Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS, COMPOSITIONS, AND METHODS FOR PROTEIN DEGRADATION
Document Type and Number:
WIPO Patent Application WO/2020/264172
Kind Code:
A1
Abstract:
Disclosed herein are compounds that target SMARCA2 and SMARCA4, causing their degradation. Also disclosed herein are compositions and methods of use in treating associated disorders and diseases.

Inventors:
QI JUN (US)
ARMSTRONG SCOTT (US)
WU LEI (CN)
Application Number:
PCT/US2020/039638
Publication Date:
December 30, 2020
Filing Date:
June 25, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DANA FARBER CANCER INST INC (US)
International Classes:
A61K31/4035; A61K31/438; A61K31/439
Domestic Patent References:
WO2018160636A12018-09-07
WO2019055657A12019-03-21
Foreign References:
US20190071415A12019-03-07
US20170362572A12017-12-21
US20190076539A12019-03-14
Other References:
See also references of EP 3989967A4
Attorney, Agent or Firm:
SMITH, DeAnn, F. et al. (US)
Download PDF:
Claims:
We claim:

or a pharmaceutically acceptable salt thereof, wherein

X is a bond, aryl, or heteroaryl;

L is an alkylene, alkenylene, or alkynylene chain comprising 1 to 35 carbon atoms, optionally wherein:

at least one, but no more than ten, -CH2- moieties of L are independently replaced with a moiety selected from -C(=0)-, -C(=0)-NR3- -NR3-C(=0)-, -C(=0)-0-, -O- C(=0)-, -NR3-C(=0)-NR3-, -0-C(=0)-NR3-, -NR3-C(=0)-0-, -0-, -S-, and -NR3-, provided the number of -CH2- moieties of L is larger than the collective number of -C(=0)-, -C(=0)-NR3- -NR3-C(=0)-, -C(=0)-0-, -0-C(=0)-, -NR3-C(=0)-NR3-, -0-C(=0)-NR3-, -NR3-C(=0)-0-, -0-, -S-, and -NR3- moieties of L, and

provided there is at least one -CH2- between each -C(=0)-, -C(=0)-NR3- -NR3- C(=0)-, -C(=0)-0-, -0-C(=0)-, -NR3-C(=0)-NR3-, -0-C(=0)-NR3-, -NR3-C(=0)-0-, -0-, -S- , and -NR.3- moiety of L;

Z-L is -CH2-L, -O-CH2-L, or -NR3-CH2-L; and

R1, R2, and R3 are each independently selected from H and alkyl.

2 The compound of claim 1, wherein the compound is of Formula la, lb, Ila, or lib:

lb

lib

or a pharmaceutically acceptable salt thereof.

3. The compound of claim 1 or 2, wherein X is phenyl or pyridyl.

4. The compound of claim 3, wherein X is phenyl.

5. The compound of claim 3, wherein X is pyridyl.

6. The compound of claim 1 or 2, wherein X is a bond.

7. The compound of any one of claims 1-6, wherein Z-L is -NH-CTb-L.

8. The compound of any one of claims 1-6, wherein Z-L is -O-CH2-L.

9. The compound of any one of claims 1-8, wherein L comprises 2-25 carbon atoms.

10. The compound of any one of claims 1-9, wherein L comprises 2 carbon atoms.

11. The compound of any one of claims 1-9, wherein L comprises 3 carbon atoms.

12. The compound of any one of claims 1-9, wherein L comprises 4 carbon atoms.

13. The compound of any one of claims 1-9, wherein L comprises 5 carbon atoms.

14. The compound of any one of claims 1-9, wherein L comprises 6 carbon atoms

15. The compound of any one of claims 1-9, wherein L comprises 7 carbon atoms

16. The compound of any one of claims 1-9, wherein L comprises 8 carbon atoms

17. The compound of any one of claims 1-9, wherein L comprises 9 carbon atoms

18. The compound of any one of claims 1-9, wherein L comprises 10 carbon atoms

19. The compound of any one of claims 1-9, wherein L comprises 11 carbon atoms

20 The compound of any one of claims 1-9, wherein L comprises 12 carbon atoms

21 The compound of any one of claims 1-9, wherein L comprises 13 carbon atoms

22 The compound of any one of claims 1-9, wherein L comprises 14 carbon atoms

23. The compound of any one of claims 1-9, wherein L comprises 15 carbon atoms

24. The compound of any one of claims 1-9, wherein L comprises 16 carbon atoms

25. The compound of any one of claims 1-9, wherein L comprises 17 carbon atoms

26. The compound of any one of claims 1-9, wherein L comprises 18 carbon atoms

27. The compound of any one of claims 1-9, wherein L comprises 19 carbon atoms

28. The compound of any one of claims 1-9, wherein L comprises 20 carbon atoms

29. The compound of any one of claims 1-9, wherein L comprises 21 carbon atoms.

30. The compound of any one of claims 1-9, wherein L comprises 22 carbon atoms.

31. The compound of any one of claims 1-9, wherein L comprises 23 carbon atoms.

32. The compound of any one of claims 1-9, wherein L comprises 24 carbon atoms.

33. The compound of any one of claims 1-9, wherein L comprises 25 carbon atoms.

34. The compound of any one of claims 1-33, wherein at least one, but no more than five,

O

-CH2- moieties of L are replaced with an amide moiety (e.g., H ).

