Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR MODULATING SPLICING
Document Type and Number:
WIPO Patent Application WO/2020/163541
Kind Code:
A1
Abstract:
Described herein are small molecule splicing modulator compounds that modulate splicing of mRNA, such as pre-mRNA, encoded by genes, and methods of use of the small molecule splicing modulator compounds for modulating splicing and treating diseases and conditions.

Inventors:
LUZZIO MICHAEL (US)
LUCAS BRIAN (US)
Application Number:
PCT/US2020/016894
Publication Date:
August 13, 2020
Filing Date:
February 05, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SKYHAWK THERAPEUTICS INC (US)
International Classes:
A61K31/46; A61K31/501; C07D237/20
Domestic Patent References:
WO2017100726A12017-06-15
WO2014028459A12014-02-20
Foreign References:
US20180170923A12018-06-21
Other References:
See also references of EP 3920926A4
Attorney, Agent or Firm:
OH, Young-In (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

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

wherein,

A is -CRA=CRA-;

E is–NR-, -O-, -S-, -S(=O)-, -S(=O)2-, or -S(=O)(=NRE)-;

RE is hydrogen, substituted or unsubstituted C1-C3 alkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted C2-C3 alkynyl;

each RA is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR1, –SR1, -S(=O)R1, -S(=O)2R1, substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C4 cycloalkyl, and substituted or unsubstituted C2–C3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR3-;

Z is CR2;

W is substituted or unsubstituted C1-C3 alkylene, substituted or unsubstituted C1–C2 heteroalkylene, substituted or unsubstituted C3–C8 cycloalkylene, substituted or unsubstituted C2–C7

heterocycloalkylene, or substituted or unsubstituted C2-C3 alkenylene;

R is hydrogen;

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R2 is hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, or substituted or unsubstituted C1–C4 haloalkyl;

R3 is hydrogen, -CN, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, -C1-C4 alkylene-OR1, substituted or unsubstituted C3–C4 cycloalkyl, or substituted or unsubstituted C2–C3 heterocycloalkyl; each R11, R12, R13, R14, R16, and R17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR1, substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C1–C4 heteroalkyl;

R15 and R18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1B.

2. A compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CRA=CRA-;

each RA is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR1, –SR1, -S(=O)R1, -S(=O)2R1, substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C4 cycloalkyl, and substituted or unsubstituted C2–C3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR3-;

Z is CR2;

W is substituted or unsubstituted C1-C3 alkylene, substituted or unsubstituted C1–C2 heteroalkylene, substituted or unsubstituted C3–C8 cycloalkylene, substituted or unsubstituted C2–C7

heterocycloalkylene, or substituted or unsubstituted C2-C3 alkenylene;

R is substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 fluoroalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, or substituted or unsubstituted C2–C5 heterocycloalkyl;

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R2 is hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, or substituted or unsubstituted C1–C4 haloalkyl;

R3 is hydrogen, -CN, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, -C1-C4 alkylene-OR1, substituted or unsubstituted C3–C4 cycloalkyl, or substituted or unsubstituted C2–C3 heterocycloalkyl;

each R11, R12, R13, R14, R16, and R17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR1, substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C1–C4 heteroalkyl;

R15 and R18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1D.

3. A compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CRA=CRA-;

each RA is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR1, –SR1, -S(=O)R1, -S(=O)2R1, substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C4 cycloalkyl, and substituted or unsubstituted C2–C3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR3-;

Z is CR2;

W is substituted or unsubstituted C1-C3 alkylene, substituted or unsubstituted C1–C2 heteroalkylene, substituted or unsubstituted C3–C8 cycloalkylene, substituted or unsubstituted C2–C7

heterocycloalkylene, or substituted or unsubstituted C2-C3 alkenylene;

R is substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 fluoroalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, or substituted or unsubstituted C2–C5 heterocycloalkyl; each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R2 is hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, or substituted or unsubstituted C1–C4 haloalkyl;

R3 is hydrogen, -CN, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, -C1-C4 alkylene-OR1, substituted or unsubstituted C3–C4 cycloalkyl, or substituted or unsubstituted C2–C3 heterocycloalkyl;

each R11, R12, R13, R14, R16, and R17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR1, substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C1–C4 heteroalkyl;

R15 and R18 are the same and selected from the group consisting of F,–OR1, substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C1–C4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1F.

4. A compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CRA=CRA-;

each RA is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR1, –SR1, -S(=O)R1, -S(=O)2R1, substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C4 cycloalkyl, and substituted or unsubstituted C2–C3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR3-;

Z is CR2; W is substituted or unsubstituted C1-C3 alkylene, substituted or unsubstituted C1–C2 heteroalkylene, substituted or unsubstituted C3–C8 cycloalkylene, substituted or unsubstituted C2–C7

heterocycloalkylene, or substituted or unsubstituted C2-C3 alkenylene;

R is substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 fluoroalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, or substituted or unsubstituted C2–C5 heterocycloalkyl;

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R2 is hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, or substituted or unsubstituted C1–C4 haloalkyl;

R3 is hydrogen, -CN, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, -C1-C4 alkylene-OR1, substituted or unsubstituted C3–C4 cycloalkyl, or substituted or unsubstituted C2–C3 heterocycloalkyl;

each R11, R12, R13, R14, R16, and R17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR1, substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C1–C4 heteroalkyl;

R15 and R18 are not the same and selected from the group consisting of hydrogen, deuterium, F,–OR1, substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C1–C4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1.

5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein the compound of Formula (I) has the structure of Formula (Ia):

6. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein the compound of Formula (I) has the structure of Formula (Ib):

7. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein the compound of Formula (I) has the structure of Formula (Ic):

8. The compound of any one of claims 1–7, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is substituted or unsubstituted aryl.

9. The compound of claim 1-8, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is 2–hydroxy–phenyl substituted with 1, 2, or 3 substituents independently selected from:

deuterium, halogen, hydroxy, nitro, cyano,–SR1,–S(=O)R1,–S(=O)2R1,–N(R1)2,–C(=O)R1,–

OC(=O)R1,–C(=O)OR1,–C(=O)N(R1)2, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted C2-C7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;

wherein

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C6 alkyl, -CD3, substituted or unsubstituted C1–C6 haloalkyl, substituted or unsubstituted C1–C6 heteroalkyl, substituted or unsubstituted C3–C8 cycloalkyl, substituted or unsubstituted C2–C7 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

10. The compound of claim 9, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

11. The compound of claim 10, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted aryl, wherein if aryl is substituted then it is substituted with 1 or 2 substituents independently selected from:

deuterium, halogen, -OH, -NO2, -CN,–SR1,–S(=O)R1,–S(=O)2R1,–N(R1)2,–C(=O)R1,–OC(=O)R1,– C(=O)OR1,–C(=O)N(R1)2, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2- C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C7 cycloalkyl, and substituted or unsubstituted C2-C7

heterocycloalkyl; wherein

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

12. The compound of claim 10, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted heteroaryl, wherein if heteroaryl is substituted then it is substituted with 1 or 2 substituents independently selected from:

deuterium, halogen, -OH, -NO2, -CN,–SR1,–S(=O)R1,–S(=O)2R1,–N(R1)2,–C(=O)R1,–OC(=O)R1,– C(=O)OR1,–C(=O)N(R1)2, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2- C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C7 cycloalkyl, and substituted or unsubstituted C2-C7

heterocycloalkyl; wherein

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

13. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein ring Q is substituted or unsubstituted heteroaryl.

14. The compound of claim 13, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is substituted or unsubstituted 5- or 6-membered monocyclic heteroaryl.

15. The compound of claim 13, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is substituted or unsubstituted 6-membered monocyclic heteroaryl.

16. The compound of claim 13-15, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is 6-membered monocyclic heteroaryl selected from:

and wherein each RQ is independently

selected from hydrogen, deuterium,–F,–Cl, -CN,–OH, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2,– CF3,–OCH3, -OCH2CH3, -CH2OCH3, -OCH2CH2CH3, and -OCH(CH3)2; and ring P is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

17. The compound of any one of claims 1–7, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein:

ring wherein each RQ is independently selected from the group consisting of hydrogen, deuterium,–F,–Cl, -CN,–OH, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2,–CF3,–OCH3, - OCH2CH3, -CH2OCH3, -OCH2CH2CH3, and -OCH(CH3)2; and ring P is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

18. The compound of claim 16 or 17, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RQ is independently selected from the group consisting of hydrogen,–F,–Cl, -CN,–OH, -CH3,–CF3, and–OCH3.

19. The compound of any one of claims 16-18, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring P is substituted or unsubstituted heteroaryl.

20. The compound of any one of claims 16-19, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring P is heteroaryl selected from the group consisting of:

each RB is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C1-C6 alkyl, -CD3, substituted or unsubstituted C1–C6 fluoroalkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, deuterium substituted C1-C6 alkoxy, -OCD3, substituted or unsubstituted C3–7 cycloalkyl, substituted or unsubstituted C2–C7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;

RB1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C1-C6 alkyl, -CD3, substituted or unsubstituted C1–C6 fluoroalkyl, substituted or unsubstituted C1-C6 heteroalkyl, substituted or unsubstituted C3–7 cycloalkyl, and substituted or unsubstituted C2–C7 heterocycloalkyl; and

m is 1, 2, or 3.

21. The compound of any one of claims 16-18, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring P is heteroaryl selected from the group consisting of:

wherein,

each RB is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C1-C6 alkyl, -CD3, substituted or unsubstituted C1–C6 fluoroalkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, deuterium substituted C1-C6 alkoxy, -OCD3, substituted or unsubstituted C3–7 cycloalkyl, substituted or unsubstituted C2–C7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl;

RB1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C1-C6 alkyl, -CD3, substituted or unsubstituted C1–C6 fluoroalkyl, substituted or unsubstituted C1-C6 heteroalkyl, substituted or unsubstituted C3–7 cycloalkyl, and substituted or unsubstituted C2–C7 heterocycloalkyl; and

m is 1, 2, or 3.

22. The compound of claim 20 or 21, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RB is independently hydrogen, deuterium,–F,–Cl, -CN,–CH3,–CF3,–OH, or–OCH3.

23. The compound of claim 22, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RB is independently–F or–OCH3.

24. The compound of any one of claims 20-23, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein RB1 is hydrogen, deuterium,–CH3,–CF3, or–CD3.

25. The compound of any one of claims 20–24, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein m is 1.

26. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein ring Q is 2–naphthyl substituted at the 3 position with 0, 1, and 2 substituents independently selected from:

deuterium, halogen, -OH, -NO2, -CN,–SR1,–S(=O)R1,–S(=O)2R1,–N(R1)2,–C(=O)R1,–OC(=O)R1,– C(=O)OR1,–C(=O)N(R1)2, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2- C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted C2-C7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; wherein

each R1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C2–C5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

27. The compound of any one of claims 1–7, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is selected from the group consisting of:

28. The compound of any one of claims 1–7, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is selected from the group consisting of:

29. The compound of any one of claims 1–7, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is selected from the group consisting of:

30. The compound of any one of claims 1–7, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein ring Q is selected from the group consisting of:

RB1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C1-C6 alkyl, -CD3, substituted or unsubstituted C1–C6 fluoroalkyl, substituted or unsubstituted C1-C6 heteroalkyl, substituted or unsubstituted C3–7 cycloalkyl, and substituted or unsubstituted C2–C7 heterocycloalkyl. 31. The compound of any one of claims 1–30, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted C1-C3 alkylene.

32. The compound of claim 31, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is–CH2-.

33. The compound of claim 31, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is–CH2CH2-.

34. The compound of claim 31, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is–CH2CH2CH2-.

35. The compound of any one of claims 1–30, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted C3–C8 cycloalkylene or substituted or unsubstituted C2-C3 alkenylene.

36. The compound of claim 35, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted C3–C8 cycloalkylene.

37. The compound of claim 36, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted cyclopropylene.

38. The compound of claim 35, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted C2-C3 alkenylene.

39. The compound of claim 38, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is–CH=CH-.

40. The compound of any one of claims 1–30, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted C1-C2 heteroalkylene.

41. The compound of claim 40, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein W is substituted or unsubstituted–CH2OCH2–.

42. The compound of any one of claims 1–41, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each R16 and R17 is selected from the group consisting of hydrogen, F, –OR1, substituted or unsubstituted C1–C6 alkyl, a substituted or unsubstituted C1–C6 fluoroalkyl, and substituted or unsubstituted C1–C6 heteroalkyl.

43. The compound of any one of claims 1–41, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each R16 and R17 is selected from the group consisting of hydrogen, F, -OH, -OCH3, -OCH2CH3, -OCH2CH2OH, -OCH2CN, -OCF3, -CH3, -CH2CH3, -CH2OH, -CH2CH2OH, - CH2CN, -CH2F, -CHF2, -CF3, -CH2CH2F, -CH2CHF2, and -CH2CF3.

44. The compound of any one of claims 1–41 or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein each R16 and R17 is selected from the group consisting of hydrogen, F, -OH, -OCH3, -OCF3, -CH3, -CH2OH, -CH2F, -CHF2, and -CF3.

45. The compound of any one of claims 1–44, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R16 is hydrogen.

46. The compound of any one of claims 1–45, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R17 is hydrogen.

47. The compound of any one of claims 1–46, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R2 is hydrogen.

48. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 fluoroalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C5 cycloalkyl, or substituted or unsubstituted C2–C4 heterocycloalkyl.

49. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2, -CH2OH, - CH2CH2OH, -CH2CH2CH2OH, -C(OH)(CH3)2, -CH2CN, -CH2C(=O)OCH3, -CH2C(=O)OCH2CH3, - CH2C(=O)NHCH3, -CH2C(=O)N(CH3)2, -CH2NH2, -CH2NHCH3, -CH2N(CH3)2, -CH2F, -CHF2, -CF3, cyclopropyl, cyclobutyl, oxetanyl, aziridinyl, or azetidinyl.

50. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2, -CH2OH, - CH2CH2OH, -CH2CH2CH2OH, -CH2CN, -CH2F, -CHF2, -CF3, cyclopropyl, or oxetanyl.

51. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is -CH3, -CH2CH3, -CH2OH, -CH2CH2OH, -CH2CN, -CH2F, -CHF2, -CF3, cyclopropyl, or oxetanyl.

52. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is -CH3, -CH2CH3, -CH2OH, -CH2CH2OH, -CH2CN, cyclopropyl, or oxetanyl.

53. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is -CH3, -CH2OH, -CH2CN, -CHF2, -CF3, or cyclopropyl.

54. The compound of any one of claims 1–47, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R is -CH3, -CH2CH3, -CH2F, -CHF2, -CF3, cyclopropyl, or oxetanyl. 55. The compound of any one of claims 1-54, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein R15 and R18 are the same and selected from the group consisting of F, –OR1, substituted or unsubstituted C1–C3 alkyl, substituted or unsubstituted C1–C3 fluoroalkyl, and substituted or unsubstituted C1–C3 heteroalkyl.

56. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are the same and selected from the group consisting of F, - CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2, -CH2OH, -CH2CH2OH, -CH2NHCH3, -CH2N(CH3)2, -OH, - OCH3, -OCH2CH3, -OCH2CH2OH, -OCH2CN, -OCF3, -CH2F, -CHF2, and -CF3.

57. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are the same and selected from the group consisting of F, - CH3, -CH2OH, -OCH2CN, -OH, -OCH3, -OCH2CN, -OCF3, -CH2F, -CHF2, and -CF3.

58. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are the same and selected from the group consisting of F, - CH3, -OCH3, -OCF3, -CH2F, -CHF2, and -CF3.

59. The compound of any one of claims 1–54, wherein R15 and R18 are F.

60. The compound of any one of claims 1–54, wherein R15 and R18 are -CH3.

61. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are not the same and selected from hydrogen, deuterium, F, -CH3, -CH2CH3, -CH2CH2CH3, -CH(CH3)2, -CH2OH, -CH2CH2OH, -CH2NHCH3, -CH2N(CH3)2, -OH, - OCH3, -OCH2CH3, -OCH2CH2OH, -OCH2CN, -OCF3, -CH2F, -CHF2, and -CF3.

62. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH3, -CH2OH, -OCH2CN, -OH, -OCH3, -OCH2CN, -OCF3, -CH2F, -CHF2, and - CF3.

63. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH3, -OCH3, -OCF3, -CH2F, -CHF2, and -CF3.

64. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 and R18 are not the same and selected from the group consisting of hydrogen, F, -CH3, and -OCH3.

65. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 is hydrogen and R18 is -CH3.

66. The compound of any one of claims 1–54, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein R15 is -CH3 and R18 is hydrogen.

67. The compound of any one of claims 1-63, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein R3 is hydrogen, -CN,–OR1,–N(R1)2, substituted or unsubstituted C1– C4 alkyl, -CD3, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, -C1-C4 alkylene-OR1, or substituted or unsubstituted C3–C5 cycloalkyl.

68. The compound of any one of claims 1-63, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein R3 is substituted or unsubstituted C1–C4 alkyl, -C1-C4 alkylene-OR1, or substituted or unsubstituted C3–C5 cycloalkyl.

69. The compound of any one of claims 1-63, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein R3 is -CH3, -CH2CH3, cyclopropyl, or–CH2CH2OCH3.

70. The compound of any one of claims 1-63, or a pharmaceutically acceptable salt or pharmaceutically

acceptable solvate thereof, wherein R3 is -CH3.

71. The compound of any one of claims 1–70, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein A is -CRA=CRA-.

72. The compound of claim 71, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RA is independently hydrogen, F, Cl, -CN, -CH3, -CH2CH3, -CH2CH2CH3, - CH(CH3)2, -OH, -OCH3, -OCH2CH3, -OCF3, -CH2F, -CHF2, or -CF3.

73. The compound of claim 71, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RA is independently hydrogen, F, Cl, -CN, -CH3, -OH, -OCH3, -OCF3, -CH2F, - CHF2, or -CF3.

74. The compound of claim 71, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RA is independently hydrogen, F, Cl, -CN, -CH3, or -OCH3.

75. The compound of claim 71, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein each RA is independently hydrogen, F, Cl, or -CH3.

76. The compound of claim 71, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein RA is hydrogen.

77. The compound of any one of claims 1-76, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein at least one of R11, R12, R13, R14, R15, R16, R17, and R18 is F.

78. The compound of any one of claims 1-76, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein at least one of R11, R12, R13, R14, R15, R16, R17, and R18 comprises a fluorine.

79. A compound, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, wherein the compound is selected from Table 1A, Table 1C, Table 1E, Table 1G, or Table 1H.

80. A compound of claim 2, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, that is:

81. A compound of claim 3, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, that is:

82. A compound of claim 4, or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, that is:

83. A pharmaceutical composition comprising a compound of any one of claims 1-82 or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, and a pharmaceutically acceptable excipient or carrier.

84. A method of treating a condition or disease comprising administering a compound of any one of claims 1- 82 or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof to a subject in need thereof.

85. Use of a compound of any one of claims 1-82 or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof in the manufacture of a medicament for the treatment of a condition or disease.

Description:
METHODS AND COMPOSITIONS FOR MODULATING SPLICING CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of U.S. Provisional Application No.62/801,475 filed on February 5, 2019, U.S. Provisional Application No.62/801,381 filed on February 5, 2019, U.S. Provisional Application No.62/801,386 filed on February 5, 2019, and U.S. Provisional Application No.62/801,390 filed on February 5, 2019 the disclosures of which are hereby incorporated by reference in their entirety.

BACKGROUND

[0002] The majority of protein-coding genes in the human genome are composed of multiple exons (coding regions) that are separated by introns (non-coding regions). Gene expression results in a single precursor messenger RNA (pre-mRNA). The intron sequences are subsequently removed from the pre-mRNA by a process called splicing, which results in the mature messenger RNA (mRNA). By including different combinations of exons, alternative splicing gives rise to multiple mRNAs encoding distinct protein isoforms. The spliceosome, an intracellular complex of multiple proteins and ribonucleoproteins, catalyzes splicing.

[0003] Current therapeutic approaches to direct and control mRNA expression require methods such as gene therapy, genome editing, or a wide range of oligonucleotide technologies (antisense, RNAi, etc.). Gene therapy and genome editing act upstream of transcription of mRNA by influencing the DNA code and thereby changing mRNA expression. Oligonucleotides modulate the action of RNA via canonical base/base hybridization. The appeal of this approach is in the design of the basic pharmacophore of an oligonucleotide, which can be defined in a straightforward fashion by known base pairing to the target sequence subject. Each of these therapeutic modalities suffers from substantial technical, clinical, and regulatory challenges. Some limitations of oligonucleotides as therapeutics (e.g., antisense, RNAi) include unfavorable pharmacokinetics, lack of oral bioavailability, and lack of blood-brain-barrier penetration, with the latter precluding delivery to the brain or spinal cord after parenteral drug administration for the treatment of diseases (e.g., neurological diseases, brain cancers). In addition, oligonucleotides are not taken up effectively into solid tumors without a complex delivery system such as lipid nanoparticles. Further, most of the oligonucleotides taken up into cells and tissues remain in non-functional compartments (e.g., endosomes) and does not gain access to the cytosol and/or nucleus where the target is located.

[0004] Additionally, to anneal to a target, oligonucleotide therapies require access to complementary base pairs of the target. This approach assumes that pre-mRNA sequences exist as a linear strand of RNA in the cell. However, pre-mRNA is rarely linear; it has complex secondary and tertiary structure. Further, cis-acting elements (e.g., protein binding elements) and trans-acting factors (e.g., splicing complex components) can create additional two-dimensional and three-dimensional complexity (e.g., by binding to the pre-mRNA). These features can be potency-and efficacy-limiting for oligonucleotide therapies. INCORPORATION BY REFERENCE

[0005] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. SUMMARY

[0006] The novel small molecule splicing modulators (SMSMs) described herein do not suffer from the limitations above, nor the structural and steric hindrances that greatly limit oligonucleotide therapies (e.g., by blocking hybridization to pre-mRNA targets). Small molecules have been essential in uncovering the mechanisms, regulations, and functions of many cellular processes, including DNA replication, transcription, and translation. While several recent reports have described screens for small molecule effectors of splicing, only a small number of constitutive or alternative splicing modulators have been identified and many of the small-molecule inhibitors lack specificity, lack selectivity, lack potency, exhibit toxicity, or are not orally available. Targeting the RNA transcriptome with small-molecule modulators represents an untapped therapeutic approach to treat a variety of RNA-mediated diseases. Accordingly, there remains a need to develop small- molecule RNA modulators useful as therapeutic agents. There is need in the art for novel modulators of splicing or splicing-dependent processes. Provided herein are small molecule splicing modulators and uses thereof that fulfill this need.

[0007] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

E is–NR-, -O-, -S-, -S(=O)-, -S(=O) 2 -, or -S(=O)(=NR E )-;

R E is hydrogen, substituted or unsubstituted C 1 -C 3 alkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted C 2 -C 3 alkenyl, or substituted or unsubstituted C 2 -C 3 alkynyl;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl; X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is hydrogen;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1B.

[0008] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

E is–NR-, -O-, -S-, -S(=O)-, -S(=O) 2 -, or -S(=O)(=NR E )-;

R E is hydrogen, substituted or unsubstituted C 1 -C 3 alkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted C 2 -C 3 alkenyl, or substituted or unsubstituted C 2 -C 3 alkynyl; each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is hydrogen;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C3–C4 cycloalkyl, or substituted or unsubstituted C2–C3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1B.

[0009] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7

heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, or substituted or

unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1D.

[0010] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof: wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C1-C3 alkylene, substituted or unsubstituted C1–C2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7

heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or

unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0; c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1D.

[0011] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C1-C3 alkylene, substituted or unsubstituted C1–C2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl; each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are the same and selected from the group consisting of F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1F.

[0012] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are the same and selected from the group consisting of F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1F.

[0013] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene; R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1.

[0014] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl; ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1.

[0015] In some embodiments, the compound of Formula (I) has the structure of Formula (Ia):

[0016] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib):

[0017] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib1):

[0018] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib2):

[0019] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib3):

[0020] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib4):

[0021] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic):

[0022] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic1):

[0023] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic2):

[0024] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic3):

[0025] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic4):

[0026] In some embodiments, ring Q is substituted or unsubstituted aryl. In some embodiments, ring Q is 2– hydroxy–phenyl substituted with 1, 2, or 3 substituents independently selected from: deuterium, halogen, hydroxy, nitro, cyano,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,–C(=O)R 1 ,–OC(=O)R 1 ,–C(=O)OR 1 ,– C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, substituted or unsubstituted C 2 -C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 haloalkyl, substituted or unsubstituted C 1 – C 6 heteroalkyl, substituted or unsubstituted C 3 –C 8 cycloalkyl, substituted or unsubstituted C 2 –C 7

heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0027] In some embodiments, ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. In some embodiments, ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted aryl, wherein if aryl is substituted then it is substituted with 1 or 2 substituents independently selected from: deuterium, halogen, -OH, -NO 2 , -CN,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,– C(=O)R 1 ,–OC(=O)R 1 ,–C(=O)OR 1 ,–C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, and substituted or unsubstituted C 2 -C 7 heterocycloalkyl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0028] In some embodiments, ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted heteroaryl, wherein if heteroaryl is substituted then it is substituted with 1 or 2 substituents independently selected from: deuterium, halogen, -OH, -NO 2 , -CN,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,–C(=O)R 1 ,– OC(=O)R 1 ,–C(=O)OR 1 ,–C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, and substituted or unsubstituted C 2 -C 7 heterocycloalkyl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 – C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0029] In some embodiments, ring Q is substituted or unsubstituted heteroaryl. In some embodiments, ring Q is substituted or unsubstituted 5- or 6-membered monocyclic heteroaryl. In some embodiments, ring Q is substituted or unsubstituted 6-membered monocyclic heteroaryl.

[0030] In some embodiments, ring Q is 6-membered monocyclic heteroaryl selected from:

consisting of hydrogen, deuterium,–F,–Cl, -CN,–OH, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 ,–CF 3, – OCH 3 , -OCH 2 CH 3 , -CH 2 OCH 3 , -OCH 2 CH 2 CH 3 , and -OCH(CH 3 ) 2 ; and ring P is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. [0031] In some embodiments, ring Q is , wherein each R Q is independently selected from the group consisting of hydrogen, deuterium,–F,–Cl, -CN,–OH, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , - CH(CH 3 ) 2 ,–CF 3, –OCH 3 , -OCH 2 CH 3 , -CH 2 OCH 3 , -OCH 2 CH 2 CH 3 , and -OCH(CH 3 ) 2 ; and ring P is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

[0032] In some embodiments, R Q is independently selected from the group consisting of hydrogen,–F,–Cl, - CN,–OH, -CH 3 ,–CF 3, and–OCH 3 .

[0033] In some embodiments, ring P is substituted or unsubstituted heteroaryl.

[0034] In some embodiments, ring P is heteroaryl selected from the group consisting of:

; wherein, each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl; and m is 0, 1, 2, or 3.

[0035] In some embodiments, ring P is heteroaryl selected from the group consisting of:

wherein, each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl; and m is 0, 1, 2, or 3.

[0036] In some embodiments, each R B is independently hydrogen, deuterium,–F,–Cl, -CN,–CH 3 ,–CF 3, – OH, or–OCH 3 . In some embodiments, each R B is independently–F or–OCH 3 . In some embodiments, R B is hydrogen. In some embodiments, R B is–OCH 3 . In some embodiments, R B is–CH 3 .

[0037] In some embodiments, R B1 is hydrogen, deuterium,–CH 3 ,–CF 3, or–CD 3 .

[0038] In some embodiments, m is 1, 2, or 3. In some embodiments, m is 0 or 1. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3.

[0039] In some embodiments, ring Q is 2–naphthyl substituted at the 3 position with 0, 1, and 2 substituents independently selected from: deuterium, halogen, -OH, -NO 2 , -CN,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,– C(=O)R 1 ,–OC(=O)R 1 ,–C(=O)OR 1 ,–C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, substituted or unsubstituted C 2 -C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0040] In some embodiments, ring Q is selected from the group consisting of:

[0041] In some embodiments, ring Q is selected from the group consisting of:

[0042] In some embodiments, ring Q is selected from the group consisting of:

[0043] In some embodiments, ring Q is selected from the group consisting of:

consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or

unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl.

[0044] In some embodiments, W is substituted or unsubstituted C 1 -C 2 alkylene. In some embodiments, W is C 1 -C 2 alkylene substituted with 1, 2, 3, or 4 substituents each independently selected from F, -OH, -OCH 3 , and -CH 3 . In some embodiments, W is–CH 2 –,–CHF–,–CH(CH 3 )–,–CH(OH)–,–CH(OCH 3 )–,–CF 2 –,– CH 2 CH 2 –,–CHFCH 2 –,–CH 2 CHF–,–CH(CH 3 )CH 2 –,–CH 2 CH(CH 3 )–,–CH(OH)CH 2 –,–CH 2 CH(OH)–,– CH(OCH 3 )CH 2 –,–CH 2 CH(OCH 3 )–,–CF 2 CH 2 –, or–CH 2 CF 2 –. In some embodiments, W is–CHFCH 2 –,– CH 2 CHF–,–CF 2 CH 2 –, or–CH 2 CF 2 –.

[0045] In some embodiments, W is substituted or unsubstituted C 3 -C 4 alkylene. In some embodiments, W is C 3 -C 4 alkylene substituted with 1, 2, 3, or 4 substituents each independently selected from the group consisting of F, -OH, -OCH 3 , and -CH 3 . In some embodiments, W is–CH 2 CH 2 CH 2 –,–CHFCH 2 CH 2 –,– CH 2 CHFCH 2 –,–CH 2 CH 2 CHF–,–CF 2 CH 2 CH 2 –,–CH 2 CF 2 CH 2 –,–CH 2 CH 2 CF 2 –,–CH(OH)CH 2 CH 2 –,– CH 2 CH(OH)CH 2 –,–CH 2 CH 2 CH(OH)–,–CH(OCH 3 )CH 2 CH 2 –,–CH 2 CH(OCH 3 )CH 2 –,–CH 2 CH 2 CH(OCH 3 )–, ––CH(CH 3 )CH 2 CH 2 –,–CH 2 CH(CH 3 )CH 2 –, or–CH 2 CH 2 CH(CH 3 )–. In some embodiments, W is–

CH 2 CHFCH 2 – or–CH 2 CF 2 CH 2 –. In some embodiments, W is–CHFCH 2 CH 2 – or–CF 2 CH 2 CH 2 –.

[0046] In some embodiments, W is–CH 2 CH 2 CHF– or–CH 2 CH 2 CF 2 –.

[0047] In some embodiments, W is substituted or unsubstituted C 1 -C 3 alkylene. In some embodiments, W is– CH 2 -. In some embodiments, W is–CH 2 CH 2 -. In some embodiments, W is–CH 2 CH 2 CH 2 -. In some embodiments, W is substituted or unsubstituted C 3 –C 8 cycloalkylene or substituted or unsubstituted C 2 -C 3 alkenylene. In some embodiments, W is substituted or unsubstituted C 3 –C 8 cycloalkylene. In some embodiments, W is substituted or unsubstituted cyclopropylene. In some embodiments, W is substituted or unsubstituted C 2 -C 3 alkenylene. In some embodiments, W is–CH=CH-. In some embodiments, W is substituted or unsubstituted C 1 -C 2 heteroalkylene. In some embodiments, W is substituted or unsubstituted– CH 2 OCH 2 –.

[0048] In some embodiments, each R 16 and R 17 is selected from the group consisting of hydrogen, F,–OR 1 , substituted or unsubstituted C 1 –C 6 alkyl, a substituted or unsubstituted C 1 –C 6 fluoroalkyl, and substituted or unsubstituted C 1 –C 6 heteroalkyl. In some embodiments, each R 16 and R 17 is selected from the group consisting of hydrogen, F, -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, -OCH 2 CN, -OCF 3 , -CH 3 , -CH 2 CH 3 , -CH 2 OH, - CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , and -CH 2 CF 3 . In some embodiments, each R 16 and R 17 is selected from the group consisting of hydrogen, F, -OH, -OCH 3 , -OCF 3 , -CH 3 , -CH 2 OH, - CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 16 is hydrogen. In some embodiments, R 17 is hydrogen.

[0049] In some embodiments, R 2 is hydrogen.

[0050] In some embodiments, R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 – C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 5 cycloalkyl, or substituted or unsubstituted C 2 –C 4 heterocycloalkyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , - CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH 2 CH 2 OH, -C(OH)(CH 3 ) 2 , -CH 2 CN, - CH 2 C(=O)OCH 3 , -CH 2 C(=O)OCH 2 CH 3 , -CH 2 C(=O)NHCH 3 , -CH 2 C(=O)N(CH 3 ) 2 , -CH 2 NH 2 , -CH 2 NHCH 3 , - CH 2 N(CH 3 ) 2 , -CH 2 F, -CHF 2 , -CF 3 , cyclopropyl, cyclobutyl, oxetanyl, aziridinyl, or azetidinyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH 2 CH 2 OH, - CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , cyclopropyl, or oxetanyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , cyclopropyl, or oxetanyl. In some embodiments, R is -CH 3 , - CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, cyclopropyl, or oxetanyl. In some embodiments, R is -CH 3 , - CH 2 OH, -CH 2 CN, -CHF 2 , -CF 3 , or cyclopropyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , -CH 2 F, -CHF 2 , - CF 3 , cyclopropyl, or oxetanyl.

[0051] In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F,–OR 1 , substituted or unsubstituted C 1 –C 3 alkyl, substituted or unsubstituted C 1 –C 3 fluoroalkyl, and substituted or unsubstituted C 1 –C 3 heteroalkyl. In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 NHCH 3 , - CH2N(CH3)2, -OH, -OCH3, -OCH2CH3, -OCH2CH2OH, -OCH2CN, -OCF3, -CH2F, -CHF2, and -CF3. In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F, -CH 3 , -CH 2 OH, - OCH 2 CN, -OH, -OCH 3 , -OCH 2 CN, -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F, -CH 3 , -OCH 3 , -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are F. In some embodiments, R 15 and R 18 are -CH 3 .

[0052] In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 NHCH 3 , - CH 2 N(CH 3 ) 2 , -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, -OCH 2 CN, -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH 3 , -CH 2 OH, -OCH 2 CN, -OH, -OCH 3 , -OCH 2 CN, -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH 3 , -OCH 3 , -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, F, -CH 3 , and -OCH 3 . In some embodiments, R 15 is hydrogen and R 18 is -CH 3 . In some embodiments, R 15 is -CH 3 and R 18 is hydrogen.

[0053] In some embodiments, R 3 is hydrogen, -CN,–OR 1 ,–N(R 1 ) 2 , substituted or unsubstituted C 1 –C 4 alkyl, - CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , or substituted or unsubstituted C 3 –C 5 cycloalkyl. In some embodiments, R 3 is substituted or unsubstituted C 1 –C 4 alkyl, -C 1 -C 4 alkylene-OR 1 , or substituted or unsubstituted C 3 –C 5 cycloalkyl. In some embodiments, R 3 is -CH 3 , -CH 2 CH 3 , cyclopropyl, or–CH 2 CH 2 OCH 3 .

[0054] In some embodiments, R 3 is -CH 3 .

[0055] In some embodiments, A is -CR A =CR A -.

[0056] In some embodiments, each R A is independently hydrogen, F, Cl, -CN, -CH 3 , -CH 2 CH 3 , - CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -OH, -OCH 3 , -OCH 2 CH 3 , -OCF 3 , -CH 2 F, -CHF 2 , or -CF 3 . In some embodiments, each R A is independently hydrogen, F, Cl, -CN, -CH 3 , -OH, -OCH 3 , -OCF 3 , -CH 2 F, -CHF 2 , or -CF 3 . In some embodiments, each R A is independently hydrogen, F, Cl, -CN, -CH 3 , or -OCH 3 . In some embodiments, each R A is independently hydrogen, F, Cl, or -CH 3 . In some embodiments, R A is hydrogen.

[0057] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 are F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 are F.

[0058] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine, e.g., F or C 1 –C 4 fluoroalkyl such as CH 2 F, CF 3 , CHF 2 , and CH 3 CH 2 F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 is F or C 1 –C 4 fluoroalkyl. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprise a fluorine. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprises a fluorine. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprise a fluorine.

[0059] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine, e.g., F or C 1 –C 4 fluoroalkyl such as CH 2 F, CF 3 , CHF 2 , and CH 3 CH 2 F. In some embodiments, one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine. In some embodiments, W comprises a fluorine.

[0060] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 11 is H, D, or F. In some embodiments, R 11 is D. In some embodiments, R 11 is H. In some embodiments, R 11 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 12 is H, D, or F. In some embodiments, R 12 is D. In some embodiments, R 12 is H. In some embodiments, R 12 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 13 is H, D, or F. In some embodiments, R 13 is D. In some embodiments, R 13 is H. In some embodiments, R 13 is F. In some embodiments of a compound of Formula (I) or (I*), or a

pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 14 is H, D, or F. In some embodiments, R 14 is D. In some embodiments, R 14 is H. In some embodiments, R 14 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 15 is H, D, F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 15 is H or D. In some embodiments, R 15 is H. In some embodiments, R 15 is D. In some embodiments, R 15 is F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 15 is F, CF 3 , CHF 2 , or CH 2 F. In some embodiments, R 15 is F.

[0061] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 16 is H, D, or F. In some embodiments, R 16 is D. In In some embodiments, R 16 is H. some embodiments, R 16 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 17 is H, D, or F. In some embodiments, R 17 is D. In some embodiments, R 17 is H. In some embodiments, R 17 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 18 is H, D, F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 18 is H or D. In some embodiments, R 18 is H. In some embodiments, R 18 is D. In some embodiments, R 18 is F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 18 is F, CF 3 , CHF 2 , or CH 2 F. In some embodiments, R 18 is F.

[0062] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 11 , R 12 , R 19 , R 20 and R 16 are hydrogen. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 19 is hydrogen. In some embodiments, R 19 is H, F, -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, - OCH 2 CN, -OCF 3 , -CH 3 , -CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, - CH2CHF2, and -CH2CF3. In some embodiments, R 19 is H, F, -OH, -OCH3, -OCF3, -CH3, -CH2OH, -CH2F, - CHF 2 , and -CF 3 . In some embodiments, R 19 is F or -OCH 3 .

[0063] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 20 is hydrogen. In some embodiments, R 20 is H, F, -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, -OCH 2 CN, -OCF 3 , -CH 3 , -CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, - CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , and -CH 2 CF 3 . In some embodiments, R 20 is H, F, -OH, -OCH 3 , -OCF 3 , - CH 3 , -CH 2 OH, -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 20 is F or -OCH 3 .

[0064] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 19 is H, D, or F. In some embodiments, R 19 is D. In some embodiments, R 19 is H. In some embodiments, R 19 is F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 20 is H, D, or F. In some embodiments, R 20 is D. In some embodiments, R 20 is H. In some embodiments, R 20 is F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 16 and R 19 are H. In some embodiments, R 16 and R 19 are D. In some embodiments, R 16 and R 19 are F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 19 and R 20 are H. In some embodiments, R 19 and R 20 are D. In some embodiments, R 19 and R 20 are F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 17 and R 20 are H. In some embodiments, R 17 and R 20 are D. In some embodiments, R 17 and R 20 are F.

[0065] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 are F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 are F.

[0066] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine, e.g., F or C 1 –C 4 fluoroalkyl such as CH 2 F, CF 3 , CHF 2 , and CH 3 CH 2 F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 is F or C 1 –C 4 fluoroalkyl. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprise a fluorine. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 comprises a fluorine. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprise a fluorine.

[0067] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine, e.g., F or C1–C4 fluoroalkyl such as CH2F, CF3, CHF2, and CH3CH2F. In some embodiments, one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine. In some embodiments, W comprises a fluorine.

[0068] In some embodiments, the compound is selected from Table 1A, Table 1C, Table 1E, Table 1G, or Table 1H.

[0069] In some embodiments, a compound of Formula (I) or (I*) is a single enantiomer. In some embodiments, a compound of Formula (I) or (I*) is not racemic. In some embodiments, a compound of Formula (I) or (I*) is substantially free of other isomers. In some embodiments, a compound of Formula (I) or (I*) is a single isomer substantially free of other isomers. In some embodiments, a compound of Formula (I) or (I*) comprises 25% or less of other isomers. In some embodiments, the compound of Formula (I) or (I*) comprises 20% or less of other isomers. In some embodiments, a compound of Formula (I) or (I*) comprises 15% or less of other isomers. In some embodiments, a compound of Formula (I) or (I*) comprises 10% or less of other isomers. In some embodiments, the compound of Formula (I) or (I*) comprises 5% or less of other isomers. In some embodiments, the compound of Formula (I) or (I*) comprises 1% or less of other isomers.

[0070] In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 75%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 80%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 85%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 90%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 95%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 96%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 97%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 98%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 98%.

[0071] In some embodiments, an asymmetric carbon atom of a compound of Formula (I) or (I*) is present in enantiomerically enriched form. In certain embodiments, the asymmetric carbon atom of the compound of Formula (I) or (I*) has at least 50% enantiomeric excess, at least 60 % enantiomeric excess, at least 70 % enantiomeric excess, at least 80 % enantiomeric excess, at least 90 % enantiomeric excess, at least 95 % enantiomeric excess, or at least 99 % enantiomeric excess in the (S)- or (R)-configuration.

[0072] In one aspect, provided herein is a pharmaceutical composition comprising a compound of the disclosure or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, and a pharmaceutically acceptable excipient or carrier.

[0073] In one aspect, descried herein is a method of treating a condition or disease comprising administering a compound of the disclosure or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof to a subject in need thereof.

[0074] In one aspect, described herein is the use of a compound of the disclosure or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof in the manufacture of a medicament for the treatment of a condition or disease.

INCORPORATION BY REFERENCE

[0075] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

DETAILED DESCRIPTION

[0076] Certain specific details of this description are set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the present disclosure may be practiced without these details. In other instances, well-known structures have not been shown or described in detail to avoid unnecessarily obscuring descriptions of the embodiments. Unless the context requires otherwise, throughout the specification and claims which follow, the word“comprise” and variations thereof, such as, “comprises” and“comprising” are to be construed in an open, inclusive sense, that is, as“including, but not limited to.” Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed disclosure.

[0077] As used in this specification and the appended claims, the singular forms“a,”“an,” and“the” include plural referents unless the content clearly dictates otherwise. It should also be noted that the term“or” is generally employed in its sense including“and/or” unless the content clearly dictates otherwise. [0078] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described below.

Definitions

[0079] The terms“compound(s) of this disclosure”,“compound(s) of the present disclosure”,“small molecule steric modulator(s)”,“small molecule splicing modulator(s)”“steric modulator(s)”,“splicing modulator(s)”, “compound(s) that modify splicing” and“compound(s) modifying splicing”,“SMSM” or“small molecule that binds a target RNA,” are interchangeably used herein and refer to compounds as disclosed herein and stereoisomers, tautomers, solvates, and salts (e.g., pharmaceutically acceptable salts) thereof. The terms “compound(s) of this disclosure”,“compound(s) of the present disclosure”,“small molecule steric modulator(s)”,“small molecule splicing modulator(s)”“steric modulator(s)”,“splicing modulator(s)”, “compound(s) that modify splicing” and“compound(s) modifying splicing”,“SMSM” or“small molecule that binds a target RNA,” denote a small molecule compound that binds to a cell component (e.g., DNA, RNA, pre- mRNA, protein, RNP, snRNA, carbohydrates, lipids, co-factors, nutrients and/or metabolites) and modulates splicing of a target polynucleotide, e.g., a pre-mRNA. For example, an SMSM can bind directly or indirectly to a target polynucleotide, e.g., RNA (e.g., a pre-mRNA) with a mutated, non-mutated, bulged and/or aberrant splice site, resulting in modulation of splicing of the target polynucleotide. For example, an SMSM can bind directly or indirectly to a protein, e.g., a spliceosome protein or a ribonuclear protein, resulting in steric modulation of the protein and modulation of splicing of a target RNA. For example, an SMSM can bind directly or indirectly to a spliceosome component, e.g., a spliceosome protein or snRNA resulting in steric modulation of the spliceosome protein or snRNA and modulation of splicing of target polynucleotide. These terms specifically exclude compounds consisting of oligonucleotides. These terms include small molecule compounds that may bind to one or more secondary or tertiary structure elements of a target RNA. These sites include RNA triplexes, 3WJs, 4WJs, parallel-Y junctions, hairpins, bulge loops, pseudoknots, internal loops, and other higher- order RNA structural motifs.

[0080] The term“RNA” (ribonucleic acid) as used herein, means naturally-occurring or synthetic oligoribonucleotides independent of source (e.g., the RNA may be produced by a human, animal, plant, virus, or bacterium, or may be synthetic in origin), biological context (e.g., the RNA may be in the nucleus, circulating in the blood, in vitro, cell lysate, or isolated or pure form), or physical form (e.g., the RNA may be in single-, double-, or triple-stranded form (including RNA-DNA hybrids), may include epigenetic modifications, native post-transcriptional modifications, artificial modifications (e.g., obtained by chemical or in vitro modification), or other modifications, may be bound to, e.g., metal ions, small molecules, proteins such as chaperones, or co- factors, or may be in a denatured, partially denatured, or folded state including any native or unnatural secondary or tertiary structure such as quadruplexes, hairpins, triplexes, three way junctions (3WJs), four way junctions (4WJs), parallel-Y junctions, hairpins, bulge loops, pseudoknots, and internal loops, etc., and any transient forms or structures adopted by the RNA). In some embodiments, the RNA is 20, 22, 50, 75, or 100 or more nucleotides in length. In some embodiments, the RNA is 250 or more nucleotides in length. In some embodiments, the RNA is 350, 450, 500, 600, 750, or 1,000, 2,000, 3,000, 4,000, 5,000, 7,500, 10,000, 15,000, 25,000, 50,000, or more nucleotides in length. In some embodiments, the RNA is between 250 and 1,000 nucleotides in length. In some embodiments, the RNA is a pre-RNA, pre-miRNA, or pretranscript. In some embodiments, the RNA is a non-coding RNA (ncRNA), messenger RNA (mRNA), micro-RNA (miRNA), a ribozyme, riboswitch, lncRNA, lincRNA, snoRNA, snRNA, scaRNA, piRNA, ceRNA, pseudo- gene, viral RNA, fungal RNA, parasitic RNA, or bacterial RNA.

[0081] The term“target polynucleotide” or“target RNA,” as used herein, means any type of polynucleotide or RNA, respectively, having a splice site capable of being modulated by a small molecule compound described herein. For example, a target polynucleotide” or“target RNA,” may have a secondary or tertiary structure capable of binding a small molecule compound described herein.

[0082]“Steric alteration”,“steric modification” or“steric modulation” herein refers to changes in the spatial orientation of chemical moieties with respect to each other. A person of ordinary skill in the art would recognize steric mechanisms include, but are not limited to, steric hindrance, steric shielding, steric attraction, chain crossing, steric repulsions, steric inhibition of resonance, and steric inhibition of protonation.

[0083] Any open valency appearing on a carbon, oxygen, sulfur or nitrogen atom in the structures herein indicates the presence of hydrogen, unless indicated otherwise.

[0084] The definitions described herein apply irrespective of whether the terms in question appear alone or in combination. It is contemplated that the definitions described herein can be appended to form chemically- relevant combinations, such as e.g.“heterocycloalkylaryl”,“haloalkylheteroaryl”,“ arylalkylheterocycloalkyl”, or“alkoxyalkyl”. The last member of the combination is the radical which is binding to the rest of the molecule. The other members of the combination are attached to the binding radical in reversed order in respect of the literal sequence, e.g. the combination arylalkylheterocycloalkyl refers to a heterocycloalkyl-radical which is substituted by an alkyl which is substituted by an aryl.

[0085] When indicating the number of substituents, the term“one or more” refers to the range from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents.

[0086] The term“optional” or“optionally” denotes that a subsequently described event or circumstance can but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not.

[0087] The term“substituent” denotes an atom or a group of atoms replacing a hydrogen atom on the parent molecule.

[0088] The term“substituted” denotes that a specified group bears one or more substituents. Where any group can carry multiple substituents and a variety of possible substituents is provided, the substituents are independently selected and need not to be the same. The term“unsubstituted” means that the specified group bears no substituents. The term“optionally substituted” means that the specified group is unsubstituted or substituted by one or more substituents, independently chosen from the group of possible substituents. When indicating the number of substituents, the term“one or more” means from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents.

[0089] The following abbreviations are used throughout the specification: acetic acid (AcOH); ethyl acetate (EtOAc); butyl alcohol (n-BuOH); 1,2-dichloroethane (DCE); dichloromethane (CH 2 Cl 2 , DCM); diisopropylethylamine (Diipea); dimethylformamide (DMF); hydrogen chloride (HCl); methanol (MeOH); methoxymethyl bromide (MOMBr); N-methyl-2-pyrrolidone (NMP); methyl Iodide (MeI); n-propanol (n- PrOH); p-methoxybenzyl (PMB); triethylamine (Et 3 N); [1,1’-Bis(diphenylphosphino)ferrocene] dichloropalladium(II); (Pd(dppf)Cl 2 ); sodium ethane thiolate (EtSNa); sodium acetate (NaOAc); sodium hydride (NaH); sodium hydroxide (NaOH); tetrahydropyran (THP); tetrahydrofuran (THF).

[0090] As used herein, C 1 -C x includes C 1 -C 2 , C 1 -C 3 ... C 1 -C x . By way of example only, a group designated as “C 1 -C 4 ” indicates that there are one to four carbon atoms in the moiety, i.e. groups containing 1 carbon atom, 2 carbon atoms, 3 carbon atoms or 4 carbon atoms. Thus, by way of example only,“C 1 -C 4 alkyl” indicates that there are one to four carbon atoms in the alkyl group, i.e., the alkyl group is selected from among methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.

[0091] The term“oxo” refers to the =O substituent.

[0092] The term“thioxo” refers to the =S substituent.

[0093] The term“halo”,“halogen”, and“halide” are used interchangeably herein and denote fluoro, chloro, bromo, or iodo.

[0094] The term“alkyl” refers to a straight or branched hydrocarbon chain radical, having from one to twenty carbon atoms, and which is attached to the rest of the molecule by a single bond. An alkyl comprising up to 10 carbon atoms is referred to as a C 1 -C 10 alkyl, likewise, for example, an alkyl comprising up to 6 carbon atoms is a C 1 -C 6 alkyl. Alkyls (and other moieties defined herein) comprising other numbers of carbon atoms are represented similarly. Alkyl groups include, but are not limited to, C 1 -C 10 alkyl, C 1 -C 9 alkyl, C 1 -C 8 alkyl, C 1 -C 7 alkyl, C 1 -C 6 alkyl, C 1 -C 5 alkyl, C 1 -C 4 alkyl, C 1 -C 3 alkyl, C 1 -C 2 alkyl, C 2 -C 8 alkyl, C 3 -C 8 alkyl and C 4 -C 8 alkyl. Representative alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, 1-methylethyl (i-propyl), n- butyl, i-butyl, s-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, 1-ethyl-propyl, and the like. In some embodiments, the alkyl is methyl or ethyl. In some embodiments, the alkyl is -CH(CH 3 ) 2 or - C(CH 3 ) 3 . Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted as described below.“Alkylene” or“alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group. In some embodiments, the alkylene is -CH 2 -, -CH 2 CH 2 -, or - CH 2 CH 2 CH 2 -. In some embodiments, the alkylene is -CH 2 -. In some embodiments, the alkylene is -CH 2 CH 2 -. In some embodiments, the alkylene is -CH 2 CH 2 CH 2 -.

[0095] The term“alkoxy” refers to a radical of the formula -OR a where R a is an alkyl radical as defined. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted as described below. Representative alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, pentoxy. In some embodiments, the alkoxy is methoxy. In some embodiments, the alkoxy is ethoxy. [0096] The term“alkylamino” refers to a radical of the formula -NHR a or -NR a R a where each R a is, independently, an alkyl radical as defined above. Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted as described below.

[0097] The term“alkenyl” refers to a type of alkyl group in which at least one carbon-carbon double bond is present. In one embodiment, an alkenyl group has the formula -C(R)=CR a

2, wherein R a refers to the remaining portions of the alkenyl group, which may be the same or different. In some embodiments, R a is H or an alkyl. In some embodiments, an alkenyl is selected from ethenyl (i.e., vinyl), propenyl (i.e., allyl), butenyl, pentenyl, pentadienyl, and the like. Non-limiting examples of an alkenyl group include -CH=CH 2 , -C(CH 3 )=CH 2 , - CH=CHCH 3 , -C(CH 3 )=CHCH 3 , and -CH 2 CH=CH 2 .

[0098] The term“alkynyl” refers to a type of alkyl group in which at least one carbon-carbon triple bond is present. In one embodiment, an alkenyl group has the formula -CºC-R a , wherein R a refers to the remaining portions of the alkynyl group. In some embodiments, R a is H or an alkyl. In some embodiments, an alkynyl is selected from ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Non-limiting examples of an alkynyl group include -CºCH, -CºCCH 3 -CºCCH 2 CH 3 , -CH 2 CºCH.

[0099] The term“aromatic” refers to a planar ring having a delocalized S-electron system containing 4n+2 S electrons, where n is an integer. Aromatics can be optionally substituted. The term“aromatic” includes both aryl groups (e.g., phenyl, naphthalenyl) and heteroaryl groups (e.g., pyridinyl, quinolinyl).

[0100] The term“aryl” refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. Aryl groups can be optionally substituted. Examples of aryl groups include, but are not limited to phenyl, and naphthyl. In some embodiments, the aryl is phenyl. Depending on the structure, an aryl group can be a monoradical or a diradical (i.e., an arylene group). Unless stated otherwise specifically in the specification, the term“aryl” or the prefix“ar-”(such as in“aralkyl”) is meant to include aryl radicals that are optionally substituted. In some embodiments, an aryl group is partially reduced to form a cycloalkyl group defined herein. In some embodiments, an aryl group is fully reduced to form a cycloalkyl group defined herein.

[0101] The term“haloalkyl” denotes an alkyl group wherein at least one of the hydrogen atoms of the alkyl group has been replaced by same or different halogen atoms, particularly fluoro atoms. Examples of haloalkyl include monofluoro-, difluoro-or trifluoro-methyl, -ethyl or -propyl, for example 3,3,3-trifluoropropyl, 2- fluoroethyl, 2,2,2-trifluoroethyl, fluoromethyl, or trifluoromethyl. The term“perhaloalkyl” denotes an alkyl group where all hydrogen atoms of the alkyl group have been replaced by the same or different halogen atoms.

[0102] The term“haloalkoxy” denotes an alkoxy group wherein at least one of the hydrogen atoms of the alkoxy group has been replaced by same or different halogen atoms, particularly fluoro atoms. Examples of haloalkoxyl include monofluoro-, difluoro-or trifluoro-methoxy, -ethoxy or -propoxy, for example 3,3,3- trifluoropropoxy, 2-fluoroethoxy, 2,2,2-trifluoroethoxy, fluoromethoxy, or trifluoromethoxy. The term “perhaloalkoxy” denotes an alkoxy group where all hydrogen atoms of the alkoxy group have been replaced by the same or different halogen atoms. [0103] The term“bicyclic ring system” denotes two rings which are fused to each other via a common single or double bond (annelated bicyclic ring system), via a sequence of three or more common atoms (bridged bicydic ring system) or via a common single atom (spiro bicyclic ring system). Bicyclic ring systems can be saturated, partially unsaturated, unsaturated or aromatic. Bicyclic ring systems can comprise heteroatoms selected from N, O and S.

[0104] The terms“earhocydie” or“carbocycie” refer to a ring or ring system where the atoms forming the backbone of the ring are all carbon atoms. The term thus distinguishes carbocyclic from“heterocyclic” rings or “heterocycles” in which the ring backbone contains at least one atom which is different from carbon. In some embodiments, at least one of the two rings of a bicyclic carbocycie is aromatic. In some embodiments, both rings of a bicyclic carbocycie are aromatic. Carbocycie includes cycloalkyl and aryl.

[0105] The term“cycloalkyl” refers to a monocyclic or polycyclic non -aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. In some embodiments, cycloalkyls are saturated or partially unsaturated. In some embodiments, cycloalkyls are spirocydic or bridged compounds. In some embodiments, cycloalkyls are fused with an aromatic ring (in which case the cycloalkyl is bonded through a non-aromatic ring carbon atom). Cycloalkyl groups include groups having from 3 to 10 ring atoms. Representative cycloalkyls include, but are not limited to, cycloalkyls having from three to ten carbon atoms, from three to eight carbon atoms, from three to six carbon atoms, or from three to five carbon atoms. Monocyclic cycloalkyl radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyi, and cyclooctyl. In some embodiments, the monocyclic cycloalkyl is cyclopropyl, cyclobutyl, cyclopen tyi or cyclohexyl. In some embodiments, the monocyclic cycloalkyl is cyclopentenyl or cyclohexenyl. In some embodiments, the monocyclic cycloalkyl is cyclopentenyl. Polycyclic radicals include, for example, adamantyl, 1 ,2 -dihydronaphtha! eny 1 , 1,4-dihydronaphtha!enyl, tetrainy!, decahnyl, 3 ,4 -dihydronaphthaleny! - 1 (2H) -one, spiro[2.2 [pentyl, norbomyl and bicyde[ 1.1.1] pentyl. Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted.

[0106] The term“bridged” refers to any ring structure with two or more rings that contains a bridge connecting two bridgehead atoms. The bridgehead atoms are defined as atoms that are the part of the skeletal framework of the molecule and which are bonded to three or more other skeletal atoms. In some embodiments, the bridgehead atoms are C, N, or P. In some embodiments, the bridge is a single atom or a chain of atoms that connects two bridgehead atoms. In some embodiments, the bridge is a valence bond that connects two bridgehead atoms. In some embodiments, the bridged ring system is cycloalkyl. In some embodiments, the bridged ring system is heterocycloalkyl.

[0107] The term“fused” refers to any ring structure described herein which is fused to an existing ring structure. When the fused ring is a heterocyclyl ring or a heteroaryl ring, any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring may be replaced with one or more N, S, and O atoms. The non-limiting examples of fused heterocyclyl or heteroaryl ring structures include 6-5 fused heterocycle, 6-6 fused heterocycle, 5-6 fused heterocycle, 5-5 fused heterocycle, 7-5 fused heterocycle, and 5-7 fused heterocycle. [0108] The term“haloalkyl” refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, fluoromethyl, 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 may be optionally substituted.

[0109] The term“haloalkoxy” refers to an alkoxy radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethoxy, difluoromethoxy, fluoromethoxy, trichloromethoxy, 2,2,2-trifluoroethoxy, 1,2-difluoroethoxy, 3-bromo-2-fluoropropoxy, 1,2-dibromoethoxy, and the like. Unless stated otherwise specifically in the specification, a haloalkoxy group may be optionally substituted.

[0110] The term“fluoroalkyl” refers to an alkyl in which one or more hydrogen atoms are replaced by a fluorine atom. In one aspect, a fluoroalkyl is a C 1 -C 6 fluoroalkyl. In some embodiments, a fluoroalkyl is selected from trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like.

[0111] The term“heteroalkyl” refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen (e.g. -NH-, -N(alkyl)-, or -N(aryl)-), sulfur (e.g. -S-, -S(=O)-, or -S(=O) 2 -), or combinations thereof. In some embodiments, a heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. In some embodiments, a heteroalkyl is attached to the rest of the molecule at a heteroatom of the heteroalkyl. In some embodiments, a heteroalkyl is a C 1 -C 6 heteroalkyl. Representative heteroalkyl groups include, but are not limited to -OCH 2 OMe, -OCH 2 CH 2 OH, -OCH 2 CH 2 OMe, or -OCH 2 CH 2 OCH 2 CH 2 NH 2 .

[0112] The term“heteroalkylene” refers to an alkyl radical as described above where one or more carbon atoms of the alkyl is replaced with a O, N or S atom.“Heteroalkylene” or“heteroalkylene chain” refers to a straight or branched divalent heteroalkyl chain linking the rest of the molecule to a radical group. Unless stated otherwise specifically in the specification, the heteroalkyl or heteroalkylene group may be optionally substituted as described below. Representative heteroalkylene groups include, but are not limited to -OCH 2 CH 2 O-, - OCH 2 CH 2 OCH 2 CH 2 O-, or -OCH 2 CH 2 OCH 2 CH 2 OCH 2 CH 2 O-.

[0113] The term“heterocycloalkyl” refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen, and sulfur. Unless stated otherwise specifically in the specification, the heterocycloalkyl radical may be a monocyclic, or bicyclic ring system, which may include fused (when fused with an aryl or a heteroaryl ring, the heterocycloalkyl is bonded through a non-aromatic ring atom) or bridged ring systems. The nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized. The nitrogen atom may be optionally quaternized. The heterocycloalkyl radical is partially or fully saturated. Examples of heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, tetrahydroquinolyl, tetrahydroisoquinolyl, decahydroquinolyl, 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, 1,1-dioxo-thiomorpholinyl. The term heterocycloalkyl also includes all ring forms of carbohydrates, including but not limited to monosaccharides, disaccharides and oligosaccharides. Unless otherwise noted, heterocycloalkyls have from 2 to 12 carbons in the ring. In some embodiments, heterocycloalkyls have from 2 to 10 carbons in the ring. In some embodiments, heterocycloalkyls have from 2 to 10 carbons in the ring and 1 or 2 N atoms. In some embodiments, heterocycloalkyls have from 2 to 10 carbons in the ring and 3 or 4 N atoms. In some embodiments, heterocycloalkyls have from 2 to 12 carbons, 0-2 N atoms, 0-2 O atoms, 0-2 P atoms, and 0-1 S atoms in the ring. In some embodiments, heterocycloalkyls have from 2 to 12 carbons, 1-3 N atoms, 0-1 O atoms, and 0-1 S atoms in the ring. It is understood that when referring to the number of carbon atoms in a heterocycloalkyl, the number of carbon atoms in the heterocycloalkyl is not the same as the total number of atoms (including the heteroatoms) that make up the heterocycloalkyl (i.e. skeletal atoms of the heterocycloalkyl ring). Unless stated otherwise specifically in the specification, a heterocycloalkyl group may be optionally substituted.

[0114] The term“heterocycle” or“heterocyclic” refers to heteroaromatic rings (also known as heteroaryls) and heterocycloalkyl rings (also known as heteroalicyclic groups) that includes at least one heteroatom selected from nitrogen, oxygen and sulfur, wherein each heterocyclic group has from 3 to 12 atoms in its ring system, and with the proviso that any ring does not contain two adjacent O or S atoms. In some embodiments, heterocycles are monocyclic, bicyclic, polycyclic, spirocyclic or bridged compounds. Non-aromatic heterocyclic groups (also known as heterocycloalkyls) include rings having 3 to 12 atoms in its ring system and aromatic heterocyclic groups include rings having 5 to 12 atoms in its ring system. The heterocyclic groups include benzo-fused ring systems. Examples of non-aromatic heterocyclic groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, oxazolidinonyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, thioxanyl, piperazinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6- tetrahydropyridinyl, pyrrolin-2-yl, pyrrolin-3-yl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, 3 h-indolyl, indolin-2-onyl, isoindolin-1-onyl, isoindoline-1,3-dionyl, 3,4-dihydroisoquinolin-1(2H)-onyl, 3,4-dihydroquinolin-2(1H)- onyl, isoindoline-1,3-dithionyl, benzo[d]oxazol-2(3H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, benzo[d]thiazol-2(3H)-onyl, and quinolizinyl. Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. The foregoing groups are either C-attached (or C-linked) or N-attached where such is possible. For instance, a group derived from pyrrole includes both pyrrol-1-yl (N-attached) or pyrrol-3- yl (C-attached). Further, a group derived from imidazole includes imidazol-1-yl or imidazol-3-yl (both N- attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached). The heterocyclic groups include benzo-fused ring systems. Non-aromatic heterocycles are optionally substituted with one or two oxo (=O) moieties, such as pyrrolidin-2-one. In some embodiments, at least one of the two rings of a bicyclic heterocycle is aromatic. In some embodiments, both rings of a bicyclic heterocycle are aromatic.

[0115] The term“heteroaryl” refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. The heteroaryl is monocyclic or bicyclic. Illustrative examples of monocyclic heteroaryls include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, pyridazinyl, triazinyl, oxadiazolyl, thiadiazolyl, furazanyl, indolizine, indole, benzofuran, benzothiophene, indazole, benzimidazole, purine, quinolizine, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,8-naphthyridine, and pteridine. Illustrative examples of monocyclic heteroaryls include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, pyridazinyl, triazinyl, oxadiazolyl, thiadiazolyl, and furazanyl. Illustrative examples of bicyclic heteroaryls include indolizine, indole, benzofuran, benzothiophene, indazole, benzimidazole, purine, quinolizine, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,8-naphthyridine, and pteridine. In some embodiments, heteroaryl is pyridinyl, pyrazinyl, pyrimidinyl, thiazolyl, thienyl, thiadiazolyl or furyl. In some embodiments, a heteroaryl contains 0-6 N atoms in the ring. In some embodiments, a heteroaryl contains 1-4 N atoms in the ring. In some embodiments, a heteroaryl contains 4-6 N atoms in the ring. In some embodiments, a heteroaryl contains 0-4 N atoms, 0-1 O atoms, 0-1 P atoms, and 0-1 S atoms in the ring. In some embodiments, a heteroaryl contains 1-4 N atoms, 0-1 O atoms, and 0-1 S atoms in the ring. In some embodiments, heteroaryl is a C 1 -C 9 heteroaryl. In some embodiments, monocyclic heteroaryl is a C 1 -C 5 heteroaryl. In some embodiments, monocyclic heteroaryl is a 5-membered or 6-membered heteroaryl. In some embodiments, a bicyclic heteroaryl is a C6-C9 heteroaryl. In some embodiments, a heteroaryl group is partially reduced to form a heterocycloalkyl group defined herein. In some embodiments, a heteroaryl group is fully reduced to form a heterocycloalkyl group defined herein.

[0116] The term“moiety” refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule.

[0117] The term“optionally substituted” or“substituted” means that the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from D, halogen, -CN, -NH 2 , -NH(alkyl), -N(alkyl) 2 , -OH, -CO 2 H, -CO 2 alkyl, -C(=O)NH 2 , -C(=O)NH(alkyl), -C(=O)N(alkyl) 2 , - S(=O) 2 NH 2 , -S(=O) 2 NH(alkyl), -S(=O) 2 N(alkyl) 2 , alkyl, cycloalkyl, fluoroalkyl, heteroalkyl, alkoxy, fluoroalkoxy, heterocycloalkyl, aryl, heteroaryl, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, and arylsulfone. In some other embodiments, optional substituents are independently selected from D, halogen, -CN, -NH 2 , -NH(CH 3 ), -N(CH 3 ) 2 , -OH, -CO 2 H, -CO 2 (C 1 -C 4 alkyl), -C(=O)NH 2 , - C(=O)NH(C 1 -C 4 alkyl), -C(=O)N(C 1 -C 4 alkyl) 2 , -S(=O) 2 NH 2 , -S(=O) 2 NH(C 1 -C 4 alkyl), -S(=O) 2 N(C 1 -C 4 alkyl) 2 , C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 4 fluoroalkyl, C 1 -C 4 heteroalkyl, C 1 -C 4 alkoxy, C 1 -C 4 fluoroalkoxy, -SC 1 -C 4 alkyl, -S(=O)C 1 -C 4 alkyl, and -S(=O) 2 (C 1 -C 4 alkyl). In some embodiments, optional substituents are independently selected from D, halogen, -CN, -NH 2 , -OH, -NH(CH 3 ), -N(CH 3 ) 2 , - NH(cyclopropyl), -CH 3 , - CH 2 CH 3 , -CF 3 , -OCH 3 , and -OCF 3 . In some embodiments, substituted groups are substituted with one or two of the preceding groups. In some embodiments, an optional substituent on an aliphatic carbon atom (acyclic or cyclic) includes oxo (=O).

[0118] The term“tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule. The compounds presented herein may exist as tautomers. Tautomers are compounds that are interconvertible by migration of a hydrogen atom, accompanied by a switch of a single bond and adjacent double bond. In bonding arrangements where tautomerization is possible, a chemical equilibrium of the tautomers will exist. All tautomeric forms of the compounds disclosed herein are contemplated. The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. Some examples of tautomeric interconversions include:

[0119] The term“about” or“approximately” can mean within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example,“about” can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively,“about” can mean a range of up to 20%, up to 15%, up to 10%, up to 5%, or up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, within 5-fold, or within 2-fold, of a value.

[0120] The terms“administer,”“administering”,“administration, ” and the like, as used herein, refer to the methods that may be used to enable delivery of compounds or compositions to the desired site of biological action. These methods include, but are not limited to oral routes (p.o.), intraduodenal routes (i.d.), parenteral injection (including intravenous (i.v.), subcutaneous (s.c.), intraperitoneal (i.p.), intramuscular (i.m.), intravascular or infusion (inf.)), topical (top.) and rectal (p.r.) administration. Those of skill in the art are familiar with administration techniques that can be employed with the compounds and methods described herein. In some embodiments, the compounds and compositions described herein are administered orally.

[0121] The terms“co-administration” or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time. [0122] The terms“effective amount” or“therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated: for example a reduction and/or alleviation of one or more signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses can be an amount of an agent that provides a clinically significant decrease in one or more disease symptoms. An appropriate“effective” amount may be determined using techniques, such as a dose escalation study, in individual cases.

[0123] The terms“enhance” or“enhancing,” as used herein, means to increase or prolong either in amount, potency or duration a desired effect. For example, in regard to enhancing splicing of a target, the temi “enhancing” can refer to the ability to increase or prolong splicing, either in amount, potency or duration, of a the target.

[0124] The term“subject” or“patient” encompasses mammals. Examples of mammals include, but are not limited to, any member of the mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. In one aspect, the mammal is a human. The term“animal” as used herein comprises human beings and non-human animals. In one embodiment, a“non-human animal” is a mammal, for example a rodent such as rat or a mouse. In one embodiment, a non-human animal is a mouse.

[0125] The terms“treat,”“treating” or“treatment,” as used herein, include alleviating, abating or ameliorating at least one symptom of a disease or condition, preventing additional symptoms, inhibiting the disease or condition, e.g. , arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.

[0126] The term“preventing” or“prevention” of a disease state denotes causing the clinical symptoms of the disease state not to develop in a subject that can be exposed to or predisposed to the disease state, but does not yet experience or display symptoms of the disease state.

[0127] The terms“pharmaceutical composition” and“pharmaceutical formulation” (or“formulation”) are used interchangeably and denote a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient together with one or more pharmaceutically acceptable excipients to be administered to a subject, e.g , a human in need thereof.

[0128] The term“pharmaceutical combination” as used herein, means a product that results from mixing or combining more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term“fixed combination” means that the active ingredients, e.g. , a compound described herein and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term“non-fixed combination” means that the active ingredients, e.g. a compound described herein and a co agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g., administration of three or more active ingredients.

[0129] The term“pharmaceutically acceptable” denotes an attribute of a material which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and neither biologically nor otherwise undesirable and is acceptable for veterinary as well as human pharmaceutical use.“Pharmaceutically acceptable” can refer a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.

[0130] The terms“pharmaceutically acceptable excipient”,“pharmaceutically acceptable carrier” and “therapeutically inert excipient” can be used interchangeably and denote any pharmaceutically acceptable ingredient in a pharmaceutical composition having no therapeutic activity and being non-toxic to the subject administered, such as disintegrators, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants, carriers, diluents, excipients, preservatives or lubricants used in formulating pharmaceutical products.

[0131] The term“pharmaceutically acceptable salts” denotes salts which are not biologically or otherwise undesirable. Pharmaceutically acceptable salts include both acid and base addition salts. A“pharmaceutically acceptable salt” can refer to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and/or does not abrogate the biological activity and properties of the compound. In some embodiments, pharmaceutically acceptable salts are obtained by reacting an SMSM compound of any one of Formulas (I), (Ia), (Ib), or (Ic) with an acid. Pharmaceutically acceptable salts are also obtained by reacting a compound of any one of Formulas (I), (Ia), (Ib), or (Ic) or with a base to form a salt.

[0132] The type of pharmaceutical acceptable salts, include, but are not limited to: (1) acid addition salts, formed by reacting the free base form of the compound with a pharmaceutically acceptable: inorganic acid, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid, and the like; or with an organic acid, such as, for example, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2- ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 2- naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4’- methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, butyric acid, phenylacetic acid, phenylbutyric acid, valproic acid, and the like; (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion (e.g. lithium, sodium, potassium), an alkaline earth ion (e.g. magnesium, or calcium), or an aluminum ion. In some cases, compounds described herein may coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine. In other cases, compounds described herein may form salts with amino acids such as, but not limited to, arginine, lysine, and the like. Acceptable inorganic bases used to form salts with compounds that include an acidic proton, include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.

[0133] The term“nucleic acid” as used herein generally refers to one or more nucleobases, nucleosides, or nucleotides, and the term includes polynucleobases, polynucleosides, and polynucleotides.

[0134] The term“polynucleotide”, as used herein generally refers to a molecule comprising two or more linked nucleic acid subunits, e.g., nucleotides, and can be used interchangeably with“oligonucleotide”. For example, a polynucleotide may include one or more nucleotides selected from adenosine (A), cytosine (C), guanine (G), thymine (T) and uracil (U), or variants thereof. A nucleotide generally includes a nucleoside and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more phosphate (PO 3 ) groups. A nucleotide can include a nucleobase, a five-carbon sugar (either ribose or deoxyribose), and one or more phosphate groups. Ribonucleotides include nucleotides in which the sugar is ribose. Deoxyribonucleotides include nucleotides in which the sugar is deoxyribose. A nucleotide can be a nucleoside monophosphate, nucleoside diphosphate, nucleoside triphosphate or a nucleoside polyphosphate. For example, a nucleotide can be a deoxyribonucleoside polyphosphate, such as a deoxyribonucleoside triphosphate (dNTP), Exemplary dNTPs include deoxyadenosine triphosphate (dATP), deoxycytidine triphosphate (dCTP), deoxyguanosine triphosphate (dGTP), uridine triphosphate (dUTP) and deoxythymidine triphosphate (dTTP). dNTPs can also include detectable tags, such as luminescent tags or markers (e.g., fluorophores). For example, a nucleotide can be a purine (i.e., A or G, or variant thereof) or a pyrimidine (i.e., C, T or U, or variant thereof). In some examples, a polynucleotide is deoxyribonucleic acid (DNA), ribonucleic acid (RNA), or derivatives or variants thereof. Exemplary polynucleotides include, but are not limited to, short interfering RNA (siRNA), a microRNA (miRNA), a plasmid DNA (pDNA), a short hairpin RNA (shRNA), small nuclear RNA (snRNA), messenger RNA (mRNA), precursor mRNA (pre-mRNA), antisense RNA (asRNA), and heteronuclear RNA (hnRNA), and encompasses both the nucleotide sequence and any structural embodiments thereof, such as single-stranded, double-stranded, triple-stranded, helical, hairpin, stem loop, bulge, etc. In some cases, a polynucleotide is circular. A polynucleotide can have various lengths. For example, a polynucleotide can have a length of at least about 7 bases, 8 bases, 9 bases, 10 bases, 20 bases, 30 bases, 40 bases, 50 bases, 100 bases, 200 bases, 300 bases, 400 bases, 500 bases, 1 kilobase (kb), 2 kb, 3, kb, 4 kb, 5 kb, 10 kb, 50 kb, or more. A polynucleotide can be isolated from a cell or a tissue. For example, polynucleotide sequences may comprise isolated and purified DNA/RNA molecules, synthetic DNA/RNA molecules, and/or synthetic DNA/RNA analogs.

[0135] Polynucleotides may include one or more nucleotide variants, including nonstandard nucleotide(s), non- natural nucleotide(s), nucleotide analog(s) and/or modified nucleotides. Examples of modified nucleotides include, but are not limited to diaminopurine, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl)uracil, 5-carboxymethylaminomethyl-2- thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6- isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5’- methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6- isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5- methyluracil, uracil-5- oxyacetic acid methylester, 5-methyl-2-thiouracil, 3-(3-amino- 3- N-2-carboxypropyl) uracil, (acp3)w, 2,6-diaminopurine and the like. In some cases, nucleotides may include modifications in their phosphate moieties, including modifications to a triphosphate moiety. Non-limiting examples of such modifications include phosphate chains of greater length (e.g., a phosphate chain having, 4, 5, 6, 7, 8, 9, 10 or more phosphate moieties) and modifications with thiol moieties (e.g., alpha-thiotriphosphate and beta- thiotriphosphates). Nucleic acid molecules may also be modified at the base moiety (e.g., at one or more atoms that typically are available to form a hydrogen bond with a complementary nucleotide and/or at one or more atoms that are not typically capable of forming a hydrogen bond with a complementary nucleotide), sugar moiety or phosphate backbone. Nucleic acid molecules may also contain amine -modified groups, such as amino ally 1-dUTP (aa-dUTP) and aminohexhylacrylamide-dCTP (aha-dCTP) to allow covalent attachment of amine reactive moieties, such as N-hydroxysuccinimide esters (NHS). Alternatives to standard DNA base pairs or RNA base pairs in the oligonucleotides of the present disclosure can provide higher density in bits per cubic mm, higher safety (resistant to accidental or purposeful synthesis of natural toxins), easier discrimination in photo-programmed polymerases, or lower secondary structure. Such alternative base pairs compatible with natural and mutant polymerases for de novo and/or amplification synthesis are described in Betz K, Malyshev DA, Lavergne T, Welte W, Diederichs K, Dwyer TJ, Ordoukhanian P, Romesberg FE, Marx A. Nat. Chem. Biol.2012 Jul;8(7):612-4, which is herein incorporated by reference for all purposes.

[0136] As used herein, the terms“polypeptide”,“protein” and“peptide” are used interchangeably and refer to a polymer of amino acid residues linked via peptide bonds and which may be composed of two or more polypeptide chains. The terms“polypeptide”,“protein” and“peptide” refer to a polymer of at least two amino acid monomers joined together through amide bonds. An amino acid may be the L-optical isomer or the D- optical isomer. More specifically, the terms“polypeptide”,“protein” and“peptide” refer to a molecule composed of two or more amino acids in a specific order; for example, the order as determined by the base sequence of nucleotides in the gene or RNA coding for the protein. Proteins are essential for the structure, function, and regulation of the body’s cells, tissues, and organs, and each protein has unique functions. Examples are hormones, enzymes, antibodies, and any fragments thereof. In some cases, a protein can be a portion of the protein, for example, a domain, a subdomain, or a motif of the protein. In some cases, a protein can be a variant (or mutation) of the protein, wherein one or more amino acid residues are inserted into, deleted from, and/or substituted into the naturally occurring (or at least a known) amino acid sequence of the protein. A protein or a variant thereof can be naturally occurring or recombinant.

[0137] Methods for detection and/or measurement of polypeptides in biological material are well known in the art and include, but are not limited to, Western-blotting, flow cytometry, ELISAs, RIAs, and various proteomics techniques. An exemplary method to measure or detect a polypeptide is an immunoassay, such as an ELISA. This type of protein quantitation can be based on an antibody capable of capturing a specific antigen, and a second antibody capable of detecting the captured antigen. Exemplary assays for detection and/or measurement of polypeptides are described in Harlow, E. and Lane, D. Antibodies: A Laboratory Manual, (1988), Cold Spring Harbor Laboratory Press.

[0138] Methods for detection and/or measurement of RNA in biological material are well known in the art and include, but are not limited to, Northern-blotting, RNA protection assay, RT PCR. Suitable methods are described in Molecular Cloning: A Laboratory Manual (Fourth Edition) By Michael R. Green, Joseph Sambrook, Peter MacCallum 2012, 2,028 pp, ISBN 978-1-936113-42-2.

[0139] As used here, a“small molecular weight compound” can be used interchangeably with“small molecule” or“small organic molecule”. Small molecules refer to compounds other than peptides or oligonucleotides; and typically have molecular weights of less than about 2000 Daltons, e.g., less than about 900 Daltons.

[0140] A ribonucleoprotein (RNP) refers to a nucleoprotein that contains RNA. A RNP can be a complex of a ribonucleic acid and an RNA-binding protein. Such a combination can also be referred to as a protein-RNA complex. These complexes can function in a number of biological functions that include, but are not limited to, DNA replication, gene expression, metabolism of RNA, and pre-mRNA splicing. Examples of RNPs include the ribosome, the enzyme telomerase, vault ribonucleoproteins, RNase P, heterogeneous nuclear RNPs (hnRNPs) and small nuclear RNPs (snRNPs).

[0141] Nascent RNA transcripts from protein-coding genes and mRNA processing intermediates, collectively referred to as pre-mRNA, are generally bound by proteins in the nuclei of eukaryotic cells. From the time nascent transcripts first emerge from RNA polymerase (e.g., RNA polymerase II) until mature mRNAs are transported into the cytoplasm, the RNA molecules are associated with an abundant set of splicing complex components (e.g., nuclear proteins and snRNAs). These proteins can be components of hnRNPs, which can contain heterogeneous nuclear RNA (hnRNA) (e.g., pre-mRNA and nuclear RNA complexes) of various sizes.

[0142] Splicing complex components function in splicing and/or splicing regulation. Splicing complex components can include, but are not limited to, ribonuclear proteins (RNPs), splicing proteins, small nuclear RNAs (snRNAs), small nuclear ribonucleoproteins (snRNPs), and heterogeneous nuclear ribonucleoproteins (hnRNPs). Splicing complex components include, but are not limited to, those that may be required for splicing, such as constitutive splicing, alternative splicing, regulated splicing and splicing of specific messages or groups of messages. A group of related proteins, the serine arginine rich proteins (SR proteins), can function in constitutive pre-mRNA splicing and may also regulate alternative splice-site selection in a concentration- dependent manner. SR proteins typically have a modular structure that consists of one or two RNA-recognition motifs (RRMs) and a C-terminal rich in arginine and serine residues (RS domain). Their activity in alternative splicing may be antagonized by members of the hnRNP A/B family of proteins. Splicing complex components can also include proteins that are associated with one or more snRNAs. SR proteins in human include, but are not limited to, SC35, SRp55, SRp40, SRm300, SFRS10, TASR-1, TASR-2, SF2/ASF, 9G8, SRp75, SRp30c, SRp20 and P54/SFRS11. Other splicing complex components in human that can be involved in splice site selection include, but are not limited to, U2 snRNA auxiliary factors (e.g. U2AF65, U2AF35), Urp/U2AF1- RS2, SF1/BBP, CBP80, CBP 20, SF1 and PTB/hnRNP1. hnRNP proteins in humans include, but are not limited to, A1, A2/B1, L, M, K, U, F, H, G, R, I and C1/C2. Human genes encoding hnRNPs include HNRNPA0, HNRNPA1, HNRNPA1L1, HNRNPA1L2, HNRNPA3, HNRNPA2B1, HNRNPAB, HNRNPB1, HNRNPC, HNRNPCL1, HNRNPD, HNRPDL, HNRNPF, HNRNPH1, HNRNPH2, HNRNPH3, HNRNPK, HNRNPL, HNRPLL, HNRNPM, HNRNPR, HNRNPU, HNRNPUL1, HNRNPUL2, HNRNPUL3, and FMR1. Splicing complex components may be stably or transiently associated with a snRNP or with a transcript.

[0143] The term“intron” refers to both the DNA sequence within a gene and the corresponding sequence in the unprocessed RNA transcript. As part of the RNA processing pathway, introns can be removed by RNA splicing either shortly after or concurrent with transcription. Introns are found in the genes of most organisms and many viruses. They can be located in a wide range of genes, including those that generate proteins, ribosomal RNA (rRNA), and transfer RNA (tRNA).

[0144] An“exon” can be any part of a gene that encodes a part of the final mature RNA produced by that gene after introns have been removed by RNA splicing. The term“exon” refers to both the DNA sequence within a gene and to the corresponding sequence in RNA transcripts.

[0145] A“spliceosome” can be assembled from snRNAs and protein complexes. The spliceosome can remove introns from a transcribed pre-mRNA.

[0146] “Medium effective dose” (ED 50 ) is the dose at which 50% of a population expresses a specified response.“Medium lethal dose” (LD 50 ) is the dose at which 50% of a population dies.“Medium toxic dose” (TD50) is the dose at which 50% of a population expresses a specified toxic effect. One particularly useful pharmacological indicator is the“therapeutic index” which is traditionally defined as the ratio of LD 50 to ED 50 or the ratio of TD 50 to ED 50 . Therapeutic index provides a simple and useful indicator of the benefit versus adverse effect of a drug. Those drugs which have a high therapeutic index have a large therapeutic window, i.e., the drugs may be administered over a wider range of effective doses without incurring significant adverse events. Conversely, drugs having a small therapeutic index have a small therapeutic window (small range of effective doses without incurring significant adverse events).

[0147] The term“AUC” as used herein refers to an abbreviation for“area under the curve” in a graph of the concentration of a therapeutic agent over time in a certain part or tissue, such as blood or plasma, of a subject to whom the therapeutic agent has been administered.

Small Molecule Splicing Modulators (SMSMs)

[0148] It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as agents for use in treating, preventing, or ameliorating a disease or condition associated with a target RNA. The present invention provides the unexpected discovery that certain small chemical molecules can modify splicing events in pre-mRNA molecules, herein referred to as small molecule splicing modulators (SMSMs). These SMSMs can modulate specific splicing events in specific pre-mRNA molecules. These SMSMs can operate by a variety of mechanisms to modify splicing events. For example, the SMSMs of this invention can: 1) interfere with the formation and/or function and/or other properties of splicing complexes, spliceosomes, and/or their components such as hnRNPs, snRNPs, SR-proteins and other splicing factors or elements, resulting in the prevention or induction of a splicing event in a pre-mRNA molecule. As another example; 2) prevent and/or modify post-transcriptional regulation (e.g., splicing) of gene products, such as hnRNPs, snRNPs, SR-proteins and other splicing factors, which can subsequently be involved in the formation and/or function of a spliceosome or splicing complex component; 3) prevent and/or modify phosphorylation, glycosylation and/or other modifications of gene products including, but not limited to, hnRNPs, snRNPs, SR- proteins and other splicing factors, which can subsequently be involved in the formation and/or function of a spliceosome or splicing complex component; 4) bind to and/or otherwise affect specific pre-mRNA so that a specific splicing event is prevented or induced, e.g., via a mechanism that does not involve base pairing with RNA in a sequence-specific manner. The small molecules of this invention are different from and are not related to antisense or antigene oligonucleotides.

[0149] Described herein are compounds modifying splicing of gene products for use in the treatment, prevention and/or delay of progression of diseases or conditions (e.g., cancer). Described herein are compounds modifying splicing of gene products wherein the compounds induce a transcriptionally inactive variant or transcript of a gene product. Described herein are compounds modifying splicing of gene products wherein the compounds repress a transcriptionally active variant or transcript of a gene product.

[0150] In some embodiments, a compound of Formula (I) or (I*) is made from racemic starting materials (and/or intermediate) and separated into the individual enantiomers by chiral chromatography as an intermediate or final product. Unless otherwise stated, it is understood that the absolute configuration of the separated intermediates and final compounds is not determined. In some emboeiments, the absolute stereochemistry of the enantiomers as drawn is arbitrarily assigned. In some embodiments, both enantiomers are synthesized.

[0151] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

E is–NR-, -O-, -S-, -S(=O)-, -S(=O) 2 -, or -S(=O)(=NR E )-;

R E is hydrogen, substituted or unsubstituted C 1 -C 3 alkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted C 2 -C 3 alkenyl, or substituted or unsubstituted C 2 -C 3 alkynyl;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is hydrogen;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1B.

[0152] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

E is–NR-, -O-, -S-, -S(=O)-, -S(=O) 2 -, or -S(=O)(=NR E )-; R E is hydrogen, substituted or unsubstituted C 1 -C 3 alkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted C 2 -C 3 alkenyl, or substituted or unsubstituted C 2 -C 3 alkynyl;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is hydrogen;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C1–C4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1B.

[0153] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C3–C8 cycloalkylene, substituted or unsubstituted C2–C7

heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or

unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0; c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1D.

[0154] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C2–C3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7

heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or

unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl; each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are both hydrogen or both deuterium;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1D.

[0155] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O)2R 1 , substituted or unsubstituted C1–C4 alkyl, substituted or unsubstituted C1–C4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are the same and selected from the group consisting of F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1F.

[0156] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene; R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are the same and selected from the group consisting of F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1, with the provision that the compound is not a compound in Table 1F.

[0157] In one aspect, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl; ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C1–C4 alkyl, a substituted or unsubstituted C1–C4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1.

[0158] In one aspect, provided herein is a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof:

wherein,

A is -CR A =CR A -;

each R A is independently selected from the group consisting of hydrogen, deuterium, F, Cl,–CN,–OR 1 , –SR 1 , -S(=O)R 1 , -S(=O) 2 R 1 , substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 4 cycloalkyl, and substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

ring Q is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

X is -NR 3 -;

Z is CR 2 ;

W is substituted or unsubstituted C 1 -C 3 alkylene, substituted or unsubstituted C 1 –C 2 heteroalkylene, substituted or unsubstituted C 3 –C 8 cycloalkylene, substituted or unsubstituted C 2 –C 7 heterocycloalkylene, or substituted or unsubstituted C 2 -C 3 alkenylene;

R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 –C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, or substituted or unsubstituted C 2 –C 5 heterocycloalkyl;

each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

R 2 is hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, or substituted or unsubstituted C 1 –C 4 haloalkyl;

R 3 is hydrogen, -CN, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , substituted or unsubstituted C 3 –C 4 cycloalkyl, or substituted or unsubstituted C 2 –C 3 heterocycloalkyl;

each R 11 , R 12 , R 13 , R 14 , R 16 , R 17 , R 19 , and R 20 is independently selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F,–OR 1 , substituted or unsubstituted C 1 –C 4 alkyl, a substituted or unsubstituted C 1 –C 4 fluoroalkyl, and substituted or unsubstituted C 1 –C 4 heteroalkyl;

a is 0;

b is 0;

c is 1; and

d is 1.

[0159] In some embodiments, the compound of Formula (I) has the structure of Formula (Ia):

[0160] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib):

[0161] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib1):

[0162] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib2):

[0163] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib3):

[0164] In some embodiments, the compound of Formula (I) has the structure of Formula (Ib4):

[0165] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic):

[0166] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic1):

[0167] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic2):

[0168] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic3):

[0169] In some embodiments, the compound of Formula (I) has the structure of Formula (Ic4):

[0170] In some embodiments, ring Q is substituted or unsubstituted aryl. In some embodiments, ring Q is 2– hydroxy–phenyl substituted with 1, 2, or 3 substituents independently selected from: deuterium, halogen, hydroxy, nitro, cyano,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,–C(=O)R 1 ,–OC(=O)R 1 ,–C(=O)OR 1 ,– C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, substituted or unsubstituted C 2 -C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 haloalkyl, substituted or unsubstituted C 1 – C 6 heteroalkyl, substituted or unsubstituted C 3 –C 8 cycloalkyl, substituted or unsubstituted C 2 –C 7

heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0171] In some embodiments, ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl. In some embodiments, ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted aryl, wherein if aryl is substituted then it is substituted with 1 or 2 substituents independently selected from: deuterium, halogen, -OH, -NO 2 , -CN,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,– C(=O)R 1 ,–OC(=O)R 1 ,–C(=O)OR 1 ,–C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, and substituted or unsubstituted C 2 -C 7 heterocycloalkyl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0172] In some embodiments, ring Q is 2–hydroxy–phenyl substituted with substituted or unsubstituted heteroaryl, wherein if heteroaryl is substituted then it is substituted with 1 or 2 substituents independently selected from: deuterium, halogen, -OH, -NO 2 , -CN,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,–C(=O)R 1 ,– OC(=O)R 1 ,–C(=O)OR 1 ,–C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, and substituted or unsubstituted C 2 -C 7 heterocycloalkyl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C1–C4 alkyl, -CD3, substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 – C 6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0173] In some embodiments, ring Q is substituted or unsubstituted heteroaryl. In some embodiments, ring Q is substituted or unsubstituted 5- or 6-membered monocyclic heteroaryl. In some embodiments, ring Q is substituted or unsubstituted 6-membered monocyclic heteroaryl.

