 6.63 (1H, d, J = 5.7Hz), 4.54-4.48 (1H, m), 4.15-4.06 (1H, m), 3.43-3.33 (1H, m), 3.29-3.18 (1H, m), 3.16-3.06 (1H, m), 2.23-2.15 (1H, m), 0.80-0.66 (4H, m). [0567] Example 41. Synthesis of 2-(4-methyl-1H-imidazol-2-yl)-4-[3-(4-phenyl-1H- imidazol-2-yl)chroman-6-yl]oxy-pyridine (Compound 143) [0568] Step 1 – tert-butyl 6-[[2-[4-methyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-y l]-4- pyridyl]oxy]chromane-3-carboxylate [0569] A mixture of 2-[[2-(4-bromo-2-pyridyl)-4-methyl-imidazol-1-yl]methoxy]eth yl- trimethyl-silane (285mg, 0.77mmol), tert-butyl 6-hydroxychromane-3-carboxylate (213mg, 0.85mmol) and potassium phosphate tribasic (328.5mg, 1.55mmol) in anhydrous toluene (3.8mL) was degassed under a nitrogen atmosphere for 10 minutes; then palladium (II) acetate (10.4mg, 0.05mmol) and 2-(di-tert-butylphosphino)biphenyl (27.7mg, 0.09mmol) were added. The vial was sealed and the mixture heated at 100°C for 4 hours. After cooling to room temperature, the mixture was filtered through a plug of diatomaceous earth and the filter cake washed with DCM. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-100% EtOAc in petroleum ether to give tert-butyl 6-[[2-[4-methyl-1-(2-trimethylsilylethoxymethyl)imidazol-2-y l]-4-pyridyl]oxy]chromane-3- carboxylate (330mg, 0.61mmol, 79% yield) as a dark orange oil. UPLC-MS (ES+, Short acidic): 1.92 & 1.98 min, m/z 538.8 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) consistent with a ~ 2.5 / 1 ratio of regioisomers. [0570] [0571] Step 2 - formic acid; 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]chromane-3- carboxylic acid (0.4; 1) [0572] To a solution of tert-butyl 6-[[2-[4-methyl-1-(2-trimethylsilylethoxymethyl)imidazol- 2-yl]-4-pyridyl]oxy]chromane-3-carboxylate (328mg, 0.61mmol) in DCM (4.6mL) was added trifluoroacetic acid (2.3mL, 30.04mmol) and the mixture was stirred at room temperature, in a sealed vial, overnight. The mixture was concentrated and the residue azeotroped with toluene. The residue was purified by reverse column chromatography using as eluent a gradient 0-50% acetonitrile+0.1% formic acid in water+0.1% formic acid to give formic acid; 6-[[2-(4-methyl- 1H-imidazol-2-yl)-4-pyridyl]oxy]chromane-3-carboxylic acid (0.4; 1) (148mg, 0.40mmol, 66% yield) as a white solid. UPLC-MS (ES + , Short acidic): 1.14 min, m/z 352.4 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 13.26 (1H, br s), 12.72 (1H, br s), 8.47 (1H, d, J = 5.7Hz), 8.13 (0.4H, s, formate), 7.37 (1H, d, J = 2.3Hz), 7.05 (1H, d, J = 2.8Hz), 6.98-6.93 (3H, m), 6.88 (1H, d, J = 8.8Hz), 4.36 (1H, dd, J = 10.8Hz, 3.1Hz), 4.17 (1H, dd, J = 10.8Hz, 7.7Hz), 3.07-2.95 (3H, m), 2.19 (3H, s). [0573] Step 3 - 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]-N-phenacyl- chromane-3- carboxamide [0574] A mixture of formic acid; 6-[[2-(4-methyl-1H-imidazol-2-yl)-4- pyridyl]oxy]chromane-3-carboxylic acid (0.4; 1) (148mg, 0.40mmol), 2-aminoacetophenone hydrochloride (75.7mg, 0.44mmol), propylphosphonic anhydride (358µL, 0.60mmol) and N,N-diisopropylethylamine (245 µL, 1.41mmol) in THF (4mL) was heated at 65°C for 2 hours. After cooling to room temperature, the mixture was concentrated and the residue taken up in EtOAc (30mL). The organic layer was washed with water (2x10mL) and brine (10mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo to give 6-[[2-(4-methyl-1H-imidazol- 2-yl)-4-pyridyl]oxy]-N-phenacyl-chromane-3-carboxamide (188mg, 0.40mmol, 100% yield) as an orange solid. The compound was used in the next step without further purification. UPLC- MS (ES+, Short acidic): 1.33 min, m/z 469.5 [M+H] + . [0575] Step 4 - 2-(4-methyl-1H-imidazol-2-yl)-4-[3-(4-phenyl-1H-imidazol-2-y l)chroman-6- yl]oxy-pyridine [0576] To a suspension of 6-[[2-(4-methyl-1H-imidazol-2-yl)-4-pyridyl]oxy]-N-phenacyl- chromane-3-carboxamide (188mg, 0.40mmol) in 