35. The compound of any one of claims 1-34, wherein at least one -CH2- moiety of L is

replaced with an amide moiety (

36. The compound of any one of claims 1-34, wherein at least two -CH2- moieties of L

O

are replaced with two amide moieties (e.g., H ).

37. The compound of any one of claims 1-34, wherein at least three -CH2- moieties of L

O are replaced with three amide moieties (e.g.,

38. The compound of any one of claims 1-34, wherein one, two, three, or six -CH2-

O

moieties of L are replaced with one, two, three, or six amide moieties (e.g., H ).

39. The compound of any one of claims 34-38, wherein the amide moieties are separated by at least one carbon atom (e.g., CH2 units).

40. The compound of claim 39, wherein the amide moieties are separated by at least six carbon atoms (e.g., CH2 units).

41. The compound of any one of claims 34-40, wherein the carbon atom (e.g., C(=0) unit) of the amide is attached to Z.

42. The compound of any one of claims 1-41, wherein a carbon atom of L is attached to Z.

43. The compound of any one of claims 1-42, wherein at least one, but no more than ten, -CTh- moieties of L is replaced by an oxygen atom.

44. The compound of any one of claims 1-43, wherein at least one -CTh- moiety of L is replaced by an -0-.

45. The compound of any one of claims 1-43, wherein at least two -CTh- moieties of L are replaced by at least two -0-.

46. The compound of any one of claims 1-43, wherein at least six -CTh- moieties of L are replaced by at least six -0-.

47. The compound of any one of claims 1-43, wherein one, two, or six -CTh- moieties of L are replaced by -0-.

48. The compound of any one of claims 1-43, wherein the L comprises an ethylene glycol moiety, a diethylene glycol moiety, a triethylene glycol moeity, or an oligoethylene glycol moeity.

49. The compound of any one of claims 1-48, wherein at least one -CTh- moiety of L is replaced by -NR3-.

50. The compound of claim 49, wherein R3 is H.

51. The compound of any one of claims 1-50, wherein at least one -CH2- moiety of L is replaced by an -C(=0)-.

52. A compound selected from:

or a pharmaceutically acceptable salt thereof.

53. A pharmaceutical composition comprising a compound of any one of the preceding claims and a pharmaceutically acceptable carrier.

54. A method of degrading SMARCA2 or SMARCA4, comprising contacting a cell with a compound of any one of claims 1-52 or a pharmaceutically acceptable salt thereof.

55. A method of treating a disease or disorder, comprising administering to a subject in need thereof a compound of any one of claims 1-52.

56. A method of treating a disease or disorder that benefits from degradation of

SMARCA2 or SMARCA4, comprising administering to a subject in need thereof a compound of any one of claims 1-52.

57. The method of claim 56, wherein the disease or disorder benefits from the degradation of SMARCA2.

58. The method of claim 56, wherein the disease or disorder benefits from the degradation of SMARCA4.

59. The method of any one of claims 55-58, wherein the disease or disorder is cancer.

60. The method of claim 59, wherein the cancer is selected from synovial sarcoma, lung cancer, ovarian cancer, brain cancer, kidney cancer, leukemia, non-small cell lung cancer, Burkitf s Lymphoma, childhood medulloblastoma, pancreatic adenocarcinoma, ovarian clear cell carcinoma, renal cell carcinoma, endometrial carcinomas and melanoma.

61. The method of any one of claims 55-60, wherein the method further comprises conjointly administering one or more additional chemotherapeutic agents.

Description:
COMPOUNDS. COMPOSITIONS. AND METHODS FOR PROTEIN DEGRADATION

RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Patent Application No. 62/867,642, filed on June 27, 2019; the contents of which are hereby incorporated by reference in their entirety.

BACKGROUND

Targeted protein degradation is an emerging strategy to eliminate the function of a protein of interest. To date this process has been accomplished using ligands that can bind and recruit the ligase activity of cereblon (CRBN), von Hippel-Lindau tumor suppressor (VHL), mouse double minute 2 homolog (MDM2), or inhibitor of apoptosis (IAP) proteins.

SMARCA2 and SMARCA4 (also known as transcription activator Brahma-related gene 1 (BRG1) protein), are catalytic ATPase subunits of the S Witch/ Sucrose Non-fermentable (SWI/SNF) complex, also known the Brg/Brm-associated (BAF) complex. Together with core and regulatory subunits, SMARCA2 and SMARCA4 perform ATP hydrolysis that perturbs histone-DNA contacts. This sculpting of the nucleosomal landscape at promoters provides access to transcription factors and cognate DNA elements facilitating both gene activation and repression.

Many tumors express mutated versions of SMARCA2 and SMARCA4 and these mutations have been implicated in several types of cancer. For example, synoival sarcoma (SS) typically afflicts young adults and teenagers, in which tumors grow in the extremities, often close to joints. The standard of care is surgical removal of these tumors, often in conjunction with radiotherapy. However, these treatments can result in significant loss of function in the afflicted limb, resulting in a significant decrease in the subject’s quality of life.