[0174] In some embodiments, ring Q is 6-membered monocyclic heteroaryl selected from:

wherein each R Q is independently selected from the group consisting of hydrogen, deuterium,–F,–Cl, -CN,–OH, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 ,–CF 3, – OCH 3 , -OCH 2 CH 3 , -CH 2 OCH 3 , -OCH 2 CH 2 CH 3 , and -OCH(CH 3 ) 2 ; and ring P is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

[0175] In some embodiments, ring Q is , wherein each R Q is independently selected from the group consisting of hydrogen, deuterium,–F,–Cl, -CN,–OH, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , - CH(CH 3 ) 2 ,–CF 3, –OCH 3 , -OCH 2 CH 3 , -CH 2 OCH 3 , -OCH 2 CH 2 CH 3 , and -OCH(CH 3 ) 2 ; and ring P is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

[0176] In some embodiments, R Q is independently selected from the group consisting of hydrogen,–F,–Cl, - CN,–OH, -CH 3 ,–CF 3, and–OCH 3 .

[0177] In some embodiments, ring P is substituted or unsubstituted heteroaryl.

[0178] In some embodiments, ring P is heteroaryl selected from the group consisting of:

wherein, each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl; and m is 0, 1, 2, or 3.

[0179] In some embodiments, ring P is heteroaryl selected from the group consisting of:

wherein, each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, deuterium substituted C1-C6 alkoxy, -OCD3, substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl; and m is 0, 1, 2, or 3.

[0180] In some embodiments of a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring P is heteroaryl selected from the group consisting of:

and wherein each R B is independently selected from the group consisting of

hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; and m is 0, 1, 2, 3, or 4. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 0, 1, 2, or 3. In some embodiments, m is 1, 2, 3, or 4. In some embodiments, m is 1, 2, or 3. In some embodiments, m is 2, 3, or 4. In some embodiments, m is 0 or 1. In some embodiments, m is 1 or 2. In some embodiments, m is 2 or 3. In some embodiments, m is 3 or 4.

[0181] In some embodiments of a compound of Formula (I), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring P is heteroaryl selected from the group consisting of:

wherein each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; and m is 1, 2, 3 or 4. In some embodiments, R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl. In some embodiments, m is 1, 2, or 3. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4.

[0182] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring P is heteroaryl selected from the group consisting of:

, wherein each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; and m is 0, 1, 2, 3, or 4. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 0, 1, 2, or 3. In some embodiments, m is 1, 2, 3, or 4. In some embodiments, m is 1, 2, or 3. In some embodiments, m is 2, 3, or 4. In some embodiments, m is 0 or 1. In some

embodiments, m is 1 or 2. In some embodiments, m is 2 or 3. In some embodiments, m is 3 or 4.

[0183] In some embodiments, each R B is independently hydrogen, deuterium,–F,–Cl, -CN,–CH 3 ,–CF 3, – OH, or–OCH 3 . In some embodiments, each R B is independently–F or–OCH 3 . In some embodiments, R B is hydrogen. In some embodiments, R B is–OCH 3 . In some embodiments, R B is–CH 3 .

[0184] In some embodiments, R B1 is hydrogen, deuterium,–CH 3 ,–CF 3, or–CD 3 .

[0185] In some embodiments, m is 0 or 1. In some embodiments, m is 1, 2, or 3. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3.

[0186] In some embodiments, ring Q is 2–naphthyl substituted at the 3 position with 0, 1, and 2 substituents independently selected from: deuterium, halogen, -OH, -NO 2 , -CN,–SR 1 ,–S(=O)R 1 ,–S(=O) 2 R 1 ,–N(R 1 ) 2 ,– C(=O)R 1 ,–OC(=O)R 1 ,–C(=O)OR 1 ,–C(=O)N(R 1 ) 2 , substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 7 cycloalkyl, substituted or unsubstituted C 2 -C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl; wherein each R 1 is independently hydrogen, deuterium, substituted or unsubstituted C 1 –C 4 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C1–C4 heteroalkyl, substituted or unsubstituted C3–C6 cycloalkyl, substituted or unsubstituted C 2 –C 5 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[0187] In some embodiments, ring Q is selected from the group consisting of:

and

[0188] In some embodiments, ring Q is selected from the group consisting of:

and

[0189] In some embodiments, ring Q is selected from the group consisting of:

and

[0190] In some embodiments, ring Q is selected from the group consisting of:

and wherein R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl. [0191] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring Q is selected from the group consisting of:

and wherein each of the ring Q group can be optionally substituted with 1-3

R B , wherein each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.

[0192] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring Q is selected from the group consisting of:

and wherein each of the ring Q group can be optionally

substituted with 1-3 R B , wherein each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.

[0193] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring Q is selected from the group consisting of:

and wherein each of the ring Q group can be optionally substituted with 1, 2, 3, 4, or 5 R B , wherein each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, -OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.

[0194] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, ring Q is selected from the group consisting of: [0195]

and wherein R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl, and wherein each of the ring Q group can be optionally substituted with 1, 2, 3, 4 or 5 R B , wherein each R B is independently selected from the group consisting of hydrogen, deuterium, halogen, hydroxy, cyano, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 2 -C 6 alkenyl, substituted or unsubstituted C 2 -C 6 alkynyl, substituted or unsubstituted C 1 -C 6 alkoxy, deuterium substituted C 1 -C 6 alkoxy, - OCD 3 , substituted or unsubstituted C 3–7 cycloalkyl, substituted or unsubstituted C 2 –C 7 heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl. In some embodiments, R B1 is selected from the group consisting of hydrogen, deuterium, substituted or unsubstituted C 1 -C 6 alkyl, -CD 3 , substituted or unsubstituted C 1 –C 6 fluoroalkyl, substituted or unsubstituted C 1 -C 6 heteroalkyl, substituted or unsubstituted C 3–7 cycloalkyl, and substituted or unsubstituted C 2 –C 7 heterocycloalkyl.

[0196] In some embodiments, W is substituted or unsubstituted C 1 -C 2 alkylene. In some embodiments, W is C 1 -C 2 alkylene substituted with 1, 2, 3, or 4 substituents each independently selected from F, -OH, -OCH 3 , and -CH 3 . In some embodiments, W is–CH 2 –,–CHF–,–CH(CH 3 )–,–CH(OH)–,–CH(OCH 3 )–,–CF 2 –,– CH 2 CH 2 –,–CHFCH 2 –,–CH 2 CHF–,–CH(CH 3 )CH 2 –,–CH 2 CH(CH 3 )–,–CH(OH)CH 2 –,–CH 2 CH(OH)–,– CH(OCH 3 )CH 2 –,–CH 2 CH(OCH 3 )–,–CF 2 CH 2 –, or–CH 2 CF 2 –. In some embodiments, W is–CHFCH 2 –,– CH 2 CHF–,–CF 2 CH 2 –, or–CH 2 CF 2 –.

[0197] In some embodiments, W is substituted or unsubstituted C 3 -C 4 alkylene. In some embodiments, W is C 3 -C 4 alkylene substituted with 1, 2, 3, or 4 substituents each independently selected from the group consisting of F, -OH, -OCH 3 , and -CH 3 . In some embodiments, W is–CH 2 CH 2 CH 2 –,–CHFCH 2 CH 2 –,– CH 2 CHFCH 2 –,–CH 2 CH 2 CHF–,–CF 2 CH 2 CH 2 –,–CH 2 CF 2 CH 2 –,–CH 2 CH 2 CF 2 –,–CH(OH)CH 2 CH 2 –,– CH2CH(OH)CH2–,–CH2CH2CH(OH)–,–CH(OCH3)CH2CH2–,–C H2CH(OCH3)CH2–,–CH2CH2CH(OCH3)–, ––CH(CH 3 )CH 2 CH 2 –,–CH 2 CH(CH 3 )CH 2 –, or–CH 2 CH 2 CH(CH 3 )–. In some embodiments, W is–

CH 2 CHFCH 2 – or–CH 2 CF 2 CH 2 –. In some embodiments, W is–CHFCH 2 CH 2 – or–CF 2 CH 2 CH 2 –.

[0198] In some embodiments, W is–CH 2 CH 2 CHF– or–CH 2 CH 2 CF 2 –.

[0199] In some embodiments, W is substituted or unsubstituted C 1 -C 3 alkylene. In some embodiments, W is– CH 2 -. In some embodiments, W is–CH 2 CH 2 -. In some embodiments, W is–CH 2 CH 2 CH 2 -. In some embodiments, W is substituted or unsubstituted C 3 –C 8 cycloalkylene or substituted or unsubstituted C 2 -C 3 alkenylene. In some embodiments, W is substituted or unsubstituted C 3 –C 8 cycloalkylene. In some embodiments, W is substituted or unsubstituted cyclopropylene. In some embodiments, W is substituted or unsubstituted C 2 -C 3 alkenylene. In some embodiments, W is–CH=CH-. In some embodiments, W is substituted or unsubstituted C 1 -C 2 heteroalkylene. In some embodiments, W is substituted or unsubstituted– CH 2 OCH 2 –.

[0200] In some embodiments, each R 16 and R 17 is selected from the group consisting of hydrogen, F,–OR 1 , substituted or unsubstituted C 1 –C 6 alkyl, a substituted or unsubstituted C 1 –C 6 fluoroalkyl, and substituted or unsubstituted C 1 –C 6 heteroalkyl. In some embodiments, each R 16 and R 17 is selected from the group consisting of hydrogen, F, -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, -OCH 2 CN, -OCF 3 , -CH 3 , -CH 2 CH 3 , -CH 2 OH, - CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , and -CH 2 CF 3 . In some embodiments, each R 16 and R 17 is selected from the group consisting of hydrogen, F, -OH, -OCH 3 , -OCF 3 , -CH 3 , -CH 2 OH, - CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 16 is hydrogen. In some embodiments, R 17 is hydrogen.

[0201] In some embodiments, R 2 is hydrogen.

[0202] In some embodiments, R is substituted or unsubstituted C 1 –C 4 alkyl, substituted or unsubstituted C 1 – C 4 fluoroalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, substituted or unsubstituted C 3 –C 5 cycloalkyl, or substituted or unsubstituted C 2 –C 4 heterocycloalkyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , - CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH 2 CH 2 OH, -C(OH)(CH 3 ) 2 , -CH 2 CN, - CH 2 C(=O)OCH 3 , -CH 2 C(=O)OCH 2 CH 3 , -CH 2 C(=O)NHCH 3 , -CH 2 C(=O)N(CH 3 ) 2 , -CH 2 NH 2 , -CH 2 NHCH 3 , - CH 2 N(CH 3 ) 2 , -CH 2 F, -CHF 2 , -CF 3 , cyclopropyl, cyclobutyl, oxetanyl, aziridinyl, or azetidinyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH 2 CH 2 OH, - CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , cyclopropyl, or oxetanyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , cyclopropyl, or oxetanyl. In some embodiments, R is -CH 3 , - CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, cyclopropyl, or oxetanyl. In some embodiments, R is -CH 3 , - CH 2 OH, -CH 2 CN, -CHF 2 , -CF 3 , or cyclopropyl. In some embodiments, R is -CH 3 , -CH 2 CH 3 , -CH 2 F, -CHF 2 , - CF 3 , cyclopropyl, or oxetanyl.

[0203] In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F,–OR 1 , substituted or unsubstituted C 1 –C 3 alkyl, substituted or unsubstituted C 1 –C 3 fluoroalkyl, and substituted or unsubstituted C 1 –C 3 heteroalkyl. In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 NHCH 3 , - CH2N(CH3)2, -OH, -OCH3, -OCH2CH3, -OCH2CH2OH, -OCH2CN, -OCF3, -CH2F, -CHF2, and -CF3. In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F, -CH 3 , -CH 2 OH, - OCH 2 CN, -OH, -OCH 3 , -OCH 2 CN, -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are the same and selected from the group consisting of F, -CH 3 , -OCH 3 , -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are F. In some embodiments, R 15 and R 18 are -CH 3 .

[0204] In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 NHCH 3 , - CH 2 N(CH 3 ) 2 , -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, -OCH 2 CN, -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH 3 , -CH 2 OH, -OCH 2 CN, -OH, -OCH 3 , -OCH 2 CN, -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, deuterium, F, -CH 3 , -OCH 3 , -OCF 3 , -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 15 and R 18 are not the same and selected from the group consisting of hydrogen, F, -CH 3 , and -OCH 3 . In some embodiments, R 15 is hydrogen and R 18 is -CH 3 . In some embodiments, R 15 is -CH 3 and R 18 is hydrogen.

[0205] In some embodiments, R 3 is hydrogen, -CN,–OR 1 ,–N(R 1 ) 2 , substituted or unsubstituted C 1 –C 4 alkyl, - CD 3 , substituted or unsubstituted C 1 –C 4 haloalkyl, substituted or unsubstituted C 1 –C 4 heteroalkyl, -C 1 -C 4 alkylene-OR 1 , or substituted or unsubstituted C 3 –C 5 cycloalkyl. In some embodiments, R 3 is substituted or unsubstituted C 1 –C 4 alkyl, -C 1 -C 4 alkylene-OR 1 , or substituted or unsubstituted C 3 –C 5 cycloalkyl. In some embodiments, R 3 is -CH 3 , -CH 2 CH 3 , cyclopropyl, or–CH 2 CH 2 OCH 3 .

[0206] In some embodiments, R 3 is -CH 3 .

[0207] In some embodiments, A is -CR A =CR A -.

[0208] In some embodiments, each R A is independently hydrogen, F, Cl, -CN, -CH 3 , -CH 2 CH 3 , - CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -OH, -OCH 3 , -OCH 2 CH 3 , -OCF 3 , -CH 2 F, -CHF 2 , or -CF 3 . In some embodiments, each R A is independently hydrogen, F, Cl, -CN, -CH 3 , -OH, -OCH 3 , -OCF 3 , -CH 2 F, -CHF 2 , or -CF 3 . In some embodiments, each R A is independently hydrogen, F, Cl, -CN, -CH 3 , or -OCH 3 . In some embodiments, each R A is independently hydrogen, F, Cl, or -CH 3 . In some embodiments, R A is hydrogen.

[0209] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 are F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 are F.

[0210] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine, e.g., F or C 1 –C 4 fluoroalkyl such as CH 2 F, CF 3 , CHF 2 , and CH 3 CH 2 F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 is F or C 1 –C 4 fluoroalkyl. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprise a fluorine. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprises a fluorine. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprise a fluorine.

[0211] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine, e.g., F or C 1 –C 4 fluoroalkyl such as CH 2 F, CF 3 , CHF 2 , and CH 3 CH 2 F. In some embodiments, one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , and R 18 comprises a fluorine. In some embodiments, W comprises a fluorine.

[0212] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 11 is H, D, or F. In some embodiments, R 11 is D. In some embodiments, R 11 is H. In some embodiments, R 11 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 12 is H, D, or F. In some embodiments, R 12 is D. In some embodiments, R 12 is H. In some embodiments, R 12 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 13 is H, D, or F. In some embodiments, R 13 is D. In some embodiments, R 13 is H. In some embodiments, R 13 is F. In some embodiments of a compound of Formula (I) or (I*), or a

pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 14 is H, D, or F. In some embodiments, R 14 is D. In some embodiments, R 14 is H. In some embodiments, R 14 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 15 is H, D, F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 15 is H or D. In some embodiments, R 15 is H. In some embodiments, R 15 is D. In some embodiments, R 15 is F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 15 is F, CF 3 , CHF 2 , or CH 2 F. In some embodiments, R 15 is F.

[0213] In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 16 is H, D, or F. In some embodiments, R 16 is D. In In some embodiments, R 16 is H. some embodiments, R 16 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 17 is H, D, or F. In some embodiments, R 17 is D. In some embodiments, R 17 is H. In some embodiments, R 17 is F. In some embodiments of a compound of Formula (I) or (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 18 is H, D, F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 18 is H or D. In some embodiments, R 18 is H. In some embodiments, R 18 is D. In some embodiments, R 18 is F, CH 2 F, CHF 2 , CF 3 , or CH 3 . In some embodiments, R 18 is F, CF 3 , CHF 2 , or CH 2 F. In some embodiments, R 18 is F.

[0214] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 11 , R 12 , R 19 , R 20 and R 16 are hydrogen. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 19 is hydrogen. In some embodiments, R 19 is H, F, -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, - OCH 2 CN, -OCF 3 , -CH 3 , -CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CH 2 F, - CH2CHF2, and -CH2CF3. In some embodiments, R 19 is H, F, -OH, -OCH3, -OCF3, -CH3, -CH2OH, -CH2F, - CHF 2 , and -CF 3 . In some embodiments, R 19 is F or -OCH 3 .

[0215] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 20 is hydrogen. In some embodiments, R 20 is H, F, -OH, -OCH 3 , -OCH 2 CH 3 , -OCH 2 CH 2 OH, -OCH 2 CN, -OCF 3 , -CH 3 , -CH 2 CH 3 , -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CN, -CH 2 F, - CHF 2 , -CF 3 , -CH 2 CH 2 F, -CH 2 CHF 2 , and -CH 2 CF 3 . In some embodiments, R 20 is H, F, -OH, -OCH 3 , -OCF 3 , - CH 3 , -CH 2 OH, -CH 2 F, -CHF 2 , and -CF 3 . In some embodiments, R 20 is F or -OCH 3 .

[0216] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 19 is H, D, or F. In some embodiments, R 19 is D. In some embodiments, R 19 is H. In some embodiments, R 19 is F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 20 is H, D, or F. In some embodiments, R 20 is D. In some embodiments, R 20 is H. In some embodiments, R 20 is F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 16 and R 19 are H. In some embodiments, R 16 and R 19 are D. In some embodiments, R 16 and R 19 are F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 19 and R 20 are H. In some embodiments, R 19 and R 20 are D. In some embodiments, R 19 and R 20 are F. In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, R 17 and R 20 are H. In some embodiments, R 17 and R 20 are D. In some embodiments, R 17 and R 20 are F.

[0217] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 are F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 is F. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 is F. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 are F.

[0218] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine, e.g., F or C 1 –C 4 fluoroalkyl such as CH 2 F, CF 3 , CHF 2 , and CH 3 CH 2 F. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 is F or C 1 –C 4 fluoroalkyl. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprise a fluorine. In some embodiments, at least one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 comprises a fluorine. In some embodiments, one of R 11 , R 12 , R 13 , R 14 , R 16 , R 19 , R 20 , and R 17 comprises a fluorine. In some embodiments, at least two of R 11 , R 12 , R 13 , R 14 , R 16 , and R 17 comprise a fluorine.

[0219] In some embodiments of a compound of Formula (I*), or a pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof, at least one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine, e.g., F or C1–C4 fluoroalkyl such as CH2F, CF3, CHF2, and CH3CH2F. In some embodiments, one of W, R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 19 , R 20 , and R 18 comprises a fluorine. In some embodiments, W comprises a fluorine.

[0220] In some embodiments, a compound of Formula (I) or (I*) is a single enantiomer. In some embodiments, a compound of Formula (I) or (I*) is not racemic. In some embodiments, a compound of Formula (I) or (I*) is substantially free of other isomers. In some embodiments, a compound of Formula (I) or (I*) is a single isomer substantially free of other isomers. In some embodiments, a compound of Formula (I) or (I*) comprises 25% or less of other isomers. In some embodiments, the compound of Formula (I) or (I*) comprises 20% or less of other isomers. In some embodiments, a compound of Formula (I) or (I*) comprises 15% or less of other isomers. In some embodiments, a compound of Formula (I) or (I*) comprises 10% or less of other isomers. In some embodiments, the compound of Formula (I) or (I*) comprises 5% or less of other isomers. In some embodiments, the compound of Formula (I) or (I*) comprises 1% or less of other isomers.

[0221] In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 75%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 80%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 85%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 90%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 95%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 96%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 97%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 98%. In some embodiments, a compound of Formula (I) or (I*) has a stereochemical purity of at least 98%.

[0222] In some embodiments, an asymmetric carbon atom of a compound of Formula (I) or (I*) is present in enantiomerically enriched form. In certain embodiments, the asymmetric carbon atom of the compound of Formula (I) or (I*) has at least 50% enantiomeric excess, at least 60 % enantiomeric excess, at least 70 % enantiomeric excess, at least 80 % enantiomeric excess, at least 90 % enantiomeric excess, at least 95 % enantiomeric excess, or at least 99 % enantiomeric excess in the (S)- or (R)-configuration.

[0223] Exemplary SMSM compounds are summarized in Table 1A.

Table 1A: Exemplary SMSM compounds

[0224] In some embodiments, an SMSM described herein is not a compound in Table IB.

[0225] In some embodiments, R 16 and R 17 are not simultaneously hydrogen or deuterium. In some embodiments, R 16 and R 17 are not simultaneously hydrogen.

[0226] In some embodiments, when (i) W is -CH 2 -, -CH 2 CH 2 -, -CH 2 OCH 2 -, or -CH 2 CH 2 CH 2 -, (ii) A is - CH=CH-, and (iii) R 16 and R 17 are both hydrogen, R 3 is not -CH 3 or -CH 2 CH 2 F.

[0227] In some embodiments, when (i) W is -CH 2 -, -CH 2 CH 2 -, -CH 2 OCH 2 -, or -CH 2 CH 2 CH 2 -, (ii) A is - CH=CH-, and (iii) R 15 , R 18 , R 16 and R 17 are hydrogen, then R 3 is not -CH 3 or -CH 2 CH 2 F.

[0228] In some embodiments, R 3 is not -CH 3 or -CH 2 CH 2 F.

[0229] In some embodiments, when R 16 and R 17 are both hydrogen, R 3 is not -CH 3 or -CH 2 CH 2 F.

[0230] In some embodiments, when (i) A is -CH=CH- and (ii) R 16 and R 17 are both hydrogen, then R 3 is not - CH 3 or -CH 2 CH 2 F. In some embodiments, when (i) A is -CH=CH- and (ii) R 3 is -CH 3 or -CH 2 CH 2 F, then R 16 and R 17 are not simultaneously hydrogen.

[0231] In some embodiments, ring Q is not

[0232] Exemplary SMSM compounds are summarized in Table 1C.

Table 1 C: Exemplary SMSM compounds

[0233] In some embodiments, an SMSM described herein is not a compound in Table ID.

[0234] In some embodiments, the compound of Formula (I) or (I*) is not

[0235] In some embodiments, ring Q is not

[0236] In some embodiments, when (i) W is -CH 2 CH 2 -, (ii) A is–CH=CH-, (iii) R 16 , R 17 , R 15 and R 18 are hydrogen, and (iv) R 3 is–CH 3 , then R is not–C(=O)CH 2 CH 2 NH 2 .

[0237] In some embodiments, when (i) W is -CH 2 CH 2 -, (ii) A is–CH=CH-, and (iii) R 16 , R 17 , R 15 and R 18 are hydrogen, then R is not–C(=O)CH 2 CH 2 NH 2 .

[0238] In some embodiments, R is not–C(=O)CH 2 CH 2 NH 2 . In some embodiments, R is not substituted by oxo. [0239] Exemplary SMSM compounds are summarized in Table 1E.

[0240] In some embodiments, an SMSM descriebd herein is not a compound of Table 1F.

Table 1F: List of compounds

[0241] In some embodiments, the compound of Formula (I) or (I*), when (i) W is -CH 2 CH 2 - or - CH 2 CH 2 CH 2 -, (ii) R 16 and R 17 are hydrogen, (iii) R 15 and R 18 are–CH 3 , and (iv) A is–CH=CH-, then R is not –CH 2 -C 6 H 4 -O-CH 3 or–C(=O)OC(CH 3 ) 3 . [0242] In some embodiments, the compound of Formula (I) or (I*), when (i) W is -CH 2 CH 2 - or - CH 2 CH 2 CH 2 -, (ii) R 15 and R 18 are–CH 3 , and (iii) A is–CH=CH-, then R is not–CH 2 -C 6 H 4 -O-CH 3 ,– C(=O)OC(CH 3 ) 3 , or COO .

[0243] In some embodiments, the compound of Formula (I) or (I*), when (i) W is -CH 2 CH 2 - or - CH 2 CH 2 CH 2 -, (ii) R 16 and R 17 are hydrogen, and (iii) R 15 and R 18 are–CH 3 , then R is not–CH 2 -C 6 H 4 -O-CH 3 or–C(=O)OC(CH 3 ) 3 .

[0244] In some embodiments, when (i) W is -CH 2 CH 2 - or -CH 2 CH 2 CH 2 -, (ii) A is–CH=CH-, and (iii) R 15 and R 18 are–CH 3 , then R 16 and R 17 are not simultaneously hydrogen. [0245] In some embodiments, R is not . In some embodiments, R is not - C(=O)OC(CH 3 ) 3 . In some embodiments, R is not COO .

[0246] In some embodiments, when R 15 and R 18 are–CH 17

3, R 16 and R are not both hydrogen. In some embodiments, when R 16 and R 17 are hydrogen, R 15 and R 18 are not simultanesouly–CH 3 .

[0247] In some embodiments, when (i) R 15 and R 18 are–CH 3 and (ii) W is -CH 2 CH 2 - or -CH 2 CH 2 CH 2 -, then R 16 and R 17 are not both hydrogen.

[0248] In some embodiments, when (i) R 15 and R 18 are–CH 3 and (ii) A is–CH=CH-, then R 16 and R 17 are not both hydrogen.

[0249] In some embodiments, when R 17 is -C(CH 3 ) 3, R 16 is not hydrogen. In some embodiments, when R 16 is hydrogen, R 17 is not -C(CH 3 ) 3. In some embodiments, when R 16 is -C(CH 3 ) 3, R 17 is not hydrogen. In some embodiments, when R 17 is hydrogen, R 16 is not -C(CH 3 ) 3. [0250] In some embodiments, the compound of Formula (I) or (I*) is a compound in Table 1G.

Table 1G: Exemplary SMSM compounds

[0251] In some embodiments, the compound of Formula (I) or (I*) is a compound in Table 1H.

Table 1H: Exemplary SMSM compounds

[0252] In some embodiments, an SMSM described herein, possesses one or more stereocenters and each stereocenter exists independently in either the R or S configuration. The compounds presented herein include all diastereomeric, enantiomeric, and epimeric forms as well as the appropriate mixtures thereof. The compounds and methods provided herein include all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. In certain embodiments, compounds described herein are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds/salts, separating the diastereomers and recovering the optically pure enantiomers. In some embodiments, resolution of enantiomers is carried out using covalent diastereomeric derivatives of the compounds described herein. In another embodiment, diastereomers are separated by separation/resolution techniques based upon differences in solubility. In other embodiments, separation of stereoisomers is performed by chromatography or by the forming diastereomeric salts and separation by recrystallization, or chromatography, or any combination thereof. Jean Jacques, Andre Collet, Samuel H. Wilen,“Enantiomers, Racemates and Resolutions”, John Wiley And Sons, Inc., 1981. In one aspect, stereoisomers are obtained by stereoselective synthesis.

[0253] In some embodiments, compounds described herein are prepared as prodrugs. A“prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. In some embodiments, the design of a prodrug increases the effective water solubility. An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the“prodrug”) to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial. A further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound. In certain embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.

[0254] In one aspect, prodrugs are designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. By virtue of knowledge of pharmacokinetic, pharmacodynamic processes and drug metabolism in vivo, once a pharmaceutically active compound is known, the design of prodrugs of the compound is possible. (see, for example, Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392; Silverman (1992), The Organic Chemistry of Drug Design and Drug Action, Academic Press, Inc., San Diego, pages 352-401, Rooseboom et al., Pharmacological Reviews, 56:53-102, 2004; Aesop Cho,“Recent Advances in Oral Prodrug Discovery”, Annual Reports in Medicinal Chemistry, Vol. 41, 395-407, 2006; T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series).

[0255] In some cases, some of the herein-described compounds may be a prodrug for another derivative or active compound. [0256] In some embodiments, sites on the aromatic ring portion of compounds described herein are susceptible to various metabolic reactions Therefore incorporation of appropriate substituents on the aromatic ring structures will reduce, minimize or eliminate this metabolic pathway. In specific embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, or an alkyl group.

[0257] In another embodiment, the compounds described herein are labeled isotopically (e.g. with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.

[0258] Compounds described herein include isotopically-labeled compounds, which are identical to those recited in the various formulae and structures presented herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the present compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, sulfur, fluorine and chlorine, such as, for example, 2 H, 3

3 h, 1 C, 14 C, 15 N, 18 O, 17 O, 35 S, 18 F, l. In one aspect, isotopically-labeled compounds described herein, for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. In one aspect, substitution with isotopes such as deuterium affords certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements.

[0259] In additional or further embodiments, the compounds described herein are metabolized upon administration to an organism in need to produce a metabolite that is then used to produce a desired effect, including a desired therapeutic effect.

[0260] Compounds described herein may be formed as, and/or used as, pharmaceutically acceptable salts. The type of pharmaceutical acceptable salts, include, but are not limited to: (1) acid addition salts, formed by reacting the free base form of the compound with a pharmaceutically acceptable: inorganic acid, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, metaphosphoric acid, and the like; or with an organic acid, such as, for example, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, trifluoroacetic acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, toluenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene- 1-carboxylic acid, glucoheptonic acid, 4,4’-methylenebis-(3-hydroxy-2-ene-1-carboxylic acid), 3- phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, butyric acid, phenylacetic acid, phenylbutyric acid, valproic acid, and the like; (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion (e.g. lithium, sodium, potassium), an alkaline earth ion (e.g. magnesium, or calcium), or an aluminum ion. In some cases, compounds described herein may coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine. In other cases, compounds described herein may form salts with amino acids such as, but not limited to, arginine, lysine, and the like. Acceptable inorganic bases used to form salts with compounds that include an acidic proton, include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.

[0261] It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms, particularly solvates. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. In some embodiments, solvates of compounds described herein are conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.

[0262] In some embodiments, an SMSM has a molecular weight of at most about 2000 Daltons, 1500 Daltons, 1000 Daltons or 900 Daltons. In some embodiments, an SMSM has a molecular weight of at least 100 Daltons, 200 Daltons, 300 Daltons, 400 Daltons or 500 Daltons. In some embodiments, an SMSM does not comprise a phosphodiester linkage.

Methods of Making Compounds

[0263] Compounds described herein can be synthesized using standard synthetic techniques or using methods known in the art in combination with methods described herein. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology can be employed. Compounds can be prepared using standard organic chemistry techniques such as those described in, for example, March’s Advanced Organic Chemistry, 6th Edition, John Wiley and Sons, Inc. Alternative reaction conditions for the synthetic transformations described herein may be employed such as variation of solvent, reaction temperature, reaction time, as well as different chemical reagents and other reaction conditions. The starting materials can be available from commercial sources or can be readily prepared. By way of example only, provided are schemes for preparing the exemplary SMSMs.

[0264] Scheme 1

[0265] Scheme 2

[0266] Scheme 3

[0267] Scheme 4

[0268] Scheme 5

[0269] Scheme 6

[0270] Scheme 7

[0271] Scheme 8

[0272] Scheme 9

[0273] Scheme 10

[0274] Scheme 11

[0275] Scheme 12

[0276] Scheme A

[0277] Scheme B

[0278] Scheme C

[0279] Scheme D

[0280] Scheme E

[0281] Scheme F

[0282] Scheme G

[0283] Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, “Synthetic Organic Chemistry”, John Wiley & Sons, Inc., New York; S. R. Sandler et al.,“Organic Functional Group Preparations,” 2nd Ed., Academic Press, New York, 1983; H. O. House,“Modern Synthetic Reactions”, 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist,“Heterocyclic Chemistry”, 2nd Ed., John Wiley & Sons, New York, 1992; J. March,“Advanced Organic Chemistry: Reactions, Mechanisms and Structure”, 4th Ed., Wiley Interscience, New York, 1992. Additional suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, Fuhrhop, J. and Penzlin G.“Organic Synthesis: Concepts, Methods, Starting Materials”, Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3 527-29074-5; Hoffman, R.V.“Organic Chemistry, An Intermediate Text” (1996) Oxford University Press, ISBN 0-19-509618-5; Larock, R. C.“Comprehensive Organic Transformations: A Guide to Functional Group Preparations” 2nd Edition (1999) Wiley-VCH, ISBN: 0-471-19031-4; March, J.“Advanced Organic Chemistry: Reactions, Mechanisms, and Structure” 4th Edition (1992) John Wiley & Sons, ISBN: 0-471-60180- 2; Otera, J. (editor)“Modern Carbonyl Chemistry” (2000) Wiley-VCH, ISBN: 3-527-29871-1; Patai, S.“Patai’s 1992 Guide to the Chemistry of Functional Groups” (1992) Interscience ISBN: 0-471-93022-9; Solomons, T. W. G.“Organic Chemistry” 7th Edition (2000) John Wiley & Sons, ISBN: 0-471-19095-0; Stowell, J.C., “Intermediate Organic Chemistry” 2nd Edition (1993) Wiley-Interscience, ISBN: 0-471-57456-2;“Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann’s Encyclopedia” (1999) John Wiley & Sons, ISBN: 3-527-29645-X, in 8 volumes;“Organic Reactions” (1942-2000) John Wiley & Sons, in over 55 volumes; and“Chemistry of Functional Groups” John Wiley & Sons, in 73 volumes.

[0284] In the reactions described, it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, in order to avoid their unwanted participation in reactions. A detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure).

[0285] SMSMs can be made using known techniques and further chemically modified, in some embodiments, to facilitate intranuclear transfer to, e.g., a splicing complex component, a spliceosome or a pre-mRNA molecule. One of ordinary skill in the art will appreciate the standard medicinal chemistry approaches for chemical modifications for intranuclear transfer (e.g., reducing charge, optimizing size, and/or modifying lipophilicity).

Pharmaceutical Compositions

[0286] In some embodiments, the compounds described herein are formulated into pharmaceutical compositions. Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. A summary of pharmaceutical compositions described herein can be found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins1999), herein incorporated by reference for such disclosure.

[0287] A pharmaceutical composition can be a mixture of an SMSM described herein with one or more other chemical components (i.e. pharmaceutically acceptable ingredients), such as carriers, excipients, binders, filling agents, suspending agents, flavoring agents, sweetening agents, disintegrating agents, dispersing agents, surfactants, lubricants, colorants, diluents, solubilizers, moistening agents, plasticizers, stabilizers, penetration enhancers, wetting agents, anti-foaming agents, antioxidants, preservatives, or one or more combination thereof. The pharmaceutical composition facilitates administration of the compound to an organism.

[0288] The compositions described herein can be administered to the subject in a variety of ways, including parenterally, intravenously, intradermally, intramuscularly, colonically, rectally or intraperitoneally. In some embodiments, the small molecule splicing modulator or a pharmaceutically acceptable salt thereof is administered by intraperitoneal injection, intramuscular injection, subcutaneous injection, or intravenous injection of the subject. In some embodiments, the pharmaceutical compositions can be administered parenterally, intravenously, intramuscularly or orally. The oral agents comprising a small molecule splicing modulator can be in any suitable form for oral administration, such as liquid, tablets, capsules, or the like. The oral formulations can be further coated or treated to prevent or reduce dissolution in stomach. The compositions of the present invention can be administered to a subject using any suitable methods known in the art. Suitable formulations for use in the present invention and methods of delivery are generally well known in the art. For example, the small molecule splicing modulators described herein can be formulated as pharmaceutical compositions with a pharmaceutically acceptable diluent, carrier or excipient. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions including pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, such as, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.

[0289] Pharmaceutical formulations described herein can be administrable to a subject in a variety of ways by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular, intramedullary injections, intrathecal, direct intraventricular, intraperitoneal, intralymphatic, intranasal injections), intranasal, buccal, topical or transdermal administration routes. The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations. [0290] In some embodiments, the pharmaceutical formulation is in the form of a tablet. In other embodiments, pharmaceutical formulations containing an SMSM described herein are in the form of a capsule. In one aspect, liquid formulation dosage forms for oral administration are in the form of aqueous suspensions or solutions selected from the group including, but not limited to, aqueous oral dispersions, emulsions, solutions, elixirs, gels, and syrups.

[0291] For administration by inhalation, an SMSM described herein can be formulated for use as an aerosol, a mist or a powder. For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, or gels formulated in a conventional manner. In some embodiments, an SMSM described herein can be prepared as transdermal dosage forms. In some embodiments, an SMSM described herein can be formulated into a pharmaceutical composition suitable for intramuscular, subcutaneous, or intravenous injection. In some embodiments, an SMSM described herein can be administered topically and can be formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. In some embodiments, an SMSM described herein can be formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas.

Splicing

[0292] Extensive posttranscriptional processing occurs before eukaryotic pre-mRNA matures and exits from the nucleus to the cytoplasm, including the addition of a 7-methylguanosine cap at the 5’ end, the cleavage and addition of a poly-A tail at the 3’ end as well as the removal of intervening sequences or introns by the spliceosome. The vast majority of higher eukaryotic genes contain multiple introns that are spliced out with high precision and fidelity in order to maintain the reading frame of the exons. Splicing of pre-mRNA can utilize the recognition of short consensus sequences at the boundaries and within introns and exons by an array of small nuclear ribonucleoprotein (snRNP) complexes (e.g., snRNPs U1, U2, U4, U5, U6, U11, U12m U4atc and U6 atc) and a large number of proteins, including spliceosomal proteins and positively as well as negatively acting splicing modulators.

[0293] Serine-arginine-rich (SR)-domain-containing proteins generally serve to promote constitutive splicing. They can also modulate alternative splicing by binding to intronic or exonic splicing enhancer (ISE) or ESE, respectively) sequences. Other pre-mRNA binding proteins, such as hnRNPs, regulate splicing by binding to intronic or exonic splicing suppressor (ISS or ESS, respectively) sequences and can also act as general splicing modulators. The SR protein family is a class of at least 10 proteins that have a characteristic serine/arginine rich domain in addition to an RNA-binding. SR proteins are generally thought to enhance splicing by simultaneously binding to U170K, a core component of the U1 snRNP, at the 5’ splice site, and the U2AF35 at the 3’ splice site, thus bridging the two ends of the intron. While this particular function of SR proteins seems to be redundant, as any individual SR protein can commit a pre-mRNA for constitutive splicing, the role of the various SR proteins in alternative splicing of specific pre-mRNAs is distinct due in part to their ability to recognize and bind to unique consensus sequences. Phosphorylation of the RS domain of SR proteins can lead to the regulation of their protein interactions, RNA binding, localization, trafficking, and role in alternative splicing. Several cellular kinases that phosphorylate SR proteins have been identified, including SR protein Kinase (SRPKs), Cdc2-like kinases (Clks), pre-mRNA processing mutant 4 (PRP4), and topoisomerase I. Optimal phosphorylation of SR proteins may be required for proper functioning as both hypo- and hyperphosphorylation of the RS domains may be detrimental to their role in constitutive and alternative splicing.

[0294] In higher eukaryotes, the vast majority of genes contain one or more introns, which creates a situation in which the exons are spliced together to generate mature mRNA and microRNA (miRNA). In the host nucleus, pre-mRNA splicing is the mechanism by which introns are removed from a pre-mRNA and the exons are ligated together to generate mature mRNAs and pre-miRNA that is then exported to the cytoplasm for translation into the polypeptide gene product. Splicing of pre-mRNA can occur in cis, where two exons derive from two adjacent cotranscribed sequences, or in trans, when the two exons come from different pre-mRNA transcripts. The ratio of the different protein products (isoforms) may be due to the frequency of alternative splicing events within a pre-mRNA that leads to different amounts of distinct splice variants. In some embodiments, alternative splicing of a pre-mRNA may lead to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 protein isoforms being expressed.

[0295] Aberrations in splicing are thought to be the cause of roughly half of all inherited diseases. Aberrant splicing due to mutations in consensus sequences involved in exon-intron boundary recognition is responsible for up to 15% of inherited diseases. In addition, defects in the splicing machinery itself due to the loss or gain of function of splicing factors and modulators are causes of a wide range of human ailments from cancer to neurodegenerative diseases. Both constitutive and alternative splicing are subject to regulation by upstream signaling pathways. This regulation can be essential during development, in tissue specific expression of certain isoforms, during the cell cycle and in response to extrinsic signaling molecules.

[0296] Alternative splicing allows for a single gene to express different isoforms of mRNA, thus playing a major role in contributing to the cellular complexity in higher eukaryotes without the need to expand the genome. Splicing can also be subject to regulation by upstream signaling pathways. For example, an upstream signaling pathway may modulate alternative splicing and increase or decrease expression levels of different isoforms of mRNA.

[0297] Alternative splicing events are highly regulated by numerous splicing factors in a tissue type-, developmental stage-, and signal-dependent manner. Furthermore, non-mutation based causes of splicing defects and defects in the splicing machinery itself, e.g., due to the loss/gain of function of splicing factors or their relative stoichiometry, cause of a wide range of human ailments, ranging from cancer to neurodegenerative diseases. In many diseases the disease state is caused by an alteration of the ratio of different isoforms of two or more proteins expressed from a gene. In some embodiments, the alteration in the ratio of the protein products is due to changes in the frequency of alternative splicing events within a pre-mRNA, leading to changes in the ratio of splice variants produced. In some embodiments, alternative splicing of a pre-mRNA may lead to 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 protein isoforms being expressed. In some embodiments, a change in the splice variant ratio is caused by genetic mutation. [0298] In eukaryotes, the vast majority of splicing processes are catalyzed by the spliceosome, an RNA-protein complex that occurs in unique steps and may comprise a subset of several hundred different proteins, in addition to five spliceosomal snRNAs. These factors are responsible for the accurate positioning of the spliceosome on the 5’ and 3’ splice site sequences. The reason why so many factors are needed reflects the observation that exon recognition can be affected by many pre-mRNA features such as exon length, sequence recognition, the presence of enhancer and silencer elements, the strength of upstream splicing signals, the promoter architecture, and the rate of RNA processivity, secondary and tertiary RNA structure.

[0299] All mammalian diseases are ultimately mediated by the transcriptome. Insofar as messenger mRNA (mRNA) is part of the transcriptome, and all protein expression derives from mRNAs, there is the potential to intervene in protein-mediated diseases by modulating the expression of the relevant protein and by, in turn, modulating the translation of the corresponding upstream mRNA. But mRNA is only a small portion of the transcriptome: other transcribed RNAs also regulate cellular biology either directly by the structure and function of RNA structures (e.g., ribonucleoproteins) as well as via protein expression and action, including (but not limited to) microRNA (miRNA), long noncoding RNA (lncRNA), long intergenic noncoding RNA (lincRNA), small nucleolar RNA (snoRNA), small nuclear RNA (snRNA), small Cajal body-specific RNA (scaRNA), piwi- interacting RNA (piRNA), competing endogenous (ceRNA), and pseudo-genes. Drugs that intervene at this level have the potential of modulating any and all cellular processes. Existing therapeutic modalities such as antisense RNA or siRNA, in most cases, have yet to overcome significant challenges such as drug delivery, absorption, distribution to target organs, pharmacokinetics, and cell penetration. In contrast, small molecules have a long history of successfully surmounting these barriers and these qualities, which make them suitable as drugs, are readily optimized through a series of analogues to overcome such challenges. In sharp contrast, the application of small molecules as ligands for RNA that yield therapeutic benefit has received little to no attention from the drug discovery community.

[0300] DNA sequences in the chromosome are transcribed into pre-mRNAs which contain coding regions (exons) and generally also contain intervening non-coding regions (introns). Introns are removed from pre- mRNAs through splicing. Pre-mRNA splicing proceeds by a two-step mechanism. In the first step, the 5’ splice site is cleaved, resulting in a“free” 5’ exon and a lariat intermediate. In the second step, the 5’ exon is ligated to the 3’ exon with release of the intron as the lariat product. These steps are catalyzed in a complex of small nuclear ribonucleoproteins and proteins called the spliceosome.

[0301] In most cases, the splicing reaction occurs within the same pre-mRNA molecule, which is termed cis- splicing. Splicing between two independently transcribed pre-mRNAs is termed trans-splicing.

[0302] Introns are portions of eukaryotic DNA, which intervene between the coding portions, or“exons,” of that DNA. Introns and exons are transcribed into RNA termed“primary transcript, precursor to mRNA” (or “pre-mRNA”). Introns can be removed from the pre-mRNA so that the native protein encoded by the exons can be produced (the term“native protein” as used herein refers to naturally occurring, wild type, or functional protein). The removal of introns from pre-mRNA and subsequent joining of the exons is carried out in the splicing process. [0303] The splicing process is a series of reactions, which are carried out on RNA after transcription but before translation and which are mediated by splicing factors. Thus, a“pre-mRNA” can be an RNA that contains both exons and intron(s), and a mature mRNA (“mRNA”) can be an RNA in which the intron(s) have been removed and the exons joined together sequentially so that the protein may be translated therefrom by the ribosomes.

[0304] Introns can be defined by a set of“splice elements” that are part of the splicing machinery and may be required for splicing and which are relatively short, conserved RNA segments that bind the various splicing factors, which carry out the splicing reactions. Thus, each intron is defined by a 5’ splice site, a 3’ splice site, and a branch point situated there between. Splice elements also comprise exon splicing enhancers and silencers, situated in exons, and intron splicing enhancers and silencers situated in introns at a distance from the splice sites and branch points. In addition to splice site and branch points these elements control alternative aberrant and constitutive splicing.

[0305] Initial RNA transcripts (pre-mRNA) of most eukaryotic genes are retained in the nucleus until non- coding intron sequences are removed by the spliceosome to produce mature messenger RNA (mRNA). The splicing that occurs can vary, so the synthesis of alternative protein products from the same primary transcript can be affected by tissue-specific or developmental signals. A significant fraction of human genetic diseases, including a number of cancers, are believed to result from deviations in the normal pattern of pre-mRNA splicing. The spliceosome is a complex comprising ribonucleoprotein (snRNP) particles composed of small nuclear RNAs and proteins. snRNA components of the spliceosome can promote the two transesterification reactions of splicing.

[0306] Two unique spliceosomes coexist in most eukaryotes: the U2-dependent spliceosome, which catalyzes the removal of U2-type introns, and the less abundant U12-dependent spliceosome, which is present in only a subset of eukaryotes and splices the rare U12-type class of introns. The U2-dependent spliceosome is assembled from the U1, U2, U5, and U4/U6 snRNPs and numerous non-snRNP proteins. The U2 snRNP is recruited with two weakly bound protein subunits, SF3a and SF3b, during the first ATP-dependent step in spliceosome assembly. SF3b is composed of seven conserved proteins, including PHF5D, SF3b155, SF3b145, SF3b130, SF3b49, SF3b14a, and SF3b10.

[0307] Splicing or RNA splicing typically refers to the editing of the nascent precursor messenger RNA (pre- mRNA) transcript into a mature messenger RNA (mRNA). Splicing is a biochemical process which includes the removal of introns followed by exon ligation. Sequential transesterification reactions are initiated by a nucleophilic attack of the 5’ splice site (5’ss) by the branch adenosine (branch point; BP) in the downstream intron resulting in the formation of an intron lariat intermediate with a 2’, 5’-phosphodiester linkage. This is followed by a 5’ss-mediated attack on the 3’ splice site (3’ss), leading to the removal of the intron lariat and the formation of the spliced RNA product.

[0308] Splicing can be regulated by various cis-acting elements and trans-acting factors. Cis-acting elements are sequences of the mRNA and can include core consensus sequences and other regulatory elements. Core consensus sequences typically can refer to conserved RNA sequence motifs, including the 5’ss, 3’ss, polypyrimidine tract and BP region, which can function for spliceosome recruitment. BP refers to a partially conserved sequence of pre-mRNA, generally less than 50 nucleotides upstream of the 3’ss. BP reacts with the 5’ss during the first step of the splicing reaction. Other regulatory cis-acting elements can include exonic splicing enhancer (ESE), exonic splicing silencer (ESS), intronic splicing enhancer (ISE), and intronic splicing silencer (ISS). Trans-acting factors can be proteins or ribonucleoproteins which bind to cis-acting elements.

[0309] Splice site identification and regulated splicing can be accomplished principally by two dynamic macromolecular machines, the major (U2-dependent) and minor (U12-dependent) spliceosomes. Each spliceosome contains five snRNPs: U1, U2, U4, U5 and U6 snRNPs for the major spliceosome (which processes ~95.5% of all introns); and U11, U12, U4atac, U5 and U 6 atac snRNPs for the minor spliceosome. Spliceosome recognition of consensus sequence elements at the 5’ss, 3’ss and BP sites is one of the steps in the splicing pathway, and can be modulated by ESEs, ISEs, ESSs, and ISSs, which can be recognized by auxiliary splicing factors, including SR proteins and hnRNPs. Polypyrimidine tract-binding protein (PTBP) can bind to the polypyrimidine tract of introns and may promote RNA looping.

[0310] Alternative splicing is a mechanism by which a single gene may eventually give rise to several different proteins. Alternative splicing can be accomplished by the concerted action of a variety of different proteins, termed“alternative splicing regulatory proteins,” that associate with the pre-mRNA, and cause distinct alternative exons to be included in the mature mRNA. These alternative forms of the gene’s transcript can give rise to distinct isoforms of the specified protein. Sequences in pre-mRNA molecules that can bind to alternative splicing regulatory proteins can be found in introns or exons, including, but not limited to, ISS, ISE, ESS, ESE, and polypyrimidine tract. Many mutations can alter splicing patterns. For example, mutations can be cis-acting elements, and can be located in core consensus sequences (e.g. 5’ss, 3’ss and BP) or the regulatory elements that modulate spliceosome recruitment, including ESE, ESS, ISE, and ISS.

[0311] A cryptic splice site, for example, a cryptic 5’ss and a cryptic 3’ss, can refer to a splice site that is not normally recognized by the spliceosome and therefore are in the dormant state. Cryptic splice site can be recognized or activated, for example, by mutations in cis-acting elements or trans-acting factors, or structural configurations, such as bulges.

Splicing Modulation

[0312] The present invention contemplates use of small molecules with favorable drug properties that modulate the activity of splicing of a target RNA. Provided herein are small molecule splicing modulators (SMSMs) that modulate splicing of a target polynucleotide. In some embodiments, the SMSMs bind and modulate target RNA. In some embodiments, provided herein is a library of SMSMs that bind and modulate one or more target RNAs. In some embodiments, the target RNA is mRNA. In some embodiments, the target RNA is mRNA a noncoding RNA. In some embodiments, the target RNA is a pre-mRNA. In some embodiments, the target RNA is hnRNA. In some embodiments, the small molecules modulate splicing of the target RNA. In some embodiments, a small molecule provided herein modulates splicing at a sequence of the target RNA. In some embodiments, a small molecule provided herein modulates splicing at a cryptic splice site sequence of the target RNA. In some embodiments, a small molecule provided herein binds to a target RNA. In some embodiments, a small molecule provided herein binds to a splicing complex component. In some embodiments, a small molecule provided herein binds to a target RNA and a splicing complex component.

[0313] Thus, provided herein are methods of preventing or inducing a splicing event in a pre-mRNA molecule, comprising contacting the pre-mRNA molecule and/or other elements of the splicing machinery (e.g., within a cell) with a compound provided herein to prevent or induce the splicing event in the pre-mRNA molecule. The splicing event that is prevented or induced can be, e.g., an aberrant splicing event, a constitutive splicing event or an alternate splicing event.

[0314] Further provided herein is a method of identifying a compound capable of preventing or inducing a splicing event in a pre-mRNA molecule, comprising contacting the compound with splicing elements and/or factors involved in alternative, aberrant and/or constitutive splicing as described herein (e.g., within cells) under conditions whereby a positive (prevention or induction of splicing) or negative (no prevention or induction of splicing) effect is produced and detected and identifying a compound that produces a positive effect as a compound capable of preventing or inducing a splicing event.

[0315] In some embodiments, a small molecule compound described herein in a pharmaceutically acceptable carrier prevents or induces an alternative or aberrant splicing event in a pre-mRNA molecule. As noted above, the small molecule compounds provided herein are not antisense or antigene oligonucleotides. Tables 1, 1B, 1D, and 1F show the chemical structure and name of exemplary compounds and is not intended to be all- inclusive.

[0316] In some embodiments, a composition comprises a small molecule splicing modulator compound (SMSM); wherein the SMSM interacts with an unpaired bulged nucleobase of an RNA duplex, and wherein the RNA duplex comprises a splice site. Provided herein is composition comprising a complex comprising a small molecule splicing modulator compound (SMSM) bound to an RNA duplex, wherein the SMSM interacts with an unpaired bulged nucleobase of an RNA duplex, and wherein the RNA duplex comprises a splice site. In some embodiments, the duplex RNA comprises an alpha helix. In some embodiments, the unpaired bulged nucleobase is located on an external portion of a helix of the duplex RNA. In some embodiments, the unpaired bulged nucleobase is located within an internal portion of the helix of the duplex RNA. In some embodiments, the SMSM forms one or more intermolecular interactions with the duplex RNA. In some embodiments, the SMSM forms one or more intermolecular interactions with the unpaired bulged nucleobase. In some embodiments, the intermolecular interaction is selected from the group comprising an ionic interaction, a hydrogen bond, a dipole-dipole interaction or a van der Waals interaction. In some embodiments, a first portion of the SMSM interacts with the unpaired bulged nucleobase on a first RNA strand of the RNA duplex. In some embodiments, a second portion of the SMSM interacts with one or more nucleobases of a second RNA strand of the RNA duplex, wherein the first RNA strand is not the second RNA strand. In some embodiments, a rate of exchange of the unpaired bulged nucleobase from within the interior of a helix of the duplex RNA to an exterior portion of the helix is reduced. In some embodiments, the SMSM reduces a rate of rotation of the unpaired bulged nucleobase. In some embodiments, the SMSM reduces a rate of rotation of the unpaired bulged nucleobase around a phosphate backbone of an RNA strand of the RNA duplex. In some embodiments, the SMSM modulates a distance of the unpaired bulged nucleobase from a second nucleobase of the duplex RNA. In some embodiments, the SMSM reduces the distance of the unpaired bulged nucleobase from a second nucleobase of the duplex RNA. In some embodiments, the unpaired bulged nucleobase is located within the interior of a helix of the duplex RNA of the complex. In some embodiments, the SMSM reduces a size of a bulge of the RNA duplex. In some embodiments, the SMSM removes a bulge of the RNA duplex. In some embodiments, the SMSM stabilizes a bulge of the RNA duplex. In some embodiments, the SMSM modulates splicing at the splice site of the RNA duplex. In some embodiments, the SMSM increases splicing at the splice site of the RNA duplex. In some embodiments, the SMSM reduces splicing at the splice site of the RNA duplex. In some embodiments, the unpaired bulged nucleobase has modulated base stacking within an RNA strand of the RNA duplex. In some embodiments, the unpaired bulged nucleobase has increased base stacking within an RNA strand of the RNA duplex. In some embodiments, the unpaired bulged nucleobase has decreased base stacking within an RNA strand of the RNA duplex. In some embodiments, the SMSM is not an aptamer. In some embodiments, the RNA duplex comprises pre-mRNA. In some embodiments, the unpaired bulged nucleobase is free to rotate around a phosphate backbone of an RNA strand of the RNA duplex in the absence of the SMSM.

[0317] In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to cells, wherein the SMSM kills the cells at an IC 50 of less than 50 nM. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to cells, wherein the SMSM modulates splicing at a splice site sequence of a pre-mRNA that encodes a mRNA, wherein the mRNA encodes a target protein or a functional RNA, and wherein a total amount of the mRNA is increased at least about 10% compared to the total amount of the mRNA encoding the target protein or functional RNA produced in control cells. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to cells, wherein the SMSM modulates splicing at a splice site sequence of a pre-mRNA that encodes a mRNA, wherein the mRNA encodes a target protein or a functional RNA, and wherein a total amount of the mRNA, the target protein and/or the functional RNA is at least 10% lower than the total amount of the mRNA, the target protein and/or the functional RNA in control cells.

[0318] In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to cells, wherein the SMSM modulates splicing at a splice site sequence of a pre-mRNA that encodes a first mRNA isoform associated with a disease or condition and a second mRNA isoform, wherein a total amount of the first mRNA isoform is decreased by at least about 10% compared to the total amount of the first mRNA isoform in control cells, and/or a total amount of the second mRNA isoform is increased by at least about 10% compared to the total amount of the first mRNA isoform in control cells. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to cells comprising an amount of a first mRNA isoform and an amount of a second mRNA isoform present in the cells; wherein a ratio of the first mRNA isoform to the second mRNA isoform is decreased at least 1.2 fold; wherein the first and second mRNAs are encoded by a pre-MRNA comprising a splice site sequence, and wherein the first mRNA isoform is associated with a disease or condition and a second mRNA isoform.

[0319] In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, wherein the SMSM modulates exon inclusion, exon exclusion, pseudoexon inclusion, intron retention, or splicing at a cryptic splice site of the polynucleotide, and wherein the SMSM modulates splicing of the splice site sequence. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, thereby modulating splicing of the polynucleotide, wherein the splice site sequence comprises a splice site sequence selected from the group consisting of splice site sequences of Table 2A, Table 2B, Table 2C or Table 2D. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, wherein the splice site sequence comprises a sequence selected from the group consisting of GGAguaag and AGAguaag. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, wherein the splice site sequence comprises at least one bulged nucleotide at the -3, -2, -1, +1, +2, +3, +4, +5 or +6 position of the splice site sequence. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, wherein the splice site sequence comprises a mutant nucleotide at the -3, -2, -1, +1, +2, +3, +4, +5 or +6 position of the splice site sequence.

[0320] In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, thereby modulating splicing of the polynucleotide, wherein the splice site sequence comprises a sequence selected from the group consisting of NGAgunvrn, NHAdddddn, NNBnnnnnn, and NHAddmhvk; wherein N or n is A, U, G or C; B is C, G, or U; H or h is A, C, or U; d is a, g, or u; m is a or c; r is a or g; v is a, c or g; k is g or u. In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, thereby modulating splicing of the polynucleotide, wherein the splice site sequence comprises a sequence selected from the group consisting of NNBgunnnn, NNBhunnnn, or NNBgvnnnn; wherein N or n is A, U, G or C; B is C, G, or U; H or h is A, C, or U; d is a, g, or u; m is a or c; r is a or g; v is a, c or g; k is g or u. In some embodiments, the splice site sequence comprises a sequence selected from the group consisting of NNBgurrrn, NNBguwwdn, NNBguvmvn, NNBguvbbn, NNBgukddn, NNBgubnbd, NNBhunngn,

NNBhurmhd, or NNBgvdnvn; wherein N or n is A, U, G or C; B is C, G, or U; H or h is A, C, or U; d is a, g, or u; m is a or c; r is a or g; v is a, c or g; k is g or u. In some embodiments, the nucleotide at the -3, -2, -1, +1, +2, +3, +4, +5 or +6 position of the splice site sequence is a bulged nucleotide. In some embodiments, the nucleotide at the -3, -2, -1, +1, +2, +3, +4, +5 or +6 position of the splice site sequence is mutated nucleotide. In some embodiments, the splice site sequence comprises a sequence selected from the group consisting of splice site sequences of Table 2A, Table 2B, Table 2C or Table 2D.

[0321] In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell comprising a polynucleotide with a splice site sequence, thereby modulating splicing of the polynucleotide, wherein the polynucleotide is encoded by a gene selected from the group consisting of genes of Table 2A, Table 2B, Table 2C or Table 2D. In some embodiments, the gene is SMN2. In some embodiments, modulating splicing of the polynucleotide comprises inhibiting skipping of exon 7. In some embodiments, the gene is DMD. In some embodiments, modulating splicing of the polynucleotide comprises promoting skipping of exon 51.

[0322] In some embodiments, a method of modulating splicing comprises contacting a small molecule splicing modulator compound (SMSM) to a cell; wherein the SMSM interacts with an unpaired bulged nucleobase of an RNA duplex in the cell; wherein the duplex RNA comprises a splice site sequence; and wherein the SMSM modulates splicing of the RNA duplex. In some embodiments, a method comprises modulating the relative position of a first nucleobase relative to a second nucleobase, wherein the first nucleobase and the second nucleobase are within a duplex RNA, the method comprising contacting a small molecule splicing modulator compound (SMSM) to the duplex RNA, or a pharmaceutically acceptable salt thereof, wherein the first nucleobase is an unpaired bulged nucleobase of the RNA duplex; wherein the duplex RNA comprises a splice site sequence.

[0323] In some embodiments, the duplex RNA comprises a helix. In some embodiments, the unpaired bulged nucleobase is located on an external portion of a helix of the duplex RNA prior to contacting the SMSM. In some embodiments, the SMSM forms one or more intermolecular interactions with the duplex RNA. In some embodiments, the SMSM forms one or more intermolecular interactions with the unpaired bulged nucleobase. In some embodiments, the intermolecular interaction is selected from the group comprising an ionic interaction, a hydrogen bond, a dipole-dipole interaction or a van der Waals interaction. In some

embodiments, a rate of exchange of the unpaired bulged nucleobase from within the interior of a helix of the duplex RNA to an exterior portion of the helix is reduced. In some embodiments, a rate of rotation of the unpaired bulged nucleobase is reduced. In some embodiments, a rate of rotation of the unpaired bulged nucleobase around a phosphate backbone of an RNA strand of the RNA duplex is reduced. In some embodiments, a distance of the unpaired bulged nucleobase from a second nucleobase of the duplex RNA is modulated after contacting the SMSM. In some embodiments, the distance of the unpaired bulged nucleobase from a second nucleobase of the duplex RNA is reduced. In some embodiments, the unpaired bulged nucleobase is located within the interior of the helix of the duplex RNA. In some embodiments, a size of a bulge of the RNA duplex is reduced. In some embodiments, a bulge of the RNA duplex is removed or maintained. In some embodiments, splicing at the splice site of the RNA duplex is promoted. In some embodiments, base stacking of the unpaired bulged nucleobase within an RNA strand of the RNA duplex is increased after contacting the SMSM. In some embodiments, the distance of the unpaired bulged nucleobase from a second nucleobase of the duplex RNA is increased or maintained. In some embodiments, a bulge of the RNA duplex is stabilized after contacting the SMSM. In some embodiments, the unpaired bulged nucleobase is located on an exterior portion of a helix of the duplex RNA. In some embodiments, a size of a bulge of the RNA duplex is increased. In some embodiments, splicing at the splice site of the RNA duplex is inhibited. In some embodiments, splicing is inhibited at the splice site In some embodiments, base stacking of the unpaired bulged nucleobase within an RNA strand of the RNA duplex is reduced after contacting the SMSM. In some embodiments, the RNA duplex comprises pre-mRNA.

[0324] In some embodiments, a method of treating a subject with a tumor comprises administering a small molecule splicing modulator compound (SMSM) to the subject, wherein a size of the tumor is reduced. In some embodiments, a method of treating a subject with a tumor comprises administering a small molecule splicing modulator compound (SMSM) to the subject, wherein tumor growth is inhibited by at least 20. In some embodiments, a method of the treatment, prevention and/or delay of progression of a condition or disease comprises administering a small molecule splicing modulator compound (SMSM) to a subject, wherein the SMSM modulates splicing of a splice site of a polynucleotide in a cell of the subject, wherein the condition or disease is associated with splicing of the splice site. In some embodiments, the subject has the disease or condition. In some embodiments, a method of treating a subject with a disease or condition comprises administering a small molecule splicing modulator compound (SMSM) to a subject with a disease or condition selected from the group consisting of diseases of Table 2A, Table 2B, Table 2C or Table 2D. In some embodiments, a method of treating a subject with a disease or condition comprises administering a small molecule splicing modulator compound (SMSM) to a subject with a disease or condition, wherein the SMSM is selected from the group consisting of the SMSMs of Table 1A, Table 1C, Table 1E, Table 1G, or Table 1H. In some embodiments, a method of treating a subject with a disease or condition comprises administering a small molecule splicing modulator compound (SMSM) to a subject with a disease or condition, wherein the SMSM binds to a pre-mRNA comprising a splice site sequence selected from the group consisting of splice site sequences of Table 2A, Table 2B, Table 2C or Table 2D. In some embodiments, the subject is a mammal. In some embodiments, the mammal is a human. In some embodiments, the polynucleotide is a pre- mRNA. In some embodiments, the disease or condition is spinal muscular atrophy. In some embodiments, the disease or condition is Duchenne’s muscular dystrophy. In some embodiments, the method further comprises administering an additional therapeutic molecule to the subject. In some embodiments, the SMSM is a compound described herein. In some embodiments, the SMSM is selected from the group consisting of SMSMs of Table 1A, Table 1C, Table 1E, Table 1G, or Table 1H.

[0325] In some embodiments, modulating splicing comprises preventing, inhibiting or reducing splicing at the splice site sequence of the polynucleotide. In some embodiments, modulating splicing comprises enhancing, promoting or increasing splicing at the splice site sequence of the polynucleotide. In some embodiments, the splice site sequence is a 5’ splice site sequence, a 3’ splice site sequence, a branch point splice site sequence or a cryptic splice site sequence. In some embodiments, the splice site comprises a mutation, the splice site comprises a bulge, the splice site comprises a mutation and a bulge, the splice site does not comprises a mutation, the splice site does not comprises a bulge, or the splice site does not comprises a mutation and does not comprise a bulge. In some embodiments, the bulge is a bulge caused by the mutation. In some embodiments, a bulged nucleotide is a mutant nucleotide. In some embodiments, a bulged nucleotide is not a mutant nucleotide. In some embodiments, the SMSM decreases a K D of splicing complex component to the polynucleotide. In some embodiments, the SMSM increases a K D of splicing complex component to the polynucleotide. In some embodiments, the SMSM inhibits binding of a splicing complex component to the polynucleotide at the splice site sequence, upstream of the splice site sequence or downstream of the splice site sequence. In some embodiments, the SMSM promotes binding of a splicing complex component to the polynucleotide at the splice site sequence, upstream of the splice site sequence or downstream of the splice site sequence. In some embodiments, the polynucleotide is RNA. In some embodiments, the RNA is a pre-mRNA. In some embodiments, the RNA is a heterogeneous nuclear RNA. In some embodiments, the splice site sequence is a 5’ splice site sequence, a 3’ splice site sequence, a branch point (BP) splice site sequence, an exonic splicing enhancer (ESE) sequence, an exonic splicing silencer (ESS) sequence, an intronic splicing enhancer (ISE) sequence, an intronic splicing silencer (ISS) sequence, a polypyrimidine tract sequence, or any combination thereof. In some embodiments, the polynucleotide is at least 5, 6, 7, 8, 9, 10, 15, 20, 25, 50, 100, 250, 500, 750, 1,000, 2,000, 5,000, 10,000, 50,000, 100,000, 500,000, or 1,000,000 nucleotides in length. In some embodiments, the SMSM binds to the splice site sequence of the polynucleotide. In some embodiments, the SMSM interacts with a bulge of the splice site sequence of the polynucleotide. In some embodiments, the polynucleotide comprises a cis-acting element sequence. In some embodiments, the cis-acting element sequence does not comprise a bulge. In some embodiments, the cis-acting element sequence does not comprise a mutation. In some embodiments, the cis-acting element sequence comprises a mutation, a bulge, or a combination thereof, at the cis-acting element sequence, 1-1000 nucleobases upstream of the cis-acting element sequence or 1-1000 nucleobases downstream of the cis-acting element sequence. In some embodiments, the cis-acting element sequence comprises a regulatory element sequence that modulates recruitment of a splicing complex component to the polynucleotide. In some embodiments, the cis-acting element sequence comprises a regulatory element sequence that modulates recruitment of a spliceosome to the polynucleotide. In some embodiments, the regulatory element sequence comprises an exonic splicing enhancer (ESE) sequence, an exonic splicing silencer (ESS) sequence, an intronic splicing enhancer (ISE) sequence, an intronic splicing silencer (ISS) sequence, and combinations thereof. In some embodiments, the SMSM binds to the splicing complex component. In some embodiments, the splicing complex component is 9G8, A1 hnRNP, A2 hnRNP, ASD-1, ASD-2b, ASF, B1 hnRNP, C1 hnRNP, C2 hnRNP, CBP20, CBP80, CELF, F hnRNP, FBP11, Fox-1, Fox-2, G hnRNP, H hnRNP, hnRNP 1, hnRNP 3, hnRNP C, hnRNP G, hnRNP K, hnRNP M, hnRNP U, Hu, HUR, I hnRNP, K hnRNP, KH-type splicing regulatory protein (KSRP), L hnRNP, M hnRNP, mBBP, muscle-blind like (MBNL), NF45, NFAR, Nova-1, Nova-2, nPTB, P54/SFRS11, polypyrimidine tract binding protein (PTB), PRP19 complex proteins, R hnRNP, RNPC1, SAM68, SC35, SF, SF1/BBP, SF2, SF3 a, SF3B, SFRS10, Sm proteins, SR proteins, SRm300, SRp20, SRp30c, SRP35C, SRP36, SRP38, SRp40, SRp55, SRp75, SRSF, STAR, GSG, SUP-12, TASR-1, TASR-2, TIA, TIAR, TRA2, TRA2a/b, U hnRNP, U1 snRNP, U11 snRNP, U12 snRNP, U1-C, U2 snRNP, U2AF1-RS2, U2AF35, U2AF65, U4 snRNP, U5 snRNP, U6 snRNP, Urp, YB1, or any combination thereof. In some embodiments, the splicing complex component comprises RNA. In some embodiments, the splicing complex component is a small nuclear RNA (snRNA). In some embodiments, the snRNA comprises U1 snRNA, U2 snRNA, U4 snRNA, U5 snRNA, U6 snRNA, U11 snRNA, U12 snRNA, U4atac snRNA, U5 snRNA, U6 atac snRNA, or any combination thereof. In some embodiments, the splicing complex component comprises a protein. In some embodiments, the splicing complex component comprises a small nuclear ribonucleoprotein (snRNP). In some embodiments, the snRNP comprises U1 snRNP, U2 snRNP, U4 snRNP, U5 snRNP, U6 snRNP, U11 snRNP, U12 snRNP, U4atac snRNP, U5 snRNP, U6 atac snRNP, or any combinations thereof. In some embodiments, the protein is a serine/arginine-rich (SR) protein. In some embodiments, the splice site sequence comprises a base that is mismatched to a base of a snRNA sequence. In some embodiments, a bulge is due to mismatched base pairing between the splice site sequence and a snRNA sequence.

[0326] In some embodiments, a method comprises upregulating expression of a native protein in a cell containing a DNA encoding the native protein, wherein the DNA contains a mutation or no mutation that causes downregulation of the native protein by aberrant and/or alternate splicing thereof. For example, the DNA can encode a pre-mRNA that has a mutation or an aberrant secondary or tertiary structure that causes downregulation of one or more isoforms of a protein. The method can comprise introducing into the cell a small molecule provided herein that prevents an aberrant splicing event, whereby the native intron is removed by correct splicing and the native protein is produced by the cell. In some embodiments, a method comprises introducing into a cell a small molecule provided herein that modulates an alternate splicing event to produce a protein that has a different function than the protein that would be produced without modulation of alternate splicing.

[0327] In some embodiments, a method comprises downregulating expression of a native protein in a cell containing a DNA encoding the native protein, wherein the DNA contains a mutation or no mutation that causes upregulation of the native protein by aberrant and/or alternate splicing thereof. For example, the DNA can encode a pre-mRNA that has a mutation or an aberrant secondary or tertiary structure that causes upregulation of one or more isoforms of a protein. The method can comprise introducing into the cell a small molecule provided herein that prevents an aberrant splicing event, whereby the native intron is removed by correct splicing and the native protein is produced by the cell. In some embodiments, a method comprises introducing into a cell a small molecule provided herein that modulates an alternate splicing event to produce a protein that has a different function than the protein that would be produced without modulation of alternate splicing. For example, a method can comprise preventing aberrant splicing in a pre-mRNA molecule containing a mutation or an aberrant secondary or tertiary structure and/or preventing an alternative splicing event. When present in the pre-mRNA, the mutation or aberrant secondary or tertiary structure can cause a pre-mRNA to splice incorrectly and produce an aberrant mRNA or mRNA fragment different from the mRNA ordinarily resulting from a pre-mRNA without the mutation or aberrant secondary or tertiary structure. For example, s pre-mRNA molecule can contain: (i) a first set of splice elements defining a native intron which can be removed by splicing when the mutation or aberrant secondary or tertiary structure is absent to produce a first mRNA molecule encoding a native protein, and (ii) a second set of splice elements induced by the mutation or aberrant secondary or tertiary structure which defines an aberrant intron different from the native intron, which aberrant intron is removed by splicing when the mutation or aberrant secondary or tertiary structure is present to produce an aberrant second mRNA molecule different from the first mRNA molecule. The method can comprise contacting the pre-mRNA molecule and/or other factors and/or elements of the splicing machinery as described herein (e.g., within a cell) with a compound described herein to prevent or promote an aberrant splicing event in a pre-mRNA molecule, whereby the native intron is removed by correct splicing and native protein production is increased in the cell.

[0328] In some embodiments, a method comprises upregulating expression of a RNA that would otherwise be downregulated by modulating an alternative splicing event in the RNA. The method can comprise contacting a pre-mRNA molecule and/or other elements and/or factors of the splicing machinery with a compound described herein to modulate alternate splicing events, whereby a native splicing event is inhibited and an alternate splicing event is promoted that upregulates expression of a RNA that is otherwise downregulated when under the control of the native splicing event.

[0329] In some embodiments, a method comprises downregulating expression of a RNA that would otherwise be upregulated by modulating an alternative splicing event in the RNA. The method can comprise contacting a pre-mRNA molecule and/or other elements and/or factors of the splicing machinery with a compound described herein to modulate alternate splicing events, whereby a native splicing event is inhibited and an alternate splicing event is promoted that downregulates expression of a RNA that is otherwise upregulated when under the control of the native splicing event.

[0330] The methods, compounds and compositions described herein have a variety of uses. For example, they are useful in any process where it is desired to have a means for downregulating expression of a RNA to be expressed until a certain time, after which it is desired to upregulate RNA expression. For such use, the RNA to be expressed may be any RNA encoding a protein to be produced so long as the gene contains a native intron. The RNA may be mutated by any suitable means, such as site-specific mutagenesis (see, T. Kunkel, U.S. Pat. No.4,873,192) to deliberately create an aberrant second set of splice elements which define an aberrant intron which substantially downregulates expression of the gene. A sequence encoding the RNA may be inserted into a suitable expression vector and the expression vector inserted into a host cell (e.g., a eukaryotic cell such as a yeast, insect, or mammalian cell (e.g., human, rat)) by standard recombinant techniques. The host cell can then be grown in culture by standard techniques. When it is desired to upregulate expression of the mutated gene, a suitable compound of the present invention, in a suitable formulation, can be added to the culture medium so that expression of the gene is upregulated.

[0331] Also provided herein is a method of altering the ratio of splice variants produced from a gene. The method can comprise contacting a pre-mRNA molecule and/or other elements and/or factors of the splicing machinery with a compound or compounds described herein to modulate alternative splicing events. The compound or compounds of this invention can be used to act upon 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 alternative splicing events that may occur within a pre-mRNA. In some embodiments, a first splice variant may be downregulated or inhibited and/or a second splice variant may be upregulated, resulting in an altered ratio of splice variants of the two or more RNA. In some embodiments, a first splice variant may be upregulated while a second splice variant may be unaffected, thereby altering the ratio of the RNA. In some embodiments, a first splice variant may be downregulated while a second splicing event may be unaffected thereby altering the ratio of the RNA.

[0332] The methods, compounds and formulations described herein are also useful as in vitro or in vivo tools to examine and modulate splicing events in human or animal RNAs encoded by genes, e.g., those developmentally and/or tissue regulated (e.g., alternate splicing events).

[0333] The compounds and formulations described herein are also useful as therapeutic agents in the treatment of disease involving aberrant and/or alternate splicing. Thus, in some embodiments, a method of treating a subject having a condition or disorder associated with an alternative or aberrant splicing event in a pre-mRNA molecule, comprises administering to the subject a therapeutically effective amount of a compound described herein to modulate an alternative splicing event or prevent an aberrant splicing event, thereby treating the subject. The method can, e.g., restore a correct splicing event in a pre-mRNA molecule. The method can, e.g., utilize a small molecule compound described herein in a pharmaceutically acceptable carrier.

[0334] Formulations containing the small molecules described herein can comprise a physiologically or pharmaceutically acceptable carrier, such as an aqueous carrier. Thus, formulations for use in the methods described herein include, but are not limited to, those suitable for oral administration, parenteral administration, including subcutaneous, intradermal, intramuscular, intravenous and intra-arterial administration, as well as topical administration (e.g., administration of an aerosolized formulation of respirable particles to the lungs of a patient afflicted with cystic fibrosis or lung cancer or a cream or lotion formulation for transdermal administration of patients with psoriasis). The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art. The most suitable route of administration in any given case may depend upon the subject, the nature and severity of the condition being treated, and the particular active compound, which is being used, as would be readily determined by one of skill in the art.

[0335] Also provided herein are methods for the use of a compound described herein having the characteristics set forth above for the preparation of a medicament for upregulating or downregulating RNA expression in a patient having a disorder associated with aberrant or alternate splicing of a pre-mRNA molecule, as discussed above. In some embodiments, the medicament upregulates gene expression. In other embodiments, the medicament downregulates gene expression. In the manufacture of at medicament according to the invention, the compound can be admixed with, inter alia, a pharmaceutically acceptable carrier. The carrier may be a solid or a liquid. One or more compounds may be incorporated in any combination in the formulations described herein, which may be prepared by any of the well-known techniques of pharmacy, such as admixing the components, and/or including one or more accessory therapeutic ingredients.

[0336] The present inventors identify herein low molecular weight compounds (sometimes referred to herein as small molecules, which block mRNA splicing and/or enhance (facilitate, augment) mRNA splicing. The splicing that can be regulated by the methods described herein include alternative splicing, e.g., exon skipping, intron retention, pseudoexons skipping, exon exclusion, partial intron exclusion and others. Depending on factors such as the splicing sequence and the RNA (or gene encoding the RNA) or exon involved, modulation of splicing can be accomplished in the presence of, or in the absence of, antisense oligonucleotides (AOs) that are specific for splicing sequences of interest. In some embodiments, a small molecule and an AO act synergistically.

[0337] In some aspects, a method comprises contacting a splice modulating compound (e.g., a SMSM) to a pre-mRNA that modulates splicing of the pre-mRNA to favor expression of a transcript that promotes cell proliferation. For example, an SMSM described herein can increase one or more isoforms of a transcript that promotes cell proliferation. For example, an SMSM described herein can decrease expression one or more isoforms of a transcript that prevents or inhibits cell proliferation.

[0338] In some aspects, a method comprises contacting a splice modulating compound (e.g., a SMSM) to a pre-mRNA that modulates splicing of the pre-mRNA to favor expression of a transcript that prevents or inhibits cell proliferation. For example, an SMSM described herein can increase one or more isoforms of a transcript that prevents or inhibits cell proliferation. For example, an SMSM described herein can decrease expression one or more isoforms of a transcript that promotes cell proliferation.

[0339] In some embodiments, a method of modulating splicing of pre-mRNA comprises using an SMSM to decrease expression or functionality of one or more isoforms of a transcript in a subject. The method can comprise administering an SMSM, or a composition comprising an SMSM, to a subject, wherein the SMSM binds to a pre-mRNA or a splicing complex component and modulates splicing of the pre-mRNA to favor expression of one or more isoforms of a transcript. The method can comprise administering an SMSM, or a composition comprising an SMSM, to a subject, wherein the SMSM binds to a pre-mRNA or a splicing complex component and modulates splicing of the pre-mRNA to disfavor expression of one or more isoforms of a transcript.

[0340] In some embodiments, the present invention provides a method of treating a subject afflicted with a disease or condition associated with aberrant splicing of a pre-mRNA. The method can comprise administering an SMSM, or a composition comprising an SMSM, to a subject, wherein the SMSM binds to a pre-mRNA or a splicing complex component and modulates splicing of the pre-mRNA to inhibit expression of one or more isoforms of a transcript. The method can comprise administering an SMSM, or a composition comprising an SMSM, to a subject, wherein the SMSM binds to a pre-mRNA or a splicing complex component and modulates the splicing of the pre-mRNA to increase expression of one or more isoforms of a transcript.

[0341] A number of diseases are associated with expression of an aberrant gene product (e.g., an RNA transcript or protein) of a gene. For example, aberrant amounts of a RNA transcript may lead to disease due to corresponding changes in protein expression. Changes in the amount of a particular RNA transcript may be the result of several factors. First, changes in the amount of RNA transcripts may be due to an aberrant level of transcription of a particular gene, such as by the perturbation of a transcription factor or a portion of the transcription process, resulting in a change in the expression level of a particular RNA transcript. Second, changes in the splicing of particular RNA transcripts, such as by perturbation of a particular splicing process or mutations in the gene that lead to modified splicing can change the levels of a particular RNA transcript. Changes to the stability of a particular RNA transcript or to components that maintain RNA transcript stability, such as the process of poly-A tail incorporation or an effect on certain factors or proteins that bind to and stabilize RNA transcripts, may lead to changes in the levels of a particular RNA transcript. The level of translation of particular RNA transcripts can also affect the amount of those transcripts, affecting or upregulating RNA transcript decay processes. Finally, aberrant RNA transport or RNA sequestration may also lead to changes in functional levels of RNA transcripts, and may have an effect on the stability, further processing, or translation of the RNA transcripts.

[0342] In some embodiments, provided herein are methods for modulating the amount of one, two, three or more RNA transcripts encoded by a pre-mRNA, comprising contacting a cell with an SMSM compound or a pharmaceutically acceptable salt thereof. In some embodiments, the cell is contacted with an SMSM compound or a pharmaceutically acceptable salt thereof in a cell culture. In other embodiments, the cell is contacted with an SMSM compound or a pharmaceutically acceptable salt thereof in a subject (e.g., a non-human animal subject or a human subject).

[0343] In some embodiments, provided herein are methods for treatment, prevention and/or delay of progression of a disease or condition comprising administering an effective amount of a small molecule splicing modulator as described herein to a subject, in particular to a mammal.

[0344] In some embodiments, provided herein are compositions and methods for treating a disease or condition, including steric modulator compounds or pharmaceutically acceptable salts thereof that promote prevention or correction of exon skipping of a pre-mRNA. The invention further provides compositions and methods for increasing production of mature mRNA and, in turn, protein, in cells of a subject in need thereof, for example, a subject that can benefit from increased production of protein. The invention further provides compositions and methods for decreasing production of mature mRNA and, in turn, protein, in cells of a subject in need thereof, for example, a subject that can benefit from decreased production of protein. In one embodiment, the described methods may be used to treat subjects having a disease or condition caused by a mutation in a gene, including missense, splicing, frameshift and nonsense mutations, as well as whole gene deletions, which result in deficient protein production. In another embodiment, the described methods may be used to treat subjects having a disease or condition not caused by gene mutation. In some embodiments, the compositions and methods of the present invention are used to treat subjects having a disease or condition, who can benefit from increased production of protein. In some embodiments, the compositions and methods of the present invention are used to treat subjects having a disease or condition, who can benefit from increased production of protein. In some embodiments, the compositions and methods of the present invention are used to treat subjects having a disease or condition, who can benefit from decreased production of a protein.

[0345] In some embodiments, provided herein are methods of treating a disease or condition in a subject in need thereof by increasing the expression of a target protein or functional RNA by cells of the subject, wherein the cells have a mutation that causes, e.g., exon skipping or intron inclusion, or a portion thereof, of pre-mRNA, wherein the pre-mRNA encodes the target protein or functional RNA. The method can comprise contacting cells of a subject with an SMSM compound or a pharmaceutically acceptable salt thereof that targets the pre- mRNA encoding the target protein or functional RNA or splicing complex component, whereby splicing of an exon from a pre-mRNA encoding a target protein or functional RNA is prevented or inhibited, thereby increasing a level of mRNA encoding the target protein or functional RNA, and increasing the expression of the target protein or functional RNA in the cells of the subject. In some embodiments, also disclosed herein is a method of increasing expression of a target protein by cells having a mutation or aberrant secondary or tertiary RNA structure that causes exon skipping of pre-mRNA, the pre-mRNA comprising a mutation or aberrant secondary or tertiary RNA structure that causes exon skipping. The method can comprise contacting the cells with an SMSM compound or a pharmaceutically acceptable salt thereof that targets a pre-mRNA encoding a target protein or functional RNA, whereby splicing of an exon from a pre-mRNA encoding a target protein or functional RNA is prevented or inhibited, thereby increasing the level of mRNA encoding functional protein, and increasing the expression of protein in the cells. In some embodiments, the target protein is a tumor suppressor. In some embodiments, the target protein is a tumor promoter. In some embodiments, the target protein or the functional RNA is a compensating protein or a compensating functional RNA that functionally augments or replaces a target protein or functional RNA that is deficient in amount or activity in the subject. In some embodiments, the cells are in or from a subject having a condition caused by a deficient amount or activity of the protein. In some embodiments, the deficient amount of the target protein is caused by haploinsufficiency of the target protein, wherein the subject has a first allele encoding a functional target protein, and a second allele from which the target protein is not produced, or a second allele encoding a nonfunctional target protein, and wherein an SMSM compound or a pharmaceutically acceptable salt thereof binds to a targeted portion of a pre-mRNA transcribed from the first allele. In some embodiments, the target protein is produced in a form that is fully-functional compared to the equivalent protein produced from mRNA in which an exon has been skipped or is missing. In some embodiments, the pre-mRNA is encoded by a genetic sequence with at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to a pre-mRNA. In some embodiments, an SMSM compound or a pharmaceutically acceptable salt thereof increases the amount of the target protein or the functional RNA by modulating alternative splicing of pre-mRNA transcribed from a gene encoding the functional RNA or target protein. In some embodiments, an SMSM compound or a pharmaceutically acceptable salt thereof increases the amount of the target protein or the functional RNA by modulating aberrant splicing resulting from mutation of the gene encoding the target protein or the functional RNA.

[0346] In some embodiments, the total amount of the mRNA encoding the target protein or functional RNA produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased at least about 10%, at least about 20%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, at least about 300%,at least about 400%, or at least about 500%, compared to the total amount of the mRNA encoding the target protein or functional RNA produced in a control cell.

[0347] In some embodiments, the total amount of the mRNA encoding the target protein or functional RNA produced in the cell contacted with than SMSM compound or a pharmaceutically acceptable salt thereof is increased about 20% to about 300%, about 50% to about 300%, about 100% to about 300%, about 150% to about 300%, about 20% to about 50%, about 20% to about 100%, about 20% to about 150%, about 20% to about 200%, about 20% to about 250%, about 50% to about 100%, about 50% to about 150%, about 50% to about 200%, about 50% to about 250%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 150% to about 200%, about 150% to about 250%, or about 200% to about 250%, compared to the total amount of the mRNA encoding the target protein or functional RNA produced in a control cell.

[0348] In some embodiments, the total amount of target protein produced by the cell contacted with an SMSMS compound or a pharmaceutically acceptable salt thereof is increased at least about 20%, at least about 50%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, or at least about 300%, compared to the total amount of target protein produced by a control cell. In some embodiments, the total amount of target protein produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 20% to about 300%, about 50% to about 300%, about 100% to about 300%, about 150% to about 300%, about 20% to about 50%, about 20% to about 100%, about 20% to about 150%, about 20% to about 200%, about 20% to about 250%, about 50% to about 100%, about 50% to about 150%, about 50% to about 200%, about 50% to about 250%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 150% to about 200%, about 150% to about 250%, or about 200% to about 250%, compared to the total amount of target protein produced by a control cell.

[0349] In some embodiments, a total amount of the mRNA encoding the target protein or functional RNA produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased at least about 1.1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 5-fold, or at least about 10-fold compared to the total amount of the mRNA encoding the target protein or functional RNA produced in a control cell. In some embodiments, a total amount of an mRNA encoding the target protein or functional RNA produced in a cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 1.1 to about 10-fold, about 1.5 to about 10-fold, about 2 to about 10-fold, about 3 to about 10-fold, about 4 to about 10-fold, about 1.1 to about 5-fold, about 1.1 to about 6-fold, about 1.1 to about 7-fold, about 1.1 to about 8-fold, about 1.1 to about 9-fold, about 2 to about 5-fold, about 2 to about 6-fold, about 2 to about 7-fold, about 2 to about 8-fold, about 2 to about 9-fold, about 3 to about 6-fold, about 3 to about 7-fold, about 3 to about 8-fold, about 3 to about 9-fold, about 4 to about 7-fold, about 4 to about 8-fold, or about 4 to about 9-fold, compared to a total amount of the mRNA encoding the target protein or functional RNA produced in a control cell.

[0350] In some embodiments, a total amount of target protein produced by a cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased at least about 1.1-fold, at least about 1.5- fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4- fold, at least about 5-fold, or at least about 10-fold, compared to the total amount of target protein produced by a control cell. In some embodiments, the total amount of target protein produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 1.1 to about 10-fold, about 1.5 to about 10-fold, about 2 to about 10-fold, about 3 to about 10-fold, about 4 to about 10-fold, about 1.1 to about 5-fold, about 1.1 to about 6-fold, about 1.1 to about 7-fold, about 1.1 to about 8-fold, about 1.1 to about 9-fold, about 2 to about 5-fold, about 2 to about 6-fold, about 2 to about 7-fold, about 2 to about 8-fold, about 2 to about 9-fold, about 3 to about 6-fold, about 3 to about 7-fold, about 3 to about 8-fold, about 3 to about 9- fold, about 4 to about 7-fold, about 4 to about 8-fold, or about 4 to about 9-fold, compared to a total amount of target protein produced by a control cell.

[0351] In some embodiments, the total amount of the mRNA encoding the target protein or functional RNA produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 100%, compared to the total amount of the mRNA encoding the target protein or functional RNA produced in a control cell.

[0352] In some embodiments, the total amount of the mRNA encoding the target protein or functional RNA produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100% about 90% to about 100%, about 20% to about 30%, about 20% to about 40%, about 20% to about 50%, about 20% to about 60%, about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 30% to about 40%, about 30% to about 50%, about 30% to about 60%, about 30% to about 70%, about 30% to about 80%, about 30% to about 90%, about 40% to about 50%, about 40% to about 60%, about 40% to about 70%, about 40% to about 80%, about 40% to about 90%, about 50% to about 60%, about 50% to about 70%, about 50% to about 80%, about 50% to about 90%, about 60% to about 70%, about 60% to about 80%, about 60% to about 90%, 70% to about 80%, about 70% to about 90%, or about 80% to about 90%, compared to the total amount of the mRNA encoding the target protein or functional RNA produced in a control cell.

[0353] In some embodiments, the total amount of target protein produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 100%, compared to the total amount of target protein produced by a control cell. In some embodiments, the total amount of target protein produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100% about 90% to about 100%, about 20% to about 30%, about 20% to about 40%, about 20% to about 50%, about 20% to about 60%, about 20% to about 70%, about 20% to about 80%, about 20% to about 90%, about 30% to about 40%, about 30% to about 50%, about 30% to about 60%, about 30% to about 70%, about 30% to about 80%, about 30% to about 90%, about 40% to about 50%, about 40% to about 60%, about 40% to about 70%, about 40% to about 80%, about 40% to about 90%, about 50% to about 60%, about 50% to about 70%, about 50% to about 80%, about 50% to about 90%, about 60% to about 70%, about 60% to about 80%, about 60% to about 90%, 70% to about 80%, about 70% to about 90%, or about 80% to about 90%, compared to the total amount of target protein produced by a control cell.

[0354] In some embodiments, the difference in amount between a first splice variant and a second splice variant encoding a target protein or functional RNA isoform produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 20% to about 300%, about 50% to about 300%, about 100% to about 300%, about 150% to about 300%, about 20% to about 50%, about 20% to about 100%, about 20% to about 150%, about 20% to about 200%, about 20% to about 250%, about 50% to about 100%, about 50% to about 150%, about 50% to about 200%, about 50% to about 250%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 150% to about 200%, about 150% to about 250%, about 200% to about 250%, at least about 20%, at least about 50%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, or at least about 300%, compared to the difference in amounts between the two splice variants produced by a control cell. In some embodiments, the difference in amount between a first protein isoform expressed from a first splice variant and a second protein isoform expressed from a second splice variant produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 20% to about 300%, about 50% to about 300%, about 100% to about 300%, about 150% to about 300%, about 20% to about 50%, about 20% to about 100%, about 20% to about 150%, about 20% to about 200%, about 20% to about 250%, about 50% to about 100%, about 50% to about 150%, about 50% to about 200%, about 50% to about 250%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 150% to about 200%, about 150% to about 250%, about 200% to about 250%, at least about 20%, at least about 50%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, or at least about 300%, compared to the difference in amounts between two protein isoforms produced from the splice variants produced by a control cell.