1-butanol (4mL) were added ammonium acetate (309mg, 4.01mmol) and Et3N (56µL, 0.40mmol). The vial was sealed and the mixture irradiated at 150°C for 45 minutes. After cooling to room temperature, the mixture was concentrated and the residue was purified by column chromatography using as eluent a gradient 0-20% MeOH. Fractions containing the product were combined and re-purified by reverse column chromatography using as eluent a gradient acetonitrile+0.1% formic acid. Fractions containing the product were loaded onto an SCX-2 column, which was flushed with MeOH followed by MeOH+NH 3 to give 2-(4-methyl-1H-imidazol-2-yl)-4-[3-(4-phenyl-1H-imidazol- 2-yl)chroman-6-yl]oxy-pyridine (42mg, 0.093mmol, 23% yield) as a white solid. UPLC-MS (ES+, final purity): 2.35 min, m/z 450.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 + few μL of CF 3 CO 2 D) δ/ppm: 8.65 (1H, d, J = 5.7Hz), 8.14 (1H, s), 7.84-7.80 (2H, m), 7.78 (1H, d, J = 2.3Hz), 7.58-7.51 (3H, m), 7.49-7.43 (1H, m), 7.15-7.10 (2H, m), 7.07 (1H, dd, J = 8.8Hz, 2.8Hz), 7.01 (1H, d, J = 8.8Hz), 4.65-4.58 (1H, m), 4.40 (1H, dd, J = 10.7Hz, 9.0Hz), 3.91- 3.82 (1H, m), 3.44-3.27 (2H, m), 2.33 (3H, d, J = 0.9Hz). [0577] Example 42. Synthesis of 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N- phenacyl-chromane-3-carboxamide (Compound 151) [0578] Step 1 – N-(6-chloropyrimidin-4-yl)cyclopropanecarboxamide [0579] To a solution of 6-chloropyrimidin-4-ylamine (800mg, 6.18mmol) and pyridine (1.25mL, 15.46mmol) in THF (25mL) at 0°C, under a nitrogen atmosphere, was slowly added cyclopropanecarbonyl chloride (0.7mL, 7.71mmol) and the mixture was heated at 60°C for 17 hours. After cooling to room temperature, the mixture was partitioned between water (50mL) and EtOAc (50mL). The two layers were separated and the aqueous extracted with EtOAc (2x50mL). The combined organic layers were washed with brine (50mL), dried over Na 2 SO 4 , filtered and concentrated. The residue was purified by column chromatography using as eluent a gradient 0-4% MeOH in DCM to give N-(6-chloropyrimidin-4-yl)cyclopropanecarboxamide (917mg, 4.64mmol, 75% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.35 min, m/z 197.9 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 11.54 (1H, s), 8.75 (1H, d, J = 1.0Hz), 8.10 (1H, d, J = 1.0Hz), 2.08-2.00 (1H, m), 0.93-0.86 (4H, m). [0580] Step 2 – 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxychromane-3 -carboxylic acid [0581] A solution of 6-hydroxychromane-3-carboxylic acid (198mg, 1.02mmol), N-(6- chloropyrimidin-4-yl)cyclopropanecarboxamide (200mg, 1.01mmol) and potassium carbonate (560mg, 4.05mmol) in DMSO (1.25mL) was heated 110°C overnight. The reaction mixture was cooled to room temperature and poured into a solution of citric acid (777.8mg, 4.05mmol) in water (15mL). The resulting precipitate was filtered, washed with water and dried to afford 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxychromane-3 -carboxylic acid (316mg, 0.89mmol, 88% yield) as a beige solid. Compound used directly in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.44 min, m/z 356.2 [M+H] + (94%). 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.67 (1H, br s), 11.22 (1H, s), 8.48 (1H, d, J = 0.9Hz), 7.50 (1H, d, J = 0.9Hz), 6.98 (1H, d, J = 2.8Hz), 6.89 (1H, dd, J = 8.8, 2.8Hz), 6.80 (1H, d, J = 8.8Hz), 4.32 (1H, dd, J = 10.8, 3.1Hz), 4.15 (1H, dd, J = 10.8, 7.5Hz), 3.04-2.92 (3H, m), 2.06-1.98 (1H, m), 0.89-0.80 (4H, m). [0582] Step 3 – 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacy l- chromane-3-carboxamide [0583] A mixture of 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxychromane-3 - carboxylic acid (100mg, 0.28mmol), 2-aminoacetophenone hydrochloride (53mg, 0.31mmol), propylphosphonic anhydride (250µL, 0.42mmol) and N,N-diisopropylethylamine (152µL, 0.87mmol) in