SMARCA4 plays an important role in SS18-SSX, a fusion oncogene that presents in 95% of subjects with synovial sarcoma. SS18-SSX is formed from the fusion of synovial sarcoma translocation 18 (SSI 8) and synovial sarcoma breakpoint (SSX). SSI 8 is a component of the BAF complex, where the interaction of SS18-SSX with the BAF results in the loss of function in BAF47, a known tumor suppressor. This results in the activation of the Sox2 pathway, which is crucial for the proliferation of malignant SS cells. Degradation of SMARCA4 results in the disruption of the SS18-SSX/BAF complex, and accordingly, reduces in the proliferation of malignant cells, such as synovial sarcoma cells. Thus, the targeted degradation of SMARCA2 and SMARCA4 represents an attractive method for the inhibition of SS18-SSX and the treatment of synovial sarcoma. However, to date, there are no small molecule treatments that target SMARCA2 and SMARCA4 and are approved for use in humans.

SUMMARY

Disclosed herein are compounds that selectively degrade SMARCA2 and SMARCA4. Also disclosed herein are compositions and methods of use in treating associated disorders and diseases. These diseases include lung cancer, such as non-small cell lung cancer, Burkitt’s Lymphoma, childhood medulloblastoma, pancreatic adenocarcinoma, ovarian clear cell carcinoma, renal cell carcinoma, endometrial carcinomas and melanoma.

In certain aspects, the present disclosure provides compounds of Formula (I) and Formula (II):

II or a pharmaceutically acceptable salt thereof, wherein

X is a bond, aryl, or heteroaryl; L is an alkylene, alkenylene, or alkynylene chain comprising 1 to 35 carbon atoms, for example, 1 to 35 -CH2- moieties,

optionally wherein:

at least one, but no more than ten, -CH2- moieties of L are independently replaced with a moiety selected from -C(=0)-, -C(=0)-NR3- -NR3-C(=0)-, -C(=0)-0-, -O- C(=0)-, -NR 3 -C(=0)-NR 3 -, -0-C(=0)-NR 3 -, -NR 3 -C(=0)-0-, -0-, -S-, and -NR3-, provided the number of -CH2- moieties of L is larger than the collective number of -C(=0)-, -C(=0)-NR 3 - -NR 3 -C(=0)-, -C(=0)-0-, -0-C(=0)-, -NR 3 -C(=0)-NR 3 -, -0-C(=0)-NR 3 -, -NR 3 -C(=0)-0-, -0-, -S-, and -NR3- moieties of L, and

provided there is at least one -CH2- between each -C(=0)-, -C(=0)-NR 3 - -NR3- C(=0)-,

-C(=0)-0-, -0-C(=0)-, -NR 3 -C(=0)-NR 3 -, -0-C(=0)-NR 3 -, -NR 3 -C(=0)-0-, -0-, -S- , and -NR3- moiety of L;

Z-L is -CH2-L, O-CH2-L, or -NR 3 -CH 2 -L; and

R 1 , R 2 , and R 3 are each independently selected from H and alkyl.

In certain aspects, the present disclosure provides pharmaceutical compositions of comprising compounds of formula I or formula II and at least one pharmaceutically acceptable excipient.

In certain aspects, the present disclosure provides methods of treating cancer comprising of administering to a subject in need of a treatment for cancer, an amount of a compound of formula I or formula II.

BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 depicts the testing of El against a panel of synovial sarcoma cell lines. The compound has antiproliferation effect against synovial sarcoma lines.

FIG. 2 depicts the activity of exemplary compounds of the disclosure against SMARCA4.

FIG. 3 depicts the binding of exemplary compounds of the disclosure against CRBN. FIG. 4A depicts the CRBN binding assay of compounds El to E13 in the AlphaScreen Assay using CRBN and biotinylated pomolidimide as binding partners with dose response in 200 mM NaCl, 50 mM Tris pH 7.5, 0.1% Pluronic acid, 1 mM TCEP.

FIG. 4B depicts the CRBN binding assay of compounds E7, E8, E9, E10, Ell, and E13 in 200 mM NaCl, 50 mM Tris pH 7.5, 0.1% Pluronic acid, 1 mM TCEP. FIG. 4C depicts the CRBN binding assay of compound E12 in 200 mM NaCl, 50 mM Tris pH 7.5, 0.1% Pluronic acid, 1 mM TCEP.

FIG. 4D depicts a CRBN binding assay of Lenalidomide in 200 mM NaCl, 50 mM Tris pH 7.5, 0.1% Pluronic acid, 1 mM TCEP.

FIG. 5A depicts the activity of exemplary compounds of the disclosure against SMARCA4 after 20 hours in an ATP lite assay, relative to DMSO.

FIG. 5B depicts the activity of exemplary compounds of the disclosure against SMARCA4 after 24 hours in an ATP lite assay, relative to DMSO.

FIG. 5C depicts the activity of exemplary compounds of the disclosure against SMARCA4 after 48 hours in an ATP lite assay, relative to DMSO.

FIG. 6 depicts the activity of exemplary compounds of the disclosure against SMARCA4 after 24 hours in a CCK assay, relative to DMSO.