[0355] In some embodiments, the difference in amount between a first splice variant and a second splice variant encoding a target protein or functional RNA isoform produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 1.1 to about 10-fold, about 1.5 to about 10-fold, about 2 to about 10-fold, about 3 to about 10-fold, about 4 to about 10-fold, about 1.1 to about 5-fold, about 1.1 to about 6-fold, about 1.1 to about 7-fold, about 1.1 to about 8-fold, about 1.1 to about 9-fold, about 2 to about 5-fold, about 2 to about 6-fold, about 2 to about 7-fold, about 2 to about 8-fold, about 2 to about 9-fold, about 3 to about 6-fold, about 3 to about 7-fold, about 3 to about 8-fold, about 3 to about 9-fold, about 4 to about 7- fold, about 4 to about 8-fold, about 4 to about 9-fold, at least about 1.1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 5-fold, or at least about 10-fold, compared to the difference in amounts between the two splice variants produced by a control cell. In some embodiments, the difference in amount between a first protein isoform expressed from a first splice variant and a second protein isoform expressed from a second splice variant produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is increased about 1.1 to about 10-fold, about 1.5 to about 10-fold, about 2 to about 10-fold, about 3 to about 10- fold, about 4 to about 10-fold, about 1.1 to about 5-fold, about 1.1 to about 6-fold, about 1.1 to about 7-fold, about 1.1 to about 8-fold, about 1.1 to about 9-fold, about 2 to about 5-fold, about 2 to about 6-fold, about 2 to about 7-fold, about 2 to about 8-fold, about 2 to about 9-fold, about 3 to about 6-fold, about 3 to about 7-fold, about 3 to about 8-fold, about 3 to about 9-fold, about 4 to about 7-fold, about 4 to about 8-fold, about 4 to about 9-fold, at least about 1.1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 5-fold, or at least about 10-fold, compared to the difference in amounts between two protein isoforms expressed from the splice variants produced by a control cell.

[0356] In some embodiments, a difference in amount between a first splice variant and a second splice variant encoding a target protein or functional RNA isoform produced in a cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased about 20% to about 300%, about 50% to about 300%, about 100% to about 300%, about 150% to about 300%, about 20% to about 50%, about 20% to about 100%, about 20% to about 150%, about 20% to about 200%, about 20% to about 250%, about 50% to about 100%, about 50% to about 150%, about 50% to about 200%, about 50% to about 250%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 150% to about 200%, about 150% to about 250%, about 200% to about 250%, at least about 20%, at least about 50%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, or at least about 300%, compared to the difference in amounts between the two splice variants produced by a control cell. In some embodiments, a difference in amount between a first protein isoform expressed from a first splice variant and a second protein isoform expressed from a second splice variant produced by a cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased about 20% to about 300%, about 50% to about 300%, about 100% to about 300%, about 150% to about 300%, about 20% to about 50%, about 20% to about 100%, about 20% to about 150%, about 20% to about 200%, about 20% to about 250%, about 50% to about 100%, about 50% to about 150%, about 50% to about 200%, about 50% to about 250%, about 100% to about 150%, about 100% to about 200%, about 100% to about 250%, about 150% to about 200%, about 150% to about 250%, about 200% to about 250%, at least about 20%, at least about 50%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, or at least about 300%, compared to a difference in amounts between two protein isoforms produced from the splice variants produced by a control cell.

[0357] In some embodiments, the difference in amount between a first splice variant and a second splice variant encoding a target protein or functional RNA isoform produced in the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased about 1.1 to about 10-fold, about 1.5 to about 10-fold, about 2 to about 10-fold, about 3 to about 10-fold, about 4 to about 10-fold, about 1.1 to about 5-fold, about 1.1 to about 6-fold, about 1.1 to about 7-fold, about 1.1 to about 8-fold, about 1.1 to about 9-fold, about 2 to about 5-fold, about 2 to about 6-fold, about 2 to about 7-fold, about 2 to about 8-fold, about 2 to about 9-fold, about 3 to about 6-fold, about 3 to about 7-fold, about 3 to about 8-fold, about 3 to about 9-fold, about 4 to about 7- fold, about 4 to about 8-fold, about 4 to about 9-fold, at least about 1.1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 5-fold, or at least about 10-fold, compared to the difference in amounts between the two splice variants produced by a control cell. In some embodiments, the difference in amount between a first protein isoform expressed from a first splice variant and a second protein isoform expressed from a second splice variant produced by the cell contacted with an SMSM compound or a pharmaceutically acceptable salt thereof is decreased about 1.1 to about 10-fold, about 1.5 to about 10-fold, about 2 to about 10-fold, about 3 to about 10- fold, about 4 to about 10-fold, about 1.1 to about 5-fold, about 1.1 to about 6-fold, about 1.1 to about 7-fold, about 1.1 to about 8-fold, about 1.1 to about 9-fold, about 2 to about 5-fold, about 2 to about 6-fold, about 2 to about 7-fold, about 2 to about 8-fold, about 2 to about 9-fold, about 3 to about 6-fold, about 3 to about 7-fold, about 3 to about 8-fold, about 3 to about 9-fold, about 4 to about 7-fold, about 4 to about 8-fold, about 4 to about 9-fold, at least about 1.1-fold, at least about 1.5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 5-fold, or at least about 10-fold, compared to the difference in amounts between two protein isoforms express from the splice variants produced by a control cell.

[0358] The ratio of a first isoform and a second isoform may contribute to a number of conditions or diseases. In some embodiments, a subject without a condition or disease has a first isoform to second isoform ratio of 1:1. In some embodiments, a subject with a condition or disease described herein has a first isoform to second isoform ratio of about 1:1.2, 1:1.4, 1:1.6, 1:1.8, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5. In some embodiments, a subject with a condition or disease described herein has a first isoform to second isoform ratio from about 1:1 to about 1:1.1, about 1:1 to about 1:1.2, about 1:1 to about 1:1.3, about 1:1 to about 1:1.4, about 1:1 to about 1:1.5, about 1:1 to about 1:1.6, about 1:1 to about 1:1.8, about 1:1 to about 1:2, about 1:1 to about 1:3, about 1:1 to about 1:3.5, about 1:1 to about 1:4, about 1:1 to about 1:4.5, about 1:1 to about 1:5, 1:2 to about 1:3, about 1:2 to about 1:4, about 1:2 to about 1:5, about 1:3 to about 1:4, about 1:3 to about 1:5, or about 1:4 to about 1:5.

[0359] In some embodiments, binding of an SMSM compound or a pharmaceutically acceptable salt thereof to pre-mRNA prevents splicing out of one or more exons and/or introns and/or proteins thereof, from the population of pre-mRNAs to produce mRNA encoding the target protein or functional RNA. In some embodiments, the cell comprises a population of pre-mRNAs transcribed from the gene encoding the target protein or functional RNA, wherein the population of pre-mRNAs comprises a mutation that causes the splicing out of one or more exons, and wherein an SMSM compound or a pharmaceutically acceptable salt thereof binds to the mutation that causes the splicing out of the one or more exons in the population of pre-mRNAs. In some embodiments, the binding of an SMSM compound or a pharmaceutically acceptable salt thereof to the mutation that causes the splicing out of the one or more exons prevents splicing out of the one or more exons from the population of pre-mRNAs to produce mRNA encoding the target protein or functional RNA. In some embodiments, the condition is a disease or disorder. In some embodiments, the method further comprises assessing protein expression. In some embodiments, an SMSM compound or a pharmaceutically acceptable salt thereof binds to a targeted portion of a pre-mRNA.

[0360] In some embodiments, the binding of an SMSM compound or a pharmaceutically acceptable salt thereof catalyzes the inclusion of a missing exon or removal of an undesired retained intron or portions thereof, resulting in healthy mRNA and proteins. In some embodiments, the binding of an SMSM compound or a pharmaceutically acceptable salt thereof has minimal to no effect on non-diseased cells.

[0361] In some embodiments, an SMSM kills cells at an IC 50 of less than 50 nM. In some embodiments, the cells are primary cells. In some embodiments, an SMSM kills the cells at an IC 50 of less than 48 nM, 45 nM, 40 nM, 35 nM, 30 nM, 25 nM, 20 nM, 15 nM, 10 nM, 5 nM, 3 nM, or 1 nM.

[0362] In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide of the primary cells. In some embodiments, an SMSM modulates proliferation or survival of the primary cells. In some embodiments, the primary cells are primary diseased cells. In some embodiments, the primary diseased cells are primary cancer cells. In some embodiments, the SMSM is present at a concentration of at least about 1 nM, 10 nM, 100 nM, 1 mM, 10 mM, 100 mM, 1 mM, 10 mM, 100 mM, or 1 M. In some embodiments, at least about 5%, 10%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% of the primary diseased cells are killed. In some embodiments, at least about 5%, 10%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% of the primary diseased cells undergo apoptosis. In some embodiments, at least about 5%, 10%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% of the primary diseased cells undergo necrosis. In some embodiments, proliferation is reduced or inhibited in at least about 5%, 10%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% of the primary diseased cells. In some embodiments, the primary diseased cells are non-transformed cells.

[0363] In some embodiments, an SMSM reduces a size of a tumor in a subject. In some embodiments, a size of a tumor in a subject administered an SMSM or a pharmaceutically acceptable salt thereof is reduced by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% in the subject. In some embodiments, a diameter of a tumor in a subject administered an SMSM or a pharmaceutically acceptable salt thereof is reduced by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%. In some embodiments, a volume of the tumor is reduced by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% in the subject. In some embodiments, the tumor is malignant.

[0364] In some embodiments, a method comprises contacting an SMSM to primary non-diseased cells. In some embodiments, at most about 1%, 5%, 10%, 15%, 20%, 25%, or 50% of the primary non-diseased cells are killed. In some embodiments, at most about 1%, 5%, 10%, 15%, 20%, 25%, or 50% of the primary non-diseased cells undergo apoptosis. In some embodiments, at most about 1%, 5%, 10%, 15%, 20%, 25%, or 50% of the primary non-diseased cells undergo necrosis. In some embodiments, proliferation is reduced or inhibited in at most about 1%, 5%, 10%, 15%, 20%, 25%, or 50% of the primary non-diseased cells. In some embodiments, the primary non-diseased cells are of the same tissue as the primary diseased cells. In some embodiments, the primary non- diseased cells are differentiated cells. [0365] An SMSM can modulate splicing at a splice site of a polynucleotide and does not exhibit significant toxicity. In some embodiments, an SMSM penetrates the blood brain barrier (BBB) when administered to a subject.

[0366] In some embodiments, an SMSM has a brain/blood AUC of at least about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.5, 3.0, 3.5, 40, or higher.

[0367] In some embodiments, an SMSM provided herein, e.g., an SMSM of Formula (I) or (I*), has an apparent permeability (Papp) of at least about 1, at least about 2, at least about 3, at least about 4, at least about 5, at least about 10, at least about 15, at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, or at least about 100, as determined by MDCK-MDR1 Permeability assay. In some embodiments, an SMSM provided herein has an apparent permeability of at least about 10, at least about 20, or at least about 50. In some embodiments, an SMSM provided herein, e.g., an SMSM of Formula (I) or (I*), has an Efflux Ratio (ER) of at most about 3. In some embodiments, an SMSM provided herein has an Efflux ratio within a range of from about 1, about 2, about 3 or about 4, to about 5, about 6, about 7, about 8, about 9, about 10, about 12 about 15, or about 20, as determined by MDCK-MDR1 Permeability assay. In some embodiments, an SMSM provided herein has an Efflux ratio of from about 3 to about 10. In some embodiments, an SMSM provided herein has an Efflux ratio that is at most about 3, at most about 2, or at most about 1. In some embodiments, an SMSM provided herein has an Efflux ratio of larger than about 10. In some embodiments, an SMSM provided herein has an Efflux ratio of at least about 10, at least about 20, at least about 50, at least about 100, at least about 200, or at least about 300.

[0368] In some embodiments, an SMSM has a half-life of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 hours in a human.

[0369] In some embodiments, an SMSM is stable at room temperature for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours; or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months; or at least 1, 2, 3, 4, or 5 years. In some embodiments, an SMSM is stable at 4 °C for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours; or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months; or at least 1, 2, 3, 4, or 5 years. In some embodiments, an SMSM is stable at room temperature in water or an organic solvent for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours; or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months; or at least 1, 2, 3, 4, or 5 years. In some embodiments, an SMSM is stable at 4 °C in water or an organic solvent for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours; or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months; or at least 1, 2, 3, 4, or 5 years.

[0370] In some embodiments, an SMSM has an cell viability IC 50 of 0.01-10 nM, 0.01-5 nM, 0.01-2.5 nM, 0.01-1 nM, 0.01-0.75 nM, 0.01-0.5 nM, 0.01-0.25 nM, 0.01-0.1 nM, 0.1-100 nM, 0.1-50 nM, 0.1-25 nM, 0.1- 10 nM, 0.1-7.5 nM, 0.1-5 nM, 0.1-2.5 nM, 2-1000 nM, 2-500 nM, 2-250 nM, 2-100 nM, 2-75 nM, 2-50 nM, 2- 25 nM, 2-10 nM, 10-1000 nM, 10-500 nM, 10-250 nM, 10-100 nM, 10-75 nM, 10-50 nM, 10-25 nM, 25-1000 nM, 25-500 nM, 25-250 nM, 25-100 nM, 25-75 nM, 25-50 nM, 50-1000 nM, 50-500 nM, 50-250 nM, 50-100 nM, 50-75 nM, 60-70 nM, 100-1000 nM, 100-500 nM, 100-250 nM, 250-1000 nM, 250-500 nM, or 500-1000 nM.

[0371] In some embodiments, an SMSM has an cell viability IC 50 of at most 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 30 nM, 35 nM, 40 nM, 45 nM, 50 nM, 51 nM, 52 nM, 53 nM, 54 nM, 55 nM, 56 nM, 57 nM, 58 nM, 59 nM, 60 nM, 61 nM, 62 nM, 63 nM, 64 nM, 65 nM, 66 nM, 67 nM, 68 nM, 69 nM, 70 nM, 71 nM, 72 nM, 73 nM, 74 nM, 75 nM, 76 nM, 77 nM, 78 nM, 79 nM, 80 nM, 81 nM, 82 nM, 83 nM, 84 nM, 85 nM, 90 nM, 95 nM, 100 nM, 110 nM, 120 nM, 130 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 210 nM, 220 nM, 230 nM, 240 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, 950 nM, 1 mM, or 10 mM.

[0372] In some embodiments, an SMSM reduces cell proliferation of diseased cells by more than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% when the cells are treated with the SMSM at a concentration of 2-1000 nM, 2-500 nM, 2-250 nM, 2-100 nM, 2-75 nM, 2-50 nM, 2-25 nM, 2-10 nM, 10-1000 nM, 10-500 nM, 10-250 nM, 10-100 nM, 10-75 nM, 10- 50 nM, 10-25 nM, 25-1000 nM, 25-500 nM, 25-250 nM, 25-100 nM, 25-75 nM, 25-50 nM, 50-1000 nM, 50- 500 nM, 50-250 nM, 50-100 nM, 50-75 nM, 60-70 nM, 100-1000 nM, 100-500 nM, 100-250 nM, 250-1000 nM, 250-500 nM, or 500-1000 nM for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 21, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, or 48 hours.

[0373] In some embodiments, an SMSM reduces cell proliferation of diseased cells by more than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% when the cells are treated with the SMSM at a concentration of at least 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 30 nM, 35 nM, 40 nM, 45 nM, 50 nM, 51 nM, 52 nM, 53 nM, 54 nM, 55 nM, 56 nM, 57 nM, 58 nM, 59 nM, 60 nM, 61 nM, 62 nM, 63 nM, 64 nM, 65 nM, 66 nM, 67 nM, 68 nM, 69 nM, 70 nM, 71 nM, 72 nM, 73 nM, 74 nM, 75 nM, 76 nM, 77 nM, 78 nM, 79 nM, 80 nM, 81 nM, 82 nM, 83 nM, 84 nM, 85 nM, 90 nM, 95 nM, 100 nM, 110 nM, 120 nM, 130 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 210 nM, 220 nM, 230 nM, 240 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, 950 nM, 1 mM, or 10 mM for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 21, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, or 48 hours.

[0374] In some embodiments, an SMSM reduces viability of diseased cells by more than 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. when the cells are treated with the SMSM at a concentration of 2-1000 nM, 2-500 nM, 2-250 nM, 2-100 nM, 2-75 nM, 2-50 nM, 2-25 nM, 2-10 nM, 10- 1000 nM, 10-500 nM, 10-250 nM, 10-100 nM, 10-75 nM, 10-50 nM, 10-25 nM, 25-1000 nM, 25-500 nM, 25- 250 nM, 25-100 nM, 25-75 nM, 25-50 nM, 50-1000 nM, 50-500 nM, 50-250 nM, 50-100 nM, 50-75 nM, 60- 70 nM, 100-1000 nM, 100-500 nM, 100-250 nM, 250-1000 nM, 250-500 nM, or 500-1000 nM for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 21, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, or 48 hours.

[0375] In some embodiments, an SMSM reduces viability of diseased cells by more than 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% when the cells are treated with the SMSM at a concentration of at least 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 30 nM, 35 nM, 40 nM, 45 nM, 50 nM, 51 nM, 52 nM, 53 nM, 54 nM, 55 nM, 56 nM, 57 nM, 58 nM, 59 nM, 60 nM, 61 nM, 62 nM, 63 nM, 64 nM, 65 nM, 66 nM, 67 nM, 68 nM, 69 nM, 70 nM, 71 nM, 72 nM, 73 nM, 74 nM, 75 nM, 76 nM, 77 nM, 78 nM, 79 nM, 80 nM, 81 nM, 82 nM, 83 nM, 84 nM, 85 nM, 90 nM, 95 nM, 100 nM, 110 nM, 120 nM, 130 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 210 nM, 220 nM, 230 nM, 240 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, 950 nM, 1 mM, or 10 mM for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 21, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, or 48 hours.

[0376] In some embodiments, an SMSM does not reduce viability of non-diseased cells by more than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, or 50 when the cells are treated with the SMSM at a concentration of 2-1000 nM, 2-500 nM, 2-250 nM, 2-100 nM, 2-75 nM, 2-50 nM, 2-25 nM, 2-10 nM, 10-1000 nM, 10-500 nM, 10-250 nM, 10-100 nM, 10-75 nM, 10-50 nM, 10-25 nM, 25-1000 nM, 25-500 nM, 25-250 nM, 25-100 nM, 25-75 nM, 25-50 nM, 50-1000 nM, 50-500 nM, 50-250 nM, 50-100 nM, 50-75 nM, 60-70 nM, 100-1000 nM, 100-500 nM, 100-250 nM, 250-1000 nM, 250-500 nM, or 500-1000 nM for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 21, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, or 48 hours.

[0377] In some embodiments, an SMSM does not reduce viability of non-diseased cells by more than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, or 50% when the cells are treated with the SMSM at a concentration of at least 2 nM, 3 nM, 4 nM, 5 nM, 6 nM, 7 nM, 8 nM, 9 nM, 10 nM, 11 nM, 12 nM, 13 nM, 14 nM, 15 nM, 16 nM, 17 nM, 18 nM, 19 nM, 20 nM, 21 nM, 22 nM, 23 nM, 24 nM, 25 nM, 30 nM, 35 nM, 40 nM, 45 nM, 50 nM, 51 nM, 52 nM, 53 nM, 54 nM, 55 nM, 56 nM, 57 nM, 58 nM, 59 nM, 60 nM, 61 nM, 62 nM, 63 nM, 64 nM, 65 nM, 66 nM, 67 nM, 68 nM, 69 nM, 70 nM, 71 nM, 72 nM, 73 nM, 74 nM, 75 nM, 76 nM, 77 nM, 78 nM, 79 nM, 80 nM, 81 nM, 82 nM, 83 nM, 84 nM, 85 nM, 90 nM, 95 nM, 100 nM, 110 nM, 120 nM, 130 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 210 nM, 220 nM, 230 nM, 240 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, 950 nM, 1 mM, or 10 mM for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 21, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, or 48 hours.

[0378] In some embodiments, an SMSM reduces a size of a tumor in a subject by at least 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.

[0379] In some embodiments, an SMSM inhibits tumor growth of a tumor in a subject by at least 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 30%, 35%, 40%, 45%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.

SMSM Targets

[0380] Aberrant splicing of mRNA, such as pre-mRNA, can result in a defective protein and can cause a disease or a disorder in a subject. The compositions and methods described herein can reduce this aberrant splicing of mRNA, such as pre-mRNA, and treat a disease or a disorder caused by this aberrant splicing.

[0381] Diseases associated with changes to RNA transcript amount are often treated with a focus on the aberrant protein expression. However, if the processes responsible for the aberrant changes in RNA levels, such as components of the splicing process or associated transcription factors or associated stability factors, could be targeted by treatment with a small molecule, it would be possible to restore protein expression levels such that the unwanted effects of the expression of aberrant levels of RNA transcripts or associated proteins. Therefore, there is a need for methods of modulating the amount of RNA transcripts encoded by certain genes as a way to prevent or treat diseases associated with aberrant expression of the RNA transcripts or associated proteins. Structural Targets [0382] Mutations and/or aberrant secondary or tertiary RNA structures in cis-acting elements can induce three- dimensional structural change in pre-mRNA. Mutations and/or aberrant secondary RNA structures in cis-acting elements can induce three-dimensional structural change in pre-mRNA when the pre-mRNA is, for example, bound to at least one snRNA, or at least one snRNP, or at least one other auxiliary splicing factor. For example, non-canonical base pairing of a non-canonical splice site sequence to a snRNA can form a bulge. For example, a bulge can be formed when the 5’ss is bound to U1-U12 snRNA or a portion thereof. For example, a bulge can be induced to form when 5’ss containing at least one mutation is bound to U1-U12 snRNA or a portion thereof. For example, a bulge can be formed when the cryptic 5’ss is bound to U1-U12 snRNA or a portion thereof. For example, a bulge can be induced to form when cryptic 5’ss containing at least one mutation is bound to U1-U12 snRNA or a portion thereof. For example, a bulge can be formed when the 3’ss is bound to U2 snRNA or a portion thereof. For example, a bulge can be induced to form when the 3’ss is bound to U2 snRNA or a portion thereof. For example, a bulge can be formed when the cryptic 3’ss is bound to U2 snRNA or a portion thereof. For example, a bulge can be induced to form when the cryptic 3’ss is bound to U2 snRNA or a portion thereof. The protein components of U1 and U2 may or may not present to form the bulge. Exemplary 5’ splice site mutations and/or with aberrant secondary and/or tertiary structures that can induce a bulge structure are described herein. A polynucleotide in the methods disclosed herein can contain any one of exemplary the 5’ splice site sequences described herein.

[0383] In some embodiments, a small molecule can bind to a bulge. In some embodiments, a bulge is naturally occurring. In some embodiments, a bulge is formed by non-canonical base-pairing between the splice site and the small nuclear RNA. For example, a bulge can be formed by non-canonical base-pairing between the 5’ss and U1-U12 snRNA. The bulge can comprise 1 nucleotide, 2 nucleotides, 3 nucleotides, 4 nucleotides, 5 nucleotides, 6 nucleotides, 7 nucleotides, 8 nucleotides, 9 nucleotides, 10 nucleotides, 11 nucleotides, 12 nucleotides, 13 nucleotides, 14 nucleotides, or 15 nucleotides. In some embodiments, 3-dimensional structural changes can be induced by a mutation without bulge formation. In some embodiments, a bulge may be formed without any mutation in a splice site. In some embodiments, a recognition portion can be formed by a mutation in any of the cis-acting elements. In some embodiments, a small molecule can bind to a recognition portion that is induced by a mutation. In some embodiments, a mutation and/or aberrant secondary or tertiary RNA structure at an authentic 5’ splice site can result in splicing at a cryptic 5’ splice site. In some embodiments, a mutation and/or aberrant secondary or tertiary RNA structure can be in one of the regulatory elements including ESEs, ESSs, ISEs, and ISSs.

[0384] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide in an exon. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide upstream (5’) of the splice site of the splice site sequence. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the -1 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNN*nnnnnn, wherin N* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the -2 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NN*Nnnnnnn, wherein N* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the -3 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of N*NNnnnnnn, wherin N* represents a bulged nucleotide.

[0385] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide in an intron. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide downstream (3’) of the splice site of the splice site sequence.

[0386] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +1 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNn*nnnnn, wherein n* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +2 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNnn*nnnn, wherein n* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +3 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNnnn*nnn, wherin n* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +4 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNnnnn*nn, wherin n* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +5 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNnnnnn*n, wherin n* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +6 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNnnnnnn*, wherin n* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the +7 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNnnnnnnn*, wherin n* represents a bulged nucleotide.

[0387] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with one or more bulged nucleotides at the -1, -2, -3, +1, +2, +3, +4, +5, +6 and/or +7 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNN*nnnnnn, NN*Nnnnnnn, N*NNnnnnnn, NNNn*nnnnn, NNNnn*nnnn, NNNnnn*nnn, NNNnnnn*nn, NNNnnnnn*n, NNNnnnnnn*, or NNNnnnnnnn*, wherin N* or n* represents a bulged nucleotide. [0388] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with one or more bulged nucleotides at the -1, -2, and/or -3 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNN*nnnnnn, NN*Nnnnnnn, or N*NNnnnnnn, wherin N* represents a bulged nucleotide.

[0389] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with one or more bulged nucleotides at the +1, +2, +3, +4, +5, +6 and/or +7 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NNNn*nnnnn, NNNnn*nnnn, NNNnnn*nnn, NNNnnnn*nn, NNNnnnnn*n, NNNnnnnnn*, or NNNnnnnnnn*, wherin n* represents a bulged nucleotide.

[0390] In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the -1 position relative to the splice site of the splice site sequence and a bulged nucleotide at the -2 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre-mRNA comprising a splice site sequence of NN*N*nnnnnn, wherin N* represents a bulged nucleotide. In some embodiments, a target of an SMSM is a pre-mRNA comprising a splice site sequence with a bulged nucleotide at the -2 position relative to the splice site of the splice site sequence and a bulged nucleotide at the -3 position relative to the splice site of the splice site sequence. For example, a target of an SMSM can be a pre- mRNA comprising a splice site sequence of N*N*Nnnnnnn, wherin N* represents a bulged nucleotide.

[0391] In some embodiments, an SMSM interacts with a bulged nucleotide of an RNA duplex comprising a splice site. In some embodiments, the RNA duplex comprises pre-mRNA. In some embodiments, an SMSM binds to an RNA duplex and interacts with an unpaired bulged nucleobase of an RNA duplex comprising a splice site. In some embodiments, a first portion of the SMSM interacts with the bulged nucleotide on a first RNA strand of the RNA duplex. In some embodiments, a second portion of the SMSM interacts with one or more nucleotides of a second RNA strand of the RNA duplex, wherein the first RNA strand is not the second RNA strand. In some embodiments, the SMSM forms one or more intermolecular interactions with the duplex RNA, for example, an ionic interaction, a hydrogen bond, a dipole-dipole interaction or a van der Waals interaction. In some embodiments, the SMSM forms one or more intermolecular interactions with the bulged nucleotide, for example, an ionic interaction, a hydrogen bond, a dipole-dipole interaction or a van der Waals interaction.

[0392] In some embodiments, the duplex RNA comprises an alpha helix. In some embodiments, the bulged nucleotide is located on an external portion of a helix of the duplex RNA. In some embodiments, the bulged nucleotide is located within an internal portion of the helix of the duplex RNA.

[0393] In some embodiments, a rate of exchange of the bulged nucleotide from within the interior of a helix of the duplex RNA to an exterior portion of the helix is reduced.

[0394] In some embodiments, the SMSM modulates a distance of the bulged nucleotide from a second nucleotide of the duplex RNA. In some embodiments, the SMSM reduces the distance of the bulged nucleotide from a second nucleotide of the duplex RNA. In some embodiments, the SMSM increases the distance of the bulged nucleotide from a second nucleotide of the duplex RNA. [0395] In some embodiments, the bulged nucleotide is located within the interior of a helix of the duplex RNA of the complex. In some embodiments, the bulged nucleotide has modulated base stacking within an RNA strand of the RNA duplex. In some embodiments, the bulged nucleotide has increased base stacking within an RNA strand of the RNA duplex. In some embodiments, the bulged nucleotide has decreased base stacking within an RNA strand of the RNA duplex.

[0396] In some embodiments, the SMSM modulates splicing at the splice site of the RNA duplex. In some embodiments, the SMSM increases splicing at the splice site of the RNA duplex. In some embodiments, the SMSM reduces splicing at the splice site of the RNA duplex. In some embodiments, the SMSM reduces a size of a bulge of the RNA duplex. In some embodiments, the SMSM removes a bulge of the RNA duplex. In some embodiments, the SMSM stabilizes a bulge of the RNA duplex.

[0397] In some embodiments, the unpaired bulged nucleotide is free to rotate around a phosphate backbone of an RNA strand of the RNA duplex in the absence of the SMSM. In some embodiments, the SMSM reduces a rate of rotation of the unpaired bulged nucleotide. In some embodiments, the SMSM reduces a rate of rotation of the unpaired bulged nucleotide around a phosphate backbone of an RNA strand of the RNA duplex.

[0398] In some embodiments, the SMSM is not an aptamer.

[0399] Also, provided herein is a method of modulating splicing comprising contacting a small molecule splicing modulator compound (SMSM) to a cell; wherein the SMSM interacts with an unpaired bulged nucleotide of an RNA duplex in the cell; wherein the duplex RNA comprises a splice site; and wherein the SMSM modulates splicing of the RNA duplex.

[0400] Provided herein is a method for modulating the relative position of a first nucleotide relative to a second nucleotide, wherein the first nucleotide and the second nucleotide are within a duplex RNA, the method comprising contacting a small molecule splicing modulator compound (SMSM) to the duplex RNA, or a pharmaceutically acceptable salt thereof, wherein the first nucleotide is a bulged nucleotide of the RNA duplex; wherein the duplex RNA comprises a splice site.

[0401] In some embodiments, the duplex RNA comprises a helix.

[0402] In some embodiments, the bulged nucleotide is located on an external portion of a helix of the duplex RNA prior to contacting the SMSM.

[0403] In some embodiments, SMSM forms one or more intermolecular interactions with the duplex RNA.

[0404] In some embodiments, the SMSM forms one or more intermolecular interactions with an unpaired bulged nucleotide. In some embodiments, the intermolecular interaction is selected from the group comprising an ionic interaction, a hydrogen bond, a dipole-dipole interaction or a van der Waals interaction. In some embodiments, a rate of exchange of the unpaired bulged nucleotide from within the interior of a helix of the duplex RNA to an exterior portion of the helix is reduced. In some embodiments, a rate of rotation of the unpaired bulged nucleotide is reduced. In some embodiments, a rate of rotation of the unpaired bulged nucleotide around a phosphate backbone of an RNA strand of the RNA duplex is reduced. In some embodiments, a distance of the unpaired bulged nucleotide from a second nucleotide of the duplex RNA is modulated after contacting the SMSM. In some embodiments, the distance of the unpaired bulged nucleotide from a second nucleotide of the duplex RNA is reduced. In some embodiments, unpaired bulged nucleotide is located within the interior of the helix of the duplex RNA. In some embodiments, a size of a bulge of the RNA duplex is reduced. In some embodiments, a bulge of the RNA duplex is removed or maintained.

[0405] In some embodiments, splicing at the splice site of the RNA duplex is promoted. In some embodiments, base stacking of the unpaired bulged nucleotide within an RNA strand of the RNA duplex is increased after contacting the SMSM. In some embodiments, the distance of the unpaired bulged nucleotide from a second nucleotide of the duplex RNA is increased or maintained. In some embodiments, a bulge of the RNA duplex is stabilized after contacting the SMSM. In some embodiments, the unpaired bulged nucleotide is located on an exterior portion of a helix of the duplex RNA. In some embodiments, a size of a bulge of the RNA duplex is increased. In some embodiments, splicing at the splice site of the RNA duplex is inhibited. In some embodiments, splicing is inhibited at the splice site. In some embodiments, base stacking of the unpaired bulged nucleotide within an RNA strand of the RNA duplex is reduced after contacting the SMSM.

[0406] Exemplary sites targeted by the SMSMs described herein include 5’ splice sites, 3’ splice sites, polypyrimidine tracts, branch sites, splicing enhancers and silencer elements. Mutations or aberrant secondary or tertiary RNA structures at hot spots can create mRNA sites or scaffold sequences that can be targeted. For example, many exons are flanked by the intronic dinucleotides GT and AG at the 5’ and 3’ splice sites, respectively. For example, mutations or aberrant secondary or tertiary RNA structures at these sites can cause, e.g., exclusion of an adjacent exon or inclusion of an adjacent intron. Many factors influence the complex pre- mRNA splicing process, including several hundred different proteins, at least five spliceosomal snRNAs, sequences on the mRNA, sequence length, enhancer and silencer elements, and strength of splicing signals. Exemplary sites targeted by the SMSMs described herein include secondary and sometimes tertiary structures of RNA. For example, exemplary sites targeted by the SMSMs described herein include a stem loop, hairpin, branch point sequence (BPS), polypyrimidine tract (PPT), 5’ splice site (5’ss) and 3’ splice site (3’ss), duplex snRNA and splice sites and trans acting protein binding to RNA. The target pre-mRNA can comprise a defective sequence, such as a sequence that produces a deficient protein, such as a protein with altered function such as enzyme activity, or expression, such as lack of expression. In some embodiments, the defective sequence impacts the structure of the RNA. In some embodiments, the defect sequence impacts recognition by snRNP.

[0407] In addition to consensus splice site sequences, structural constraints, including those resulting from mutations, can affect cis-acting sequences such as exonic/intronic splicing enhancers (ESE/ISE) or silencer elements (ESS/ISS).

[0408] In some embodiments, a mutation in native DNA and/or pre-mRNA, or an aberrant secondary or tertiary structure of RNA, creates a new splice site sequence. For example, a mutation or aberrant RNA structure may cause native regions of the RNA that are normally dormant, or play no role as splicing elements, to become activated and serve as splice sites or splice elements. Such splice sites and elements can be referred to as “cryptic”. For example, a native intron may become divided into two aberrant introns, with a new exon situated there between. For example, a mutation may create a new splice site between a native 5’ splice site and a native branch point. For example, a mutation may activate a cryptic branch point sequence between a native splice site and a native branch point. For example, a mutation may create a new splice site between a native branch point and a native splice site and may further activate a cryptic splice site and a cryptic branch point sequentially upstream from the aberrant mutated splice site.

[0409] In some embodiments, a mutation or misexpression of trans-acting proteins that regulate splicing activity may cause native regions of the RNA that are normally dormant, or play no role as splicing elements, to become activated and serve as splice sites or splice elements. For example, a mutation or misexpression of an SR protein may cause native regions of the RNA that are normally dormant, or play no role as splicing elements, to become activated and serve as splice sites or splice elements.

[0410] In some embodiments, a mutation in native DNA and/or pre-mRNA inhibits splicing at a splice site. For example, a mutation may result in a new splice site upstream from (i.e., 5’ to) a native splice site sequence and downstream from (i.e., 3’ to) a native branch point sequence. The native splice site sequence and the native branch point sequence may serve as members of both the native set of splice site sequences and the aberrant set of splice site sequences.

[0411] In some embodiments, a native splice element (e.g., a branch point) is also a member of the set of aberrant splice elements. For example, SMSMs provided herein can block the native element and activate a cryptic element (e.g., a cryptic 5’ss, a cryptic 3’ss or a cryptic branch point), which may recruit remaining members of the native set of splice elements to promote correct splicing over incorrect splicing. In some embodiments, an activated cryptic splice element is in an intron. In some embodiments, an activated cryptic splice element is in an exon. The compounds and methods provided herein can be used to block or activate a variety of different splice elements, depending on the type of aberrant splice element (e.g., mutated splice element or non-mutated splice element) and/or depending on regulation of a splice element (e.g., regulation by upstream signaling pathways). For example, the compounds and methods provided herein can block a mutated element, a non-mutated element, a cryptic element, or a native element; it may block a 5’ splice site, a 3’ splice site, or a branch point.

[0412] In some embodiments, an alternate splicing event can be modulated by employing the compounds provided herein. For example, a compound provided herein can be introduced into a cell in which a gene is present that encodes a pre-mRNA that comprises alternate splice sites. In some embodiments, in the absence of the compound, a first splicing event occurs to produce a gene product having a particular function. For example, in the presence of the compound provided herein, the first splicing event can be inhibited. In some embodiments, in the presence of the compound provided herein, the first splicing event can be inhibited and a second or alternate splicing event occurs, resulting in expression of the same gene to produce a gene product having a different function.

[0413] In some embodiments, a first inhibited splicing event (e.g., a splicing event inhibited by a mutation, a mutation-induced bulge or a non-mutation induced bulge), is promoted or enhanced in the presence of a compound provided herein. In some embodiments, the first inhibited splicing event (e.g., a splicing event inhibited by a mutation, a mutation-induced bulge or a non-mutation induced bulge), is promoted or enhanced in the presence of a compound provided herein. For example, the inhibition of the first splicing event (e.g., a splicing event inhibited by a mutation, a mutation-induced bulge or a non-mutation induced bulge) can be restored to a corresponding first splicing event that is uninhibited, in the presence of a compound provided herein; or the inhibition of the first splicing event can be decreased, in the presence of a compound provided herein. In some embodiments, a second or alternate splicing event occurs, resulting in expression of the same gene to produce a gene product having a different function.

Target Polynucleotides

[0414] The compounds described herein can modulate splicing of gene products, such as those described herein. In some embodiments, the compounds described herein are use in the treatment, prevention and/or delay of progression of diseases or conditions (e.g., cancer and neurodegenerative diseases). In some embodiments, the compounds described herein can modulate splicing and induce a transcriptionally inactive variant or transcript of a gene product, such as those described herein. In some embodiments, the compounds described herein modulate splicing and repress a transcriptionally active variant or transcript of a gene product, such as those described herein.

[0415] Modulation of splicing by the compounds described herein includes, but is not limited to, modulation of naturally occurring splicing, splicing of an RNA expressed in a diseased cell, splicing of cryptic splice site sequences of an RNA or alternative splicing. Modulation of splicing by the compounds described herein can restore or promote correct splicing or a desired splicing event. Modulation of splicing by the compounds described herein includes, but is not limited to, prevention of aberrant splicing events, e.g., splicing events caused by mutations or aberrant secondary or tertiary structures of RNA that are associated with conditions and diseases. In some embodiments, the compounds described herein prevent or inhibit splicing at a splice site sequence. In some embodiments, the compounds described herein promote or increase splicing at a splice site sequence. In some embodiments, the compounds described herein modulate splicing at a specific splice site sequence.

[0416] The compositions and methods described herein can be used to modulate splicing of a target RNA, e.g., pre-mRNAs, encoded by genes. Examples of genes encoding a target RNA, e.g., a pre-mRNA, include, but are not limited to the genes described herein. Examples of genes encoding a target RNA of the compositions and methods described herein, e.g., a pre-mRNA, include, but are not limited to ABCA4, ABCD1, ACADM, ACADSB, ADA, ADAMTS13, AGL, AGT, ALB, ALDH3A2, ALG6, ANGPTL3, APC, APOA1, APOB, APOC3, AR, ATM, ATP7A, ATP7B, ATR, ATXN2, ATXN3, B2M, BCL2–like 11 (BIM), BMP2K, BRCA1, BRCA2, BTK, C3, CACNA1B, CACNA1C, CALCA, CAT, CD33, CD46, CDH1, CDH23, CFB, CFTR, CHM, CLCN1, COL11A1, COL11A2, COL1A1, COL1A2, COL2A1, COL3A1, COL4A5, COL6A1, COL7A1, COL9A2, COLQ, CREBBP, CSTB, CUL4B, CYBB, CYP17, CYP19, CYP27A1, DES, DGAT2, DMD, DUX4, DYSF, EGFR, EMD, ETV4, F11, F13A1, F5, F7, F8, FAH, FANCA, FANCC, FANCG, FBN1, FECH, FGA, FGFR2, FGG, FIX, FLNA, FOXM1, FRAS1, GALC, GBA, GCGR, GH1, GHR, GHV, GLA, HADHA, HBA2, HBB, HEXA, HEXB, HLCS, HMBS, HMGCL, HNF1A, HPRT1, HPRT2, HSF4, HSPG2, HTT, IDH1, IDS, IKBKAP, IL7RA, INSR, ITGB2, ITGB3, ITGB4, JAG1, KLKB1, KRAS, KRT5, L1CAM, LAMA2, LAMA3, LDLR, LGALS3, LMNA, LPA, LPL, LRRK2, MADD, MAPT, MET, MLH1, MSH2, MST1R, MTHFR, MUT, MVK, NF1, NF2, NR1H4, OAT, OPA1, OTC, OXT, PAH, PBGD, PCCA, PDH1, PGK1, PHEX, PKD2, PKLR, PKM1, PKM2, PLEKHM1, PLKR, POMT2, PRDM1, PRKAR1A, PROC, PSEN1, PTCH1, PTEN, PYGM, RP6KA3, RPGR, RSK2, SBCAD, SCN5A, SCNA, SERPINA1, SH2D1A, SLC12A3, SLC6A8, SMN2, SOD1, SPINK5, SPTA1, TMPRSS6, TP53, TRAPPC2, TSC1, TSC2, TSHB, TTN, TTR, UBE3A, UGT1A1 and USH2A.

[0417] Examples of genes encoding a target RNA, e.g., a pre-mRNA, include, but are not limited to the genes in Table 2B. Examples of genes encoding a target RNA of the compositions and methods described herein, e.g., a pre-mRNA, include, but are not limited to ABCD1, APOB, AR, ATM, BRCA1, C3, CFTR, COL1A1, COL3A1, COL6A1, COL7A1, CYP19, CYP27A1, DMD, F5, F7, FAH, FBN1, FGA, GCK, GHV, HBA2, HBB, HMGCL, HPRT1, HXA, IDS, ITGB2, ITGB3, KRT5, LDLR, LMNA, LPL, MTHFR, NF1, NF2, PBGD, PGK1, PKD1, PTEN, RPGR, TP53, TSC2, UGT1A1 and YGM.

[0418] Examples of genes encoding a target RNA, e.g., a pre-mRNA, include, but are not limited to the genes in Table 2C. Examples of genes encoding a target RNA of the compositions and methods described herein, e.g., a pre-mRNA, include, but are not limited to genes encoding a target RNA, e.g., a pre-mRNA, with a splice site comprising a splice site sequence of AGAguaag. Examples of genes encoding a target RNA of the compositions and methods described herein, e.g., a pre-mRNA, include, but are not limited to ABCA9, ABCB1, ABCB5, ACADL, ACSS2, ADAL, ADAM10, ADAM15, ADAMTS20, ADAMTS6, ADAMTS9, ADCY10, ADCY8, AFP, AGL, AHCTF1, AKAP10, AKAP3, ALAS1, ALS2CL, AMBRA1, ANK3, ANTXR2, ANXA10, ANXA11, AP2A2, AP4E1, APOB, ARFGEF1, ARFGEF2, ARHGAP1, ARHGAP18, ARHGEF18, ARHGEF2, ARPC3, ARS2, ASH1L, ASNSD1, ASPM, ATAD5, ATG4A, ATP11C, ATP6V1G3, BBOX1, BCS1L, BMPR2, BRCC3, BRSK2, C10orf137, C11orf70, C12orf51, C13orf1, C13orf15, C14orf118, C15orf29, C15orf42, C16orf33, C16orf38, C16orf48, C18orf8, C19orf42, C1orf107, C1orf114, C1orf130, C1orf149, C1orf27, C1orf71, C1orf94, C1R, C20orf74, C21orf70, C3orf23, C4orf18, C5orf34, C8B, C8orf33, C9orf114, C9orf86, C9orf98, CA11, CAB39, CACNA2D1, CALCOCO2, CAMK1D, CAMKK1, CAPN9, CAPSL, CBX1, CBX3, CCDC102B, CCDC11, CCDC15, CCDC18, CCDC5, CCDC81, CD4, CDC14A, CDC16, CDC2L5, CDC42BPB, CDCA8, CDH10, CDH11, CDH24, CDH8, CDH9, CDK5RAP2, CDK8, CELSR3, CENPI, CENTB2, CENTG2, CEP110, CEP170, CEP192, CETP, CFH, CHAF1A, CHD9, CHIC2, CHN1, CLIC2, CLINT1, CLPB, CMIP, CNOT1, CNOT7, COG3, COL11A1, COL12A1, COL14A1, COL19A1, COL1A1, COL1A2, COL22A1, COL24A1, COL25A1, COL29A1, COL2A1, COL3A1, COL4A1, COL4A2, COL4A5, COL4A6, COL5A2, COL9A1, COMTD1, COPA, COPB2, COPS7B, COPZ2, CPSF2, CPXM2, CR1, CREBBP, CRKRS, CSE1L, CT45-6, CUBN, CUL5, CXorf41, CYP3A4, CYP3A43, CYP3A5, DCC, DCTN3, DDA1, DDX1, DDX24, DDX4, DENND2D, DEPDC2, DHFR, DHRS7, DIP2A, DMD, DNAH3, DNAH8, DNAI1, DNAJA4, DNAJC13, DNAJC7, DNTTIP2, DOCK11, DOCK4, DPP4, DSCC1, DYNC1H1, ECM2, EDEM3, EFCAB3, EFCAB4B, EIF3A, ELA1, ELA2A, EMCN, EML5, ENPP3, EPB41L5, EPHA3, EPHB1, EPHB3, EPS15, ERCC8, ERGIC3, ERMN, ERMP1, ERN1, ERN2, ETS2, EVC2, EXO1, EXOC4, F3, FAM13A1, FAM13B1, FAM13C1, FAM184A, FAM19A1, FAM20A, FAM23B, FAM65C, FANCA, FANCM, FANK1, FAR2, FBXO15, FBXO18, FBXO38, FEZ2, FGFR1OP, FGFR1OP2, FGFR2, FGR, FLJ35848, FLJ36070, FLNA, FN1, FNBP1L, FOLH1, FRAS1, FUT9, FZD3, FZD6, GAB1, GALNT3, GART, GAS2L3, GCG, GJA1, GLT8D1, GNAS, GNB5, GOLGB1, GOLT1A, GOLT1B, GPATCH1, GPR160, GRAMD3, GRHPR, GRIA1, GRIA3, GRIA4, GRIN2B, GRM3, GRM4, GRN, GSDMB, GSTCD, GTPBP4, HDAC3, HDAC5, HDX, HEPACAM2, HERC1, HIPK3, HNRNPH1, HSPA9, HSPG2, HTT, ICA1, IFI44L, IL1R2, IL5RA, IMMT, INPP5D, INTU, IPO4, IPO8, ISL2, IWS1, JAK1, JAK2, KATNAL2, KCNN2, KCNT2, KIAA0256, KIAA0586, KIAA1033, KIAA1219, KIAA1622, KIF15, KIF16B, KIF5A, KIF5B, KIF9, KIN, KIR2DL5B, KIR3DL2, KIR3DL3, KLF12, KLF3, KPNA5, KREMEN1, KRIT1, KRTCAP2, L1CAM, L3MBTL, L3MBTL2, LACE1, LAMA2, LAMB1, LGMN, LHCGR, LHX6, LIMCH1, LIMK2, LMBRD1, LMBRD2, LMLN, LMO2, LOC390110, LPCAT2, LRP4, LRPPRC, LRRC19, LRRC42, LUM, LVRN, LYST, MADD, MAGI1, MAGT1, MALT1, MAP4K4, MAPK8IP3, MAPK9, MATN2, MCF2L2, MDGA2, MEGF10, MEGF11, MEMO1, MGAM, MGAT4A, MGC34774, MIB1, MIER2, MKL2, MLANA, MLL5, MLX, MME, MPI, MRAP2, MRPL39, MRPS28, MRPS35, MTDH, MTF2, MUC2, MYB, MYCBP2, MYH2, MYO19, MYO3A, MYO9B, MYOM2, MYOM3, NAG, NARG1, NARG2, NCOA1, NDFIP2, NEDD4, NEK1, NEK5, NFIA, NFIX, NFRKB, NKAP, NLRC3, NLRC5, NME7, NOL10, NOS1, NOS2A, NOTCH1, NPM1, NR4A3, NRXN1, NSMAF, NSMCE2, NT5C3, NUBP1, NUBPL, NUMA1, NUP160, NUP98, NUPL1, OBFC2B, OLIG2, OSBPL11, OSBPL8, OSGEPL1, PADI4, PAH, PAN2, PAPOLG, PARVB, PAWR, PCNX, PCOTH, PDCD4, PDE8B, PDIA3, PDK4, PDS5A, PDS5B, PHACTR4, PHKB, PHLDB2, PHTF1, PIAS1, PIGF, PIGN, PIGT, PIK3C2G, PIK3CG, PIK3R1, PIWIL3, PKHD1L1, PLCB1, PLCB4, PLCG1, PLD1, PLEKHA5, PLEKHA7, PLXNC1, POLN, POLR3D, POMT2, POSTN, PPFIA2, PPP1R12A, PPP3CB, PPP4C, PPP4R1L, PPP4R2, PRAME, PRC1, PRIM1, PRIM2, PRKG1, PRMT7, PROCR, PROSC, PROX1, PRPF40B, PRPF4B, PRRG2, PSD3, PSMAL, PTK2, PTK2B, PTPN11, PTPN22, PTPN3, PTPN4, PTPRD, PTPRK, PTPRM, PUS10, PVRL2, QRSL1, RAB11FIP2, RAB23, RB1CC1, RBM39, RBM45, REC8, RFC4, RHPN2, RLN3, RNF32, RNFT1, ROCK1, ROCK2, RP1, RP11-265F1, RP13-36C9., RPAP3, RPN1, RTEL1, RYR3, SAAL1, SAE1, SCN11A, SCN1A, SCN3A, SCO1, SCYL3, SDK2, SEC24A, SEC24D, SEC31A, SEL1L, SENP3, SENP6, SENP7, SETD3, SETD4, SGCE, SGOL2, SGPL1, SH3PXD2A, SH3PXD2B, SH3RF2, SH3TC2, SIPA1L2, SIPA1L3, SKAP1, SKIV2L2, SLC13A1, SLC28A3, SLC38A1, SLC38A4, SLC39A10, SLC4A2, SMARCA1, SMARCA5, SMC5, SNRK, SNRP70, SNX6, SPAG9, SPATA13, SPATA4, SPATS1, SPECC1L, SPP2, SRP72, SSX3, SSX5, SSX9, STAG1, STAMBPL1, STARD6, STK17B, STX3, STXBP1, SUCLG2, SULF2, SUPT16H, SYCP1, SYTL5, TAF2, TBC1D3G, TBC1D8B, TBCEL, TBK1, TCEB3, TCF12, TCP11L2, TDRD3, TEAD1, TET2, TFRC, TG, THOC2, TIAL1, TIAM2, TIMM50, TLK2, TMEM156, TMEM27, TMF1, TNFRSF10A, TNFRSF10B, TNFRSF8, TNK2, TNKS, TNKS2, TOM1L1, TOP2B, TP53INP1, TP63, TRAF3IP3, TRIM44, TRIM65, TRIML1, TRIML2, TRPM7, TTC17, TTLL5, TTN, TTPAL, UHRF1BP1, UNC45B, UNC5C, USP38, USP39, USP6, UTP15, UTP18, UTRN, UTX, UTY, UVRAG, UXT, VAPA, VPS29, VPS35, VTI1A, VTI1B, VWA3B, WDFY2, WDR17, WDR26, WDR44, WDR67, WDTC1, WRNIP1, WWC3, XRN1, XRN2, XX-FW88277, YARS, ZBTB20, ZC3HAV1, ZC3HC1, ZNF114, ZNF365, ZNF37A, ZNF618 and ZWINT.

[0419] Examples of genes encoding a target RNA, e.g., a pre-mRNA, include, but are not limited to the genes in Table 2D. Examples of genes encoding a target RNA of the compositions and methods described herein, e.g., a pre-mRNA, include, but are not limited to genes encoding a target RNA, e.g., a pre-mRNA, with a splice site comprising a splice site sequence of GGAgtaag. Examples of genes encoding a target RNA of the compositions and methods described herein, e.g., a pre-mRNA, include, but are not limited to ABCC9, ACTG2, ADAM22, ADAM32, ADAMTS12, ADCY3, ADRBK2, AFP, AKNA, APOH, ARHGAP26, ARHGAP8, ATG16L2, ATP13A5, B4GALNT3, BBS4, BRSK1, BTAF1, C11orf30, C11orf65, C14orf101, C15orf60, C1orf87, C2orf55, C4orf29, C6orf118, C9orf43, CACHD1, CACNA1G, CACNA1H, CAPN3, CARKD, CCDC131, CCDC146, CD1B, CDK6, CEL, CGN, CGNL1, CHL1, CLEC16A, CLK1, CLPTM1, CMYA5, CNGA3, CNTN6, COL11A1, COL15A1, COL17A1, COL1A1, COL2A1, CRYZ, CSTF3, CYFIP2, CYP24A1, CYP4F2, CYP4F3, DAZ2, DCBLD1, DCUN1D4, DDEF1, DDX1, DHRS9, DMTF1, DOCK10, DPP3, DPY19L2P2, DVL3, EFNA4, EFTUD2, EPHA4, EPHB2, ERBB4, ERCC1, FAM134A, FAM161A, FAM176B, FCGBP, FGD6, FKBP3, GAPDH, GBGT1, GFM1, GPR158, GRIA1, GSTCD, GSTO2, HCK, HLA-DPB1, HLA-G, HLTF, HP1BP3, HPGD, HSF2BP, INTS3, IQGAP2, ITFG1, ITGAL, ITGB1, ITIH1, ITPR2, JMJD1C, KALRN, KCNN2, KIAA0528, KIAA0564, KIAA1166, KIAA1409, KIAA1787, KIF3B, KLHL20, KLK12, LAMA1, LARP7, LENG1, LOC389634, LRWD1, LYN, MAP2K1, MCM6, MEGF10, MGAM, MGAT5, MGC16169, MKKS, MPDZ, MRPL11, MS4A13, MSMB, MTIF2, NDC80, NEB, NEK11, NFE2L2, NFKBIL2, NKAIN2, NLRC3, NLRC5, NLRP13, NLRP7, NLRP8, NT5C, NUDT5, NUP88, OBFC2A, OPN4, OPTN, PARD3, PBRM1, PCBP4, PDE10A, PDLIM5, PDXK, PDZRN3, PELI2, PGM2, PIP5K1A, PITRM1, PKIB, PMFBP1, POMT2, PRKCA, PRODH, PRUNE2, PTPRN2, PTPRT, RALBP1, RALGDS, RBL2, RFT1, RFTN1, RIF1, RMND5B, RNF11, RNGTT, RPS6KA6, RRM1, RRP1B, RTF1, RUFY1, SCN2A, SCN4A, SCN8A, SDK1, SEZ6, SFRS12, SH3BGRL2, SIVA1, SLC22A17, SLC25A14, SLC6A11, SLC6A13, SLC6A6, SMTN, SNCAIP, SNX6, STAT6, SUPT6H, SV2C, SYCP2, SYT6, TAF2, TBC1D26, TBC1D29, TBPL1, TECTB, TEK, TGM7, TGS1, TM4SF20, TM6SF1, TMEM194A, TMEM77, TOM1L2, TP53BP2, TP53I3, TRPM3, TRPM5, TSPAN7, TTLL9, TUSC3, TXNDC10, UCK1, USH2A, USP1, UTP20, VPS39, WDR16, ZC3H7A, ZFYVE1, ZNF169 and ZNF326.

[0420] The SMSM compounds and methods of their use described herein can modulate splicing, such as aberrant splicing of polynucleotide encoded by a gene, e.g., an ABCA4, ABCA9, ABCB1, ABCB5, ABCC9, ABCD1, ACADL, ACADM, ACADSB, ACSS2, ACTG2, ADA, ADAL, ADAM10, ADAM15, ADAM22, ADAM32, ADAMTS12, ADAMTS13, ADAMTS20, ADAMTS6, ADAMTS9, ADCY10, ADCY3, ADCY8, ADRBK2, AFP, AGL, AGT, AHCTF1, AKAP10, AKAP3, AKNA, ALAS1, ALB, ALDH3A2, ALG6, ALS2CL, AMBRA1, ANGPTL3, ANK3, ANTXR2, ANXA10, ANXA11, AP2A2, AP4E1, APC, APOA1, APOB, APOC3, APOH, AR, ARFGEF1, ARFGEF2, ARHGAP1, ARHGAP18, ARHGAP26, ARHGAP8, ARHGEF18, ARHGEF2, ARPC3, ARS2, ASH1L, ASNSD1, ASPM, ATAD5, ATG16L2, ATG4A, ATM, ATP11C, ATP13A5, ATP6V1G3, ATP7A, ATP7B, ATR, ATXN2, ATXN3, B2M, B4GALNT3, BBOX1, BBS4, BCL2–like 11 (BIM), BCS1L, BMP2K, BMPR2, BRCA1, BRCA2, BRCC3, BRSK1, BRSK2, BTAF1, BTK, C10orf137, C11orf30, C11orf65, C11orf70, C12orf51, C13orf1, C13orf15, C14orf101, C14orf118, C15orf29, C15orf42, C15orf60, C16orf33, C16orf38, C16orf48, C18orf8, C19orf42, C1orf107, C1orf114, C1orf130, C1orf149, C1orf27, C1orf71, C1orf87, C1orf94, C1R, C20orf74, C21orf70, C2orf55, C3, C3orf23, C4orf18, C4orf29, C5orf34, C6orf118, C8B, C8orf33, C9orf114, C9orf43, C9orf86, C9orf98, CA11, CAB39, CACHD1, CACNA1B, CACNA1C, CACNA1G, CACNA1H, CACNA2D1, CALCA, CALCOCO2, CAMK1D, CAMKK1, CAPN3, CAPN9, CAPSL, CARKD, CAT, CBX1, CBX3, CCDC102B, CCDC11, CCDC131, CCDC146, CCDC15, CCDC18, CCDC5, CCDC81, CD1B, CD33, CD4, CD46, CDC14A, CDC16, CDC2L5, CDC42BPB, CDCA8, CDH1, CDH10, CDH11, CDH23, CDH24, CDH8, CDH9, CDK5RAP2, CDK6, CDK8, CEL, CELSR3, CENPI, CENTB2, CENTG2, CEP110, CEP170, CEP192, CETP, CFB, CFH, CFTR, CGN, CGNL1, CHAF1A, CHD9, CHIC2, CHL1, CHM, CHN1, CLCN1, CLEC16A, CLIC2, CLINT1, CLK1, CLPB, CLPTM1, CMIP, CMYA5, CNGA3, CNOT1, CNOT7, CNTN6, COG3, COL11A1, COL11A2, COL12A1, COL14A1, COL15A1, COL17A1, COL19A1, COL1A1, COL1A2, COL22A1, COL24A1, COL25A1, COL29A1, COL2A1, COL3A1, COL4A1, COL4A2, COL4A5, COL4A6, COL5A2, COL6A1, COL7A1, COL9A1, COL9A2, COLQ, COMTD1, COPA, COPB2, COPS7B, COPZ2, CPSF2, CPXM2, CR1, CREBBP, CRKRS, CRYZ, CSE1L, CSTB, CSTF3, CT45-6, CUBN, CUL4B, CUL5, CXorf41, CYBB, CYFIP2, CYP17, CYP19, CYP24A1, CYP27A1, CYP3A4, CYP3A43, CYP3A5, CYP4F2, CYP4F3, DAZ2, DCBLD1, DCC, DCTN3, DCUN1D4, DDA1, DDEF1, DDX1, DDX24, DDX4, DENND2D, DEPDC2, DES, DGAT2, DHFR, DHRS7, DHRS9, DIP2A, DMD, DMTF1, DNAH3, DNAH8, DNAI1, DNAJA4, DNAJC13, DNAJC7, DNTTIP2, DOCK10, DOCK11, DOCK4, DPP3, DPP4, DPY19L2P2, DSCC1, DUX4, DVL3, DYNC1H1, DYSF, ECM2, EDEM3, EFCAB3, EFCAB4B, EFNA4, EFTUD2, EGFR, EIF3A, ELA1, ELA2A, EMCN, EMD, EML5, ENPP3, EPB41L5, EPHA3, EPHA4, EPHB1, EPHB2, EPHB3, EPS15, ERBB4, ERCC1, ERCC8, ERGIC3, ERMN, ERMP1, ERN1, ERN2, ETS2, ETV4, EVC2, EXO1, EXOC4, F11, F13A1, F3, F5, F7, F8, FAH, FAM134A, FAM13A1, FAM13B1, FAM13C1, FAM161A, FAM176B, FAM184A, FAM19A1, FAM20A, FAM23B, FAM65C, FANCA, FANCC, FANCG, FANCM, FANK1, FAR2, FBN1, FBXO15, FBXO18, FBXO38, FCGBP, FECH, FEZ2, FGA, FGD6, FGFR1OP, FGFR1OP2, FGFR2, FGG, FGR, FIX, FKBP3, FLJ35848, FLJ36070, FLNA, FN1, FNBP1L, FOLH1, FOXM1, FRAS1, FUT9, FZD3, FZD6, GAB1, GALC, GALNT3, GAPDH, GART, GAS2L3, GBA, GBGT1, GCG, GCGR, GCK, GFM1, GH1, GHR, GHV, GJA1, GLA, GLT8D1, GNAS, GNB5, GOLGB1, GOLT1A, GOLT1B, GPATCH1, GPR158, GPR160, GRAMD3, GRHPR, GRIA1, GRIA3, GRIA4, GRIN2B, GRM3, GRM4, GRN, GSDMB, GSTCD, GSTO2, GTPBP4, HADHA, HBA2, HBB, HCK, HDAC3, HDAC5, HDX, HEPACAM2, HERC1, HEXA, HEXB, HIPK3, HLA-DPB1, HLA-G, HLCS, HLTF, HMBS, HMGCL, HNF1A, HNRNPH1, HP1BP3, HPGD, HPRT1, HPRT2, HSF2BP, HSF4, HSPA9, HSPG2, HTT, HXA, ICA1, IDH1, IDS, IFI44L, IKBKAP, IL1R2, IL5RA, IL7RA, IMMT, INPP5D, INSR, INTS3, INTU, IPO4, IPO8, IQGAP2, ISL2, ITFG1, ITGAL, ITGB1, ITGB2, ITGB3, ITGB4, ITIH1, ITPR2, IWS1, JAG1, JAK1, JAK2, JMJD1C, KALRN, KATNAL2, KCNN2, KCNT2, KIAA0256, KIAA0528, KIAA0564, KIAA0586, KIAA1033, KIAA1166, KIAA1219, KIAA1409, KIAA1622, KIAA1787, KIF15, KIF16B, KIF3B, KIF5A, KIF5B, KIF9, KIN, KIR2DL5B, KIR3DL2, KIR3DL3, KLF12, KLF3, KLHL20, KLK12, KLKB1, KPNA5, KRAS, KREMEN1, KRIT1, KRT5, KRTCAP2, L1CAM, L3MBTL, L3MBTL2, LACE1, LAMA1, LAMA2, LAMA3, LAMB1, LARP7, LDLR, LENG1, LGALS3, LGMN, LHCGR, LHX6, LIMCH1, LIMK2, LMBRD1, LMBRD2, LMLN, LMNA, LMO2, LOC389634, LOC390110, LPA, LPCAT2, LPL, LRP4, LRPPRC, LRRC19, LRRC42, LRRK2, LRWD1, LUM, LVRN, LYN, LYST, MADD, MAGI1, MAGT1, MALT1, MAP2K1, MAP4K4, MAPK8IP3, MAPK9, MAPT, MATN2, MCF2L2, MCM6, MDGA2, MEGF10, MEGF11, MEMO1, MET, MGAM, MGAT4A, MGAT5, MGC16169, MGC34774, MIB1, MIER2, MKKS, MKL2, MLANA, MLH1, MLL5, MLX, MME, MPDZ, MPI, MRAP2, MRPL11, MRPL39, MRPS28, MRPS35, MS4A13, MSH2, MSMB, MST1R, MTDH, MTF2, MTHFR, MTIF2, MUC2, MUT, MVK, MYB, MYCBP2, MYH2, MYO19, MYO3A, MYO9B, MYOM2, MYOM3, NAG, NARG1, NARG2, NCOA1, NDC80, NDFIP2, NEB, NEDD4, NEK1, NEK11, NEK5, NF1, NF2, NFE2L2, NFIA, NFIX, NFKBIL2, NFRKB, NKAIN2, NKAP, NLRC3, NLRC5, NLRP13, NLRP7, NLRP8, NME7, NOL10, NOS1, NOS2A, NOTCH1, NPM1, NR1H4, NR4A3, NRXN1, NSMAF, NSMCE2, NT5C, NT5C3, NUBP1, NUBPL, NUDT5, NUMA1, NUP160, NUP88, NUP98, NUPL1, OAT, OBFC2A, OBFC2B, OLIG2, OPA1, OPN4, OPTN, OSBPL11, OSBPL8, OSGEPL1, OTC, OXT, PADI4, PAH, PAN2, PAPOLG, PARD3, PARVB, PAWR, PBGD, PBRM1, PCBP4, PCCA, PCNX, PCOTH, PDCD4, PDE10A, PDE8B, PDH1, PDIA3, PDK4, PDLIM5, PDS5A, PDS5B, PDXK, PDZRN3, PELI2, PGK1, PGM2, PHACTR4, PHEX, PHKB, PHLDB2, PHTF1, PIAS1, PIGF, PIGN, PIGT, PIK3C2G, PIK3CG, PIK3R1, PIP5K1A, PITRM1, PIWIL3, PKD1, PKD2, PKHD1L1, PKIB, PKLR, PKM1, PKM2, PLCB1, PLCB4, PLCG1, PLD1, PLEKHA5, PLEKHA7, PLEKHM1, PLKR, PLXNC1, PMFBP1, POLN, POLR3D, POMT2, POSTN, PPFIA2, PPP1R12A, PPP3CB, PPP4C, PPP4R1L, PPP4R2, PRAME, PRC1, PRDM1, PRIM1, PRIM2, PRKAR1A, PRKCA, PRKG1, PRMT7, PROC, PROCR, PRODH, PROSC, PROX1, PRPF40B, PRPF4B, PRRG2, PRUNE2, PSD3, PSEN1, PSMAL, PTCH1, PTEN, PTK2, PTK2B, PTPN11, PTPN22, PTPN3, PTPN4, PTPRD, PTPRK, PTPRM, PTPRN2, PTPRT, PUS10, PVRL2, PYGM, QRSL1, RAB11FIP2, RAB23, RALBP1, RALGDS, RB1CC1, RBL2, RBM39, RBM45, REC8, RFC4, RFT1, RFTN1, RHPN2, RIF1, RLN3, RMND5B, RNF11, RNF32, RNFT1, RNGTT, ROCK1, ROCK2, RP1, RP11- 265F1, RP13-36C9, RP6KA3, RPAP3, RPGR, RPN1, RPS6KA6, RRM1, RRP1B, RSK2, RTEL1, RTF1, RUFY1, RYR3, SAAL1, SAE1, SBCAD, SCN11A, SCN1A, SCN2A, SCN3A, SCN4A, SCN5A, SCN8A, SCNA, SCO1, SCYL3, SDK1, SDK2, SEC24A, SEC24D, SEC31A, SEL1L, SENP3, SENP6, SENP7, SERPINA1, SETD3, SETD4, SEZ6, SFRS12, SGCE, SGOL2, SGPL1, SH2D1A, SH3BGRL2, SH3PXD2A, SH3PXD2B, SH3RF2, SH3TC2, SIPA1L2, SIPA1L3, SIVA1, SKAP1, SKIV2L2, SLC12A3, SLC13A1, SLC22A17, SLC25A14, SLC28A3, SLC38A1, SLC38A4, SLC39A10, SLC4A2, SLC6A11, SLC6A13, SLC6A6, SLC6A8, SMARCA1, SMARCA5, SMC5, SMN2, SMTN, SNCAIP, SNRK, SNRP70, SNX6, SOD1, SPAG9, SPATA13, SPATA4, SPATS1, SPECC1L, SPINK5, SPP2, SPTA1, SRP72, SSX3, SSX5, SSX9, STAG1, STAMBPL1, STARD6, STAT6, STK17B, STX3, STXBP1, SUCLG2, SULF2, SUPT16H, SUPT6H, SV2C, SYCP1, SYCP2, SYT6, SYTL5, TAF2, TBC1D26, TBC1D29, TBC1D3G, TBC1D8B, TBCEL, TBK1, TBPL1, TCEB3, TCF12, TCP11L2, TDRD3, TEAD1, TECTB, TEK, TET2, TFRC, TG, TGM7, TGS1, THOC2, TIAL1, TIAM2, TIMM50, TLK2, TM4SF20, TM6SF1, TMEM156, TMEM194A, TMEM27, TMEM77, TMF1, TMPRSS6, TNFRSF10A, TNFRSF10B, TNFRSF8, TNK2, TNKS, TNKS2, TOM1L1, TOM1L2, TOP2B, TP53, TP53BP2, TP53I3, TP53INP1, TP63, TRAF3IP3, TRAPPC2, TRIM44, TRIM65, TRIML1, TRIML2, TRPM3, TRPM5, TRPM7, TSC1, TSC2, TSHB, TSPAN7, TTC17, TTLL5, TTLL9, TTN, TTPAL, TTR, TUSC3, TXNDC10, UBE3A, UCK1, UGT1A1, UHRF1BP1, UNC45B, UNC5C, USH2A, USP1, USP38, USP39, USP6, UTP15, UTP18, UTP20, UTRN, UTX, UTY, UVRAG, UXT, VAPA, VPS29, VPS35, VPS39, VTI1A, VTI1B, VWA3B, WDFY2, WDR16, WDR17, WDR26, WDR44, WDR67, WDTC1, WRNIP1, WWC3, XRN1, XRN2, XX-FW88277, YARS, YGM, ZBTB20, ZC3H7A, ZC3HAV1, ZC3HC1, ZFYVE1, ZNF114, ZNF169, ZNF326, ZNF365, ZNF37A, ZNF618 or ZWINT gene. [0421] For example, provided herein are splice modulating compounds that modulate splicing, such as aberrant splicing of ABCA4, ABCA9, ABCB1, ABCB5, ABCC9, ABCD1, ACADL, ACADM, ACADSB, ACSS2, ACTG2, ADA, ADAL, ADAM10, ADAM15, ADAM22, ADAM32, ADAMTS12, ADAMTS13, ADAMTS20, ADAMTS6, ADAMTS9, ADCY10, ADCY3, ADCY8, ADRBK2, AFP, AGL, AGT, AHCTF1, AKAP10, AKAP3, AKNA, ALAS1, ALB, ALDH3A2, ALG6, ALS2CL, AMBRA1, ANGPTL3, ANK3, ANTXR2, ANXA10, ANXA11, AP2A2, AP4E1, APC, APOA1, APOB, APOC3, APOH, AR, ARFGEF1, ARFGEF2, ARHGAP1, ARHGAP18, ARHGAP26, ARHGAP8, ARHGEF18, ARHGEF2, ARPC3, ARS2, ASH1L, ASNSD1, ASPM, ATAD5, ATG16L2, ATG4A, ATM, ATP11C, ATP13A5, ATP6V1G3, ATP7A, ATP7B, ATR, ATXN2, ATXN3, B2M, B4GALNT3, BBOX1, BBS4, BCL2–like 11 (BIM), BCS1L, BMP2K, BMPR2, BRCA1, BRCA2, BRCC3, BRSK1, BRSK2, BTAF1, BTK, C10orf137, C11orf30, C11orf65, C11orf70, C12orf51, C13orf1, C13orf15, C14orf101, C14orf118, C15orf29, C15orf42, C15orf60, C16orf33, C16orf38, C16orf48, C18orf8, C19orf42, C1orf107, C1orf114, C1orf130, C1orf149, C1orf27, C1orf71, C1orf87, C1orf94, C1R, C20orf74, C21orf70, C2orf55, C3, C3orf23, C4orf18, C4orf29, C5orf34, C6orf118, C8B, C8orf33, C9orf114, C9orf43, C9orf86, C9orf98, CA11, CAB39, CACHD1, CACNA1B, CACNA1C, CACNA1G, CACNA1H, CACNA2D1, CALCA, CALCOCO2, CAMK1D, CAMKK1, CAPN3, CAPN9, CAPSL, CARKD, CAT, CBX1, CBX3, CCDC102B, CCDC11, CCDC131, CCDC146, CCDC15, CCDC18, CCDC5, CCDC81, CD1B, CD33, CD4, CD46, CDC14A, CDC16, CDC2L5, CDC42BPB, CDCA8, CDH1, CDH10, CDH11, CDH23, CDH24, CDH8, CDH9, CDK5RAP2, CDK6, CDK8, CEL, CELSR3, CENPI, CENTB2, CENTG2, CEP110, CEP170, CEP192, CETP, CFB, CFH, CFTR, CGN, CGNL1, CHAF1A, CHD9, CHIC2, CHL1, CHM, CHN1, CLCN1, CLEC16A, CLIC2, CLINT1, CLK1, CLPB, CLPTM1, CMIP, CMYA5, CNGA3, CNOT1, CNOT7, CNTN6, COG3, COL11A1, COL11A2, COL12A1, COL14A1, COL15A1, COL17A1, COL19A1, COL1A1, COL1A2, COL22A1, COL24A1, COL25A1, COL29A1, COL2A1, COL3A1, COL4A1, COL4A2, COL4A5, COL4A6, COL5A2, COL6A1, COL7A1, COL9A1, COL9A2, COLQ, COMTD1, COPA, COPB2, COPS7B, COPZ2, CPSF2, CPXM2, CR1, CREBBP, CRKRS, CRYZ, CSE1L, CSTB, CSTF3, CT45-6, CUBN, CUL4B, CUL5, CXorf41, CYBB, CYFIP2, CYP17, CYP19, CYP24A1, CYP27A1, CYP3A4, CYP3A43, CYP3A5, CYP4F2, CYP4F3, DAZ2, DCBLD1, DCC, DCTN3, DCUN1D4, DDA1, DDEF1, DDX1, DDX24, DDX4, DENND2D, DEPDC2, DES, DGAT2, DHFR, DHRS7, DHRS9, DIP2A, DMD, DMTF1, DNAH3, DNAH8, DNAI1, DNAJA4, DNAJC13, DNAJC7, DNTTIP2, DOCK10, DOCK11, DOCK4, DPP3, DPP4, DPY19L2P2, DSCC1, DUX4, DVL3, DYNC1H1, DYSF, ECM2, EDEM3, EFCAB3, EFCAB4B, EFNA4, EFTUD2, EGFR, EIF3A, ELA1, ELA2A, EMCN, EMD, EML5, ENPP3, EPB41L5, EPHA3, EPHA4, EPHB1, EPHB2, EPHB3, EPS15, ERBB4, ERCC1, ERCC8, ERGIC3, ERMN, ERMP1, ERN1, ERN2, ETS2, ETV4, EVC2, EXO1, EXOC4, F11, F13A1, F3, F5, F7, F8, FAH, FAM134A, FAM13A1, FAM13B1, FAM13C1, FAM161A, FAM176B, FAM184A, FAM19A1, FAM20A, FAM23B, FAM65C, FANCA, FANCC, FANCG, FANCM, FANK1, FAR2, FBN1, FBXO15, FBXO18, FBXO38, FCGBP, FECH, FEZ2, FGA, FGD6, FGFR1OP, FGFR1OP2, FGFR2, FGG, FGR, FIX, FKBP3, FLJ35848, FLJ36070, FLNA, FN1, FNBP1L, FOLH1, FOXM1, FRAS1, FUT9, FZD3, FZD6, GAB1, GALC, GALNT3, GAPDH, GART, GAS2L3, GBA, GBGT1, GCG, GCGR, GCK, GFM1, GH1, GHR, GHV, GJA1, GLA, GLT8D1, GNAS, GNB5, GOLGB1, GOLT1A, GOLT1B, GPATCH1, GPR158, GPR160, GRAMD3, GRHPR, GRIA1, GRIA3, GRIA4, GRIN2B, GRM3, GRM4, GRN, GSDMB, GSTCD, GSTO2, GTPBP4, HADHA, HBA2, HBB, HCK, HDAC3, HDAC5, HDX, HEPACAM2, HERC1, HEXA, HEXB, HIPK3, HLA-DPB1, HLA-G, HLCS, HLTF, HMBS, HMGCL, HNF1A, HNRNPH1, HP1BP3, HPGD, HPRT1, HPRT2, HSF2BP, HSF4, HSPA9, HSPG2, HTT, HXA, ICA1, IDH1, IDS, IFI44L, IKBKAP, IL1R2, IL5RA, IL7RA, IMMT, INPP5D, INSR, INTS3, INTU, IPO4, IPO8, IQGAP2, ISL2, ITFG1, ITGAL, ITGB1, ITGB2, ITGB3, ITGB4, ITIH1, ITPR2, IWS1, JAG1, JAK1, JAK2, JMJD1C, KALRN, KATNAL2, KCNN2, KCNT2, KIAA0256, KIAA0528, KIAA0564, KIAA0586, KIAA1033, KIAA1166, KIAA1219, KIAA1409, KIAA1622, KIAA1787, KIF15, KIF16B, KIF3B, KIF5A, KIF5B, KIF9, KIN, KIR2DL5B, KIR3DL2, KIR3DL3, KLF12, KLF3, KLHL20, KLK12, KLKB1, KPNA5, KRAS, KREMEN1, KRIT1, KRT5, KRTCAP2, L1CAM, L3MBTL, L3MBTL2, LACE1, LAMA1, LAMA2, LAMA3, LAMB1, LARP7, LDLR, LENG1, LGALS3, LGMN, LHCGR, LHX6, LIMCH1, LIMK2, LMBRD1, LMBRD2, LMLN, LMNA, LMO2, LOC389634, LOC390110, LPA, LPCAT2, LPL, LRP4, LRPPRC, LRRC19, LRRC42, LRRK2, LRWD1, LUM, LVRN, LYN, LYST, MADD, MAGI1, MAGT1, MALT1, MAP2K1, MAP4K4, MAPK8IP3, MAPK9, MAPT, MATN2, MCF2L2, MCM6, MDGA2, MEGF10, MEGF11, MEMO1, MET, MGAM, MGAT4A, MGAT5, MGC16169, MGC34774, MIB1, MIER2, MKKS, MKL2, MLANA, MLH1, MLL5, MLX, MME, MPDZ, MPI, MRAP2, MRPL11, MRPL39, MRPS28, MRPS35, MS4A13, MSH2, MSMB, MST1R, MTDH, MTF2, MTHFR, MTIF2, MUC2, MUT, MVK, MYB, MYCBP2, MYH2, MYO19, MYO3A, MYO9B, MYOM2, MYOM3, NAG, NARG1, NARG2, NCOA1, NDC80, NDFIP2, NEB, NEDD4, NEK1, NEK11, NEK5, NF1, NF2, NFE2L2, NFIA, NFIX, NFKBIL2, NFRKB, NKAIN2, NKAP, NLRC3, NLRC5, NLRP13, NLRP7, NLRP8, NME7, NOL10, NOS1, NOS2A, NOTCH1, NPM1, NR1H4, NR4A3, NRXN1, NSMAF, NSMCE2, NT5C, NT5C3, NUBP1, NUBPL, NUDT5, NUMA1, NUP160, NUP88, NUP98, NUPL1, OAT, OBFC2A, OBFC2B, OLIG2, OPA1, OPN4, OPTN, OSBPL11, OSBPL8, OSGEPL1, OTC, OXT, PADI4, PAH, PAN2, PAPOLG, PARD3, PARVB, PAWR, PBGD, PBRM1, PCBP4, PCCA, PCNX, PCOTH, PDCD4, PDE10A, PDE8B, PDH1, PDIA3, PDK4, PDLIM5, PDS5A, PDS5B, PDXK, PDZRN3, PELI2, PGK1, PGM2, PHACTR4, PHEX, PHKB, PHLDB2, PHTF1, PIAS1, PIGF, PIGN, PIGT, PIK3C2G, PIK3CG, PIK3R1, PIP5K1A, PITRM1, PIWIL3, PKD1, PKD2, PKHD1L1, PKIB, PKLR, PKM1, PKM2, PLCB1, PLCB4, PLCG1, PLD1, PLEKHA5, PLEKHA7, PLEKHM1, PLKR, PLXNC1, PMFBP1, POLN, POLR3D, POMT2, POSTN, PPFIA2, PPP1R12A, PPP3CB, PPP4C, PPP4R1L, PPP4R2, PRAME, PRC1, PRDM1, PRIM1, PRIM2, PRKAR1A, PRKCA, PRKG1, PRMT7, PROC, PROCR, PRODH, PROSC, PROX1, PRPF40B, PRPF4B, PRRG2, PRUNE2, PSD3, PSEN1, PSMAL, PTCH1, PTEN, PTK2, PTK2B, PTPN11, PTPN22, PTPN3, PTPN4, PTPRD, PTPRK, PTPRM, PTPRN2, PTPRT, PUS10, PVRL2, PYGM, QRSL1, RAB11FIP2, RAB23, RALBP1, RALGDS, RB1CC1, RBL2, RBM39, RBM45, REC8, RFC4, RFT1, RFTN1, RHPN2, RIF1, RLN3, RMND5B, RNF11, RNF32, RNFT1, RNGTT, ROCK1, ROCK2, RP1, RP11-265F1, RP13-36C9, RP6KA3, RPAP3, RPGR, RPN1, RPS6KA6, RRM1, RRP1B, RSK2, RTEL1, RTF1, RUFY1, RYR3, SAAL1, SAE1, SBCAD, SCN11A, SCN1A, SCN2A, SCN3A, SCN4A, SCN5A, SCN8A, SCNA, SCO1, SCYL3, SDK1, SDK2, SEC24A, SEC24D, SEC31A, SEL1L, SENP3, SENP6, SENP7, SERPINA1, SETD3, SETD4, SEZ6, SFRS12, SGCE, SGOL2, SGPL1, SH2D1A, SH3BGRL2, SH3PXD2A, SH3PXD2B, SH3RF2, SH3TC2, SIPA1L2, SIPA1L3, SIVA1, SKAP1, SKIV2L2, SLC12A3, SLC13A1, SLC22A17, SLC25A14, SLC28A3, SLC38A1, SLC38A4, SLC39A10, SLC4A2, SLC6A11, SLC6A13, SLC6A6, SLC6A8, SMARCA1, SMARCA5, SMC5, SMN2, SMTN, SNCAIP, SNRK, SNRP70, SNX6, SOD1, SPAG9, SPATA13, SPATA4, SPATS1, SPECC1L, SPINK5, SPP2, SPTA1, SRP72, SSX3, SSX5, SSX9, STAG1, STAMBPL1, STARD6, STAT6, STK17B, STX3, STXBP1, SUCLG2, SULF2, SUPT16H, SUPT6H, SV2C, SYCP1, SYCP2, SYT6, SYTL5, TAF2, TBC1D26, TBC1D29, TBC1D3G, TBC1D8B, TBCEL, TBK1, TBPL1, TCEB3, TCF12, TCP11L2, TDRD3, TEAD1, TECTB, TEK, TET2, TFRC, TG, TGM7, TGS1, THOC2, TIAL1, TIAM2, TIMM50, TLK2, TM4SF20, TM6SF1, TMEM156, TMEM194A, TMEM27, TMEM77, TMF1, TMPRSS6, TNFRSF10A, TNFRSF10B, TNFRSF8, TNK2, TNKS, TNKS2, TOM1L1, TOM1L2, TOP2B, TP53, TP53BP2, TP53I3, TP53INP1, TP63, TRAF3IP3, TRAPPC2, TRIM44, TRIM65, TRIML1, TRIML2, TRPM3, TRPM5, TRPM7, TSC1, TSC2, TSHB, TSPAN7, TTC17, TTLL5, TTLL9, TTN, TTPAL, TTR, TUSC3, TXNDC10, UBE3A, UCK1, UGT1A1, UHRF1BP1, UNC45B, UNC5C, USH2A, USP1, USP38, USP39, USP6, UTP15, UTP18, UTP20, UTRN, UTX, UTY, UVRAG, UXT, VAPA, VPS29, VPS35, VPS39, VTI1A, VTI1B, VWA3B, WDFY2, WDR16, WDR17, WDR26, WDR44, WDR67, WDTC1, WRNIP1, WWC3, XRN1, XRN2, XX- FW88277, YARS, YGM, ZBTB20, ZC3H7A, ZC3HAV1, ZC3HC1, ZFYVE1, ZNF114, ZNF169, ZNF326, ZNF365, ZNF37A, ZNF618 or a ZWINT mRNA, such as pre-mRNA.

[0422] In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ABCA4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ABCA9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ABCB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ABCB5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ABCC9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ABCD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ACADL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ACADM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ACADSB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ACSS2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ACTG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAM10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAM15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAM22. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAM32. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ADAMTS12. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAMTS13. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAMTS20. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ADAMTS6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADAMTS9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADCY10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADCY3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADCY8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ADRBK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AFP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of AGL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AGT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AHCTF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AKAP10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AKAP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AKNA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ALAS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ALB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ALDH3A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ALG6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ALS2CL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of AMBRA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ANGPTL3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ANK3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ANTXR2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ANXA10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ANXA11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AP2A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of AP4E1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of APC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of APOA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of APOB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of APOC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of APOH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of AR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARFGEF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARFGEF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARHGAP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARHGAP18. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ARHGAP26. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARHGAP8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARHGEF18. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ARHGEF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARPC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ARS2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ASH1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ASNSD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ASPM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATAD5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATG16L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATG4A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATP11C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATP13A5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATP6V1G3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATP7A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ATP7B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATXN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ATXN3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of B2M. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of B4GALNT3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BBOX1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BBS4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BCL2–like 11 (BIM). In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BCS1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BMP2K. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BMPR2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BRCA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BRCA2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BRCC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BRSK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BRSK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BTAF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of BTK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C10orf137. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C11orf30. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C11orf65. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C11orf70. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C12orf51. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C13orf1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C13orf15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C14orf101. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C14orf118. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of C15orf29. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C15orf42. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C15orf60. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C16orf33. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C16orf38. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C16orf48. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C18orf8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C19orf42. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf107. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf114. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf130. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf149. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf27. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf71. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf87. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1orf94. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C1R. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C20orf74. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C21orf70. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C2orf55. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of C3orf23. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C4orf18. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C4orf29. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C5orf34. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C6orf118. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C8B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C8orf33. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C9orf114. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C9orf43. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C9orf86. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of C9orf98. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CA11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAB39. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CACHD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CACNA1B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CACNA1C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CACNA1G. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CACNA1H. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CACNA2D1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CALCA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CALCOCO2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAMK1D. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAMKK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAPN3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAPN9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAPSL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CARKD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CAT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CBX1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CBX3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CCDC102B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of CCDC11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CCDC131. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CCDC146. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of CCDC15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CCDC18. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CCDC5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CCDC81. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CD1B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CD33. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CD4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of CD46. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDC14A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDC16. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDC2L5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDC42BPB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDCA8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH23. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH24. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDH9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDK5RAP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDK6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CDK8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CEL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CELSR3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CENPI. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CENTB2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CENTG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CEP110. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CEP170. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CEP192. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CETP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CFB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CFH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CFTR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CGN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CGNL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CHAF1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CHD9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CHIC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of CHL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CHM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CHN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLCN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLEC16A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLIC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLINT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLPB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CLPTM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CMIP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of CMYA5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CNGA3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CNOT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CNOT7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CNTN6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COG3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL11A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of COL11A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL12A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL14A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of COL15A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL17A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL19A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of COL1A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL1A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL22A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL24A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL25A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL29A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL2A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL3A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL4A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL4A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL4A5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL4A6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL5A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL6A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL7A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL9A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COL9A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COLQ. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COMTD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COPA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COPB2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COPS7B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of COPZ2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CPSF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CPXM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CR1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CREBBP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CRKRS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CRYZ. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CSE1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CSTB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CSTF3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of CT45-6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CUBN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CUL4B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CUL5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CXorf41. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYBB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYFIP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP17. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP19. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP24A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP27A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP3A4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP3A43. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP3A5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP4F2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of CYP4F3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DAZ2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DCBLD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of DCC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DCTN3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DCUN1D4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DDA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DDEF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DDX1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DDX24. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DDX4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DENND2D. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DEPDC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DES. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of DGAT2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DHFR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DHRS7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DHRS9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DIP2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DMD. For example, the SMSM compounds and methods of their use described herein can modulate splicing of exon 51a pre-mRNA of DMD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DMTF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNAH3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNAH8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNAI1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNAJA4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNAJC13. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNAJC7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DNTTIP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DOCK10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DOCK11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DOCK4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DPP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DPP4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DPY19L2P2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DSCC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DUX4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DVL3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of DYNC1H1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of DYSF. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ECM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EDEM3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EFCAB3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EFCAB4B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EFNA4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EFTUD2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EGFR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EIF3A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ELA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ELA2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of EMCN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EMD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EML5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ENPP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of EPB41L5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EPHA3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EPHA4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EPHB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EPHB2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EPHB3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EPS15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERBB4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERCC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERCC8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERGIC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERMN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERMP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ERN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ETS2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ETV4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EVC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of EXO1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of EXOC4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of F11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of F13A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of F3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of F5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of F7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of F8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM134A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM13A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM13B1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM13C1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM161A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM176B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of FAM184A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM19A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM20A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of FAM23B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAM65C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FANCA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FANCC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FANCG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FANCM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FANK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FAR2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FBN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FBXO15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FBXO18. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FBXO38. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FCGBP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FECH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FEZ2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FGA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FGD6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FGFR1OP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of FGFR1OP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FGFR2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FGG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FGR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of FIX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FKBP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FLJ35848. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FLJ36070. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FLNA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FNBP1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FOLH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FOXM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FRAS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FUT9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FZD3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of FZD6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GAB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GALC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GALNT3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GAPDH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GART. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GAS2L3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GBA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GBGT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GCG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GCGR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GCK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GFM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GHR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GHV. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GJA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GLA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GLT8D1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GNAS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GNB5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GOLGB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GOLT1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GOLT1B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GPATCH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GPR158. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GPR160. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRAMD3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRHPR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRIA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRIA3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRIA4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRIN2B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRM3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRM4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GRN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of GSDMB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GSTCD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GSTO2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of GTPBP4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HADHA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HBA2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HBB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HCK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HDAC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HDAC5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HDX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of HEPACAM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HERC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HEXA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HEXB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HIPK3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HLA-DPB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HLA-G. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HLCS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HLTF. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HMBS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HMGCL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HNF1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HNRNPH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HP1BP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HPGD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HPRT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HPRT2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HSF2BP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HSF4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HSPA9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HSPG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HTT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of HXA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ICA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IDH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IDS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IFI44L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IKBKAP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IL1R2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IL5RA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IL7RA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IMMT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of INPP5D. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of INSR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of INTS3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of INTU. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IPO4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of IPO8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IQGAP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ISL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ITFG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ITGAL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ITGB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ITGB2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ITGB3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ITGB4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ITIH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ITPR2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of IWS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of JAG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of JAK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of JAK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of JMJD1C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KALRN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KATNAL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KCNN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KCNT2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA0256. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA0528. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA0564. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA0586. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA1033. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA1166. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of KIAA1219. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA1409. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIAA1622. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of KIAA1787. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIF15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIF16B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIF3B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIF5A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIF5B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIF9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIR2DL5B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIR3DL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KIR3DL3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KLF12. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KLF3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KLHL20. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KLK12. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KLKB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KPNA5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KRAS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of KREMEN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KRIT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KRT5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of KRTCAP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of L1CAM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of L3MBTL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of L3MBTL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of LACE1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LAMA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LAMA2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LAMA3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LAMB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LARP7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LDLR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LENG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LGALS3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LGMN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LHCGR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LHX6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LIMCH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LIMK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LMBRD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LMBRD2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LMLN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LMNA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of LMO2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LOC389634. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LOC390110. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LPA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LPCAT2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LPL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LRP4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LRPPRC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LRRC19. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LRRC42. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LRRK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LRWD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LUM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LVRN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LYN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of LYST. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MADD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MAGI1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MAGT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MALT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MAP2K1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MAP4K4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MAPK8IP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MAPK9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MAPT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MATN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MCF2L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MCM6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MDGA2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MEGF10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MEGF11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MEMO1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MET. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MGAM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MGAT4A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MGAT5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MGC16169. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MGC34774. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MIB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MIER2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MKKS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MKL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MLANA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MLH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MLL5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MLX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MME. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MPDZ. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MPI. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MRAP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MRPL11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MRPL39. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MRPS28. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MRPS35. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MS4A13. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MSH2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MSMB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MST1R. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MTDH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MTF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MTHFR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MTIF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MUC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MUT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MVK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYCBP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYH2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of MYO19. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYO3A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYO9B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYOM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of MYOM3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NAG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NARG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NARG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NCOA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NDC80. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NDFIP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NEB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NEDD4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NEK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of NEK11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NEK5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of NFE2L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NFIA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NFIX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NFKBIL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NFRKB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NKAIN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NKAP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NLRC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NLRC5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NLRP13. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NLRP7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NLRP8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NME7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NOL10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NOS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NOS2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NOTCH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NPM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of NR1H4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NR4A3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NRXN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NSMAF. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NSMCE2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NT5C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NT5C3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUBP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUBPL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUDT5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUMA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUP160. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUP88. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUP98. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of NUPL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OAT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OBFC2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OBFC2B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OLIG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OPA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OPN4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OPTN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OSBPL11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OSBPL8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OSGEPL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OTC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of OXT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PADI4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PAH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PAN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PAPOLG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PARD3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PARVB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PAWR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PBGD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PBRM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PCBP4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PCCA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PCNX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PCOTH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PDCD4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDE10A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDE8B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDIA3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDK4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDLIM5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDS5A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDS5B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDXK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PDZRN3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PELI2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PGK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PGM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PHACTR4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PHEX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PHKB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PHLDB2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PHTF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIAS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIGF. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIGN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIGT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIK3C2G. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIK3CG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIK3R1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIP5K1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PITRM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PIWIL3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PKD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PKD2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PKHD1L1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PKIB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PKLR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PKM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PKM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLCB1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PLCB4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLCG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLEKHA5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLEKHA7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLEKHM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLKR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PLXNC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PMFBP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of POLN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of POLR3D. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of POMT2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of POSTN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PPFIA2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PPP1R12A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PPP3CB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PPP4C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PPP4R1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PPP4R2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRAME. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRDM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRIM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRIM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRKAR1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRKCA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRKG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRMT7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PROC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PROCR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRODH. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PROSC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PROX1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRPF40B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRPF4B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRRG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PRUNE2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PSD3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PSEN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PSMAL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTCH1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTEN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PTK2B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPN11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPN22. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPN3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPN4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPRD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPRK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPRM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPRN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PTPRT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PUS10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of PVRL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of PYGM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of QRSL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RAB11FIP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RAB23. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RALBP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RALGDS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RB1CC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RBL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RBM39. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RBM45. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of REC8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RFC4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RFT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of RFTN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RHPN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RIF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RLN3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of RMND5B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RNF11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RNF32. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RNFT1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RNGTT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ROCK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ROCK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RP11-265F1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RP13-36C9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RP6KA3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RPAP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RPGR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RPN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RPS6KA6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RRM1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RRP1B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RSK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of RTEL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RTF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RUFY1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of RYR3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SAAL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SAE1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SBCAD. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN11A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN3A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN4A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN5A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCN8A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCNA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SCO1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SCYL3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SDK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SDK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SEC24A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SEC24D. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SEC31A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SEL1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SENP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SENP6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SENP7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SERPINA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SETD3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SETD4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SEZ6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SFRS12. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SGCE. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SGOL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SGPL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SH2D1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SH3BGRL2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SH3PXD2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SH3PXD2B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SH3RF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SH3TC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SIPA1L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SIPA1L3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SIVA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SKAP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SKIV2L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC12A3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC13A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SLC22A17. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC25A14. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC28A3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SLC38A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC38A4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC39A10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC4A2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC6A11. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC6A13. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC6A6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SLC6A8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SMARCA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SMARCA5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SMC5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SMN2. For example, the SMSM compounds and methods of their use described herein can modulate splicing of exon 7 of a pre-mRNA of SMN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SMTN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SNCAIP. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SNRK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SNRP70. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SNX6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SOD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPAG9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPATA13. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SPATA4. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPATS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPECC1L. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPINK5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SPTA1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SRP72. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SSX3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SSX5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SSX9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of STAG1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of STAMBPL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of STARD6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of STAT6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of STK17B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of STX3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of STXBP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SUCLG2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SULF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SUPT16H. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SUPT6H. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SV2C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SYCP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SYCP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of SYT6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of SYTL5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TAF2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBC1D26. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBC1D29. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBC1D3G. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBC1D8B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBCEL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TBPL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TCEB3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TCF12. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of TCP11L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TDRD3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TEAD1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TECTB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TEK. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TET2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TFRC. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TGM7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TGS1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of THOC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TIAL1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TIAM2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TIMM50. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TLK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of TM4SF20. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TM6SF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TMEM156. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TMEM194A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TMEM27. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TMEM77. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TMF1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TMPRSS6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TNFRSF10A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TNFRSF10B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of TNFRSF8. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TNK2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TNKS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TNKS2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TOM1L1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TOM1L2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TOP2B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TP53. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TP53BP2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TP53I3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TP53INP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TP63. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRAF3IP3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRAPPC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of TRIM44. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRIM65. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRIML1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRIML2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRPM3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRPM5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TRPM7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TSC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TSC2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TSHB. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TSPAN7. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TTC17. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TTLL5. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TTLL9. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TTN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TTPAL. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TTR. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of TUSC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of TXNDC10. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UBE3A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UCK1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UGT1A1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UHRF1BP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UNC45B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UNC5C. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of USH2A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of USP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of USP38. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of USP39. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of USP6. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UTP15. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UTP18. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UTP20. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of UTRN. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UTX. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UTY. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UVRAG. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of UXT. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of VAPA. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of VPS29. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of VPS35. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of VPS39. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of VTI1A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of VTI1B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of VWA3B. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDFY2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDR16. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDR17. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDR26. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDR44. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDR67. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WDTC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WRNIP1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of WWC3. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of XRN1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of XRN2. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of XX-FW88277. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of YARS. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of YGM. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre- mRNA of ZBTB20. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZC3H7A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZC3HAV1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZC3HC1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZFYVE1. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZNF114. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZNF169. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZNF326. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZNF365. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZNF37A. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZNF618. In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing of a pre-mRNA of ZWINT.