THF (2.8mL) was heated at 65°C for 1 hour. After cooling to room temperature, the mixture was concentrated and the residue dissolved in EtOAc (30mL). The organic layer was washed with water (2x10mL) and brine (10mL), dried over Na 2 SO 4 , filtered and concentrated to afford 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacy l- chromane-3-carboxamide (131mg, 0.28mmol, 98% yield) as a beige solid. The compound used in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.61 min, m/z 473.2 [M+H] + . [0584] Step 4 – 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N-phenacy l- chromane-3-carboxamide [0585] To a suspension of 6-[6-(cyclopropanecarbonylamino)pyrimidin-4-yl]oxy-N- phenacyl-chromane-3-carboxamide (131mg, 0.28mmol) in 1-butanol (2.7mL) were added ammonium acetate (213mg, 2.77mmol) and triethylamine (39µL, 0.28mmol). The vial was sealed and the mixture irradiated at 150°C for 45 min. The mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0-20% MeOH in DCM. Fractions containing the product was re-purified by reverse column chromatography using as eluent gradient 0-40% acetonitrile+0.1% formic acid in water+0.1% formic acid. Fractions containing the product were loaded onto a SCX-2 column, which was flushed at first with MeOH and the MeOH/NH 3 to give N-[6-[3-(4-phenyl-1H-imidazol-2-yl)chroman-6- yl]oxypyrimidin-4-yl]cyclopropanecarboxamide (28mg, 0.062mmol, 22% yield) as a white solid. UPLC-MS (ES+, final purity): 2.94 min, m/z 454.3 [M+H] + . 1 H NMR (400MHz, DMSO- d 6 ) δ/ppm: 12.30 (1H, br s), 11.23 (1H, s), 8.50 (1H, d, J = 1.0Hz), 7.76-7.70 (2H, m), 7.56 (1H, br s), 7.52 (1H, d, J = 1.0Hz), 7.38-7.32 (2H, m), 7.22-7.16 (1H, m), 7.03 (1H, d, J = 2.8Hz), 6.94 (1H, dd, J = 8.8Hz, 2.8Hz), 6.86 (1H, d, J = 8.8Hz), 4.55-4.49 (1H, m), 4.18-4.10 (1H, m), 3.47-3.38 (1H, m), 3.29-3.19 (1H, m), 3.16-3.08 (1H, m), 2.07-1.99 (1H, m), 0.89- 0.82 (4H, m). [0586] Example 43. Synthesis of 5-[3-(5-isopropyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin- 2-yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 133) [0587] Step 1 – tert-butyl 4-hydroxyiminopiperidine-1-carboxylate [0588] A mixture of 1-Boc-4-piperidone (2g, 10.04mmol), hydroxylamine hydrochloride (1.4g, 20.08mmol) and potassium acetate (1.97g, 20.07mmol) in EtOH (20mL) was heated at 90°C overnight. After cooling to room temperature, the mixture was concentrated and the residue taken up in water (20mL). The aqueous layer was extracted with EtOAc (3x20mL). The combined organic layers were washed with sat aq. NaHCO 3 (25mL) and brine (25mL), separated, dried over Na 2 SO 4 , filtered and concentrated to give tert-butyl 4- hydroxyiminopiperidine-1-carboxylate (2.067g, 9.64mmol, 96% yield) as a white solid. UPLC-MS (ES+, Short acidic): 1.36 min, m/z 215.0 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 ) δ/ppm: 10.44 (1H, s), 3.44-3.35 (4H, m), 2.46-2.41 (2H, m), 2.24-2.19 (2H, m), 1.40 (9H, s). [0589] Step 2 – tert-butyl 3-amino-4,4-diethoxy-piperidine-1-carboxylate [0590] A mixture of tert-butyl 4-hydroxyiminopiperidine-1-carboxylate (2.06g, 9.63mmol), potassium carbonate (2.66g, 19.28mmol) and p-toluenesulfonyl chloride (1.84g, 9.64mmol) in THF (50mL) was stirred at room temperature for 24 hours then heated at 40°C for 20 hours. After cooling to room temperature, the mixture was filtered, and the solvent removed under reduce pressure. The residue was dissolved in EtOH (20mL) and added dropwise to a mixture of potassium ethoxide (1.62g, 19.27mmol) and Na 2 SO 4 anhydrous (5.47g, 38.54mmol) in EtOH (20mL) at 0°C under a nitrogen atmosphere. The mixture was stirred at room temperature for 1.5 hours then heated at 60°C for 1 hour. After cooling to room temperature, the mixture was filtered and the filter cake was washed with EtOH. The filtrate was concentrated and the residue purified