FIG. 7 depicts the activity of one dose of exemplary compounds of the disclosure against SMARCA4 after 48 hours, relative to DMSO.

FIG. 8 depicts the activity of two doses of exemplary compounds of the disclosure against SMARCA4 after 48 hours, relative to DMSO.

FIG. 9 depicts the activity of exemplary compounds of the disclosure against SMARCA4 after 72 hours in an ATP lite assay, relative to DMSO.

FIG. 10 depicts the binding of exemplary compounds of the disclosure against SMARCA4, relative to DMSO.

FIG. 11 depicts the binding of exemplary compounds of the disclosure against CRBN, relative to DMSO.

FIG. 12 depicts the binding of exemplary compounds of the disclosure against SMARCA4, relative to DMSO.

FIG. 13 depicts the binding of exemplary compounds of the disclosure against SMARCA4, relative to DMSO.

FIG. 14 depicts the binding of exemplary compounds of the disclosure against CRBN, relative to DMSO.

FIG. 15 depicts the activity of exemplary compounds of the disclosure against a RN2 cell line after 72 hours, relative to DMSO.

FIG. 16 depicts the activity of exemplary compounds of the disclosure against a MV411 cell line after 72 hours, relative to DMSO. FIG. 17 depicts the binding of exemplary compounds of the disclosure against SMARCA4, relative to DMSO.

FIG. 18 depicts the binding of exemplary compounds of the disclosure against CRBN, relative to DMSO.

FIG. 19 depicts the activity of exemplary compounds of the disclosure against a MV411 cell line after 72 hours, relative to DMSO.

FIG. 20 depicts the activity of exemplary compounds of the disclosure against a A549 cell line after 72 hours, relative to DMSO.

FIG. 21A depicts the activity of exemplary compounds of the disclosure against a SMARCA4 dependent cell line, relative to DMSO.

FIG. 21B depicts the activity of exemplary compounds of the disclosure against a SMARCA4 independent cell line, relative to DMSO.

FIG. 22 depicts the activity of exemplary compounds of the disclosure against a MV411 cell line after 72 hours, relative to DMSO.

FIG. 23 depicts the activity of exemplary compounds of the disclosure against a A549 cell line after 72 hours, relative to DMSO.

FIG. 24 depicts the binding of exemplary compounds of the disclosure against SMARCA4 relative to DMSO.

FIG. 25 depicts the binding of exemplary compounds of the disclosure against CRBN, relative to DMSO.

FIG. 26 depicts the binding of exemplary compounds of the disclosure against a RN2 cell line after 72 hours, relative to DMSO.

FIG. 27 depicts the binding of exemplary compounds of the disclosure against a MV411 cell line after 72 hours, relative to DMSO.

FIG. 28A depicts the activity of exemplary compounds of the disclosure against a SMAR.CA4 dependent cell line, relative to DMSO.

FIG. 28B depicts the activity of exemplary compounds of the disclosure against a SMAR.CA4 dependent cell line, relative to DMSO.

FIG. 29A depicts the binding of exemplary compounds of the disclosure against a MV411 wild type cell line after 24 hours, relative to DMSO.

FIG. 29B depicts the binding of exemplary compounds of the disclosure against a MV411 CRBN knock out cell line after 24 hours, relative to DMSO. FIG. 30A depicts the binding of exemplary compounds of the disclosure against a NOMOl cell line after 24 hours, relative to DMSO.

FIG. 30B depicts the binding of exemplary compounds of the disclosure against a THP1 cell line after 24 hours, relative to DMSO.

FIG. 31A depicts the binding of exemplary compounds of the disclosure against a MV411 wild type cell line after 72 hours, relative to DMSO.

FIG. 31B depicts the binding of exemplary compounds of the disclosure against a MV411 CRBN knock out cell line after 72 hours, relative to DMSO.

FIG. 32A depicts the binding of exemplary compounds of the disclosure against a NOMOl cell line after 72 hours, relative to DMSO.

FIG. 32B depicts the binding of exemplary compounds of the disclosure against a THP1 cell line after 72 hours, relative to DMSO.

FIG. 33A depicts the binding of exemplary compounds of the disclosure against a OCIAML2 cell line after 72 hours, relative to DMSO.

FIG. 33B depicts the binding of exemplary compounds of the disclosure against a OCIAML3 cell line after 72 hours, relative to DMSO.

FIG. 34 depicts the binding of exemplary compounds of the disclosure against SMARCA4, relative to DMSO.

PET ATT, ED DESCRIPTION

Overview

Ubiquitination is a post-translational modification of proteins that is critical to many cellular processes, including protein degradation by the proteasome, cell cycle progression, transcriptional regulation, DNA repair and signal transduction. Ubiquitination requires the sequential action of three enzymes. El, or ubiquitin-activating enzyme, catalyzes the ATP- dependent activation of ubiquitin and formation of a thioester bond between the ubiquitin C terminus and the catalytic cysteine on the El. Ubiquitin is then transferred to a catalytic cysteine of one of the ~40 E2s (ubiquitin-conjugating enzymes) and through the E3 (ubiquitin ligase) to the substrate. CRBN interacts with the DNA damage-binding protein-1 (DDB1), Cullin 4 (Cul4A or Cul4B), and regulator of Cullins 1 (RoCl) to form the functional E3 ubiquitin ligase complex. In this complex, CRBN functions as a substrate receptor of E3 ubiquitin ligase complex and targets proteins for proteolysis through a ubiquitin-proteasome pathway. The SMARCA2 and SMARCA4 degraders described herein are a group of synthetic molecules designed to recruit a specific ubiquitin ligase (e.g., cereblon) to a chosen target protein (e.g., SMARCA2 or SMARCA4). These degraders act to bring the target protein and the ligase into close proximity to enable facile degradation through the ubiquitination process.