[0423] In some embodiments, the SMSM compounds and methods of their use described herein can modulate splicing, such as alternative splicing of a polynucleotide encoded by MAPT gene. In some embodiments, alternative splicing of the MAPT pre-mRNA may lead to the expression of 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 isoforms of the tau protein. In some embodiments, alternative splicing of the MAPT pre-mRNA may lead to the expression of 6 isoforms of the tau protein. In some embodiments, the 6 isoforms of tau include 3 four-repeat (4R) isoforms and 3 three-repeat (3R) isoforms of the tau protein. In the 3R tau isoforms exon 10 is excluded from the splice variants. For example, a 3R tau isoform in which exon 10 is excluded may include exon 2 and/or exon 3. In the 4R tau isoforms exon 10 is included in the splice variants. For example, a 4R tau isoform in which exon 10 is included may include exon 2 and/or exon 3. The inclusion or exclusion of exon 10 may depend on alternative splicing events in a stem loop occurring at the exon 10 intron 10 junction. In some embodiments, a mutation occurring at the 5’ss results in inclusion of exon 10 in an mRNA encoding the tau protein. In some embodiments, a mutation in an ISS region of the stem loop results in exclusion of exon 10 from the mRNA encoding the tau protein. In some embodiments, a mutation at the 5’ss destabilizes the stem loop, thereby decreasing exon 10 inclusion in the mRNA of tau. In some embodiments, a mutation at the 5’ss inhibits binding of a spliceosome component to the pre-mRNA, thereby decreasing exon 10 inclusion in the mRNA of tau. In some embodiments, a mutation at the ISS region of the stem loop inhibits binding of a spliceosome component to the pre-mRNA, thereby increasing exon 10 inclusion in the mRNA of tau.

[0424] The ratio of 3R to 4R tau isoforms may contribute to a number of conditions or diseases. In some embodiments, a subject without a condition or disease has a 3R to 4R ratio of 1:1. In some embodiments, a subject with a condition or disease described herein has a 3R to 4R ratio of about 1:1.2, 1:1.4, 1:1.6, 1:1.8, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5. In some embodiments, a subject with a condition or disease described herein has a 3R to 4R ratio from about 1:1 to about 1:1.1, about 1:1 to about 1:1.2, about 1:1 to about 1:1.3, about 1:1 to about 1:1.4, about 1:1 to about 1:1.5, about 1:1 to about 1:1.6, about 1:1 to about 1:1.8, about 1:1 to about 1:2, about 1:1 to about 1:3, about 1:1 to about 1:3.5, about 1:1 to about 1:4, about 1:1 to about 1:4.5, about 1:1 to about 1:5, 1:2 to about 1:3, about 1:2 to about 1:4, about 1:2 to about 1:5, about 1:3 to about 1:4, about 1:3 to about 1:5, or about 1:4 to about 1:5. In some embodiments, a subject with a condition or disease described herein has a 4R to 3R ratio of about 1:1.2, 1:1.4, 1:1.6, 1:1.8, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5 or 1:5. In some embodiments, a subject with a condition or disease described herein has a 4R to 3R ratio from about 1:1 to about 1:1.1, about 1:1 to about 1:1.2, about 1:1 to about 1:1.3, about 1:1 to about 1:1.4, about 1:1 to about 1:1.5, about 1:1 to about 1:1.6, about 1:1 to about 1:1.8, about 1:1 to about 1:2, about 1:1 to about 1:3, about 1:1 to about 1:3.5, about 1:1 to about 1:4, about 1:1 to about 1:4.5, about 1:1 to about 1:5, 1:2 to about 1:3, about 1:2 to about 1:4, about 1:2 to about 1:5, about 1:3 to about 1:4, about 1:3 to about 1:5, or about 1:4 to about 1:5.

[0425] In some aspects, the SMSM compounds are used to modulate alternative splicing of tau pre-mRNA. In some embodiments, the SMSM compound binds to the stem loop of exon 10 of the tau pre-mRNA, reducing binding affinity of a spliceosome component to the 5’ss, thereby increasing exclusion of exon 10 in the mRNA of tau and increasing the ratio of 3R:4R tau isoforms. In some embodiments, the SMSM compound binds to the stem loop of exon 10 of the tau pre-mRNA, increasing binding affinity of a spliceosome component to the 5’ss, thereby increasing inclusion of exon 10 in the mRNA of tau and decreasing the ratio of 3R:4R tau isoforms. In some embodiments, the SMSM compound binds to the stem loop of exon 10 of the tau pre-mRNA, reducing binding affinity of a spliceosome component to the ISS region, thereby increasing inclusion of exon 10 in the mRNA of tau and decreasing the ratio of 3R:4R tau isoforms. In some embodiments, the SMSM compound binds to the stem loop of exon 10 of the tau pre-mRNA, increasing binding affinity of a spliceosome component to the ISS region, thereby reducing inclusion of exon 10 in the mRNA of tau and increasing the ratio of 3R:4R tau isoforms. In some embodiments, the SMSM compound restores the ratio of 3R:4R to 1:1. In some embodiments, the SMSM compound alters the ratio from 3R>4R to 4R>3R. In some embodiments, the SMSM compound alters the ratio from 3R<4R to 4R<3R. In some embodiments, the SMSM compound binds to the stem loop of exon 10 of the tau pre-mRNA, increasing the thermodynamic stability of the stem loop, thereby reducing inclusion of exon 10 in the mRNA of tau and increasing the ratio of 3R:4R tau isoforms. In some embodiments, the SMSM compound binds to the stem loop of exon 10 of the tau pre-mRNA, decreasing the thermodynamic stability of the stem loop, thereby increasing inclusion of exon 10 in the mRNA of tau and decreasing the ratio of 3R:4R tau isoforms.

[0426] Mutations and/or aberrant secondary or tertiary RNA structures in cis-acting elements of splicing can alter splicing patterns. Mutations and/or aberrant secondary or tertiary RNA structures can be found in core consensus sequences, including 5’ss, 3’ss, and BP regions, or other regulatory elements, including ESEs, ESSs, ISEs, and ISSs. Mutations in cis-acting elements can result in multiple diseases. Exemplary diseases are described below. The present disclosure provides splice modulating compounds and methods that target pre- mRNA containing one or more mutations and/or aberrant secondary or tertiary RNA structures in cis-acting elements. In some embodiments, the present disclosure provides methods and small molecule binding agents that target pre-mRNA containing one or more mutations and/or aberrant secondary or tertiary RNA structures in splice sites or BP regions. In some embodiments, the present disclosure provides methods and small molecule binding agents that target pre-mRNA containing one or more mutations and/or aberrant secondary or tertiary RNA structures in other regulatory elements, for example, ESEs, ESSs, ISEs, and ISSs.

[0427] In some embodiments, splicing at a splice site sequence of a polynucleotide of primary cells is modulated. In some embodiments, splicing at a splice site sequence of a polynucleotide of cells of a tumor is modulated. In some embodiments, the SMSM modulates splicing at a cryptic splice site sequence. In some embodiments, an SMSM modulates splicing of splice site of a polynucleotide. In some embodiments, wherein the polynucleotide is transcribed from the gene. In some embodiments, SMSM modulates exon inclusion in the polynucleotide and splicing of the splice site sequence. In some embodiments, the SMSM modulates pseudoexons inclusion in the polynucleotide and splicing of the splice site sequence. In some embodiments, the SMSM modulates splicing at a cryptic splice site sequence of a polynucleotide.

[0428] In some embodiments, an SMSM modulates splicing by preventing, inhibiting or reducing splicing of the polynucleotide. In some embodiments, an SMSM modulates splicing by preventing, inhibiting or reducing splicing at the splice site sequence. In some embodiments, an SMSM decreases affinity of a splicing complex component to the polynucleotide. In some embodiments, an SMSM decreases affinity of a splicing complex component to the polynucleotide at the splice site sequence, upstream of the splice site sequence or downstream of the splice site sequence. In some embodiments, an SMSM inhibits or reduces a rate of catalysis of splicing of the polynucleotide. In some embodiments, an SMSM inhibits or reduces a rate of catalysis of splicing of the polynucleotide at the splice site sequence. In some embodiments, an SMSM increases steric hindrance between a splicing complex component and the polynucleotide. In some embodiments, an SMSM increases steric hindrance between a splicing complex component and the polynucleotide at the splice site sequence, upstream of the splice site sequence or downstream of the splice site sequence. In some embodiments, an SMSM increases steric hindrance between a first splicing complex component and a second splicing complex component. In some embodiments, an SMSM prevents, inhibits, disrupts or reduces binding of a first splicing complex component and a second splicing complex component.

[0429] In some embodiments, an SMSM decreases affinity of a first splicing complex component to a second splicing complex component. In some embodiments, an SMSM prevents, inhibits, disrupts or reduces binding of a splicing complex component to the polynucleotide. In some embodiments, an SMSM prevents, inhibits, disrupts or reduces binding of a splicing complex component to the polynucleotide at the splice site sequence, upstream of the splice site sequence or downstream of the splice site sequence.

[0430] In some embodiments, an SMSM modulates splicing by promoting or increasing splicing of the polynucleotide. In some embodiments, an SMSM modulates splicing by promoting or increasing splicing the splice site sequence. In some embodiments, an SMSM increases affinity of a splicing complex component to the polynucleotide. In some embodiments, an SMSM increases affinity of a splicing complex component to the polynucleotide at the splice site sequence, upstream of the splice site sequence or downstream of the splice site sequence. In some embodiments, an SMSM increases a rate of catalysis of splicing of the polynucleotide. In some embodiments, an SMSM increases a rate of catalysis of splicing of the polynucleotide at the splice site sequence. In some embodiments, an SMSM decreases or reduces steric hindrance between a splicing complex component and the polynucleotide. In some embodiments, an SMSM decreases steric hindrance between a splicing complex component and the polynucleotide at the splice site sequence, 1-1000 nucleobases bases upstream of the splice site sequence or 1-1000 nucleobases downstream of the splice site sequence. In some embodiments, an SMSM decreases or reduces steric hindrance between a first splicing complex component and a second splicing complex component. In some embodiments, an SMSM promotes or increases binding of a first splicing complex component and a second splicing complex component. In some embodiments, an SMSM increases affinity of a first splicing complex component to a second splicing complex component. In some embodiments, an SMSM promotes or increases binding of a splicing complex component to the polynucleotide. In some embodiments, an SMSM promotes or increases binding of a splicing complex component to the polynucleotide at the splice site sequence, 1-1000 nucleobases upstream of the splice site sequence or 1-1000 nucleobases downstream of the splice site sequence. In some embodiments, an SMSM binds to a splicing complex component, the polynucleotide, or a combination thereof. In some embodiments, an SMSM binds to the polynucleotide at the splice site sequence, 1-1000 nucleobases upstream of the splice site sequence or 1- 1000 nucleobases downstream of the splice site sequence. In some embodiments, an SMSM structurally modulates a splicing complex component, the polynucleotide, or both. In some embodiments, an SMSM promotes or increases steric hindrance, steric shielding, steric attraction, chain crossing, steric repulsions, steric inhibition of resonance, steric inhibition of protonation, or a combination thereof of the polynucleotide, a splicing complex component or a combination thereof. In some embodiments, binding of an SMSM to a polynucleotide or a splicing complex component decreases conformational stability of a splice site sequence. In some embodiments, binding of an SMSM to a polynucleotide increases conformational stability of a splice site sequence.

[0431] In some embodiments, an SMSM modulates exon skipping of a target polynucleotide, such as a pre- mRNA. For example, an SMSM can inhibit exon skipping of a target polynucleotide, such as a pre-mRNA. For example, an SMSM can promote exon skipping of a target polynucleotide, such as a pre-mRNA. In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with exon skipping of the polynucleotide, such as a pre-mRNA. In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with aberrant exon skipping of the polynucleotide, such as a pre-mRNA.

[0432] In some embodiments, an SMSM modulates exon inclusion of a target polynucleotide, such as a pre- mRNA. For example, an SMSM can inhibit exon inclusion of a target polynucleotide, such as a pre-mRNA. For example, an SMSM can promote exon inclusion of a target polynucleotide, such as a pre-mRNA. In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with exon inclusion of the polynucleotide, such as a pre-mRNA. In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with aberrant exon inclusion of the polynucleotide, such as a pre-mRNA.

[0433] In some embodiments, an SMSM modulates nonsense mediated degradation (NMD) of a target polynucleotide, such as a pre-mRNA. For example, an SMSM can inhibit nonsense mediated degradation (NMD) of a target polynucleotide, such as a pre-mRNA or an mRNA. In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with NMD of the polynucleotide, such as a pre-mRNA or an mRNA.

[0434] In some embodiments, an SMSM modulates intron inclusion of a target polynucleotide. For example, an SMSM can inhibit intron inclusion of a target polynucleotide, such as a pre-mRNA. For example, an SMSM can promote intron inclusion of a target polynucleotide, such as a pre-mRNA. In some embodiments, an SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with intron inclusion of the polynucleotide. In some embodiments, the SMSM modulates splicing at a splice site sequence of a polynucleotide in a cell of a subject with a disease or condition associated with intron inclusion of the polynucleotide.

[0435] In some embodiments, an SMSM modulates splicing at splice site sequence of a polynucleotide, such as a pre-mRNA, wherein the splice site sequence comprises a sequence selected from the group consisting of NGAgunvrn, NHAdddddn, NNBnnnnnn, and NHAddmhvk; wherein N or n is A, U, G or C; B is C, G, or U; H or h is A, C, or U; d is a, g, or u; m is a or c; r is a or g; v is a, c or g; k is g or u.

[0436] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence NNBgunnnn, NNBhunnnn, or NNBgvnnnn. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence NNBgurrrn, NNBguwwdn, NNBguvmvn, NNBguvbbn, NNBgukddn, NNBgubnbd, NNBhunngn, NNBhurmhd, or NNBgvdnvn; wherein N or n is A, U, G or C; B is C, G, or U; H or h is A, C, or U; d is a, g, or u; m is a or c; r is a or g; v is a, c or g; k is g or u.

[0437] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence of Table 2A, Table 2B, Table 2C or Table 2D. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence AAAauaagu, AAAguaagua, AAAguacau, AAAguaga, AAAguaug, AAAguaugu, AAAgugagug, AAAgugaguu, AACaugagga, AACguaagu, AACgugacu, AACgugauu, AAGaugagc, AAGauuugu, AAGgaugag, AAGgcaaaa, AAGgcaaggg, AAGgcaggga, AAGggaaaa, AAGguaugag, AAGguaaag, AAGguaaau, AAGguaaca, AAGguaacaug, AAGguaacu, AAGguaagcc, AAGguaagcg, AAGguaauaa, AAGguaaugu, AAGguaaugua, AAGguacag, AAGguacgg, AAGguacug, AAGguagacc, AAGguagag, AAGguagcg, AAGguagua, AAGguagug, AAGguauac, AAGguauau, AAGguauauu, AAGguauca, AAGguaucg, AAGguaucu, AAGguauga, AAGguaugg, AAGguaugu, AAGguauuu, AAGgucaag, AAGgucaau, AAGgucucu, AAGgucuggg, AAGgucugu, AAGgugaccuu, AAGgugagau, AAGgugaguc, AAGgugccu, AAGgugggcc, AAGgugggu, AAGguggua, AAGguguau, AAGgugucu, AAGgugugc, AAGgugugu, AAGguguua, AAGguuaag, AAGguuagc, AAGguuagug, AAGguuca, AAGguuuaa, AAGguuuau, AAGguuugg, AAGuuaagg, AAGuuaaua, AAGuuagga, AAUguaaau, AAUguaagc, AAUguaagg, AAUguaauu, AAUguaugu, AAUgugagu, AAUgugugu, ACAguaaau, ACAgugagg, ACAguuagu, ACAguuuga, ACCaugagu, ACCgugaguu, ACGauaagg, ACGcuaagc, ACGguagcu, ACGgugaac, ACGgugagug, ACUguaaau, ACUguaacu, ACUguauu, ACUgugagug, AGAguaag, AGAguaaga, AGAguaagg, AGAguaagu, AGAguagau, AGAguaggu, AGAgugaau, AGAgugagc, AGAgugagu, AGAgugcgu, AGCguaagg, AGCguaagu, AGCguacgu, AGCguaggu, AGCgugagu, AGGguaauga, AGGguagac, AGGguauau, AGGgugaau, AGGgugagg, AGGgugauc, AGGgugcaa, AGGgugucu, AGUguaagc, AGUguaagu, AGUgugagu, AGUgugaguac, AUAgucagu, AUAgugaau, AUCgguaaaa, AUCguuaga, AUGguaaaa, AUGguaacc, AUGguacau, AUGguaugu, AUGguauuu, AUGgucauu, AUGgugacc, AUUuuaagc, CAAGguaccu, CAAguaaac, CAAguaacu, CAAguaagc, CAAguaagg, CAAguaagua, CAAguaau, CAAguaugu, CAAguauuu, CAAgugaaa, CAAgugagu, CACgugagc, CACguuggu, CAGauaacu, CAGaugagg, CAGaugagu, CAGauuggu, CAGcugugu, CAGgcgagu, CAGgcuggu, CAGguaaggc, CAGguaaaa, CAGguaaag, CAGguaaccuc, CAGguaagac, CAGguaagc, CAGguaagu, CAGguaau, CAGguaaugc, CAGguaaugu, CAGguacaa, CAGguacag, CAGguacagu, CAGguaccg, CAGguacug, CAGguagag, CAGguagcaa, CAGguaggagg, CAGguaggc, CAGguagguga, CAGguagua, CAGguagug, CAGguauag, CAGguauau, CAGguaucc, CAGguauga, CAGguaugg, CAGguaugu, CAGguauug, CAGgucaau, CAGgucagug, CAGgucuga, CAGgucugga, CAGgucuggu, CAGgucuuu, CAGgugacu, CAGgugagc, CAGgugaggg, CAGgugagugg, CAGgugaua, CAGgugcac, CAGgugcag, CAGgugcgc, CAGgugcug, CAGguggau, CAGgugggug, CAGgugua, CAGguguag, CAGguguau, CAGguguga, CAGgugugu, CAGguuaag, CAGguugau, CAGguugcu, CAGguuggc, CAGguuguc, CAGguuguu, CAGguuuagu, CAGguuugc, CAGguuugg, CAGuuuggu, CAUggaagac, CAUguaau, CAUguaauu, CAUguaggg, CAUguauuu, CCAguaaac, CCAgugaga, CCGguaacu, CCGgugaau, CCGgugacu, CCGgugagg, CCUauaagu, CCUaugagu, CCUguaaau, CCUguaagc, CCUguaauu, CCUgugaau, CCUgugauu, CGAguccgu, CGCauaagu, CGGguaau, CGGguauau, CGGguaugg, CGGgucauaauc, CGGgugggu, CGGguguau, CGGgugugu, CGUgugaau, CGUgugggu, CUGguauga, CUGgugaau, CUGgugaguc, CUGgugaguuc, CUGgugcau, CUGgugcuu, CUGguguga, CUGguuugu, CUGuuaag, CUGuugaga, GAAggaagu, GAAguaaac, GAAguaaau, GAAgucugg, GAAguggg, GAAgugugu, GAAuaaguu, GACaugagg, GAGaucugg, GAGaugagg, GAGCAGguaagcu, GAGcugcag, GAGgcaggu, GAGgcgugg, GAGgcuccc, GAGguggguuu, GAGguaaag, GAGguaaga, GAGguaagag, GAGguaagcg, GAGguaauac, GAGguaauau, GAGguaaugu, GAGguacaa, GAGguagga, GAGguauau, GAGguauga, GAGguaugg, GAGgucuggu, GAGgugaag, GAGgugagg, GAGgugca, GAGgugccu, GAGgugcggg, GAGgugcug, GAGguguac, GAGguguau, GAGgugugc, GAGgugugu, GAGuuaagu, GAUaugagu, GAUguaaau, GAUguaagu, GAUguaauu, GAUguaua, GAUgugacu, GAUgugagg, GAUgugauu, GCAguaaau, GCAguagga, GCAguuagu, GCGaugagu, GCGgagagu, GCGguaaaa, GCGguaauca, GCGgugacu, GCGgugagca, GCGgugagcu, GCGguggga, GCGguuagu, GCUguaaau, GCUguaacu, GCUguaauu, GGAguaag, GGAguaagg, GGAguaagu, GGAguaggu, GGAgugagu, GGAguuagu, GGCguaagu, GGCgucagu, GGGauaagu, GGGaugagu, GGGguaagug, GGGguaaau, GGGguaacu, GGGguacau, GGGgugacg, GGGgugagug, GGGgugcau, GGGguuggga, GGUguaagu, GUAgugagu, GUGguaagu, GUGguaagug, GUGgugagc, GUGgugagu, GUGgugauc, GUGguugua, GUUauaagu, GUUCUCAgugug, GUUguaaau, GUUuugguga, uAGCAGguaagca, uGGguaccug, UAGaugcgu, UAGguaaag, UAGguaccc, UAGguaggu, UAGguauau, UAGguauc, UAGguauga, UAGguauug, UAGgucaga, UAGgugcau, UAGguguau, UCAguaaac, UCAguaaau, UCAguaagu, UCAgugauu, UCAgugug, UCCgugaau, UCCgugacu, UCCgugagc, UCUguaaau, UGAgugaau, UGGauaagg, UGGguaaag, UGGguacca, UGGguaugc, UGGguggau, UGGguggggg, UGGgugggug, UGGgugugg, UGGguuagu, UGUgcaagu, UGUguaaau, UGUguacau, UUAguaaau, UUCauaagu, UUGguaaag, UUGguaaca, UUGguacau, UUGguagau, UUGgugaau, UUGgugagc, UUUauaagc or UUUgugagc.

[0438] ABCA4, ABCA9, ABCB1, ABCB5, ABCC9, ABCD1, ACADL, ACADM, ACADSB, ACSS2, ACTG2, ADA, ADAL, ADAM10, ADAM15, ADAM22, ADAM32, ADAMTS12, ADAMTS13, ADAMTS20, ADAMTS6, ADAMTS9, ADCY10, ADCY3, ADCY8, ADRBK2, AFP, AGL, AGT, AHCTF1, AKAP10, AKAP3, AKNA, ALAS1, ALB, ALDH3A2, ALG6, ALS2CL, AMBRA1, ANGPTL3, ANK3, ANTXR2, ANXA10, ANXA11, AP2A2, AP4E1, APC, APOA1, APOB, APOC3, APOH, AR, ARFGEF1, ARFGEF2, ARHGAP1, ARHGAP18, ARHGAP26, ARHGAP8, ARHGEF18, ARHGEF2, ARPC3, ARS2, ASH1L, ASNSD1, ASPM, ATAD5, ATG16L2, ATG4A, ATM, ATP11C, ATP13A5, ATP6V1G3, ATP7A, ATP7B, ATR, ATXN2, ATXN3, B2M, B4GALNT3, BBOX1, BBS4, BCL2–like 11 (BIM), BCS1L, BMP2K, BMPR2, BRCA1, BRCA2, BRCC3, BRSK1, BRSK2, BTAF1, BTK, C10orf137, C11orf30, C11orf65, C11orf70, C12orf51, C13orf1, C13orf15, C14orf101, C14orf118, C15orf29, C15orf42, C15orf60, C16orf33, C16orf38, C16orf48, C18orf8, C19orf42, C1orf107, C1orf114, C1orf130, C1orf149, C1orf27, C1orf71, C1orf87, C1orf94, C1R, C20orf74, C21orf70, C2orf55, C3, C3orf23, C4orf18, C4orf29, C5orf34, C6orf118, C8B, C8orf33, C9orf114, C9orf43, C9orf86, C9orf98, CA11, CAB39, CACHD1, CACNA1B, CACNA1C, CACNA1G, CACNA1H, CACNA2D1, CALCA, CALCOCO2, CAMK1D, CAMKK1, CAPN3, CAPN9, CAPSL, CARKD, CAT, CBX1, CBX3, CCDC102B, CCDC11, CCDC131, CCDC146, CCDC15, CCDC18, CCDC5, CCDC81, CD1B, CD33, CD4, CD46, CDC14A, CDC16, CDC2L5, CDC42BPB, CDCA8, CDH1, CDH10, CDH11, CDH23, CDH24, CDH8, CDH9, CDK5RAP2, CDK6, CDK8, CEL, CELSR3, CENPI, CENTB2, CENTG2, CEP110, CEP170, CEP192, CETP, CFB, CFH, CFTR, CGN, CGNL1, CHAF1A, CHD9, CHIC2, CHL1, CHM, CHN1, CLCN1, CLEC16A, CLIC2, CLINT1, CLK1, CLPB, CLPTM1, CMIP, CMYA5, CNGA3, CNOT1, CNOT7, CNTN6, COG3, COL11A1, COL11A2, COL12A1, COL14A1, COL15A1, COL17A1, COL19A1, COL1A1, COL1A2, COL22A1, COL24A1, COL25A1, COL29A1, COL2A1, COL3A1, COL4A1, COL4A2, COL4A5, COL4A6, COL5A2, COL6A1, COL7A1, COL9A1, COL9A2, COLQ, COMTD1, COPA, COPB2, COPS7B, COPZ2, CPSF2, CPXM2, CR1, CREBBP, CRKRS, CRYZ, CSE1L, CSTB, CSTF3, CT45-6, CUBN, CUL4B, CUL5, CXorf41, CYBB, CYFIP2, CYP17, CYP19, CYP24A1, CYP27A1, CYP3A4, CYP3A43, CYP3A5, CYP4F2, CYP4F3, DAZ2, DCBLD1, DCC, DCTN3, DCUN1D4, DDA1, DDEF1, DDX1, DDX24, DDX4, DENND2D, DEPDC2, DES, DGAT2, DHFR, DHRS7, DHRS9, DIP2A, DMD, DMTF1, DNAH3, DNAH8, DNAI1, DNAJA4, DNAJC13, DNAJC7, DNTTIP2, DOCK10, DOCK11, DOCK4, DPP3, DPP4, DPY19L2P2, DSCC1, DUX4, DVL3, DYNC1H1, DYSF, ECM2, EDEM3, EFCAB3, EFCAB4B, EFNA4, EFTUD2, EGFR, EIF3A, ELA1, ELA2A, EMCN, EMD, EML5, ENPP3, EPB41L5, EPHA3, EPHA4, EPHB1, EPHB2, EPHB3, EPS15, ERBB4, ERCC1, ERCC8, ERGIC3, ERMN, ERMP1, ERN1, ERN2, ETS2, ETV4, EVC2, EXO1, EXOC4, F11, F13A1, F3, F5, F7, F8, FAH, FAM134A, FAM13A1, FAM13B1, FAM13C1, FAM161A, FAM176B, FAM184A, FAM19A1, FAM20A, FAM23B, FAM65C, FANCA, FANCC, FANCG, FANCM, FANK1, FAR2, FBN1, FBXO15, FBXO18, FBXO38, FCGBP, FECH, FEZ2, FGA, FGD6, FGFR1OP, FGFR1OP2, FGFR2, FGG, FGR, FIX, FKBP3, FLJ35848, FLJ36070, FLNA, FN1, FNBP1L, FOLH1, FOXM1, FRAS1, FUT9, FZD3, FZD6, GAB1, GALC, GALNT3, GAPDH, GART, GAS2L3, GBA, GBGT1, GCG, GCGR, GCK, GFM1, GH1, GHR, GHV, GJA1, GLA, GLT8D1, GNAS, GNB5, GOLGB1, GOLT1A, GOLT1B, GPATCH1, GPR158, GPR160, GRAMD3, GRHPR, GRIA1, GRIA3, GRIA4, GRIN2B, GRM3, GRM4, GRN, GSDMB, GSTCD, GSTO2, GTPBP4, HADHA, HBA2, HBB, HCK, HDAC3, HDAC5, HDX, HEPACAM2, HERC1, HEXA, HEXB, HIPK3, HLA-DPB1, HLA-G, HLCS, HLTF, HMBS, HMGCL, HNF1A, HNRNPH1, HP1BP3, HPGD, HPRT1, HPRT2, HSF2BP, HSF4, HSPA9, HSPG2, HTT, HXA, ICA1, IDH1, IDS, IFI44L, IKBKAP, IL1R2, IL5RA, IL7RA, IMMT, INPP5D, INSR, INTS3, INTU, IPO4, IPO8, IQGAP2, ISL2, ITFG1, ITGAL, ITGB1, ITGB2, ITGB3, ITGB4, ITIH1, ITPR2, IWS1, JAG1, JAK1, JAK2, JMJD1C, KALRN, KATNAL2, KCNN2, KCNT2, KIAA0256, KIAA0528, KIAA0564, KIAA0586, KIAA1033, KIAA1166, KIAA1219, KIAA1409, KIAA1622, KIAA1787, KIF15, KIF16B, KIF3B, KIF5A, KIF5B, KIF9, KIN, KIR2DL5B, KIR3DL2, KIR3DL3, KLF12, KLF3, KLHL20, KLK12, KLKB1, KPNA5, KRAS, KREMEN1, KRIT1, KRT5, KRTCAP2, L1CAM, L3MBTL, L3MBTL2, LACE1, LAMA1, LAMA2, LAMA3, LAMB1, LARP7, LDLR, LENG1, LGALS3, LGMN, LHCGR, LHX6, LIMCH1, LIMK2, LMBRD1, LMBRD2, LMLN, LMNA, LMO2, LOC389634, LOC390110, LPA, LPCAT2, LPL, LRP4, LRPPRC, LRRC19, LRRC42, LRRK2, LRWD1, LUM, LVRN, LYN, LYST, MADD, MAGI1, MAGT1, MALT1, MAP2K1, MAP4K4, MAPK8IP3, MAPK9, MAPT, MATN2, MCF2L2, MCM6, MDGA2, MEGF10, MEGF11, MEMO1, MET, MGAM, MGAT4A, MGAT5, MGC16169, MGC34774, MIB1, MIER2, MKKS, MKL2, MLANA, MLH1, MLL5, MLX, MME, MPDZ, MPI, MRAP2, MRPL11, MRPL39, MRPS28, MRPS35, MS4A13, MSH2, MSMB, MST1R, MTDH, MTF2, MTHFR, MTIF2, MUC2, MUT, MVK, MYB, MYCBP2, MYH2, MYO19, MYO3A, MYO9B, MYOM2, MYOM3, NAG, NARG1, NARG2, NCOA1, NDC80, NDFIP2, NEB, NEDD4, NEK1, NEK11, NEK5, NF1, NF2, NFE2L2, NFIA, NFIX, NFKBIL2, NFRKB, NKAIN2, NKAP, NLRC3, NLRC5, NLRP13, NLRP7, NLRP8, NME7, NOL10, NOS1, NOS2A, NOTCH1, NPM1, NR1H4, NR4A3, NRXN1, NSMAF, NSMCE2, NT5C, NT5C3, NUBP1, NUBPL, NUDT5, NUMA1, NUP160, NUP88, NUP98, NUPL1, OAT, OBFC2A, OBFC2B, OLIG2, OPA1, OPN4, OPTN, OSBPL11, OSBPL8, OSGEPL1, OTC, OXT, PADI4, PAH, PAN2, PAPOLG, PARD3, PARVB, PAWR, PBGD, PBRM1, PCBP4, PCCA, PCNX, PCOTH, PDCD4, PDE10A, PDE8B, PDH1, PDIA3, PDK4, PDLIM5, PDS5A, PDS5B, PDXK, PDZRN3, PELI2, PGK1, PGM2, PHACTR4, PHEX, PHKB, PHLDB2, PHTF1, PIAS1, PIGF, PIGN, PIGT, PIK3C2G, PIK3CG, PIK3R1, PIP5K1A, PITRM1, PIWIL3, PKD1, PKD2, PKHD1L1, PKIB, PKLR, PKM1, PKM2, PLCB1, PLCB4, PLCG1, PLD1, PLEKHA5, PLEKHA7, PLEKHM1, PLKR, PLXNC1, PMFBP1, POLN, POLR3D, POMT2, POSTN, PPFIA2, PPP1R12A, PPP3CB, PPP4C, PPP4R1L, PPP4R2, PRAME, PRC1, PRDM1, PRIM1, PRIM2, PRKAR1A, PRKCA, PRKG1, PRMT7, PROC, PROCR, PRODH, PROSC, PROX1, PRPF40B, PRPF4B, PRRG2, PRUNE2, PSD3, PSEN1, PSMAL, PTCH1, PTEN, PTK2, PTK2B, PTPN11, PTPN22, PTPN3, PTPN4, PTPRD, PTPRK, PTPRM, PTPRN2, PTPRT, PUS10, PVRL2, PYGM, QRSL1, RAB11FIP2, RAB23, RALBP1, RALGDS, RB1CC1, RBL2, RBM39, RBM45, REC8, RFC4, RFT1, RFTN1, RHPN2, RIF1, RLN3, RMND5B, RNF11, RNF32, RNFT1, RNGTT, ROCK1, ROCK2, RP1, RP11-265F1, RP13-36C9, RP6KA3, RPAP3, RPGR, RPN1, RPS6KA6, RRM1, RRP1B, RSK2, RTEL1, RTF1, RUFY1, RYR3, SAAL1, SAE1, SBCAD, SCN11A, SCN1A, SCN2A, SCN3A, SCN4A, SCN5A, SCN8A, SCNA, SCO1, SCYL3, SDK1, SDK2, SEC24A, SEC24D, SEC31A, SEL1L, SENP3, SENP6, SENP7, SERPINA1, SETD3, SETD4, SEZ6, SFRS12, SGCE, SGOL2, SGPL1, SH2D1A, SH3BGRL2, SH3PXD2A, SH3PXD2B, SH3RF2, SH3TC2, SIPA1L2, SIPA1L3, SIVA1, SKAP1, SKIV2L2, SLC12A3, SLC13A1, SLC22A17, SLC25A14, SLC28A3, SLC38A1, SLC38A4, SLC39A10, SLC4A2, SLC6A11, SLC6A13, SLC6A6, SLC6A8, SMARCA1, SMARCA5, SMC5, SMN2, SMTN, SNCAIP, SNRK, SNRP70, SNX6, SOD1, SPAG9, SPATA13, SPATA4, SPATS1, SPECC1L, SPINK5, SPP2, SPTA1, SRP72, SSX3, SSX5, SSX9, STAG1, STAMBPL1, STARD6, STAT6, STK17B, STX3, STXBP1, SUCLG2, SULF2, SUPT16H, SUPT6H, SV2C, SYCP1, SYCP2, SYT6, SYTL5, TAF2, TBC1D26, TBC1D29, TBC1D3G, TBC1D8B, TBCEL, TBK1, TBPL1, TCEB3, TCF12, TCP11L2, TDRD3, TEAD1, TECTB, TEK, TET2, TFRC, TG, TGM7, TGS1, THOC2, TIAL1, TIAM2, TIMM50, TLK2, TM4SF20, TM6SF1, TMEM156, TMEM194A, TMEM27, TMEM77, TMF1, TMPRSS6, TNFRSF10A, TNFRSF10B, TNFRSF8, TNK2, TNKS, TNKS2, TOM1L1, TOM1L2, TOP2B, TP53, TP53BP2, TP53I3, TP53INP1, TP63, TRAF3IP3, TRAPPC2, TRIM44, TRIM65, TRIML1, TRIML2, TRPM3, TRPM5, TRPM7, TSC1, TSC2, TSHB, TSPAN7, TTC17, TTLL5, TTLL9, TTN, TTPAL, TTR, TUSC3, TXNDC10, UBE3A, UCK1, UGT1A1, UHRF1BP1, UNC45B, UNC5C, USH2A, USP1, USP38, USP39, USP6, UTP15, UTP18, UTP20, UTRN, UTX, UTY, UVRAG, UXT, VAPA, VPS29, VPS35, VPS39, VTI1A, VTI1B, VWA3B, WDFY2, WDR16, WDR17, WDR26, WDR44, WDR67, WDTC1, WRNIP1, WWC3, XRN1, XRN2, XX- FW88277, YARS, YGM, ZBTB20, ZC3H7A, ZC3HAV1, ZC3HC1, ZFYVE1, ZNF114, ZNF169, ZNF326, ZNF365, ZNF37A, ZNF618 or a ZWINT

[0439] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence of Table 2A. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence AAAauaagu, AAAguaagua, AAAguacau, AAAguaga, AAAguaug, AAAguaugu, AAAgugagug, AAAgugaguu, AACaugagga, AACguaagu, AACgugacu, AACgugauu, AAGaugagc, AAGauuugu, AAGgaugag, AAGgcaaaa, AAGgcaaggg, AAGgcaggga, AAGggaaaa, AAGgtatgag, AAGguaaag, AAGguaaau, AAGguaaca, AAGguaacaug, AAGguaacu, AAGguaagcc, AAGguaagcg, AAGguaauaa, AAGguaaugu, AAGguaaugua, AAGguacag, AAGguacgg, AAGguacug, AAGguagacc, AAGguagag, AAGguagcg, AAGguagua, AAGguagug, AAGguauac, AAGguauau, AAGguauauu, AAGguauca, AAGguaucg, AAGguaucu, AAGguauga, AAGguaugg, AAGguaugu, AAGguauuu, AAGgucaag, AAGgucaau, AAGgucucu, AAGgucuggg, AAGgucugu, AAGgugaccuu, AAGgugagau, AAGgugaguc, AAGgugccu, AAGgugggcc, AAGgugggu, AAGguggua, AAGguguau, AAGgugucu, AAGgugugc, AAGgugugu, AAGguguua, AAGguuaag, AAGguuagc, AAGguuagug, AAGguuca, AAGguuuaa, AAGguuuau, AAGguuugg, AAGuuaagg, AAGuuaaua, AAGuuagga, AAUguaaau, AAUguaagc, AAUguaagg, AAUguaauu, AAUguaugu, AAUgugagu, AAUgugugu, ACAguaaau, ACAgugagg, ACAguuagu, ACAguuuga, ACCaugagu, ACCgugaguu, ACGauaagg, ACGcuaagc, ACGguagcu, ACGgugaac, ACGgugagug, ACUguaaau, ACUguaacu, ACUguauu, ACUgugagug, AGAguaaga, AGAguaagg, AGAguaagu, AGAguagau, AGAguaggu, AGAgugaau, AGAgugagc, AGAgugagu, AGAgugcgu, AGCguaagg, AGCguaagu, AGCguacgu, AGCguaggu, AGCgugagu, AGGguaauga, AGGguagac, AGGguauau, AGGgugaau, AGGgugagg, AGGgugauc, AGGgugcaa, AGGgugucu, AGUguaagc, AGUguaagu, AGUgugagu, AGUgugaguac, AUAgucagu, AUAgugaau, AUCgguaaaa, AUCguuaga, AUGguaaaa, AUGguaacc, AUGguacau, AUGguaugu, AUGguauuu, AUGgucauu, AUGgugacc, AUUuuaagc, CAAGguaccu, CAAguaaac, CAAguaacu, CAAguaagc, CAAguaagg, CAAguaagua, CAAguaau, CAAguaugu, CAAguauuu, CAAgugaaa, CAAgugagu, CACgugagc, CACguuggu, CAGauaacu, CAGaugagg, CAGauuggu, CAGcugugu, CAGgcuggu, CAGgtaaggc, CAGguaaaa, CAGguaaag, CAGguaaccuc, CAGguaagac, CAGguaagc, CAGguaagu, CAGguaau, CAGguaaugc, CAGguaaugu, CAGguacaa, CAGguacag, CAGguacagu, CAGguaccg, CAGguacug, CAGguagag, CAGguagcaa, CAGguaggagg, CAGguaggc, CAGguagguga, CAGguagua, CAGguagug, CAGguauag, CAGguauau, CAGguaucc, CAGguauga, CAGguaugg, CAGguaugu, CAGguauug, CAGgucaau, CAGgucagug, CAGgucuga, CAGgucugga, CAGgucuggu, CAGgucuuu, CAGgugagc, CAGgugaggg, CAGgugagugg, CAGgugaua, CAGgugcac, CAGgugcag, CAGgugcgc, CAGgugcug, CAGguggau, CAGgugggug, CAGgugua, CAGguguag, CAGguguau, CAGguguga, CAGgugugu, CAGguuaag, CAGguugau, CAGguugcu, CAGguuggc, CAGguuguc, CAGguuguu, CAGguuuagu, CAGguuugc, CAGguuugg, CAGuuuggu, CAUggaagac, CAUguaau, CAUguaauu, CAUguaggg, CAUguauuu, CCAguaaac, CCAgugaga, CCGguaacu, CCGgugaau, CCGgugacu, CCGgugagg, CCUauaagu, CCUaugagu, CCUguaaau, CCUguaagc, CCUguaauu, CCUgugaau, CCUgugauu, CGAguccgu, CGCauaagu, CGGguaau, CGGguauau, CGGguaugg, CGGgucauaauc, CGGgugggu, CGGguguau, CGGgugugu, CGUgugaau, CGUgugggu, CUGguauga, CUGgugaau, CUGgugaguc, CUGgugaguuc, CUGgugcau, CUGgugcuu, CUGguguga, CUGguuugu, CUGuuaag, CUGuugaga, GAAggaagu, GAAguaaac, GAAguaaau, GAAgucugg, GAAguggg, GAAgugugu, GAAuaaguu, GACaugagg, GAGaucugg, GAGaugagg, GAGCAGguaagcu, GAGcugcag, GAGgcaggu, GAGgcgugg, GAGgcuccc, GAGgtgggttt, GAGguaaag, GAGguaaga, GAGguaagag, GAGguaagcg, GAGguaauac, GAGguaauau, GAGguaaugu, GAGguacaa, GAGguagga, GAGguauau, GAGguauga, GAGguaugg, GAGgucuggu, GAGgugaag, GAGgugagg, GAGgugca, GAGgugccu, GAGgugcggg, GAGgugcug, GAGguguac, GAGguguau, GAGgugugc, GAGgugugu, GAGuuaagu, GAUaugagu, GAUguaaau, GAUguaagu, GAUguaauu, GAUguaua, GAUgugacu, GAUgugagg, GAUgugauu, GCAguaaau, GCAguagga, GCAguuagu, GCGaugagu, GCGgagagu, GCGguaaaa, GCGguaauca, GCGgugacu, GCGgugagca, GCGgugagcu, GCGguggga, GCGguuagu, GCUguaaau, GCUguaacu, GCUguaauu, GGAguaagg, GGAguaagu, GGAguaggu, GGAgugagu, GGAguuagu, GGCguaagu, GGCgucagu, GGGauaagu, GGGaugagu, GGGgtaagtg, GGGguaaau, GGGguaacu, GGGguacau, GGGgugacg, GGGgugagug, GGGgugcau, GGGguuggga, GGUguaagu, GUUCUCAgugug, UCAgugug, GUAgugagu, GUGguaagu, GUGguaagug, GUGgugagc, GUGgugagu, GUGgugauc, GUGguugua, GUUauaagu, GUUguaaau, GUUuugguga, UAGCAGguaagca, TGGgtacctg, UAGaugcgu, UAGguaaag, UAGguaccc, UAGguaggu, UAGguauau, UAGguauc, UAGguauga, UAGguauug, UAGgucaga, UAGgugcau, UAGguguau, UCAguaaac, UCAguaaau, UCAguaagu, UCAgugauu, UCCgugaau, UCCgugacu, UCCgugagc, UCUguaaau, UGAgugaau, UGGauaagg, UGGguaaag, UGGguacca, UGGguaugc, UGGguggau, UGGguggggg, UGGgugggug, UGGgugugg, UGGguuagu, UGUgcaagu, UGUguaaau, UGUguacau, UUAguaaau, UUCauaagu, UUGguaaag, UUGguaaca, UUGguacau, UUGguagau, UUGgugaau, UUGgugagc, UUUauaagc or UUUgugagc.

[0440] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence of Table 2B. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence AAAauaagu, AAGaugagc, AAGauuugu, AAGgaugag, AAGgcaaaa, AAGuuaagg, AAGuuaaua, AAGuuagga, ACCaugagu, ACGauaagg, ACGcuaagc, AGGguauau, AGGgugagg, AGGgugauc, AGGgugucu, AUGgugacc, AUUuuaagc, CAAgugagu, CACgugagc, CACguuggu, CAGauaacu, CAGaugagg, CAGaugagu, CAGauuggu, CAGcugugu, CAGgcgagu, CAGgcuggu, CAGgugacu, CAGguugau, CAGguugcu, CAGguuggc, CAGguuguu, CAGuuuggu, CAUguaggg, CAUguauuu, CCGgugaau, CCUauaagu, CCUaugagu, CCUgugaau, CGCauaagu, CGGguguau, CUGuuaag, CUGuugaga, GAAggaagu, GAAguaaau, GAAgucugg, GAAguggg, GAAgugugu, GAAuaaguu, GACaugagg, GAGaucugg, GAGaugagg, GAGgcaggu, GAGgcgugg, GAGgcuccc, GAGguaaga, GAGguagga, GAGgugagg, GAGuuaagu, GAUaugagu, GAUaugagu, GCAguagga, GCGaugagu, GCGgagagu, GCGgugacu, GCGguuagu, GCUguaacu, GGGaugagu, GUAgugagu, GUGgugagc, GUGgugauc, UAGaugcgu, UGGauaagg, UGGguacca, UGGguggau, UGGgugggug, UGUgcaagu, UUCauaagu, UUGguaaca, UUUauaagc or UUUgugagc.

[0441] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence of Table 2C or Table 2D. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence NGAguaag.

[0442] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence of Table 2C. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence AGAguaag.

[0443] In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence of Table 2D. In some embodiments, an SMSM modulates splicing of a splice site sequence comprising a sequence GGAguaag. Table 2A– Exemplary targets

Methods of Treatment

[0444] The compositions and methods described herein can be used for treating a human disease or disorder associated with aberrant splicing, such as aberrant pre-mRNA splicing. The compositions and methods described herein can be used for treating a human disease or disorder by modulating mRNA, such as pre-mRNA. In some embodiments, the compositions and methods described herein can be used for treating a human disease or disorder by modulating splicing of a nucleic acid even when that nucleic acid is not aberrantly spliced in the pathogenesis of the disease or disorder being treated.

[0445] Provided herein are methods of treating cancer or a non-cancer disease or condition in a mammal in need thereof. The method can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof, to a mammal with a cancer or a non-cancer disease or condition. In some embodiments, the present disclosure relates to the use of an SMSM as described herein for the preparation of a medicament for the treatment, prevention and/or delay of progression of cancer or a non-cancer disease or condition. In some embodiments, the present disclosure relates to the use of a steric modulator as described herein for the treatment, prevention and/or delay of progression of cancer or a non- cancer disease or condition.

[0446] In some embodiments, an effective amount in the context of the administration of an SMSM compound or a pharmaceutically acceptable salt thereof, or composition or medicament thereof refers to an amount of an SMSM compound or a pharmaceutically acceptable salt thereof to a patient which has a therapeutic effect and/or beneficial effect. In certain specific embodiments, an effective amount in the context of the administration of an SMSM compound or a pharmaceutically acceptable salt thereof, or composition or medicament thereof to a patient results in one, two or more of the following effects: (i) reduces or ameliorates the severity of a disease; (ii) delays onset of a disease; (iii) inhibits the progression of a disease; (iv) reduces hospitalization of a subject; (v) reduces hospitalization length for a subject; (vi) increases the survival of a subject; (vii) improves the quality of life of a subject; (viii) reduces the number of symptoms associated with a disease; (ix) reduces or ameliorates the severity of a symptom associated with a disease; (x) reduces the duration of a symptom associated with a disease associated; (xi) prevents the recurrence of a symptom associated with a disease; (xii) inhibits the development or onset of a symptom of a disease; and/or (xiii) inhibits of the progression of a symptom associated with a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to restore the amount of a RNA transcript of a gene to the amount of the RNA transcript detectable in healthy patients or cells from healthy patients. In other embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to restore the amount an RNA isoform and/or protein isoform of gene to the amount of the RNA isoform and/or protein isoform detectable in healthy patients or cells from healthy patients.

[0447] In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to decrease the aberrant amount of an RNA transcript of a gene which associated with a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to decrease the amount of the aberrant expression of an isoform of a gene. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to result in a substantial change in the amount of an RNA transcript (e.g., mRNA transcript), alternative splice variant or isoform.

[0448] In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to increase or decrease the amount of an RNA transcript (e.g., an mRNA transcript) of gene which is beneficial for the prevention and/or treatment of a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to increase or decrease the amount of an alternative splice variant of an RNA transcript of gene which is beneficial for the prevention and/or treatment of a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to increase or decrease the amount of an isoform of gene which is beneficial for the prevention and/or treatment of a disease.

[0449] A method of treating cancer in a subject in need thereof can comprise administering to the subject a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof. A method of treating a non-cancer disease or condition in a subject in need thereof can comprise administering to the subject a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof.

[0450] In some embodiments, the present disclosure relates to a method for the treatment, prevention and/or delay of progression of cancer or a non-cancer disease or condition comprising administering an effective amount of a SMSM as described herein to a subject, in particular to a mammal.

[0451] In some embodiments, an effective amount in the context of the administration of an SMSM compound or a pharmaceutically acceptable salt thereof, or composition or medicament thereof refers to an amount of an SMSM compound or a pharmaceutically acceptable salt thereof to a patient which has a therapeutic effect and/or beneficial effect. In certain specific embodiments, an effective amount in the context of the administration of an SMSM compound or a pharmaceutically acceptable salt thereof, or composition or medicament thereof to a patient results in one, two or more of the following effects: (i) reduces or ameliorates the severity of a disease; (ii) delays onset of a disease; (iii) inhibits the progression of a disease; (iv) reduces hospitalization of a subject; (v) reduces hospitalization length for a subject; (vi) increases the survival of a subject; (vii) improves the quality of life of a subject; (viii) reduces the number of symptoms associated with a disease; (ix) reduces or ameliorates the severity of a symptom associated with a disease; (x) reduces the duration of a symptom associated with a disease associated; (xi) prevents the recurrence of a symptom associated with a disease; (xii) inhibits the development or onset of a symptom of a disease; and/or (xiii) inhibits of the progression of a symptom associated with a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to restore the amount of a RNA transcript of a gene to the amount of the RNA transcript detectable in healthy patients or cells from healthy patients. In other embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to restore the amount an RNA isoform and/or protein isoform of gene to the amount of the RNA isoform and/or protein isoform detectable in healthy patients or cells from healthy patients.

[0452] In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to decrease the aberrant amount of an RNA transcript of a gene which associated with a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to decrease the amount of the aberrant expression of an isoform of a gene. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to result in a substantial change in the amount of an RNA transcript (e.g., mRNA transcript), alternative splice variant or isoform.

[0453] In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to increase or decrease the amount of an RNA transcript (e.g., an mRNA transcript) of gene which is beneficial for the prevention and/or treatment of a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to increase or decrease the amount of an alternative splice variant of an RNA transcript of gene which is beneficial for the prevention and/or treatment of a disease. In some embodiments, an effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof is an amount effective to increase or decrease the amount of an isoform of gene which is beneficial for the prevention and/or treatment of a disease. Non- limiting examples of effective amounts of an SMSM compound or a pharmaceutically acceptable salt thereof are described herein. For example, the effective amount may be the amount required to prevent and/or treat a disease associated with the aberrant amount of an mRNA transcript of gene in a human subject. In general, the effective amount will be in a range of from about 0.001 mg/kg/day to about 500 mg/kg/day for a patient having a weight in a range of between about 1 kg to about 200 kg. The typical adult subject is expected to have a median weight in a range of between about 70 and about 100 kg.

[0454] In one embodiment, an SMSM described herein can be used in the preparation of medicaments for the treatment of diseases or conditions described herein. In addition, a method for treating any of the diseases or conditions described herein in a subject in need of such treatment, can involve administration of pharmaceutical compositions that includes at least one SMSM described herein or a pharmaceutically acceptable salt, thereof, in a therapeutically effective amount to a subject.

[0455] In certain embodiments, an SMSM described herein can be administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition. Amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient’s health status, weight, and response to the drugs, and the judgment of the treating physician. Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation clinical trial. In prophylactic applications, compositions containing an SMSM described herein can be administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition. In certain embodiments, the dose of drug being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a“drug holiday”). Doses employed for adult human treatment typically range of 0.01mg-5000 mg per day or from about 1 mg to about 1000 mg per day. In some embodiments, a desired dose is conveniently presented in a single dose or in divided doses.

[0456] For combination therapies described herein, dosages of the co-administered compounds can vary depending on the type of co-drug(s) employed, on the specific drug(s) employed, on the disease or condition being treated and so forth. In additional embodiments, when co-administered with one or more other therapeutic agents, the compound provided herein is administered either simultaneously with the one or more other therapeutic agents, or sequentially. If administration is simultaneous, the multiple therapeutic agents can be, by way of example only, provided in a single, unified form, or in multiple forms.

Conditions and Diseases

[0457] The present disclosure relates to a pharmaceutical composition comprising a SMSM described herein for use in the treatment, prevention and/or delay of progression of a disease, disorder or condition. In some embodiments, the present disclosure relates to a pharmaceutical composition comprising a SMSM described herein for use in the treatment, prevention and/or delay of progression of a disease, disorder or condition in Table 2A, Table 2B, Table 2C and Table 2D.

[0458] A method of treating, preventing, or delaying a non-cancer disease or condition disease can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a disease, disorder or condition in Table 2A, Table 2B, Table 2C and Table 2D.

[0459] In some embodiments, the present disclosure relates to a pharmaceutical composition comprising a SMSM described herein for use in the treatment, prevention and/or delay of progression of cancer.

[0460] A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a liquid cancer. A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a leukemia or lymphoma. A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a leukemia, acute myeloid leukemia, colon cancer, gastric cancer, macular degeneration, acute monocytic leukemia, breast cancer, hepatocellular carcinoma, cone-rod dystrophy, alveolar soft part sarcoma, myeloma, skin melanoma, prostatitis, pancreatitis, pancreatic cancer, retinitis, adenocarcinoma, adenoiditis, adenoid cystic carcinoma, cataract, retinal degeneration, gastrointestinal stromal tumor, Wegener’s granulomatosis, sarcoma, myopathy, prostate adenocarcinoma, Hodgkin’s lymphoma, ovarian cancer, non-Hodgkin’s lymphoma, multiple myeloma, chronic myeloid leukemia, acute lymphoblastic leukemia, renal cell carcinoma, transitional cell carcinoma, colorectal cancer, chronic lymphocytic leukemia, anaplastic large cell lymphoma, kidney cancer, breast cancer, cervical cancer.

[0461] A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a solid cancer or solid tumor.

[0462] In some embodiments, the tumor is selected from the group consisting of adenocarcinoma, melanoma (e.g., metastatic melanoma), liver cancer (e.g., hepatocellular carcinoma, hepatoblastoma, liver carcinoma), prostate cancer (e.g., prostate adenocarcinoma, androgen-independent prostate cancer, androgen-dependent prostate cancer, prostate carcinoma), sarcoma (e.g., leiomyosarcoma, rhabdomyosarcoma), brain cancer (e.g., glioma, a malignant glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, primary brain lymphoma, anaplastic astrocytoma, juvenile pilocytic astrocytoma, a mixture of oligodendroglioma and astrocytoma elements), breast cancer (e.g., triple negative breast cancer, metastatic breast cancer, breast carcinoma, breast sarcoma, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, inflammatory breast cancer), Paget’s disease, juvenile Paget’s disease, lung cancer (e.g., KRAS-mutated non-small cell lung cancer, non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma, small cell lung cancer, lung carcinoma), pancreatic cancer (e.g., insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, carcinoid tumor, islet cell tumor, pancreas carcinoma), skin cancer (e.g., skin melanoma, basal cell carcinoma, squamous cell carcinoma, melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, acrallentiginous melanoma, skin carcinoma), cervical cancer (e.g., squamous cell carcinoma, adenocarcinoma, cervical carcinoma), ovarian cancer (e.g., ovarian epithelial carcinoma, borderline tumor, germ cell tumor, stromal tumor, ovarian carcinoma), cancer of the mouth, cancer of the nervous system (e.g., cancer of the central nervous system, a CNS germ cell tumor), goblet cell metaplasia, kidney cancer (e.g., renal cell cancer, adenocarcinoma, hypernephroma, Wilms’ tumor, fibrosarcoma, transitional cell cancer (renal pelvis and/or uterer), renal cell carcinoma, renal carcinoma), bladder cancer (e.g., transitional cell carcinoma, squamous cell cancer, carcinosarcoma), stomach cancer (e.g., fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, liposarcoma, fibrosarcoma, carcinosarcoma), uterine cancer (e.g., endometrial cancer, endometrial carcinoma, uterine sarcoma), cancer of the esophagus (e.g., squamous cancer, adenocarcinoma, adenoid cyctic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma, esophageal carcinomas), colon cancer (e.g., colon carcinoma), cancer of the rectum (e.g., rectal cancers), colorectal cancer (e.g., colorectal carcinoma, metastatic colorectal cancer, hereditary nonpolyposis colorectal cancer, KRAS mutated colorectal cancer), gallbladder cancer (e.g., adenocarcinoma, cholangiocarcinoma, papillary cholangiocarcinoma, nodular cholangiocarcinoma, diffuse cholangiocarcinoma), testicular cancer (e.g., germinal tumor, seminoma, anaplastic testicular cancer, classic (typical) testicular cancer, spermatocytic testicular cancer, nonseminoma testicular cancer), embryonal carcinoma (e.g., teratoma carcinoma, choriocarcinoma (yolk-sac tumor)), gastric cancer (e.g., gastrointestinal stromal tumor, cancer of other gastrointestinal tract organs, gastric carcinomas), bone cancer (e.g., connective tissue sarcoma, bone sarcoma, cholesteatoma-induced bone osteosarcoma, Paget’s disease of bone, osteosarcoma, chondrosarcoma, Ewing’s sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcoma, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi’s sarcoma, leiomyosarcoma, alveolar soft part sarcoma), liposarcoma, lymphangiosarcoma, neurilemmoma, rhabdomyosarcoma, synovial sarcoma, cancer of the lymph node (e.g., lymphangioendotheliosarcoma), adenoid cystic carcinoma, vaginal cancer (e.g., squamous cell carcinoma, adenocarcinoma, melanoma), vulvar cancer (e.g., squamous cell carcinoma, melanoma, adenocarcinoma, sarcoma, Paget’s disease), cancer of other reproductive organs, thyroid cancer (e.g., papillary thyroid cancer, follicular thyroid cancer, medullary thyroid cancer, anaplastic thyroid cancer, thyroid carcinoma), salivary gland cancer (e.g., adenocarcinoma, mucoepidermoid carcinoma), eye cancer (e.g., ocular melanoma, iris melanoma, choroidal melanoma, cilliary body melanoma, retinoblastoma), penal cancers, oral cancer (e.g. squamous cell carcinoma, basal cancer), pharynx cancer (e.g., squamous cell cancer, verrucous pharynx cancer), cancer of the head, cancer of the neck, cancer of the throat, cancer of the chest, cancer of the spleen, cancer of skeletal muscle, cancer of subcutaneous tissue, adrenal cancer, pheochromocytoma, adrenocortical carcinoma, pituitary cancer, Cushing’s disease, prolactin-secreting tumor, acromegaly, diabetes insipidus, myxosarcoma, osteogenic sarcoma, endotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, ependyoma, optic nerve glioma, primitive neuroectodermal tumor, rhabdoid tumor, renal cancer, glioblastoma multiforme, neurofibroma, neurofibromatosis, pediatric cancer, neuroblastoma, malignant melanoma, carcinoma of the epidermis, polycythemia vera, Waldenstrom’s macroglobulinemia, monoclonal gammopathy of undetermined significance, benign monoclonal gammopathy, heavy chain disease, pediatric solid tumor, Ewing’s sarcoma, Wilms tumor, carcinoma of the epidermis, HIV- related Kaposi’s sarcoma, rhabdomyosarcoma, thecomas, arrhenoblastomas, endometrial carcinoma, endometrial hyperplasia, endometriosis, fibrosarcomas, choriocarcinoma, nasopharyngeal carcinoma, laryngeal carcinoma, hepatoblastoma, Kaposi’s sarcoma, hemangioma, cavernous hemangioma, hemangioblastoma, retinoblastoma, glioblastoma, Schwannoma, neuroblastoma, rhabdomyosarcoma, osteogenic sarcoma, leiomyosarcoma, urinary tract carcinoma, abnormal vascular proliferation associated with phakomatoses, edema (such as that associated with brain tumors), Meigs’ syndrome, pituitary adenoma, primitive neuroectodermal tumor, medullblastoma, and acoustic neuroma.

[0463] A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with basal cell carcinoma, goblet cell metaplasia, or a malignant glioma. A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a cancer of the liver, breast, lung, prostate, cervix, uterus, colon, pancreas, kidney, stomach, bladder, ovary, or brain.

[0464] A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a cancer of the head, neck, eye, mouth, throat, esophagus, esophagus, chest, bone, lung, kidney, colon, rectum or other gastrointestinal tract organs, stomach, spleen, skeletal muscle, subcutaneous tissue, prostate, breast, ovaries, testicles or other reproductive organs, skin, thyroid, blood, lymph nodes, kidney, liver, pancreas, and brain or central nervous system.

[0465] Specific examples of cancers that can be prevented and/or treated in accordance with present disclosure include, but are not limited to, the following: renal cancer, kidney cancer, glioblastoma multiforme, metastatic breast cancer; breast carcinoma; breast sarcoma; neurofibroma; neurofibromatosis; pediatric tumors; neuroblastoma; malignant melanoma; carcinomas of the epidermis; leukemias such as but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, erythroleukemia leukemias and myclodysplastic syndrome, chronic leukemias such as but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as but not limited to Hodgkin’s disease, non-Hodgkin’s disease; multiple myelomas such as but not limited to smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma and extramedullary plasmacytoma; Waldenstrom’s macroglobulinemia; monoclonal gammopathy of undetermined significance; benign monoclonal gammopathy; heavy chain disease; bone cancer and connective tissue sarcomas such as but not limited to bone sarcoma, myeloma bone disease, multiple myeloma, cholesteatoma-induced bone osteosarcoma, Paget’s disease of bone, osteosarcoma, chondrosarcoma, Ewing’s sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi’s sarcoma, leiomyosarcoma, liposarcoma, lymphangio sarcoma, neurilemmoma, rhabdomyosarcoma, and synovial sarcoma; brain tumors such as but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, and primary brain lymphoma; breast cancer including but not limited to adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, Paget’s disease (including juvenile Paget’s disease) and inflammatory breast cancer; adrenal cancer such as but not limited to pheochromocytom and adrenocortical carcinoma; thyroid cancer such as but not limited to papillary or follicular thyroid cancer, medullary thyroid cancer and anaplastic thyroid cancer; pancreatic cancer such as but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; pituitary cancers such as but limited to Cushing’s disease, prolactin-secreting tumor, acromegaly, and diabetes insipius; eye cancers such as but not limited to ocular melanoma such as iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma; vaginal cancers such as squamous cell carcinoma, adenocarcinoma, and melanoma; vulvar cancer such as squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget’s disease; cervical cancers such as but not limited to, squamous cell carcinoma, and adenocarcinoma; uterine cancers such as but not limited to endometrial carcinoma and uterine sarcoma; ovarian cancers such as but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor; cervical carcinoma; esophageal cancers such as but not limited to, squamous cancer, adenocarcinoma, adenoid cyctic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell(small cell) carcinoma; stomach cancers such as but not limited to, adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; colon cancers; KRAS mutated colorectal cancer; colon carcinoma; rectal cancers; liver cancers such as but not limited to hepatocellular carcinoma and hepatoblastoma, gallbladder cancers such as adenocarcinoma; cholangiocarcinomas such as but not limited to papillary, nodular, and diffuse; lung cancers such as KRAS-mutated non-small cell lung cancer, non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma and small-cell lung cancer; lung carcinoma; testicular cancers such as but not limited to germinal tumor, seminoma, anaplastic, classic (typical), spermatocytic, nonseminoma, embryonal carcinoma, teratoma carcinoma, choriocarcinoma (yolk-sac tumor), prostate cancers such as but not limited to, androgen-independent prostate cancer, androgen-dependent prostate cancer, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; penal cancers; oral cancers such as but not limited to squamous cell carcinoma; basal cancers; salivary gland cancers such as but not limited to adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; pharynx cancers such as but not limited to squamous cell cancer, and verrucous; skin cancers such as but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, acrallentiginous melanoma; kidney cancers such as but not limited to renal cell cancer, adenocarcinoma, hypernephroma, fibrosarcoma, transitional cell cancer (renal pelvis and/or uterer); renal carcinoma; Wilms’ tumor; bladder cancers such as but not limited to transitional cell carcinoma, squamous cell cancer, adenocarcinoma, carcinosarcoma. In addition, cancers include myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangioendotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma and papillary adenocarcinomas.

[0466] A method of treating, preventing, or delaying cancer can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with a pediatric solid tumor, Ewing’s sarcoma, Wilms tumor, neuroblastoma, neurofibroma, carcinoma of the epidermis, malignant melanoma, cervical carcinoma, colon carcinoma, lung carcinoma, renal carcinoma, breast carcinoma, breast sarcoma, metastatic breast cancer, HIV-related Kaposi’s sarcoma, prostate cancer, androgen- independent prostate cancer, androgen-dependent prostate cancer, neurofibromatosis, lung cancer, non-small cell lung cancer, KRAS-mutated non-small cell lung cancer, malignant melanoma, melanoma, colon cancer, KRAS-mutated colorectal cancer, glioblastoma multiforme, renal cancer, kidney cancer, bladder cancer, ovarian cancer, hepatocellular carcinoma, thyroid carcinoma, rhabdomyosarcoma, acute myeloid leukemia, or multiple myeloma. [0467] In some embodiments, cancers and conditions associated therewith that are prevented and/or treated in accordance with the present disclosure are breast carcinomas, lung carcinomas, gastric carcinomas, esophageal carcinomas, colorectal carcinomas, liver carcinomas, ovarian carcinomas, thecomas, arrhenoblastomas, cervical carcinomas, endometrial carcinoma, endometrial hyperplasia, endometriosis, fibrosarcomas, choriocarcinoma, head and neck cancer, nasopharyngeal carcinoma, laryngeal carcinomas, hepatoblastoma, Kaposi’s sarcoma, melanoma, skin carcinomas, hemangioma, cavernous hemangioma, hemangioblastoma, pancreas carcinomas, retinoblastoma, astrocytoma, glioblastoma, Schwannoma, oligodendroglioma, medulloblastoma, neuroblastomas, rhabdomyosarcoma, osteogenic sarcoma, leiomyosarcomas, urinary tract carcinomas, thyroid carcinomas, Wilm’s tumor, renal cell carcinoma, prostate carcinoma, abnormal vascular proliferation associated with phakomatoses, edema (such as that associated with brain tumors), or Meigs’ syndrome. In specific embodiment, the cancer an astrocytoma, an oligodendroglioma, a mixture of oligodendroglioma and an astrocytoma elements, an ependymoma, a meningioma, a pituitary adenoma, a primitive neuroectodermal tumor, a medullblastoma, a primary central nervous system (CNS) lymphoma, or a CNS germ cell tumor.

[0468] In some embodiments, the cancer treated in accordance with the present disclosure is an acoustic neuroma, an anaplastic astrocytoma, a glioblastoma multiforme, or a meningioma. In some embodiments, the cancer treated in accordance with the present disclosure is a brain stem glioma, a craniopharyngioma, an ependyoma, a juvenile pilocytic astrocytoma, a medulloblastoma, an optic nerve glioma, primitive neuroectodermal tumor, or a rhabdoid tumor.

[0469] A method of treating, preventing, or delaying a condition or disease can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with acute myeloid leukemia, ALS, Alzheimer’s disease, argyrophilic grain disease, cancer metabolism, chronic lymphocytic leukemia, colorectal carcinoma, corticobasal degeneration, cystic fibrosis, dilated cardiomyopathy, Duchenne muscular dystrophy, Ehlers-Danlos syndrome, endometrial cancer, Fabry’s disease, familial dysautonomia, familial hypercholesterolemia, familial persistent hyperinsulinemic hypoglycemia, frontotemporal dementia, FTDP-17, gucher’s disease, glioma, globular glial tauopathy, HIV-1, Huntington’s disease, Hutchinson-Gilford progeria syndrome, hypercholesterolemia, Leber congenital amaurosis, migraine, multiple sclerosis, myelodysplastic syndromes, NASH, Niemann-Pick’s, non-small cell lung cancer, pain, Parkinson’s disease, phenylketonuria, Pick’s disease, progressive supranuclear palsy, spinal muscular atrophy, spinocerebellar ataxia type 2, or Wilson’s disease.

[0470] A method of treating, preventing, or delaying a non-cancer disease or condition disease can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with atypical hemolytic uremic syndrome (aHUS), cystic fibrosis, muscular dystrophy, polycystic autosomal-dominant kidney disease, cancer-induced cachexia, benign prostatic hyperplasia, rheumatoid arthritis, psoriasis, atherosclerosis, obesity, retinopathies (including diabetic retinopathy and retinopathy of prematurity), retrolental fibroplasia, neovascular glaucoma, age-related macular degeneration, exudative macular degeneration, thyroid hyperplasias (including Grave’s disease), corneal and other tissue transplantation, epidemic keratoconjunctivitis, Vitamin A deficiency, contact lens overwear, atopic keratitis, superior limbic keratitis, and pterygium keratitis sicca, viral infections, inflammation associated with viral infections, chronic inflammation, lung inflammation, nephrotic syndrome, preeclampsia, ascites, pericardial effusion (such as that associated with pericarditis), pleural effusion, Sjogren’s syndrome, acne rosacea, phylectenulosis, syphilis, lipid degeneration, chemical burns, bacterial ulcers, fungal ulcers, Herpes simplex infection, Herpes zoster infections, protozoan infections, Mooren’s ulcer, Terrien’s marginal degeneration, marginal keratolysis, systemic lupus, polyarteritis, trauma, Wegener’s sarcoidosis, Paget’s disease, scleritis, Stevens-Johnson’s disease, pemphigoid, radial keratotomy, Eales’ disease, Behcet’s disease, sickle cell anemia, pseudoxanthoma elasticum, Stargardt’s disease, pars planitis, chronic retinal detachment, vein occlusion, artery occlusion, carotid obstructive disease, chronic uveitis/vitritis, ocular histoplasmosis, Mycobacteria infections, Lyme’s disease, Best’s disease, myopia, optic pits, hyperviscosity syndromes, toxoplasmosis, sarcoidosis, trauma, post-laser complications, diseases associated with rubeosis (neovascularization of the iris and of the angle), and diseases caused by the abnormal proliferation of fibrovascular or fibrous tissue, including all forms of prolific vitreoretinopathy. Certain examples of non- neoplastic conditions that can be prevented and/or treated in accordance with the methods described herein include viral infections, including but not limited to, those associated with viruses belonging to Flaviviridae, flavivirus, pestivirus, hepacivirus, West Nile virus, hepatitis C virus (HCV) or human papilloma virus (HPV), cone-rod dystrophy, prostatitis, pancreatitis, retinitis, cataract, retinal degeneration, Wegener’s granulomatosis, myopathy, adenoiditis, germ cell tumors, combined methylmalonic aciduria and homocystinuria, cb1C type, Alzheimer’s disease, hyperprolinemia, acne, tuberculosis, succinic semialdehyde dehydrogenase deficiency, esophagitis, mental retardation, glycine encephalopathy, Crohn’s disease, spina bifida, autosomal recessive disease, schizophrenia, neural tube defects, myelodysplastic syndromes, amyotropic lateral sclerosis, neuronitis, Parkinson’s disease, talipes equinovarus, dystrophinopathies, cerebritis, bladder related disorders, cleft lip, cleft palate, cervicitis, spasticity, lipoma, scleroderma, Gitelman syndrome, poliomyelitis, paralysis, Aagenaes syndrome, oculomotor nerve paralysis, and spinal muscular atrophy.

[0471] A method of treating, preventing, or delaying a non-cancer disease or condition disease can comprise administering a therapeutically effective amount of a compound described herein or a pharmaceutically acceptable salt thereof to a subject with atypical hemolytic uremic syndrome (aHUS), Hutchinson-Gilford progeria syndrome (HGPS), Limb girdle muscular dystrophy type 1B, Familial partial lipodystrophy type 2, Frontotemporal dementia with parkinsonism chromosome 17, Richardson’s syndrome, PSP-Parkinsonism, Argyrophilic grain disease, Corticobasal degeneration, Pick’s disease, Globular glial tauopathy, Guadeloupean Parkinsonism, Myotonic dystrophy, Down Syndrome, Neonatal Hypoxia-Ischemia, Familial Dysautonomia, Spinal muscular atrophy, Hypoxanthine phosphoribosyltransferase deficiency, Ehlers-Danlos syndrome, Occipital Horn Syndrome, Fanconi Anemia, Marfan Syndrome, thrombotic thrombocytopenic purpura, glycogen Storage Disease Type III, cystic fibrosis, neurofibromatosis, Tyrosinemia (type I), Menkes Disease, Analbuminemia, Congenital acetylcholinesterase deficiency, Haemophilia B deficiency (coagulation factor IX deficiency), Recessive dystrophic epidermolysis bullosa, Dominant dystrophic epidermolysis bullosa, Somatic mutations in kidney tubular epithelial cells, Neurofibromatosis type II, X-linked adrenoleukodystrophy (X- ALD), FVII deficiency, Homozygous hypobetalipoproteinemia, Ataxia-telangiectasia, Androgen Sensitivity, Common congenital afibrinogenemia, Risk for emphysema, Mucopolysaccharidosis type II (Hunter syndrome), Severe type III osteogenesis imperfecta, Ehlers-Danlos syndrome IV, Glanzmann thrombasthenia, Mild Bethlem myopathy, Dowling-Meara epidermolysis bullosa simplex, Severe deficiency of MTHFR, Acute intermittent porphyria, Tay-Sachs Syndrome, Myophosphorylase deficiency (McArdle disease), Chronic Tyrosinemia Type 1, Mutation in placenta, Leukocyte adhesion deficiency, Hereditary C3 deficiency, Neurofibromatosis type I, Placental aromatase deficiency, Cerebrotendinous xanthomatosis, Duchenne and Becker muscular dystrophy, Severe factor V deficiency, Alpha-thalassemia, Beta-thalassemia, Hereditary HL deficiency, Lesch-Nyhan syndrome, Familial hypercholesterolemia, Phosphoglycerate kinase deficiency, Cowden syndrome, X-linked retinitis pigmentosa (RP3), Crigler-Najjar syndrome type 1, Chronic tyrosinemia type I, Sandhoff disease, Maturity onset diabetes of the young (MODY), Familial tuberous sclerosis, Polycystic kidney disease 1, or Primary Hyperthyroidism.

[0472] In some embodiments, non-cancer diseases that can be prevented and/or treated in accordance with the disclosure of WO2016/19638 6 al, WO2016/12834 3 a1, WO2015/02487 6 a2 and EP3053577A1. In some embodiments, non-cancer diseases that can be prevented and/or treated include, but are not limited to, atypical hemolytic uremic syndrome (aHUS), Hutchinson-Gilford progeria syndrome (HGPS), Limb girdle muscular dystrophy type 1B, Familial partial lipodystrophy type 2, Frontotemporal dementia with parkinsonism chromosome 17, Richardson’s syndrome, PSP-Parkinsonism, Argyrophilic grain disease, Corticobasal degeneration, Pick’s disease, Globular glial tauopathy, Guadeloupean Parkinsonism, Myotonic dystrophy, Down Syndrome, Neonatal Hypoxia-Ischemia, Familial Dysautonomia, Spinal muscular atrophy, Hypoxanthine phosphoribosyltransferase deficiency, Ehlers-Danlos syndrome, Occipital Horn Syndrome, Fanconi Anemia, Marfan Syndrome, thrombotic thrombocytopenic purpura, glycogen Storage Disease Type III, cystic fibrosis, neurofibromatosis, Tyrosinemia (type I), Menkes Disease, Analbuminemia, Congenital acetylcholinesterase deficiency, Haemophilia B deficiency (coagulation factor IX deficiency), Recessive dystrophic epidermolysis bullosa, Dominant dystrophic epidermolysis bullosa, Somatic mutations in kidney tubular epithelial cells, Neurofibromatosis type II, X-linked adrenoleukodystrophy (X-ALD), FVII deficiency, Homozygous hypobetalipoproteinemia, Ataxia-telangiectasia, Androgen Sensitivity, Common congenital afibrinogenemia, Risk for emphysema, Mucopolysaccharidosis type II (Hunter syndrome), Severe type III osteogenesis imperfecta, Ehlers-Danlos syndrome IV, Glanzmann thrombasthenia, Mild Bethlem myopathy, Dowling-Meara epidermolysis bullosa simplex, Severe deficiency of MTHFR, Acute intermittent porphyria, Tay-Sachs Syndrome, Myophosphorylase deficiency (McArdle disease), Chronic Tyrosinemia Type 1, Mutation in placenta, Leukocyte adhesion deficiency, Hereditary C3 deficiency, Neurofibromatosis type I, Placental aromatase deficiency, Cerebrotendinous xanthomatosis, Duchenne and Becker muscular dystrophy, Severe factor V deficiency, Alpha-thalassemia, Beta-thalassemia, Hereditary HL deficiency, Lesch-Nyhan syndrome, Familial hypercholesterolemia, Phosphoglycerate kinase deficiency, Cowden syndrome, X-linked retinitis pigmentosa (RP3), Crigler-Najjar syndrome type 1, Chronic tyrosinemia type I, Sandhoff disease, Maturity onset diabetes of the young (MODY), Familial tuberous sclerosis, or Polycystic kidney disease 1. Methods of Administering

[0473] The compositions described herein can be administered to the subject in a variety of ways, including parenterally, intravenously, intradermally, intramuscularly, colonically, rectally or intraperitoneally. In some embodiments, the small molecule splicing modulator or a pharmaceutically acceptable salt thereof is administered by intraperitoneal injection, intramuscular injection, subcutaneous injection, or intravenous injection of the subject. In some embodiments, the pharmaceutical compositions can be administered parenterally, intravenously, intramuscularly or orally. The oral agents comprising a small molecule splicing modulator can be in any suitable form for oral administration, such as liquid, tablets, capsules, or the like. The oral formulations can be further coated or treated to prevent or reduce dissolution in stomach. The compositions of the present invention can be administered to a subject using any suitable methods known in the art. Suitable formulations for use in the present invention and methods of delivery are generally well known in the art. For example, the small molecule splicing modulators described herein can be formulated as pharmaceutical compositions with a pharmaceutically acceptable diluent, carrier or excipient. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions including pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, such as, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.

[0474] Pharmaceutical formulations described herein can be administrable to a subject in a variety of ways by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular, intramedullary injections, intrathecal, direct intraventricular, intraperitoneal, intralymphatic, intranasal injections), intranasal, buccal, topical or transdermal administration routes. The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.

[0475] In some embodiments, the pharmaceutical compositions described herein are administered orally. In some embodiments, the pharmaceutical compositions described herein are administered topically. In such embodiments, the pharmaceutical compositions described herein are formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, shampoos, scrubs, rubs, smears, medicated sticks, medicated bandages, balms, creams or ointments. In some embodiments, the pharmaceutical compositions described herein are administered topically to the skin. In some embodiments, the pharmaceutical compositions described herein are administered by inhalation. In some embodiments, the pharmaceutical compositions described herein are formulated for intranasal administration. Such formulations include nasal sprays, nasal mists, and the like. In some embodiments, the pharmaceutical compositions described herein are formulated as eye drops. In some embodiments, the pharmaceutical compositions described herein are: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by inhalation to the mammal; and/or (e) administered by nasal administration to the mammal; or and/or (f) administered by injection to the mammal; and/or (g) administered topically to the mammal; and/or (h) administered by ophthalmic administration; and/or (i) administered rectally to the mammal; and/or (j) administered non-systemically or locally to the mammal. In some embodiments, the pharmaceutical compositions described herein are administered orally to the mammal. In certain embodiments, an SMSM described herein is administered in a local rather than systemic manner. In some embodiments, an SMSM described herein is administered topically. In some embodiments, an SMSM described herein is administered systemically.

[0476] Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.

[0477] For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser that contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.

[0478] Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.

[0479] SMSMs suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against contamination from microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.

Dosing and Schedules

[0480] The SMSMs utilized in the methods of the invention can be, e.g., administered at dosages that may be varied depending upon the requirements of the subject the severity of the condition being treated and/or imaged, and/or the SMSM being employed. For example, dosages can be empirically determined considering the type and stage of disease diagnosed in a particular subject and/or the type of imaging modality being used in conjunction with the SMSMs. The dose administered to a subject, in the context of the present invention should be sufficient to affect a beneficial diagnostic or therapeutic response in the subject. The size of the dose also can be determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a SMSM in a particular subject.

[0481] It is advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. Toxicity and therapeutic efficacy of such compounds can be determined by procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 . Compounds that exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue to minimize potential damage to uninfected cells and, thereby, reduce side effects.

[0482] Therapeutic index data obtained from cell culture assays and/or animal studies can be used in predicting the therapeutic index in vivo and formulating a range of dosages for use in subjects, such as human subjects. The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the concentration of the test compound which achieves a half-maximal inhibition of symptoms as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography. Various animal models and clinical assays for evaluating effectiveness of a particular SMSM in preventing or reducing a disease or condition are known in the art may be used in the present invention. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g. Fingl et al, 1975, In: The Pharmacological Basis of Therapeutics. Ch.1 pi).

[0483] In some aspects, the SMSMs provided have a therapeutic index (LD 50 /ED 50 ) of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 10000, or 100000 or more. In some aspects, the SMSMs provided have a therapeutic index (LD 50 /ED 50 ) of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 10000, or 100000 or more as determined in cell culture.

[0484] In some aspects, the SMSMs provided have an IC 50 viability / EC 50 splicing value of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 10000, or 100000 or more. In some aspects, the SMSMs provided have an IC 50 viability / EC 50 splicing value of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 10000, or 100000 or more as determined in cell culture.

[0485] A dosage of using an SMSM when administered may be at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 grams/m 2 in humans, or a dosage in another subject comparable to that in humans. A dosage ("dosage X") of an SMSM in a subject other than a human is comparable to a dosage ("dosage Y") of the SMSM in humans if the serum concentration of the scavenger in the subject post administration of the SMSM at dosage X is equal to the serum concentration of the SMSM in humans post administration of the compound at dosage Y.

[0486] Within the scope of the present description, the effective amount of an SMSM compound or a pharmaceutically acceptable salt thereof for use in the manufacture of a medicament, the preparation of a pharmaceutical kit or in a method for preventing and/or treating a disease in a human subject in need thereof, is intended to include an amount in a range of from about 1 mg to about 50 grams.

[0487] The compositions of the present invention can be administered as frequently as necessary, including hourly, daily, weekly or monthly.

[0488] In any of the aforementioned aspects are further embodiments comprising single administrations of an effective amount of an SMSM described herein, including further embodiments in which (i) the compound is administered once; (ii) the compound is administered to the mammal multiple times over the span of one day; (iii) continually; or (iv) continuously.

[0489] In any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of an SMSM described herein, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours. In further or alternative embodiments, the method comprises a drug holiday, wherein the administration of an SMSM described herein is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed. In one embodiment, the length of the drug holiday varies from 2 days to 1 year.

Combination Therapies

[0490] In certain instances, it is appropriate to administer at least one SMSM described herein in combination with another therapeutic agent. For example, a compound SMSM described herein can be co-administered with a second therapeutic agent, wherein SMSM and the second therapeutic agent modulate different aspects of the disease, disorder or condition being treated, thereby providing a greater overall benefit than administration of either therapeutic agent alone.

[0491] In some embodiments, an SMSM described herein can be used in combination with an anti-cancer therapy. In some embodiments, a steric modulator is used in combination with conventional chemotherapy, radiotherapy, hormonal therapy, and/or immunotherapy. In some embodiments, an SMSM described herein can be used in combination with conventional chemotherapeutic agents including alkylating agents (e.g., temozolomide, cyclophosphamide, ifosfamide, chlorambucil, busulfan, melphalan, mechlorethamine, uramustine, thiotepa, nitrosoureas, etc.), anti-metabolites (e.g., 5-fluorouracil, azathioprine, methotrexate, leucovorin, capecitabine, cytarabine, floxuridine, fludarabine, gemcitabine, pemetrexed, raltitrexed, etc.), plant alkaloids (e.g., vincristine, vinblastine, vinorelbine, vindesine, podophyllotoxin, paclitaxel, docetaxel, etc.), topoisomerase inhibitors (e.g., irinotecan, topotecan, amsacrine, etoposide (VP16), etoposide phosphate, teniposide, etc.), antitumor antibiotics (e.g., doxorubicin, adriamycin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, mitoxantrone, plicamycin, etc.), platinum-based compounds (e.g. cisplatin, oxaloplatin, carboplatin, etc.), EGFR inhibitors (e.g., gefitinib, erlotinib, etc.), and the like.

[0492] In some embodiments, an SMSM may be administered in combination with one or more other SMSMs.

[0493] A SMSM may be administered to a subject in need thereof prior to, concurrent with, or following the administration of chemotherapeutic agents. For instance, SMSMs may be administered to a subject at least 8 hours, 7 hours, 6 hours, 5 hours, 4 hours, 3 hours, 2 hours, 1.5 hours, 1 hour, or 30 minutes before the starting time of the administration of chemotherapeutic agent(s). In certain embodiments, they may be administered concurrent with the administration of chemotherapeutic agent(s). In other words, in these embodiments, SMSMs are administrated at the same time when the administration of chemotherapeutic agent(s) starts. In other embodiments, SMSMs may be administered following the starting time of administration of chemotherapeutic agent(s) (e.g., at least 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours or 8 hours after the starting time of administration of chemotherapeutic agents). Alternatively, SMSMs may be administered at least 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours or 8 hours after the completion of administration of chemotherapeutic agents. Generally, these SMSMs are administered for a sufficient period of time so that the disease or condition is prevented or reduced. Such sufficient period of time may be identical to, or different from, the period during which chemotherapeutic agent(s) are administered. In certain embodiments, multiple doses of SMSMs are administered for each administration of a chemotherapeutic agent or a combination of multiple chemotherapeutic agents.

[0494] In certain embodiments, an appropriate dosage of a SMSM is combined with a specific timing and/or a particular route to achieve the optimum effect in preventing or reducing the disease or condition. For instance, an SMSM may be administered to a human orally at least 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours 8 hours, 9 hours, 10 hours, 11 hours or 12 hours; or at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days; or at least 1 week, 2 weeks, 3 weeks or 4 weeks; or at least 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months or 12 months; prior to or after the beginning or the completion, of the administration of a chemotherapeutic agent or a combination of chemotherapeutic agents.

Subjects

[0495] The subjects that can be treated with the SMSMs and methods described herein can be any subject that produces mRNA that is subject to alternative splicing, e.g., the subject may be a eukaryotic subject, such as a plant or an animal. In some embodiments, the subject is a mammal, e.g., human. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human animal. In some embodiments, the subject is a fetus, an embryo, or a child. In some embodiments, the subject is a non-human primate such as chimpanzee, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like.