by column chromatography using as eluent a gradient 0-8% MeOH in DCM to give tert-butyl 3-amino-4,4-diethoxy-piperidine-1-carboxylate (1.181g, 4.10mmol, 43% yield) as a thick yellow oil. UPLC-MS (ES+, Short acidic): 1.25 min, m/z 289.1 [M+H] + . [0591] Step 3 – tert-butyl 4,4-diethoxy-3-[[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4 - yl)oxy]chromane-3-carbonyl]amino]piperidine-1-carboxylate [0592] A mixture of 6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chromane- 3- carboxylic acid (585mg, 1.72mmol), tert-butyl 3-amino-4,4-diethoxy-piperidine-1-carboxylate (741mg, 2.57mmol), propylphosphonic anhydride (2.05mL, 3.44mmol) and N,N- diisopropylethylamine (0.9mL, 5.17mmol) in THF (17.2mL) was heated at 65°C for 2 hours. The mixture was concentrated and the residue dissolved in EtOAc (100mL). The organic layer was washed with water (2x30mL) and brine (30mL), dried over Na 2 SO 4 , filtered and concentrated to afford tert-butyl 4,4-diethoxy-3-[[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin- 4-yl)oxy]chromane-3-carbonyl]amino]piperidine-1-carboxylate (1049.7mg, 1.72mmol, 100% yield) as a light orange foam. The compound was used directly in the next step without further purification. UPLC-MS (ES+, Short acidic): 1.79 min, m/z 611.7 [M+H] + . [0593] Step 4 – tert-butyl 2-[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4-yl)oxy]chroma n- 3-yl]-3,4,6,7-tetrahydroimidazo[4,5-c]pyridine-5-carboxylate [0594] A mixture of tert-butyl 4,4-diethoxy-3-[[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin- 4-yl)oxy]chromane-3-carbonyl]amino]piperidine-1-carboxylate (1.05g, 1.72mmol), ammonium acetate (2.65g, 34.38mmol) and p-toluenesulfonic acid monohydrate (65.4mg, 0.34mmol) in 1-butanol (11.5mL) was irradiated at 150°C for 1.5 hours. Additional ammonium acetate (2.65g, 34.38mmol) and p-toluenesulfonic acid monohydrate (65.4mg, 0.34mmol) were added and the mixture was further irradiated at 150°C for 2 hours. The mixture was concentrated and the residue dissolved in EtOAc (100mL). The organic layer was washed with sat. aq. NaHCO 3 (30mL), water (30mL) and brine (30mL), dried over Na 2 SO 4 , filtered and the solvent removed in vacuo. The residue was purified by column chromatography using as eluent a gradient 0-10% MeOH in DCM to give tert-butyl 2-[6-[(7-oxo-6,8-dihydro-5H-1,8- naphthyridin-4-yl)oxy]chroman-3-yl]-3,4,6,7-tetrahydroimidaz o[4,5-c]pyridine-5-carboxylate (214mg, 0.41mmol, 24% yield) as an off-white solid. UPLC-MS (ES+, Short acidic): 1.27 min, m/z 518.4 [M+H] + . [0595] Step 5 – 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chrom an-6-yl]oxy- 3,4-dihydro-1H-1,8-naphthyridin-2-one [0596] To a mixture of tert-butyl 2-[6-[(7-oxo-6,8-dihydro-5H-1,8-naphthyridin-4- yl)oxy]chroman-3-yl]-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin e-5-carboxylate (214mg, 0.41mmol) in DCM (2.5mL) was added TFA (0.5mL, 6.53mmol) and stirred at 25°C in a sealed vial for 1.5 hours. The mixture was concentrated and the residue azeotroped with toluene. The residue was dissolved in water and loaded onto an SCX-2 column. The column was washed with MeOH and then 2N NH 3 in methanol to elute the product. The filtrate was concentrated and dried to afford 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chrom an-6-yl]oxy- 3,4-dihydro-1H-1,8-naphthyridin-2-one (84mg, 0.20mmol, 49% yield) as a yellow solid. The compound was used in the next step without further purification. UPLC-MS (ES+, Short acidic): 0.97 min, m/z 418.1 [M+H] + . [0597] Step 6 – 5-[3-(5-isopropyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2- yl)chroman-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one [0598] A mixture of 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2-yl)chrom an-6- yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (65mg, 