In certain aspects, the degraders are comprised of two“hooks” linked by a chemical linker. The first hook is a ligase-recruiting ligand (e.g., Lenalidomide) whilst the second is a ligand (e.g., PFI-3) that binds the target protein (e.g., SMARCA2 or SMARCA4).

In certain aspects, the disclosed compounds target SMARCA2 and SMARCA4 proteins for degradation utilizing the ubiquitination E3 ligase, Cereblon.

Compounds

In certain aspects, the present disclosure provides compounds of Formula (I) and Formula (II):

II

or a pharmaceutically acceptable salt thereof, wherein

X is a bond, aryl, or heteroaryl; L is an alkylene, alkenylene, or alkynylene chain comprising 1 to 35 carbon atoms, for example, 1 to 35 -CH2- moieties,

optionally wherein:

at least one, but no more than ten, -CH2- moieties of L are independently replaced with a moiety selected from -C(=0)-, -C(=0)-NR3- -NR3-C(=0)-, -C(=0)-0-, -O- C(=0)-, -NR3-C(=0)-NR 3 -, -0-C(=0)-NR 3 -, -NR 3 -C(=0)-0-, -0-, -S-, and -NR3, provided the number of -CH2- moieties of L is larger than the collective number of -C(=0)-, -C(=0)-NR3- -NR3-C(=0)-, -C(=0)-0-, -0-C(=0)-, -NR3-C(=0)-NR 3 -, -0-C(=0)-NR3-, -NR3-C(=0)-0-, -0-, -S-, and -NR3- moieties of L, and

provided there is at least one -CH2- between each -C(=0)-, -C(=0)-NR3- -NR3- C(=0)-,

-C(=0)-0-, -0-C(=0)-, -NR3-C(=0)-NR 3 -, -0-C(=0)-NR 3 -, -NR3-C(=0)-0-, -0-, -S- , and -NR3- moiety of L;

Z-L is -CH2-L, -O-CH2-L, or -NR 3 -CH 2 -L; and

R 1 , R 2 , and R 3 are each independently selected from H and alkyl.

In certain embodiments, the compound of Formula I is a compound of Formula la or lb, or the compound of Formula II is a compound of Formula Ila or lib:

la

lib

or a pharmaceutically acceptable salt thereof.

In certain embodiments, X is aryl or heteroaryl, for example, X is phenyl or pyridyl. In certain embodiments, X is phenyl, for example, p-phenyl, preferably p-phenyl with no additional substituents. In other embodiments, X is pyridyl, for example, 3,6-pyridyl, preferably 3,6-pyridyl with no additional substituents. In yet other embodiments, X is a bond.

In certain embodiments, Z-L is -NH-CH2-L. In other embodiments, Z-L is -O-CH2-L.

In certain embodiments, L is an alkylene, alkenylene, or alkynylene chain comprising 3 to 35 carbon atoms, for example, 13-25 carbon atoms. In certain embodiments, L comprises 2 carbon atoms. In other embodiments, L comprises 3 carbon atoms. In other embodiments, L comprises 4 carbon atoms. In other embodiments, L comprises 5 carbon atoms. In yet other embodiments, L comprises 6 carbon atoms. In yet other embodiments, L comprises 7 carbon atoms. In yet other embodiments, L comprises 8 carbon atoms. In yet other embodiments, L comprises 9 carbon atoms. In yet other embodiments, L comprises 10 carbon atoms. In yet other embodiments, L comprises 11 carbon atoms. In yet other embodiments, L comprises 12 carbon atoms. In yet other embodiments, L comprises 13 carbon atoms. In yet other embodiments, L comprises 14 carbon atoms. In yet other embodiments, L comprises 15 carbon atoms. In yet other embodiments, L comprises 16 carbon atoms. In yet other embodiments, L comprises 17 carbon atoms. In yet other embodiments, L comprises 18 carbon atoms. In yet other embodiments, L comprises 19 carbon atoms. In yet other embodiments, L comprises 20 carbon atoms. In yet other embodiments, L comprises 21 carbon atoms. In yet other embodiments, L comprises 22 carbon atoms. In yet other embodiments, L comprises 23 carbon atoms. In yet other embodiments, L comprises 24 carbon atoms. In yet other embodiments, L comprises 25 carbon atoms.

In certain embodiments, the invention relates to any one of the compounds described herein, wherein L comprises 1 to 35 -CH2- moieties, optionally wherein at least one, but no more than ten, -CH2- moieties of L are independently replaced with a moiety selected from - C(=0)-,

-C(=0)-NR 3 - -NR 3 -C(=0)-, -C(=0)-0-, -0-C(=0)-, -NR 3 -C(=0)-NR 3 -, -0-C(=0)-NR 3 -, - NR 3 -C(=0)-0-, -0-, -S-, and -NR3-.