[0496] In some embodiments, the subject is prenatal (e.g., a fetus), a child (e.g., a neonate, an infant, a toddler, a preadolescent), an adolescent, a pubescent, or an adult (e.g., an early adult, a middle aged adult, a senior citizen). The human subject can be between about 0 months and about 120 years old, or older. The human subject can be between about 0 and about 12 months old; for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months old. The human subject can be between about 0 and 12 years old; for example, between about 0 and 30 days old; between about 1 month and 12 months old; between about 1 year and 3 years old; between about 4 years and 5 years old; between about 4 years and 12 years old; about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 years old. The human subject can be between about 13 years and 19 years old; for example, about 13, 14, 15, 16, 17, 18, or 19 years old. The human subject can be between about 20 and about 39 year old; for example, about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, or 39 years old. The human subject can be between about 40 to about 59 years old; for example, about 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, or 59 years old. The human subject can be greater than 59 years old; for example, about 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, or 120 years old. The human subjects can include living subjects or deceased subjects. The human subjects can include male subjects and/or female subjects.

Assays [0497] Gene expression experiments often involve measuring the relative amount of gene expression products, such as mRNA, expressed in two or more experimental conditions. This is because altered levels of a specific sequence of a gene expression product can suggest a changed need for the protein coded for by the gene expression product, perhaps indicating a homeostatic response or a pathological condition.

[0498] In some embodiments, a method can comprise measuring, assaying or obtaining expression levels of one or more genes. In some cases, the method provides a number or a range of numbers, of genes that the expression levels of the genes can be used to diagnose, characterize or categorize a biological sample. In some embodiments, the gene expression data corresponds to data of an expression level of one or more biomarkers that are related to a disease or condition. The number of genes used can be between about 1 and about 500; for example about 1-500, 1-400, 1-300, 1-200, 1-100, 1-50, 1-25, 1-10, 10-500, 10-400, 10-300, 10-200, 10-100, 10-50, 10-25, 25-500, 25-400, 25-300, 25-200, 25-100, 25-50, 50-500, 50-400, 50-300, 50-200, 50-100, 100- 500, 100-400, 100-300, 100-200, 200-500, 200-400, 200-300, 300-500, 300-400, 400-500, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, or any included range or integer. For example, at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 33, 35, 38, 40, 43, 45, 48, 50, 53, 58, 63, 65, 68, 100, 120, 140, 142, 145, 147, 150, 152, 157, 160, 162, 167, 175, 180, 185, 190, 195, 200, 300, 400, 500 or more total genes can be used. The number of genes used can be less than or equal to about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 33, 35, 38, 40, 43, 45, 48, 50, 53, 58, 63, 65, 68, 100, 120, 140, 142, 145, 147, 150, 152, 157, 160, 162, 167, 175, 180, 185, 190, 195, 200, 300, 400, 500, or more.

[0499] In some embodiments, relative gene expression, as compared to normal cells and/or tissues of the same organ, can be determined by measuring the relative rates of transcription of RNA, such as by production of corresponding cDNAs and then analyzing the resulting DNA using probes developed from the gene sequences as corresponding to a genetic marker. Thus, the levels of cDNA produced by use of reverse transcriptase with the full RNA complement of a cell suspected of being cancerous produces a corresponding amount of cDNA that can then be amplified using polymerase chain reaction, or some other means, such as linear amplification, isothermal amplification, NASB, or rolling circle amplification, to determine the relative levels of resulting cDNA and, thereby, the relative levels of gene expression. General methods for determining gene expression product levels are known to the art and may include but are not limited to one or more of the following: additional cytological assays, assays for specific proteins or enzyme activities, assays for specific expression products including protein or RNA or specific RNA splice variants, in situ hybridization, whole or partial genome expression analysis, microarray hybridization assays, SAGE, enzyme linked immuno-absorbance assays, mass-spectrometry, immuno-histochemistry, blotting, microarray, RT-PCR, quantitative PCR, sequencing, RNA sequencing, DNA sequencing (e.g., sequencing of cDNA obtained from RNA); Next-Gen sequencing, nanopore sequencing, pyrosequencing, or Nanostring sequencing. Gene expression product levels may be normalized to an internal standard such as total mRNA or the expression level of a particular gene including but not limited to glyceraldehyde 3-phosphate dehydrogenase, or tubulin.

[0500] Gene expression data generally comprises the measurement of the activity (or the expression) of a plurality of genes, to create a picture of cellular function. Gene expression data can be used, for example, to distinguish between cells that are actively dividing, or to show how the cells react to a particular treatment. Microarray technology can be used to measure the relative activity of previously identified target genes and other expressed sequences. Sequence based techniques, like serial analysis of gene expression (SAGE, SuperSAGE) are also used for assaying, measuring or obtaining gene expression data. SuperSAGE is especially accurate and can measure any active gene, not just a predefined set. In an RNA, mRNA or gene expression profiling microarray, the expression levels of thousands of genes can be simultaneously monitored to study the effects of certain treatments, diseases, and developmental stages on gene expression.

[0501] In accordance with the foregoing, the expression level of a gene, marker, gene expression product, mRNA, pre-mRNA, or a combination thereof may be determined using northern blotting and employing the sequences as identified herein to develop probes for this purpose. Such probes may be composed of DNA or RNA or synthetic nucleotides or a combination of these and may advantageously be comprised of a contiguous stretch of nucleotide residues matching, or complementary to, a sequence corresponding to a genetic marker. Such probes will most usefully comprise a contiguous stretch of at least 15-200 residues or more including 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 175, or 200 nucleotides or more. Thus, where a single probe binds multiple times to the transcriptome of experimental cells, whereas binding of the same probe to a similar amount of transcriptome derived from the genome of control cells of the same organ or tissue results in observably more or less binding, this is indicative of differential expression of a gene, marker, gene expression product, mRNA, or pre-mRNA comprising, or corresponding to, sequences corresponding to a genetic marker from which the probe sequence was derived.

[0502] In some embodiments of the present invention, gene expression may be determined by microarray analysis using, for example, Affymetrix arrays, cDNA microarrays, oligonucleotide microarrays, spotted microarrays, or other microarray products from Biorad, Agilent, or Eppendorf. Microarrays provide particular advantages because they may contain a large number of genes or alternative splice variants that may be assayed in a single experiment. In some cases, the microarray device may contain the entire human genome or transcriptome or a substantial fraction thereof allowing a comprehensive evaluation of gene expression patterns, genomic sequence, or alternative splicing. Markers may be found using standard molecular biology and microarray analysis techniques as described in Sambrook Molecular Cloning a Laboratory Manual 2001 and Baldi, P., and Hatfield, W.G., DNA Microarrays and Gene Expression 2002.

[0503] Microarray analysis generally begins with extracting and purifying nucleic acid from a biological sample, (e.g. a biopsy or fine needle aspirate) using methods known to the art. For expression and alternative splicing analysis it may be advantageous to extract and/or purify RNA from DNA. It may further be advantageous to extract and/or purify mRNA from other forms of RNA such as tRNA and rRNA. In some embodiments, RNA samples with RIN d5.0 are typically not used for multi-gene microarray analysis, and may instead be used only for single-gene RT-PCR and/or TaqMan assays. Microarray, RT-PCR and TaqMan assays are standard molecular techniques well known in the relevant art. TaqMan probe-based assays are widely used in real-time PCR including gene expression assays, DNA quantification and SNP genotyping.

[0504] Various kits can be used for the amplification of nucleic acid and probe generation of the subject methods. In some embodiments, Ambion WT-expression kit can be used. Ambion WT-expression kit allows amplification of total RNA directly without a separate ribosomal RNA (rRNA) depletion step. With the Ambion® WT Expression Kit, samples as small as 50 ng of total RNA can be analyzed on Affymetrix® GeneChip® Human, Mouse, and Rat Exon and Gene 1.0 ST Arrays. In addition to the lower input RNA requirement and high concordance between the Affymetrix® method and TaqMan® real-time PCR data, the Ambion® WT Expression Kit provides a significant increase in sensitivity. For example, a greater number of probe sets detected above background can be obtained at the exon level with the Ambion® WT Expression Kit as a result of an increased signal-to-noise ratio. Ambion WT-expression kit may be used in combination with additional Affymetrix labeling kit.

[0505] In some embodiments, AmpTec Trinucleotide Nano mRNA Amplification kit (6299-A15) can be used in the subject methods. The ExpressArt® TRinucleotide mRNA amplification Nano kit is suitable for a wide range, from 1 ng to 700 ng of input total RNA. According to the amount of input total RNA and the required yields of a RNA, it can be used for 1-round (input >300 ng total RNA) or 2-rounds (minimal input amount 1 ng total RNA), with a RNA yields in the range of >10 mg. AmpTec’s proprietary TRinucleotide priming technology results in preferential amplification of mRNAs (independent of the universal eukaryotic 3’-poly(A)-sequence), combined with selection against rRNAs. This kit can be used in combination with cDNA conversion kit and Affymetrix labeling kit.

[0506] In some embodiments, gene expression levels can be obtained or measured in an individual without first obtaining a sample. For example, gene expression levels may be determined in vivo, that is in the individual. Methods for determining gene expression levels in vivo are known to the art and include imaging techniques such as CAT, MRI; NMR; PET; and optical, fluorescence, or biophotonic imaging of protein or RNA levels using antibodies or molecular beacons. Such methods are described in US 2008/0044824, US 2008/0131892, herein incorporated by reference. Additional methods for in vivo molecular profiling are contemplated to be within the scope of the present invention.

[0507] Provided herein are methods for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of one, two, three or more RNA transcripts (e.g., pre-mRNA or mRNA transcripts or isoforms thereof) of one, two, three or more genes.

[0508] In one embodiment, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of an RNA transcript, comprising: (a) contacting a cell with an SMSM compound or a pharmaceutically acceptable salt thereof, and (b) determining the amount of the RNA transcript produced by the cell, wherein an alteration in the amount of the RNA transcript in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof relative to the amount of the RNA transcript in the absence of an SMSM compound or a pharmaceutically acceptable salt thereof or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript. In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of an RNA transcript (e.g., an mRNA transcript), comprising: (a) contacting a first cell with an SMSM compound or a pharmaceutically acceptable salt thereof, (b) contacting a second cell with a negative control (e.g., a vehicle control, such as PBS or DMSO); and (c) determining the amount of the RNA transcript produced by the first cell and the second cell; and (d) comparing the amount of the RNA transcript produced by the first cell to the amount of the RNA transcript expressed by the second cell, wherein an alteration in the amount of the RNA transcript produced by the first cell relative to the amount of the RNA transcript produced by the second cell indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript. In some embodiments, the contacting of the cell with the compound occurs in cell culture. In other embodiments, the contacting of the cell with the compound occurs in a subject, such as a non-human animal subject. In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of an RNA transcript (e.g., an mRNA transcript), comprising: (a) culturing a cell in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof; and (b) determining the amount of the two or more RNA transcripts splice variants produced by the cell, wherein an alteration in the amount of the two or more RNA transcripts in the presence of the compound relative to the amount of the two or more RNA transcripts splice variants in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of the RNA transcript.

[0509] In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of an RNA transcript (e.g., an mRNA transcript), comprising: (a) culturing a cell in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof; (b) isolating two or more RNA transcript splice variants from the cell after a certain period of time; and (c) determining the amount of the two or more RNA transcript splice variants produced by the cell, wherein an alteration in the amount of the two or more RNA transcript in the presence of the compound relative to the amount of the two or more RNA transcript splice variants in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of the RNA transcript. In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of an RNA transcript (e.g., an mRNA transcript), comprising (a) culturing a first cell in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof; (b) culturing a second cell in the presence of a negative control (e.g., a vehicle control, such as PBS or DMSO); (c) isolating two or more RNA transcript splice variants produced by the first cell and isolating two or more RNA transcript splice variants produced by the second cell; (d) determining the amount of the two or more RNA transcript splice variants produced by the first cell and the second cell; and (e) comparing the amount of the two or more RNA transcript splice variants produced by the first cell to the amount of the two or more RNA transcript splice variants produced by the second cell, wherein an alteration in the amount of the two or more RNA transcript splice variants produced by the first cell relative to the amount of the two or more RNA transcript splice variants produced by the second cell indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of the RNA transcript.

[0510] In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of an RNA transcript (e.g., an mRNA transcript), comprising: (a) contacting a cell-free system with an SMSM compound or a pharmaceutically acceptable salt thereof, and (b) determining the amount of the RNA transcript produced by the cell- free system, wherein an alteration in the amount of the RNA transcript in the presence of the compound relative to the amount of the RNA transcript in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript. In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of an RNA transcript (e.g., an mRNA transcript), comprising: (a) contacting a first cell-free system with an SMSM compound or a pharmaceutically acceptable salt thereof, (b) contacting a second cell-free system with a negative control (e.g., a vehicle control, such as PBS or DMSO); and (c) determining the amount of the RNA transcript produced by the first cell-free system and the second cell-free system; and (d) comparing the amount of the RNA transcript produced by the first cell-free system to the amount of the RNA transcript expressed by the second cell-free system, wherein an alteration in the amount of the RNA transcript produced by the first cell- free system relative to the amount of the RNA transcript produced by the second cell-free system indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript. In some embodiments, the cell-free system comprises purely synthetic RNA, synthetic or recombinant (purified) enzymes, and protein factors. In other embodiments, the cell-free system comprises RNA transcribed from a synthetic DNA template, synthetic or recombinant (purified) enzymes, and protein factors. In other embodiments, the cell-free system comprises purely synthetic RNA and nuclear extract. In other embodiments, the cell-free system comprises RNA transcribed from a synthetic DNA template and nuclear extract. In other embodiments, the cell-free system comprises purely synthetic RNA and whole cell extract. In other embodiments, the cell-free system comprises RNA transcribed from a synthetic DNA template and whole cell extract. In some embodiments, the cell-free system additionally comprises regulatory RNAs (e.g., microRNAs).

[0511] In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of an RNA transcript (e.g., an mRNA transcript), comprising: (a) contacting a cell-free system with an SMSM compound or a pharmaceutically acceptable salt thereof; and (b) determining the amount of two or more RNA transcript splice variants produced by the cell-free system, wherein an alteration in the amount of the two or more RNA transcript splice variants in the presence of the compound relative to the amount of the two or more RNA transcript splice variants in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of the RNA transcript. In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of an RNA transcript (e.g., an mRNA transcript), comprising: (a) contacting a first cell-free system with an SMSM compound or a pharmaceutically acceptable salt thereof; (b) contacting a second cell-free system with a negative control (e.g., a vehicle control, such as PBS or DMSO); and (c) determining the amount of two or more RNA transcript splice variants produced by the first cell-free system and the second cell-free system; and (d) comparing the amount of the two or more RNA transcript splice variants produced by the first cell-free system to the amount of the RNA transcript expressed by the second cell-free system, wherein an alteration in the amount of the two or more RNA transcript splice variants produced by the first cell-free system relative to the amount of the two or more RNA transcript splice variants produced by the second cell-free system indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the splicing of the RNA transcript. In some embodiments, the cell-free system comprises purely synthetic RNA, synthetic or recombinant (purified) enzymes, and protein factors. In other embodiments, the cell-free system comprises RNA transcribed from a synthetic DNA template, synthetic or recombinant (purified) enzymes, and protein factors. In other embodiments, the cell-free system comprises purely synthetic RNA and nuclear extract. In other embodiments, the cell-free system comprises RNA transcribed from a synthetic DNA template and nuclear extract. In other embodiments, the cell-free system comprises purely synthetic RNA and whole cell extract. In other embodiments, the cell-free system comprises RNA transcribed from a synthetic DNA template and whole cell extract. In some embodiments, the cell-free system additionally comprises regulatory RNAs (e.g., microRNAs).

[0512] In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of an RNA transcript (e.g., an mRNA transcript), comprising: (a) culturing a cell in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof, (b) isolating the RNA transcript from the cell after a certain period of time; and (c) determining the amount of the RNA transcript produced by the cell, wherein an alteration in the amount of the RNA transcript in the presence of the compound relative to the amount of the RNA transcript in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript. In some embodiments, provided herein is a method for determining whether an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of an RNA transcript (e.g., an mRNA transcript), comprising (a) culturing a first cell in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof, (b) culturing a second cell in the presence of a negative control (e.g., a vehicle control, such as PBS or DMSO); (c) isolating the RNA transcript produced by the first cell and isolating the RNA transcript produced by the second cell; (d) determining the amount of the RNA transcript produced by the first cell and the second cell; and (e) comparing the amount of the RNA transcript produced by the first cell to the amount of the RNA transcript produced by the second cell, wherein an alteration in the amount of the RNA transcript produced by the first cell relative to the amount of the RNA transcript produced by the second cell indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript.

[0513] In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a primary cell from a subject. In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a primary cell from a subject with a disease. In specific embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a primary cell from a subject with a disease associated with an aberrant amount of an RNA transcript for a particular gene. In some specific embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a primary cell from a subject with a disease associated with an aberrant amount of an isoform of a particular gene. In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a fibroblast, an immune cell, or a muscle cell. In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a diseased cell.

[0514] In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is from a cell line. In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a cell line derived from a subject with a disease. In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is from a cell line known to have aberrant RNA transcript levels for a particular gene. In specific embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is from a cell line derived from a subject with a disease known to have aberrant RNA transcript levels for a particular gene. In some embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is a diseased cell line. In some specific embodiments, the cell contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof is from a cell line derived from a subject with a disease known to have an aberrant amount of an RNA isoform and/or protein isoform of a particular gene. Non-limiting examples of cell lines include 293, 3T3, 4T1, 721, 9L, A2780, A172, A20, A253, A431, A-549, A-673, ALC, B 16, B35, BCP-1, BEAS-2B, bEnd.3, BHK, BR 293, BT20, BT483, BxPC3, C2C12, C 3 h-10T1/2, C6/36, C6, Cal-27, CHO, COR-L23, COS, COV-434, CML Tl, CMT, CRL7030, CT26, D17, DH82, DU145, DuCaP, EL4, EM2, EM3, EMT6, FM3, H1299, H69, HB54, HB55, HCA2, HEK-293, HeLa, Hepalclc7, HL-60, HMEC, Hs578T, HsS78Bst, HT-29, HTB2, HUVEC, Jurkat, J558L, JY, K562, Ku812, KCL22, KG1, KYOl, LNCap, Ma- Mel, MC-38, MCF-7, MCF-IOA, MDA-MB-231, MDA-MB-468, MDA-MB-435, MDCK, MG63, MOR/0.2R, MONO-MAC 6, MRC5, MTD-1A, NCI-H69, NIH-3T3, NALM- 1, NSO, NW-145, OPCN, OPCT, PNT-1A, PNT-2, Raji, RBL, RenCa, RIN-5F, RMA, Saos-2, Sf21, Sf9, SiHa, SKBR3, SKOV-3, T2, T-47D, T84, THP1, U373, U87, U937, VCaP, Vero, VERY, W138, WM39, WT-49, X63, YAC-1, and YAR cells. In one embodiment, the cells are from a patient.

[0515] In some embodiments, a dose-response assay is performed. In one embodiment, the dose response assay comprises: (a) contacting a cell with a concentration of an SMSM compound or a pharmaceutically acceptable salt thereof; (b) determining the amount of the RNA transcript produced by the cell, wherein an alteration in the amount of the RNA transcript in the presence of the compound relative to the amount of the RNA transcript in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript; (c) repeating steps (a) and (b), wherein the only experimental variable changed is the concentration of the compound or a form thereof; and (d) comparing the amount of the RNA transcript produced at the different concentrations of the compound or a form thereof. In some embodiments, the dose response assay comprises: (a) culturing a cell in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof, (b) isolating the RNA transcript from the cell after a certain period of time; (c) determining the amount of the RNA transcript produced by the cell, wherein an alteration in the amount of the RNA transcript in the presence of the compound relative to the amount of the RNA transcript in the absence of the compound or the presence of a negative control (e.g., a vehicle control such as PBS or DMSO) indicates that an SMSM compound or a pharmaceutically acceptable salt thereof modulates the amount of the RNA transcript; (d) repeating steps (a), (b), and (c), wherein the only experimental variable changed is the concentration of the compound or a form thereof; and (e) comparing the amount of the RNA transcript produced at the different concentrations of the compound or a form thereof. In some embodiments, the dose-response assay comprises: (a) contacting each well of a microtiter plate containing cells with a different concentration of an SMSM compound or a pharmaceutically acceptable salt thereof; (b) determining the amount of an RNA transcript produced by cells in each well; and (c) assessing the change of the amount of the RNA transcript at the different concentrations of the compound or form thereof.

[0516] In some embodiments described herein, the cell is contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof, or a tissue sample is contacted with an SMSM compound or a pharmaceutically acceptable salt thereof, or a negative control for a period of 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 48 hours, 72 hours or more. In other embodiments described herein, the cell is contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof, or a tissue sample is contacted with an SMSM compound or a pharmaceutically acceptable salt thereof, or a negative control for a period of 15 minutes to 1 hour, 1 to 2 hours, 2 to 4 hours, 6 to 12 hours, 12 to 18 hours, 12 to 24 hours, 28 to 24 hours, 24 to 48 hours, 48 to 72 hours.

[0517] In some embodiments described herein, the cell is contacted or cultured with a concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, or a tissue sample is contacted with a concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, wherein the concentration is 0.01 mM, 0.05 mM, 1 mM, 2 mM, 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 50 mM, 75 mM, 100 mM, or 150 mM. In other embodiments described herein, the cell is contacted or cultured with concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, or a tissue sample is contacted with a concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, wherein the concentration is 175 mM, 200 mM, 250 mM, 275 mM, 300 mM, 350 mM, 400 mM, 450 mM, 500 mM, 550 mM 600 mM, 650 mM, 700 mM, 750 mM, 800 mM, 850 mM, 900 mM, 950 mM or 1 mM. In some embodiments described herein, the cell is contacted or cultured with concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, or a tissue sample is contacted with a concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, wherein the concentration is 5 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 150 nM, 200 nM, 250 nM, 300 nM, 350 nM, 400 nM, 450 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, or 950 nM. In some embodiments described herein, the cell is contacted or cultured with concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, or a tissue sample is contacted with a concentration of an SMSM compound or a pharmaceutically acceptable salt thereof, wherein the concentration is between 0.01 mM to 0.1 mM, 0.1 mM to 1 mM, 1 mM to 50 mM, 50 mM to 100 mM, 100 mM to 500 mM, 500 mM to 1 nM, 1 nM to 10 nM, 10 nM to 50 nM, 50 nM to 100 nM, 100 nM to 500 nM, 500 nM to 1000 nM.

[0518] Techniques known to one skilled in the art may be used to determine the amount of an RNA transcript. In some embodiments, the amount of one, two, three or more RNA transcripts is measured using deep sequencing, such as ILLUMINA® RNASeq, ILLUMINA® next generation sequencing (NGS), ION TORRENT™ RNA next generation sequencing, 454™ pyrosequencing, or Sequencing by Oligo Ligation Detection (SOLID™). In other embodiments, the amount of multiple RNA transcripts is measured using an exon array, such as the GENECHIP® human exon array. In some embodiments, the amount of one, two, three or more RNA transcripts is determined by RT-PCR. In other embodiments, the amount of one, two, three or more RNA transcripts is measured by RT-qPCR. Techniques for conducting these assays are known to one skilled in the art.

[0519] In some embodiments, a statistical analysis or other analysis is performed on data from the assay utilized to measure an RNA transcript. In some embodiments, a student t-test statistical analysis is performed on data from the assay utilized to measure an RNA transcript to determine those RNA transcripts that have an alternation in amount in the presence of the compound relative to the amount in the absence of the compound or presence of a negative control. In specific embodiments, the student t-test value of those RNA transcripts with the alternation is 10%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1%. In some specific embodiments, p value of those RNA transcripts with the alternation is 10%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1%. In certain specific embodiments, the student t-test and p values of those RNA transcripts with the alteration are 10%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1% and 10%, 5%, 4%, 3%, 2%, 1%, 0.5% or 0.1%), respectively.

[0520] In some embodiments, a further analysis is performed to determine how an SMSM compound or a pharmaceutically acceptable salt thereof is changing the amount of an RNA transcript. In specific embodiments, a further analysis is performed to determine if an alternation in the amount of an RNA transcript in the presence of an SMSM compound or a pharmaceutically acceptable salt thereof relative the amount of the RNA transcript in the absence of the compound or a form thereof, or the presence of a negative control is due to changes in transcription, splicing, and/or stability of the RNA transcript. Techniques known to one skilled in the art may be used to determine whether an SMSM compound or a pharmaceutically acceptable salt thereof changes, e.g., the transcription, splicing and/or stability of an RNA transcript. [0521] In some embodiments, the stability of one or more RNA transcripts is determined by serial analysis of gene expression (SAGE), differential display analysis (DD), RNA arbitrarily primer (RAP)-PCR, restriction endonuclease-lytic analysis of differentially expressed sequences (READS), amplified restriction fragment- length polymorphism (ALFP), total gene expression analysis (TOGA), RT-PCR, RT-qPCR, high-density cDNA filter hybridization analysis (HDFCA), suppression subtractive hybridization (SSH), differential screening (DS), cDNA arrays, oligonucleotide chips, or tissue microarrays. In other embodiments, the stability of one or more RNA transcripts is determined by Northern blots, RNase protection, or slot blots.

[0522] In some embodiments, the transcription in a cell or tissue sample is inhibited before (e.g., 5 minutes, 10 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 36 hours, 48 hours, or 72 hours before) or after (e.g., 5 minutes, 10 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 36 hours, 48 hours, or 72 hours after) the cell or the tissue sample is contacted or cultured with an inhibitor of transcription, such as a-amanitin, DRB, flavopiridol, triptolide, or actinomycin-D. In other embodiments, the transcription in a cell or tissue sample is inhibited with an inhibitor of transcription, such as a-amanitin, DRB, flavopiridol, triptolide, or actinomycin-D, while the cell or tissue sample is contacted or cultured with an SMSM compound or a pharmaceutically acceptable salt thereof.

[0523] In some embodiments, the level of transcription of one or more RNA transcripts is determined by nuclear run-on assay or an in vitro transcription initiation and elongation assay. In some embodiments, the detection of transcription is based on measuring radioactivity or fluorescence. In some embodiments, a PCR- based amplification step is used.

[0524] In some embodiments, the amount of alternatively spliced forms of the RNA transcripts of a particular gene are measured to see if there is an alteration in the amount of one, two or more alternatively spliced forms of the RNA transcripts of the gene. In some embodiments, the amount of an isoform encoded by a particular gene is measured to see if there is an alteration in the amount of the isoform. In some embodiments, the levels of spliced forms of RNA are quantified by RT-PCR, RT-qPCR, or northern blotting. In other embodiments, sequence-specific techniques may be used to detect the levels of an individual splice form. In some embodiments, splicing is measured in vitro using nuclear extracts. In some embodiments, detection is based on measuring radioactivity or fluorescence. Techniques known to one skilled in the art may be used to measure alterations in the amount of alternatively spliced forms of an RNA transcript of a gene and alterations in the amount of an isoform encoded by a gene.

Biological samples

[0525] A sample, e.g., a biological sample can be taken from a subject and examined to determine whether the subject produces mRNA that is subject to alternative splicing. A biological sample can comprise a plurality of biological samples. The plurality of biological samples can contain two or more biological samples; for examples, about 2-1000, 2-500, 2-250, 2-100, 2-75, 2-50, 2-25, 2-10, 10-1000, 10-500, 10-250, 10-100, 10-75, 10-50, 10-25, 25-1000, 25-500, 25-250, 25-100, 25-75, 25-50, 50-1000, 50-500, 50-250, 50-100, 50-75, 60-70, 100-1000, 100-500, 100-250, 250-1000, 250-500, 500-1000, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, or more biological samples. The biological samples can be obtained from a plurality of subjects, giving a plurality of sets of a plurality of samples. The biological samples can be obtained from about 2 to about 1000 subjects, or more; for example, about 2-1000, 2-500, 2-250, 2-100, 2-50, 2-25, 2- 20, 2-10, 10-1000, 10-500, 10-250, 10-100, 10-50, 10-25, 10-20, 15-20, 25-1000, 25-500, 25-250, 25-100, 25- 50, 50-1000, 50-500, 50-250, 50-100, 100-1000, 100-500, 100-250, 250-1000, 250-500, 500-1000, or at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 68, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, to 1000 or more subjects.

[0526] The biological samples can be obtained from human subjects. The biological samples can be obtained from human subjects at different ages. The human subject can be prenatal (e.g., a fetus), a child (e.g., a neonate, an infant, a toddler, a preadolescent), an adolescent, a pubescent, or an adult (e.g., an early adult, a middle aged adult, a senior citizen). The human subject can be between about 0 months and about 120 years old, or older. The human subject can be between about 0 and about 12 months old; for example, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months old. The human subject can be between about 0 and 12 years old; for example, between about 0 and 30 days old; between about 1 month and 12 months old; between about 1 year and 3 years old; between about 4 years and 5 years old; between about 4 years and 12 years old; about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 years old. The human subject can be between about 13 years and 19 years old; for example, about 13, 14, 15, 16, 17, 18, or 19 years old. The human subject can be between about 20 and about 39 year old; for example, about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, or 39 years old. The human subject can be between about 40 to about 59 years old; for example, about 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, or 59 years old. The human subject can be greater than 59 years old; for example, about 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, or 120 years old. The human subjects can include living subjects or deceased subjects. The human subjects can include male subjects and/or female subjects.

[0527] Biological samples can be obtained from any suitable source that allows determination of expression levels of genes, e.g., from cells, tissues, bodily fluids or secretions, or a gene expression product derived therefrom (e.g., nucleic acids, such as DNA or RNA; polypeptides, such as protein or protein fragments). The nature of the biological sample can depend upon the nature of the subject. If a biological sample is from a subject that is a unicellular organism or a multicellular organism with undifferentiated tissue, the biological sample can comprise cells, such as a sample of a cell culture, an excision of the organism, or the entire organism. If a biological sample is from a multicellular organism, the biological sample can be a tissue sample, a fluid sample, or a secretion. [0528] The biological samples can be obtained from different tissues. The term tissue is meant to include ensembles of cells that are of a common developmental origin and have similar or identical function. The term tissue is also meant to encompass organs, which can be a functional grouping and organization of cells that can have different origins. The biological sample can be obtained from any tissue. Suitable tissues from a plant can include, but are not limited to, epidermal tissue such as the outer surface of leaves; vascular tissue such as the xylem and phloem, and ground tissue. Suitable plant tissues can also include leaves, roots, root tips, stems, flowers, seeds, cones, shoots, stobili, pollen, or a portion or combination thereof.

[0529] The biological samples can be obtained from different tissue samples from one or more humans or non- human animals. Suitable tissues can include connective tissues, muscle tissues, nervous tissues, epithelial tissues or a portion or combination thereof. Suitable tissues can also include all or a portion of a lung, a heart, a blood vessel (e.g., artery, vein, capillary), a salivary gland, a esophagus, a stomach, a liver, a gallbladder, a pancreas, a colon, a rectum, an anus, a hypothalamus, a pituitary gland, a pineal gland, a thyroid, a parathyroid, an adrenal gland, a kidney, a ureter, a bladder, a urethra, a lymph node, a tonsil, an adenoid, a thymus, a spleen, skin, muscle, a brain, a spinal cord, a nerve, an ovary, a fallopian tube, a uterus, vaginal tissue, a mammary gland, a testicle, a vas deferens, a seminal vesicle, a prostate, penile tissue, a pharynx, a larynx, a trachea, a bronchi, a diaphragm, bone marrow, a hair follicle, or a combination thereof. A biological sample from a human or non- human animal can also include a bodily fluid, secretion, or excretion; for example, a biological sample can be a sample of aqueous humour, vitreous humour, bile, blood, blood serum, breast milk, cerebrospinal fluid, endolymph, perilymph, female ejaculate, amniotic fluid, gastric juice, menses, mucus, peritoneal fluid, pleural fluid, saliva, sebum, semen, sweat, tears, vaginal secretion, vomit, urine, feces, or a combination thereof. The biological sample can be from healthy tissue, diseased tissue, tissue suspected of being diseased, or a combination thereof.

[0530] In some embodiments, the biological sample is a fluid sample, for example a sample of blood, serum, sputum, urine, semen, or other biological fluid. In certain embodiments the sample is a blood sample. In some embodiments the biological sample is a tissue sample, such as a tissue sample taken to determine the presence or absence of disease in the tissue. In certain embodiments the sample is a sample of thyroid tissue.

[0531] The biological samples can be obtained from subjects in different stages of disease progression or different conditions. Different stages of disease progression or different conditions can include healthy, at the onset of primary symptom, at the onset of secondary symptom, at the onset of tertiary symptom, during the course of primary symptom, during the course of secondary symptom, during the course of tertiary symptom, at the end of the primary symptom, at the end of the secondary symptom, at the end of tertiary symptom, after the end of the primary symptom, after the end of the secondary symptom, after the end of the tertiary symptom, or a combination thereof. Different stages of disease progression can be a period of time after being diagnosed or suspected to have a disease; for example, at least about, or at least, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27 or 28 days; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 weeks; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 months; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49 or 50 years after being diagnosed or suspected to have a disease. Different stages of disease progression or different conditions can include before, during or after an action or state; for example, treatment with drugs, treatment with a surgery, treatment with a procedure, performance of a standard of care procedure, resting, sleeping, eating, fasting, walking, running, performing a cognitive task, sexual activity, thinking, jumping, urinating, relaxing, being immobilized, being emotionally traumatized, being shock, and the like.

[0532] The methods of the present disclosure provide for analysis of a biological sample from a subject or a set of subjects. The subject(s) may be, e.g., any animal (e.g., a mammal), including but not limited to humans, non-human primates, rodents, dogs, cats, pigs, fish, and the like. The present methods and compositions can apply to biological samples from humans, as described herein.

[0533] A biological sample can be obtained by methods known in the art such as the biopsy methods provided herein, swabbing, scraping, phlebotomy, or any other suitable method. The biological sample can be obtained, stored, or transported using components of a kit of the present disclosure. In some cases, multiple biological samples, such as multiple thyroid samples, can be obtained for analysis, characterization, or diagnosis according to the methods of the present disclosure. In some cases, multiple biological samples, such as one or more samples from one tissue type (e.g., thyroid) and one or more samples from another tissue type (e.g., buccal) can be obtained for diagnosis or characterization by the methods of the present disclosure. In some cases, multiple samples, such as one or more samples from one tissue type (e.g., thyroid) and one or more samples from another tissue (e.g., buccal) can be obtained at the same or different times. In some cases, the samples obtained at different times are stored and/or analyzed by different methods. For example, a sample can be obtained and analyzed by cytological analysis (e.g., using routine staining). In some cases, a further sample can be obtained from a subject based on the results of a cytological analysis. The diagnosis of cancer or other condition can include an examination of a subject by a physician, nurse or other medical professional. The examination can be part of a routine examination, or the examination can be due to a specific complaint including, but not limited to, one of the following: pain, illness, anticipation of illness, presence of a suspicious lump or mass, a disease, or a condition. The subject may or may not be aware of the disease or condition. The medical professional can obtain a biological sample for testing. In some cases the medical professional can refer the subject to a testing center or laboratory for submission of the biological sample. The methods of obtaining provided herein include methods of biopsy including fine needle aspiration, core needle biopsy, vacuum assisted biopsy, incisional biopsy, excisional biopsy, punch biopsy, shave biopsy or skin biopsy. In some cases, the methods and compositions provided herein are applied to data only from biological samples obtained by FNA. In some cases, the methods and compositions provided herein are applied to data only from biological samples obtained by FNA or surgical biopsy. In some cases, the methods and compositions provided herein are applied to data only from biological samples obtained by surgical biopsy. A biological sample can be obtained by non-invasive methods, such methods including, but not limited to: scraping of the skin or cervix, swabbing of the cheek, saliva collection, urine collection, feces collection, collection of menses, tears, or semen. The biological sample can be obtained by an invasive procedure, such procedures including, but not limited to: biopsy, alveolar or pulmonary lavage, needle aspiration, or phlebotomy. The method of biopsy can further include incisional biopsy, excisional biopsy, punch biopsy, shave biopsy, or skin biopsy. The method of needle aspiration can further include fine needle aspiration, core needle biopsy, vacuum assisted biopsy, or large core biopsy. Multiple biological samples can be obtained by the methods herein to ensure a sufficient amount of biological material. Methods of obtaining suitable samples of thyroid are known in the art and are further described in the ATA Guidelines for thyroid nodule management (Cooper et al. Thyroid Vol.16 No.22006), herein incorporated by reference in its entirety. Generic methods for obtaining biological samples are also known in the art and further described in for example Ramzy, Ibrahim Clinical Cytopathology and Aspiration Biopsy 2001 which is herein incorporated by reference in its entirety. The biological sample can be a fine needle aspirate of a thyroid nodule or a suspected thyroid tumor. The fine needle aspirate sampling procedure can be guided by the use of an ultrasound, X-ray, or other imaging device.

[0534] In some cases, the subject can be referred to a specialist such as an oncologist, surgeon, or endocrinologist for further diagnosis. The specialist can likewise obtain a biological sample for testing or refer the individual to a testing center or laboratory for submission of the biological sample. In any case, the biological sample can be obtained by a physician, nurse, or other medical professional such as a medical technician, endocrinologist, cytologist, phlebotomist, radiologist, or a pulmonologist. The medical professional can indicate the appropriate test or assay to perform on the sample, or the molecular profiling business of the present disclosure can consult on which assays or tests are most appropriately indicated. The molecular profiling business can bill the individual or medical or insurance provider thereof for consulting work, for sample acquisition and or storage, for materials, or for all products and services rendered.

[0535] A medical professional need not be involved in the initial diagnosis or sample acquisition. An individual can alternatively obtain a sample through the use of an over the counter kit. The kit can contain a means for obtaining said sample as described herein, a means for storing the sample for inspection, and instructions for proper use of the kit. In some cases, molecular profiling services are included in the price for purchase of the kit. In other cases, the molecular profiling services are billed separately.

[0536] A biological sample suitable for use by the molecular profiling business can be any material containing tissues, cells, nucleic acids, genes, gene fragments, expression products, gene expression products, and/or gene expression product fragments of an individual to be tested. Methods for determining sample suitability and/or adequacy are provided. The biological sample can include, but is not limited to, tissue, cells, and/or biological material from cells or derived from cells of an individual. The sample can be a heterogeneous or homogeneous population of cells or tissues. The biological sample can be obtained using any method known to the art that can provide a sample suitable for the analytical methods described herein.

[0537] Obtaining a biological sample can be aided by the use of a kit. A kit can be provided containing materials for obtaining, storing, and/or shipping biological samples. The kit can contain, for example, materials and/or instruments for the collection of the biological sample (e.g., sterile swabs, sterile cotton, disinfectant, needles, syringes, scalpels, anesthetic swabs, knives, curette blade, liquid nitrogen, etc.). The kit can contain, for example, materials and/or instruments for the storage and/or preservation of biological samples (e.g., containers; materials for temperature control such as ice, ice packs, cold packs, dry ice, liquid nitrogen; chemical preservatives or buffers such as formaldehyde, formalin, paraformaldehyde, glutaraldehyde, alcohols such as ethanol or methanol, acetone, acetic acid, HOPE fixative (Hepes-glutamic acid buffer-mediated organic solvent protection effect), heparin, saline, phosphate buffered saline, TAPS, bicine, Tris, tricine, TAPSO, HEPES, TES, MOPS, PIPES, cadodylate, SSC, MES, phosphate buffer; protease inhibitors such as aprotinin, bestatin, calpain inhibitor I and II, chymostatin, E-64, leupeptin, alpha-2-macroglobulin, pefabloc SC, pepstatin, phenylmethanesufonyl fluoride, trypsin inhibitors; DNAse inhibitors such as 2-mercaptoethanol, 2-nitro-5- thicyanobenzoic acid, calcium, EGTA, EDTA, sodium dodecyl sulfate, iodoacetate, etc.; RNAse inhibitors such as ribonuclease inhibitor protein; double-distilled water; DEPC (diethyprocarbonate) treated water, etc.). The kit can contain instructions for use. The kit can be provided as, or contain, a suitable container for shipping. The shipping container can be an insulated container. The shipping container can be self -addressed to a collection agent (e.g., laboratory, medical center, genetic testing company, etc.). The kit can be provided to a subject for home use or use by a medical professional. Alternatively, the kit can be provided directly to a medical professional.

[0538] One or more biological samples can be obtained from a given subject. In some cases, between about 1 and about 50 biological samples are obtained from the given subject; for example, about 1-50, 1-40, 1-30, 1-25, 1-20, 1-15, 1-10, 1-7, 1-5, 5-50, 5-40, 5-30, 5-25, 5-15, 5-10, 10-50, 10-40, 10-25, 10-20, 25-50, 25-40, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 biological samples can be obtained from the given subject. Multiple biological samples from the given subject can be obtained from the same source (e.g., the same tissue), e.g., multiple blood samples, or multiple tissue samples, or from multiple sources (e.g., multiple tissues). Multiple biological samples from the given subject can be obtained at the same time or at different times. Multiple biological samples from the given subject can be obtained at the same condition or different condition. Multiple biological samples from the given subject can be obtained at the same disease progression or different disease progression of the subject. If multiple biological samples are collected from the same source (e.g., the same tissue) from the particular subject, the samples can be combined into a single sample. Combining samples in this way can ensure that enough material is obtained for testing and/or analysis. EXAMPLES

[0539] These examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein. The starting materials and reagents used for the synthesis of the compounds described herein may be synthesized or can be obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Acros Organics, Fluka, and Fischer Scientific.

A. Biological Examples

Example A1: Splicing Assay (MAPTau, MADD, FOXM1)

[0540] Various cells lines are treated with SMSMs described herein. RNA is then isolated, cDNA synthesized, qPCR performed and the levels of various mRNA targets of the SMSMs are determined. In some instances, RNA is isolated, cDNA synthesized, qPCR performed and the levels of mRNA isoforms in the various cell samples is determined.

Materials

[0541] Cells to Ct kit: ThermoFisher, AM1728. TaqMan Gene Expression Master Mix: ThermoFisher, 4369542. PPIA probe/primer: ThermoFisher, Hs03045993_gH, VIC-MGB_PL.

Probe/primer sequences:

[0542] FoxM1

[0543] MADD

[0544] MAPTau

ll l h ( ) b / i

Cells:

[0545] Cells used include 93–T449, A–375, A–673, ASPC–1, BxPC–3, CCL–136, Daoy, DU-145, G-401, Hep-3B, IMR–32, K-562, LP–LoVo, MDA–MB–157, MDA–MB–231–luc, MDA-MB-468, MG–63, Ms751, NCI-H358, PACA–2, PANC–1, PC-3, RGX–MPC–11, RGX–PACA–2, SH-SY5Y, SJSA, SKOV3, SNU–16, SW872 (HTB–92), TOLEDO, T.T, U–118, U–251MG, U–87MG, and Z–138 cells.

[0546] On the day of the experiment, a 96-well plate is seeded with the cell lines of interest. The cells are diluted with full growth media to a concentration of 2.0 x 10 5 cells/mL and 100 mL of cells are added to each well (20,000 cells per well). The cells are treated with a compound immediately after plating.

[0547] The compounds are then added to the cell plate using the HP compound dispenser. In the initial experiment, a top concentration of 10 mM and an 8 point 4-fold dilution scheme is used. The stock compounds are made at a concentration of 5 mM, and the DMSO concentration is set to 0.2%. DMSO is used to normalize all the compound-containing wells and the untreated cells.

[0548] The treated cells are incubated at 37 °C in a 5% CO 2 incubator for the desired amount of time. Plates are placed in a plastic bag with a wet paper towel to prevent evaporation.

[0549] RNA is isolated using the Cells to C T kit (ThermoFisher, AM1728). The cells are washed once with 100 mL cold PBS.50 mL of lysis buffer is added to each well/tube (49.5 mL lysis buffer + 0.5 mL DNase I per well/tube). The lysis reaction is mixed and incubated at room temperature for 5 minutes.5 mL of stop solution is added directly into each cell lysis reaction and mixed by pipetting up and down 5 times. The plates/tubes are incubated at room temperature for 2 minutes then placed on ice if the cDNA synthesis is to be performed immediately. Otherwise, the plates/tubes are stored at -80 °C. cDNA synthesis reactions are then performed. Reverse Transcription (RT) Master mix is prepared according to the table below.

[0550] 40 mL RT master mix is added to PCR tubes or plate wells.10 mL of RNA is added to each tube/well. The RT thermal cycler program is then run and tubes or plate wells are incubated at 37 °C for 1 hour, then at 95 °C for 5 minutes to deactivate the enzyme.

[0551] The qPCR is performed using a QuantStudio 6 instrument (ThermoFisher) and the following cycling conditions and according to the tables below. All samples and standards are analyzed in triplicate. Cycle 1: 2 minutes at 50 °C. Cycle 2: 10 minutes at 95 °C. Cycle 3 (repeat 40 times): 15 seconds at 95 °C, 1 minute at 60 °C.

Isoform 1 or isoform 2 standard samples

[0552] The determined isoform 2 and isoform 1 quantities are then be used to determine the isoform 2: isoform 2 ratio at the various compound concentrations. The PPIA quantities are used in the normalization to account of cell proliferation effects of the compounds.

Standard construction

PPIA standard (5834 bps)

[0553] The G Blocks are inserted into the pCI-neo mammalian expression vector (Promega) at the EcoRI and NotI restriction sites (bolded) using Infusion cloning technology (Clontech). The plasmids are then purified using standard miniprep or maxiprep kits (Macherey Nagel).

Standard curve preparation

[0554] The dilution necessary to make the top standard is calculated. A top concentration 200,000,000 copies/mL of the stock plasmid is prepared in TE buffer. A series of 10-fold dilutions, also in TE, are then made. A total of 5 mL of each standard is used in a qPCR well to generate samples containing 10 9 copies, 10 8 copies, 10 7 copies, 10 6 copies, 10 5 copies, 10 4 copies, 10 3 copies, 10 2 copies, 10 1 copies, and 0 copies.

[0555] An assay to measure the FOXM1 A2 mRNA and FOXM1 BC mRNA, or MADD WT (isoform 4) mRNA and MADD Ex16 (isoform 3) mRNA, or MAPTau 4R mRNA and MAPTau 3R mRNA simultaneously in cell wells is performed. The RNA values are measured relative to DMSO control and also included a housekeeping gene, PPIA to ensure data is consistent. The mRNA values are measured after 24 hours of incubation with the SMSM compounds. The SMSMs dose dependently increased FOXM1 A2 levels while concomitantly decreasing FOXM1 BC levels with EC 50 and IC 50 values in the nanomolar range.

Example A2: SMN2 Splicing Assay - Monitoring expression levels of SMN2 splice variant using real-time quantitative PCR

[0556] Various cells lines are treated with the SMSMs described herein. RNA is then isolated, cDNA synthesized, qPCR performed and the levels of various mRNA targets of the SMSMs are determined. In some instances, RNA is isolated, cDNA synthesized, qPCR performed and the levels of mRNA isoforms in the various cell samples are determined.

Materials

[0557] Cells to Ct kit: ThermoFisher, AM1728. TaqMan Gene Expression Master Mix: ThermoFisher, 4369542. PPIA probe/primer: ThermoFisher, Hs03045993_gH, VIC-MGB_PL.

Probe/primer sequences:

[0558] The table below summarizes primers that can be used.

[0559] SMA type I patient cells (GM03813 (Coriell))

Protocol

[0560] On the day of the experiment, a 96-well plate is seeded with the cell lines of interest. The cells are diluted with full growth media to a concentration of 2.0 x 10 5 cells/mL and 100 mL of cells are added to each well (20,000 cells per well). The cells are treated with a compound immediately after plating.

[0561] The compounds are then added to the cell plate using the HP compound dispenser. In the initial experiment, a top concentration of 10 mM and an 8 point 4-fold dilution scheme is used. The stock compounds are made at a concentration of 5 mM, and the DMSO concentration is set to 0.2%. DMSO is used to normalize all the compound-containing wells and the untreated cells.

[0562] The treated cells are incubated at 37 °C in a 5% CO 2 incubator for the desired amount of time. Plates are placed in a plastic bag with a wet paper towel to prevent evaporation.

[0563] RNA is isolated using the Cells to C T kit (ThermoFisher, AM1728). The cells are washed once with 100 mL cold PBS.50 mL of lysis buffer is added to each well/tube (49.5 mL lysis buffer + 0.5 mL DNase I per well/tube). The lysis reaction is mixed and incubated at room temperature for 5 minutes.5 mL of stop solution is added directly into each cell lysis reaction and mixed by pipetting up and down 5 times. The plates/tubes are incubated at room temperature for 2 minutes then placed on ice if the cDNA synthesis is to be performed immediately. Otherwise, the plates/tubes are stored at -80 °C.

[0564] cDNA synthesis reactions are then performed. 40 mL RT master mix is added to PCR tubes or plate wells.10 mL of RNA is added to each tube/well. The RT thermal cycler program is then run and tubes or plate wells are incubated at 37 °C for 1 hour, then at 95 °C for 5 minutes to deactivate the enzyme.

[0565] The qPCR is performed using a QuantStudio 6 instrument (ThermoFisher) and the following cycling conditions and according to the tables below. All samples and standards are analyzed in triplicate. Cycle 1: 2 minutes at 50 °C. Cycle 2: 10 minutes at 95 °C. Cycle 3 (repeat 40 times): 15 seconds at 95 °C, 1 minute at 60 °C.

SMN2 FL or SMN2 D7 standard samples

[0566] The determined SMN2 D7 and SMN2 FL quantities are then be used to determine the SMN2 D7 :SMN2 FL ratio at the various compound concentrations. The PPIA quantities are used in the normalization to account of cell proliferation effects of the compounds.

Standard construction

PPIA standard (5834 bps)

G Block sequence (IDT DNA)

SMN2 FL standard G Block sequence (IDT DNA) are used

SMN2 D7 standard G Block sequence (IDT DNA) are used

[0567] The G Blocks are inserted into the pCI-neo mammalian expression vector (Promega) at the EcoRI and NotI restriction sites (bolded) using Infusion cloning technology (Clontech). The plasmids are then purified using standard miniprep or maxiprep kits (Macherey Nagel).

Standard curve preparation

[0568] The dilution to make the top standard is calculated. A top concentration 200,000,000 copies/mL of the stock plasmid is prepared in TE buffer. A series of 10-fold dilutions, also in TE, are then made. A total of 5 mL of each standard is used in a qPCR well to generate samples containing 10 9 copies, 10 8 copies, 10 7 copies, 10 6 copies, 10 5 copies, 10 4 copies, 10 3 copies, 10 2 copies, 10 1 copies, and 0 copies.

[0569] An assay to measure the SMN2 FL mRNA and SMN2 D7 mRNA simultaneously in cell wells is performed. The RNA values are measured relative to DMSO control and also included a housekeeping gene, PPIA to ensure data is consistent. The mRNA values are measured after 24 hours of incubation with the SMSM compounds. The SMSMs dose dependently increased the SMN2 FL values while concomitantly decreasing the SMN2 D7 values at the same with EC 50 and IC 50 values in the nanomolar range.

[0570] Additionally, to monitor expression levels of SMN2 splice variant using real-time quantitative PCR, SMA type I patient cells (GM03813 (Coriell)) are plated at 5,000 cells/well in 200 ml Dulbecco’s modified Eagle’s medium (DMEM) with GlutaMAX and 10% fetal bovine serum (FBS) (Life Technologies, Inc.) in 96- well plates, and incubated for 6 hours in a cell culture incubator. Cells are then treated with SMSMs at different concentrations (0.5% DMSO) in duplicate for 24 hours. After removal of the supernatant, cells are lysed in Cells-To-Ct lysis buffer (Life Technologies, Inc.) according to the manufacturer’s recommendations. The mRNA levels of SMN2 FL, SMN2 D7 are quantified using Taqman-based RT-qPCR and SMN2-specific primers and probes. The SMN2 forward and reverse primers are each used at a final concentration of 0.4 mM. The SMN2 probe is used at a final concentration of 0.15 mM. RT-qPCR is carried out at the following temperatures for indicated times: Step 1: 48°C (15 min); Step 2: 95 °C (10 min); Step 3: 95 °C (15 sec); Step 4: 60 °C (1 min); Steps 3 and 4 are repeated for 40 cycles. The Ct values for each mRNA are converted to mRNA abundance using actual PCR efficiencies.

Example A3: IKBKAP Splicing Assay

[0571] Various cells lines are treated with the SMSMs described herein. RNA is then isolated, cDNA synthesized, qPCR performed and the levels of IKBKAP targets of the SMSMs are determined. Materials

[0572] Cells to Ct kit: ThermoFisher, AM1728. TaqMan Gene Expression Master Mix: ThermoFisher, 4369542. PPIA probe/primer: ThermoFisher, Hs03045993_gH, VIC-MGB_PL.

Probe/primer sequences:

[0573] IKBKAP

IKBKAP WT probe/primer: IDT DNA

Protocol

[0574] On the day of the experiment, a 96-well plate is seeded with the cell lines of interest. The cells are diluted with full growth media to a concentration of 2.0 x 10 5 cells/mL and 100 mL of cells are added to each well (20,000 cells per well). The cells are treated with a compound immediately after plating.

[0575] The compounds are then added to the cell plate using the HP compound dispenser. In the initial experiment, a top concentration of 10 mM and an 8 point 4-fold dilution scheme is used. The stock compounds are made at a concentration of 5 mM, and the DMSO concentration is set to 0.2%. DMSO is used to normalize all the compound-containing wells and the untreated cells.

[0576] The treated cells are incubated at 37 °C in a 5% CO 2 incubator for the desired amount of time. Plates are placed in a plastic bag with a wet paper towel to prevent evaporation.

[0577] RNA is isolated using the Cells to C T kit (ThermoFisher, AM1728). The cells are washed once with 100 mL cold PBS.50 mL of lysis buffer is added to each well/tube (49.5 mL lysis buffer + 0.5 mL DNase I per well/tube). The lysis reaction is mixed and incubated at room temperature for 5 minutes.5 mL of stop solution is added directly into each cell lysis reaction and mixed by pipetting up and down 5 times. The plates/tubes are incubated at room temperature for 2 minutes then placed on ice if the cDNA synthesis is to be performed immediately. Otherwise, the plates/tubes are stored at -80 °C.

[0578] cDNA synthesis reactions are then performed. 40 mL RT master mix is added to PCR tubes or plate wells.10 mL of RNA is added to each tube/well. The RT thermal cycler program is then run and tubes or plate wells are incubated at 37 °C for 1 hour, then at 95 °C for 5 minutes to deactivate the enzyme.

[0579] The qPCR is performed using a QuantStudio 6 instrument (ThermoFisher) and the following cycling conditions and according to the tables below. All samples and standards are analyzed in triplicate. Cycle 1: 2 minutes at 50 °C. Cycle 2: 10 minutes at 95 °C. Cycle 3 (repeat 40 times): 15 seconds at 95 °C, 1 minute at 60 °C.

IKBKAP FL or IKBKAP D20 standard samples

[0580] The determined IKBKAP FL and IKBKAP D20 isoform quantities are then used to determine the IKBKAP FL : IKBKAP D20 ratio at increasing SMSM compound concentrations. The PPIA quantities are used in the normalization to account of cell proliferation effects of the compounds.

Standard construction

PPIA standard (5834 bps)

[0581] The G Blocks are inserted into the pCI-neo mammalian expression vector (Promega) at the EcoRI and NotI restriction sites (bolded) using Infusion cloning technology (Clontech). The plasmids are then purified using standard miniprep or maxiprep kits (Macherey Nagel).

Standard curve preparation

[0582] The dilution necessary to make the top standard is calculated. A top concentration 200,000,000 copies/mL of the stock plasmid is prepared in TE buffer. A series of 10-fold dilutions, also in TE, are then made. A total of 5 mL of each standard is used in a qPCR well to generate samples containing 10 9 copies, 10 8 copies, 10 7 copies, 10 6 copies, 10 5 copies, 10 4 copies, 10 3 copies, 10 2 copies, 10 1 copies, and 0 copies.

[0583] An assay to measure the IKBKAP FL mRNA and IKBKAP D20 mRNA simultaneously in cell wells is performed. The RNA values are measured relative to DMSO control and also included a housekeeping gene, PPIA to ensure data is consistent. The mRNA values are measured after 24 hours of incubation with the SMSM compounds.

Example A4: Cell viability and proliferation

[0584] Small molecule splicing modulators are tested in a dose-response assay using different cancer cell lines. Cells are first plated in 96-well plastic tissue culture plates (10,000 cells per well). The cells are treated with 500 nM of SMSM or vehicle alone (DMSO) for 48 hours. Following treatment, the cells are washed with PBS, stained with a crystal violet staining solution, and allowed to dry for 48-72 hrs. After drying, sodium citrate buffer is added to each well and allowed to incubate for 5 min at room temperature. The absorbance is measured at 450nM using a microplate reader (Biorad; Hercules, CA). The relative cell proliferation for each of the cancer cell lines is determined.

[0585] To measure cell viability, cells are plated in 96-well plastic tissue culture plates at a density of 5xl0 3 cells/well. Twenty-four hours after plating, cells are treated with various SMSMs. After 72 hours, the cell culture media are removed and plates are stained with 100 mL/well of a solution containing 0.5% crystal violet and 25% methanol, rinsed with deionized water, dried overnight, and resuspended in 100 mL citrate buffer (0.1 M sodium citrate in 50% ethanol) to assess plating efficiency. Intensity of crystal violet staining, assessed at 570 nm and quantified using a Vmax Kinetic Microplate Reader and Softmax software (Molecular Devices Corp., Menlo Park, CA), are directly proportional to cell number. Data are normalized to vehicle-treated cells and are presented as the mean ± SE from representative experiments. SMSMs that are effective are determined for various cells lines.

[0586] Small molecule splicing modulators are tested in a dose-response assay using cancer cells and NHDF cells.

[0587] Cancer cells or NHDF cells are first plated in 96-well plastic tissue culture plates (10,000 cells per well). The cells are treated with vehicle alone (DMSO), or increasing concentrations of SMSM compounds for 72 h. Following treatment, cell proliferation is determined using a crystal violet assay. The relative cell proliferation at each concentration is determined.

Example A5: Monitoring expression levels of FOXM1 splice variants using real-time quantitative PCR

[0588] Human fibroblasts are plated at 10,000 cells/well in 200 mL DMEM with GlutaMAX and 10% FBS in 96-well plates in a cell culture incubator (37 °C, 5% CO2, 100% relative humidity). Cells are then treated with SMSMs at different concentrations (0.1-1000 nM, each in 0.5% DMSO) in triplicate for 24 hours. RNA extraction is performed as per instructions in the Cells-to-CT™ Kits (Ambion®, Applied Biosystems). RNA samples are frozen at -20 °C until further analysis. Relative expression levels of full-length FOXM1 (FOXM1 FL ) or FOXM1 lacking exon VIIa (FOXM1 DVIIa ) with GAPDH for internal control, is measured using one-step multiplex reverse transcription-polymerase chain reaction (RT-PCR). TaqMan® FAM probes are used for relative quantitation of FOXM1 FL or FOXM1 DVIIa expression levels and TaqMan® VIC probes are used for relative quantitation of human GAPDH levels. The fidelity of the amplification methods is determined using the ''Ct relative quantification method for quantitative PCR.

Example A6: Maximum tolerable dose study

[0589] Survival of mice after administration of SMSMs after 10 or 11 days is assessed.

[0590] Tolerance of the drug treatments is determined by measuring the weight of the mice during the period of drug administration. Body weight is measured prior to tumor inoculation and prior to the treatment administration and then daily. The changes in the final weight of the mice for the SMSM treatments are determined.

Example A7: Dose range and time course studies [0591] Dose range and time course studies comparing anti-neoplastic effects of SMSMs against vehicle are conducted.

[0592] Exemplary experimental groups used for this study are shown in the table below.

[0593] Female NCrNu mice are used. Age range of enrolment is 7-10 weeks. A total of 75 animals are for the studies.

[0594] Each mouse are inoculated into a right flank with the single cell suspension of 95% viable tumor cells (5 x 10 6 cells/ mouse) in serum-free RPMI 1640 Media for tumor development. Treatments are administered when mean tumor size reached approximately 75 mm 3 .

[0595] An acclimation period of a minimum of 72 hrs is allowed between animal receipt and tumor inoculation in order to accustom the animals to the laboratory environment. Immunodeficient NCrNu mice are maintained in a pathogen-free environment. Animals are fed a diet of Irradiated Mouse pellet feed Purina rodent diet #5053 (Fisher Feeds, Bound Brook, NJ) and chlorinated water from a reverse osmosis (RO) system (4-6 ppm).

[0596] Before commencement of treatment, all animals are weighed and assigned to treatment groups using a randomization procedure. Mice are randomized into groups based upon their tumor sizes to ensure that each group had approximately the same mean tumor size and range of tumor size.

[0597] After inoculation, the animals are checked daily for morbidity and mortality. At the time of routine monitoring, the animals are checked for any effects of tumor growth on normal behavior such as mobility, food and water consumption, body weight gain/loss, eye/hair matting and any other abnormal effects. Deaths and observed clinical signs are recorded. Animals that are observed to be in a continuing deteriorating condition or bearing a tumor exceeding 2,000 mm 3 in size are euthanized.

[0598] Body weight is measured prior to tumor inoculation and prior to the treatment administration and then daily. Tumor size are measured 2 -3 times per week in two dimensions using a caliper, and the volume are expressed in mm 3 using the formula: V = 0.5 x a x b 2 where a and b are the long and short diameters of the tumor, respectively.

[0599] Studies are terminated when the tumor size in the vehicle treated group reached 2,000mm 3 . Each mouse is bled at 2 hrs after the last dose and at least 50 ml of plasma are collected from each mouse. All of the collected plasma samples and retainer dosing solutions for each dose level are used for bioanalytical measurements. All tumors are also collected and weighed. One necrosis-free tumor fragment of approximately 50 mg is taken from each tumor and flash-frozen for RNA isolation. The remaining tumor is flash frozen for PK analysis.

Example A8: In vivo SMSM treatment effect on inhibition of tumor growth

[0600] Studies are performed to assess the effects of in vivo SMSM treatment on various tumors. Studies are also performed to assess the effects of in vivo SMSM treatment on mRNA levels. Immunocompromised nude mice with pre-existing cancer xenografts are treated with vehicle or SMSMs. Tumor tissues from subcutaneous xenografts are broken into a powder using a BioPulverizer (Biospec Products, Inc.). After SMSM treatment, mRNA is isolated from the xenografts and is analyzed by qRT-PCR.

[0601] Tumor size is measured 2 times per week in two dimensions using a caliper. Studies are terminated when the tumor size in the vehicle treated group reached 2,000 mm 3 . Each mouse is bled at 2 hrs after the last dose and at least 50 ml of plasma is collected from each mouse. All of the collected plasma samples and retainer dosing solutions for each dose level are used for bioanalytical measurements. All tumors are also collected and weighed. One necrosis-free tumor fragment of approximately 50 mg is taken from each tumor and flash-frozen for RNA isolation. The remaining tumor is flash frozen for PK analysis.

[0602] The effects of in vivo SMSM treatments on pre-existing subcutaneous cancer xenografts are assessed. For these in vivo experiments, 1 x 10 6 cancer cells (cells re-suspended in 100 ml PBS are subcutaneously injected into the flanks of nude mice. When the tumor reached approximately 100 mm 3 (volume=(¾)(p)(length/2)(width/2) 2 ), the SMSM treatments are initiated.

Example A9: Quantitative Splicing Assay (HTT)

[0603] GM04724 (CAG 70/20) Huntington’s disease patient lymphoblasts (Coriell) are plated in 96-wel v- bottom plates at 50,000 cells/well. Immediately after plating, cells are dosed with compound for 24 h at concentrations ranging from 2.5uM to 0.15nM (0.1% DMSO). Treated cells are lysed and cDNA synthesized using the Fast Advanced Cells-to-Ct kit (Thermofisher A35378) according to the manufacturer’s instructions. 2 uL of each cDNA are used in qPCR reactions to confirm the compound-induced inclusion of a cryptic exon within intron 49 of the Huntingtin (HTT) transcripts. The qPCR reactions are prepared in 384-well plates in 10 uL volume, using TaqMan™ Fast Advanced Master Mix [ThermoFisher; 4444965] with primers and probes shown in the table below. Reactions are run in a Quant Studio 6 qPCR instrument with default settings. Probe/primer sequences:

HTT 49b FAM

Example A10: mHTT protein assay

[0604] Compounds are tested on GM04724 (CAG 70/20) Huntington’s disease patient lymphoblast cells at doses ranging from 10 mM to 0.6 nM.4,500 cells/well are seeded in 384 well plates. One plate replica is carried out for parallel viability testing by CellTiter Glo (CTG). Compounds are incubated for 48 hours. mHTT protein levels are assessed by the 2B7–MW1 assay via Mesoscale Discovery (MSD) as previously reported (Macdonald et al., 2014). The antibody pair is comprised of previously characterized monoclonals (2B7 and MW1) interrogating two regions for HTT conformation and biological properties: the N17 domain and the polyQ domain (Baldo et al., 2012; Ko et., 2001).2B7–MW1 is dependent on subject/animal specific levels of HTT at the time of treatment.2B7–MW1 is dependent on polyQ expansion (e.g., the higher the expansion the higher the signal) and on mHTT size (e.g., a similar polyQ will give higher signal with smaller HTT size). The viability readout is carried out by CTG according to the manufacturer’s instructions.

Example A11: Quantitative Splicing Assay (SMN)

[0605] Spinal muscular atrophy (SMA) patient fibroblasts (GM03813, Coriell) are plated in 96-well plates at 50,000 cells/well. Immediately after plating, cells are dosed with compounds for 24 h at concentrations ranging from 2.5mM to 0.6nM (0.1% DMSO). Treated cells are lysed and cDNA synthesized using the Fast Advanced Cells-to-Ct kit (Thermofisher A35378) according to the manufacturer’s instructions.2 mL of each cDNA are used in qPCR reactions. The qPCR reactions are prepared in 384-well plates in 10 mL volume, using TaqMan™ Fast Advanced Master Mix (ThermoFisher; 4444965) with primers and probes shown in the table below. Reactions are run in a Quant Studio 6 qPCR instrument with default settings.

Probe/primer sequences:

Example A12: SMN protein assay [0606] Compounds are tested on spinal muscular atrophy (SMA) patient fibroblasts (GM03813, Coriell) at doses ranging from 2.5 mM to 0.6 nM.7000 cells/well are seeded in 96-well plates. Compounds are incubated for 48 hours and the cells were lysed with 100 mL of lysis buffer.20 mL of lysate is used for SMN protein measurement by Mesoscale Discovery (MSD) assay developed by PharmOptima (Michigan). A standard curve prepared with SMN protein ranging from 1mg/ml to 19.5pg/ml is used in each MSD plate to calculate the absolute SMN protein amount in each sample.

[0607] One plate with 700 cells/well is prepared for parallel viability testing by Cell Tier Glo reagents (Promega, G7572/G7573 (CTG). The viability readout is carried out according to the manufacturer’s instructions.

Example A13: Assessment of blood-brain-barrier (BBB) penetration potential via an MDCK-MDR1 Permeability assay

[0608] The permeability of compounds is assessed for BBB penetration potential by use of an MDCK-MDR1 assay (Catalog EA203) performed by Absorption Systems, Exton PA. See,“Evaluation of the MDR- MDCK cell line as a permeability screen for the blood-brain barrier,” Wang, Q. Rager, J.D.;

Weinstein, K.; Kardos, P.S.; Dobson, G.L.; Li, J.; Hidalgo, I.J.

[0609] Experimental Procedure: MDR1-MDCK cell monolayers are grown to confluence on collagen-coated, microporous membranes in 12-well assay plates. The permeability assay buffer is Hanks’ balanced salt solution containing 10 mM HEPES and 15 mM glucose at a pH of 7.4. The buffer in the receiver chamber also contained 1% bovine serum albumin. The dosing solution concentration is 5 mM of test article in the assay buffer. Cell monolayers are dosed on the apical side (A-to-B) or basolateral side (B-to-A) and incubated at 37°C with 5% CO 2 in a humidified incubator. Samples are taken from the donor and receiver chambers at 120 minutes. Each determination is performed in duplicate. The flux of lucifer yellow is also measured post-experimentally for each monolayer to ensure no damage is inflicted to the cell monolayers during the flux period. All samples are assayed by LC-MS/MS using electrospray ionization. Analytical conditions are outlined below.

[0610] The apparent permeability (Papp) and percent recovery are calculated as follows:

Papp = (dC r /dt) × V r /(A × C A ) (1)

Percent Recovery = 100 × ((V al

r × C final

r ) + (Vd × C fin

d ))/(Vd × CN) (2)

where,

dCr /dt is the slope of the cumulative concentration in the receiver compartment versus time in mM s -1 ; V r is the volume of the receiver compartment in cm 3 ;

V d is the volume of the donor compartment in cm 3 ;

A is the area of the insert (1.13 cm 2 for 12-well);

CA is the average of the nominal dosing concentration and the measured 120 minute donor concentration in mM;

C N is the nominal concentration of the dosing solution in mM; C final

r is the cumulative receiver concentration in mM at the end of the incubation period;

C final

d is the concentration of the donor in mM at the end of the incubation period.

Efflux ratio (ER) is defined as Papp (B-to-A) / Papp (A-to-B).

[0611] Analytical Method: Liquid Chromatography. Column: Waters ACQUITY UPLC BEH Phenyl 30 × 2.1 mm, 1.7 mm; M.P. Buffer: 25 mM ammonium formate buffer, pH 3.5; Aqueous Reservoir (A): 90% water, 10% buffer; Organic Reservoir (B): 90% acetonitrile, 10% buffer; Flow Rate: 0.7 mL/minute; Gradient Program:

Total Run Time: 1.00 minute

Autosampler: 2 mL injection volume

Wash1: water/methanol/2-propanol:1/1/1; with 0.2% formic acid

Wash2: 0.1% formic acid in water

Brain Penetration

Potential Classification: A-B P app ³ 3.0 and Efflux Ratio < 3.0: High

A -B Papp ³ 3.0 and 10 > Efflux Ratio ³ 3.0: Moderate

A -B Papp ³ 3.0 and Efflux Ratio ³ 10: Low

A-B P app < 3.0: Low

B. Chemical Synthesis Examples

[0612] Compounds described herein can be synthesized using standard synthetic techniques or using methods known in the art in combination with methods described herein. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology can be employed. Compounds can be prepared using standard organic chemistry techniques such as those described in, for example, March’s Advanced Organic Chemistry, 6th Edition, John Wiley and Sons, Inc. Alternative reaction conditions for the synthetic transformations described herein may be employed such as variation of solvent, reaction temperature, reaction time, as well as different chemical reagents and other reaction conditions. The starting materials can be available from commercial sources or can be readily prepared. By way of example only, provided are schemes for preparing the Examples described herein.

[0613] The following abbreviations are used: DCM– dichloromethane; DIPEA– N,N-diisopropylethylamine; DMSO - dimethyl sulfoxide; DMF - N,N-dimethylformamide; EDCI - N-(3-Dimethylaminopropyl)-NĄ - ethylcarbodiimide; HOBt̢ 1-hydroxybenzotriazole; THF- tetrahydrofuran; Et 2 O - diethyl ether; EtOAc - ethyl acetate; EtOH - ethyl alcohol; LCMS– liquid chromatography mass spectrometer; Ms– mesylate; MeCN– acetonitrile; MeOH - methyl alcohol; MTBE – methyl tert-butyl ether ;SFC - supercritical fluid chromatography; TMSCl– trimethylsilyl chloride; h– hour; min– minute; rt– room temperature (22-25 °C); g– grams; mL - milliliters; mg– milligrams; mmol– millimoles.

[0614] Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, “Synthetic Organic Chemistry”, John Wiley & Sons, Inc., New York; S. R. Sandler et al.,“Organic Functional Group Preparations,” 2nd Ed., Academic Press, New York, 1983; H. O. House,“Modern Synthetic Reactions”, 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist,“Heterocyclic Chemistry”, 2nd Ed., John Wiley & Sons, New York, 1992; J. March,“Advanced Organic Chemistry: Reactions, Mechanisms and Structure”, 4th Ed., Wiley Interscience, New York, 1992. Additional suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, Fuhrhop, J. and Penzlin G.“Organic Synthesis: Concepts, Methods, Starting Materials”, Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3527–29074–5; Hoffman, R.V.“Organic Chemistry, An Intermediate Text” (1996) Oxford University Press, ISBN 0–19–509618–5; Larock, R. C.“Comprehensive Organic Transformations: A Guide to Functional Group Preparations” 2nd Edition (1999) Wiley–VCH, ISBN: 0–471–19031–4; March, J.“Advanced Organic Chemistry: Reactions, Mechanisms, and Structure” 4th Edition (1992) John Wiley & Sons, ISBN: 0–471– 60180–2; Otera, J. (editor)“Modern Carbonyl Chemistry” (2000) Wiley–VCH, ISBN: 3–527–29871–1; Patai, S.“Patai’s 1992 Guide to the Chemistry of Functional Groups” (1992) Interscience ISBN: 0–471–93022–9; Solomons, T. W. G.“Organic Chemistry” 7th Edition (2000) John Wiley & Sons, ISBN: 0–471–19095–0; Stowell, J.C.,“Intermediate Organic Chemistry” 2nd Edition (1993) Wiley–Interscience, ISBN: 0–471–57456– 2;“Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann’s Encyclopedia” (1999) John Wiley & Sons, ISBN: 3–527–29645–X, in 8 volumes;“Organic Reactions” (1942–2000) John Wiley & Sons, in over 55 volumes; and“Chemistry of Functional Groups” John Wiley & Sons, in 73 volumes.

[0615] In the reactions described, it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, in order to avoid their unwanted participation in reactions. A detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure).

[0616] Examples can be made using known techniques and further chemically modified, in some embodiments, to facilitate intranuclear transfer to, e.g., a splicing complex component, a spliceosome or a pre–mRNA molecule. One of ordinary skill in the art will appreciate the standard medicinal chemistry approaches for chemical modifications for intranuclear transfer (e.g., reducing charge, optimizing size, and/or modifying lipophilicity). [0617] In some embodiments, the compounds made in the examples below are made from racemic starting materials (and/or intermediates) and separated into the individual enantiomers by chiral chromatography as final products or intermediates. Unless otherwise stated, it is understood that the absolute configuration of the separated intermediates and final compounds as drawn is arbitrarily assigned and was not determined.

[0618] The following examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein. The starting materials and reagents used for the synthesis of the compounds described herein may be synthesized or can be obtained from commercial sources, such as, but not limited to, Sigma– Aldrich, Acros Organics, Fluka, and Fischer Scientific.

[0619] Stereochemistry: (±) indicates that the product is a racemic mixture of enantiomers. For example (±) (1S,2S,3R,5R) indicates that the relative product stereochemistry shown is based on known stereochemistry of similar compounds and or reactions and the product is a racemic mixture of enantiomers of both (1S,2S,3R,5R) and (1R,2R,3S,5S) stereoisomers.

[0620] A compound in which the absolute stereochemistry of separated enantiomers is undetermined is represented as being either of the single enantiomers, for example (1S,2S,3R,5R) or (1R,2R,3S,5S) or drawn as being either possible single enantiomer. In such cases, the product is pure and a single enantiomer, but absolute stereochemistry is not identified, but relative stereochemistry is known and indicated.

Example B1. Synthesis and List of Boronic Acid Intermediates.

Synthesis of (4-oxo-4,5-dihydrothieno[3,2-c]pyridin-2-yl)boronic acid (B1).

[0621] i-PrMgCl.LiCl (20 mL, 26.08 mmol) was added drop wise into a solution of 2-bromothieno[3,2- c]pyridin-4(5H)-one (1g, 4.34 mmol) in THF (15 mL) at 0 °C, and the resulting reaction mixture was stirred for 1 h then was allowed to warm to room temperature. Then (MeO) 3 B (895 mg, 8.69 mmol) was added and the reaction mixture was stirred overnight. HCl/dioxane was added to quench the reaction, the reaction mixture was concentrated and purified by silica gel chromatography (4% MeOH in DCM) to obtain the the title compound. LCMS: m/z 245.1[M+H] + ; t R = 1.14min.

Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso quinoline (B2).

[0622] Step 1: Synthesis of 7-bromo-6-methoxyisoquinoline.2,2-dimethoxyethanamine (3.7 g, 34.9 mmol) and Na2SO4 (3.3 g, 23.3 mmol) were added to a stirred solution of 3-bromo-4-methoxybenzaldehyde (5 g, 23.3 mmol) in 50 mL of toluene. The reaction mixture was heated to reflux for 6 h using a Dean-stark apparatus. Solvent and excess reagents were distilled off. The crude product was dissolved in THF (50 mL). ClCOOCH 3 (2.2 g, 23.3 mmol) was added dropwise at 0 °C. After stirring for 5 min, P(OEt) 3 (4.6 g, 27.9 mmol) was added dropwise. The mixture was stirred for 18 h at room temperature. Then the solvents were distilled off. Excess reagents were removed by repeated addition of toluene and evaporation of the solvents. TiCl 4 (17.6 g, 93.0 mmol) and CHCl 3 (25 mL) were added. The mixture was heated to reflux for 48 h. The mixture was poured on ice and the pH was adjusted to 9 by using aqueous ammonia. The resulting mixture was extracted with EtOAc followed by removal off the solvents. The residue was purified by flash silica gel column chromatography (0-70% EtOAc/petroleum ether) to give the title compound (1.3 g, 16% yield) as a white solid. LCMS: m/z 240.0 [M+H] + ; t R = 1.38 min.

[0623] Step 2: Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso quinoline (B2). A mixture of 7-bromo-6-methoxyisoquinoline (200 mg, 0.844 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (321 mg, 1.266 mmol), Pd(dppf)Cl 2 (124 mg, 0.169 mmol) and KOAc (166 mg, 1.69 mmol) in 1,4-dioxane (7 mL) was degassed and stirred at 105 °C for 8 h under N 2 protection. The reaction was cooled to room temperature and the resulting solution of crude boronic ester was used directly in the following step. LCMS: m/z 286.0 [M+H] + ; t R = 1.80 min.

Synthesis of 1-bromo-4-iodo-2-(methoxymethoxy)benzene (B3).

[0624] Step 1: Synthesis of 1-bromo-4-iodo-2-(methoxymethoxy)benzene. MOMBr (1.25 g, 10 mmol) was added to a stirred solution of 2-bromo-5-iodophenol (1.5 g, 5 mmol) and K 2 CO 3 (1.38 g, 10 mmol) in 20 mL of DMF at 0 °C. The mixture was then stirred at room temperature for 16 h, quenched with 20 mL of H2O and extracted with EtOAc (20 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel column (0-5% EtOAc/petroleum ether) to give of 1-bromo-4-iodo-2- (methoxymethoxy)benzene as colorless liquid (1.45 g, 79% yield). LCMS: tR = 1.50 min.

[0625] Step 2: Synthesis of 4-(4-bromo-3-(methoxymethoxy)phenyl)-1-(tetrahydro-2H-pyran- 2-yl)-1H- pyrazole. A mixture of 1-bromo-4-iodo-2-(methoxymethoxy)benzene (3.3 g, 9.6 mmol), 1-(tetrahydro-2H- pyran-2-yl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1H-pyrazole (2.67 g, 9.6 mmol), Pd(dppf)Cl 2 (866 mg, 1 mmol) and K 2 CO 3 (2.66 g, 19.3 mmol) in 40 mL of dioxane and 4 mL of H 2 O was degassed and stirred at 105 o C for 8 h. After cooling to room temperature, the mixture was extracted with EtOAc (30 mL X 3). The combined organic solvents were concentrated and purified by silica gel column (10-50%

EtOAc/petroleum ether) to give 4-(4-bromo-3-(methoxymethoxy)phenyl)-1-(tetrahydro-2H-pyran- 2-yl)-1H- pyrazole as colorless oil (2.5 g, 69% yield). LCMS: m/z 367.1 [M+H] + ; t R = 2.03 min.

[0626] Step 3: Synthesis of 4-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2- yl)phenyl)-1- (tetrahydro-2H-pyran-2-yl)-1H-pyrazole (B3). A mixture of 4-(4-bromo-3- (methoxymethoxy)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyra zole (1.5 g, 4.1 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(1,3,2-dioxaborolane) (2.1 g, 8.2 mmol), Pd(dppf)Cl 2 (369 mg, 0.41 mmol) and KOAc (804 mg, 8.2 mmol) in 20 mL of dioxane was degassed and stirred at 105 o C for 8 h. The mixture was filtered, concentrated, and purified by silica gel column to give 4-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl)-1-(tetrahydro- 2H-pyran-2-yl)-1H-pyrazole as colorless oil (0.81 g, 48% yield). LCMS: m/z 415.3 [M+H] + ; t R = 2.10 min.

Synthesis of 5-methoxy-N,N-dimethyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxabor olan-2-yl)benzofuran-2- carboxamide (B4)

[0627] Step 1: Synthesis of 6-bromo-5-methoxybenzofuran-2-carboxylic acid. Into a 40-mL sealed tube, was placed ethyl 6-bromo-5-methoxybenzofuran-2-carboxylate (2.00 g, 6.68 mmol, 1.00 equiv), THF (20 mL), H 2 O (2.0 mL), and LiOH (0.46 g, 19.2 mmol, 2.87 equiv). The resulting solution was stirred for 1 h at room temperature. The pH value of the solution was adjusted to pH 2 with HCl (2 mol/L). The resulting solution was extracted with ethyl acetate, dried over anhydrous sodium sulfate, and concentrated. This resulted in the title compound (1.5 g, 80%) as an off-white solid.

[0628] Step 2: Synthesis of 6-bromo-5-methoxy-N,N-dimethylbenzofuran-2-carboxamide. To a solution of dimethylamine hydrochloride (0.40 g, 5.0 mmol, 1.0 equiv) in DMF (20 mL) was added Et 3 N (1.52 g, 15.0 mmol, 3 equiv), the reaction mixture was stirred at room temperature for 15 min. Then 6-bromo-5- methoxybenzofuran-2-carboxylic acid (1.50 g, 5.0 mmol,1 equiv) and HATU (2.28 g, 5.0 mmol,1.2 equiv) was added and the reaction mixture was stirred for 3 h. The resulting mixture was diluted with water (30 mL) and the aqueous layer was extracted with ethyl acetate (20 mLx2). The combined organic layers were washed with brine (30 mLx5) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with petroleum ether/ ethyl acetate (100:1~1:1) to afford the title compound (1.33 g, 90%) as a white solid.

[0629] Step 3: Synthesis of 5–methoxy-N,N-dimethyl-6-(4,4,5,5–tetramethyl-1,3,2–di oxaborolan-2-yl)- 1–benzofuran-2-carboxamide (B4). To a solution of 6-bromo-5-methoxy-N,N-dimethyl-1-benzofuran-2- carboxamide (800 mg, 2.68 mmol, 1.0 equiv) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.02 g, 4.025 mmol, 1.5 equiv) in dioxane (10 mL) was added Pd(dppf)Cl 2 (196 mg, 0.268 mmol, 0.10 equiv) and KOAc (790 mg, 8.10 mmol, 3.0 equiv). After stirring for 3 h at 100 °C under a nitrogen atmosphere, the reaction was quenched by the addition of 50 mL of water. The resulting solution was extracted with 3x50 mL of ethyl acetate, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford the title compound (1.5 g). The crude product was used directly in the next step without further purification. Synthesis of 2-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2-yl)phenyl)-5- methyloxazole (B5).

[0630] Step 1: Synthesis of 4-bromo-3-hydroxy-N-(prop-2-yn-1-yl)benzamide. To a solution of prop-2- yn-1-amine (1.52 g, 27.7 mmol), HATU (10.5 g, 27.7 mmol), DIPEA (5.96 g, 46.08 mmol) in 50 mL of DMF was added 4-bromo-3-hydroxybenzoic acid (5 g, 23.04 mmol). The mixture was then stirred at room temperature overnight, quenched with 100 mL of LiCl (aqueous solution), and extracted with EtOAc (50 mLx3). The combined organic solvents were washed with brine, and dried over anhydrous Na 2 SO 4 , and concentrated in vacuum to afford the title compound (5.8 g, 100% yield) as yellow solid (100% yield). LCMS: m/z 255.1 [M+H] + ; t R = 1.59 min.

[0631] Step 2: Synthesis of 2-bromo-5-(5-methyloxazol-2-yl)phenol. FeCl 3 (1.80 mg, 11.07 mmol) was added to a stirred solution of 4-bromo-3-hydroxy-N-(prop-2-yn-1-yl)benzamide (5.60 g, 22.1 mmol) in 50 mL of 1,2-dichloroethane at room temperature. The mixture was then stirred at 80 o C for 2 h, quenched with 100 mL of H 2 O, and extracted with EtOAc (50 mL X 3). The combined organic solvents were dried over anhydrous Na2SO4, concentrated, and purified by silica gel column (0-16% methanol/dichloromethane) to give the title compound (3.20 g, 57.1% yield) as a white solid. LCMS: m/z 255.1 [M+H] + ; t R = 1.87min.

[0632] Step 3: Synthesis of 2-(4-bromo-3-(methoxymethoxy)phenyl)-5-methyloxazole. NaH (1.32 g, 33.2 mmol, 60% in mineral oil) was added to a stirred solution of 2-bromo-5-(5- methyloxazol-2-yl)phenol (2.10 g, 8.3 mmol) in 30 mL of THF at room temperature over 30 mins, bromo(methoxy)methane (1555 mg, 12.45 mmol) was added and mixture was then stirred at room temperature for 1 h, quenched with 30 mL of H 2 O, and extracted with EtOAc (20 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated, and purified by silica gel column (0-38% ethyl acetate/petroleum ether) to afford the title compound as a yellow liquid (3.70 g, 98.7% yield). LCMS: m/z 298.1 [M+H] + ; t R = 2.12 min.

[0633] Step 4: Synthesis of 2-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2- yl)phenyl)-5-methyloxazole (B5). To a solution of AcOK (2.38 g, 24.24 mmol), Pd(dppf)Cl 2 (1.77 g, 2.42 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (6.16 g, 24.24 mmol) in 40 mL of dioxane was added 2-(4-bromo-3- (methoxymethoxy) phenyl)-5-methyloxazole (3.60 g, 12.12 mmol). The mixture was then stirred at 100 o C for 2 h, concentrated, and purified by silica gel column (0-15% Ethyl

acetate/Petroleum ether) to give the title compound (4.20 g, 90% yield) as yellow solid (90% yield). LCMS: m/z 346.1 [M+H] + ; t R = 2.20 min.

Synthesis of 1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)-1H-imidazole-4- carbonitrile (B6).

[0634] Step 1: Synthesis of 1-(3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile. The mixture of 5- fluoro-2-nitrophenol (4.0 g, 25.4 mmol), 1H-imidazole-4-carbonitrile (3.56 g, 38.2 mol) and Cs 2 CO 3 (12.46 g, 38.22 mmol) in 100 mL of DMF was stirred at 120 °C for 16 h. The solid was filtered off, the filtrate was concentrated under vacuum to give 5 g of 1-(3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile (68% yield), which was used directly to next step. LCMS: m/z 231.2 [M+H] + ; t R = 1.20 min.

[0635] Step 2: Synthesis of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile. A mixture of 1- (3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile (4 g, 17.4 mmol), Fe (2.92 g, 52.2 mol) and NH 4 Cl (2.8 g, 52.2 mmol) in 40 mL of EtOH and 20 mL of water was stirred at 80 °C for 2 h. The solid was filtered off. The filtrate was concentrated and purified by silica gel chromatography (80% EtOAc/petroleum ether) to give 3.2 g of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile. (92% yield). LCMS: m/z 201.2 [M+H] + ; t R = 1.28 min.

[0636] Step 3: Synthesis of 1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)-1H- imidazole-4-carbonitrile (B6). HCl (4.5 mL, 12 N aqueous solution) and water (4.5 mL) was added to the solution of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile (1 g, 5.0 mmol) in 18 mL of MeOH at 0 °C. NaNO 2 (0.38 g, 5.5 mmol) in water (3 mL) was then added. After stirring at 0 °C for 30 min,

4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolan e) (2.54 g, 10.0 mmol) was added. The mixture was stirred at room temperature overnight, quenched with H 2 O (50 mL) and extracted with CH 2 Cl 2 (30 mL X 3). The combined organic layers were concentrated and purified with silica gel chromatography (0-100%

EtOAc/petroleum ether) to give 170 mg of (1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenyl)-1H-imidazole-4-carbonitrile (15 % yield). LCMS: m/z 230.1 [M+H] + ; t R = 1.26 min.

Synthesis of 2-(4-chloro-5-fluoro-2-(methoxymethoxy)phenyl)-4,4,5,5-tetra methyl-1,3,2-dioxaborolane (B7).

[0637] Step 1: Synthesis of 2-bromo-5-chloro-4-fluorophenol. Into a 500-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-chloro-4-fluorophenol (30.0 g, 205 mmol, 1.0 equiv), CH 2 Cl 2 (300 mL), Br 2 (39.2 g, 246 mmol, 1.2 equiv). The resulting solution was stirred overnight at room temperature. The reaction was then quenched by the addition of 500 mL of water/ice. The resulting solution was extracted with 3x500 mL of CH 2 Cl 2 and the organic layers combined. The resulting mixture was washed with 1 x500 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (0-20%). This resulted in of 2-bromo-5-chloro-4-fluorophenol (25 g, 54.27%) as a yellow solid.

[0638] Step 2: Synthesis of 1-bromo-4-chloro-5-fluoro-2-(methoxymethoxy)benzene. Into a 250 mL 3- necked round-bottom flask were added 2-bromo-5-chloro-4-fluorophenol (8.00 g, 35.5 mmol, 1.0 equiv) and THF (110 mL). To the above mixture was added NaH (60 wt%, 1.70 g, 42.5 mmol, 1.2 equiv) in portions at 0 °C. The resulting mixture was stirred for additional 30 min, then the above mixture was added

bromo(methoxy)methane (8.00 g, 64.0 mmol, 1.8 equiv) dropwise at 0 °C. The resulting mixture was stirred for 2 h and allowed to warm to room temperature. The reaction was cooled to 0 °C and quenched by the addition of saturated NH 4 Cl aq. (70 mL). The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column

chromatography, eluting with petroleum ether to afford 1-bromo-4-chloro-5-fluoro-2- (methoxymethoxy)benzene (8.1g, 84.7%) as a colorless oil.

[0639] Step 3: Synthesis of 2-(4-chloro-5-fluoro-2-(methoxymethoxy)phenyl)-4,4,5,5-tetra methyl-1,3,2- dioxaborolane (B7). Into a 500-mL 3-necked round-bottom flask, was placed a solution of 1-bromo-4-chloro- 5-fluoro-2- (methoxymethoxy)benzene (20.0 g, 74.2 mmol, 1.0 equiv) in dioxane (200 mL),

bis(pinacolato)diboron (28.3 g, 111 mmol, 1.5 equiv), Pd(dppf)Cl 2 (2.72 g, 3.71 mmol, 0.05 equiv), KOAc (14.6 g, 148 mmol, 2 equiv). The resulting solution was stirred for 3 h at 100 °C. The resulting mixture was concentrated and residue was applied onto a silica gel column with ethyl acetate/petroleum ether (3:100). This resulted in 2-[4-chloro-5-fluoro-2-(methoxymethoxy)phenyl]-4,4,5,5-tetra methyl-1,3,2-dioxaborolane (13 g, 55.34%) as a solid.

Synthesis of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (B8).

[0640] Step 1: Syntheisis of 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole. K 2 CO 3 (4.04 g, 29.2 mmol ) was added to a solution of 4-fluoro-2-methoxy-1-nitrobenzene (5 g, 29.2 mmol) and 4,5- dibromo-2H-1,2,3-triazole (6.63 g, 29.2 mmol) in DMF (100 mL). The resulting mixture was stirred at 80 °C for 16 hours. After cooling to room temperature, the mixture was poured into ice-water (100 mL) and extracted with EtOAc. The organic layers were washed with water (100 mL), dried over anhydrous MgSO 4 , concentrated in vacuum to give 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole (10 g , 97%) as a white solid. LCMS: m/z 378.9 [M+H] + ; t R =1.250 min.

[0641] Step 2: Synthesis of 2-methoxy-4-(2H-1,2,3-triazol-2-yl)aniline. Pd/C (1 g 10% on activated carbon) was added to a solution of 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole (10 g, 26.5 mmol) in MeOH (150 mL). The mixture was stirred under hydrogen atmosphere for 5 h and filtered. The filtrate was concentrated in vacuum to give 2-methoxy-4-(2H-1,2,3-triazol-2-yl)aniline (5 g, yield 98%) as a white solid. LCMS: m/z 191 [M+H] + ; t R =0.574 min.