0.16mmol), acetone (22.9µL, 0.31mmol), sodium triacetoxyborohydride (66.1mg, 0.31mmol) and acetic acid (17.9µL, 0.31mmol) in DMF (1.5mL) was stirred at 25°C. Additional acetone (22.9µL, 0.31mmol) and sodium triacetoxyborohydride (66.1mg, 0.31mmol) were added after 23 hours and 30 hours and the mixture was further stirred at 25°C for 48 hours. The mixture was concentrated and the residue purified by reverse column chromatography using as eluent a gradient 0-40% (MeCN+0.1% formic acid) in (H 2 O+0.1% formic acid). Fractions containing the product were dried and loaded onto a SCX-2 column, which was flushed at first with MeOH and then 2N NH 3 in MeOH to elute 5-[3-(5-isopropyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2- yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (20.9mg, 0.046mmol, 29% yield) as a white solid. UPLC-MS (ES+, final purity): 2.05 min, m/z 460.2 [M+H] + . 1 H NMR (400MHz, DMSO-d 6 + 3 drops of CF 3 CO 2 D) δ/ppm: 8.03 (1H, d, J = 6.1Hz), 7.04 (1H, d, J = 2.7Hz), 6.97 (1H, dd, J = 8.8Hz, 2.7Hz), 6.91 (1H, d, J = 8.8Hz), 6.36 (1H, d, J = 6.1Hz), 4.50- 4.44 (2H, m), 4.44-4.37 (2H, m), 3.92-3.84 (1H, m), 3.83-3.68 (2H, m), 3.45-3.20 (3H, m), 3.07-2.91 (4H, m), 2.61-2.55 (2H, m), 1.33 (6H, d, J = 6.6Hz). Exchangeable protons not seen. [0599] Example 44. Synthesis of 5-[3-(5-acetyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2- yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (Compound 135) [0600] Compound 135 was synthesised in analogy with Example 43 with the exception of Step 6 as follows: To a solution of 5-[3-(4,5,6,7-tetrahydro-3H-imidazo[4,5-c]pyridin-2- yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (36mg, 0.09mmol) and triethylamine (12μL, 0.09mmol) in DMF (0.8mL) was added acetic anhydride (8.2μL, 0.09mmol) at 0°C and the mixture was stirred at this temperature for 2 hours. The mixture was concentrated and the residue purified by column chromatography using as eluent a gradient 0- 40% (MeCN+0.1% formic acid) in (H2O+0.1% formic acid). Fractions containing the product were concentrated and loaded onto a SCX-2 column, which was flushed at first with MeOH and then NH 3 2N in MeOH to give 5-[3-(5-acetyl-3,4,6,7-tetrahydroimidazo[4,5-c]pyridin-2- yl)chroman-6-yl]oxy-3,4-dihydro-1H-1,8-naphthyridin-2-one (15.5mg, 0.034mmol, 39% yield) as a white solid. UPLC-MS (ES+, final purity): 2.22 and 2.25 min, m/z 460.2 [M+H] + . 1H NMR (400MHz, DMSO-d 6 ) δ/ppm: 12.08 (1H, br s), 10.45 (1H, s), 7.95 (1H, d, J = 5.8Hz), 6.97 (1H, d, J = 2.6Hz), 6.90 (1H, dd, J = 8.8Hz, 2.6Hz), 6.86 (1H, d, J = 8.8Hz), 6.25 (1H, d, J = 5.8Hz), 4.46-4.31 (3H, m), 4.11-4.01 (1H, m), 3.75-3.63 (2H, m), 3.18-3.09 (1H, m), 3.09- 3.00 (1H, m), 2.95-2.89 (2H, m), 2.66-2.61 (1H, m), 2.56-2.51 (3H, m), 2.08 & 2.05 (3H, 2s, acetamide rotamers).1H missing (underneath water / DMSO peak). [0601] Example 45. Chiral Separation of Selected Compounds [0602] Conditions disclosed in table below can be used to separate stereoisomers of the racemic compound as shown: [0603] Example 46. Biological Assay [0604] HCT-116 AlphaLISA SureFire pERK1/2 Cellular Assay [0605] The human HCT-116 colorectal carcinoma cell line (ATCC CCL-247) endogenously expresses the KRAS G13D mutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. To determine whether compounds inhibit constitutive ERK phosphorylation in HCT-116 cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). Assay read outs took place 2 or 24 hours after dosing with compounds. On the first day, HCT-116 cells were harvested, resuspended in growth medium (McCoys5A with Glutamax (Life Technologies 36600021) and 10% heat-inactivated fetal bovine serum (Sigma F9665)), and counted. Cells were plated in 100 µl per well in each well of a 96-well culture dish (Sigma CLS3598) to a final density of 30,000 (2hr read) or 15,000 (24hr read) cells per well and incubated over night at 37°C and 5% CO 2 . On day 2, the growth medium was exchanged for dosing medium (McCoys5A with Glutamax (Life Technologies 36600021) and 1% heat- inactivated fetal bovine serum (Sigma F9665)) and the cells were dosed with compounds to produce a 10-point dose response, where the top concentration was 1 µM and subsequent concentrations were at 1/3 log dilution intervals. A matched DMSO control was included. The cells were subsequently incubated for either 2 or 24 hours at 37°C and 5% CO 2 . After incubation, media was removed and the cells were incubated with lysis buffer containing phosphatase inhibitors for 15 minutes at room temperature. Cell lysates were transferred to a ½ area 96 well white Optiplate TM (Perkin Elmer 6005569) and incubated with anti-mouse IgG acceptor beads, a biotinylated anti-ERK1/2 rabbit antibody recognizing both phosphorylated and non-phosphorylated ERK1/2, a mouse antibody targeted to the Thr202/Tyr204 epitope and recognizing phosphorylated ERK proteins only, and streptavidin-coated donor beads. The biotinylated antibody binds to the streptavidin-coated donor beads and the phopsho-ERK1/2 antibody binds to the acceptor beads. Plates were read on an EnVision reader (Perkin Elmer) and excitation of the beads at 680 nm with a laser induced the release of singlet oxygen molecules from the donor beads that trigger energy transfer to the acceptor beads in close proximity, producing a signal that can be measured at 570 nm. Both antibodies bound to phosphorylated ERK proteins, bringing the donor and acceptor beads into close proximity. All data were analyzed using the Dotmatics or GraphPad Prism software packages. Inhibition of ERK phosphorylation was assessed by determination of the absolute IC 50 value, which is defined as the concentration of compound required to decrease the level of phosphorylated ERK proteins by 50% when compared to DMSO control. [0606] WiDr AlphaLISA SureFire pERK1/2 Cellular Assay [0607] The human WiDr colorectal adenocarcinoma cell line (ATCC CCL-218) endogenously expresses the BRAF V600E mutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. To determine whether compounds inhibit constitutive ERK phosphorylation in WiDr cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). The main procedure is essentially the same as for HCT-116 cells (above), with the following adjustments to the growth medium (Eagle's Minimum Essential Medium (Sigma M2279) with 1x Glutamax (Life Technologies 35050038), 1x Sodium-Pyruvate (Sigma S8636), and 10% heat-inactivated fetal bovine serum (Sigma F9665)), the dosing medium (Eagle's Minimum Essential Medium (Sigma M2279) with 1x Glutamax (Life Technologies 35050038), 1x Sodium-Pyruvate (Sigma S8636), and 1% heat-inactivated fetal bovine serum (Sigma F9665)), and the seeding densities (2hr: 50,000 cells per well; 24hr: 35,000 cells per well). Moreover, the compounds were dosed in ½ log dilution intervals with the top concentration of 10 µM. [0608] HCT-116 AlphaLISA SureFire pERK1/2 Cellular Assay (Dimer) [0609] The human HCT-116 colorectal carcinoma cell line (ATCC CCL-247) endogenously expresses the KRAS G13D mutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. First generation RAF inhibitors can promote RAF dimer formation in KRAS mutant tumours leading to a paradoxical activation of the pathway. To determine whether compounds can circumvent this problem and inhibit RAF dimers in HCT- 116 cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p-T202/Y204 assay kit ALSU-PERK-A10K). The main procedure is essentially the same as described above, with the following adjustments: Cells were seeded with the seeding density of 30,000 cells per well. On the second day (the day of dosing) no medium change was performed and the cells were dosed