In certain embodiments, the invention relates to any one of the compounds described herein, wherein at least one, but no more than ten, -CH2- moieties of L are independently replaced with a moiety selected from -C(=0)-, -C(=0)-NR 3 - -NR 3 -C(=0)-, -C(=0)-0-, -O- C(=0)-, -NR 3 -C(=0)-NR 3 -, -0-C(=0)-NR 3 -, -NR 3 -C(=0)-0-, -O-, -S-, and -NR 3 -.

In certain preferred embodiments, at least one, but no more than five, -CH2- moieties

O

of L are replaced with an amide moiety (e.g., H ). in certain embodiments, at least one

O

-CH2- moiety of L is replaced with an amide moiety (e.g., H ). in certain embodiments,

O

at least two -CH2- moieties of L are replaced with two amide moieties (e.g., H ). i n certain embodiments, at least three -CH2- moieties of L are replaced with three amide

O moieties (e.g., In certain embodiments, one, two, three, or six -CH2- moieties of L

O

are replaced with one, two, three, or six amide moieties (e.g., H ). i n certain

embodiments, the amide moieties are separated by at least one carbon atom (e.g., CH2 units). In certain embodiments, the amide moieties are separated by at least six carbon atoms (e.g., CH2 units). In certain embodiments, the carbon atom (e.g., C(=0)unit) of the amide is attached to Z.

In certain embodiments, at least one, but no more than ten, -CH2- moieties of L are replaced by at least one, but no more than ten -0-. In certain embodiments, at least one -CH2- moiety of L is replaced by -0-. In certain embodiments, at least two -CH2- moieties of L are replaced by at least two -0-. In certain embodiments, at least six -CH2- moieties of L are replaced by at least six -0-. In certain embodiments, one, two, or six methylene moieties of L are replaced by -0-. In certain embodiments, L comprises an ethylene glycol moiety, a diethylene glycol moiety, a triethylene glycol moeity, or an oligoethylene glycol moeity, for example, a diethylene glycol moiety.

In certain embodiments, at least one -CH2- moiety of L is replaced by -NR3-. In certain embodiments, R 3 is H.

In certain embodiments, at least one -CH2- moiety of L is replaced by an -C(=0)-.

In certain embodiments, R 1 is H.

In certain embodiments, R 2 is H.

In certain embodiments, the invention relates to a compound of Formula (I). In other embodiments, the invention relates to a compound of Formula (II).

In certain embodiments, a compound of Formula (I) or Formula (II) is selected from:

or a pharmaceutically acceptable salt thereof.

In certain embodiments, disclosed herein are pharmaceutical compositions, comprising a compound of Formula (I) or Formula (II), and one or more pharmaceutically acceptable excipients. In certain embodiments, the pharmaceutical compositions may be used in treating or preventing a condition or disease as described herein.

Methods of Use

In one aspect, the present disclosure provides methods of degrading SMARCA2 or SMARCA4, comprising contacting a cell with a compound of the disclosure or a

pharmaceutically acceptable salt thereof.

In another aspect, the present disclosure provides methods of treating a disease or disorder, comprising administering to a subject in need thereof a compound of the disclosure. In certain embodiments, the disease or disorder is cancer. In certain embodiments, the cancer is selected from synovial sarcoma, lung cancer, ovarian cancer, brain cancer, kidney cancer, leukemia, non-small cell lung cancer, Burkitf s Lymphoma, childhood medulloblastoma, pancreatic adenocarcinoma, ovarian clear cell carcinoma, renal cell carcinoma, endometrial carcinomas and melanoma.

In yet another aspect, the present disclosure provides methods of treating a disease or disorder that benefits from degradation of SMARCA2 or SMARCA4, comprising

administering to a subject in need thereof a compound of the disclosure. In certain

embodiments, the disease or disorder benefits from the degradation of SMARCA2. In certain embodiments, the disease or disorder benefits from the degradation of SMARCA4. In certain embodiments, the disease or disorder is cancer. In certain embodiments, the cancer is selected from synovial sarcoma, lung cancer, ovarian cancer, brain cancer, kidney cancer, leukemia, non-small cell lung cancer, Burkitf s Lymphoma, childhood medulloblastoma, pancreatic adenocarcinoma, ovarian clear cell carcinoma, renal cell carcinoma, endometrial carcinomas and melanoma.

In certain embodiments, the methods disclosed herein further comprise conjointly administering one or more additional chemotherapeutic agents.

Definitions

Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art of the present disclosure. The following references provide one of skill with a general definition of many of the terms used in this disclosure: Singleton et ah, Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise.

In this disclosure, "comprises," "comprising," "containing" and "having" and the like can have the meaning ascribed to them in U.S. Patent law and can mean " includes,"

"including," and the like; "consisting essentially of or "consists essentially" likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.

Unless specifically stated or obvious from context, as used herein, the term "or" is understood to be inclusive. Unless specifically stated or obvious from context, as used herein, the terms "a", "an", and "the" are understood to be singular or plural.