[0642] Step 3: Synthesis of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid. A pre-cooled (- 15°C) solution of t-BuONO (2.61 g, 25.3 mmol) and 2-methoxy-4-(2H-1,2,3- triazol-2-yl)aniline (4.0 g, 21 mmol) in AcOH (80 mL) was added dropwise to a precooled solution of TfOH (3.79 g, 25.3 mmol) in AcOH (80 mL). The reaction was stirred for 10~20 min at 10-15°C, then poured into cold Et 2 O (1000 mL). The precipitated diazonium salt was collected by filtration and dried in vacuum to give 2-methoxy-4-(2H-1,2,3- triazol-2-yl)benzenediazonium, trifluoromethanesulfonic salt (7.4 g, yield 95%) as a white solid. LCMS: m/z 202.2 [M+] + ; t R = 0.737 min. The 2-methoxy-4-(2H-1,2,3-triazol-2-yl)benzenediazonium,

trifluoromethanesulfonic salt (7.4 g, 21 mmol) was disolved in water (150 mL). Hypodiboric acid (4.74 g, 52.7 mmol) was added. The reaction mixture was stirred at 25 °C for 3 h. The precipitate was collected by filtration and dried in vacuum to give (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (4.6 g, yield 99%) as a white solid. LCMS: m/z 220.2 [M+H] + ; t R =1.127 min.

[0643] Step 4: Synthesis of (2-hydroxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid. BBr 3 (4.6 mL, 18.26 mmol, 4M) was added to a solution of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (1000 mg, 4.57 mmol) in DCM (4 mL). The reaction mixture was stirred at 20 °C for 18 h under N 2 atmosphere, concentrated in vacuum and purified by silica gel column (10-25% EtOAc/petroleum ether) to give (2- hydroxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (450 mg, 59 % yield) as a yellow solid. LCMS: m/z 206.2 [M+H] + ; t R = 0.898 min.

Synthesis of (4-(4-cyano-1H-imidazol-1-yl)-2-methoxyphenyl)boronic acid (B9).

[0644] Step 1: Synthesis of 1-(3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile. The mixture of 5- fluoro-2-nitrophenol (4 g, 25.4 mmol), 1H-imidazole-4-carbonitrile (3.56 g, 38.2 mol) and Cs 2 CO 3 (12.46 g, 38.2 mmol) in 100 mL of DMF was stirred at 120 o C for 16 h. The solid was filtered off, the filtrate was concentrated under vacuum to give 5 g of 1-(3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile (68% yield), which was used directly to next step. LCMS: m/z 231.2 [M+H] + ; t R = 1.20 min.

[0645] Step 2: Synthesis of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile. A mixture of 1- (3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile (4 g, 17.4 mmol), Fe (2.92 g, 52.2 mol) and NH 4 Cl (2.8 g, 52.2 mmol) in 40 mL of EtOH and 20 mL of water was stirred at 80 o C for 2 h. The solid was filtered off. The filtrate was concentrated and purified by silica gel chromatography (80% EtOAc/petroleum ether) to give 3.2 g of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile. (92% yield). LCMS: m/z 201.2 [M+H] + ; t R = 1.28 min. [0646] Step 3: Synthesis of 1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)-1H- imidazole-4-carbonitrile. HCl (4.5 mL, 12 N aqueous solution) and water (4.5 mL) was added to the solution of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile (1 g, 5.0 mmol) in 18 mL of MeOH at 0 o C. NaNO 2 (0.38 g, 5.5 mmol) in water (3 mL) was then added. After stirring at 0 o C for 30 min, 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(1,3,2-dioxaborolane) (2.54 g, 10.0 mmol) was added. The mixture was stirred at room temperature overnight, quenched with H 2 O (50 mL) and extracted with CH 2 Cl 2 (30 mL X 3). The combined organic layers were concentrated and purified with silica gel chromatography (0-100% EtOAc/petroleum ether) to give 170 mg of (1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl )phenyl)-1H-imidazole- 4-carbonitrile (15 % yield). LCMS: m/z 230.1 [M+H] + ; t R = 1.26 min.

Synthesis of 5-(4-ethyl-1H-1,2,3-triazol-1-yl)-2-(4,4,5,5-tetramethyl-1,3 ,2-dioxaborolan-2-yl)phenol (B10).

[0647] Step 1: Synthesis of 2-(benzyloxy)-4-fluoro-1-nitrobenzene. Benzyl bromide (56 g, 0.33 mol) was added to a solution of 5-fluoro-2-nitrophenol (40 g, 0.25 mol) and K 2 CO 3 (69 g, 0.5 mol) in DMF (400 mL). The mixture was stirred at room temperature for 1 h, quenched with water and extracted with EtOAc. The combined organic solvents were washed with LiCl aqueous solution (300 mL X 2), concentrated and purified by silica gel chromatography (20% EtOAc/petroleum ether) to give 61 g of 2-(benzyloxy)-4-fluoro-1- nitrobenzene (97% yield). LCMS: m/z 270.0 [M+Na] + ; t R = 1.87 min.

[0648] Step 2: Synthesis of 4-azido-2-(benzyloxy)-1-nitrobenzene. NaN 3 (41 g, 0.63 mol) was added to a stirred solution of 2-(benzyloxy)-4-fluoro-1-nitrobenzene (61 g, 0.25 mol) in DMF (800 mL). The mixture was stirred at room temperature for 24 h and 60 o C for 3 h. After cooling to room temperature, the mixture was quenched with water and extracted with EtOAc. The combined organic solvents were washed with LiCl aqueous solution (500 ml X 2), dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (20% EtOAc/petroleum ether) to give 65 g of 4-azido-2-(benzyloxy)-1-nitrobenzene. (98.5% yield). LCMS: m/z 293.0 [M+Na] + ; t R = 1.93 min.

[0649] Step 3: Synthesis of 1-(3-(benzyloxy)-4-nitrophenyl)-4-ethyl-1H-1,2,3-triazole. A mixture of 4- azido-2-(benzyloxy)-1-nitrobenzene (2.7 g, 10.0 mmol), pent-2-ynoic acid (0.98 g, 10.0 mmol), Cu 2 O (143 mg, 1.0 mmol) in 40 mL of CH 3 CN was degassed and stirred at 80 o C for 2 h. After cooling to room temperature, the mixture were concentrated and purified by silica gel column (0-100% EtOAc/petroleum ether) to give 3.2 g of 1-(3-(benzyloxy)-4-nitrophenyl)-4-ethyl-1H-1,2,3-triazole as yellow solid (89% yield). LCMS: m/z 325.2 [M+H] + ; t R = 1.89 min. [0650] Step 4: Synthesis of 2-amino-5-(4-ethyl-1H-1,2,3-triazol-1-yl)phenol. To a solution of 1-(3- (benzyloxy)-4-nitrophenyl)-4-ethyl-1H-1,2,3-triazole (2.2 g, 6.8 mmol) in 40 mL of methanol was added Pd/C (0.8 g, 10% in activated carbon). The mixture was stirred at room temperature under hydrogen atmosphere for 2 h and filtered through a pad of Celite® brand filter agent. The filtrate was concentrated and purified by silica gel column (0-10% EtOAc/petroleum ether) to give 1.2 g 2-amino-5-(4-ethyl-1H-1,2,3-triazol-1-yl)phenol (87% yield). LCMS: m/z 205.2 [M+H] + ; t R = 1.23 min.

[0651] Step 5: Synthesis of 5-(4-ethyl-1H-1,2,3-triazol-1-yl)-2-(4,4,5,5-tetramethyl-1,3 ,2-dioxaborolan- 2-yl)phenol.2.7 mL of HCl (37% aqueous solution) was added to a solution of 2-amino-5-(4-ethyl-1H-1,2,3- triazol-1-yl)phenol (600 mg, 3.0 mmol) in 18 mL of methanol and a solution of NaNO 2 (228 mg , 3.3 mmol) in 7.2 mL of water added at 0 °C. The mixture was stirred at 0 °C for 30 min followed by addition of 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (2.29 g , 9.0 mmol). The mixture was then stirred at room temperature for 16 h, extracted with dichloromethane (50 mL X 2). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel column (0-100% EtOAc/petroleum ether) to give 283 mg of 5-(4-ethyl-1H-1,2,3-triazol-1-yl)-2-(4,4,5,5-tetramethyl-1,3 ,2-dioxaborolan-2- yl)phenol (30% yield). LCMS: m/z 316.1 [M+H] + ; t R = 1.91 min.

Synthesis of 3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)benzonitrile (B11).

[0652] Step 1: Synthesis of 4-bromo-3-(methoxymethoxy)benzonitrile. NaH (1.62 g, 40.6 mmol, 60% in mineral oil) was added to a stirred solution of 4-bromo-3-hydroxybenzonitrile (4 g, 20.3 mmol) in 100 mL of DMF at 0 o C. After stirring at 0 o C for 30 min, MOMBr (5.08 g, 40.6 mmol) was added. The mixture was then stirred at room temperature for 2 h, quenched with NH 4 Cl aqueous solution (10 mL), extracted with EtOAc (50 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give 4.8 g of 4-bromo-3- (methoxymethoxy)benzonitrile as colorless oil (98% yield). LCMS: m/z 241.9 [M+H] + ; t R = 1.81 min.

[0653] Synthesis of 3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)benzonitrile. A mixture of 4-bromo-3-(methoxymethoxy)benzonitrile (2 g, 8.3 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (2.74 g, 10.8 mmol), Pd(dppf)Cl 2 (607 mg, 0.83 mmol) and KOAc (1.63 g, 16.6 mmol) in 50 mL of dioxane was degassed and stirred at 100 o C for 2 h. After cooling to room temperature, the mixture was extracted with EtOAc (30 mL X 3). The combined organic solvents were concentrated and purified by silica gel column (0-20% EtOAc/petroleum ether) to give 2.27 g of 3-(methoxymethoxy)-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile as colorless oil (95% yield). LCMS: m/z 290.3 [M+H] + ; t R = 1.96 min.

Synthesis of 5-(methoxymethoxy)-2-methyl-6-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2- yl)benzo[d]oxazole (B12).

[0654] Step 1: Synthesis of 2-amino-4-methoxyphenol. A mixture of 4-methoxy-2-nitrophenol (25 g, 0.15 mol) and Pd/C (2.5 g) in MeOH (500 mL) was stirred at 20 o C under hydrogen atmosphere (balloon) for 2 days. The reaction mixture was filtered and the filtrate was concentrated in vacuum to afford 2-amino-4- methoxyphenol as brown solid (20.0 g, yield 97.2 %). LCMS: m/z 140.1 [M+H] + ; tR = 0.93 min.

[0655] Step 2: Synthesis of 5-methoxy-2-methylbenzo[d]oxazole. A mixture of 2-amino-4-methoxyphenol (20.0 g, 0.14 mol) in trimethyl orthoacetate (50 mL) was stirred at 100 o C for 1 h. The mixture was concentrated, and the residue was purified by silica gel chromatography (0-35% EtOAc/petroleum) to give 5- methoxy-2-methylbenzo[d]oxazole as orange oil (17.5 g, yield 74.6 %). LCMS: m/z 164.1 [M+H] + ; t R = 1.41 min.

[0656] Step 3: Synthesis of 6-bromo-5-methoxy-2-methylbenzo[d]oxazole. NBS (19.6 g, 0.11 mol) was added to a stirred solution of 5-methoxy-2-methylbenzo[d]oxazole (17.5 g, 0.11 mol) in AcOH (150 mL). After stirring at 20 o C for 18 h, the mixture was quenched with ice water, neutralized with Na 2 CO 3 aqueous solution, extracted with EtOAc (200 mL X 3). The extracts were concentrated, and the residue was purified by silica gel chromatography (0-10% EtOAc/petroleum) to give 6-bromo-5-methoxy-2-methylbenzo[d]oxazole as pink solid (20.5 g, yield 77.0 %). 1 H NMR (500 MHz, CDCl 3 ) d 7.68 (s, 1H), 7.17 (s, 1H), 3.93 (s, 3H), 2.61 (s, 3H). LCMS: m/z 242.1; 243.9 [M+H] + ; t R = 1.70 min.

[0657] Step 4: Synthesis of 6-bromo-2-methylbenzo[d]oxazol-5-ol. BBr 3 (210 mL, 0.21 mol, 1N in CH 2 Cl 2 ) was added to 6-bromo-5-methoxy-2-methylbenzo[d]oxazole (20.5 g, 0.085 mol) in CH 2 Cl 2 (30 mL). at 0 o C. This resulting mixture was stirred at 0 o C for 10 min, then stirred at 20 o C for 3 days. The mixture was quenched with ice water, neutralized with NaHCO 3 aqueous, extracted with EtOAc (360 mL X 3). The extracts were concentrated, and the residue was purified by silica gel chromatography (0-20% MeOH/CH 2 Cl 2 ) to give 6-bromo-2-methylbenzo[d]oxazol-5-ol as grey solid (19.0 g, yield 98.6 %). LCMS: m/z 228.0; 230.0 [M+H] + ; t R = 1.50 min.

[0658] Step 5: Synthesis of 6-bromo-5-(methoxymethoxy)-2-methylbenzo[d]oxazole. MOMBr (15.6 g, 0.12 mol) was added drop wise to a solution of 6-bromo-2-methylbenzo[d]oxazol-5-ol (19.0 g, 0.08 mol) and DIPEA (37.7 g, 0.19 mol) in CH 3 CN (300 mL) at 5 o C. After stirring for 30 min, the mixture was quenched with ice water, extracted with EtOAc (200 mL X 3). The extracts were washed with brine (300 mL), concentrated and purified by silica gel chromatography (0-25% EtOAc/petroleum) to give 6-bromo-5- (methoxymethoxy)-2-methylbenzo[d]oxazole as pink solid (18.0 g, yield 79.5 %). LCMS: m/z 272.0; 274.0 [M+H] + ; t R = 1.77 min.

[0659] Step 6: Synthesis of 5-(methoxymethoxy)-2-methyl-6-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan- 2-yl)benzo[d]oxazole (B12). A mixture of 6-bromo-5-(methoxymethoxy)-2-methylbenzo[d]oxazole (50 g, 0.02 mol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (28.0 g, 0.11 mol), Pd(dppf)Cl 2 (1.1 g, 15 mmol) and KOAc (10.8 g, 0.11 mol) in 1, 4-dioxane (300 mL) was degassed and stirred at 100 o C 50 h. The mixture was quenched with ice water, extracted with EtOAc (200 mL X 3). The extracts were washed with brine (200 mL), concentrated and purified by silica gel chromatography (0-25% EtOAc/petroleum) to give 5- (methoxymethoxy)-2-methyl-6-(4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl)benzo[d]oxazole as pink solid (4.0 g, yield 68 %). LCMS: m/z 320.2 [M+H] + ; t R = 1.86 min.

Synthesis of 6-(methoxymethoxy)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)-4H-chromen-4-one (B13).

[0660] Step 1: Synthesis of 1-(4-bromo-2,5-dihydroxyphenyl)ethenone. Into a 1-L 3-necked round-bottom flask, was placed 1-(4-bromo-2,5-dimethoxyphenyl)ethanone (30.00 g, 115.786 mmol, 1 equiv), DCM (300.00 mL). This was followed by the addition of BBr 3 (1M in DCM, 347.97 mL, 347.97 mmol, 3 equiv) dropwise with stirring at 0 o C. The resulting solution was stirred for 3 h at room temperature. The reaction was then quenched by the addition of 500 mL of NaHCO 3 /Water. The resulting solution was extracted with 3x500 mL of dichloromethane and concentrated. The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1;1). This resulted in 14 g (52.3%) of 1-(4-bromo-2,5- dihydroxyphenyl)ethanone as a yellow solid.

[0661] Step 2: Synthesis of 7-bromo-6-hydroxychromen-4-one. Into a 50-mL 3-necked round-bottom flask, was placed 1-(4-bromo-2,5-dihydroxyphenyl)ethanone (462 mg, 2.0 mmol) and triethyl orthoformate (1.80 mL). This was followed by the addition of perchloric acid (0.2 mL) dropwise with stirring at 0 °C. The resulting solution was stirred overnight at room temperature. The reaction was then quenched by the addition of 10 mL of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the combined organic layers were concentrated. The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1:9-1:1). This resulted in 100 mg (20.75%) of 7-bromo-6-hydroxychromen-4-one as a brown solid.

[0662] Step 3: Synthesis of 7-bromo-6-(methoxymethoxy)chromen-4-one. Into a 50 mL 3-necked round- bottom flask were added 7-bromo-6-hydroxychromen-4-one (366 mg, 1.52 mmol, 1.0 equiv) and THF (6 mL) at 0 °C. To the above mixture was added NaH (91 mg, 3.8 mmol, 60 wt% in mineral oil) in portions at 0 °C. The resulting mixture was stirred for additional 30 min at 0 o C. Then to the above mixture was added MOMBr (285 mg, 2.3 mmol, 1.5 equiv) at 0 °C. The resulting mixture was stirred for additional 1 h at room temperature. The resulting mixture was diluted with water (20 mL). The resulting mixture was extracted with EtOAc (3 x 20mL). The organics were combined and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with hexane/EtOAc (9:1-1:1) to afford 7-bromo-6-(methoxymethoxy)chromen-4- one (340 mg, 45.63%) as a brown solid.

[0663] Step 4: Synthesis of 6-(methoxymethoxy)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)-4H- chromen-4-one (B13). Into a 100 mL round-bottom flask were added 7-bromo-6- (methoxymethoxy)chromen-4-one (1.04 g, 3.65 mmol, 1.0 equiv), bis(pinacolato)diboron (66.80 mg, 0.263 mmol, 1.5 equiv), Pd(dppf)Cl 2 (267 mg, 0.365 mmol, 0.10 equiv), KOAc (712 mg, 7.26 mmol, 2 equiv) and 1,4-dioxane (15 mL). The resulting mixture was stirred overnight at 100 °C under nitrogen atmosphere. The resulting solution was used in the next step directly without work up. LCMS: m/z 333 [M+H] + .

Synthesis of 7-(methoxymethoxy)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)quinoline (B14).

[0664] Step 1: Synthesis of 6-bromo-7-methoxyquinoline. H 2 SO 4 (10.5 mL ) in 12 mL of H 2 O was added 4- bromo-3-methoxyaniline (10.5 g, 51.7 mmol) and propane-1,2,3-triol (12.5 g, 135.7 mmol). The mixture was heated to 110 °C, 3-nitrobenzenesulfonic acid (10 g, 49.5 mmol) was added portion-wise. Then 15 mL of H 2 O, 15 mL of propane-1,2,3-triol and 15 mL of H 2 SO 4 were added successively. The mixture was stirred at 140 o C for 3 h. After cooling to room temperature, the mixture was poured onto ice, and the pH was adjusted to 8 by addition NH 3 H 2 O. The mixture was extracted with EtOAc (100 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (25- 100% EtOAc/petroleum ether) to give 9.9 g of 6-bromo-7-methoxyquinoline as a gray solid (83% yield). LCMS: m/z 240.1 [M+H] + ; t R = 1.54 min.

[0665] Step 2: Synthesis of 6-bromoquinolin-7-ol. To a solution of 6-bromo-7-methoxyquinoline (2 g, 8.4 mmol) in CH 2 Cl 2 (4 mL) at 25 o C was added BBr 3 (40 mL, 1N in CH 2 Cl 2 ) and stirred at 25 o C for 16 h, monitored by LCMS. Then water (15 mL) and ammonia methanol solution was added to pH to 8~9. The precipitate was collected by filtration to give 1.02 g of 6-bromoquinolin-7-ol. LCMS: m/z 226.0 [M+H] + ; t R = 1.15 min.

[0666] Step 3: Synthesis of 6-bromo-7-(methoxymethoxy)quinoline. NaH (363 mg, 9.07 mmol, 60% in mineral oil) was added to a stirred solution of 6-bromoquinolin-7-ol (1.02 g, 4.53 mmol) in 65 mL of THF at 25 o C. After stirring at 25 o C for 30 min, MOMBr (623 mg, 4.98 mmol) was added. The mixture was then stirred at room temperature for 1 h, quenched with NH 4 Cl aqueous solution (20 mL), extracted with EtOAc (60 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-50% EtOAc/petroleum ether) to give 780 mg of 6-bromo-7- (methoxymethoxy)quinoline as colorless oil (64% yield), LCMS: LCMS: m/z 270.0 [M+H] + ; t R = 1.67 min.

[0667] Step 4: Synthesis of 7-(methoxymethoxy)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2- yl)quinoline (B14). A mixture of 6-bromo-7-(methoxymethoxy)quinoline (100 mg, 0.42 mmol),

4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolan e) (159 mg, 0.63 mmol), Pd(dppf)Cl 2 (61 mg, 0.084 mmol) and KOAc (82 mg, 0.84 mmol) in 4 mL of dioxane was degassed and stirred at 100 o C for 2 h. After cooling to room temperature, the mixture was used directly to next step.

6-(methoxymethoxy)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2-yl)quinoline (B15).

[0668] Step 1: Synthesis of 7-bromo-6-methoxyquinoline. A mixture of 3-bromo-4-methoxyaniline (10 g, 49.49 mmol), 3-nitrobenzenesulfonic acid (11.06 g, 54.4 mmol), propane-1,2,3-triol (45.6 g, 495 mmol) and H 2 SO 4 (48.5 g, 495 mmol) in water (60 mL) was heated to 140 o C for 16 h. LCMS showed most of the starting material disappeared, and the mixture was poured into ice water (200 mL). NH 3 .H 2 O was added dropwise to the mixture to make the pH to about 8. The mixture was extracted with ethyl acetate (150 mL). The organic phase was washed with sodium chloride solution, dried, concentrated, and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give 7-bromo-6-methoxyquinoline (6.81 g, 57% yield) as white solid. LCMS: m/z 237.9 [M+H] + ; t R = 1.44 min.

[0669] Step 2: Synthesis of 7-bromoquinolin-6-ol. To a mixture of 7-bromo-6-methoxyquinoline (6.3 g, 26.46 mmol) in DCM (80 mL) was added 1M BBr 3 in DCM (66 mL) dropwise at -78 o C. The mixture was warmed to room temperature and stirred for 16 h. LCMS showed most of the starting material disappeared, and the mixture was quenched with ice water dropwise at 0 o C. Sodium hydroxide solution was added to the mixture to make the pH to about 7. The solid was filtered to give 8.2 g of 7-bromoquinolin-6-ol as white solid (contained boric acid). LCMS: m/z 224.0 [M+H] + ; t R = 1.12 min.

[0670] Step 3: Synthesis of 7-bromo-6-(methoxymethoxy)quinoline. To a solution of 7-bromoquinolin-6- ol (2 g, 8.93 mmol) and DIPEA (2.31 g, 17.85 mmol) in DCM (30 mL) was added MOMBr (1.67 g, 13.39 mmol) at 0 o C. The solution was stirred at room temperature for 2 h. LCMS showed most of the starting material disappeared, and the mixture was quenched with sodium bicarbonate solution (10 mL). The mixture was extracted with ethyl acetate (50 mL) and water (40 mL). The organic layer was dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-25% EtOAc/petroleum ether) to give 7- bromo-6-(methoxymethoxy)quinoline (920 mg, 38 % yield) as light yellow solid. LCMS: m/z 268.0 [M+H] + ; t R = 1.53 min. [0671] Step 4: Synthesis of 6-(methoxymethoxy)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2- yl)quinoline. A mixture of 7-bromo-6-(methoxymethoxy)quinoline (1.1 g, 4.1 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.56 g, 6.15 mmol), Pd(dppf)Cl 2 (300.2 mg, 0.41 mmol) and KOAc (1.21 g, 7.68 mmol) in dioxane (15 mL) was stirred at 100 o C under N 2 for 16 h, concentrated and purified by silica gel chromatography (0-40% EtOAc/petroleum ether) to give 6-(methoxymethoxy)-7-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline (800 mg, 61% yield) as light yellow oil. LCMS: m/z 234.0 [M- 81] + ; t R = 1.40 min.

Synthesis of 2-fluoro-5-(methoxymethoxy)-N-methyl-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)benzamide (B16).

[0672] Step 1: 4-bromo-2-fluoro-5-hydroxybenzoic acid. Into a 2-L 3-necked round-bottom flask, was placed 2-fluoro-5-hydroxybenzoic acid (40 g, 256 mmol), CHCl 3 (400 mL). This was followed by the addition of Br 2 (38.3 mL, 748 mmol, 3 equiv) in CH 3 COOH (400 mL) dropwise with stirring at 0 o C over 30 min. The resulting solution was stirred overnight at room temperature. The reaction was quenched with Na 2 S 2 O 3 at room temperature. The resulting mixture was extracted with EtOAc (3 x 1L), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column

chromatography, eluting with PE/EtOAc (1:1) to afford 4-bromo-2-fluoro-5-hydroxybenzoic acid (40 g, 66.4%) as a yellow solid.

[0673] Step 2: Synthesis of 4-bromo-2-fluoro-5-hydroxy-N-methylbenzamide. Into a 50-mL 3-necked round-bottom flask, was placed 4-bromo-2-fluoro-5-hydroxybenzoic acid (15 g, 63.8 mmol, 1.0 equiv), THF (150 mL), isopropyl chloroformate (19.48 g, 158.9 mmol, 2.3 equiv), TEA (15 g, 148 mmol, 2.3 equiv). The resulting solution was stirred for 0.5 hr at 0 o C. The solids were filtered out. To the filtrate was added MeNH 2 (1M in MeOH, 225 mL) at 0 o C. The resulting solution was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure and diluted with water (100 mL). The resulting mixture was extracted with EtOAc (3 x 100 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (9:1-1:1) to afford 4-bromo-2-fluoro-5-hydroxy-N-methylbenzamide (7g, 44.21%) as a white solid. ES, m/z : 248 (M+1).

[0674] Step 3: Synthesis of 4-bromo-2-fluoro-5-(methoxymethoxy)-N-methylbenzamide. Into a 50 mL 3- necked round-bottom flask were added 4-bromo-2-fluoro-5-hydroxy-N-methylbenzamide(5.00 g, 20.157 mmol, 1.00 equiv) and THF (75.00 mL) at 0 o C. To the above mixture was added NaH (60%, 532.10 mg, 22.173 mmol, 1.10 equiv) in portions at 0oC. The resulting mixture was stirred for 30 min at 0oC. Then to the above mixture was added MOMBr (3.02 g, 24.183 mmol, 1.20 equiv) in portions at 0 o C. The resulting mixture was stirred for additional 1 h at room temperature. The resulting mixture was diluted with water (100 mL). The resulting mixture was extracted with EtOAc (3 x 100 mL). The organics were combined and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with hexane/ EtOAc (9:1-1:1) to afford 4-bromo-2- fluoro-5-(methoxymethoxy)-N-methylbenzamide (5.0g, 84.92%) as a light yellow solid. ES, m/z : 292 (M+1).

[0675] Step 4: 2-fluoro-5-(methoxymethoxy)-N-methyl-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)benzamide (B16). Into a 250 mL round-bottom flask were added 4-bromo-2-fluoro-5-(methoxymethoxy)- N-methylbenzamide (5 g, 17.1 mmol), bis(pinacolato)diboron (6.56 g, 25 mmol), Pd(dppf)Cl 2 (1.4 g, 1.9 mmol), K 2 CO 3 (4.75 g, 34 mmol) and 1,4-dioxane (50 mL). The mixture was stirred overnight at 100 °C under nitrogen atmosphere. The resulting mixture was diluted with water (100 mL) and extracted with EtOAc (3 x 100 mL). The organics were combined and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (5:1-1:1) to afford 2-fluoro-5-(methoxymethoxy)-N-methyl-4-(4,4,5,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)benzamide (3 g, 71.42%) as a yellow solid. ES, m/z : 340 (M+1).

Synthesis of 2-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2-yl)phenyl)-5- methyloxazole (B17).

[0676] Step 1: Synthesis of 4-bromo-3-hydroxy-N-(prop-2-yn-1-yl)benzamide. To a solution of prop-2- yn-1-amine (1.52 g, 27.65 mmol), HATU (10.51 g, 27.65 mmol), DIPEA(5.96 g, 46.08 mmol) in 50 mL of DMF was added 4-bromo-3-hydroxybenzoic acid (5 g, 23.04 mmol). The mixture was then stirred at room temperature overnight, quenched with 100 mL of LiCl (aqueous solution), extracted with EtOAc (50 mL X 3). The combined organic solvents were washed with brine and dried over anhydrous Na 2 SO 4 , concentrated in vacuum to give 5.8 g of 4-bromo-3-hydroxy-N-(prop-2-yn-1-yl)benzamide as yellow solid (100% yield). LCMS: m/z 255.1 [M+H] + ; t R = 1.59 min.

[0677] Step 2: Synthesis of 2-bromo-5-(5-methyloxazol-2-yl)phenol. FeCl 3 (1795 mg, 11.07 mmol) was added to a stirred solution of 4-bromo-3-hydroxy-N-(prop-2-yn-1-yl)benzamide (5.60 g, 22.13 mmol) in 50 mL of 1,2-dichloroethane at room temperature. The mixture was then stirred at 80 o C for 2 h, quenched with 100 mL of H 2 O, extracted with EtOAc (50 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel column (0-16% methanol/dichloromethane) to give 3.20 g of 2-bromo-5-(5-methyloxazol-2-yl)phenol as white solid (57.1% yield). LCMS: m/z 255.1 [M+H] + ; t R = 1.87min.

[0678] Step 3: Synthesis of 2-(4-bromo-3-(methoxymethoxy)phenyl)-5-methyloxazole. NaH (1.33 mg, 33.2 mmol, 60% in mineral oil) was added to a stirred solution of 2-bromo-5-(5- methyloxazol-2-yl)phenol (2.10 g, 8.3 mmol) in 30 mL of THF at room temperature for 30 mins, bromo(methoxy)methane (1.56 g, 12.45 mmol) was added. The mixture was then stirred at room temperature for 1 h, quenched with 30 mL of H 2 O, extracted with EtOAc (20 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel column (0-38% Ethyl acetate/Petroleum ether) to give 3.70 g of 2-(4- bromo-3-(methoxymethoxy)phenyl)-5-methyloxazole as yellow liquid (99% yield). LCMS: m/z 298.1

[M+H] + ; t R = 2.12 min.

[0679] Step 4: Synthesis of 2-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan-2- yl)phenyl)-5-methyloxazole (B17). To a solution of AcOK (2.38 gg, 24.24 mmol), Pd(dppf)Cl 2 (1.77 g, 2.42 mmol), (4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (6.16 g, 24.24 mmol) in 40 mL of dioxane was added 2-(4-bromo-3- (methoxymethoxy) phenyl)-5-methyloxazole (3.60 g, 12.12 mmol). The mixture was then stirred at 100 °C for 2 h, concentrated and purified by silica gel column (0-15% Ethyl

acetate/Petroleum ether) to give 4.20 g of 2-(3-(methoxymethoxy)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaboro lan- 2-yl)phenyl)-5-methyloxazole as yellow solid (90% yield).

Synthesis of 6-(methoxymethoxy)-2-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)-4H- chromen-4-one (B18).

[0680] Step 1: Synthesis of 1-[4-bromo-2-hydroxy-5-(methoxymethoxy)phenyl]ethenone. Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 1-(4- bromo-2,5-dihydroxyphenyl)ethanone (10.0g, 43.28 mmol, 1.00 equiv), acetone (100.00 mL), K 2 CO 3 (6.58g, 23.81 mmol, 1.10 equiv), bromo(methoxymethoxy)methane (6.7g, 47.6mmol, 1.10 equiv). The resulting solution was stirred for 2 hr at room temperature. The solids were filtered out. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (3:1). The collected fractions were combined and concentrated. This resulted in 7 g (58.8%) of 1-[4-bromo-2-hydroxy-5-(methoxymethoxy)phenyl]ethanone as a yellow oil. (ES, m/z): [M+H] + =274.

[0681] Step 2: Synthesis of 1-[4-bromo-2-hydroxy-5-(methoxymethoxy) phenyl]butane-1,3-dione. Into a 50 ml 3-necked round-bottom flask were added THF (80 mL) and NaH (2.79 g, 116.3 mmol, 60 wt%) at 0 °C. The mixture of ethyl acetate (6.41 g, 72.7 mmol, 2.5 equiv) and 1-[4-bromo-2-hydroxy-5- (methoxymethoxy) phenyl]ethanone (8.00 g, 29.08 mmol, 1.0 equiv) dissolved in 3ml THF was added slowly at 0 degrees C. Then the reaction was allowed to stir for 2 h at room temperature.

The reaction was quenched by the addition of ice water at rt. The resulting mixture was extracted with ethyl acetate (100ml x3). The combined organic layers were washed with NaCl aq, dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography. This resulted in 1-[4-bromo-2-hydroxy-5-(methoxymethoxy) phenyl]butane-1,3- dione (9 g, 97.6%) as yellow oil. (ES, m/z): [M+1] + =316

[0682] Step 3: Synthesis of 7-bromo-6-hydroxy-2-methylchromen-4-one. Into a 500-mL round-bottom flask, was placed 1-[4-bromo-2-hydroxy-5-(methoxymethoxy)phenyl]butane-1,3-dio ne (9.00 g, 28.38 mmol, 1.0 equiv), isopropyl alcohol (200 mL), Amberlyst 15 (9.0 g). The resulting solution was stirred for 3 h at 80 °C. The solids were filtered out. The resulting mixture was concentrated under vacuum. This resulted in 7.2 g (99.5%) of 7-bromo-6-hydroxy-2-methylchromen-4-one as a light yellow solid. (ES, m/z): [M+H] + =254.

[0683] Step 4: Synthesis of 7-bromo-6-(methoxymethoxy)-2-methylchromen-4-one. Into a 150-mL 3- necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed bromo(methoxymethoxy)methane (2.92 g, 18.82 mmol, 1.2 equiv), 7-bromo-6-hydroxy-2-methylchromen-4- one (4.0 g, 15.68 mmol, 1.0 equiv), NaH (0.45 g, 18.75 mmol, 95 wt% in mineral oil, 1.20 equiv), THF (40 mL). The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 50 mL of ice/salt. The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/hexane (20%). The collected fractions were combined and concentrated under vacuum. This resulted in 3 g (60.8%) of 7-bromo-6-(methoxymethoxy)-2-methylchromen-4-one as a yellow solid. (ES, m/z): [M+H] + =298.

Synthesis of 6-(methoxymethoxy)-2-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)chromen-4- one (B18).

[0684] Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed bis(pinacolato)diboron (1.32 g, 5.22 mmol, 1.20 equiv), 7-bromo-6-(methoxymethoxy)- 2-methylchromen-4-one (1.30 g, 4.35 mmol, 1.0 equiv), KOAc (1.28 g, 13.04 mmol, 3.0 equiv), 1,4-dioxane (13.0 mL), Pd(dppf)Cl 2 CH 2 Cl 2 (0.35 g, 0.435 mmol, 0.10 equiv). The resulting solution was stirred for 2 h at 100 °C in an oil bath. The resulting mixture was concentrated under vacuum. This resulted in 1.5 g (crude) of 6-(methoxymethoxy)-2-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)chromen-4-one as a brown solid. (ES, m/z): [M+1] + =347.

Synthesis of 7-(methoxymethoxy)-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)chromen-4- one (B19).

[0685] Step 1: Synthesis of 1-(5-bromo-2,4-dihydroxyphenyl)ethenone. Into a 2-L 3-necked round-bottom flask, was placed 2 (20.00 g, 131.5 mmol, 1.00 equiv), chloroform (1 L), Bu 4 NBr 3 (69.72 g, 144.595 mmol, 1.10 equiv). The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 200 mL of 5% solution of sodium thiosulfate. The resulting mixture was washed with 1x300 mL of HCl (1M). The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/hexane (20%). This resulted in 20 g (65.8%) of 1-(5-bromo-2,4-dihydroxyphenyl)ethanone as a yellow solid. m/z: 231(M+H + ).

[0686] Step 2: Synthesis of 1-[5-bromo-2-hydroxy-4-(methoxymethoxy)phenyl]ethenone. Into a 250-mL 3-necked round-bottom flask, was placed 1-(5-bromo-2,4-dihydroxyphenyl)ethanone (5.00 g, 21.64 mmol, 1.00 equiv), K 2 CO 3 (3.59 g, 26.0 mmol, 1.20 equiv), acetone (100 mL), methane, bromomethoxy- (2.84 g, 22.73 mmol, 1.05 equiv). The resulting solution was stirred overnight at room temperature. The solids were filtered out. The resulting solution was diluted with 1 L of DCM. The resulting mixture was washed with 2x200 mL of NaHCO 3 . The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. This resulted in 5 g (84.0%) of 1-[5-bromo-2-hydroxy-4-(methoxymethoxy)phenyl]ethanone as yellow oil. LCMS: m/z:275 (M+H + ).

[0687] Step 3: Synthesis of 1-[5-bromo-2-hydroxy-4-(methoxymethoxy)phenyl]butane-1,3-dio ne. Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 1-[5-bromo-2-hydroxy-4-(methoxymethoxy)phenyl]ethanone (6.00 g, 21.810 mmol, 1.00 equiv), THF (50.00 mL, 12.343 mmol, 67.91 equiv). This was followed by the addition of NaH (2.09 g, 87.2 mmol, 4.0 equiv) in several batches at 0 degrees C in 10 min. To this was added EtOAc (3.84 g, 43.58 mmol, 2.0 equiv) at 0 degrees C. The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 20 mL of water. The resulting solution was extracted with 2x100 mL of ethyl acetate dried over anhydrous sodium sulfate and concentrated under vacuum. This resulted in 6 g (86.7%) of 1-[5-bromo-2- hydroxy-4-(methoxymethoxy)phenyl]butane-1,3-dione as a solid. m/z: 317 (M+H + ).

[0688] Step 4: Synthesis of 6-bromo-7-hydroxy-2-methylchromen-4-one. Into a 500-mL 3-necked round- bottom flask, was placed 1-[5-bromo-2-hydroxy-4-(methoxymethoxy)phenyl]butane-1,3-dio ne (6.0 g, 18.9 mmol, 1.00 equiv), i-PrOH (200 mL), Amberlyst-15 (20 g). The resulting solution was stirred for 1 h at 85 °C. The solids were filtered off and washed with 100 mL of hot MeOH. The resulting mixture was concentrated under vacuum. This resulted in 4 g (82.9%) of 6-bromo-7-hydroxy-2-methylchromen-4-one as a yellow solid. m/z:255(M+H + ).

[0689] Step 5: Synthesis of 6-bromo-7-(methoxymethoxy)-2-methylchromen-4-one. Into a 100-mL 3- necked round-bottom flask, was placed 6-bromo-7-hydroxy-2-methylchromen-4-one (4.00 g, 15.68mmol, 1.0 equiv), THF (40.00 mL), NaH (60%, 0.56 g, 23.4 mmol, 1.5 equiv), methane, MOMBr (2.94 g, 23.5mmol, 1.5 equiv). The resulting solution was stirred for 2 h at room temperature. The resulting solution was diluted with 30 mL of water. The resulting solution was extracted with 2x100 mL of ethyl acetate dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/hexane (20%). This resulted in 3 g (64.0%) of 6-bromo-7-(methoxymethoxy)-2-methylchromen-4-one as a yellow solid. m/z: 299(M+H + ).

[0690] Step 6: Synthesis of 7-(methoxymethoxy)-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)chromen-4-one (B19). Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 6-bromo-7-(methoxymethoxy)-2-methylchromen-4-one (3.0 g, 10.03 mmol, 1.0 equiv), bis(pinacolato)diboron (3.06 g, 12.05 mmol, 1.2 equiv), Pd(dppf)Cl 2 (734 mg, 1.0 mmol, 0.10 equiv), KOAc (1.97 g, 20.06 mmol, 2.0 equiv), 1,4-dioxane (30 mL). The resulting solution was stirred for 2 hr at 100 °C. The resulting solution was diluted with 300 mL of EtOAc. The resulting mixture was washed with 2x50 ml of water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/hexane (50%). This resulted in 2 g (57.60%) of 7-(methoxymethoxy)-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)chromen-4-one as yellow oil. m/z:347(M+H + ).

Synthesis of 6-(methoxymethoxy)-3-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl) quinazolin- 4(3H)-one (B20).

[0691] Step 1: Synthesis of 4-bromo-5-methoxy-2-nitrobenzoate. Into a 100-mL round-bottom flask, was placed methyl 4-bromo-5-fluoro-2-nitrobenzoate (25.0 g, 89.9 mmol, 1.0 equiv), MeOH (250 mL), MeONa (5.83 g, 107.9 mmol, 1.2 equiv). The resulting solution was stirred for overnight at 30 o C. The resulting mixture was concentrated under vacuum. The residual was then quenched by the addition of 100 mL of water. The resulting solution was extracted with 2x100 mL of ethyl acetate and concentrated under vacuum. This resulted in 25 g (95.9%) of methyl 4-bromo-5-methoxy-2-nitrobenzoate as a white solid.

[0692] Step 2: Synthesis of 4-bromo-5-methoxy-2-nitrobenzoic acid.4-bromo-5-methoxy-2-nitrobenzoic acid. Into a 500-mL round-bottom flask, was placed methyl 4-bromo-5-methoxy-2-nitrobenzoate (25.0 g, 86.19 mmol, 1.0 equiv), MeOH (250 mL), H 2 O (50 mL), LiOH (4.13 g, 173 mmol, 2.0 equiv). The resulting solution was stirred for 2 h at room temperature. The resulting mixture was concentrated under vacuum. The reaction was then quenched by the addition of 250 mL of water. The pH value of the solution was adjusted to 5 with HCl (1 mol/L). The solids were collected by filtration. This resulted in 23 g (96.7%) of 4-bromo-5- methoxy-2-nitrobenzoic acid as a white solid.

[0693] Step 3: Synthesis of 4-bromo-5-methoxy-N-methyl-2- nitrobenzamide. Into a 250-mL round- bottom flask, was placed 4-bromo-5-methoxy-2-nitrobenzoic acid (10.00 g, 36.2 mmol, 1.0 equiv), DMF (100 mL), methanamine, hydrochloride (3.67 g, 54.34 mmol, 1.50 equiv), HATU (20.66 g, 54.33 mmol, 1.50 equiv), DIEA (14.05 g, 108.71 mmol, 3.0 equiv). The resulting solution was stirred for 1 h at room temperature. The reaction was then quenched by the addition of 500 mL of water/ice. The solids were collected by filtration and then dried under vacuum. This resulted in 9 g (86%) of 4-bromo-5-methoxy-N- methyl-2- nitrobenzamide as a white solid.

[0694] Step 4: Synthesis of 2-amino-4-bromo-5-methoxy-N-methylbenzamide. Into a 500-mL round- bottom flask, was placed 4-bromo-5-methoxy-N-methyl-2-nitrobenzamide (9.00 g, 31.13 mmol, 1.0 equiv), THF (200 mL), H2O (50 mL), NH4Cl (16.65 g, 311.3 mmol, 10.0 equiv), Zn (20.36 g, 311.32 mmol, 10.0 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 300 mL of water. The solids were filtered out. The filtrate was extracted with 2x200 mL of ethyl acetate and the organic layers combined and concentrated under vacuum. This resulted in 10 g (crude) of 2- amino-4-bromo-5-methoxy-N-methylbenzamide as a light yellow solid. [0695] Step 5: Synthesis of 7-bromo-6-methoxy-3-methylquinazolin-4-one. Into a 250-mL 3-necked round-bottom flask, was placed 2-amino-4-bromo-5-methoxy-N-methylbenzamide (5.00 g, 19.30 mmol, 1.0 equiv), CH(OMe) 3 (50 mL), MeOH (50 mL), p-TsOH (0.33 g, 1.93 mmol, 0.10 equiv). The resulting solution was stirred for 4 h at 75 o C in an oil bath. The resulting mixture was concentrated under vacuum. The residual was slurried in 200 mL of water. The solids were collected by filtration. This resulted in 3.6 g (69.3%) of 7- bromo-6-methoxy-3-methylquinazolin-4-one as a white solid.

[0696] Step 6: Synthesis of 7-bromo-6-hydroxy-3-methylquinazolin-4(3H)-one. To a solution of 7- bromo-6-methoxy-3-methylquinazolin-4(3H)-one (3.6 g, 13.43 mmol, 1.0 equiv) in CH 2 Cl 2 (20 mL) at 25 o C was added BBr 3 (30 mL, 1N in CH 2 Cl 2 ) and stirred at 25 o C for 2 h, monitored by LCMS. Then water (15 mL) was added. The organic layer was extracted with water (100 mL X 2). The combined aqueous layers were adjusted with saturated K 2 CO 3 aqueous solution to pH to 8~9 and extracted with dichloromethane (240 mL). The organic layers were dried over anhydrous Na 2 SO 4 , filtered and concentrated to give 7-bromo-6-hydroxy- 3-methylquinazolin-4(3H)-one (3.2 g, crude) as a yellow solid, which was used directly to next step.

[0697] Step 7: Synthesis of 7-bromo-6-(methoxymethoxy)-3-methylquinazolin-4(3H)-one. Into a 50-mL round-bottom flask, was placed 7-bromo-6-hydroxy-3-methylquinazolin-4(3H)-one (3.2 g, 12.598 mmol, 1.00 equiv), THF (30.00 mL). The solution was stirred at 0 o C and NaH (0.36 g, 15.118 mmol, 1.20 equiv) was added slowly. The resulting solution was stirred for 0.5 h at 0 °C. Then bromomethoxy-methane (1.39 g, 11.11 mmol, 1.1 equiv) was added slowly. The resulting solution was allowed to stir for an additional 1 h at 0 o C. The reaction was then quenched by the addition of 20 mL of water/ice. The resulting solution was extracted with 2x30 mL of ethyl acetate and then concentrated under vacuum. This resulted in 2 g (crude) of 7-bromo-6-(methoxymethoxy)- 3-methylquinazolin-4(3H)-one as off-white solid.

[0698] Step 8: Synthesis of 6-(methoxymethoxy)-3-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl) quinazolin-4(3H)-one (B20). Into a 40 mL vial were added 7-bromo-6-(methoxymethoxy)-3- methylquinazolin-4(3H)-one (2.00 g, 6.710 mmol, 1.00 equiv), bis(pinacolato)diboron (2.54 g, 10.0 mmol, 1.5 equiv), Pd(dppf)Cl 2 .CH 2 Cl 2 (0.46 g, 0.536 mmol, 0.08 equiv), 1,4-dioxane (25 mL) and KOAc (1.32 g, 13.4 mmol, 2.0 equiv). The resulting mixture was stirred for overnight at 100 o C under nitrogen atmosphere. The reaction was then quenched by the addition of 20 mL of water/ice. The resulting solution was extracted with 2x30 mL of ethyl acetate and then concentrated under vacuum. This resulted in 1.2g (crude) of 6- (methoxymethoxy)-3-methyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxa borolan-2-yl) quinazolin-4(3H)-one as a light green oil.

Synthesis of 2-fluoro-5-(methoxymethoxy)-N,N-dimethyl-4-(4,4,5,5-tetramet hyl-1,3,2-dioxaborolan-2- yl)benzamide (B22).

[0699] Step 1: Synthesis of 4-bromo-2-fluoro-5-hydroxybenzoic acid. Into a 1-L 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 2-fluoro-5-hydroxybenzoic acid (50.0 g, 320.3 mmol, 1.0 equiv), CHCl 3 (500 mL). This was followed by the addition of Br 2 (61.4 g, 384.4 mmol, 1.2 equiv) at 0 °C over 30 min. The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 300 mL of water (Na 2 S 2 O 3 ). The resulting solution was extracted with 3x500 mL of dichloromethane dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column and eluted with ethyl acetate/hexane (20%). This resulted in 45 g (59.8%) of 4-bromo-2-fluoro-5-hydroxybenzoic acid as yellow oil. m/z: 238(M+H + ).

[0700] Step 2: Synthesis of 4-bromo-2-fluoro-5-hydroxy-N,N-dimethylbenzamide. Into a 1-L 3-necked round-bottom flask, was placed 4-bromo-2-fluoro-5-hydroxybenzoic acid (20.0 g, 85.1 mmol, 1.0 equiv), THF (300 mL), triethylamine (19.8 g, 195.7 mmol, 2.3 equiv). This was followed by the addition of a solution of isopropyl chloroformate (26.0 g, 211.9 mmol, 2.5 equiv) in MeOH (300 mL) dropwise with stirring at 0 o C in 20 min. To this was added dimethylamine (7.60 g, 168.5 mmol, 2 equiv) at 0 o C. The resulting solution was stirred for 2 h at room temperature. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column and eluted with ethyl acetate/hexane (50%). This resulted in 7 g (31.4%) of 4- bromo-2-fluoro-5-hydroxy-N,N-dimethylbenzamide as a yellow solid. LCMS m/z: 262 (M+H + ).

[0701] Step 3: Synthesis of 4-bromo-2-fluoro-5-(methoxymethoxy)-N,N-dimethylbenzamide. Into a 250- mL 3-necked round-bottom flask, was placed 4-bromo-2-fluoro-5-hydroxy-N,N-dimethylbenzamide (8.0 g, 30.53 mmol, 1.0 equiv), THF (100 mL), NaH (1.48 g, 61.6 mmol, 2.0 equiv), methane, bromomethoxy- (5.72 g, 45.77 mmol, 1.5 equiv). The resulting solution was stirred for 2 h at room temperature. The resulting solution was diluted with 500 mL of EtOAc. The resulting mixture was washed with 2 x100 ml of water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column eluting with ethyl acetate/hexane (50%). This resulted in 6 g (64.2%) of 4-bromo-2- fluoro-5-(methoxymethoxy)-N,N-dimethylbenzamide as colorless oil. m/z:306 (M+H).

[0702] Step 4: Synthesis of 2-fluoro-5-(methoxymethoxy)-N,N-dimethyl-4-(4,4,5,5-tetramet hyl-1,3,2- dioxaborolan-2-yl)benzamide (B22). Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed Pd(dppf)Cl 2 (977.2 mg, 1.34 mmol, 0.07 equiv), 4-bromo-2- fluoro-5-hydroxy-N,N-dimethylbenzamide (5.0 g, 19.08 mmol, 1.0 equiv), bis(pinacolato)diboron (5.81 g, 22.8 mmol, 1.2 equiv), KOAc (3.74 g, 38.16 mmol, 2.0 equiv), Dioxane (100 mL). The resulting solution was stirred overnight at 100 °C. The resulting solution was diluted with 500 mL of EtOAc. The resulting mixture was washed with 2x100 ml of water. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column eluting with ethyl acetate/hexane (50%). This resulted in 3 g (44.5%) of 2-fluoro-5-(methoxymethoxy)-N,N-dimethyl-4-(4,4,5,5-tetramet hyl-1,3,2- dioxaborolan-2-yl)benzamide as yellow oil. m/z:354.1(M+H + ).

Synthesis of 2-[4-chloro-5-fluoro-2-(methoxymethoxy)phenyl]-4,4,5,5-tetra methyl-1,3,2-dioxaborolane (B23).

[0703] Step 1: Synthesis of 2-bromo-5-chloro-4-fluorophenol. Into a 500-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 3-chloro-4-fluorophenol (30.0 g, 204.7 mmol, 1.0 equiv), CH 2 Cl 2 (300 mL), Br 2 (39.26 g, 245.65 mmol, 1.2 equiv). The resulting solution was stirred overnight at room temperature. The reaction was then quenched by the addition of 500 mL of water/ice. The resulting solution was extracted with 3x500 mL of CH 2 Cl 2 and the organic layers combined. The resulting mixture was washed with 1 x500 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column eluting with ethyl acetate/petroleum ether (0-20%). This resulted in 25 g (54.2%) of 2-bromo-5-chloro-4-fluorophenol as a yellow solid.

[0704] Step 2: Synthesis of 1-bromo-4-chloro-5-fluoro-2-(methoxymethoxy)benzene. Into a 250 mL 3- necked round-bottom flask were added 2-bromo-5-chloro-4-fluorophenol (8.00 g, 35.49 mmol, 1.0 equiv) and THF (110 mL). To the above mixture was added NaH (1.70 g, 42.50 mmol, 1.20 equiv, 60wt%) in portions at 0 °C. The resulting mixture was stirred for additional 30 min at 0 °C. To the above mixture was added bromo(methoxy)methane (8.00 g, 64.02 mmol, 1.8 equiv) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature. The reaction was quenched by the addition of saturated NH 4 Cl aq. (70 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE to afford 1-bromo-4- chloro-5-fluoro-2-(methoxymethoxy)benzene (8.1g, 84.7%) as a colorless oil.

[0705] Step 3: Synthesis of 2-[4-chloro-5-fluoro-2-(methoxymethoxy)phenyl]-4,4,5,5-tetra methyl-1,3,2- dioxaborolane (B23). Into a 500-mL 3-necked round-bottom flask, was placed a solution of 1-bromo-4- chloro-5-fluoro-2-(methoxymethoxy)benzene (20.0 g, 74.21 mmol, 1.0 equiv) in dioxane (200 mL), bis(pinacolato)diboron (28.27 g, 111.33 mmol, 1.5 equiv), Pd(dppf)Cl 2 (2.72 g, 3.71 mmol, 0.05 equiv), KOAc (14.57 g, 148.4 mmol, 2 equiv). The resulting solution was stirred for 3 h at 100 °C. The resulting mixture was concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (3:100). This resulted in 13 g (55.3%) of 2-[4-chloro-5-fluoro-2-(methoxymethoxy)phenyl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane as a solid.

Synthesis of (6-(methoxymethoxy)-4-oxo-3-(2,2,2-trifluoroethyl)-3,4-dihyd roquinazolin-7-yl)boronic acid (B26).

[0706] Step 1: Synthesis of 4-Bromo-5-hydroxy-2-nitrobenzoic acid. KOH (17 g, 303 mmol) was added to a suspension of 4-bromo-5-fluoro-2-nitrobenzoic acid (20 g, 75.8 mmol) in water (100 mL). The mixture was heated to 80 ºC for 5 h. After cooled to room temperature, the mixture was acidified to pH of 4 by HCl aqueous solution. The precipitate was collected by filtration and dried on air to give 20 g of crude product 4- bromo-5-hydroxy-2-nitrobenzoic acid as yellow solid (91% yield). LCMS: m/z 283.9 [M+Na] + ; t R = 1.47 min.

[0707] Step 2: Synthesis of 4-bromo-5-hydroxy-2-nitro-N-(2,2,2-trifluoroethyl)benzamide. 2,2,2- Ttrifluoroethanamine (11.2 g, 113.7 mmol), DIPEA (19.6 g, 151.6 mmol) and HATU (43.2 g, 113.7 mmol) were added to a suspension of 4-bromo-5-hydroxy-2-nitrobenzoic acid (20 g, 75.8 mmol) in 50 mL of DMF and 50 mL of CH 3 CN. The mixture was stirred at room temperature for 1 h. The reaction mixture was concentrated to give crude 4-bromo-5-hydroxy-2-nitro-N-(2,2,2-trifluoroethyl)benzamide as yellow oil, which was used directly to next step. LCMS: m/z 342.9 [M+H] + ; t R = 1.58 min.

[0708] Step 3: Synthesis of 2-amino-4-bromo-5-hydroxy-N-(2,2,2-trifluoroethyl)benzamide. Raney- Nickel (1.17 g, 20.0 mmol) was added to a solution of 4-bromo-5-hydroxy-2-nitro-N-(2,2,2- trifluoroethyl)benzamide (crude from above) and hydrazine hydrate (11 g, 220 mmol) in MeOH (200 mL). The mixture was stirred at room temperature for 2 h and filtered through Celite® brand filter agent. The filtrate was concentrated. The residue was purified on silica gel chromatography (0-50% EtOAc/petroleum ether) to give 19 g (total yield 76% for 3 steps) of 2-amino-4-bromo-5-hydroxy-N-(2,2,2- trifluoroethyl)benzamide as off-white solid. LCMS: m/z 313.1 [M+H] + ; t R = 0.89 min.

[0709] Step 4: Synthesis of 7-bromo-6-hydroxy-3-(2,2,2-trifluoroethyl)quinazolin-4(3H)-o ne. To a solution of 2-amino-4-bromo-5-hydroxy-N-(2,2,2-trifluoroethyl)benzamide (13.5 g, 43.3 mmol) and triethoxymethane (19.2 g, 129.8 mmol) in EtOH (80 mL) was added catalytic amount of 4- methylbenzenesulfonic acid (0.39 g, 2.2 mmol). The mixture was heated to 80 ºC for 2 h, concentrated and the residue was triturated with petroleum ether. The solid was collected by filtration to give 9.4 g of 7-bromo-6- hydroxy-3-(2,2,2-trifluoroethyl)quinazolin-4(3H)-one as pink solid. The filtrate was concentrated and the residue was purified by silica gel chromatography (0-40% EtOAc/petroleum ether) to give 1.6 g of 7-bromo- 6-hydroxy-3-(2,2,2-trifluoroethyl)quinazolin-4(3H)-one as pink solid (79% yield). LCMS: m/z 325.1 [M+H] + ; t R = 1.74 min.

[0710] Step 5: Synthesis of 7-bromo-6-(methoxymethoxy)-3-(2,2,2-trifluoroethyl)quinazoli n-4(3H)-one. NaH (2.1 g, 52.5 mmol, 60% in mineral oil) was added to a solution of 7-bromo-6-hydroxy-3-(2,2,2- trifluoroethyl)quinazolin-4(3H)-one (11.5 g, 35.5 mmol) in DMF (40 mL) at 0 ºC. The mixture was stirred for 30 min at 0 ºC, then bromomethyl methyl ether (3.5 mL, 42.8 mmol) was added. The resulting mixture was warmed to room temperature and stirred for 2 h, quenched with water and extracted with EtOAc (80 mL x 3). The extracts were washed with water and brine, and concentrated. The residue was triturated with

EtOAc/petroleum ether (50%). The precipitate was filtered to give 8 g of 7-bromo-6-(methoxymethoxy)-3- (2,2,2-trifluoroethyl)quinazolin-4(3H)-one as white solid. The filtrate was concentrated and purified silica gel chromatography (0-20% EtOAc/CH 2 Cl 2 ) to give an additional 3.8 g of 7-bromo-6-(methoxymethoxy)-3- (2,2,2-trifluoroethyl)quinazolin-4(3H)-one as white solid (90% yield). LCMS: m/z 369.0 [M+H] + ; t R = 1.97 min.

[0711] Step 6: Synthesis of 6-(methoxymethoxy)-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)-3- (2,2,2-trifluoroethyl)quinazolin-4(3H)-one (B26). To a solution of 7-bromo-6-(methoxymethoxy)-3-(2,2,2- trifluoroethyl)quinazolin -4(3H)-one (7.8 g, 21.2 mmol) in dioxane ( 60 mL) were added KOAc (6.6 g, 67.4 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (11.4 g, 45 mmol) and Pd(dppf)Cl 2 (823 mg, 1.13 mmol). The mixture was heated to 100 ºC under N 2 for 5 h. After concentration, the mixture was purified through a short silica gel column (EtOAc/petroleum ether, 50%) to give 8 g of crude product. LCMS showed mainly boronic acid signal. LCMS: m/z 333.1 [M+H] + ; t R = 1.60 min; m/z 415.0 [M+H] + ; t R = 2.05 min. Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)qui noxaline (B27).

[0712] Step 1: Synthesis of N-(5-bromo-4-methoxy-2-nitrophenyl)acetamide.3-Bromo-4-metho xyaniline (5 g, 0.025 mmol) was added to the acetic anhydride (11.7 mL). The reaction was stirred for 1 h at room temperature. The mixture was added dropwise to the stirred solution of acetic anhydride (9.5 mL) and AcOH (6.5 mL) in HNO 3 (1.8 mL, 0.028 mmol) at 0 o C. Then reaction was stirred for 2 h at room temperature, quenched with saturated NaHCO 3 aqueous solution. The mixture was extracted by EtOAc. The combined organic solvents were concentrated under reduced pressure. The residue was purified by silica gel flash column chromatography (0-10% EtOAc/petroleum ether) to give 6.5 g of N-(5-bromo-4-methoxy-2- nitrophenyl)acetamide (88% yield) as a black solid. LCMS: m/z 289.0 [M+H] + ; t R = 1.68 min.

[0713] Step 2: Synthesis of 5-bromo-4-methoxy-2-nitroaniline. KOH (2.4 g, 43.241 mmol) was added to a solution of N-(5-bromo-4-methoxy-2-nitrophenyl)acetamide (2.5 g, 8.648 mmol) in 25 mL of EtOH. The reaction mixture was stirred for 2 h at 80 °C, and concentrated under reduced pressure. EtOAc and water was added and the organic layer separated. The organic phase was dried over Na 2 SO 4 and concentrated to give 2.0 g crude of 5-bromo-4-methoxy-2-nitroaniline (88% yield) as a black solid. LCMS: m/z [M+H] + ; t R = 1.70 min.

[0714] Step 3: Synthesis of 4-bromo-5-methoxybenzene-1,2-diamine. Fe (5.2 g, 93.1 mmol) and AcOH (5 mL) was added to a solution of 5-bromo-4-methoxy-2-nitroaniline (2.3 g, 9.310 mmol) in 20 mL of THF. The reaction mixture was stirred for 2 h at 80 °C. After cooling to room temperature, the mixture was filtered, and the filtrate was concentrated under reduced pressure. Water was added and the mixture was extracted with EtOAc. The combined organic solvents were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by flash silica gel column chromatography (0-50% EtOAc/petroleum ether) to give 1.5 g of 4-bromo-5-methoxybenzene-1,2-diamine (41% yield) as a black solid. LCMS: m/z 217.0 [M+H] + ; t R = 1.44 min. [0715] Step 4: Synthesis of 6-bromo-7-methoxyquinoxaline.1,4-dioxane-2,3-diol (0.603 g, 5.017 mmol) was added to a solution of 4-bromo-5-methoxybenzene-1,2-diamine (1.089 g, 5.0 mmol) in 25 mL of EtOH. The reaction mixture was stirred overnight at room temperature, concentrated under reduced pressure, and purified by silica gel column chromatography (0-20% EtOAc/petroleum ether) to give 841 mg of 6-bromo-7- methoxyquinoxaline (70% yield) as a red solid. LCMS: m/z 241.0[M+H] + ; t R = 1.65 min.

[0716] Step 5: Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)qui noxaline (B27). A mixture of 6-bromo-7-methoxyquinoxaline (200 mg, 0.837 mmol), 4,4,4',4',5,5,5',5'-octamethyl- 2,2'-bi(1,3,2-dioxaborolane) (320 mg, 1.260 mmol), Pd(dppf)Cl 2 (120 mg, 0.170 mmol), AcOK (160 mg 1.680 mmol) in 5 mL of dioxane was degassed and heated at 105 o C for 2 h under nitrogen atmoshphere. After cooling to room temperature, the crude boronic ester was usded directly to next step.

Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cin noline (B28).

[0717] Step 1: Synthesis of 4-bromo-5-methoxy-2-nitroaniline. NBS (11.6 g, 65 mmol) was added to a solution of 5-methoxy-2-nitroaniline (10 g, 59.5 mmol) in acetonitrile (100 mL) under N 2 protection. The reaction mixture was stirred and refluxed overnight. After cooling to room temperature, the mixture was quenched with water,and concentrated to give the crude product. The crude peoduct was washed with water to give 14 g of 4-bromo-5-methoxy-2-nitroaniline (89% yield) as a brown solid. LCMS: m/z 247.1 [M+H] + ; t R = 1.69 min.

[0718] Step 2: Synthesis of 1-bromo-4-iodo-2-methoxy-5-nitrobenzene. H 2 SO 4 (conc, 2.25 mL) was added dropwise to a solution of 4-bromo-5-methoxy-2-nitroaniline (4 g, 16.3 mmol) in acetonitrile (100 mL) under N 2 protection at -20 °C. Then NaNO 2 (2.25 g, 2 eq) was added slowly. The reaction mixture was stirred at -20 °C for 30 min, KI (10.8 g, 4 eq) was added at the same temperature.100 mL of water was added to quench the reaction and the resulting mixture was extracted with DCM (180 mL X 3). The combined organic phases were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-50% EtOAc/petroleum ether) to give 2.5 g of 1-bromo-4-iodo-2-methoxy-5-nitrobenzene (42.8% yield) as a yellow solid. LCMS: m/z 358.2 [M+H] + ; tR = 1.88 min.

[0719] Step 3: Synthesis of 5-bromo-2-iodo-4-methoxyaniline. Fe (550 mg, 1.0 equiv) and HCl (0.98 ml, 1M) were added to a solution of 1-bromo-4-iodo-2-methoxy-5-nitrobenzene (3.5g, 9.8 mmol) in EtOH (10 mL). The reaction mixture was stirred at 80 °C for 2 h, filtered and the filtrate’s pH was adjusted to pH 8-9 by saturated K 2 CO 3 aqueous solution. Then the mixture was extracted with DCM (50 mL X 3). The combined organic phases were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give 3 g of 5-bromo-2-iodo-4-methoxyaniline (93.6% yield) as a yellow solid. LCMS: m/z 328.1 [M+H] + ; t R = 1.75 min. [0720] Step 4: Synthesis of (E)-1-(5-bromo-2-iodo-4-methoxyphenyl)-3,3-diethyltriaz-1-en e.5-bromo-2- iodo-4-methoxyaniline (4.2 g, 12.8 mmol) was dissolved in a minimum amount of MeCN. HCl (12 M, 8.5 mL, conc.) and ice (10 g) were added. The suspension was cooled to -5 °C, and a solution of NaNO 2 (1.9 g, 2.2 equiv) in 1.5 mL of water and 0.5 mL of CH 3 CN was added slowly during which the temperature was kept between -5 and -2 °C. Once the addition was complete, the solution was stirred at -5 °C for 30 min, and then it was transferred slowly via cannula to a solution of Et 2 NH (13.3 mL, 10 equiv) and K 2 CO 3 (8.8 g, 5 equiv) in 3:1 water/CH 3 CN (0.1 M) and then cooled to 0 °C. Once the transfer was complete, the mixture was allowed to gradually warm to room temperature overnight, diluted with water and extracted with Et 2 O. The combined organic layers were dried over MgSO 4 , filtered, concentrated and purified by silica gel chromatography (0- 20% EtOAc/petroleum ether) to give 4 g of (E)-1-(5-bromo-2-iodo-4-methoxyphenyl)-3,3-diethyltriaz-1-en e (80% yield). LCMS: m/z 412.1 [M+H] + ; t R = 2.21 min.

[0721] Step 5: Synthesis of (E)-1-(5-bromo-4-methoxy-2-((trimethylsilyl)ethynyl)phenyl)- 3,3- diethyltriaz-1-ene. A solution of (E)-1-(5-bromo-2-iodo-4-methoxyphenyl)-3,3-diethyltriaz-1-en e (4 g, 9.73 mmol), Pd (PPh 3 ) 2 Cl 2 (273 mg, 0.04 equiv), CuI (130 mg, 0.07 equiv) and (trimethylsilyl) acetylene (1.43 mL, 10.2 mmol) in triethylamine (100 mL) was immediately degassed and heated to 50 °C with stirring overnight. After cooling to room temperature, the mixture was filtered, concentrated and purified by silica gel chromatography (0-5% EtOAc/petroleum ether) to give 3.2 g of (E)-1-(5-bromo-4-methoxy-2- ((trimethylsilyl)ethynyl)phenyl)-3,3-diethyltriaz-1-ene (86% yield) as a white solid. LCMS: m/z 382.1

[M+H] + ; t R = 2.53 min.

[0722] Step 6: Synthesis of (E)-1-(5-bromo-2-ethynyl-4-methoxyphenyl)-3,3-diethyltriaz-1 -ene. To a solution of (E)-1-(5-bromo-4-methoxy-2-((trimethylsilyl)ethynyl)phenyl)- 3,3-diethyltriaz-1-ene (3.2 g, 8.4 mmol) in 100 mL of THF and 33 mL of H 2 O was added K 2 CO 3 (11.6 g, 84 mmol). The mixture was stirred at room temperature under N 2 protection for 4 h. Water was added to quench the reaction. The resulting mixture was extracted with EtOAc (100 ml X 3). The combined organic phases were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-10% EtOAc/petroleum ether) to give 2.5 g of (E)-1-(5-bromo-2-ethynyl-4-methoxyphenyl)-3,3-diethyltriaz-1 -ene (96% yield) as a yellow solid. LCMS: m/z 310.2 [M+H] + ; t R = 2.10 min.

[0723] Step 7: Synthesis of 7-bromo-6-methoxycinnoline. A solution of (E)-1-(5-bromo-2-ethynyl-4- methoxyphenyl)-3,3-diethyltriaz-1-ene (2.5 g, 8 mmol) in 1,2-dichlorobenzene (100 mL) was stirred at 200 °C under N 2 protection for 4 h. After cooling to room temperature, the mixture was concentrated and purified by silica gel chromatography (0-50% EtOAc/petroleum ether) to give 450 mg of 7-bromo-6-methoxycinnoline (23% yield) as a yellow solid. LCMS: m/z 239.1 [M+H] + ; t R = 1.55 min.

[0724] Step 8: Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cin noline (B28). To a solution of 7-bromo-6-methoxycinnoline (450 mg, 1.89 mmol) in dioxane (100 mL) was added

Bis(pinacolato)diboron (720 mg, 2.84 mmol), Pd(dppf)Cl 2 (276 mg, 0.378 mmol) and KOAc (370 mg, 3.78 mmol). The reaction mixture was stirred at 105 o C under N 2 protection overnight. After cooling to room temperature, the crude 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)cin noline (400 mg) was directly in next step. LCMS: m/z 205.1 [M+H] + ; t R = 1.08 min.

Synthesis of (2-hydroxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (B29).

[0725] Step 1: Syntheisis of 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole. K 2 CO 3 (4.04 g, 29.2 mmol ) was added to a solution of 4-fluoro-2-methoxy-1-nitrobenzene (5 g, 29.2 mmol) and 4,5- dibromo-2H-1,2,3-triazole (6.63 g, 29.2 mmol) in DMF (100 mL). The resulting mixture was stirred at 80 °C for 16 h. After cooling to room temperature, the mixture was poured into ice-water (100 mL) and extracted with EtOAc. The organic layers were washed with water (100 mL), dried over anhydrous MgSO 4 , concentrated in vacuum to give 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole (10 g , 97%) as a white solid. LCMS: m/z 378.9 [M+H] + ; t R =1.250 min.

[0726] Step 2: Synthesis of 2-methoxy-4-(2H-1,2,3-triazol-2-yl)aniline. Pd/C (1 g 10% on activated carbon) was added to a solution of 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole (10 g, 26.5 mmol) in MeOH (150 mL). The mixture was stirred under hydrogen atmosphere for 5 h and filtered. The filtrate was concentrated in vacuum to give 2-methoxy-4-(2H-1,2,3-triazol-2-yl)aniline (5 g, yield 98%) as a white solid. LCMS: m/z 191 [M+H] + ; t R =0.574 min.

[0727] Step 3: Synthesis of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid. A pre-cooled (- 15°C) solution of t-BuONO (2.61 g, 25.3 mmol) and 2-methoxy-4-(2H-1,2,3- triazol-2-yl)aniline (4.0 g, 21 mmol) in AcOH (80 mL) was added dropwise to a precooled solution of TfOH (3.79 g, 25.3 mmol) in AcOH (80 mL). The reaction was stirred for 10~20 min at 10-15°C, then poured into cold Et 2 O (1000 mL). The precipitated diazonium salt was collected by filtration and dried in vacuum to give 2-methoxy-4-(2H-1,2,3- triazol-2-yl)benzenediazonium, trifluoromethanesulfonic salt (7.4 g, yield 95%) as a white solid. LCMS: m/z 202.2 [M+] + ; t R = 0.737 min. The 2-methoxy-4-(2H-1,2,3-triazol-2-yl)benzenediazonium,

trifluoromethanesulfonic salt (7.4 g, 21 mmol) was disolved in water (150 mL). Hypodiboric acid (4.74 g, 52.7 mmol) was added. The reaction mixture was stirred at 25 °C for 3 h. The precipitate was collected by filtration and dried in vacuum to give (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (4.6 g, yield 99%) as a white solid. LCMS: m/z 220.2 [M+H] + ; t R =1.127 min.

[0728] Step 4: Synthesis of (2-hydroxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid. BBr 3 (4.6 mL, 18.26 mmol, 4M) was added to a solution of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (1000 mg, 4.57 mmol) in DCM (4 mL). The reaction mixture was stirred at 20 °C for 18 h under N 2 atmosphere, concentrated in vacuum and purified by silica gel column (10-25% EtOAc/petroleum ether) to give (2- hydroxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (450 mg, 59 % yield) as a yellow solid. LCMS: m/z 206.2 [M+H] + ; t R = 0.898 min.

Synthesis of (2-methoxy-4-((1-methyl-1H-pyrazol-4-yl)oxy)phenyl)boronic acid (B30). [0729] Step 1: Synthesis of 4-(3-methoxy-4-nitrophenoxy)-1-methyl-1H-pyrazole. A mixture of 4-fluoro- 2-methoxy-1-nitrobenzene (2.1 g, 12.2 mmol), 1-methyl-1H-pyrazol-4-ol (1.0 g, 10.2 mmol) and K 2 CO 3 (2.82 g, 20.4 mmol) in DMSO (50 mL) was degassed and stirred at 110 °C under nitrogen for 16 h. After cooling to the ambient temperature, the mixture was extracted with ethyl acetate (20 mL x 2). The combined organic layers were dried over anhydrous sodium sulfate. The residue was purified with silica gel chromatography (10% of ethyl acetate in petroleum ether) to give 4-(3-methoxy-4-nitrophenoxy)-1-methyl-1H-pyrazole (2.83 g, 90% yield) as a white solid . LCMS: m/z 250.1 [M+H] + ; t R = 1.62 min.

[0730] Step 2: Synthesis of 2-methoxy-4-((1-methyl-1H-pyrazol-4-yl)oxy)aniline. Pd/C (150 mg, 10% on activated carbon) was added to a solution of 4-(3-methoxy-4-nitrophenoxy)-1-methyl-1H-pyrazole (1.416 g) in methanol (30 mL). The resulting mixture was stirred at 25 ć under hydrogen for 3 h, filtered, concentrated and purified with silica gel chromatography (33% ethyl acetate/petroleum ether) to give 2-methoxy-4-((1- methyl-1H-pyrazol-4-yl)oxy)aniline (1.01 g, 86% yield) as a brown oil. LCMS: m/z 220.1 [M+H] + ; t R = 0.67 min.

[0731] Step 3: Synthesis of 2-methoxy-4-((1-methyl-1H-pyrazol-4-yl)oxy)benzenediazonium, triflic salt. TfOH (805 mg, 5.364 mmol) in acetic acid (25 mL) was added to a solution of 2-methoxy-4-((1-methyl-1H- pyrazol-4-yl)oxy)aniline (1.0 g, 4.47 mmol) and t-BuONO (1.31 g, 11.175 mmol) in acetic acid (25 mL). The resulting mixture was stirred at 10 o C under nitrogen for 20 mins. The mixture was poured to cold diethyl ether (70 mL), the precipitated diazonium salt was collected by filtration and died in vacuum to give 2- methoxy-4-((1-methyl-1H-pyrazol-4-yl)oxy)benzenediazonium, triflic salt (1.65 g , 92% yield) as a brown oil. LCMS: m/z 231.1 [M+H] + ; t R = 0.38 min.

[0732] Step 4: Synthesis of (2-methoxy-4-((1-methyl-1H-pyrazol-4- yl)oxy)phenyl)boronic acid. 2- methoxy-4-((1-methyl-1H-pyrazol-4-yl)oxy)benzenediazonium, triflic salt (1.652 g, 4.127 mmol) was dissolved in water (12 mL). B 2 (OH) 4 (925 mg, 10.317 mmol) was added, and the resulting mixture was stirred at 25 o C under nitrogen for 3 h. The mixture was extracted with ethyl acetate (20 mL X 2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and then concentrated under reduced pressure. The residue was purified with silica gel chromatography (20% of methanol in dichloromethane) to give (2-methoxy-4-((1-methyl-1H-pyrazol-4- yl)oxy)phenyl)boronic acid (407 mg, 33% yield) as a brown oil. LCMS: m/z 249.1 [M+H] + ; t R = 1.09 min.

Synthesis of 4-(4-methoxybenzyloxy)-5-(4,4,5,5-tetramethyl-1,3,2-dioxabor olan-2 yl)picolinonitrile (B31).

[0733] Step 1: Synthesis of 3-bromo-4-(4-methoxybenzyloxy)pyridine. NaH (0.6 g, 15.0 mmol, 60% in mineral oil) was added to a solution of (4-methoxyphenyl)methanol (1.38 g, 10.0 mmol) in DMF (30 mL) at 0 o C. After stirring for 20 min, 3-bromo-4-chloropyridine (1.78 g, 9.3 mmol) was added in portions. The mixture was stirred at 80 ºC for 2 h. After cooling to room temperature, the reaction was quenched with water and the precipitate was collected by filtration and dried in vacuo to give 3-bromo-4-(4- methoxybenzyloxy)pyridine as white solid (2.71 g), which was used directly to next step. LCMS: m/z 294.1 [M+H] + . t R = 1.11 min.

[0734] Step 2: Synthesis of 3-bromo-4-(4-methoxybenzyloxy)pyridine 1-oxide. m-CPBA (2.8 g, 16.3 mmol) was added to a solution of 3-bromo-4-(4-methoxybenzyloxy)pyridine (2.71 g, 9.2 mmol) in CH 2 Cl 2 (30 mL) at room temperature. The mixture was stirred for 20 h and quenched with 1N NaOH aqueous solution and extracted with CH 2 Cl 2 (50 mL X 2). The organic layers were concentrated to give 3-bromo-4-(4- methoxybenzyloxy)pyridine 1-oxide (2.9 g), which was used directly to next step. LCMS: m/z 312.1 [M+H] + . t R =1.24 min.

[0735] Step 3: Synthesis of 5-bromo-4-(4-methoxybenzyloxy)picolinonitrile. To a solution of 3-bromo-4- (4-methoxybenzyloxy)pyridine 1-oxide (2.71 g, 8.7 mmol) in CH 2 Cl 2 (30 mL) were added TMSCN (1.4 g, 14.1 mmol) and dimethylcarbamic chloride (1.6 g, 14.9 mmol). The solution was stirred at room temperature for 20 h, quenched with aqueous NaHCO 3 solution and extracted with CH 2 Cl 2 (50 mL x 2). The combined organic layers were concentrated and purified on silica chromatography (0-40% EtOAc/petroleum ether) to give 5-bromo-4-(4-methoxybenzyloxy)picolinonitrile as white solid (0.52 g, t R = 2.09 min) and 3-bromo-4-(4- methoxybenzyloxy)picolinonitrile as white solid (0.57 g, t R = 2.03 min). LCMS: m/z 343.0 [M+Na] + .

[0736] Step 4: Synthesis of 4-((4-methoxybenzyl)oxy)-5-(4,4,5,5-tetramethyl-1,3,2-dioxab orolan -2- yl)picolinonitrile (B31). A mixture of 5-bromo-4-(4-methoxybenzyloxy)picolinonitrile (0.45 g, 1.41 mmol), bis(pinacolato)diboron (716 mg, 2.82 mmol), Pd(dppf)Cl 2 (50 mg ,0.07 mmol) and KOAc (414 mg, 4.22 mmol) in dioxane (10 mL) was degassed and stirred at 100 o C under argon for 2 h. After cooling to room temperature, the mixture was quenched with water (30 mL) and extracted with EtOAc (20 mL X 2). The extracts were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 , filtered and the filtrate was concentrated and purified by silica gel chromatography (0-50% EtOAc/petroleum ether) to give 4-((4- methoxybenzyl)oxy)- 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)picolinonitri le as red oil (0.20 g).

LCMS: m/z 367.2 [M+H] + , t R = 2.12 min.

Synthesis of 2-fluoro-4-(methoxymethoxy)-5-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)benzonitrile (B32).

[0737] Step 1: Synthesis of 5-bromo-2-fluoro-4-hydroxybenzonitrile. To a mixture of 2-fluoro-4- hydroxybenzonitrile (1.37 g, 10 mmol) in CH 3 CN (20 mL) was added triflic acid (1.80 g, 12 mmol) and NBS (1.88 g, 11 mmol) portionwise at -30 o C. After addition, the resulting mixture was stirred at room temperature overnight, diluted with water (40 mL), and extracted with EtOAc (40 mL X 3). The combined organic layers were dried over anhydrous Na 2 SO 4 , concentrated in vacuo, and purified by reverse-phase chromatography (eluting with 20% CH 3 CN in water with water (0.01% TFA)) to give 678 mg of 5-bromo-2-fluoro-4- hydroxybenzonitrile (16 % yield) as a yellow solid. LCMS: m/z 216.0 [M+H] + , t R = 1.65 min. [0738] Step 2: Synthesis of 5-bromo-2-fluoro-4-(methoxymethoxy)benzonitrile. To a solution of 5- bromo-2-fluoro-4-hydroxybenzonitrile (678 mg, 3.1 mmol) in THF (15 mL) was added NaH (174 mg, 4.35 mmol, 60% in mineral oil) at 0 o C. After stirring for 0.5 h, MOMBr (584 mg, 4.67 mmol) was added at 0 o C. The mixture was stirred at room temperature for 1.5 h, quenched with water (20 mL), and extracted with EtOAc (20 mL X 3). The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (10% EtOAc/petroleum ether) to give 615 mg of 5- bromo-2-fluoro-4-(methoxymethoxy)benzonitrile (75% yield) as a white solid. LC-MS: m/z 260.0 [M+H] + , t R = 2.00 min.

[0739] Step 3: Synthesis of 2-fluoro-4-(methoxymethoxy)-5-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)benzonitrile (B32). A mixture of 5-bromo-2-fluoro-4-(methoxymethoxy)benzonitrile (615 mg, 2.4mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.2 g, 4.7 mmol), Pd(dppf)Cl 2 (194 mg, 0.24 mmol) and KOAc (698 mg, 7.1 mmol) in dioxane (8 mL) was stirred at 100 o C for 2 h under nitrogen atmoshphere. The mixture was concentrated and purified by silica gel chromatography (5% MeOH/CH 2 Cl 2 ) to give 727 mg of 2-fluoro-4-(methoxymethoxy)-5-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)benzonitrile (71% yield) as a white solid. LCMS showed boronic acid signal. LC-MS: m/z 226.1 [M+H] + , t R = 1.13 min.

Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso quinoline (B33).

[0740] Step 1: Synthesis of 2-amino-5-bromo-4-methoxybenzoic acid. NBS (6.4 g, 56 mmol) in 5 mL of DMF was added to a solution of 2-amino-4-methoxybenzoic acid (6 g, 56 mmol) in 30 mL of DMF at 0 o C. After stirring at 0 o C for 1 h, the mixture allow to warm up to room temperature and stired for 16 h. Saturated Na2SO3 (80 mL) was added and the mixture was acidified with 1 N HCl aqueous solution till pH = 3. The precipitate was collected by filtration and dried in vacuum to give 8.5 g of 2-amino-5-bromo-4- methoxybenzoic acid as white solid (96% yield), which was used directly to next step. LCMS: m/z 246.0 [M+H] + ; t R = 1.12 min.

[0741] Step 2: Synthesis of 6-bromo-7-methoxyquinazolin-4-ol. Formimidamide acetic acid (9.4 g, 90.2 mmol) was added to a solution of 2-amino-5-bromo-4-methoxybenzoic acid (8.5 g, 34.7 mmol) in 2- methoxyethan-1-ol (20 mL). The mixture was stirred at 135 o C for 16 h. After cooling to room to room temperature, the mixture was poured into 20 mL of H 2 O. The precipitate was collected by filtration and dried in vacuum to give 7.6 g of 6-bromo-7-methoxyquinazolin-4-ol (86% yield) as yellow solid, which was used directly to next step. LCMS: m/z 257.0 [M+H] + ; t R = 1.48 min.

[0742] Step 3: Synthesis of 6-bromo-4-chloro-7-methoxyquinazoline. DMF (1 m) was added to a stirred solution of 6-bromo-7-methoxyquinazolin-4-ol (7.6 g, 29.8 mmol) in 150 mL of SOCl 2 . The mixture was stirred at 80 o C for 3 h and concentrated.50 mL of satturated NaHCO3 aqueous solution was added. The resulting mixture was extracted with EtOAc (50 mL X 3). The combined orgainc solvents were washed with brine (50 mL), dried over anhhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-70% CH 2 Cl 2 /petroleum ether) to give 6.1 g of 6-bromo-4-chloro-7-methoxyquinazoline as a white solid (75% yiled). LCMS: m/z 273.0 [M+H] + ; t R = 1.90 min.

[0743] Step 4: Synthesis of 6-bromo-7-methoxyquinazoline. TsNHNH 2 (5.81 g, 31.2 mmol) was added to a solution of 6-bromo-4-chloro-7-methoxyquinazoline (6.1 g, 22 mmol) in 80 mL of CH 2 Cl 2 . After stirring at 85 °C for 3 h, the mixture was concentrated. Then EtOH (100 mL) and NaOH aqueous solution (80 mL, 2N) were added. The mixture was then stirred at 85 °C for 3 h. After cooling to room temperature, the mixture was extracted with 2-methoxy-2-methylpropane (50 mL X 4). The combined organic solvents were washed with brine (50 mL), concentrated and purified by silica gel chromatography (0-45% EtOAc/petroleum ether) to give 5.3 g of 6-bromo-7-methoxyquinazoline as yellow solid (97% yield). LCMS: m/z 239.0 [M+H] + ; t R = 1.57 min.

[0744] Step 5: Synthesis of 6-bromoquinazolin-7-ol. A mixture of 6-bromo-7-methoxyquinazoline (4 g, 16.7 mmol), decanethiol (8.76 g, 50 mmol) and K 2 CO 3 (6.94 g, 50 mmol) in 10 mL of NMP was stirred at 120 o C for 4 h. The mixture was purified by prep-HPLC (0-50% H 2 O (0.02% TFA/MeOH) afford 1.2 g of 6- bromoquinazolin-7-ol as yellow solid (32% yield). LCMS: m/z 225.0 [M+H] + ; t R = 1.28 min.

[0745] Step 6: Synthesis of 6-bromo-7-(methoxymethoxy)quinazoline. NaH (373 mg, 9.33 mmol, 60% in mineral oil) was added to a stirred solution of 6-bromoquinazolin-7-ol (1.05 g, 4.67 mmol) in 20 mL of THF at 0 o C. After stirring for 30 min, MOMBr (758 mg, 6.1 mmol) was added. The mixture was then stirred at room temperature, quenched with water (5 mL) and extracted with EtOAc (20 mL X 3). The combined organic solvents were concentrated and purified by silica gel chromatography (0-20% EtOAc/CH2Cl2) to give 440 mg of 6-bromo-7-(methoxymethoxy)quinazoline as yellow solid (35% yiled). LCMS: m/z 269.0 [M+H] + ; t R = 1.28 min.

[0746] Step 7: Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso quinoline (B33). A mixture of 6-bromo-7-(methoxymethoxy)quinazoline (120 mg, 0.446 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(1,3,2-dioxaborolane) (170 mg, 0.669 mmol), Pd(dppf)Cl 2 (66 mg, 0.089 mmol) and KOAc (88 mg, 0.892 mmol) in 1,4-dioxane (3 mL) was degassed and stirred at 105 o C for 8 h under N 2 . The reaction was cooled to room temperature and used in the next step without any work-up. LCMS: m/z 317.2 [M+H] + ; t R = 1.67 min.

Synthesis of 6-(methoxymethoxy)-1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)-1H-indazole (B34).

[0747] Step 1: Synthesis of 5-bromo-6-methoxy-1-methyl-1H-indazole. NaH (704 mg, 17.6 mmol, 60% in mineral oil) was added to a stirred solution of 5-bromo-6-methoxy-1H-indazole (2.0 g, 8.8 mmol) in 50 mL of DMF at room temperature. After stirring room temperature for 30 min, CH 3 I (1.9 g, 13.2 mmol) was added. The mixture was then stirred at room temperature for 2 h, quenched with ice water, extracted with EtOAc (100 mL X 3). The combined organic solvents were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-50% EtOAc/petroleum ether) to give 1.5 g of 5- bromo-6-methoxy-1-methyl-1H-indazole as white solid (71% yield). LCMS: m/z 241.0 [M+H] + ; t R = 1.58 min.

[0748] Step 2: Synthesis of 5-bromo-1-methyl-1H-indazol-6-ol. To a solution of 5-bromo-6-methoxy-1- methyl-1H-indazole (1.5 g, 6.2 mmol) in CH 2 Cl 2 (10 mL) at room temperature was added BBr 3 (40 mL, 1N in CH 2 Cl 2 ). The reaction mixture was stirred at 50 o C for 20 h, then quenched with ice water. Saturated aqueous sodium bicarbonate was added to adjust pH to 8~9. The mixture was extracted with CH 2 Cl 2 (100 mL X 3). The combined organic solvents were washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel column (0-10% MeOH/CH 2 Cl 2 ) to give 800 mg of 5-bromo-1-methyl-1H-indazol-6- ol as yellow solid (56% yield). LCMS: m/z 227.1 [M+H] + ; t R =1.78 min.

[0749] Step 3: Synthesis of 5-bromo-6-(methoxymethoxy)-1-methyl-1H-indazole. NaH (280 mg, 7.0 mmol, 60% in mineral oil) was added to a stirred solution of 5-bromo-1-methyl-1H-indazol-6-ol (800 mg, 3.5 mmol) in 10 mL of DMF at room temperature. After stirring room temperature for 30 min, MOMBr (657 mg, 5.3 mmol) was added. The mixture was then stirred at room temperature for 2 h, quenched with ice water, extracted with EtOAc (100 mL X 3). The combined organic solvents were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-25% EtOAc/petroleum ether) to give 950 mg of 5-bromo-6-(methoxymethoxy)-1-methyl-1H-indazole as yellow oil (99% yield). LCMS: m/z 271.2 [M+H] + ; tR = 1.62 min.

[0750] Step 4: Synthesis of 6-(methoxymethoxy)-1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)-1H-indazole (B34). A mixture of 5-bromo-6-(methoxymethoxy)-1-methyl-1H-indazole (200 mg, 0.74 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (284 mg 1.12 mmol), Pd 2 (dba) 3 (128 mg, 0.14 mmol), X-Phos (134 mg, 0.28 mmol), and KOAc (146 mg, 1.48 mmol) in 6 mL of dioxane was degassed and stirred at 100 o C for 2 h. The mixture was cooled to room temerature. The crude boronic ester was used directly to next step. LCMS: m/z 337.2 [M+H] + ; t R = 1.73 min.

Synthesis of 3-(methoxymethoxy)-N-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2-yl)benzamide (B35).

[0751] Step 1: Synthesis of 4-bromo-3-hydroxy-N-methylbenzamide. To the solution of 4-bromo-3- hydroxybenzoic acid (8.0 g, 37.0 mmol) in DMF (100 mL) were added HATU (15.5 g, 40.8 mmol), MeNH 2 HCl (7.4 g, 110.0 mmol) and DIPEA (11.9 g, 92.2 mmol). The mixture was stirred at room temperature for 3 h, quenched with H 2 O (300 mL), and extracted with EtOAc (200 mL X 2). The combined organic phases were dried over anhydrous Na 2 SO 4 , concentrated, and purified by silica gel chromatography (0-5%

EtOAc/petroleum ether) to give 8.0 g of 4-bromo-3-hydroxy-N-methylbenzamide as a white solid (95% yield). LCMS: m/z 232.1 [M+H] + ; t R = 1.39 min.

[0752] Step 2: Synthesis of 4-bromo-3-(methoxymethoxy)-N-methylbenzamide. NaH (4.72 g, 117.9 mmol, 60% in mineral oil) was added to a stirred solution of 4-bromo-3-hydroxy-N-methylbenzamide (9.0 g, 39.3 mmol) in 100 mL of THF at 25 o C. After stirring at 25 o C for 30 min, MOMBr (5.8 g, 46.7 mmol) was added. The mixture was then stirred at room temperature for 16 h, quenched with NH 4 Cl aqueous solution (100 mL), and extracted with EtOAc (100 mL X 2). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-25% EtOAc/petroleum ether) to give 7.0 g of 4-bromo-3-(methoxymethoxy)-N-methylbenzamide as colorless oil (84% yield), LCMS: LCMS: m/z 274.0 [M+H] + ; t R = 1.71 min.