with 1 µM of Encorafenib for 1 hour (at 37°C and 5% CO 2 ) to induce RAF dimers and promote paradoxical dimer-dependent pERK signalling. After incubation, the cells were washed, 100 µl fresh growth medium was added, and cells were dosed with compounds of interest to produce a 10-point dose response, where the top concentration was 10 µM and subsequent concentrations are at 1/2 log dilution intervals. Cells were incubated for another hour at 37°C and 5% CO 2 before lysis and processing with the pERK AlphaLISA® SureFire® kit as described above. [0610] A375 AlphaLISA SureFire pERK1/2 Cellular Assay (Monomer) [0611] The human A375 melanoma cell line (ATCC CRL-1619) endogenously expresses the BRAF V600E mutation, which leads to constitutive activation of the MAP kinase pathway and phosphorylation of ERK. In BRAF V600E mutant tumours, BRAF signals as a monomer to activate ERK. To determine whether compounds can inhibit BRAF monomers in A375 cells, they were tested using AlphaLISA® SureFire® technology (Perkin Elmer p-ERK1/2 p- T202/Y204 assay kit ALSU-PERK-A10K). The main procedure is essentially the same as described above for HCT-116 cells, with the following adjustments: The A375 cells were cultivated and dosed in Dulbecco's modified Eagle's medium containing 4.5 g/L D-glucose (Sigma D6546), 10% heat-inactivated fetal bovine serum (Sigma F9665), and 1% Sodium- Pyruvate (Sigma S8636), and seeded with a seeding density of 30,000 cells per well. No media exchange was performed before dosing with compounds to produce a 10-point dose response, where the top concentration was 10 µM and subsequent concentrations were at 1/2 log dilution intervals. Subsequently, the cells were incubated for 1 hour at 37°C and 5% CO 2 before lysis. [0612] HCT-116 CellTiter-Glo 3D Cell Proliferation Assay [0613] The human HCT-116 colorectal carcinoma cell line (ATCC CCL-247) endogenously expresses the KRAS G13D mutation, which leads to enhanced survival and proliferative signaling. To determine whether compounds inhibit the proliferation of HCT-116 cells, they are tested using the CellTiter-Glo® 3D Cell Viability Assay Kit (Promega G9683). On the first day, HCT-116 cells were harvested, resuspended in growth medium (McCoys5A with Glutamax (Life Technologies 36600021) with 10% heat-inactivated fetal bovine serum (Sigma F9665)), and counted. Cells were plated in 100 µl per well in each well of a Corning 700796- well clear round bottom Ultra-Low Attachment plate (VWR 444-1020) to a final density of 1000 cells per well. Cells were seeded for pre- and post-treatment readouts. The cells were then incubated at 37°C and 5% CO 2 for 3 days (72 hours) to allow spheroid formation. After 72 hours, the plate seeded for a pre-treatment read was removed from the incubator to allow equilibration to room temperature for 30 minutes, before CellTitre-Glo® reagent was added to each well. The plates were incubated at room temperature for 5 minutes shaking at 300 rpm, followed by an incubation of 25 minutes on the benchtop before being read on the Envision reader (Perkin Elmer) as described below. On the same day, the cells plated for the post- treatment readout were dosed with compounds to produce a 9-point dose response, where the top concentration was 15 µM and following concentrations were at 1/2 log dilution intervals. These cells were subsequently incubated at 37°C and 5% CO 2 for another 4 days (96 hours). After 4 days, the plate was removed from the incubator to allow equilibration to room temperature for 30 minutes and treated with CellTitre Glo® reagent as stated above. The method allows the quantification of ATP present in the wells, which is directly proportional to the amount of viable -hence metabolically active- cells in 3D cells cultures. The CellTitre Glo® reagent lyses the cells and contains