The term "and/or" is used in this disclosure to mean either "and" or "or" unless indicated otherwise.

It is understood that substituents and substitution patterns on the compounds of the present invention can be selected by one of ordinary skilled person in the art to result chemically stable compounds which can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.

As used herein, the term“substituted” refers to the replacement of one to six hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: hydroxyl, hydroxyalkyl, alkoxy, halogen, alkyl, nitro, silyl, acyl, acyloxy, aryl, cycloalkyl, heterocyclyl, amino, aminoalkyl, cyano, haloalkyl, haloalkoxy, -OCO-CH2-O- alkyl, -0P(0)(0-alkyl)2 or -CH2-0P(0)(0-alkyl)2. Preferably,“substituted” refers to the replacement of one to four hydrogen radicals in a given structure with the substituents mentioned above. More preferably, one to three hydrogen radicals are replaced by the substituents as mentioned above. It is understood that the substituent can be further substituted.

The term“acyl” is art-recognized and refers to a group represented by the general formula hydrocarbylC(O)-, preferably alkylC(O)-.

The term“acylamino” is art-recognized and refers to an amino group substituted with an acyl group and may be represented, for example, by the formula hydrocarbylC(0)NH-.

The term“acyloxy” is art-recognized and refers to a group represented by the general formula hydrocarbylC(0)0-, preferably alkylC(0)0-.

The term“alkoxy” refers to an alkyl group, preferably a lower alkyl group, having an oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.

The term“alkoxyalkyl” refers to an alkyl group substituted with an alkoxy group and may be represented by the general formula alkyl-O-alkyl.

The term“alkenyl”, as used herein, refers to an aliphatic group containing at least one double bond and is intended to include both "unsubstituted alkenyls" and "substituted alkenyls", the latter of which refers to alkenyl moieties having substituents replacing a hydrogen on one or more carbons of the alkenyl group. Such substituents may occur on one or more carbons that are included or not included in one or more double bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed below, except where stability is prohibitive. For example, substitution of alkenyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl groups is contemplated. An“alkyl” group or“alkane” is a straight chained or branched non-aromatic hydrocarbon which is completely saturated. Typically, a straight chained or branched alkyl group has from 1 to about 20 carbon atoms, preferably from 1 to about 10 unless otherwise defined. Examples of straight chained and branched alkyl groups include methyl, ethyl, n- propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, pentyl and octyl. A C1-C 6 straight chained or branched alkyl group is also referred to as a "lower alkyl" group.

Moreover, the term "alkyl" (or "lower alkyl") as used throughout the specification, examples, and claims is intended to include both "unsubstituted alkyls" and "substituted alkyls", the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents, if not otherwise specified, can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. For instance, the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), -CF3, -CN and the like. Exemplary substituted alkyls are described below. Cycloalkyls can be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl- substituted alkyls, -CF3, -CN, and the like.

The term“Cx- y ” when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy is meant to include groups that contain from x to y carbons in the chain. For example, the term“Cx- y alkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups such as

trifluoromethyl and 2,2,2-tirfluoroethyl, etc. Co alkyl indicates a hydrogen where the group is in a terminal position, a bond if internal. The terms“C2- y alkenyl” and“C2- y alkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.

The term“alkylamino”, as used herein, refers to an amino group substituted with at least one alkyl group.

The term“alkylthio”, as used herein, refers to a thiol group substituted with an alkyl group and may be represented by the general formula alkylS-.

The term“alkynyl”, as used herein, refers to an aliphatic group containing at least one triple bond and is intended to include both "unsubstituted alkynyls" and "substituted alkynyls", the latter of which refers to alkynyl moieties having substituents replacing a hydrogen on one or more carbons of the alkynyl group. Such substituents may occur on one or more carbons that are included or not included in one or more triple bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed above, except where stability is prohibitive. For example, substitution of alkynyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl groups is contemplated.

The term“amide”, as used herein, refers to a group

wherein each R 10 independently represents a hydrogen or hydrocarbyl group, or two R 10 are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure.

The terms“amine” and“amino” are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by wherein each R 10 independently represents a hydrogen or a hydrocarbyl group, or two R 10 are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure. The term“aminoalkyl”, as used herein, refers to an alkyl group substituted with an amino group.

The term“aralkyl”, as used herein, refers to an alkyl group substituted with an aryl group. The term“aryl” as used herein include substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon. Preferably, the ring is a 5- to 7- membered ring, more preferably a 6-membered ring. The term“aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.

Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like.

The term“carbamate” is art-recognized and refers to a group wherein R 9 and R 10 independently represent hydrogen or a hydrocarbyl group, such as an alkyl group, or R 9 and R 10 taken together with the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure.

The terms“carbocycle”, and“carbocyclic”, as used herein, refers to a saturated or unsaturated ring in which each atom of the ring is carbon. The term carbocycle includes both aromatic carbocycles and non-aromatic carbocycles. Non-aromatic carbocycles include both cycloalkane rings, in which all carbon atoms are saturated, and cycloalkene rings, which contain at least one double bond.