[0753] Step 3: Synthesis of 3-(methoxymethoxy)-N-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)benzamide (B35). A mixture of 4-bromo-3-(methoxymethoxy)-N-methylbenzamide (300 mg, 0.41 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (418 mg, 0.82 mmol), Pd(dppf)Cl 2 (160 mg, 0.11 mmol) and KOAc (216 mg, 1.1 mmol) in 14 mL of dioxane was degassed and stirred at 100 o C for 2 h. After cooling to room temperature, the mixture was used in next step without further purification.

Synthesis of 5-(methoxymethoxy)-3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)benzo[d]oxazol-2(3H)-one (B36).

[0754] Step 1: Synthesis of 2-amino-4-methoxyphenol. A mixture of 4-methoxy-2-nitrophenol (10 g, 59.17 mmol) and Pd/C (3 g, 10% on activated carbon) in MeOH (25 mL) was stirred at room temperature for 5 h under H 2 atmosphere. Then the mixture was filtered and concentrated to give 8.5 g of 2-amino-4- methoxyphenol as yellow oil (97% yield), which was used directly to next step. LCMS: m/z 140.1 [M+H] + ; tR = 0.35 min.

[0755] Step 2: Synthesis of 5-methoxybenzo[d]oxazol-2(3H)-one. A mixture of 2-amino-4-methoxyphenol (18.5 g, 133 mmol) and CDI (28.05 g, 172.9 mmol) in THF (50 ml) was stirred at 70 o C for 2 h. After cooling to room temperature, the mixture was quenched with water (400 mL), and extracted with EtOAc (500 mL X 3). The combined organic solvents were washed with brine (400 ml X 3), dried over anhydrous Na 2 SO 4 and concentrated to give 31.5 g of 5-methoxybenzo[d]oxazol-2(3H)-one as colorless oil (98% yield), which was used directly to next step. LCMS: m/z 166.2 [M+H] + ; t R = 1.61 min.

[0756] Step 3: Synthesis of 6-bromo-5-methoxybenzo[d]oxazol-2(3H)-one. NBS (20.0 g, 112.1 mmol) was added to a mixture of 5-methoxybenzo[d]oxazol-2(3H)-one (18.5 g, 112.1 mmol) and in DMF (50 ml). The mixture was stirred at room temperature for 1 h, quenched with water (200 mL), and extracted with EtOAc (250 mL X 3). The combined organic solvents were washed with LiCl aqueous solution (200 mL X 3), dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-10%

EtOAc/petroleum ether) to give 22.8 g of 6-bromo-5-methoxybenzo[d]oxazol-2(3H)-one as brown solid (91% yield). LCMS: m/z 245.9 [M+H] + ; t R = 1.76 min.

[0757] Step 4: Synthesis of 6-bromo-5-methoxy-3-methylbenzo[d]oxazol-2(3H)-one. MeI (26.5 g, 0.18 mol) was added to a mixture of 6-bromo-5-methoxybenzo[d]oxazol-2(3H)-one (22.8 g, 0.09 mol) and K 2 CO 3 (25.8 g, 0.18 mol) in DMF (100 mL). The mixture was stirred at room temperature overnight, quenched with water (200 mL), and extracted with EtOAc (80 mL X 3). The combined organic solvents were washed with LiCl aqueous solution (200 ml X 3), dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-30% EtOAc/petroleum ether) to give 23.2 g of 6-bromo-5-methoxy-3- methylbenzo[d]oxazol-2(3H)-one as brown solid (96% yield). LCMS: m/z 259.0 [M+H] + ; t R = 1.72 min.

[0758] Step 5: Synthesis of 6-bromo-5-hydroxy-3-methylbenzo[d]oxazol-2(3H)-one. To a solution of 6- bromo-5-methoxy-3-methylbenzo[d]oxazol-2(3H)-one (2.3 g, 8.91 mmol) in CH 2 Cl 2 (20 mL) was added BBr 3 (20 mL, 1N in CH 2 Cl 2 ). The mixture was stirred at room temperature for 1 hour, quenched with water (100 mL) and the pH value was adjusted to 9~10 with K 2 CO 3 . The mixture was extracted with CH 2 Cl 2 /MeOH (10:1, v/v, 120 mL X 3). The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure to give 2 g of 6-bromo-5-hydroxy-3-methylbenzo[d]oxazol-2(3H)- one as yellow oil (71% yield), which was used directly to next step. LCMS: m/z 245.9 [M+H] + ; t R = 1.51 min.

[0759] Step 6: Synthesis of 6-bromo-5-(methoxymethoxy)-3-methylbenzo[d]oxazol-2(3H)-one. NaH (656 mg, 16.4 mmol, 60% in mineral oil) was added to a stirred solution of 6-bromo-5-hydroxy-3- methylbenzo[d]oxazol-2(3H)-one (2.0 g, 8.2 mmol) in 20 mL of DMF at 0 o C. After stirring at 0 o C for 30 min, MOMBr (2.0g, 16.4 mmol) was added. The mixture was then stirred at room temperature for 2 h, quenched with NH 4 Cl aqueous solution (50 mL), and extracted with EtOAc (80 mL X 3). The combined organic solvents were with LiCl aqueous solution (50 mL X 3), dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-10% EtOAc/petroleum ether) to give 2.0 g of 6-bromo-5- (methoxymethoxy)-3-methylbenzo[d]oxazol-2(3H)-one as yellow oil (72% yield). LCMS: 289.0 [M+H] + ; t R = 1.72 min.

[0760] Step 7. Synthesis of 5-(methoxymethoxy)-3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dio xaborolan-2- yl)benzo[d]oxazol-2(3H)-one (B36). A mixture of 6-bromo-5-(methoxymethoxy)-3-methylbenzo[d]oxazol- 2(3H)-one (2 g, 6.94 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (2.65 g, 10.41 mmol), Pd(dppf)Cl 2 (51 mg, 0.694 mmol) and KOAc (2.04 g, 20.82 mmol) in 30 mL of dioxane was degassed and stirred at 100 o C for 2 h. After cooling to room temperature, the mixture was concentrated and purified by silica gel column (0-50% EtOAc/petroleum ether) to give 2.0 g of 5-(methoxymethoxy)-3-methyl-6-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2(3H)-one as yellow oil (87% yield). LCMS: m/z 336.2 [M+H] + ; t R = 1.81 min.

Synthesis of (4-oxo-4,5-dihydrothieno[3,2-c]pyridin-2-yl)boronic acid (B39).

[0761] i-PrMgCl.LiCl (20 mL, 26.08 mmol) was dropped into a solution of 2-bromothieno[3,2-c]pyridin- 4(5H)-one (1 g, 4.34 mmol) in THF (15 mL) at 0 °C. The resulting reaction mixture was stirred for 1 h, then warmed up to room temperature. Then B(OMe) 3 (895 mg, 8.69 mmol) was added and the reaction mixture was stirred overnight. HCl/dioxane was added to quench the reaction, and the mixture was concentrated and purified by silica gel chromatography (4% MeOH/CH 2 Cl 2 ) to obtain (4-oxo-4,5-dihydrothieno[3,2-c]pyridin- 2-yl)boronic acid. LCMS: m/z 245.1[M+H] + ; t R = 1.14min.

Synthesis of (7-oxo-6,7-dihydrothieno[2,3-c]pyridin-2-yl)boronic acid (B40).

[0762] Step 1: Synthesis of (E)-3-(5-bromothiophen-3-yl)acrylic acid. To a solution of 5-bromothiophene- 3-carbaldehyde (5 g, 26.2 mmol) and piperidine (1.12 g, 13.1 mmol ) in pyridine (50 mL) was added malonic acid (8.17 g, 78.5 mmol ). The reaction mixture was refluxed for 3 h, cooled to room temperature and concentrated. The residue was diluted with H 2 O (100 mL), and the precipitated was formed. The suspension was acidified to pH 2 with 6 M HCl. The precipitate was collected by filtration, washed with H 2 O (3 X 30 mL) and dissolved in EtOAc (20 mL). The solution was dried over anhydrous Na 2 SO 4 and concentrated to give 5 g of (E)-3-(5-bromothiophen-3-yl)acrylic acid as a tan solid (82% yield). LCMS: m/z 232.9 [M+H] + ; t R = 1.65 min.

[0763] Step 2: Synthesis of (E)-3-(5-bromothiophen-3-yl)acryloyl azide. To a solution of (E)-3-(5- bromothiophen-3-yl)acrylic acid (1 g, 4.29 mmol ) and Et 3 N (1.3 g, 12.88 mmol ) in DCM (20 mL) was added diphenylphosphoryl azide (2.36 g, 8.58 mmol ) at 0 o C. The mixture was stirred at room temperature for 4 h, quenched with H 2 O (100 mL), and the aqueous phase was extracted with DCM (100 mL X 3). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0-5% ethyl acetate/petroleum ether) to give 900 mg of (E)-3-(5-bromothiophen-3-yl)acryloyl azide as white soild (82% yield). LCMS: m/z 232.0 [M-N 2 ] + ; t R = 1.95 min.

[0764] Step 3: Synthesis of 2-bromothieno[2,3-c]pyridin-7(6H)-one. To a solution of (E)-3-(5- bromothiophen-3-yl)acryloyl azide (900 mg, 3.49 mmol ) in Ph 2 O was added n-Bu 3 N (968 mg, 5.23 mmol ). The mixture was stirred for 2 h at 240 o C. The mixture was cooled to room temperature and hexane (20 mL) and EtOAc (15 mL) were added. The precipitate was collected by filtration to give 600 mg of 2- bromothieno[2,3-c]pyridin-7(6H)-one as white solid (75% yield). LCMS: m/z 231.9 [M+H] + ; t R = 1.44 min.

[0765] Step 4: Synthesis of (7-oxo-6,7-dihydrothieno[2,3-c]pyridin-2-yl)boronic acid (B40). To a solution of 2-bromothieno[2,3-c]pyridin-7(6H)-one (500 mg, 2.18 mmol ) in THF was added n-BuLi (1.3 mL, 2.5 N in hexanes) at -78 o C. After stirring for 30 mins, B(OMe) 3 (454 mg, 4.36 mmol) was added at -78 o C. The reaction was stirred at -78 o C for 3 h, quenched with MeOH and concentrated under reduced pressure to give 500 mg of (7-oxo-6,7-dihydrothieno[2,3-c]pyridin-2-yl)boronic acid as green oil (crude), which was used directly for the next step. LCMS: m/z 196.1 [M+H]+; t R = 0.35 min.

Synthesis of (6-methyl-7-oxo-6,7-dihydrothieno[2,3-c]pyridin-2-yl)boronic acid (B41).

[0766] Step 1: Synthesis of 2-bromo-6-methylthieno[2,3-c]pyridin-7(6H)-one. To a solution of 2- bromothieno[2,3-c]pyridin-7(6H)-one (1 g, 4.35 mmol ) in THF was added MeI (1.24 g, 8.70 mmol ) and Cs 2 CO 3 (2.84 g, 8.70 mmol ). The mixture was stirred at room temperature for 16 h. The reaction was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0-15% ethyl acetate in dichloromethane) to give 900 mg of 2-bromo-6-methylthieno[2,3-c]pyridin-7(6H)-one as a white soild (86% yield). LCMS: m/z 246.0 [M+H] + ; t R = 1.57 min.

[0767] Step 2: Synthesis of (6-methyl-7-oxo-6,7-dihydrothieno[2,3-c]pyridin-2-yl)boronic acid (B41). To a solution of 2-bromo-6-methylthieno[2,3-c]pyridin-7(6H)-one (400 mg, 1.65 mmol ) in THF was added n- BuLi (0.98 mL, 2.5 N in hexanes) at -78 o C. After stirring for 30 minutes, B(OMe) 3 (343 mg, 3.3 mmol) was added at -78 o C. The reaction was stirred at same temperature for 3 h, then was quenched with MeOH and concentrated in vacuo to give 400 mg of (6-methyl-7-oxo-6,7-dihydrothieno[2,3-c]pyridin-2-yl)boronic acid as yellow oil (crude), which used directly for the next step. LCMS: m/z 210.1 [M+H]+; t R = 0.35 min &0.48 min.

Synthesis of 1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)-1H-imidazole-4- carbonitrile (B42).

[0768] Step 1: Synthesis of 1-(3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile. A mixture of 5- fluoro-2-nitrophenol (4 g, 25.4 mmol), 1H-imidazole-4-carbonitrile (3.56 g, 38.22 mol) and Cs 2 CO 3 (12.46 g, 38.22 mmol) in 100 mL of DMF was stirred at 120 o C for 16 h. The solid was filtered off, the filtrate was concentrated under vacuum to give 5 g of 1-(3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile (68% yield), which was used directly to next step. LCMS: m/z 231.2 [M+H] + ; t R = 1.20 min.

[0769] Step 2: Synthesis of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile. A mixture of 1- (3-hydroxy-4-nitrophenyl)-1H-imidazole-4-carbonitrile (4 g, 17.4 mmol), Fe (2.92 g, 52.2 mol) and NH4Cl (2.8 g, 52.2 mmol) in 40 mL of EtOH and 20 mL of water was stirred at 80 oC for 2 h. The solid was filtered off. The filtrate was concentrated and purified by silica gel chromatography (80% EtOAc/petroleum ether) to give 3.2 g of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile. (92% yield). LCMS: m/z 201.2 [M+H] + ; t R = 1.28 min.

[0770] Step 3: Synthesis of 1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)-1H- imidazole-4-carbonitrile (B42). HCl (4.5 mL, 12 N aqueous solution) and water (4.5 mL) was added to the solution of 1-(4-amino-3-hydroxyphenyl)-1H-imidazole-4-carbonitrile (1 g, 5.0 mmol) in 18 mL of MeOH at 0 °C. NaNO 2 (0.38 g, 5.5 mmol) in water (3 mL) was then added. After stirring at 0 o C for 30 min,

4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolan e) (2.54 g, 10.0 mmol) was added. The mixture was stirred at room temperature overnight, quenched with H 2 O (50 mL) and extracted with CH 2 Cl 2 (30 mL X 3). The combined organic layers were concentrated and purified with silica gel chromatography (0-100%

EtOAc/petroleum ether) to give 170 mg of (1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenyl)-1H-imidazole-4-carbonitrile (15 % yield). LCMS: m/z 230.1 [M+H] + ; t R = 1.26 min.

Synthesis of 8-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4H -pyrido[1,2-a]pyrimidin-4- one (B43).

[0771] Step 1: Synthesis of 5-bromo-4-methoxypyridin-2-amine. Into a 250-mL 3-necked round-bottom flask, was placed a solution of 4-methoxypyridin-2-amine (10.0 g, 80.6 mmol, 1.0 equiv) in AcOH (100 mL). This was followed by the addition of Br 2 (19.31 g, 120.828 mmol, 1.5 equiv) dropwise with stirring at 0 °C. The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 100 mL of water/ice. The pH value of the solution was adjusted to 8 with K 2 CO 3 . The resulting solution was extracted with 3x50 mL of dichloromethane dried over anhydrous sodium sulfate and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:1). This resulted in 9.2 g (56.3%) of 5-bromo-4-methoxypyridin-2-amine as a white solid.

[0772] Step 2: Synthesis of 5-[[(5-bromo-4-methoxypyridin-2-yl)amino]methylidene]-2,2- dimethyl-1,3- dioxane-4,6-dione. Into a 50-mL 3-necked round-bottom flask, was placed a solution of 2,2-dimethyl-1,3- dioxane-4,6-dione (0.71 g, 4.925 mmol, 1 equiv) in EtOH (10 mL), and triethyl orthoformate (2.19 g, 14.775 mmol, 3 equiv), and the resulting solution was stirred for 4 h at 80 °C. Then 5-bromo-4-methoxypyridin-2- amine (1.00 g, 4.93 mmol, 1.0 equiv) was added. The resulting solution was allowed to react, with stirring, for an additional 2 h at 80 °C. The solids were collected by filtration. This resulted in 667 mg (37.92%) of 5-[[(5- bromo-4-methoxypyridin-2-yl)amino]methylidene]-2,2- dimethyl-1,3-dioxane-4,6-dione as a white solid.

[0773] Step 3: Synthesis of 7-bromo-8-methoxy-3H,9aH-pyrido[1,2-a]pyrimidin-4-one. Into a 250-mL round-bottom flask, was placed a solution of 5-[[(5-bromo-4-methoxypyridin-2-yl)amino]methylidene]- 2,2- dimethyl-1,3-dioxane-4,6-dione (12.00 g, 33.6 mmol, 1.0 equiv) in diphenyl ether (100 mL). The resulting solution was stirred for 4 h at 250 °C. The reaction mixture was gradually cooled to room temperature. The reaction mixture was applied onto a silica gel column with ethyl acetate/petroleum ether (4:1). This resulted in 3.6 g (41.7%) of 7-bromo-8-methoxy-3H,9aH-pyrido[1,2-a]pyrimidin-4-one as a brown solid.

Synthesis of 8-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4H -pyrido[1,2-a]pyrimidin-4- one (B43).

[0774] Into a 40-mL vial purged and maintained with an inert atmosphere of nitrogen, was placed 7-bromo- 8-methoxypyrido[1,2-a]pyrimidin-4-one (2.00g, 7.84 mmol, 1.0 equiv), 1,4-dioxane (20 mL),

bis(pinacolato)diboron (2.39 g, 9.41 mmol, 1.2 equiv), KOAc (1.54 g, 15.7 mmol, 2 equiv), Pd(dppf)Cl 2 (0.29 g, 0.392 mmol, 0.05 equiv). The resulting solution was stirred for 3 h at 100 °C. The resulting mixture was used in the next step directly without any further purification.

Synthesis of 7-methoxy-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)isoquinolin-1(2H)-one (B44).

[0775] Step 1: Synthesis of 4-bromo-3-methoxybenzoyl chloride. Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 4-bromo-3-methoxybenzoic acid (20.00 g, 86.56 mmol, 1.0 equiv), DCM (200 mL), and DMF (2.0 mL, 27.5 mmol, 0.30 equiv). This was followed by the addition of oxalyl chloride (13.20 g, 104 mmol, 1.2 equiv) dropwise with stirring at 0 °C. The resulting solution was stirred for 2 h at 25 °C. The resulting mixture was concentrated under vacuum. This resulted in 24.2 g (98.6%) of 4-bromo-3-methoxybenzoyl chloride as a white solid.

[0776] Step 2: Synthesis of (4-bromo-3-methoxyphenyl)formamido 2,2-dimethylpropanoate. Into a 500- mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 4- bromo-3-methoxybenzoyl chloride (27.4 g, 109.8 mmol, 1.0 equiv), ethyl acetate (300 mL), and O- pivaloylhydroxylammonium trifluoromethanesulfonate (29.20 g, 109.69 mmol, 1.0 equiv). This was followed by the addition of a solution of NaHCO 3 (18.50 g, 220.2 mmol, 2.0 equiv) in H 2 O (50 mL) dropwise with stirring at 0 °C. The resulting solution was stirred for 2 h at 0 °C. The aqueous phase was extracted with 200 mL of ethyl acetate and the organic layers combined. The combined organic phase was washed with 300 mL saturated NaHCO 3 aqueous solution, dried over Na 2 SO 4 and concentrated under reduced pressure. The residue was applied onto a silica gel column with ethyl acetate/hexane (1:1) as eluent. This resulted in 27.4 g (75.6%) of (4-bromo-3-methoxyphenyl)formamido 2,2-dimethylpropanoate as yellow oil. LCMS: m/z [M+H] + =330.

[0777] Step 3: Synthesis of 6-bromo-7-methoxy-2H-isoquinolin-1-one. Into a 500-mL 3-necked round- bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (4-bromo-3- methoxyphenyl)formamido 2,2-dimethylpropanoate (25.0 g, 75.72 mmol, 1.0 equiv), vinyl acetate (9.80 g, 113.8 mmol, 1.5 equiv), MeOH (300 mL), CsOAc (4.51 g, 23.5 mmol, 0.3 equiv),

bis[(pentamethylcyclopentadienyl)dichloro-rhodium] (468 mg, 0.757 mmol, 0.01 equiv). The resulting solution was stirred overnight at 40 °C. The resulting mixture was concentrated under vacuum to leave the residue. The residue was recrystallized from 100 mL of MTBE to afford 15 g (78%) of 6-bromo-7-methoxy- 2H-isoquinolin-1-one as a white solid. LCMS: m/z:[M+H] + =254.

[0778] Step 4: Synthesis of 6-bromo-7-methoxy-2-methylisoquinolin-1-one. Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 6-bromo-7- methoxy-2H-isoquinolin-1-one (5.00 g, 19.68 mmol, 1.0 equiv), and 100 mL THF. This was followed by the addition of NaH (1.20 g, 30.003 mmol, 1.52 equiv, 60wt%), in portions at 0 °C. To this was added CH 3 I (3.40 g, 21.8 mmol, 1.11 equiv) dropwise with stirring at 0 °C. The resulting solution was stirred for 4 h at 25 °C. The reaction was then quenched by the addition of 100 mL of water/ice. The resulting solution was extracted with 3x100 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate. The solids were filtered out, and the filtrated was concentrated under vacuum. This resulted in 3.2 g of 6- bromo-7-methoxy-2-methylisoquinolin-1-one as a yellow solid. LCMS: m/z [M] + =268; 1 H NMR (300 MHz, DMSO-d 6 ) d 8.02 (s, 1H), 7.68 (s, 1H), 7.47– 7.05 (m, 2H), 6.56 (d, J = 7.3 Hz, 1H), 3.95 (s, 3H), 2.51 (dt, J = 3.8, 1.9 Hz, 3H).

[0779] Step 5: Synthesis of 7-methoxy-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl) isoquinolin-1(2H)-one (B44). Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 6-bromo-7-methoxy-2-methylisoquinolin-1(2H)-one (1.33 g, 4.97 mmol, 1.0 equiv), potassium acetate (0.97 g, 9.93 mmol, 2.0 equiv), bis(pinacolato)diboron (2.52 g, 9.93 mmol, 2.00 equiv), Pd(dppf)Cl 2 (0.36 g, 0.497 mmol, 0.10 equiv), and 1,4-dioxane (25 mL). The resulting solution was stirred for 2 hat 100 °C in an oil bath. The reaction mixture was cooled down to ambient temperature and quenched by the addition of 30 mL of water. The resulting solution was extracted with 2x30 mL of dichloromethane, and the organic phase was combined, dried over Na 2 SO 4 and concentrated. This resulted in 3.0 g (crude, calculated as 3.476 mmol, 70%) of 7-methoxy-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl) isoquinolin-1(2H)-one as a dark green semi-solid, which was used directly in the next step. Synthesis of 7-methoxy-3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan -2-yl)quinazolin-4(3H)-one (B45).

[0780] Step 1: Synthesis of 2-amino-5-bromo-4-methoxybenzoic acid. Into a 250-mL round-bottom flask, was placed 2-amino-4-methoxybenzoic acid (2.00 g, 11.96 mmol, 1.0 equiv), dimethylformamide (60 mL). This was followed by the addition of N-bromosuccinimide (2.30 g, 12.9 mmol, 1.1 equiv) in several batches at 0 °C. The resulting solution was stirred for 1 h at room temperature. The reaction mixture was cooled to 0 °C with a water/ice bath. The reaction was then quenched by the addition of 13 mL of saturated aqueous

NaHCO 3 . The pH value of the solution was adjusted to 3 with HCl (6 mol/L). The resulting solution was extracted with 3x25 mL of ethyl acetate. The organics were combined and washed with 3x25mL of water and 25x1mL of brine. The combined organic layers were dried over anhydrous sodium sulfate and concentrated. This resulted in 2.5 g (84.9%) of 2-amino-5-bromo-4-methoxybenzoic acid as a light brown solid.

[0781] Step 2: Synthesis of 6-bromo-7-hydroxy-3-methylquinazolin-4-one. Into a 40-mL sealed tube, was placed 2-amino-5-bromo-4-methoxybenzoic acid (5.00 g, 20.32 mmol, 1.0 equiv), and methylformamide (8 mL). The resulting solution was stirred for 3 h at 180 °C. The reaction mixture was cooled with a water/ice bath and then 25 mL of water was added. The solids were collected by filtration. This resulted in 1.0 g (19.3%) of 6-bromo-7-hydroxy-3-methylquinazolin-4-one as a light brown solid. LCMS: m/z:[M+H] + =269.0.

[0782] Step 3: Synthesis of 7-methoxy-3-methyl-4-oxoquinazolin-6-ylboronic acid (B45). Into a 40-mL sealed tube, was placed 6-bromo-7-methoxy-3-methylquinazolin-4-one (1.00 g, 3.72 mmol, 1.0 equiv), (BPin) 2 (1.43 g, 5.63 mmol, 1.5 equiv), KOAc (729.4 mg, 7.43 mmol, 2.0 equiv), Pd(dppf)Cl 2 (163.2 mg, 0.223 mmol, 0.06 equiv), and 1,4-dioxane (20 mL). The resulting solution was stirred for 3 h at 100 °C. The reaction mixture was cooled with a water/ice bath. The resulting solution was diluted with of water and then extracted with 3x20ml of dichloromethane. The organics were combined, dried over anhydrous sodium sulfate, and concentrated to dryness under reduced pressure. This resulted in 500 mg (57.5%, crude) of 7- methoxy-3-methyl-4-oxoquinazolin-6-ylboronic acid as a light brown solid. LCMS: m/z:[M+H] + 235.1.

[0783] Additional boronic acid or ester compounds below were made following the general synthetic methods applied to synthesize B1-B45.

Example B2: Synthesis of 2-amino-pyridazine intermediates

[0784] Example B2a: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-2-fluoro-3-(methylamino)-8- azabicyclo[3.2.1]octane-8-carboxylate (INT-1).

[0785] Step 1: Synthesis of (±) tert-butyl (1S,2S,5R)-2-fluoro-3-oxo-8-azabicyclo[3.2.1]octane-8- carboxylate. TMSCl (19.2 g, 17.78 mmol) and triethylamine (17.78 g, 17.78 mmol) was added to a stirred solution of tert-butyl 3-oxo-8-azabicyclo[3.2.1]octane-8-carboxylate (20 g, 8.89 mmol) in 270 mL of DMF. The mixture was stirred at 100 o C for 16 h. Water (300 mL) was added to the reaction, then the mixture was extracted with ethyl acetate (100 mL X 3). The combined organic phases were dried and concentrated, then purified by silica gel chromatography (10% EtOAc/petroleum ether) to give the title compound (21 g, 79% yield). LCMS: m/z 298.2 [M+H] + ; t R = 2.33 min.

[0786] Step 2: Synthesis of (±) tert-butyl (1S,2S,5S)-2-fluoro-8-aza-bicyclo[3.2.1]octan-3-one.

Selectfluor TM (14.16 g, 40 mmol) was added to a solution of (±) tert-butyl 3-((trimethylsilyl)oxy)-8- azabicyclo[3.2.1]oct-2-ene-8-carboxylate (6 g, 20 mmol) in 120 mL of dry CH 3 CN at 0 o C. After addition, the mixture was stirred at rt for 2 h. The mixture was concentrated, then purified by silica gel chromatography (50% EtOAc/petroleum ether) to give the title compound. (3.84 g, 78% yield). LCMS: m/z 188.2 [M-55] + ; t R = 1.86 min.

[0787] Step 3: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-(benzylamino)-2-fluoro-8- azabicyclo[3.2.1]octane-8-carboxylate. A mixture of (±) tert-butyl (1S,2S,5S)-2-fluoro-8-aza- bicyclo[3.2.1]octan-3-one (10 g, 41 mmmol) and benzylamine (7.6 g, 82 mmol) in 160 mL of toluene was refluxed for 4 h. The mixture was concentrated.160 mL of methanol was added. NaBH 4 (3.1 g, 82 mmol) was added in small portions at 0 o C. The mixture was stirred for 2 h at rt and concentrated. H 2 O (80 mL) was added and the resulting mixture was extracted with EtOAc (60 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified on silica gel chromatography (10-50%

EtOAc/petroleum ether) to yield the title compound as a white solid (7.3 g, 53% yield). LCMS: m/z 335.1 [M+H] + ; t R = 1.80 min.

[0788] Step 4: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-(benzyl(methyl)amino)-2-fluoro-8- azabicyclo[3.2.1]octane-8-carboxylate. To a solution of (±) tert-butyl (1S,2R,3R,5R)-3-(benzylamino)-2- fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate (800 mg, 2.395 mmol, 1 equiv) in MeOH (100 ml) was added formaldehyde (143 mg, 4.8 mmol). The reaction mixture was stirred at rt for 0.5 h, then NaBH 3 CN (301 mg, 4.8) was added. The mixture was stirred for additional 3 h, monitored by LCMS. Solvent was evaporated and the resulting crude was purified by column chromatography on silica gel chromarography (50%

EtOAc/petroleum ether) to yield the title compound (800 mg, 2.30 mmol, 96% yield) as white powder.

LCMS: m/z 349.1 [M+H] + ; t R = 1.42 min.

[0789] Step 5: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-2-fluoro-3-(methylamino)-8- azabicyclo[3.2.1]octane-8-carboxylate. To a solution of (±) tert-butyl (1S,2R,3R,5R)-3- (benzyl(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-c arboxylate (800 mg, 1 equiv) in EtOH (100 mL) was added Pd/C (80 mg, 10%). The reaction mixture was stirred under H 2 atmosphere for 2 h and monitored by LCMS. The mixture was filtered and the filtrate was concentrated under reduced pressure to give the title compound (500 mg, 84% yield) as colorless oil, which was directly used in next step. LCMS: m/z 203.1 [M-55] + ; t R = 1.19 min.

[0790] Step 6: Synthesis of tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amino)-2- fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate (INT-1). DIPEA (3.97 g, 30.7 mmol) was added to a solution of 3,6-dichloropyridazine (2.2 g, 14.7 mmol) and (±) (1S,2R,3R,5R)-tert-butyl 3-amino-2-fluoro-8- azabicyclo[3.2.1]octane-8-carboxylate (3.0 g, 12.3 mmol) in DMSO (30 mL). The mixture was stirred at 120 o C for 16 h. After cooling to room temperature, the mixture was quenched with H 2 O (50 mL) and extracted with EtOAc (30 mL X 3). The combined organic layers were concentrated and purified with silica gel chromatography (10-25% EtOAc/petroleum ether) to yield the title compound (3.05 g, 8.65 mmol, 58 % yield). LCMS: m/z 357.1 [M+H] + ; t R = 1.79 min.

Example B2b: Synthesis of (±) tert-butyl exo-3-amino-6,6-difluoro-8-azabicyclo[3.2.1]octane-8- carboxylate (INT-2)

[0791] Step 1: Synthesis of (±) 6-hydroxy-8-benzyl 8-azabicyclo[3.2.1]octan-3-one.2,5-Dimethoxy-2,5- dihydrofuran (97.5 g, 750 mmol) was dissolved in water (650 mL) and treated with aqueous hydrochloric acid (3.75 ml, 2M) under an atmosphere of nitrogen. The mixture was heated to 99 °C with stirring and aqueous methanol (about 100 mL) was distilled from the reaction mixture. The reaction was cooled to ambient temperature, acetone dicarboxylic acid (146 g, 1.33 mol) wase added in one portion followed by a solution of sodium hydrogen phosphate (53.25 g, 375 mmol) and sodium hydroxide (15.0 g, 375 mmol) in water (500 mL).1,4-Dioxane (100 mL) was added and a solution of benzylamine hydrochloride (71.75 g, 502 mmol) in water (330 mL) was added dropwise over 10 minutes. The mixture was rapidly stirred for a further 4 h, acidified with aqueous hydrochloric acid (2M). Dichloromethane (500 mL) was added and the reaction mixture stirred for 10 minutes. The aqueous phase was separated and filtered through a pad of Celite® brand filter agent. The filtrate was extracted with dichloromethane (500 mL x 3). The aqueous phase was collected, basified with potassium carbonate and extracted with ethyl acetate (1000 mL x 3). The organic fractions were combined, dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to give the title compound (36 g, 21% yield) as a brown oil, containing a mixture of exo- and endo-6-hydroxy-8-benzyl 8- azabicyclo[3.2.1]octan-3-one, which was used directly to next step. LCMS: m/z 232.1 [M+H] + ; t R = 1.52 min.

[0792] Step 2: Synthesis of (±) 8-benzyl-6-((tert-butyldimethylsilyl)oxy)-8-azabicyclo[3.2.1 ]octan-3-one. TBSCl (18.7 g, 124.6 mmol) was added to a stirred solution of (±) 6-hydroxy-8-benzyl 8- azabicyclo[3.2.1]octan-3-one (24 g, 104 mmol) and imidazole (10.6 g, 156 mmol) in CH 2 Cl 2 (500 mL) at rt. The mixture was stirred at rt for 18 h, concentrated and purified by silica gel chromatography (14%

EtOAc/petroleum ether) to give the title compound as a brown oil (28 g, 78% yield). LCMS: m/z 346.2

[M+H] + ; t R = 2.46 min.

[0793] Step 3: Synthesis of (±) (1S,3R,5R)-8-benzyl-6-((tert-butyldimethylsilyl)oxy)-8- azabicyclo[3.2.1]octan-3-ol. NaBH 4 (8.8 g, 232 mmol) was added to a stirred solution of (±) 8-benzyl-6- ((tert-butyldimethylsilyl)oxy)-8-azabicyclo[3.2.1]octan-3-on e (40 g, 116 mmol) in MeOH (600 mL) at rt. The mixture was stirred at rt for 2 h, and concentrated to remove most of the solvent.500 mL of water was added. The resulting mxiture was extracted with EtOAc (500 mL x 2). The combined organic solvents were concentrated and purified by silica gel chromatography (0-24% EtOAc/petroleum ether) to give the title compound (38 g, 94% yield) as a brown oil LCMS: m/z 348.1 [M+H] + ; t R = 1.57 min.

[0794] Step 4: Synthesis of (±) (1S,3R,5R)-8-benzyl-6-((tert-butyldimethylsilyl)oxy)-8- azabicyclo[3.2.1]octan-3-yl acetate. Ac 2 O (7.9 g, 78 mmol) was added to a stirred solution of (±) 8-benzyl-6- ((tert-butyldimethylsilyl)oxy)-8-azabicyclo[3.2.1]octan-3-ol (18 g, 52 mmol), Et 3 N (10.5 g, 104 mmol) and DMAP (634 mg, 122 mmol) in 200 mL of THF at 0 o C. After addition, the mixture was stirred at rt for 18 h, concentrated and purified by silica gel chromatography (0-14% EtOAc/petroleum ether) to give the title compound (16 g, 78% yield). LCMS: m/z 390.1 [M+H] + ; t R = 2.57 min.

[0795] Step 5: Synthesis of (±) (1R,3R,5R)-8-benzyl-6-hydroxy-8-azabicyclo[3.2.1]octan-3-yl acetate. TBAF (43.3 mL, 43.3 mmol, 1 M solution in THF) was added to a stirred solution of (±) 8-benzyl-6-((tert- butyldimethylsilyl)oxy)-8-azabicyclo[3.2.1]octan-3-yl acetate (13 g, 33.4 mmol) in 100 mL of THF. The mixture was stirred at rt for 4 h and 30 mL of H 2 O was added. The mixture was extracted with EtOAc (50 mL x 3). The combined organic solvents were concentrated and purified by silica gel chromatography (0-40% EtOAc/petroleum ether) to give the title compound (8.7 g, 91% yield). LCMS: m/z 276.2 [M+H] + ; t R = 1.76 min.

[0796] Step 6: Synthesis of (±) (1S,3R,5R)-8-benzyl-6-oxo-8-azabicyclo[3.2.1]octan-3-yl acetate. Dess- Martin periodinane (13.9 g, 32.7 mmol) was added to a stirred solution of (±) 8-benzyl-6-hydroxy-8- azabicyclo[3.2.1]octan-3-yl acetate (6 g, 21.8 mmol) in 60 mL of CH 2 Cl 2 at rt. The mixture was stirred at rt for 16 h, filtered, concentrated and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give the title compound (4.45 g, 60% yield). LCMS: m/z 274.1 [M+H] + ; t R = 1.85 min.

[0797] Step 7: Synthesis of (±) (1S,3R,5R)-8-benzyl-6,6-difluoro-8-azabicyclo[3.2.1]octan-3- yl acetate. DAST (52.5 g, 326 mmol) was added to a stirred solution of (±) 8-benzyl-6-oxo-8-azabicyclo[3.2.1]octan-3-yl acetate (8.9 g, 32.6 mmol) in 90 mL of CH 2 Cl 2 under nitrogen atmosphere. The mixture was stirred at 60 o C (oil bath) for 12 h. After cooling to rt, the mixture was quenched with H 2 O, extracted with CH 2 Cl 2 (30 mL x 3), concentrated and purified by silica gel chromatography (0-28% EtOAc/petroleum ether) to give the title compound (5.3 g, 55% yield). LCMS: m/z 296.2 [M+H] + ; t R = 2.20 min.

[0798] Step 8: Synthesis of (±) tert-butyl (1S,3R,5R)-3-acetoxy-6,6-difluoro-8-azabicyclo[3.2.1]octane- 8- carboxylate. A mixture of (±) 8-benzyl-6,6-difluoro-8-azabicyclo[3.2.1]octan-3-yl acetate (1.9 g, 6.44 mmol), Pd/C (500 mg, 10% on activated carbon) and (Boc) 2 O (1.69 g, 7.73 mmol) in 50 mL of MeOH was stirred under H 2 at balloon pressure for 16 h. The mixture was filtered, concentrated and purified by silica gel chromatography (0-37% EtOAc/petroleum ether) to give the title compound (1.5 g, 76% yield). LCMS: m/z 328.1 [M+23] + ; t R = 1.94 min.

[0799] Step 9: Synthesis of (±) tert-butyl (1S,3R,5R)-6,6-difluoro-3-hydroxy-8-azabicyclo[3.2.1]octane- 8-carboxylate. K 2 CO 3 (497 mg, 3.6 mmol) was added to a stirred solution of (±) tert-butyl 3-acetoxy-6,6- difluoro-8-azabicyclo[3.2.1]octane-8-carboxylate (550 mg, 1.8 mmo) in 5 mL of MeOH. The mixture was stirred at rt for 3 h, filtered, concentrated and purified by silica gel chromatography (0-30% EtOAc/petroleum ether) to give the title compound (280 mg, 68% yield). LCMS: m/z 208.1 [M-55] + ; t R = 1.62 min.

[0800] Step 10: Synthesis of (±) tert-butyl (1S,5R)-6,6-difluoro-3-oxo-8-azabicyclo[3.2.1]octane-8- carboxylate. Dess-Martin periodinane (7.2 g, 16.9 mmol) was added to a stirred solution of (±) tert-butyl 6,6-difluoro-3-hydroxy-8-azabicyclo[3.2.1]octane-8-carboxyla te (3.1 g, 11.3 mmol) in 50 mL of CH 2 Cl 2 at rt. The mixture was stirred at rt for 16 h, filtered, concentrated and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give the title compound (1.97 g, 67% yield). LCMS: m/z 279.0 [M+18] + ; t R = 1.28 min.

[0801] Step 11: Synthesis of (±) tert-butyl (1S,5R)-6,6-difluoro-3-(hydroxyimino)-8- azabicyclo[3.2.1]octane-8-carboxylate. A mixture of (±) tert-butyl 6,6-difluoro-3-oxo-8- azabicyclo[3.2.1]octane-8-carboxylate (1.5 g, 5.7 mmol), hydroxylamine hydrochloride (595 mg, 8.6 mmol) and AcONa (708 mg, 8.6 mmol) in 50 mL of EtOH was stirred at 80 o C for 2 h. The reaction was cooled to rt, and concentrated in vacuo.50 mL of EtOAc and 20 mL of H 2 O was added, the organic phase was separated, washed with brine, dried over anhydrous Na 2 SO 4 and concentrated to give the title compound (1.2 g, 93% yield) as yellow solid, which was used directly to next step. LCMS: m/z 221.1 [M-55] + ; t R = 1.71 min.

[0802] Step 12: Synthesis of (±) tert-butyl (1S,3R,5R)-3-amino-6,6-difluoro-8-azabicyclo[3.2.1]octane-8- carboxylate. Na (333 mg, 14.5 mmol) was added in portions over 10 min to a mixture of (±) tert-butyl 6,6- difluoro-3-(hydroxyimino)-8-azabicyclo[3.2.1]octane-8-carbox ylate (400 mg, 1.45 mmol) in 20 mL of n- PrOH at 105 o C. After addition, the mixture was stirred at 105 o C for additional 30 min, cooled to rt and quenched with H 2 O (50 mL). The mixture was extracted with EtOAc (50 mL x 2). The combined organic layers were dried and concentrated under reduced pressure to afford the title compound (324 mg, 85% yield) as a yellow oil. LCMS: m/z 207.2 [M-55] + ; t R = 1.30 min.

[0803] Step 13: Synthesis of (±) tert-butyl (1S,3R,5R)-3-((6-chloropyridazin-3-yl)amino)-6,6-difluoro-8- azabicyclo[3.2.1]octane-8-carboxylate. A mixture of tert-butyl exo-3-amino-6,6-difluoro-8- azabicyclo[3.2.1]octane-8-carboxylate (5.0 g, 19.0 mmol), 3,6-dichloropyridazine (5.65 g, 38.0 mmol) and DIPEA (7.4 g, 57.0 mmol) in 100 mL of DMSO was stirred at 120 o C for 16 h. The mixture was cooled to rt, quenched with H 2 O and extracted with ethyl acetate (2x100 mL). The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by silica gel column (0-60% EtOAc/petroleum ether) to afford the title compound (8.8 g, 61.6% yield) as a yellow solid LCMS: m/z 319.3 [M-56+H] + ; t R = 1.87 min.

Step 14: Synthesis of (±) tert-butyl(1S,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amin o)-6,6-difluoro-8- azabicyclo[3.2.1]octane-8-carboxylate (INT-2).

[0804] NaH (1.6 g, 40.10 mmol, 60% in mineral oil) was added to a stirred solution of (±) tert- butyl(1S,3R,5R)-3-((6-chloropyridazin-3-yl)amino)-6,6-difluo ro-8-azabicyclo[3.2.1]octane-8-carboxylate (5.0 g, 13.37 mmol) in 120 mL of DMF at 0 o C. After stirring for 15 min at 0 o C, CH 3 I (3.8 g, 26.7 mmol) was added. The mixture was then stirred at room temperature for 1 h, quenched with H 2 O and extracted with EtOAc (2x100 mL). The combined organic phases were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated under reduced pressure and purified by silica gel column (0-60% EtOAc/petroleum ether) to afford the title compound (4.0 g, 77% yield) as a yellow solid. LCMS: m/z 389.2 [M+H] + ; t R = 1.95 min. Example B2c: Synthesis of (±) tert-butyl exo-3-amino-6,6-difluoro-8-azabicyclo[3.2.1]octane-8- carboxylate (INT-3)

[0805] Step 1: Synthesis of tert-butyl 3-((trimethylsilyl)oxy)-9-azabicyclo[3.3.1]non-2-ene-9- carboxylate. Chlorotrimethylsilane (34g, 313.8 mmol) was added to a stirred solution of tert-butyl 3-oxo-9- azabicyclo[3.3.1]nonane-9-carboxylate (50 g, 209.2 mmol) and Et 3 N (42 g, 418.4 mmol) in 500 mL of DMF at 0 o C under nitrogen protection. After the addition, the mixture was then stirred at 100 o C for 18 h. The mixture was cooled to room temperature, quenched with H 2 O (500 mL), and extracted with EtOAc (200 mL X 3). The combined organic solvents were washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , concentrated, and purified by silica gel column (0-5% EtOAc/petroleum ether) to give the title compound (46 g, 71% yield) as a colorless oil. LCMS: m/z 212.3 [M-100] + ; t R = 2.32 min.

[0806] Step 2: Synthesis of (±) tert-butyl (1S,2S,5R)-2-fluoro-3-oxo-9-azabicyclo[3.3.1]nonane-9- carboxylate. Selectfuoro (62.8 g, 177.5 mmol) was added in three portions to a stirred solution of tert-butyl 3- ((trimethylsilyl)oxy)-9-azabicyclo[3.3.1]non-2-ene-9-carboxy late (46 g, 147.9 mmol) in CH 3 CN (460 mL) at 0 o C under nitrogen protection. The mixture was then stirred at room temperature for 3 h, quenched with H 2 O (400 mL), and extracted with EtOAc (300 mL X 3). The combined organic solvents were concentrated and the residue purified by silica gel silica gel chromatography (0-10% EtOAc/petroleum ether) to give the title compound (19 g, 50% yield) as a white solid LCMS: m/z 202.1 [M-55] + ; t R = 1.86 min.

[0807] Step 3: Synthesis of (±) tert-butyl (1S,2S,3S,5R)-2-fluoro-3-hydroxy-9-azabicyclo[3.3.1]nonane-9 - carboxylate. NaBH 4 (6.65 g, 175.1 mmol) was added to a mixture of (±) tert-butyl (1S,2S,5R)-2-fluoro-3- oxo-9-azabicyclo[3.3.1]nonane-9-carboxylate (30 g, 116.6 mmol) and in methanol (300 mL). The mixture was stirred at room temperature for 1 h, and concentrated in vacuum to remove methanol. Water (300 mL) was added and the mixture was extracted with EtOAc (100 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel chromatography (0-10% EtOAc/petroleum ether) to give (±) tert-butyl (1S,2S,3S,5R)-2-fluoro-3-hydroxy-9-azabicyclo[3.3.1]nonane-9 -carboxylate (20 g, 66% yield) as a white solid; LCMS: m/z 204.3 [M-55] + ; t R = 1.72 min. Also isolated was a minor product, (±) tert-butyl (1S,2S,3R,5R)-2-fluoro-3-hydroxy-9-azabicyclo[3.3.1]nonane-9 -carboxylate (7.5 g, 25% yield) as a white solid. LCMS: m/z 204.1 [M-55] + ; t R = 1.65 min. (67% yield).

[0808] Step 4: Synthesis of (±) tert-butyl (1S,2S,3S,5R)-2-fluoro-3-((methylsulfonyl)oxy)-9- azabicyclo[3.3.1]nonane-9-carboxylate. Methanesulfonyl chloride (17.68 g, 154 mmol) was added to a solution of (±) tert-butyl (1S,2S,3S,5R)-2-fluoro-3-hydroxy-9-azabicyclo[3.3.1]nonane-9 -carboxylate (20 g, 77 mmol) and triethylamine (15.6 g, 154 mmol) in N,N-dimethylformamide (100 mL) under an atmosphere of nitrogen at 0 0 C. The mixture was allowed to warm and stirred at room temperature for 6 h, quenched with water (200 mL), and extracted with ethyl acetate (300 mL X 3). The combined organic solvents were washed with LiCl solution (200 mL X 3), dried over anhydrous Na 2 SO 4 , filtered and concentrated to give the title compound (34.7g, 90% yield) as a white solid, which was used directly to next step. LCMS: m/z 282.0 [M- 55] + ; t R = 1.98 min.

[0809] Step 5: Synthesis of (±) tert-butyl (1S,2S,3R,5R)-3-azido-2-fluoro-9-azabicyclo[3.3.1]nonane-9- carboxylate. A mixture of (±) tert-butyl (1S,2S,3R,5R)-2-fluoro-3-((methylsulfonyl)oxy)-9- azabicyclo[3.3.1]nonane-9-carboxylate (34.7 g, 103 mmol) and NaN 3 (26.8 g, 412 mmol) in DMSO (100 mL) was stirred at 120 0 C for 48 h. After cooled to room temperature, the mixture was quenched with water (300 mL), and extracted with ethyl acetate (500 mL X 3). The combined organic solvents were washed with brine (300 mL X 3), dried over anhydrous Na2SO4, filtered and concentrated to give the title compound (23.8 g, 98% yield), as a yellow oil, which was used directly in the next step. LCMS: m/z 229.1 [M-55] + ; t R = 2.06 min.

[0810] Step 6: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-amino-2-fluoro-9-azabicyclo[3.3.1]nonane-9- carboxylate. A mixture of (±) tert-butyl (1S,2S,3R,5R)-3-azido-2-fluoro-9-azabicyclo[3.3.1]nonane-9- carboxylate (23.8 g, 84 mmol) and Pd/C (3 g, 10% on activated carbon) in EtOAc (85 mL) was stirred at room temperature for 5 h under H 2 atmosphere. Then the mixture was filtered and concentrated to give the title compound (21 g, 97% yield), as a yellow oil which was used directly in the next step. LCMS: m/z 203.2 [M- 55] + ; t R = 1.22 min.

[0811] Step 7: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)amino)-2-fluoro-9- azabicyclo[3.3.1]nonane-9-carboxylate. A mixture of (±) tert-butyl (1S,2R,3R,5R)-3-amino-2-fluoro-9- azabicyclo[3.3.1]nonane-9-carboxylate as (23.4 g, 90 mmol), 3,6-dichloropyridazine (20.3 g, 140 mmol) and DIPEA (46.6 g, 360 mmol) in DMSO (100 mL) was stirred at 120 o C overnight. After cooling to room temperature, the mixture was quenched with 500 mL of water, extracted with EtOAc (800 mL X 3). The combined organic solvents were washed with water (500 mL), concentrated, and purified by silica gel column (20% EtOAc/petroleum ether) to give the title compound tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3- yl)amino)-2-fluoro-9-azabicyclo[3.3.1]nonane-9-carboxylate (6.0 g, 18% yield) as white solid. LCMS: m/z 371.0 [M+H] + ; t R = 1.98 min. [0812] Step 8: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amino)-2- fluoro-9-azabicyclo[3.3.1]nonane-9-carboxylate (INT-3). NaH (1.3 g, 32 mmol, 60% in mineral oil) was added to a stirred solution of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)amino)-2-fluoro-9- azabicyclo[3.3.1]nonane-9-carboxylate (6 g, 16 mmol) in DMF (25.0 mL) at 0 o C under nitrogen protection and stirred at room temperature for 20 min. MeI (4.6 g, 32 mmol) was added and the mixture was stirred at room temperature for 1 h. The reaction was quenched with H 2 O (200 mL) and extracted with EtOAc (300 mL X 3). The combined organic solvents were washed with LiCl aqueous solution (200 ml X 3), concentrated and purified by silica gel silica gel chromatography (0-10% EtOAc/petroleum ether) to give the title compound (5.42 g, 87% yield) as a yellow solid. LCMS: m/z 385.1 [M+H] + ; t R = 2.12 min.

Examble B2d: Synthesis of (±) tert-butyl (1S,5S,6R,7R)-6-fluoro-7-(methylamino)-3-oxa-9- azabicyclo[3.3.1]nonane-9-carboxylate (INT-4).

[0813] Step 1: Synthesis of (±) tert-butyl (1S,5S,6S)-6-fluoro-7-oxo-3-oxa-9-azabicyclo[3.3.1]nonane-9- carboxylate. LiHMDS ( 155.6 mL) was added to a stirred solution of tert-butyl-7-oxo-3-oxa-9- azabicyclo[3.3.1]nonane-9-carboxylate (25 g, 103.7 mmol) in THF (500 mL) at -78 °C. After stirring at -78°C for 20 min, NFSI (39.2 g, 124.4 mmol) in THF (100mL) was added dropwise. The reaction mixture was stirred at -78 °C for 2 h, quenched with saturated NH 4 Cl aqueous solution (200 mL), extracted with EtOAc (300 mL X 3). The combined organic solvents were washed with brine (200 mL), dried over anhydrous MgSO 4 , concentrated and purified by silica gel column (5-20% EtOAc/petroleum ether) to give the title compound (12 g, yield:45%) as a white solid. LCMS: m/z 260.2 [M+1] + ; t R =1.633 min.

[0814] Step 2: Synthesis of (±) tert-butyl (1S,5S,6R,7R)-6-fluoro-7-(methylamino)-3-oxa-9- azabicyclo[3.3.1]nonane- 9-carboxylate. AcOH (5 drops) was added to a solution of (±) tert-butyl

(1S,5S,6S)-6-fluoro-7-oxo-3-oxa-9-azabicyclo[3.3.1]nonane -9-carboxylate (5 g, 19.32 mmol) and MeNH 2 (19.4 mL, 38.6 mmol, 2N in THF) in MeOH (100 mL). The reaction was stirred at 20 °C for 16 h. MgCl 2 (5.38 g, 58 mmol) was added to reaction mixture. After stirring at 20 °C for 15 min, NaBH 4 (5.57 g, 154.6 mmol) was added. The reaction was stirred at 20°C for further 15 min. The reaction was quenched by water (100 mL) and extracted with DCM (300 mL X 2). The combined organic solvents were washed with brine (100 mL), dried over anhydrous MgSO 4 , concentrated in vacuum and purified by silica gel column (1-20% MeOH/CH 2 Cl 2 ) to give the title compound (1.5 g, 28% yield) as a colorless oil. LCMS: m/z 275.0 [M+1] + ; t R =1.549 min.

[0815] Step 3: Synthesis of tert-butyl (1S,5S,6R,7R)-7-((6-chloropyridazin-3-yl)(methyl)amino)-6- fluoro-3-oxa-9-azabicyclo[3.3.1]nonane-9-carboxylate (INT-4). A mixture of tert-butyl (1S,5S,6R,7R)-6- fluoro-7-(methylamino)-3-oxa-9-azabicyclo [3.3.1]nonane- 9-carboxylate (1.5 g, 5.77 mmol), 3,6- dichloropyridazine (1.29 g, 8.65 mmol) and DIPEA (2.23 g, 17.3 mmol) in DMSO (20 mL) was stirred at 135°C for 96 h under N 2 atmosphere. The reaction mixture was concentrated in vacuum and purified by silica gel column (20-50% EtOAc/petroleum ether) to give the title compound (360 mg, 16.2 % yield) as a white solid. LCMS: m/z 387 [M+H] + .

Example B2d: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(cyclopropyl)amino) - 2-fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate (INT-5).

[0816] Step 1: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)amino)-2-fluoro-8- azabicyclo[3.2.1]octane-8-carboxylate. Into a 1-L round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl (1S,2R,3R,5R)-3-amino-2-fluoro-8-azabicyclo[3.2.1]octane-8- carboxylate (100 g, 409 mmol, 1.0 equiv), 3,6-dichloropyridazine (122 g, 819 mmol, 2.0 equiv), DIEA (156 g, 1.23 mol, 3.0 equiv), DMSO (1 L). The resulting solution was stirred for 18 h at 130 °C. The reaction mixture was cooled to room temperature. The resulting solution was diluted with 2 L of H 2 O. The resulting solution was extracted with 3x2 L of ethyl acetate and the organic layers were combined. The combined organic layers were washed with 2x1L of brine and dried over anhydrous sodium sulfate. The solids were filtered out. The resulting mixture was concentrated under vacuum. The crude product was purified by silica gel column chromatography (gradient elution with EtOAc in petroleum ether = 0 to 50%). This resulted in the title compound (63 g) as a yellow solid.

[0817] Step 2: (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(cyclopropyl)amino) -2-fluoro-8- azabicyclo[3.2.1]octane-8-carboxylate. To a solution of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin- 3-yl)amino)-2-fluoro-8-azabicyclo [3.2.1]octane-8-carboxylate (20.0 g, 56.05 mmol, 1.0 equiv.) in 1,2- dichloroethane (400 mL) were added cyclopropylboronic acid (14.4 g, 168 mmol, 3.0 equiv.), Cu(OAc) 2 (13.2 g, 72.9 mmol, 1.3 equiv.), 2,2'-bipyridine (11.38 g, 72.9 mmol, 1.3 equiv.), Na 2 CO 3 (17.82 g, 168 mmol, 3.0 equiv.) and 4A molecular sieves (15 g) at room temperature. The mixture was heated to 50 ºC for 16 h under air atmosphere, then the reaction mixture cooled to room temperature. The mixture was filtered through Celite® brand filter agent and the filtrate was concentrated and purified on silica flash column with gradient elution with EtOAc in petroleum ether = 0 to 60%) to afford the title compound 7.0 g (31.4%) as an off-white solid. LCMS: m/z 397 [M+H] + .

Example B3: General Synthesis Method F - Heterocycle Suzuki and Chiral Purification of Penultimate Intermediates

Specific Example of General Method F: Synthesis of 4-(2-fluoro-4-(6-(((1S,2S,3R,5R)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)-5- hydroxyphenyl)-1-methylpyridin-2(1H)- one and 4-(2-fluoro-4-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1 ]octan-3- yl)(methyl)amino)pyridazin-3-yl)-5-hydroxyphenyl)-1-methylpy ridin-2(1H)-one.

[0818] Step 1: Synthesis of 1-bromo-4-chloro-5-fluoro-2-(methoxymethoxy)benzene: Into a 500-mL 4- necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed NaH (3.19 g, 133 mmol, 1.20 equiv), THF (250 mL). This was followed by the addition of 2-bromo-5-chloro-4- fluorophenol (25.00 g, 111 mmol, 1.00 equiv) dropwise with stirring at 0 °C. The reaction mixture was stirred for 30 min at 0 °C. To this was added a solution of bromo(methoxy)methane (16.63 g, 133 mmol, 1.20 equiv) in THF (50 mL) dropwise with stirring at 0 °C. The resulting solution was stirred overnight at rt. The reaction was then quenched by the addition of 500 mL of water/ice. The resulting solution was extracted with 3x500 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 500 mL of brine, dried over anhydrous sodium sulfate, and concentrated under vacuum. The residue was purified by silica gel flash chromatography eluting with ethyl acetate/petroleum ether (0-20%) to afford the title compound (25 g, 83.7%) as a yellow oil.

[0819] Step 2: Synthesis of 2-(4-chloro-5-fluoro-2-(methoxymethoxy)phenyl)-4,4,5,5-tetra methyl-1,3,2- dioxaborolane: Into a 250-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 1-bromo-4-chloro-5-fluoro-2-(methoxymethoxy)benzene (10.00 g, 37.1 mmol, 1.00 equiv), dioxane (100 mL), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (11.3 g, 44.5 mmol, 1.20 equiv), KOAc (7.28 g, 74.2 mmol, 2.00 equiv), and Pd(dppf)Cl 2 (1.36 g, 1.86 mmol, 0.05 equiv). The resulting solution was stirred overnight at 100 °C. The reaction was then quenched by the addition of 200 mL of water. The resulting solution was extracted with 3x200 mL of ethyl acetate and the organic layers combined, washed with brine, dried over anhydrous sodium sulfate, and concentrated under vacuum. The residue was purified by silica gel flash chromatography eluting with ethyl acetate/petroleum ether (0-20%) to afford the title compound (7 g, 59.6% yield) as an off-white solid.

[0820] Step 3: Synthesis of (±) tert-butyl (1R,2S,3S,5S)-3-((6-(4-chloro-5-fluoro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate: Into a 50-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (±) tert-butyl (1S,2R,3R,5R)-3-[(6-chloropyridazin-3-yl)(methyl)amino]-2-fl uoro-8- azabicyclo[3.2.1]octane-8- carboxylate (INT-1, 8.20 g, 22.1 mmol, 1.00 equiv), 1,4-dioxane (20.0 mL), 2-[4- chloro-5-fluoro-2-(methoxymethoxy)phenyl] -4,4,5,5-tetramethyl-1,3,2-dioxaborolane (7.00 g, 22.1 mmol, 1.00 equiv), K 2 CO 3 (6.11 g, 44.2 mmol, 2.00 equiv), Pd(dppf)Cl 2 (0.81 g, 1.11 mmol, 0.05 equiv), H 2 O (4.00 mL). The resulting solution was stirred overnight at 100 degrees C. The reaction was then quenched by the addition of 100 mL of water. The resulting solution was extracted with 3x100 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 1 x100 ml of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by silica gel flash chromatography eluting with ethyl acetate/petroleum ether (0-80%) to afford the title compound (5.0 g, 43.1% yield) as a yellow oil.

[0821] Step 4: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(5-fluoro-2-(methoxymethoxy)-4- (1-methyl-2-oxo-1,2-dihydropyridin-4-yl)phenyl)pyridazin-3-y l)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate: Into a 50-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed (±) tert-butyl (1R,2S,3S,5S)-3-([6-[4-chloro-5-fluoro-2- (methoxymethoxy)phenyl]pyridazin-3-yl](methyl)amino) -2-fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate (5.00 g, 9.524 mmol, 1.00 equiv), Dioxane (10.00 mL), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridin-2-one (2.69 g, 11.429 mmol, 1.20 equiv), K 2 CO 3 (2.63 g, 19.048 mmol, 2.00 equiv), Pd(dppf)Cl 2 (0.35 g, 0.476 mmol, 0.05 equiv), H 2 O (2.00 mL). The resulting solution was stirred overnight at 100 degrees C. The reaction was then quenched by the addition of 100 mL of water. The resulting solution was extracted with 3x100 mL of ethyl acetate and the organic layers combined. The resulting mixture was washed with 1 x100 of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (0-80%) to afford the title compound as a yellow solid.

[0822] Step 5: Chiral purification to isolate tert-butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(5-fluoro-2- (methoxymethoxy)-4-(1-methyl-2-oxo-1,2-dihydropyridin-4-yl)p henyl)pyridazin-3-yl)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate and tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(5-fluoro-2- (methoxymethoxy)-4-(1-methyl-2-oxo-1,2-dihydropyridin-4-yl)p henyl)pyridazin-3-yl)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate (PENT-1A & PENT-1B): Chiral Separation method - (±) tert- butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(5-fluoro-2-(methoxymethoxy)-4- (1-methyl-2-oxo-1,2-dihydropyridin-4- yl)phenyl)pyridazin-3-yl)(methyl)amino)-8-azabicyclo[3.2.1]o ctane-8-carboxylate, 600 mg, was purified by chiral SFC purification by the following method to give the two title compounds (PENT-1A, t R = 4.01 min, 650 mg) and (PENT-1B, t R = 5.65 min, 600 mg). Column: CHIRALPAK IA, 2*25cm, 5 Pm; Mobile Phase A: CO 2 , Mobile Phase B: MeOH: DCM=8:1; Flow rate: 40 mL/min; Gradient: 50% B; wavelength 220 nm.

[0823] Step 6: Synthesis of 4-(2-fluoro-4-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1 ]octan-3- yl)(methyl)amino)pyridazin-3-yl)-5-hydroxyphenyl)-1-methylpy ridin-2(1H)-one and 4-(2-fluoro-4-(6- (((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)(meth yl)amino)pyridazin-3-yl)-5- hydroxyphenyl)-1-methylpyridin-2(1H)-one (Compound 1A & Compound 1B): Into a 25-mL round- bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl

(1S,2R,3R,5R)-2-fluoro-3-([6-[5-fluoro-2-(methoxymethoxy)-4- (1-methyl-2-oxopyridin-4-yl)phenyl] pyridazin-3-yl](methyl)amino)-8-azabicyclo[3.2.1]octane-8-ca rboxylate (100.0 mg, 1 equiv), HCl (gas) in 1,4-dioxane (2.00 mL). The resulting solution was stirred for 3 h at rt. The reaction was then quenched by the addition of 20 mL of aqueous NaHCO 3 . The resulting solution was extracted with 3x20 mL of

dichloromethane. The resulting mixture was washed with 1 x20 mL brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions: Column, XB-C18; mobile phase, 0.1%NH 4 HCO 3 /H 2 O: ACN=10-60%; Detector, 254 nm; to give the title compound as Compound 1A (27 mg, 35.6%) as a yellow solid.

[0824] 1 H NMR (300 MHz, DMSO-d6) d 13.32 (s, 1H), 8.32 (d, J = 9.9 Hz, 1H), 7.94 (d, J = 12.4 Hz, 1H), 7.79 (d, J = 7.1 Hz, 1H), 7.41 (d, J = 9.9 Hz, 1H), 7.09 (d, J = 6.8 Hz, 1H), 6.59 (s, 1H), 6.46 (dd, J = 6.9, 2.1 Hz, 1H), 5.12 (s, 1H), 4.66 (d, J = 52.1 Hz, 1H), 3.56 (s, 2H), 3.47 (s, 3H), 3.06 (d, J = 1.8 Hz, 3H), 2.26 (t, J = 12.3 Hz, 1H), 1.79 (s, 2H), 1.70 (d, J = 12.6 Hz, 2H), 1.56 (d, J = 8.6 Hz, 1H). LCMS: m/z 454.3 [M+H] + .

[0825] Following the procedure above but with PENT-1B (100 mg, 0.167 mmol) gave the title compound as Compound 1B (26 mg, 34.3% yield) as a yellow solid. 1 H NMR (300 MHz, DMSO-d6) d 13.33 (s, 1H), 8.31 (d, J = 10.0 Hz, 1H), 7.94 (d, J = 12.4 Hz, 1H), 7.79 (d, J = 7.1 Hz, 1H), 7.40 (d, J = 9.9 Hz, 1H), 7.09 (d, J = 6.9 Hz, 1H), 6.59 (t, J = 1.7 Hz, 1H), 6.45 (dt, J = 7.1, 1.9 Hz, 1H), 5.00 (s, 1H), 4.64 (d, J = 52.2 Hz, 1H), 3.53 (s, 2H), 3.47 (s, 3H), 3.05 (d, J = 1.8 Hz, 3H), 2.31– 2.17 (m, 1H), 1.79 (s, 2H), 1.68 (d, J = 12.3 Hz, 2H), 1.55 (s, 1H). LCMS: m/z 454.3 [M+H] + .

[0826] The examples in Table 3 were synthesized using a sequence analogous to that used to synthesize Compound 1A and Compound 1B by general Method F.

Table 3. Additional Compounds Prepared by General Method F.

Example B4: General Synthesis Method - Heterocycle Suzuki (Reverse) and Chiral Purification of Penultimate Intermediates.

Specific Example of General Method T: Compound 2A & 2B: Synthesis of 2-(6-(((1S,3R,5R)-6,6- difluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridaz in-3-yl)-5-(6-methoxypyridazin-4- yl)phenol and 2-(6-(((1R,3S,5S)-6,6-difluoro-8-azabicyclo[3.2.1]octan-3-yl )(methyl)amino)pyridazin-3- yl)-5-(6-methoxypyridazin-4-yl)phenol.

[0827] Step 1: Synthesis of (±) tert-butyl (1S,3R,5R)-3-((6-(4-chloro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-6,6-dif luoro-8-azabicyclo[3.2.1]octane-8- carboxylate: A mixture of (±) tert-butyl(1S,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amin o)-6,6-difluoro- 8-azabicyclo[3.2.1]octane-8-carboxylate (400 mg, 1.03 mmol), 4-chloro-2-(methoxymethoxy)phenylboronic acid (335 mg, 1.55 mmol), Pd(dppf)Cl 2 (154 mg, 0.21 mmol) and K 2 CO 3 (284 mg, 2.06 mmol) in 4 mL of dioxane and 1 mL of H 2 O was degassed and stirred at 110 o C for 2 h. After cooling to rt, the mixture was concentrated and purified by silica gel column (10-60% EtOAc/petroleum ether) to give the title compound (350 mg, 64.8% yield) as a yellow solid LCMS: m/z 525.2 [M+H] + ; t R = 2.23 min.

[0828] Step 2: Synthesis of (±) tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2-(methoxymethoxy)-4-(6- methoxypyridazin-4-yl)phenyl)pyridazin-3-yl)(methyl)amino)-8 -azabicyclo[3.2.1]octane-8-carboxylate: A mixture of (±) tert-butyl (1S,3R,5R)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyridazi n-3- yl)(methyl)amino)-6,6-difluoro-8-azabicyclo[3.2.1]octane-8-c arboxylate (380 mg, 0.73 mmol),

4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolan e) (241 mg, 0.95 mmol), Pd 2 (dba) 3 (137 mg, 0.15 mmol), X-Phos (142 mg, 0.30 mmol) and KOAc (143 mg, 1.46 mmol) in 3 mL of 1,4-dioxane was degassed and stirred at 110 o C for 2 h. After cooling to room temperature, 5-chloro-3-methoxypyridazine (137 mg, 0.95 mmol), K 2 CO 3 (202 mg, 1.46 mmol) and H 2 O (0.5 mL) were added. The mixture was stirred at 110 o C for 2 h, concentrated and purified by silica gel column (10-80% EtOAc/petroleum ether) to give the title compound (350 mg, 80% yield) as a yellow solid. LCMS: m/z 599.3 [M+H] + ; t R = 1.91 min.

[0829] Step 3: Chiral purification to isolate tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2- (methoxymethoxy)-4-(6-methoxypyridazin-4-yl)phenyl)pyridazin -3-yl)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate and tert-butyl (1R,3S,5S)-6,6-difluoro-3-((6-(2- (methoxymethoxy)-4-(6-methoxypyridazin-4-yl)phenyl)pyridazin -3-yl)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate (PENT-2A & PENT-2B): (±) tert-butyl (1S,3R,5R)-6,6-difluoro-3- ((6-(2-(methoxymethoxy)-4-(6-methoxypyridazin-4-yl)phenyl)py ridazin-3-yl)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate, 350 mg, was purified by chiral SFC purification by the following method to give the two title compounds (PENT-2A, t R = 1.42 min, 170 mg) and (PENT-2B, t R = 1.81 min, 180 mg). Instrument: SFC-80 (Thar, Waters); Column: OJ 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO 2 / MeOH(0.2%Methanol Ammonia) = 75/25; Flow rate: 80 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 3.0 min; Sample solution: 100 mg dissolved in 15 mL methanol; Injection volume: 1.0 mL.

[0830] Step 4: Synthesis of 2-(6-(((1S,3R,5R)-6,6-difluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-5-(6-methoxypyridazin-4-yl) phenol and 2-(6-(((1R,3S,5S)-6,6-difluoro- 8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)- 5-(6-methoxypyridazin-4-yl)phenol (Compound 2A & Compound 2B): To the mixture of PENT-2A (100 mg, 0.17 mmol) in CH 2 Cl 2 (3 mL) was added 4N HCl in dioxane (1.5 mL). The mixture was stirred at room temperature for 1 h and concentrated to dryness, then dissolved in water and saturated NaHCO3 aqueous solution was added until pH to 8-9. The mixture was extracted with CH 2 Cl 2 /MeOH (10 mL X 3, 10:1 v/v). The combined organic solvents were concentrated and dried by lyophilization to give the title compound as Compound 2A (46 mg, 61%) as yellow solid (61% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 9.35 (s, 1H), 8.33 (d, J = 10.0 Hz, 1H), 8.05 (d, J = 8.3 Hz, 1H), 7.56 (s, 1H), 7.49 (d, 2H), 7.34 (d, J = 9.9 Hz, 1H), 5.12 (s, 2H), 4.08 (s, 3H), 3.64 (s, 1H), 3.46 (d, J = 13.7 Hz, 2H), 3.00 (s, 4H), 2.42– 2.28 (m, 2H), 1.98– 1.91 (m, 1H), 1.85– 1.73 (m, 2H), 1.75– 1.61 (m, 1H). LCMS: m/z 455.3 [M+H] + ; t R = 1.69 min.

[0831] Following the procedure above but with PENT-2B (100 mg, 0.17 mmol) gave the title compound as Compound 2B (32 mg, 42.1% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) d 9.33 (s, 1H), 8.36 (d, J = 9.7 Hz, 1H), 8.04 (d, J = 8.5 Hz, 1H), 7.53 (s, 1H), 7.45 (d, J = 15.0 Hz, 2H), 7.31 (d, J = 9.6 Hz, 1H), 5.12 (s, 1H), 4.08 (s, 3H), 3.63 (s, 1H), 3.44 (d, J = 13.5 Hz, 1H), 2.99 (s, 3H), 2.89 (s, 1H), 2.42– 2.28 (m, 2H), 1.98– 1.89 (m, 1H), 1.84– 1.72 (m, 2H), 1.75– 1.63 (m, 1H). LCMS: m/z 455.2 [M+H] + ; t R = 1.69 min. Example B5. General Synthesis Method K - Heterocycle Suzuki (Reverse) and Chiral Purification of Aryl Halide Intermediates.

[0832] Specific Example of General Method K: Compound 28A & 28B: Synthesis of 5-(4-(6- (((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3-yl)(meth yl)amino)pyridazin-3-yl)-3- hydroxyphenyl)pyrazine-2-carbonitrile and 5-(4-(6-(((1R,2R,3S,5S)-2-fluoro-9-azabicyclo[3.3.1]nonan-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)pyrazine-2- carbonitrile.

[0833] Step 1: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-(4-chloro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-9-azabicyclo[3.3.1]nonane-9- carboxylate: A mixture of (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amino)-2-fl uoro- 9-azabicyclo[3.3.1]nonane-9-carboxylate (INT-3, 1.10 g, 2.60 mmol), 4-chloro-2- (methoxymethoxy)phenylboronic acid (843 mg, 3.90 mmol), Pd(dppf)Cl 2 (190 mg, 0.26 mmol) and K 2 CO 3 (720 mg, 5.20 mmol) in 1,4-dioxane (12 mL) and H 2 O (4 mL) was strirred at 110 °C for 2 h under N 2 atmosphere. After cooling to room temperature , the mixture was concentrated and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give of the title compound (1.11 g, 82% yield) as a yellow solid. LCMS: m/z 521.3 [M+H] + ; t R = 1.80 min.

[0834] Step 2: Chiral purification to isolate of tert-butyl (1S,2R,3R,5R)-3-((6-(4-chloro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-9-azabicyclo[3.3.1]nonane-9- carboxylate and tert-butyl (1R,2S,3S,5S)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyrid azin-3- yl)(methyl)amino)-2-fluoro-9-azabicyclo[3.3.1]nonane-9-carbo xylate (HETX-28A & HETX-28B): Racemic intermediat, (±) tert-butyl (1S,2R,3R,5R)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyrid azin-3- yl)(methyl)amino)-2-fluoro-9-azabicyclo[3.3.1]nonane-9-carbo xylate, 1570 mg, was purified by chiral SFC purification by the following method to give the two title compounds as HETX-28A (t R = 0.61 min, 820 mg) and HETX-28B (t R = 2.1 min, 720 mg). Instrument: SFC-80 (Thar, Waters); Column: AD 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO 2 /MEOH/ACN (0.2%Methanol Ammonia)

=40/30/30; Flow rate: 80 g/min. Back pressure: 100 bar; Detection wavelength: 280 nm; Cycle time: 15 min; Sample solution: 1570 mg dissolved in 25 mL methanol; Injection volume: 4 mL.

[0835] Step 3: Synthesis of tert-butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(2-(methoxymethoxy)-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)pyridazin-3-yl)(m ethyl)amino)-9-azabicyclo[3.3.1]nonane- 9-carboxylate and tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(2-(methoxymethoxy)-4-(4,4,5,5- tetramethyl- 1,3,2-dioxaborolan-2-yl)phenyl)pyridazin-3-yl)(methyl)amino) -9-azabicyclo[3.3.1]nonane-9-carboxylate (BOR-28A & BOR-28B): A mixture of HETX-28A (110 mg, 0.21 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (54 mg, 0.315 mmol), Pd 2 (dba) 3 (38 mg, 0.042 mmol), X-phos (40 mg, 0.084 mmol) and KOAc (42 mg, 0.42 mmol) in 1,4-dioxane (5.0 mL) was strirred at 100 °C for 2 h under N 2 atmosphere. After cooling to room temperature, the crude mixture containing the title compound as BOR-28A was used directly in the next step. LCMS: m/z 613.2 [M+H] + ; t R = 2.04 min.

[0836] Following the above procedure, but with HETX-28B (100 mg, 0.19 mmol), gave a crude solution of the title compound as BOR-28B, which was used without further purification in the next step. LCMS: m/z 613.2 [M+H] + ; t R = 1.86 min.

[0837] Step 4: Synthesis of tert-butyl (1S,2R,3R,5R)-3-((6-(4-(5-cyanopyrazin-2-yl)-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-9-azabicyclo[3.3.1]nonane-9- carboxylate and tert-butyl (1R,2S,3S,5S)-3-((6-(4-(5-cyanopyrazin-2-yl)-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-9-azabicyclo[3.3.1]nonane-9- carboxylate (PENT-28A & PENT-28B): To the crude solution of BOR-28A from step 3 (0.16 mmol), was added 2-chloropyrazine (34 mg, 0.24 mmol), Pd(dppf)Cl 2 (12 mg, 0.016 mmol) and K 2 CO 3 (45 mg, 0.32 mmol) in 1,4-dioxane (5 mL) and H 2 O (1 mL) was strirred at 110 ° C for 2 h under N 2 atmosphere. After cooling to room temperature, the mixture was concentrated and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give the title compound as PENT-28A (50 mg, 52% yield) as a yellow oil. LCMS: m/z 590.3 [M+H] + ; t R = 1.80 min.

[0838] Following the procedure above, but with BOR-28B (0.16 mmol), gave the title compound as PENT- 28B (50 mg, 52% yield) as a yellow oil. LCMS: m/z 590.1 [M+H] + ; t R = 1.82 min.

[0839] Step 5: Synthesis of 5-(4-(6-(((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)pyrazine-2- carbonitrile 5-(4-(6-(((1R,2R,3S,5S)-2- fluoro-9-azabicyclo[3.3.1]nonan-3-yl)(methyl)amino)pyridazin -3-yl)-3-hydroxyphenyl)pyrazine-2- carbonitrile (Compound 28A & Compound 28B): To a solution of PENT-28A in (50 mg, 0.08mmol) in CH 2 Cl 2 (4.0 mL) was added TFA (2 mL). The mixture was stirred at 25 o C for 1 h, then concentrated. The crude solid was dissolved into water (3 mL) and neutralized with saturated K 2 CO 3 aqueous solution. The precipitate was collected by filtration, washed with water, and dried under reduced pressure to give the title compound as Compound 28A (20 mg, 67% yield) as a yellow solid. 1 H NMR (500 MHz, MeOD-d 4 ) d 9.34 (s, 1H), 9.07 (s, 1H), 8.23 (d, J = 9.9 Hz, 1H), 7.99 (d, J = 8.3 Hz, 1H), 7.87– 7.75 (m, 2H), 7.37 (d, J = 9.9 Hz, 1H), 6.02– 5.85 (m, 1H), 3.42– 3.38 (m, 2H), 3.17 (s, 3H), 2.66– 2.58 (m, 1H), 2.11– 1.77 (m, 7H). LCMS: m/z 446.1 [M+H] + ; t R = 1.38 min.

[0840] Following the procedure above, but with PENT-28B, gave the title compound as Compound 28B (20.3 mg, 54% yield) as a yellow solid. 1 H NMR (500 MHz, MeOD-d 4 ) d 9.34 (s, 1H), 9.07 (s, 1H), 8.23 (d, J = 9.9 Hz, 1H), 7.99 (d, J = 8.3 Hz, 1H), 7.87– 7.75 (m, 2H), 7.37 (d, J = 9.9 Hz, 1H), 6.02– 5.85 (m, 1H), 3.42– 3.38 (m, 2H), 3.17 (s, 3H), 2.66– 2.58 (m, 1H), 2.11– 1.77 (m, 7H). LCMS: m/z 446.1 [M+H] + ; t R = 1.38 min.

[0841] The examples in Table 4 were synthesized using a sequence analogous to that used to synthesize Compound 28A and Compound 28B by general Method K.

Table 4. Additonal Compounds Prepared by General method K

SMSM# Structure Chiral Chiral Intermediate

HPLC or SFC Purification Method Retention Time (min) Instrument: SFC-200 (Thar,

Waters)

Column: AD 50*250mm, 10um

23A (Daicel) 0.83

Column temperature: 35 °C

Mobile phase: CO2/ETOH

(1%Methanol Ammonia) = 65/35

Flow rate: 120 g/min

Back pressure: 100 bar

Detection wavelength: 214 nm

23B Cycle time: 5.7 min 1.61

Sample solution: 3000 mg dissolved

in 80mL MeOH

Injection volume: 3.5 mL

Instrument: SFC-80 (Thar, Waters)

Column: AD 20*250mm, 10um

(Daicel)

24A Column temperature: 35 ºC 0.54

Mobile phase: CO2/MEOH/ACN

(0.2%Methanol Ammonia)

=40/30/30

Flow rate: 80 g/min

Back pressure: 100 bar

Detection wavelength: 280 nm

24B Cycle time: 15 min 2.10

Sample solution: 1570mg dissolved

in 25ml Methanol

Injection volume: 4 mL

Instrument: SFC-200 (Thar,

Waters)

25A Column: AD 20*250mm, 10um

(Daicel) 1.05

Column temperature: 35 ºC

Mobile phase: [0842] Example B6. General Synthesis Method G - Heterocycle Suzuki and Chiral Purification of Aryl Halide Intermediates

Specific Example of General Method G, Compound 3A & 3B: Synthesis of 2-(6-(((1S,3R,5R)-6,6- difluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridaz in-3-yl)-5-(1-methyl-1H-pyrazol-4- yl)phenol and 2-(6-(((1R,3S,5S)-6,6-difluoro-8-azabicyclo[3.2.1]octan-3-yl )(methyl)amino)pyridazin-3- yl)-5-(1-methyl-1H-pyrazol-4-yl)phenol..

[0843] Step 1: Chiral purification to isolate tert-butyl (1S,3R,5R)-3-((6-(4-chloro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-6,6-dif luoro-8-azabicyclo[3.2.1]octane-8- carboxylate and tert-butyl (1R,3S,5S)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyridazi n-3- yl)(methyl)amino)-6,6-difluoro-8-azabicyclo[3.2.1]octane-8-c arboxylate (HEX-3A & HEX-3B): (±) tert- butyl (1S,3R,5R)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyridazi n-3-yl)(methyl)amino)-6,6-difluoro-8- azabicyclo[3.2.1]octane-8-carboxylate, 350 mg, was purified by chiral SFC purification by the following method to give the two title compounds HETX-3A (t R = 1.63 min, 140 mg) and HETX-3B (t R = 1.94 min, 135 mg). Instrument: SFC-80 (Thar, Waters); Column: OJ 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO2/ MeOH(0.2%Methanol Ammonia) = 75/25; Flow rate: 80 g/min; Back pressure: 100 bar; Detection wavelength: 285 nm; Cycle time: 4.5 min; Sample solution: 460 mg dissolved in 25 mL methanol; Injection volume: 1.0 mL.

[0844] Step 2: Synthesis of tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2-(methoxymethoxy)-4-(1-methy l- 1H-pyrazol-4-yl)phenyl)pyridazin-3-yl)(methyl)amino)-8-azabi cyclo[3.2.1]octane-8-carboxylate and tert-butyl (1R,3S,5S)-6,6-difluoro-3-((6-(2-(methoxymethoxy)-4-(1-methy l-1H-pyrazol-4- yl)phenyl)pyridazin-3-yl)(methyl)amino)-8-azabicyclo[3.2.1]o ctane-8-carboxylate (PENT-3A & PENT- 3B): A mixture of HETX-3A (140 mg, 0.27 mmol), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-1H-pyrazole (85 mg, 0.41 mmol), X-phos-Pd 2nd G (36 mg, 0.05 mmol) and K 3 PO 4 (114 mg, 0.54 mmol) in 4 mL of dioxane and 1 mL of H 2 O was degassed and stirred at 110 o C for 2 h. After cooling to room temperature, the mixture was concentrated and purified by silica gel column (10-90% EtOAc/petroleum ether) to give the title compound as PENT-3A (85 mg, 55.8% yield) as a yellow solid. LCMS: m/z 571.3 [M+H] + ; tR = 1.90 min.

[0845] Following the procedure above, but with HETX-3B (135 mg, 0.26 mmol) gave the title compound as PENT-3B (80 mg, 54.5% yield) as a yellow solid. LCMS: m/z 571.3 [M+H] + ; t R = 1.90 min.

[0846] Step 3: Synthesis of 2-(6-(((1S,3R,5R)-6,6-difluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-5-(1-methyl-1H-pyrazol-4-yl )phenol and 2-(6-(((1R,3S,5S)-6,6- difluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridaz in-3-yl)-5-(1-methyl-1H-pyrazol-4- yl)phenol (Compound 3A & Compound 3B): To a mixture of PENT-3A (85 mg, 0.15 mmol) in CH 2 Cl 2 (5 mL) was added 4N HCl in dioxane (2.5 mL). The mixture was stirred at room temperature for 1 h and concentrated to dryness. The residue was dissolved in water and saturated NaHCO 3 aqueous solution was added till pH to 8-9. The mixture was extracted with CH 2 Cl 2 /MeOH (10 mL X 3, 10:1 v/v). The combined organic solvents were concentrated and dried by lyophilization to give the title compound as Compound 3A (40 mg, 62.8% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 8.22 (d, J = 10.1 Hz, 1H), 8.20 (s, 1H), 7.91 (s, 1H), 7.84 (d, J = 8.3 Hz, 1H), 7.30 (d, J = 9.9 Hz, 1H), 7.15 (s, 1H), 7.13 (d, 1H), 5.08 (s, 1H), 3.87 (s, 3H), 3.63 (s, 1H), 3.44 (d, J = 13.1 Hz, 1H), 2.96 (d, J = 17.8 Hz, 3H), 2.86 (d, J = 26.5 Hz, 1H), 2.42 – 2.29 (m, 2H), 2.01– 1.87 (m, 1H), 1.87– 1.74 (m, 2H), 1.73– 1.60 (m, 1H). LCMS: m/z 427.2 [M+H] + ; t R = 1.64 min. [0847] Following the procedure above but with PENT-3B (80 mg, 0.088 mmol) gave the title compound as Compound 3B (33 mg, 55.1% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 8.22 (d, J = 10.0 Hz, 1H), 8.20 (s, 1H), 7.91 (s, 1H), 7.84 (d, J = 8.2 Hz, 1H), 7.30 (d, J = 9.9 Hz, 1H), 7.17– 7.10 (m, 2H), 5.08 (s, 1H), 3.87 (s, 3H), 3.63 (s, 1H), 3.44 (d, J = 12.5 Hz, 1H), 3.30 (s, 1H), 2.97 (s, 3H), 2.38– 2.30 (m, 2H), 1.97 – 1.87 (m, 1H), 1.86– 1.74 (m, 2H), 1.74– 1.65 (m, 1H). LCMS: m/z 427.2 [M+H] + ; t R = 1.64 min.

[0848] The examples in Table 5A and Table 5B were synthesized using a sequence analogous to that used to synthesize Compound 3A and Compound 3B by general Method G.

Table 5A. Intermediates Prepared by General Method G.

Table 5B. Additional examples prepared by General method G.

[0849] Example B7. General Synthesis Method L. Convergent Suzuki and Chiral Purification of Penultimate Intermediates

Specific Example of General Method L, Compound 4A & 4B: Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro- 8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)- 5-(1H-imidazol-1-yl)phenol and 2-(6- (((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)(meth yl)amino)pyridazin-3-yl)-5-(1H-imidazol-1- yl)phenol.

[0850] Step 1: Synthesis of 1-bromo-4-iodo-2-(methoxymethoxy)benzene. MOMBr (1.25 g, 10 mmol) was added to a stirred solution of 2-bromo-5-iodophenol (1.5 g, 5 mmol) and K 2 CO 3 (1.38 g, 10 mmol) in 20 mL of DMF at 0 o C. The mixture was then stirred at room temperature for 16 h, quenched with 20 mL of water, extracted with EtOAc (3x20 mL). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated, and purified by silica gel column (0-5% EtOAc/petroleum ether) to give the title compound (1.45 g, 79% yield) as a colorless liquid. LCMS: m/z 343.1 [M+H] + ; t R = 1.50 min.

[0851] Step 2: Synthesis of 1-(4-bromo-3-(methoxymethoxy)phenyl)-1H-imidazole. A mixture of 1- bromo-4-iodo-2-(methoxymethoxy)benzene (3.42 g, 10 mmol), 1H-imidazole (1.36 g, 20 mmol), CuI (380 mg, 2 mmol) and Cs 2 CO 3 (6.52 g, 20 mmol) in 50 mL of DMF was stirred at 100 o C for 16 h. Water (300 mL) was added to the reaction, then the mixture was extracted with ethyl acetate (3x100 mL). The combined organic phases were dried and concentrated, then purified by silica gel chromatography (80%

EtOAc/petroleum ether) to give the title compound (2.5 g, 88% yield). LCMS: m/z 283.2 [M+H] + ; t R = 1.03 min.

[0852] Step 3: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-(4-(1H-imidazol-1-yl)-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate. A mixture of 1-(4-bromo-3-(methoxymethoxy)phenyl)-1H-imidazole (282 mg, 1.0 mmol), bis(pinacolato)diboron (380 mg, 1.5 mmol), Pd(dppf)Cl 2 (73 mg, 0.1 mmol) and KOAc (294 mg, 3.0 mmol) in 10 mL of dioxane was degassed and stirred at 110 o C for 2 h. The mixture was cooled to room temperature, and (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amino)-2-fl uoro-8- azabicyclo[3.2.1]octane-8-carboxylate INT-1 (250 mg, 0.7 mmol), Pd(dppf)Cl 2 (73 mg, 0.1 mmol), K 2 CO 3 (194 mg, 1.4 mmol) and water (2 mL) were added. The mixture was degassed and stirred at 110 o C for 2 h, then cooled to rt, concentrated, and purified by silica gel column (80% EtOAc/petroleum ether), to give the title compound (240 mg, 45% yield) as a yellow oil (240 mg, 45% yield). LCMS: m/z 540.2 [M+H] + ; t R = 1.89 min.

[0853] Step 4: Chiral purification to isolate tert-butyl (1S,2R,3R,5R)-3-((6-(4-(1H-imidazol-1-yl)-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate and tert-butyl (1R,2S,3S,5S)-3-((6-(4-(1H-imidazol-1-yl)-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate (PENT-4A & PENT-4B). (±) tert-butyl (1S,2R,3R,5R)-3-((6-(4-(1H-imidazol-1-yl)-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8-carboxylate, 240 mg, was purified by chiral SFC purification by the following method to give the two title compounds (PENT-4A, t R = 1.16 min, 80 mg) and (PENT-4B, t R = 2.15 min, 70 mg). Instrument: SFC-80 (Thar, Waters); Column: AD 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO 2 /MeOH

(0.2%Methanol Ammonia) = 45/55; Flow rate: 80 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 4.5 min; Sample solution: 240 mg dissolved in 20 mL methanol; Injection volume: 1.9 mL.

[0854] Step 5: Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-5-(1H-imidazol-1-yl)phenol and 2-(6-(((1R,2R,3S,5S)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)-5- (1H-imidazol-1-yl)phenol (Compound 4A & Compound 4B).2 mL of HCl in dioxane (4 N) was added to a stirred solution of PENT-4A (80 mg, 0.15 mmol) in 1 mL of CH 2 Cl 2 . The mixture was stirred at room temperature for 2 h, and concentrated under reduced pressure. The residue was dissolved in water, the pH value was adjusted to ~9 by using K 2 CO 3 aqueous solution, the solid was collected by filtration, and dried under reduced pressure to give the title compound as Compound 4A (32 mg, 54% yield) as a yellow solid 1 H NMR (500 MHz, DMSO-d 6 ) d 8.38 (d, J = 9.1 Hz, 1H), 8.30 (s, 1H), 8.00 (d, J = 8.3 Hz, 1H), 7.78 (s, 1H), 7.33 (d, J = 8.7 Hz, 1H), 7.19 (s, 1H), 7.09 (s, 2H), 5.18– 4.95 (m, 1H), 4.74– 4.51 (m, 1H), 3.52 (s, 2H), 3.03 (s, 3H), 2.37 (s, 1H), 2.28– 2.16 (m, 1H), 1.85– 1.59 (m, 4H), 1.58– 1.46 (m, 1H). LCMS: m/z 395.2 [M+H] + ; t R = 1.53 min.

[0855] Following the procedure above, but with PENT-4B (70 mg, 0.13 mmol) gave the title compound as Compound 4B (27 mg, 54% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 8.38 (d, J = 9.1 Hz, 1H), 8.30 (s, 1H), 8.00 (d, J = 8.3 Hz, 1H), 7.78 (s, 1H), 7.33 (d, J = 8.7 Hz, 1H), 7.19 (s, 1H), 7.09 (s, 2H), 5.18– 4.95 (m, 1H), 4.74– 4.51 (m, 1H), 3.52 (s, 2H), 3.03 (s, 3H), 2.37 (s, 1H), 2.28– 2.16 (m, 1H), 1.85– 1.59 (m, 4H), 1.58– 1.46 (m, 1H). LCMS: m/z 395.2 [M+H] + ; t R = 1.53 min.

[0856] The examples in Table 6 were synthesized using a sequence analogous to that used to synthesize Compound 4A and Compound 4B by general Method L.

Table 6. Examples Prepared by General Method L.