luciferin and a luciferase (Ultra-Glo TM Recombinant Luciferase), which in the presence of ATP and oxygen can produce bioluminescence from luciferin. Therefore, plates were read on an EnVision reader (Perkin Elmer) and luminescence signals were recorded. Cell proliferation was determined on 4 days after dosing relative to the pre-treatment read. All data were analyzed using the Dotmatics or GraphPad Prism software packages. Inhibition of proliferation was assessed by determination of the GI50 value, which was defined as the concentration of compound required to decrease the level of cell proliferation by 50% when compared to DMSO control. [0614] WiDr CellTiter-Glo 3D Cell Proliferation Assay [0615] The human WiDr colorectal adenocarcinoma cell line (ATCC CCL-218) endogenously expresses the BRAF V600E mutation, which leads to enhanced survival and proliferative signaling. To determine whether compounds inhibit the proliferation of WiDr cells, they were tested using the CellTiter-Glo® 3D Cell Viability Assay Kit (Promega G9683) as stated for HCT-116 cells, with the following adjustments to the growth medium: Eagle's Minimum Essential Medium (Sigma M2279) with 1x Glutamax (Life Technologies 35050038), 1x Sodium-Pyruvate (Sigma S8636) and 10% heat-inactivated fetal bovine serum (Sigma F9665). [0616] Microsomal Stability Assay [0617] The stability studies were performed manually using the substrate depletion approach. Test compounds were incubated at 37 °C with cryo-preserved mouse or human liver microsomes (Corning) at a protein concentration of 0.5 mg.mL -1 and a final substrate concentration of 1 µM. Aliquots were removed from the incubation at defined timepoints and the reaction was terminated by adding to ice-cold organic solvent. Compound concentrations were determined by LC-MS/MS analysis. The natural log of the percentage of compound remaining was plotted against each time point and the slope determined. The half-life (t 1/2 ) and CL int were calculated using Equations 1 and 2, respectively. Data analysis was performed using Excel (Microsoft, USA). [0618] t 1/2 (min)=0.693/-slope (1) [0619] CL int (µL/min/mg) = (LN(2)/t1/2(min))*1000/microsomal protein (mg/mL) (2) [0620] HLM (human liver microsomes) and MLM (mouse liver microsomes) stability assay results are described in Tables 1-2. [0621] Plasma Protein Binding Assay [0622] The plasma protein binding was determined by the equilibrium dialysis method. A known concentration of compound (5 µM) in previously frozen human or mouse plasma (Sera Labs) was dialysed against phosphate buffer using a RED device (Life Technologies) for 4 hours at 37 °C. The concentration of compound in the protein containing (PC) and protein free (PF) sides of the dialysis membrane were determined by LC-MS/MS and the %free compound was determined by equation 4. Data analysis was performed using Excel (Microsoft, USA). [0623] % free = (1-((PC-PF)/PC)) x 100 (4) [0624] hPPB (human plasma protein binding) and mPPB (mouse plasma protein binding) results are described in Tables 1-2. [0625] FeSSIF Solubility Assay [0626] 1 mL of fed state simulated intestinal fluid (FeSSIF), prepared using FaSSIF/FeSSIF/FaSSGF powder (Biorelevant.com) and pH 5 acetate buffer, was added to 1.0 mg of compound and then incubated for 24 h (Bioshake iQ, 650 rpm, 37 °C). Following filtration under positive pressure, the concentration of compound in solution was assessed by LC-UV in comparison to the response for a calibration standard of known concentration (250 μM). FeSSIF solubility results are described in Tables 1-2.

[0629] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. [0630] While the invention has been described in connection with proposed specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth and as follows in the scope of the appended claims.