Example B8. General Synthesis Method J. Convergent Suzuki and Chiral Purification of Penultimate Intermediates, No Phenol Protecting Group

Specific Example of General Method J, Compound 5A & 5B: Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro- 8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)- 5-(1H-imidazol-1-yl)phenol and 2-(6- (((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)(meth yl)amino)pyridazin-3-yl)-5-(1H-imidazol-1- yl)phenol

[0857] Step 1: Syntheisis of 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole. K 2 CO 3 (4.04 g, 29.2 mmol ) was added to a solution of 4-fluoro-2-methoxy-1-nitrobenzene (5 g, 29.2 mmol) and 4,5- dibromo-2H-1,2,3-triazole (6.63 g, 29.2 mmol) in DMF (100 mL). The resulting mixture was stirred at 80 °C for 16 h. After cooling to room temperature, the mixture was poured into ice-water (100 mL) and extracted with EtOAc. The organic layers were washed with water (100 mL), dried over anhydrous MgSO 4 , and concentrated in vacuum to give the title compound (10 g , 97%) as a white solid. LCMS: m/z 378.9 [M+H] + ; t R =1.250 min.

[0858] Step 2: Synthesis of 2-methoxy-4-(2H-1,2,3-triazol-2-yl)aniline. Pd/C (1 g, 10 wt% on activated carbon) was added to a solution of 4,5-dibromo-2-(3-methoxy-4-nitrophenyl)-2H-1,2,3-triazole (10 g, 26.5 mmol) in MeOH (150 mL). The mixture was stirred under hydrogen atmosphere for 5 h and filtered. The filtrate was concentrated in vacuum to give the title compound (5 g, yield 98%) as a white solid. LCMS: m/z 191 [M+H] + ; t R =0.574 min.

[0859] Step 3: Synthesis of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid. A pre-cooled (- 15°C) solution of t-BuONO (2.61 g, 25.3 mmol) and 2-methoxy-4-(2H-1,2,3- triazol-2-yl)aniline (4.0 g, 21 mmol) in AcOH (80 mL) was added dropwise to a precooled solution of TfOH (3.79 g, 25.3 mmol) in AcOH (80 mL). The reaction was stirred for 10~20 min at 10-15 °C, then poured into cold Et 2 O (1000 mL). The precipitated diazonium salt was collected by filtration and dried in vacuum to give 2-methoxy-4-(2H-1,2,3- triazol-2-yl)benzenediazonium, trifluoromethanesulfonic salt (7.4 g, 95% yield) as a white solid. LCMS: m/z 202.2 [M+] + ; t R = 0.737 min. The 2-methoxy-4-(2H-1,2,3-triazol-2-yl)benzenediazonium,

trifluoromethanesulfonic salt (7.4 g, 21 mmol) was disolved in water (150 mL). Hypodiboric acid (4.74 g, 52.7 mmol) was added, and the reaction mixture was stirred at 25 °C for 3 h. The precipitate was collected by filtration and dried in vacuum to give the title compound (4.6 g, yield 99%) as a white solid. LCMS: m/z 220.2 [M+H] + ; t R =1.127 min.

[0860] Step 4: Synthesis of (2-hydroxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid. BBr 3 (4.6 mL, 18.26 mmol, 4M) was added to a solution of (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (1000 mg, 4.57 mmol) in DCM (4 mL). The reaction mixture was stirred at 20 °C for 18 h under N 2 atmosphere, concentrated in vacuum, and purified by silica gel column (10-25% EtOAc/petroleum ether) to give the title compound (450 mg, 59 % yield) as a yellow solid. LCMS: m/z 206.2 [M+H] + ; t R = 0.898 min.

[0861] Step 5: Synthesis of (±) tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(2-hydroxy-4-(2H-1,2,3-triazol- 2- yl)phenyl)pyridazin-3-yl)(methyl)amino)-9-azabicyclo[3.3.1]n onane-9-carboxylate. A mixture of (±) tert- butyl (1R,2S,3S,5S)-3-((6-chloropyridazin-3-yl) (methyl)amino)-2-fluoro-9-azabicyclo[3.3.1]nonane-9- carboxylate (INT-3, 639 mg, 1.83 mmol), (2-methoxy-4-(2H-1,2,3-triazol-2-yl)phenyl)boronic acid (450 mg, 2.20 mmol), K 2 CO 3 (505 mg, 3.67 mmol) and Pd(dppf)Cl 2 (201 mg, 0.275 mmol) in dioxane (8 mL) and water (2 mL) was degassed and stirred at 100 °C for 2 h under N 2 atmosphere. The reaction mixture was concentrated in vacuum and the residue was purified by silica gel column (20-50% EtOAc/petroleum ether) to give the title compound (450 mg, 42% yield) as a white solid. [0862] Step 6: Chiral separation to isolate tert-butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(2-hydroxy-4-(2H- 1,2,3-triazol-2-yl)phenyl)pyridazin-3-yl)(methyl)amino)-9-az abicyclo[3.3.1]nonane-9-carboxylate and tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(2-hydroxy-4-(2H-1,2,3-triazol- 2-yl)phenyl)pyridazin-3- yl)(methyl)amino)-9-azabicyclo[3.3.1]nonane-9-carboxylate (PENT-5A & PENT-5B). (±) tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(2-hydroxy-4-(2H-1,2,3-triazol- 2-yl)phenyl)pyridazin-3-yl)(methyl)amino)-9- azabicyclo[3.3.1]nonane-9-carboxylate, 450 mg, was purified by chiral SFC purification to give the two title compounds as (PENT-5A, t R = 1.55 min, 150 mg) and (PENT-5B, t R = 1.87 min, 130 mg) as white solids. LCMS: m/z 510.3 [M+H] + ; t R = 1.51 min. Instrument: SFC-150 (Thar, Waters); Column: IC 20*250mm, 10Pm (Daicel); Column temperature: 35 ºC; Mobile phase: CO2/MeOH(0.2% methanol ammonia) = 40/60; Flow rate: 120 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 5 min; Sample solution: 450 mg dissolved in 50 mL methanol and DCM; Injection volume: 1.9 mL.

[0863] Step 7: Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3-yl) (methyl) amino)pyridazin-3-yl)-5-(2H-1,2,3-triazol-2-yl)phenol and 2-(6-(((1R,2R,3S,5S)-2-fluoro-9- azabicyclo[3.3.1]nonan-3-yl)(methyl)amino)pyridazin-3-yl)-5- (2H-1,2,3-triazol-2-yl)phenol (Compound 5A & Compound 5B). HCl/dioxane (10 mL, 4N) was added to a mixture of PENT-5A (80 mg, 0.157 mmol) in CH 2 Cl 2 (2 mL) and MeOH (2 mL). The mixture was stirred at 25 °C for 2 h and concentrated. The residue was dissolved in water (2 mL), and aqueous K 2 CO 3 solution was added to adjust to pH = 10. The resulting solid was collected by filtration and dried under reduced pressure to give the title compound as Compound 5A (40 mg, 62% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 8.30 (d, J = 10.5 Hz, 1H), 8.14 (s, 2H), 8.09 (d, J = 9.0 Hz, 1H), 7.59-7.57 (m, 2H), 7.38 (d, J = 9.5 Hz, 1H), 5.78-5.68 (m, 1H), 4.77 (d, J = 54.0 Hz, 1H), 3.23-3.16 (m, 2H), 3.06 (d, J = 1.5 Hz, 3H), 2.43-2.36 (m, 2H), 1.91-1.84 (m, 3H), 1.73-1.58 (m, 4H). LCMS: m/z 410.2 [M+H] + ; t R = 1.77 min.

[0864] Following the procedure above, but starting with PENT-5B (80 mg, 0.157 mmol) gave the title compound as Compound 5B (48 mg, 75% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 8.30 (d, J = 10.0 Hz, 1H), 8.14 (s, 2H), 8.09 (d, J = 9.5 Hz, 1H), 7.59-7.57 (m, 2H), 7.38 (d, J = 10.0 Hz, 1H), 5.80- 5.65 (m, 1H), 4.77 (d, J = 51.0 Hz, 1H), 3.23-3.16 (m, 2H), 3.06 (d, J = 1.5 Hz, 3H), 2.40-2.37 (m, 2H), 1.91- 1.85 (m, 3H), 1.71-1.58 (m, 4H). LCMS: m/z 410.2 [M+H] + ; t R = 1.84 min.

[0865] The examples in Table 7 were synthesized using a sequence analogous to that used to synthesize Compound 5A and Compound 5B by general Method J.

Table 7. Examples prepared by General Method J.

[0866] Example B9. General Synthesis Method R. Convergent Suzuki and Chiral Purification of Aryl Halide Intermediates, no Phenol

Specific Example of General Method R, Compound 6A & 6B: Synthesis of 2-(6-(((1S,2S,3R,5R)-2- fluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin -3-yl)thieno[3,2-c]pyridin-4(5H)-one and 2-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl) (methyl)amino)pyridazin-3-yl)thieno[3,2- c]pyridin-4(5H)-one.

[0867] Step 1. Chiral separation to isolate tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate and tert-butyl (1R,2S,3S,5S)-3-((6- chloropyridazin-3-yl)(methyl)amino)-2-fluoro-8-azabicyclo[3. 2.1]octane-8-carboxylate (HETX-6A & HETX-6B). Racemic intermediate, (±) tert-butyl (1S,2R,3R,5R)-3-((6-chloropyridazin-3-yl)(methyl)amino)- 2-fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate, (INT-1, 6.2 g) was purified by chiral SFC purification to give the two title compounds as (HETX-6A, tR = 1.05 min, 2.6 g) and (HETX-6B, tR = 2.07 min, 2.6 g). Instrument: SFC-200 (Thar, Waters); Column: AD 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO 2 /MeOH(0.2%Methanol Ammonia) = 80/20; Flow rate: 120 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 4.1 min; Sample solution: 6000 mg dissolved in 110 mL MeOH; Injection volume: 2 mL.

[0868] Step 2. Synthesis of tert-butyl (1S,2R,3R,5R)-2-fluoro-3-(methyl(6-(4-oxo-4,5-dihydrothieno[ 3,2- c]pyridin-2-yl)pyridazin-3-yl)amino)-8-azabicyclo[3.2.1]octa ne-8-carboxylate and tert-butyl

(1R,2S,3S,5S)-2-fluoro-3-(methyl(6-(4-oxo-4,5-dihydrothie no[3,2-c]pyridin-2-yl)pyridazin-3-yl)amino)- 8-azabicyclo[3.2.1]octane-8-carboxylate (PENT-6A & PENT-6B). A mixture of HETX-6A (150 mg, 0.40 mmol), (4-oxo-4,5-dihydrothieno[3,2-c]pyridin-2-yl)boronic acid (B1, 255 mg, 1.31mmol), X-phos-Pd 2nd G (79 mg, 0.10 mmol) and K 3 PO 4 (171 mg, 0.81 mmol) in DMF (10 mL) was degassed with nitrogen and then stirred at 100 °C for 2 h. The mixture was filtered, poured into 50 mL of water and extracted with EtOAc (5 mLX3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , and concentrated. The crude product was purified by silica gel chromatography (2~5% MeOH in DCM) to give the title compound as PENT-6A (110 mg, 0.22 mmol, 55% yield) as a yellow solid. LCMS: m/z 486.1 [M+H] + ; t R = 1.67 min.

[0869] Following the procedure above, but starting with HETX-6B (150 mg, 0.40 mmol), gave the title compound as PENT-6B (120 mg, 61% yield) as a yellow solid. LCMS: m/z 486.1 [M+H] + ; t R = 1.67 min. [0870] Step 3. Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)thieno[3,2-c]pyridin-4(5H)-o ne and 2-(6-(((1R,2R,3S,5S)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)thi eno[3,2-c]pyridin-4(5H)-one (Compound 6A & Compound 6B). A mixture of PENT-6A (100 mg, 0.20 mmol) in HCl/dioxane (10 mL) was stirred at 25 °C for 3 h. The solvent was concentrated, the residue was basified by NH 3 /MeOH, and concentrated. The residue was purified by silica gel chromatography (4% MeOH in DCM) to obtain the title compound as

Compound-6A (47.1 mg, 0.12 mmol, Yield: 59%). 1 H NMR (400 MHz, DMSO-d 6 ) d 8.20 (d, J = 9.6 Hz, 1H), 8.07(s, 1H), 7.59 (d, J = 7.2 Hz, 1H), 7.17 (d, J = 8.0 Hz, 1H), 6.90 (d, J = 7.8 Hz, 1H), 5.23-5.10 (m, 1H), 4.67-4.47 (m, 1H), 4.45 (t, J = 5.2Hz, 1H), 3.52 (s, 3H), 3.39-3.37 (m, 2H), 3.00 (s, 3H), 2.25-2.18 (m, 1H), 1.75-1.51 (m, 2H), 1.70-1.65 (m, 2H), 1.58-1.25 (m, 1H); LCMS: m/z 385.9 [M+H] + ; t R = 1.38 min.

[0871] Following the procedure above, but starting with PENT-6B (110 mg, 0.22 mmol), gave the title compound as Compound-6B (58.3 mg, 0.15 mmol, 66% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 8.20 (d, J = 9.6 Hz, 1H), 8.07(s, 1H), 7.59 (d, J = 7.2 Hz, 1H), 7.17 (d, J = 8.0 Hz, 1H), 6.90 (d, J = 7.8 Hz, 1H), 5.23-5.10 (m, 1H), 4.67-4.47 (m, 1H), 4.45 (t, J = 5.2Hz, 1H), 3.52 (s, 3H), 3.39-3.37 (m, 2H), 3.00 (s, 3H), 2.25-2.18 (m, 1H), 1.75-1.51 (m, 2H), 1.70-1.65 (m, 2H), 1.58-1.25 (m, 1H); LCMS: m/z 385.9 [M+H] + ; t R = 1.38 min.

[0872] The examples in Table 8 were synthesized using a sequence analogous to that used to synthesize Compound 6A and Compound 6B by general Method R.

Table 8. Additional examples prepared by general method G.

Example B10. General Synthesis Method Q: Convergent Suzuki, OMOM Deprotection, ArylChloride Intermediate Chiral Purification

Specific Example of General Method Q, Compounds 8A & 8B: Synthesis of 2-(6-(((1S,5S,6S,7R)-6-fluoro- 3-oxa-9-azabicyclo[3.3.1]nonan-7-yl)(methyl)amino)pyridazin- 3-yl)-5-(1H-pyrazol-3-yl)phenol and 2-(6- (((1R,5R,6R,7S)-6-fluoro-3-oxa-9-azabicyclo[3.3.1]nonan-7-yl )(methyl)amino)pyridazin-3-yl)-5-(1H- pyrazol-3-yl)phenol.

[0873] Step 2: Chiral purification to isolate tert-butyl (1S,5S,6R,7R)-7-((6-chloropyridazin-3- yl)(methyl)amino)-6-fluoro-3-oxa-9-azabicyclo[3.3.1]nonane-9 -carboxylate and tert-butyl (1S,5S,6R,7R)- 7-((6-chloropyridazin-3-yl)(methyl)amino)-6-fluoro-3-oxa-9-a zabicyclo[3.3.1]nonane-9-carboxylate (HETX-8A & HETX-8B). Racemic intermediate, (±) tert-butyl (1S,5S,6R,7R)-7-((6-chloropyridazin-3- yl)(methyl)amino)-6-fluoro-3-oxa-9-azabicyclo[3.3.1]nonane-9 -carboxylate, INT-4, 360 mg, was purified by chiral SFC purification to give the two title compounds as HETX-8A (t R = 0.77 min, 160 mg, >99.5% ee) and HETX-8B (t R = 2.17 min, 110 mg, 98% ee) as white solids. Instrument: SFC-80 (Thar, Waters); Column: AD 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO 2 /MeOH(0.2%Methanol

Ammonia) = 50/50; Flow rate: 80 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 5.0 min; Sample solution: 360 mg; dissolved in 30 mL methanol; Injection volume: 1.9 mL.

[0874] Step 3: Synthesis of tert-butyl (1S,5S,6R,7R)-6-fluoro-7-((6-(2-(methoxymethoxy)-4-(1- (tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)phenyl)pyridazin- 3-yl)(methyl)amino)-3-oxa-9- azabicyclo[3.3.1]nonane-9-carboxylate and of tert-butyl (1S,5S,6R,7R)-6-fluoro-7-((6-(2- (methoxymethoxy)-4-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol- 3-yl)phenyl)pyridazin-3- yl)(methyl)amino)-3-oxa-9-azabicyclo[3.3.1]nonane-9-carboxyl ate (PENT-8A & PENT-8B). A mixture of HETX-8A (80 mg, 0.207 mmol), 4-(3-(methoxymethoxy)-4 -(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (107 mg, 0.259 mmol), K 2 CO 3 (57.2 mg, 0.414 mmol) and Pd(dppf)Cl 2 (22.7 mg, 0.03 mmol) in a solution of dioxane (2 mL) and water (0.5 mL) was degassed and stirred at 100 °C for 2 h under N 2 atmosphere. The reaction mixture was concentrated in vacuum and the residue was purified by silica gel column (20-80% EtOAc/petroleum ether) to give the title compound as PENT-8A (110 mg, 83% yield) as a white solid. LCMS: m/z 639.3 [M+1] + ; t R =1.56 min. [0875] A mixture of HETX-8B (55 mg, 0.143 mmol), 4-(3-(methoxymethoxy)-4 -(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl)-1-(tetrahydro-2H-pyran-2-yl)-1H-py razole (74 mg, 0.178 mmol), K 2 CO 3 (39.5 mg, 0.286 mmol) and Pd(dppf)Cl 2 (15.7 mg, 0.02 mmol) in a solution of dioxane (2 mL) and water (0.5 mL) was degassed and stirred at 100 °C for 2 h under N 2 atmosphere. The reaction mixture was concentrated in vacuum and the residue was purified by silica gel column (20-80% EtOAc/petroleum ether) to give the title compound as PENT-8B (65 mg, 71% yield) as a white solid. LCMS: m/z 639.3 [M+1] + ; t R =1.56 min.

[0876] Step 4: Synthesis of 2-(6-(((1S,5S,6S,7R)-6-fluoro-3-oxa-9-azabicyclo[3.3.1]nonan -7- yl)(methyl)amino)pyridazin-3-yl)-5-(1H-pyrazol-3-yl)phenol and 2-(6-(((1R,5R,6R,7S)-6-fluoro-3-oxa-9- azabicyclo[3.3.1]nonan-7-yl)(methyl)amino)pyridazin-3-yl)-5- (1H-pyrazol-3-yl)phenol (Compound 8A & Compound 8B). To a flask with PENT-8A (110 mg, 0.172 mmol) in DCM (1 mL) wa added 4M HCl in dioxane (2 mL). The reaction mixture was stirred at 20°C for 1 h, concentrated in vacuo, basified 7M ammonia in MeOH and purified by Prep-TLC (DCM: 7M ammonia MeOH 30:1) to give the title compound as Compound 8A (21 mg, 30 % yield) as a yellow solid. 1 H NMR (400 MHz, Methanol-d 4 ) d 8.38– 8.26 (m, 1H), 8.04 (s, 2H), 7.76 (d, J = 8.3 Hz, 1H), 7.63 (d, J = 9.8 Hz, 1H), 7.28 (dd, J = 8.2, 1.7 Hz, 1H), 7.23 (d, J = 1.6 Hz, 1H), 6.26– 6.09 (m, 1H), 5.47– 5.32 (m, 1H), 4.32– 4.03 (m, 4H), 4.02– 3.93 (m, 1H), 3.84– 3.77 (m, 1H), 3.20 (s, 3H), 2.97– 2.84 (m, 1H), 2.33– 2.23 (m, 1H). LCMS: m/z 411.2 [M+1] + ; t R =1.47 min.

[0877] Following the procedure above, but starting with PENT-8B (65 mg, 0.102 mmol) gave the title compound as Compound 8B (25 mg, 60% yield) as a yellow solid. 1 H NMR (400 MHz, Methanol-d 4 ) d 8.12 (d, J = 9.9 Hz, 1H), 8.02 (s, 2H), 7.76 (d, J = 8.8 Hz, 1H), 7.32 (d, J = 9.9 Hz, 1H), 7.20 (dd, J = 6.1, 1.7 Hz, 2H), 6.11– 5.87 (m, 1H), 5.15– 4.94 (m, 1H), 4.22– 3.74 (m, 4H), 3.24– 3.04 (m, 4H), 2.75– 2.60 (m, 1H), 1.95– 1.83 (m, 1H). LCMS: m/z 411.2[M+1] + ; t R =1.47 min.

[0878] The examples in Table 9A and Table 9B were synthesized using a sequence analogous to that used to synthesize Compound 8A and Compound 8B by general Method Q.

Table 9A. Intermediates Prepared by General Method Q.

Table 9B. Additional Examples Prepared by General Method Q.

[0879] Example B11. General Synthesis Method S: Convergent Suzuki, OMe Deprotection, Penultimate Chiral Purification.

Specific Example of General Synthesis Method S, Compound 9A & 9B: Synthesis of 6-(6- (((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)(meth yl)amino)pyridazin-3-yl)-5-hydroxy-N,N- dimethylbenzofuran-2-carboxamide and 6-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-5-hydroxy-N,N-dimethylbenzo furan-2-carboxamide.

[0880] Step 1: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-(2-(dimethylcarbamoyl)-5- methoxybenzofuran-6-yl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate. To a solution of 5-methoxy-N,N-dimethyl-6-(4,4,5,5–tetramethyl–1,3,2–di oxaborolan–2–yl)-1- benzofuran-2-carboxamide (B4, 1.40 g, 4.06 mmol, 1.00 equiv) and (±) tert-butyl (1S,2R,3R,5R)-3-[(6- chloropyridazin-3-yl)(methyl)amino]-2-fluoro-8-azabicyclo[3. 2.1]octane-8-carboxylate (1.203 mg, 3.24 mmol, 0.80 equiv) in dioxane (15 mL) and water (1.5 mL) were added Pd(dppf)Cl 2 (298 mg, 0.406 mmol, 0.10 equiv) and K 3 PO 4 (2.58 g, 12.17 mmol, 3.00 equiv). After stirring for 12 h at 100 °C under nitrogen atmosphere, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluting with ethyl acetate/ petroleum ether (2:1) to afford the crude product as a white solid. The crude product was further purified by reverse phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 20% to 60% gradient in 30 min; detector, UV 254 nm to afford the title compound (600 mg, 33% yield).

[0881] Step 2: Chiral purification to isolate tert-butyl (1S,2R,3R,5R)-3-((6-(2-(dimethylcarbamoyl)-5- methoxybenzofuran-6-yl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate and tert-butyl (1R,2S,3S,5S)-3-((6-(2-(dimethylcarbamoyl)-5-methoxybenzofur an-6- yl)pyridazin-3-yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1 ]octane-8-carboxylate (PENT-9A & PENT- 9B). Racemic intermediate, (±) tert-butyl (1S,2R,3R,5R)-3-((6-(2-(dimethylcarbamoyl)-5- methoxybenzofuran-6-yl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8-carboxylate (600 mg), was purified by chiral SFC purification to give the two title compounds as PENT-9A (t R = 3.12 min, 120 mg) and PENT-9B (t R = 4.91 min, 140 mg). Column: CHIRALPAK IA, 2*25cm,5 Pm; Mobile Phase A: Hex (10mM NH3-MeOH), Mobile Phase B:IPA; Flow rate: 20 mL/min; Gradient: 30 A to 30 B in 50 min; wavelength: 254/220 nm.

[0882] Step 3: 6-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl) (methyl)amino)pyridazin-3- yl)-5-hydroxy-N,N-dimethylbenzofuran-2-carboxamide and 6-(6-(((1R,2R,3S,5S)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)-5- hydroxy-N,N-dimethylbenzofuran-2- carboxamide (Compound 9A & Compound 9B). To a stirred solution of PENT-9A (120 mg, 0.217 mmol, 1.00 equiv) in DCM (4.0 mL) was added BBr 3 (163 mg, 0.650 mmol, 3.00 equiv) dropwise in portions at 0 °C. Then the reaction mixture was warmed to 25 °C and stirred for 12 h. The reaction was then quenched by the slow addition of 10 mL of water/ice and the pH value of the mixture was adjusted to 9 with the saturated NaHCO 3 solution. Then the resulting solution was extracted with dichloromethane (15 mLx3). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by reverse phase flash chromatography with the following conditions: Column, C18 silica gel; mobile phase, MeOH in water, 10% to 0% gradient in 30 min; detector, UV 254 nm to afford the title compound as Compound 9A (20 mg, 21% yield) as a yellow solid. 1 H NMR (300 MHz, Methanol-d4) d 8.23~8.20 (d, J = 9.9 Hz, 1H), 8.04 (s, 1H), 7.36 ~ 7.29 (m, 2H), 7.20 (s, 1H), 5.40 ~ 5.19 (m, 1H), 4.68 ~ 4.58 (m, 1H), 3.69 (s, 2H), 3.39 (s, 3H), 3.19 ~ 3.10 (m, 6H), 2.48~2.34 (td, J = 12.8, 3.2 Hz, 1H), 2.08~1.86 (m, 4H), 1.74 ~ 1.63 (m, 1H). LCMS: m/z 440.4 [M+H] + .

[0883] Following the procedure above, but starting with PENT-9B (140 mg, 0.253 mmol) gave the title compound as Compound 9B (18 mg, 16% overall yield) as a yellow solid. : 1 H NMR (300 MHz, Methanol- d4) d 8.23 ~ 8.20 (d, J = 10.0 Hz, 1H), 8.03 (s, 1H), 7.36 ~ 7.28 (m, 2H), 7.19 (s, 1H), 5.39 ~ 5.20 (m, 1H), 4.67~ 4.62 (m, 1H), 3.69 (s, 2H), 3.39 (s, 3H), 3.21 ~ 3.09 (m, 6H), 2.41 (td, J = 12.9, 3.1 Hz, 1H), 2.05 ~ 1.86 (m, 4H), 1.73 ~ 1.63 (m, 1H). LCMS: m/z 440.4 [M+H] + .

[0884] The examples in Table 10 were synthesized using a sequence analogous to that used to synthesize Compound 9A and Compound 9B by general Method S.

Table 10. Examples Prepared by General Method S.

Example B12. General Synthesis Method N: Convergent Suzuki with Chiral Halo-Pyridazines, Methyl Ether Deprotection

Specific Example of General Method N, Compound 7A & 7B: Synthesis of 7-(6-(((1S,2S,3R,5R)-2- fluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin -3-yl)isoquinolin-6-ol and 7-(6- (((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)(meth yl)amino)pyridazin-3-yl)isoquinolin-6-ol.

[0885] Step 1a: Synthesis of 7-bromo-6-methoxyisoquinoline.2,2-dimethoxyethanamine (3.7 g, 34.877mmol) and Na 2 SO 4 (3.3 g, 23.251 mmol) were added to a stirred solution of 3-bromo-4- methoxybenzaldehyde (5 g, 23.251 mmol) in 50 mL of toluene. The reaction mixture was heated to reflux for 6 h using a Dean-stark apparatus. Solvent and excess reagents were distilled off. The crude product was dissolved in THF (50 mL). ClCOOCH 3 (2.2 g, 23.251 mmol) was added dropwise at 0 °C. After stirring for 5 min, P(OEt) 3 (4.6 g, 27.901 mmol) was added dropwise. The mixture was stirred for 18 h at room temperature. Then the solvents were distilled off. Excess reagents were removed by repeated addition of toluene and evaporation of the solvents. TiCl 4 (17.6 g, 93.004 mmol) and CHCl 3 (25 mL) were added. The mixture was heated to reflux for 48 h. The mixture was poured on ice and the pH was adjusted to 9 by using aqueous ammonia. The resulting mixture was extracted with EtOAc followed by removal off the solvents. The residue was purified by flash silica gel column chromatography (0-70% EtOAc/petroleum ether) to give 1.3 g 7- bromo-6-methoxyisoquinoline (16% yield) as a white solid. LCMS: m/z 240.0 [M+H] + ; t R = 1.38 min.

[0886] Step 1b: Synthesis of 6-methoxy-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso quinoline (B2). A mixture of 7-bromo-6-methoxyisoquinoline (200 mg, 0.844 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (321 mg, 1.266 mmol), Pd(dppf)Cl 2 (124 mg, 0.169 mmol) and KOAc (166 mg, 1.69 mmol) in 1,4-dioxane (7 mL) was degassed and stirred at 105 o C for 8 h under N 2 protection. The reaction was cooled to room temperature and used in the next step without any work-up. LCMS: m/z 286.0 [M+H] + ; t R = 1.80 min.

[0887] Step 2: Synthesis of tert-butyl (1S,2R,3R,5R)-2-fluoro-3-(methyl(6-(4-oxo-4,5-dihydrothieno[ 3,2- c]pyridin-2-yl)pyridazin-3-yl)amino)-8-azabicyclo[3.2.1]octa ne-8-carboxylate and tert-butyl

(1R,2S,3S,5S)-2-fluoro-3-(methyl(6-(4-oxo-4,5-dihydrothie no[3,2-c]pyridin-2-yl)pyridazin-3-yl)amino)- 8-azabicyclo[3.2.1]octane-8-carboxylate (PENT-7A & PENT-7B). To the crude solution of 6-methoxy-7- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoquinoline (B2, 240 mg, 0.844 mmol) in 1,4-dioxane (7 mL) and HETX-6A (180 mg, 0.485 mmol) was added Pd(dppf)Cl 2 (100 mg, 0.136 mmol), K 2 CO 3 (200 mg, 1.45 mmol) and water (1.5 mL). The mixture was degassed and heated at 100 o C for 2 h under nitrogen

atmoshphere. After cooling to room temperature, the mixture was concentrated and purified by silica gel chromatography (0~100% EtOAc/petroleum ether) to give the title compound as PENT-7A (180 mg, 69% yield for 2 steps) as a brown oil. LCMS: m/z 494.3 [M+H] + ; t R = 1.90 min.

[0888] Following the procedure above, but starting with HETX-6B (180 mg, 0.485 mmol), gave the title compound as PENT-7B (200 mg, 75% yield for 2 steps) as a brown oil. LCMS: m/z 494.3 [M+H] + ; t R = 1.92 min.

[0889] Step 3: Synthesis of 7-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)isoquinolin-6-ol and 7-(6-(((1S,2S,3R,5R)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)iso quinolin-6-ol (Compound 7A &

Compound 7B). BBr 3 (5 mL, 1N in CH 2 Cl 2 ) was added to the mixture of PENT-7A (180 mg, 0.365 mmol) in DCM (2 mL). The mixture was stirred at room temperature for 8 h, The reaction was quenched with MeOH (2 mL) and concentrated. The residue was treated with NH 3 /MeOH (7.0 M, 3 mL) and purified by silica gel chromatography (0~100% MeOH/CH 2 Cl 2 ) to give the title compound as Compound 7A (20 mg, 14.5% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) d 9.18 (s, 1H), 8.72 (s, 1H), 8.40 (d, J = 9.9 Hz, 1H), 8.34 (d, J = 5.8 Hz, 1H), 7.60 (d, J = 5.8 Hz, 1H), 7.44 (d, J = 9.9 Hz, 1H), 7.28 (s, 1H), 5.27– 4.99 (m, 1H), 4.65 (d, J = 52.0 Hz, 1H), 3.61– 3.48 (m, 2H), 3.07 (s, 3H), 2.30– 2.20 (m, 1H), 1.90– 1.49 (m, 5H). LCMS: m/z 380.3; [M+H] + ; t R = 1.17 min.

[0890] Following the procedure above, but with PENT-7B, (200 mg, 0.405 mmol) and BBr 3 (7 mL, 1N in DCM) gave the title compound as Compound 7B (30 mg, 19% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 13.62 (s, 1H), 9.19 (s, 1H), 8.74 (s, 1H), 8.41 (d, J = 9.8 Hz, 1H), 8.35 (d, J = 5.8 Hz, 1H), 7.62 (d, J = 5.8 Hz, 1H), 7.48 (d, J = 9.9 Hz, 1H), 7.31 (s, 1H), 5.27– 5.03 (m, 1H), 4.77 (d, J = 51.3 Hz, 1H), 3.78 – 3.65 (m, 2H), 3.08 (s, 3H), 2.41– 2.29 (m, 1H), 1.95– 1.62 (m, 5H). LCMS: m/z 380.3; [M+H] + ; t R = 1.60 min.

[0891] The examples in Table 11 were synthesized using a sequence analogous to that used to synthesize Compound 7A and Compound 7B by general Method N.

Table 11. Additional Examples Prepared by General Method N.

Example B13. General Synthesis Method X: N-Methylation of Final Products

Specific Example of General Method X, Compounds 92A & 92B: Synthesis of azetidin-1-yl(6-(6- (cyclopropyl((1S,2S,3R,5R)-2-fluoro-8-methyl-8-azabicyclo[3. 2.1]octan-3-yl)amino)pyridazin-3-yl)-5- hydroxybenzofuran-2-yl)methanone and azetidin-1-yl(6-(6-(cyclopropyl((1R,2R,3S,5S)-2-fluoro-8- methyl-8-azabicyclo[3.2.1]octan-3-yl)amino)pyridazin-3-yl)-5 -hydroxybenzofuran-2-yl)methanone

[0892] Step 1: Synthesis of azetidin-1-yl(6-(6-(cyclopropyl((1S,2S,3R,5R)-2-fluoro-8-met hyl-8- azabicyclo[3.2.1]octan-3-yl)amino)pyridazin-3-yl)-5-hydroxyb enzofuran-2-yl)methanone and azetidin- 1-yl(6-(6-(cyclopropyl((1R,2R,3S,5S)-2-fluoro-8-methyl-8-aza bicyclo[3.2.1]octan-3-yl)amino)pyridazin- 3-yl)-5-hydroxybenzofuran-2-yl)methanone. Into a 50-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed Compound 57A (150 mg, 0.314 mmol, 1.0 equiv), DCM (3 mL), and HCHO (18.9 mg, 0.628 mmol, 2.0 equiv), NaBH 3 CN (39.5 mg, 0.628 mmol, 2.0 equiv). The resulting solution was stirred for 3 h at room temperature. The reaction was then quenched by the addition of 20 mL of water. The resulting solution was extracted with 3x20 mL of dichloromethane and the organic layers combined. The resulting mixture was washed with 1x20 ml of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was purified by Flash- Prep-HPLC with the following conditions: Column, XB-C18; mobile phase, 0.1%NH 4 HCO 3 /H2O: ACN=10- 60%; Detector, 254 nm. This resulted in the title compound as Compound 92A (28 mg, 18.1% yield) as a yellow solid. 1 H-NMR (300 MHz, DMSO-d6) d 13.12 (s, 1H), 8.37 (d, J = 9.9 Hz, 1H), 8.22 (s, 1H), 7.68 (d, J = 9.8 Hz, 1H), 7.34 (s, 1H), 7.21 (s, 1H), 4.81 (s, 1H), 4.59 (s, 3H), 4.08 (s, 2H), 3.27 (s, 2H), 2.66 (t, J = 12.5 Hz, 1H), 2.41– 2.30 (m, 3H), 2.23 (s, 3H), 2.01 (s, 2H), 1.82– 1.49 (m, 3H), 1.02 (s, 3H), 0.44 (s, 1H).

LCMS: m/z 492.4 [M+H] + .

[0893] Following the procedure above with Compound 57B (150 mg, 0.314 mmol), gave the title compound as Compound 92B (26 mg, 16.8% yield) as a yellow solid. 1 H-NMR (300 MHz, DMSO-d6) d 13.12 (s, 1H), 8.38 (d, J = 9.9 Hz, 1H), 8.22 (s, 1H), 7.68 (d, J = 9.8 Hz, 1H), 7.34 (d, J = 0.8 Hz, 1H), 7.21 (s, 1H), 4.81 (s, 1H), 4.67– 4.53 (m, 3H), 4.09 (t, J = 7.7 Hz, 2H), 3.30 (d, J = 12.9 Hz, 2H), 2.66 (t, J = 12.4 Hz, 1H), 2.36 (p, J = 7.5 Hz, 3H), 2.23 (s, 3H), 2.01 (s, 2H), 1.81– 1.52 (m, 3H), 1.08– 0.97 (m, 3H), 0.45 (d, J = 9.1 Hz, 1H). (ES, m/z) ). LCMS: m/z 492.5 [M+H] + .

[0894] The examples in Table 12 were synthesized using a sequence analogous to that used to synthesize Compound 57A and Compound 57B by general Method X.

Table 12. Additional Examples Prepared by General Method X

Example B14. General Procedure Y: Convergent Suzuki, OSEM protecting group, Penultimate Separation

Selected Example of General Method Y, Compounds 121A & 121B: Synthesis of 7-(6-(((1S,2S,3R,5R)-2- fluoro-9-azabicyclo[3.3.1]nonan-3-yl)(methyl)amino)pyridazin -3-yl)isoquinolin-6-ol and 7-(6- (((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3-yl)(meth yl)amino)pyridazin-3-yl)isoquinolin-6-ol.

[0895] Step 1: Synthesis of 7-bromo-6-((2-(trimethylsilyl)ethoxy)methoxy)isoquinoline. NaH (134 mg, 3.35 mmol, 60wt% in mineral oil) was added to a stirred solution of 7-bromoisoquinolin-6-ol (500 mg, 2.23 mmol) in DMF (20 mL) and THF (45 mL) at rt. After stirring at rt for 30 min, SEMCl (373 mg, 2.23 mmol) was added. The mixture was stirred at room temperature for 1 h, quenched with NH 4 Cl aqueous solution (20 mL), extracted with EtOAc (60 mL X 3). The combined organic solvents were dried over anhydrous Na 2 SO 4 , concentrated, and purified by silica gel chromatography (0-20% EtOAc/petroleum ether) to give 7-bromo-6- ((2-(trimethylsilyl)ethoxy)methoxy)isoquinoline as colorless oil (619 mg, 84% yield), LCMS: LCMS: m/z 354.0 [M+H] + ; t R = 2.21 min.

Step 2: Synthesis of (±) (1S,2R,3R,5R)-tert-butyl 2-fluoro-3-(methyl(6-(6-((2- (trimethylsilyl)ethoxy)methoxy)isoquinolin-7-yl)pyridazin-3- yl)amino)-9-azabicyclo[3.3.1]nonane-9- carboxylate. A mixture of 7-bromo-6-((2-(trimethylsilyl)ethoxy)methoxy)isoquinoline (240 mg, 0.678 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (258 mg 1.016 mmol), Pd(dppf)Cl 2 (99 mg, 0.135 mmol) and KOAc (133 mg, 1.35 mmol) in 4 mL of dioxane was degassed and stirred at 100 o C for 2 h. The mixture was cooled to room temerature, (±) (1S,2R,3R,5R)-tert-butyl 3-((6-chloropyridazin-3-yl)(methyl)

[0896] amino)-2-fluoro-9-azabicyclo[3.3.1]nonane-9-carboxylate (INT-3, 182 mg, 0.474 mmol), Pd(dppf)Cl 2 (69 mg, 0.0948 mmol), K 2 CO 3 (131 mg, 0.948 mmol), 6 mL of dioxane and 1 mL of H 2 O were added. The mixture was degassed and stirred at 100 o C for 2 h, concentrated and purified by silica gel column (100% EtOAc/petroleum ether) to give the title compound as a white solid (181 mg, 51% yield). LCMS: m/z 624.3 [M+H] + ; t R = 2.35 min.

[0897] Step 3: Synthesis of (±) 7-(6-(((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3- yl)(methyl)amino)pyridazin-3-yl)isoquinolin-6-ol. To the mixture of (±) (1S,2R,3R,5R)-tert-butyl 2-fluoro- 3-(methyl(6-(6-((2-(trimethylsilyl)ethoxy)methoxy)isoquinoli n-7-yl)pyridazin-3-yl)amino)-9- azabicyclo[3.3.1]nonane-9-carboxylate (201 mg, 0.323 mmol) in CH 2 Cl 2 (2 mL) and MeOH (1 mL), was added 4N HCl in dioxane (5 mL). The mixture was stirred at 25 o C for 2 h. The mixture was concentrated to dryness, then dissolved in water and saturated K 2 CO 3 aqueous solution was added till pH to 8-9. The mixture was extracted with CH 2 Cl 2 /MeOH 10:1 (v/v, 100 mL X 3). The combined organic solvents were concentrated and purified by silica gel column (15% MeOH/ CH 2 Cl 2 ) to give the title compound as a yellow solid (95 mg, 75% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 13.60 (s, 1H), 9.19 (s, 1H), 8.74 (s, 1H), 8.41 (d, J = 9.9 Hz, 1H), 8.35 (d, J = 5.8 Hz, 1H), 7.62 (d, J = 5.8 Hz, 1H), 7.44 (d, J = 9.9 Hz, 1H), 7.31 (s, 1H), 5.88– 5.69 (m, 1H), 4.77 (d, J = 50.8 Hz, 1H), 3.30– 3.20 (m, 2H), 3.08 (d, J = 1.5 Hz, 3H), 2.46– 2.36 (m, 1H), 2.00– 1.56 (m, 7H). LCMS: m/z 394.1 [M+H] + ; t R = 1.43 min.

[0898] Step 4: Chiral purification to isolate 7-(6-(((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3- yl)(methyl)amino)pyridazin-3-yl)isoquinolin-6-ol and 7-(6-(((1S,2S,3R,5R)-2-fluoro-9- azabicyclo[3.3.1]nonan-3-yl)(methyl)amino)pyridazin-3-yl)iso quinolin-6-ol (Compound 121A & Compound 121B). Racemic intermediate, (±) 7-(6-(((1S,2S,3R,5R)-2-fluoro-9-azabicyclo[3.3.1]nonan-3- yl)(methyl)amino)pyridazin-3-yl)isoquinolin-6-ol, 95 mg, was purified by chiral chromatography under the following conditions to provide the title compounds as Compound 121A (27 mg, t R = 2.42 min) and

Compound 121B (19 mg, t R = 3.77 min) as yellow solids. Instrument: SFC-150 (Waters);

Column: AD 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO 2 /(

MeOH/ACN(0.2% Methanol Ammonia)=1:1 ) = 40/60; Flow rate: 120 g/min; Back pressure: 100 bar;

Detection wavelength: 214 nm; Cycle time: 4 min; Sample solution: 60mg dissolved in 20 mL methanol; Injection volume: 1.9 mL Compound 121A: 1 H NMR (400 MHz, DMSO-d6) d 13.60 (s, 1H), 9.19 (s, 1H), 8.74 (s, 1H), 8.41 (d, J = 9.9 Hz, 1H), 8.35 (d, J = 5.8 Hz, 1H), 7.62 (d, J = 5.8 Hz, 1H), 7.44 (d, J = 9.9 Hz, 1H), 7.31 (s, 1H), 5.88– 5.69 (m, 1H), 4.77 (d, J = 50.8 Hz, 1H), 3.30– 3.20 (m, 2H), 3.08 (d, J = 1.5 Hz, 3H), 2.46– 2.36 (m, 1H), 2.00– 1.56 (m, 7H). LCMS: m/z 394.1 [M+H] + ; t R = 1.43 min. Compound 121B: 1 H NMR (400 MHz, DMSO-d6) d 13.60 (s, 1H), 9.19 (s, 1H), 8.74 (s, 1H), 8.41 (d, J = 9.9 Hz, 1H), 8.35 (d, J = 5.8 Hz, 1H), 7.62 (d, J = 5.8 Hz, 1H), 7.44 (d, J = 9.9 Hz, 1H), 7.31 (s, 1H), 5.88– 5.69 (m, 1H), 4.77 (d, J = 50.8 Hz, 1H), 3.30– 3.20 (m, 2H), 3.08 (d, J = 1.5 Hz, 3H), 2.46– 2.36 (m, 1H), 2.00– 1.56 (m, 7H). LCMS: m/z 394.1 [M+H] + ; t R = 1.60 min.

[0899] The examples in Table 13 were synthesized using a sequence analogous to that used to synthesize Compound 121A and Compound 121B by general Method Y.

Table 13. Additional Examples Prepared by General Method Y.

Example B15. Compound 10A & 10B: Synthesis of 5-ethynyl-2-(6-(((1S,2S,3R,5R)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)phe nol and 5-ethynyl-2-(6-(((1R,2R,3S,5S)-2- fluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin -3-yl)phenol.

[0900] Step 1: Synthesis of (±) tert-butyl (1S,2R,3R,5R)-3-((6-(4-chloro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate. A mixture of (±) tert-butyl (1R,2S,3S,5S)-3-((6-chloropyridazin-3-yl)(methyl)amino)-2-fl uoro- 8-azabicyclo[3.2.1]octane-8-carboxylate (INT-1, 2.4 g, 6.47 mmol), (4-chloro-2- (methoxymethoxy)phenyl)boronic acid (2.80 g, 12.9 mmol), Pd(dppf)Cl 2 (1.18 g, 1.62 mmol) and K 2 CO 3 (1.79 g, 12.94 mmol) in dioxane (30 mL) and water (10 mL) was degassed and stirred at 100 o C under nitrogen for 3 h. After cooling to the ambient temperature, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (30 mLx2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified with silica gel chromatography (40% of ethyl acetate in petroleum ether) to give the title compound (2.0 g, 61% yield) as a white solid. LCMS: m/z 507.2 [M+H] + ; t R = 2.03 min.

[0901] Step 2: Chiral purification to isolate tert-butyl (1S,2R,3R,5R)-3-((6-(4-chloro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate and tert-butyl (1R,2S,3S,5S)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyrid azin-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate (HETX-10A & HETX-10B).

Racemic intermediate, (±) tert-butyl (1S,2R,3R,5R)-3-((6-(4-chloro-2-(methoxymethoxy)phenyl)pyrid azin-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate, 3 g, was purified by chiral SFC purification to give the two title compounds as HETX-10A (t R = 1.55 min, 1.4 g) and HETX-10B (t R = 2.30 min, 1.4 mg). Instrument: SFC-150 (Thar, Waters); Column: SC 20*250mm, 10um (Regis); Column temperature: 35 ºC; Mobile phase: CO 2 /MeOH(0.2% methanol ammonia) = 60/40; Flow rate: 100 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 3 min; Sample solution: 3000mg dissolved in 100 ml nethanol; Injection volume: 1 mL.

[0902] Step 3: Synthesis of tert-butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(2-(methoxymethoxy)-4- ((triisopropylsilyl)ethynyl)phenyl)pyridazin-3-yl)(methyl)am ino)-8-azabicyclo[3.2.1]octane-8- carboxylate and tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(2-(methoxymethoxy)-4- ((triisopropylsilyl)ethynyl)phenyl)pyridazin-3-yl)(methyl)am ino)-8-azabicyclo[3.2.1]octane-8- carboxylate (HET-10A & HET-10B). To a solution of HETX-10A (200 mg, 0.395 mmol) in ACN (5 mL) was added ethynyltriisopropylsilane (360 mg, 1.98 mmol), (MeCN) 2 PdCl 2 (21 mg, 0.08 mmol), X-Phos (76 mg, 0.16 mmol) and Cs2CO3 (258 mg, 0.79 mmol). The reaction was stirred at 95 o C in sealed tube for 3 h under nitrogen atmoshphere. The mixture was concentrated in vacuo, and the resdiue was purified by silica gel chromatography (0-25% EtOAc/petroleum ether) to give the title compound as HET-10A (220 mg, 86% yield). LCMS: m/z 653.4 [M+H] + ; t R = 3.46 min.

[0903] Following the procedure above, but starting with HETX-10B (150 mg, 0.296 mmol), gave the title compound as HET-10B (170 mg, 88% yield). LCMS: m/z 653.0 [M+H] + ; t R = 3.77 min.

[0904] Step 4: Synthesis of tert-butyl (1S,2R,3R,5R)-3-((6-(4-ethynyl-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-2-fluor o-8-azabicyclo[3.2.1]octane-8- carboxylate and tert-butyl (1R,2S,3S,5S)-3-((6-(4-ethynyl-2-(methoxymethoxy)phenyl)pyri dazin-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate (PENT-10A & PENT-10B). To a stirred solution of HET-10A (170 mg, 0.261 mmol) in THF (5 mL) was added TBAF (2 mL, 1 N in THF). The reaction was stirred at 25 o C for 2 h, and diluted with H 2 O (20mL), and the aqueous phase was extracted with EtOAc (20 mLx3). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na 2 SO 4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0-40%

EtOAc/petroleum ether) to give the title compound as PENT-10A (80 mg, 62% yield). LCMS: m/z 497.3 [M+H] + ; t R = 2.06 min.

[0905] Following the procedure above, but starting with HET-10B (170 mg, 0.296 mmol), gave the title compound as PENT-10B (30 mg, 23% yield). LCMS: m/z 497.3 [M+H] + ; t R = 2.79 min. [0906] Step 5: Synthesis of 5-ethynyl-2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]o ctan-3- yl)(methyl)amino)pyridazin-3-yl)phenol and 5-ethynyl-2-(6-(((1R,2R,3S,5S)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)phe nol (Compound 10A & Compound 10B). To the mixture of PENT-10A (80 mg, 0.04 mmol) in CH 2 Cl 2 (10 mL) was added HCl (4N in dioxane, 5 mL). The mixture was stirred at room temperature for 2 h and concentrated to dryness. The residue was dissolved in MeOH and 7N NH 3 /MeOH was added till pH to 8-9. The mixture was concentrated and purified by silica gel chromatography (0-10% MeOH/dichloromethane) to afford the title compound as Compound 10A (31 mg, 54% yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 13.58 (s, 1H), 8.24 (d, J = 10.0 Hz, 1H), 7.90 (d, J = 8.5 Hz, 1H), 7.38 (d, J = 9.5 Hz, 1H), 7.04– 7.01 (m, 2H), 5.09– 4.99 (m, 1H), 4.70– 4.56 (m, 1H), 4.25 (s, 1H), 3.52 (s, 2H), 3.04 (d, J = 1.2 Hz, 3H), 2.27– 2.20 (m, 1H), 1.81– 7.75 (m, 2H), 1.73– 1.62 (m, 2H), 1.57– 1.51 (m, 1H). LCMS: m/z 353.2 [M+H] + , t R = 1.78 min.

[0907] Following the procedure above, but starting with PENT-10B (60 mg, 0.33 mmol) gave the title compound as Compound 10B (30 mg, 70% overall yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 13.57 (s, 1H), 8.24 (d, J = 10.0 Hz, 1H), 7.90 (d, J = 8.5 Hz, 1H), 7.39 (d, J = 10.2 Hz, 1H), 7.02 (d, J = 7.5 Hz, 2H), 5.10– 5.0 (m, 1H), 4.66 (d, J = 52.0 Hz, 1H), 4.26 (s, 1H), 3.57 (s, 2H), 3.04 (d, J = 1.5 Hz, 3H), 2.29– 2.21 (m, 1H), 1.88– 1.64 (m, 4H), 1.61– 1.52 (m, 1H). LCMS: m/z 353.2 [M+H] + , t R = 1.79 min. Example 119B: Synthesis of 2-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)phenol or 2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)phenol.

[0908] Step 1: Synthesis of tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-8-azabi cyclo [3.2.1]octane-8-carboxylate or of tert-butyl (1S,2R,3R,5R)-2-fluoro-3-((6-(2-(methoxymethoxy)phenyl)pyrid azin-3-yl)(methyl)amino)-8- azabicyclo [3.2.1]octane-8-carboxylate. To a flask with HETX-10B (100 mg, 0.197 mmol) and oxetan-3- ylboronic acid (60 mg, 0.592 mmol) in a mixture solution of dioxane (2 mL) and water (1 mL), K 3 PO 4 (84 mg, 0.394 mmol) and XPhos Pd G2 (23 mg, 0.03 mmol) were added. The reaction mixture was stirred at 100 °C for 2 h under N 2 atmosphere, concentrated in vacuum and the residue was purified by silica gel chromatography (5-25% of ethyl acetate in petroleum ether) to give the title compound as PENT-119B (67 mg, 72 % yield) as a white solid. LCMS: m/z 473.3[M+1] + ; t R =1.59 min.

[0909] Step 2: Synthesis of 2-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)phenol or 2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)phenol (Compound 119B). To a flask with PENT-119B in DCM (1 mL) was added TFA (1 mL) at 0 °C. The reaction mixture was stirred at 20 °C for 30 min, concentrated under vacuum, then basified with 7M ammonia in MeOH. The mixture was concentrated an purified by Prep-TLC (DCM: 7M ammonia MeOH 20:1, v/v) to the title compound as Compound 119B (25.2 mg, 36.2 % yield) as a yellow solid. 1 H NMR (500 MHz, DMSO-d 6 ) d 13.44 (s, 1H), 8.24 (s, 1H), 7.89 (s, 1H), 7.33 (d, J = 59.8 Hz, 2H), 6.94 (s, 2H), 5.37– 4.90 (m, 1H), 4.83– 4.46 (m, 1H), 3.62– 3.47 (m, 2H), 3.04 (s, 3H), 2.33– 2.12 (m, 1H), 2.02– 1.53 (m, 5H). LCMS: m/z 329.2 [M+1] + ; t R = 0.845 min. Compound 120A & 120B: Synthesis of 5-(difluoromethyl)-2-(6-(((1S,2S,3R,5R)-2-fluoro-8- azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)phe nol and of 5-(difluoromethyl)-2-(6- (((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)(meth yl)amino)pyridazin-3-yl)phenol.

[0910] Step 1. Synthesis of (±) (1R,2S,3S,5S)-tert-butyl 2-fluoro-3-((6-(4-formyl-2-methoxyphenyl) pyridazin-3-yl)(methyl)amino)-8-azabicyclo[3.2.1]octane-8-ca rboxylate.4-formyl-2- methoxyphenylboronic acid (480 mg, 2.67 mmol), Pd(dppf)Cl 2 (178 mg, 0.243 mmol) and K 2 CO 3 (670 mg, 4.86 mmol) were added to a mixture of (±) (1R,2S,3S,5S)-tert-butyl 3-((6-chloropyridazin-3- yl)(methyl)amino)-2-fluoro -8-azabicyclo[3.2.1]octane-8-carboxylate (INT-1, 900 mg, 2.43 mmol) in 8 mL of dioxane and 1 mL of H 2 O. The resulting mixture was stirred at 110 °C for 2 h under N 2 . After cooling to room temperature, the mixture was poured into water (60 mL) and extracted with EtOAc (30 mL x 3). The combined extracts were washed with brine (30 mL x 3), dried over anhydrous Na 2 SO 4 , filtered, and concentrated. The residue was purified by silica gel chromatography (0-50% EtOAc/petroleum ether) to give the title compound (800 mg, 70.05%) as a yellow solid. LCMS: m/z 471.3 [M+H] + ; t R = 1.29 min.

[0911] Step 2: Synthesis of (±) (1S,2R,3R,5R)-tert-butyl 3-((6-(4-(difluoromethyl)-2-methoxyphenyl) pyridazin-3-yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]oc tane-8-carboxylate. Diethylaminosulphur trifluoride (4.1 g, 25.5 mmol) was added slowly to a solution of (±) (1R,2S,3S,5S)-tert-butyl 2-fluoro-3-((6-(4- formyl-2-methoxyphenyl)pyridazin-3- yl)(methyl)amino)-8-azabicyclo[3.2.1]octane-8-carboxylate (800 mg, 1.70 mmol) in dry DCM (20 mL) at 0 °C. The resulting mixture was stirred at room temperature for 24 h, then was poured into ice water (100 mL) and extracted with EtOAc (30 mL x 3). The combined extracts were washed with brine (30 mL x 3), dried over Na2SO4, filtered, and concentrated. The residue was purified by silica flash chromatography (0-50% EtOAc/petroleum ether) to give the title compound (600 mg, 71.77%) as a yellow solid.

[0912] Step 3: Chiral purification to isolate (1S,2R,3R,5R)-tert-butyl 3-((6-(4-(difluoromethyl)-2- methoxyphenyl) pyridazin-3-yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]oc tane-8-carboxylate and (1R,2S,3S,5S)-tert-butyl 3-((6-(4-(difluoromethyl)-2-methoxyphenyl) pyridazin-3-yl)(methyl)amino)-2- fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate (NPENT-120A & NPENT-120B). Racemic intermediate, (±) (1S,2R,3R,5R)-tert-butyl 3-((6-(4-(difluoromethyl)-2-methoxyphenyl) pyridazin-3-yl)(methyl)amino)-2- fluoro-8-azabicyclo[3.2.1]octane-8-carboxylate, 600 mg, was purified by chiral-HPLC to give the title compound NPENT-120A (266 mg, t R = 1.74 min) and NPENT-120B (274 mg, t R = 2.53 min) as a colorless oils. LCMS: m/z 493.3 [M+H] + .

[0913] Chiral HPLC condition: Instrument: SFC-80 (Thar, Waters); Column: OJ 20*250mm, 10um (Daicel); Column temperature: 35 ºC; Mobile phase: CO2/ MeOH(0.2%Methanol Ammonia) = 85/15; Flow rate: 80 g/min; Back pressure: 100 bar; Detection wavelength: 214 nm; Cycle time: 3.8 min; Sample solution: 840 mg dissolved in 25 ml methanol; Injection volume: 0.6 ml

Step 4: Synthesis of (1S,2R,3R,5R)-tert-butyl 3-((6-(4-(difluoromethyl)-2-hydroxyphenyl) pyridazin-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate and (1R,2S,3S,5S)-tert-butyl 3-((6- (4-(difluoromethyl)-2-hydroxyphenyl) pyridazin-3-yl)(methyl)amino)-2-fluoro-8- azabicyclo[3.2.1]octane-8-carboxylate (PENT-120A & PENT-120B).

[0914] A mixture of NPENT-120A (150 mg, 0.3 mmol), 1-decanethiol (261 mg, 1.5 mmol) and K 2 CO 3 (83 mg, 0.6 mmol) in NMP (2 mL) was stirred at 150 o C for 8 h in a sealed tube. The resulting mixture was poured into ice water (20 mL) and extracted with EtOAc (15 mL x 3). The combined extracts were washed with brine (30 mL x 3), dried over Na 2 SO 4 , filtered, and concentrated. The residue was purified by silica flash chromatography (0-50% EtOAc/petroleum ether) to give the title compound as PENT-120A (120 mg, 82.2%) as a yellow solid. LCMS: m/z 479.3 [M+H] + ; t R = 2.11 min.

[0915] Following the procedure above, but with NPENT-120B (150 mg, 0.3 mmol), gave the title compound as PENT-120B (120 mg, 82.2%) as a yellow solid. LCMS: m/z 479.3 [M+H] + ; t R = 2.11 min.

[0916] Step 5: Synthesis of 5-(difluoromethyl)-2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicycl o [3.2.1]octan- 3-yl)(methyl)amino)pyridazin-3-yl)phenol and 5-(difluoromethyl)-2-(6-(((1R,2R,3S,5S)-2-fluoro-8- azabicyclo [3.2.1]octan-3-yl)(methyl)amino)pyridazin-3-yl)phenol (Compound 120A and Compound 120B).

[0917] TFA (1 mL) was added to a solution of PENT-120A (150 mg, 0.31 mmol) in CH 2 Cl 2 (3 mL). The mixture was stirred at room temperature for 1 h. The mixture was adjusted to pH 7 - 8 by adding 8N

NH 3 /Methanol. The residue was purified by Prep-HPLC (ACN and H 2 O with 0.05% NH 4 HCO 3 as mobile phase) to give the title compound as Compound 120A (73.2 mg, 62.56%) as a white solid. 1 H NMR (400 MHz, MeOD) d 8.15 (d, J = 9.9 Hz, 1H), 7.88 (d, J = 8.6 Hz, 1H), 7.32 (d, J = 9.9 Hz, 1H), 7.11 (d, J = 7.0 Hz, 2H), 6.89-6.60 (m, 1H), 5.41– 5.19 (m, 1H), 4.81-4.66 (m, 1H), 3.68 (s, 2H), 3.13 (d, J = 1.7 Hz, 3H), 2.44-2.37 (m, 1H), 2.09– 1.82 (m, 4H), 1.73– 1.57 (m, 1H). LCMS: m/z 379.2 [M+H] + ; t R = 1.79 min.

[0918] Following the procedure above, but with PENT-120B, gave the title compound as Compound 120B (71.7 mg, 61.28%) as a white solid. 1 H NMR (400 MHz, MeOD) d 8.15 (d, J = 9.9 Hz, 1H), 7.88 (d, J = 8.6 Hz, 1H), 7.32 (d, J = 9.9 Hz, 1H), 7.11 (d, J = 7.0 Hz, 2H), 6.89-6.60 (m, 1H), 5.41– 5.19 (m, 1H), 4.81-4.66 (m, 1H), 3.68 (s, 2H), 3.13 (d, J = 1.7 Hz, 3H), 2.44-2.37 (m, 1H), 2.09– 1.82 (m, 4H), 1.73– 1.57 (m, 1H). LCMS: m/z 379.2 [M+H] + ; t R = 1.79 min.

Compound 122: Synthesis of 5-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-2-(1-methyl-1H-pyrazol-4-yl )pyrimidin-4-ol.

[0919] Step 1: Synthesis of 2-chloro-5-iodo-4-((4-methoxybenzyl)oxy)pyrimidine. To a solution of (4- methoxyphenyl)methanol (1.0 g, 7.24 mmol) and 2,4-dichloro-5-iodopyrimidine (2.39 g, 8.69 mmol) in THF (20 mL) at 0 °C was added NaH (434.2 mg, 10.86 mmol, 60 wt% in mineral oil). The mixture was stirred at room temperature for 16 h, quenched with water (5 mL) and extracted with ethyl acetate (40 mL x 2). The combined organic layers were washed with sodium chloride solution, dried, concentrated, purified by silica gel chromatography (0 to 5% ethyl acetate/petroleum ether) to give 2-chloro-5-iodo-4-((4- methoxybenzyl)oxy)pyrimidine (1.6 g, 58% yield) as a white solid. LCMS: t R = 1.65 min.

[0920] Step 2: Synthesis of (±) tert-butyl (1R,2S,3S,5S)-2-fluoro-3-(methyl(6-(tributylstannyl)pyridazi n- 3-yl)amino)-8-azabicyclo[3.2.1]octane-8-carboxylate. A mixture of tert-butyl (1R,2S,3S,5S)-3-((6- chloropyridazin-3-yl)(methyl)amino)-2-fluoro-8-azabicyclo[3. 2.1]octane-8-carboxylate (INT-1, 600 mg, 1.62 mmol), Bu 3 Sn-SnBu 3 (1.88 g, 3.24 mmol), Pd 2 (dba) 3 (148.2 mg, 0.16 mmol) and PCy 3 (90.7 mg, 0.32 mmol) in toluene (10 mL) was stirred at 110 °C under N 2 for 16 h. The mixture was filtered, the filtrate was concentrated in vacuum and purified by combiflash (Al 2 O 3 , basic) using ethyl acetate /petroleum ether from 0 to 10% to give the title compound (458 mg, 45 % yield) as a light yellow oil. LCMS: m/z 627.0 [M+1] + , t R =3.93 min.

[0921] Step 3: Synthesis of (±) tert-butyl (1R,2S,3S,5S)-3-((6-(2-chloro-4-((4- methoxybenzyl)oxy)pyrimidin-5-yl)pyridazin-3-yl)(methyl)amin o)-2-fluoro-8-azabicyclo[3.2.1]octane-8- carboxylate. A mixture of (±) tert-butyl (1R,2S,3S,5S)-2-fluoro-3-(methyl(6-(tributylstannyl)pyridazi n-3- yl)amino)-8-azabicyclo[3.2.1]octane-8-carboxylate (130 mg, 0.35 mmol), 2-chloro-5-iodo-4-((4- methoxybenzyl)oxy)pyrimidine (370.1 mg, 0.41 mmol) and Pd(PPh 3 ) 4 (39.9 mg, 0.03 mmol) in toluene (6 mL) was stirred under N 2 at 110 o C for 16 h. The mixture was concentrated in vacuum and purified by prep- TLC (petroleum ether/ethyl acetate = 2/1) to the title compound (80 mg, 39 % yield) as brown solid. LCMS: m/z 585.0 [M+1] + , t R =2.06 min.

[0922] Step 4: Synthesis of (±) tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(4-hydroxy-2-(1-methyl-1H- pyrazol-4-yl)pyrimidin-5-yl)pyridazin-3-yl)(methyl)amino)-8- azabicyclo[3.2.1]octane-8-carboxylate. A mixture of (±) tert-butyl (1R,2S,3S,5S)-3-((6-(2-chloro-4-((4-methoxybenzyl)oxy)pyrimi din-5-yl)pyridazin-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate (323 mg, 0.552 mmol), 1-methyl-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (230 mg, 1.116 mmol), Pd(dppf)Cl 2 (40.4 mg, 0.05 mmol) and K2CO3 (229 mg, 1.66 mmol) in dioxane (3 mL) and water (1 mL) was stirred at 100 °C for 16 h under N 2 . The reaction mixture was concentrated in vacuum, and purified by silica gel chromatography (0 to 4% MeOH/dichloromethane) to give the title compound (135 mg). LCMS: m/z 511.0 [M+1] + , t R = 1.62 min).

[0923] Step 5: (±) Synthesis of 5-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-2-(1-methyl-1H-pyrazol-4-yl )pyrimidin-4-ol (Compound 122). A mixture of (±) tert-butyl (1R,2S,3S,5S)-2-fluoro-3-((6-(4-hydroxy-2-(1-methyl-1H-pyraz ol-4-yl)pyrimidin-5- yl)pyridazin-3-yl)(methyl)amino)-8-azabicyclo[3.2.1]octane-8 -carboxylate (143 mg, 0.28 mmol) and HCl/dioxane (10 mL) in DCM (5 mL) was stirred at room temperature for 2 h. The mixture was concentrated, and neutralized with 0.5 mL of 7M ammonia MeOH. The crude product was purified by silica gel column (1- 10% MeOH/DCM) to give the title compound (82.5 mg, 63.9 % yield) as pale yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) d 8.70 (s, 1H), 8.55 (s, 1H), 8.29-8.20 (m, 2H), 7.20 (d, J = 9.6 Hz, 1H), 5.39-5.18 (m, 1H), 5.08-4.87 (m, 1H), 4.16-3.98 (m, 2H), 3.92 (s, 3H), 3.39 (s, 1H), 3.01 (s, 3H), 2.13-1.86 (m, 4H), 1.84-1.74 (m, 1H). LCMS: m/z 411.0 [M+1] + ; t R = 1.26 min.

Compound 123A & 123B: Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3- yl)(methyl)amino) pyridazin-3-yl)-5-(1-methyl-1H-imidazol-2-yloxy)phenol and 2-(6-(((1R,2R,3S,5S)-2- fluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino) pyridazin-3-yl)-5-(1-methyl-1H-imidazol-2- yloxy)phenol. [0924] Step 1: Synthesis of 2-(4-bromo-3-methoxyphenoxy)-1-methyl-1H-imidazole. A mixture of 2- chloro-1-methyl-1H-imidazole (5.0 g, 48.2 mmol) and 4-bromo-3-methoxyphenol (6.0 g, 30.0 mmol) in DIPEA (120 mL) was sealed and heated to 150 °C for 8 h. The mixture was concentrated, and the residue was purified by Prep-HPLC (80% MeOH/H 2 O) to give 2-(4-bromo-3-methoxyphenoxy)-1-methyl-1H-imidazole (4.05 g, 43.1% yield) as an oil. LCMS: m/z 283.0 [M+H] + ; t R = 1.65 min.

[0925] Step 2: Synthesis of 2-bromo-5-(1-methyl-1H-imidazol-2-yloxy)phenol. BBr 3 (25 mL, 25.0 mmol, 1N in CH 2 Cl 2 ) was added to a solution of 2-(4-bromo-3-methoxyphenoxy)-1-methyl-1H-imidazole (2.5 g, 8.8 mmol) in CH 2 Cl 2 (15 mL). Then the mixture was stirred at room temperature overnight. MeOH (10 mL) was added to quench the reaction. The resulting mixture was concentrated and purified by silica gel

chromatography (5-10% MeOH in CH 2 Cl 2 ) to yield 2-bromo-5-(1-methyl-1H-imidazol-2-yloxy)phenol (1.3 g, 52.3% yield) as solid. LCMS: m/z 269.0 [M+H] + ; t R = 1.38 min.

[0926] Step 3: Synthesis of 2-(4-bromo-3-((2-(trimethylsilyl)ethoxy)methoxy) phenoxy)-1-methyl-1H- imidazole. NaH (220 mg, 5.5 mmol, 60 wt% in mineral oil) was added to a solution of 2-bromo-5-(1-methyl- 1H-imidazol-2-yloxy)phenol (1 g, 3.7 mmol) in DMF (20 mL). The mixture was stirred for another 30 min at 0 o C, then SEMCl (900 mg, 5.5 mmol) was added. The mixture was stirred at room temperature for 3 h, quenched with 30 mL of water and extracted with ethyl acetate (40 mL X 3). The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , concentrated and purified by silica gel

chromatography (10-25% ethyl acetate in petroleum ether) to give 2-(4-bromo-3-((2- (trimethylsilyl)ethoxy)methoxy)phenoxy)-1-methyl-1H-imidazol e (700 mg, 47% yield). LCMS: m/z 399.1 [M+H] + ; tR = 1.81 min.

[0927] Step 4: Synthesis of 1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) -3-((2- (trimethylsilyl)ethoxy)methoxy)phenoxy)-1H-imidazole. A mixture of 2-(4-bromo-3-((2- (trimethylsilyl)ethoxy)methoxy)phenoxy)-1-methyl-1H-imidazol e (500 mg, 1.25 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(1,3,2-dioxaborolane) (476 mg, 1.88 mmol), potassium acetate (246 mg, 2.5 mmol) and Pd(dppf)Cl 2 (108 mg, 0.12 mmol) in 1,4-dioxane (20.0 mL) was degassed and stirred at 100 °C overnight. The mixture was concentrated and purified by silica gel chromatography (10-25% ethyl acetate in petroleum ether) to give 1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 3-((2- (trimethylsilyl)ethoxy)methoxy)phenoxy)-1H-imidazole (375 mg, 58% yield). LCMS: m/z 447.3 [M+H] + ; t R = 1.82 min.

[0928] Step 5: Synthesis of (1S,2R,3R,5R)-tert-butyl 2-fluoro-3-(methyl(6-(4-(1-methyl-1H- imidazol-2- yloxy)-2-((2-(trimethylsilyl)ethoxy)methoxy)phenyl)pyridazin -3-yl)amino)-8-azabicyclo[3.2.1]octane-8- carboxylate and (1R,2S,3S,5S)-tert-butyl 2-fluoro-3-(methyl(6-(4-(1-methyl-1H- imidazol-2-yloxy)-2-((2- (trimethylsilyl)ethoxy)methoxy)phenyl)pyridazin-3-yl)amino)- 8-azabicyclo[3.2.1]octane-8-carboxylate. A mixture of 1-methyl-2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 3-((2- (trimethylsilyl)ethoxy)methoxy)phenoxy)-1H-imidazole (100 mg, 0.27 mmol), HETX-6A (144 mg, 0.32 mmol), potassium carbonate (93 mg, 0.67 mmol) and Pd(dppf)Cl 2 (23 mg, 0.03 mmol) in 5 mL of 1,4-dioxane and 1 mL of H 2 O was degassed and stirred at 110 0 C for 2 h. The mixture was concentrated and purified by silica gel chromatography (5-10% methanol in CH 2 Cl 2 ) to yield the title compound as PENT-123A (100 mg, 57% yield). LCMS: m/z 655.3 [M+H] + ; t R = 1.83 min.

[0929] Following the procedure above, but with HETX-6B (100 mg, 0.27 mmol), gave the title compound as PENT-123B (100 mg, 57% yield). LCMS: m/z 655.0 [M+H] + ; t R = 2.10 min.

[0930] Step 6: Synthesis of 2-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl)

(methyl)amino)pyridazin-3-yl)-5-(1-methyl-1H-imidazol-2-y loxy)phenol and 2-(6-(((1R,2R,3S,5S)-2- fluoro-8-azabicyclo[3.2.1]octan-3-yl) (methyl)amino)pyridazin-3-yl)-5-(1-methyl-1H-imidazol-2- yloxy)phenol (Compound 123A & Compound 123B). To the solution of PENT-123A (100 mg, 0.15 mmol) in CH 2 Cl 2 (2.0 mL) was added HCl (3 mL, 4.0 N in 1,4-dioxane). The mixture was stirred for 1 h and concentrated and the residue was dissolved into water (2.0 mL) and neutralized till pH~9 with K 2 CO 3 . The resulting mixture was extracted with CH 2 Cl 2 (10 mL X 2) and the organic layers were concentrated and purified by silica gel chromatography (5-20% methanol in CH 2 Cl 2 ) to give the title compound as Compound 123A (41 mg, 63% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 13.78 (s, 1H), 8.20 (d, J = 10.0 Hz, 1H), 7.91 (d, J = 10.0 Hz, 1H), 7.40 (d, J = 9.9 Hz, 1H), 7.00 (d, J = 1.5 Hz, 1H), 6.78– 6.61 (m, 3H), 5.06 (d, J = 23.9 Hz, 1H), 4.75 (d, J = 51.3 Hz, 1H), 3.66-3.64 (m, 2H), 3.49 (s, 3H), 3.03 (s, 3H), 2.32-2.25 (m, 1H), 1.91– 1.67 (m, 4H), 1.62-1.58 (m, 1H). LCMS: m/z 425.1 [M+H] + ; t R = 1.14 min.

[0931] Following the procedure above, but with PENT-123B (100 mg, 0.27 mmol), gave the title compound as Compound 123B (36.5 mg, 57.2% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 13.78 (s, 1H), 8.20 (d, J = 10.0 Hz, 1H), 7.91 (d, J = 10.0 Hz, 1H), 7.40 (d, J = 9.9 Hz, 1H), 7.00 (d, J = 1.5 Hz, 1H), 6.78– 6.61 (m, 3H), 5.06 (d, J = 23.9 Hz, 1H), 4.75 (d, J = 51.3 Hz, 1H), 3.66-3.64 (m, 2H), 3.49 (s, 3H), 3.03 (s, 3H), 2.32- 2.25 (m, 1H), 1.91– 1.67 (m, 4H), 1.62-1.58 (m, 1H). LCMS: m/z 425.1 [M+H] + ; t R = 1.14 min.

Compound 124A: Synthesis of N-(tert-butyl)-1-(4-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo [3.2.1]octan- 3-yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)-1H-imida zole-4-carboxamide or N-(tert-butyl)-1- (4-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl )(methyl)amino)pyridazin-3-yl)-3- hydroxyphenyl)-1H-imidazole-4-carboxamide.

[0932] Step 1: Synthesis of (1S,2R,3R,5R)-tert-butyl 3-((6-(4-(4-cyano-1H-imidazol-1-yl)-2- hydroxyphenyl)pyridazin-3-yl)(methyl)amino)-2-fluoro-8-azabi cyclo[3.2.1]octane-8-carboxylate or (1R,2S,3S,5S)-tert-butyl 3-((6-(4-(4-cyano-1H-imidazol-1-yl)-2-hydroxyphenyl)pyridazi n-3- yl)(methyl)amino)-2-fluoro-8-azabicyclo[3.2.1]octane-8-carbo xylate (PENT-69A). A mixture of HETX- 6A (100 mg, 0.27 mmol), (1-(3-hydroxy-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl )phenyl)-1H- imidazole-4-carbonitrile (B6, 168 mg, 0.54 mmol), Pd(dppf)Cl 2 (23 mg, 0.03 mmol) and K 2 CO 3 (76 mg, 0.54 mmol) in dioxane (8 mL) and water (2 mL) was degassed and stirred at 110 o C for 2 h. The mixture was concentrated and purified by silica gel column (80% EtOAc/petroleum ether) to give 100 mg of the title compound as PENT-69A as a yellow solid (71% yield). LCMS: m/z 520.2 [M+H] + ; t R = 1.95 min.

[0933] Step 2: Synthesis of N-(tert-butyl)-1-(4-(6-(((1S,2S,3R,5R)-2-fluoro-8-azabicyclo [3.2.1]octan-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)-1H-imidazo le-4-carboxamide or N-(tert-butyl)-1- (4-(6-(((1R,2R,3S,5S)-2-fluoro-8-azabicyclo[3.2.1]octan-3-yl )(methyl)amino)pyridazin-3-yl)-3- hydroxyphenyl)-1H-imidazole-4-carboxamide (Compound 123).1 mL of TFA was added to a stirred solution of PENT-69A (100 mg, 0.193 mmol) in 1 mL of CH 2 Cl 2 . The mixture was stirred at room temperature for 2 h and concentrated under reduced pressure. The residue was dissolved in water. pH value was adjusted to ~9 by using K 2 CO 3 aqueous solution. The mixture was extracted with 10% MeOH/CH 2 Cl 2 (20 mL X 3). The combined organic phases were dried and purified by reverse HPLC, to give Compound 69A (28 mg, 35% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 14.23 (s, 1H), 8.83 (d, J = 1.2 Hz, 1H), 8.62 (d, J = 1.2 Hz, 1H), 8.33 (d, J = 10.0 Hz, 1H), 8.10 (d, J = 8.7 Hz, 1H), 7.43 (d, J = 10.0 Hz, 1H), 7.37 (d, J = 2.3 Hz, 1H), 7.30 (dd, J = 8.6, 2.3 Hz, 1H), 5.13– 4.90 (m, 1H), 4.66-4.62 (m, 1H), 3.53 (s, 2H), 3.04 (s, 3H), 2.32– 2.19 (m, 2H), 1.82– 1.50 (m, 4H). LCMS: m/z 420.2 [M+H] + ; t R = 1.64 min.

[0934] The title compound was also isolated as Compound 123 (15 mg, 18% yield). 1 H NMR (400 MHz, DMSO-d 6 ) d 14.14 (s, 1H), 8.45– 8.38 (m, 1H), 8.32 (d, J = 10.0 Hz, 1H), 8.25 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.42 (d, J = 9.7 Hz, 1H), 7.36 (s, 1H), 7.32 (d, J = 8.5 Hz, 1H), 7.19 (s, 1H), 5.16– 4.93 (m, 1H), 4.75– 4.54 (m, 1H), 3.54 (s, 2H), 3.05 (s, 3H), 2.25 (t, J = 11.6 Hz, 1H), 1.88– 1.62 (m, 4H), 1.62– 1.50 (m, 1H), 1.40 (s, 9H). LCMS: m/z 494.2 [M+H] + ; t R = 1.75 min.

Compound 127A & 127B: Synthesis of (R)-4-(4-(6-(((1R,3S,5S)-6,6-difluoro-8-azabicyclo[3.2.1]oct an-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)piperidin-2 -one and (S)-4-(4-(6-(((1R,3S,5S)-6,6- difluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridaz in-3-yl)-3-hydroxyphenyl)piperidin-2-one and (S)-4-(4-(6-(((1S,3R,5R)-6,6-difluoro-8-azabicyclo[3.2.1]oct an-3-yl)(methyl)amino)pyridazin-3-yl)- 3-hydroxyphenyl)piperidin-2-one and (R)-4-(4-(6-(((1S,3R,5R)-6,6-difluoro-8-azabicyclo[3.2.1]oct an-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)piperidin-2 -one (Compounds 127A, Compound 127B, Compound 128A, and Compound 128B).

[0935] Step 1: Synthesis of (±) tert-butyl (1S,3R,5R)-3-((6-(4-chloro-5-fluoro-2- (methoxymethoxy)phenyl)pyridazin-3-yl)(methyl)amino)-6,6-dif luoro-8-azabicyclo[3.2.1]octane-8- carboxylate. Into a 50-mL 3-necked round-bottom flask, was placed a solution of INT-2 (3.80 g, 9.773 mmol, 1.00 equiv) in dioxane (4 mL), and H 2 O (1 mL), 2-[4-chloro-5-fluoro-2-(methoxymethoxy)phenyl]- 4,4,5,5-tetramethyl-1,3,2- dioxaborolane (B7, 3.09 g, 9.76 mmol, 1.0 equiv), Pd(dppf)Cl 2 (0.36 g, 0.49 mmol, 0.05 equiv), K 2 CO 3 (2.70 g, 19.5 mmol, 2 equiv). The resulting solution was stirred for 3 h at 100 °C. The resulting mixture was concentrated. The residue was applied onto a silica gel column with ethyl

acetate/petroleum ether (15:100). To give the title compound (3.0 g, 57% yield) as a yellow oil.

Step 2: Synthesis of tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2-(methoxymethoxy)-4-(2-oxo-1 ,2- dihydropyridin-4-yl)phenyl)pyridazin-3-yl)(methyl)amino)-8-a zabicyclo[3.2.1]octane-8-carboxylate and tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2-(methoxymethoxy)-4-(2-oxo-1 ,2-dihydropyridin-4- yl)phenyl)pyridazin-3-yl)(methyl)amino)-8-azabicyclo[3.2.1]o ctane-8-carboxylate. Into a 100-mL 3- necked round-bottom flask, was placed a solution of (±) tert-butyl (1S,3R,5R)-3-([6-[4-chloro-5-fluoro-2- (methoxymethoxy)phenyl]pyridazin-3-yl](methyl)amino)-6,6-dif luoro-8-azabicyclo[3.2.1]octane-8- carboxylate (2.80 g, 5.16 mmol, 1.0 equiv) in dioxane (20 mL), and H 2 O (4 mL), 1-methyl-2-oxopyridin-4- ylboronic acid (2.50 g, 16.3 mmol, 3.2 equiv), K 2 CO 3 (2.12 g, 15.3 mmol, 3 equiv), Pd(dppf)Cl 2 (0.338 g, 0.463 mmol, 0.09 equiv). The resulting solution was stirred for 3 h at 100 °C. The resulting solution was extracted with 2x10 mL of ethyl acetate dried over anhydrous sodium sulfate and concentrated. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (7:3) to provide the crude product, which was further purified by prep-SFC. This resulted in isolation of the title compounds as PENT-22A (750 mg , t R = 4.05 min) and PENT-22B (700 mg, 28%, t R = 5.66 min).

[0936] Column: CHIRALPAK AD-H, 2.0cm I.D.*25cm L; Mobile Phase A: CO2, Mobile Phase B: IPA; Flow rate: 45 mL/min; Gradient: 35% B; 220 nm; RT1: 4.05 min; RT2: 5.66 min.

[0937] Step 3: Synthesis of (±) tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2-hydroxy-4-((R)-2- oxopiperidin-4-yl)phenyl)pyridazin-3-yl)(methyl)amino)-8-aza bicyclo[3.2.1]octane-8-carboxylate and (±) tert-butyl (1S,3R,5R)-6,6-difluoro-3-((6-(2-hydroxy-4-((S)-2-oxopiperid in-4-yl)phenyl)pyridazin-3- yl)(methyl)amino)-8-azabicyclo[3.2.1]octane-8-carboxylate. Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of H2, was placed PENT-22A (700 mg, 1 equiv), MeOH (10 ml), PtO 2 (250 mg). The resulting solution was stirred for 2 days at 45 °C, and concentrated. This resulted in 600 mg (85.05%) of the title compound as a colorless oil. The residue was purified by Chiral-Prep-HPLC with the following conditions to provide the two title compounds as PENT-127A (110 mg, t R = 9.2 min) and PENT- 127B (100 mg, t R = 13.2 min) as colorless oils. Chiral HPLC Purification Conditions: CHIRAL ART

Cellulose-SB, 2*25cm,5um; Mobile Phase A: Hexane--HPLC, Mobile Phase B: EtOH--HPLC; Flow rate: 25 mL/min; Gradient: 20% B to 20% B in 18 min; Detection wavelength 300/210 nm.

[0938] Following the procedure above, but with PENT-22B (700 mg), gave the two title compounds as PENT-128A (110 mg, tR = 8.0 min) and PENT-128B (100 mg, tR = 9.0 min) as colorless oils.

[0939] Step 4: Synthesis of (R)-4-(4-(6-(((1R,3S,5S)-6,6-difluoro-8-azabicyclo[3.2.1]oct an-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)piperidin-2 -one and (S)-4-(4-(6-(((1R,3S,5S)-6,6- difluoro-8-azabicyclo[3.2.1]octan-3-yl)(methyl)amino)pyridaz in-3-yl)-3-hydroxyphenyl)piperidin-2-one and (S)-4-(4-(6-(((1S,3R,5R)-6,6-difluoro-8-azabicyclo[3.2.1]oct an-3-yl)(methyl)amino)pyridazin-3-yl)- 3-hydroxyphenyl)piperidin-2-one and (R)-4-(4-(6-(((1S,3R,5R)-6,6-difluoro-8-azabicyclo[3.2.1]oct an-3- yl)(methyl)amino)pyridazin-3-yl)-3-hydroxyphenyl)piperidin-2 -one (Compounds 127A, Compound 127B, Compound 128A, and Compound 128B). Into a 8-mL vial, was placed PENT-127A (110 mg), HCl (gas) in 1,4-dioxane (5.0 mL). The resulting solution was stirred overnight at room temperature. The residue was dissolved in 5 mL of MeOH. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, 5MMNH 4 HCO 3 /ACN=25 increasing to 5MMNH 4 HCO 3 /ACN=45 within 25 min; Detector, 254. This resulted in isolation of one of the 4 possible diastereomeric title compounds as a pure enantiomer as Compound 127A (58.2 mg) as an off-white solid. 1 H- NMR (400 MHz, DMSO-d 6 ) d 13.36 (s, 1H), 8.21 (d, J = 10.0 Hz, 1H), 7.73 (d, J = 11.9 Hz, 1H), 7.29 (d, J = 9.8 Hz, 1H), 5.09 (s, 1H), 3.62 (s, 1H), 3.50– 3.36 (m, 2H), 3.30 (s, 1H), 2.97 (s, 3H), 2.86 (s, 4H), 2.46– 2.37 (m, 2H), 2.37– 2.27 (m, 2H), 2.00 (d, J = 5.0 Hz, 3H), 1.81 (d, J = 13.5 Hz, 2H), 1.68 (dd, J = 11.8, 5.6 Hz, 1H). LCMS: m/z 620. [0940] Following the procedure above with PENT-127B (100 mg), gave Compound 127B (35.6 mg) as an off white solid. 1 H-NMR (400 MHz, DMSO-d 6 ) d 13.36 (s, 1H), 8.21 (d, J = 10.0 Hz, 1H), 7.73 (d, J = 11.9 Hz, 1H), 7.29 (d, J = 9.8 Hz, 1H), 5.09 (s, 1H), 3.62 (s, 1H), 3.50– 3.36 (m, 2H), 3.30 (s, 1H), 2.97 (s, 3H), 2.86 (s, 4H), 2.46– 2.37 (m, 2H), 2.37– 2.27 (m, 2H), 2.00 (d, J = 5.0 Hz, 3H), 1.81 (d, J = 13.5 Hz, 2H), 1.68 (dd, J = 11.8, 5.6 Hz, 1H). LCMS: m/z 620.

[0941] Following the procedure above with PENT-128A (110 mg), gave Compound 128A (53.2 mg) as an off white solid. 1 H-NMR (400 MHz, DMSO-d 6 ) d 13.36 (s, 1H), 8.21 (d, J = 10.0 Hz, 1H), 7.73 (d, J = 11.9 Hz, 1H), 7.29 (d, J = 9.8 Hz, 1H), 5.09 (s, 1H), 3.62 (s, 1H), 3.50– 3.36 (m, 2H), 3.30 (s, 1H), 2.97 (s, 3H), 2.86 (s, 4H), 2.46– 2.37 (m, 2H), 2.37– 2.27 (m, 2H), 2.00 (d, J = 5.0 Hz, 3H), 1.81 (d, J = 13.5 Hz, 2H), 1.68 (dd, J = 11.8, 5.6 Hz, 1H). LCMS: m/z 620.

[0942] Following the procedure above with PENT-128B (100 mg), gave Compound 128B (47.2 mg) as an off white solid. 1 H-NMR (400 MHz, DMSO-d 6 ) d 13.36 (s, 1H), 8.21 (d, J = 10.0 Hz, 1H), 7.73 (d, J = 11.9 Hz, 1H), 7.29 (d, J = 9.8 Hz, 1H), 5.09 (s, 1H), 3.62 (s, 1H), 3.50– 3.36 (m, 2H), 3.30 (s, 1H), 2.97 (s, 3H), 2.86 (s, 4H), 2.46– 2.37 (m, 2H), 2.37– 2.27 (m, 2H), 2.00 (d, J = 5.0 Hz, 3H), 1.81 (d, J = 13.5 Hz, 2H), 1.68 (dd, J = 11.8, 5.6 Hz, 1H). LCMS: m/z 620.

Example B16. Compounds of the disclosure

[0943] Table 14 shows additional compounds that were synthesized using the methods described above. In some cases, an SMSM provided herein can be designated by more than one SMSM # in different parts of the application; for example, the same compound can appear more than once in and Table 1A, Table 1C, Table 1E, Table 1G, or Table 1H, and Table 14, in the examples, and in the schemes.

Table 14. List of compounds

Example A14. mHTT protein assay

[0944] Compounds were tested on GM04724 (CAG 70/20) Huntington’s disease patient lymphoblast cells at doses ranging from 10 mM to 0.6 nM. 4,500 cells/well were seeded in 384 well plates. One plate replica was carried out for parallel viability testing by CellTiter Glo (CTG). Compounds were incubated for 48 hours. mHTT protein levels were assessed by the 2B7–MW1 assay via Mesoscale Discovery (MSD) as previously reported (Macdonald et al., 2014). The antibody pair is comprised of previously characterized monoclonals (2B7 and MW1) interrogating two regions for HTT conformation and biological properties: the N17 domain and the polyQ domain (Baldo et al., 2012; Ko et., 2001).2B7–MW1 is dependent on subject/animal specific levels of HTT at the time of treatment.2B7–MW1 is dependent on polyQ expansion (e.g., the higher the expansion the higher the signal) and on mHTT size (e.g., a similar polyQ will give higher signal with smaller HTT size). The viability readout was carried out by CTG according to the manufacturer’s instructions.

[0945] The results of the mHTT protein assay and permeability assay are shown in Table 15. It is to be understood that the absolute stereochemistry for compounds of Table 14 is not determined and is arbitrarily assigned, and may in fact correspond to the enantiomer of the compound drawn. In some embodiments, the relative stereochemistry is indicated.

[0946] Example A15. HTT Quantitative Splicing Assay

[0947] GM04724 (CAG 70/20) Huntington’s disease patient lymphoblasts (Coriell) are plated in 96-wel v- bottom plates at 50,000 cells/well. Immediately after plating, cells are dosed with compound for 24 h at concentrations ranging from 2.5uM to 0.15nM (0.1% DMSO). Treated cells are lysed and cDNA synthesized using the Fast Advanced Cells-to-Ct kit (Thermofisher A35378) according to the manufacturer’s instructions. 2 uL of each cDNA are used in qPCR reactions to confirm the compound-induced inclusion of a cryptic exon within intron 49 of the Huntingtin (HTT) transcripts. The qPCR reactions are prepared in 384-well plates in 10 uL volume, using TaqMan™ Fast Advanced Master Mix [ThermoFisher; 4444965] with primers and probes shown in the table below. Reactions are run in a Quant Studio 6 qPCR instrument with default settings

[0948] Probe/primer sequences:

HTTcryp49b-FAM:

Example B19. SMN protein assay

[0949] Compounds were tested on spinal muscular atrophy (SMA) patient fibroblasts (GM03813, Coriell) at doses ranging from 2.5 mM to 0.6 nM.7000 cells/well were seeded in 96-well plates. Compounds were incubated for 48 hours and the cells were lysed with 100 mL of lysis buffer.20 mL of lysate was used for SMN protein measurement by Mesoscale Discovery (MSD) assay developed by PharmOptima (Michigan). A standard curve prepared with SMN protein ranging from 1mg/ml to 19.5pg/ml was used in each MSD plate to calculate the absolute SMN protein amount in each sample.

[0950] One plate with 700 cells/well was prepared for parallel viability testing by Cell Tier Glo reagents (Promega, G7572/G7573 (CTG). The viability readout was carried out according to the manufacturer’s instructions. The assay results for some exemplary compounds are shown in Table 15.

Example B20. SMN Quantitative Splicing Assay

[0951] Spinal muscular atrophy (SMA) patient fibroblasts (GM03813, Coriell) are plated in 96-well plates at 50,000 cells/well. Immediately after plating, cells are dosed with compounds for 24 h at concentrations ranging from 2.5mM to 0.6nM (0.1% DMSO). Treated cells are lysed and cDNA synthesized using the Fast Advanced Cells-to-Ct kit (Thermofisher A35378) according to the manufacturer’s instructions.2 mL of each cDNA are used in qPCR reactions. The qPCR reactions are prepared in 384-well plates in 10 mL volume, using TaqMan™ Fast Advanced Master Mix (ThermoFisher; 4444965) with primers and probes shown in the table below. Reactions are run in a Quant Studio 6 qPCR instrument with default settings.

[0952] Probe/primer sequences:

Table 15. Assay data for Compounds

[0953] The examples and embodiments described herein are for illustrative purposes only and various modifications or changes suggested to persons skilled in the art are to be included within the spirit and purview of this application and scope of the appended claims.