The term“carbocycle” includes 5-7 membered monocyclic and 8-12 membered bicyclic rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated and aromatic rings. Carbocycle includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings. The term“fused carbocycle” refers to a bicyclic carbocycle in which each of the rings shares two adjacent atoms with the other ring. Each ring of a fused carbocycle may be selected from saturated, unsaturated and aromatic rings. In an exemplary embodiment, an aromatic ring, e.g., phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, is included in the definition of carbocyclic. Exemplary“carbocycles” include cyclopentane, cyclohexane, bicyclo[2.2.1]heptane, 1,5-cyclooctadiene, 1,2,3,4-tetrahydronaphthalene, bicyclo[4.2.0]oct-3-ene, naphthalene and adamantane. Exemplary fused carbocycles include decalin, naphthalene, 1,2,3,4-tetrahydronaphthalene, bicyclo[4.2.0]octane, 4,5,6,7-tetrahydro- lH-indene and bicyclo[4.1.0]hept-3-ene.“Carbocycles” may be susbstituted at any one or more positions capable of bearing a hydrogen atom.

A“cycloalkyl” group is a cyclic hydrocarbon which is completely saturated.

“Cycloalkyl” includes monocyclic and bicyclic rings. Typically, a monocyclic cycloalkyl group has from 3 to about 10 carbon atoms, more typically 3 to 8 carbon atoms unless otherwise defined. The second ring of a bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. Cycloalkyl includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings. The term“fused cycloalkyl” refers to a bicyclic cycloalkyl in which each of the rings shares two adjacent atoms with the other ring. The second ring of a fused bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. A“cycloalkenyl” group is a cyclic hydrocarbon containing one or more double bonds.

The term“carbocyclylalkyl”, as used herein, refers to an alkyl group substituted with a carbocycle group.

The term“carbonate” is art-recognized and refers to a group -OCO2-R 10 , wherein R 10 represents a hydrocarbyl group.

The term“carboxy”, as used herein, refers to a group represented by the

formula -CO2H.

The term“ester”, as used herein, refers to a group -C(0)OR 10 wherein R 10 represents a hydrocarbyl group.

The term“ether”, as used herein, refers to a hydrocarbyl group linked through an oxygen to another hydrocarbyl group. Accordingly, an ether substituent of a hydrocarbyl group may be hydrocarbyl-O-. Ethers may be either symmetrical or unsymmetrical.

Examples of ethers include, but are not limited to, heterocycle-O-heterocycle and aryl-O- heterocycle. Ethers include“alkoxyalkyl” groups, which may be represented by the general formula alkyl-O-alkyl.

The terms“halo” and“halogen” as used herein means halogen and includes chloro, fluoro, bromo, and iodo.

The terms“hetaralkyl” and“heteroaralkyl”, as used herein, refers to an alkyl group substituted with a hetaryl group.

The term "heteroalkyl", as used herein, refers to a saturated or unsaturated chain of carbon atoms and at least one heteroatom, wherein no two heteroatoms are adjacent. The terms“heteroaryl” and“hetaryl” include substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The terms“heteroaryl” and“hetaryl” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is

heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.

The term“heteroatom” as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur.

The terms“heterocyclyl”,“heterocycle”, and“heterocyclic” refer to substituted or unsubstituted non-aromatic ring structures, preferably 3- to 10-membered rings, more preferably 3- to 7-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The terms “heterocyclyl” and“heterocyclic” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Heterocyclyl groups include, for example, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like.

The term“heterocyclylalkyl”, as used herein, refers to an alkyl group substituted with a heterocycle group.

The term“hydrocarbyl”, as used herein, refers to a group that is bonded through a carbon atom that does not have a =0 or =S substituent, and typically has at least one carbon- hydrogen bond and a primarily carbon backbone, but may optionally include heteroatoms. Thus, groups like methyl, ethoxyethyl, 2-pyridyl, and trifluoromethyl are considered to be hydrocarbyl for the purposes of this application, but substituents such as acetyl (which has a =0 substituent on the linking carbon) and ethoxy (which is linked through oxygen, not carbon) are not. Hydrocarbyl groups include, but are not limited to aryl, heteroaryl, carbocycle, heterocyclyl, alkyl, alkenyl, alkynyl, and combinations thereof. The term“hydroxyalkyl”, as used herein, refers to an alkyl group substituted with a hydroxy group.

The term“lower” when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy is meant to include groups where there are ten or fewer non-hydrogen atoms in the substituent, preferably six or fewer. A“lower alkyl”, for example, refers to an alkyl group that contains ten or fewer carbon atoms, preferably six or fewer. In certain embodiments, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy substituents defined herein are respectively lower acyl, lower acyloxy, lower alkyl, lower alkenyl, lower alkynyl, or lower alkoxy, whether they appear alone or in combination with other

substituents, such as in the recitations hydroxyalkyl and aralkyl (in which case, for example, the atoms within the aryl group are not counted when counting the carbon atoms in the alkyl substituent).

The terms“polycyclyl”,“polycycle”, and“polycyclic” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls) in which two or more atoms are common to two adjoining rings, e.g., the rings are“fused rings”. Each of the rings of the polycycle can be substituted or unsubstituted. In certain embodiments, each ring of the poly cycle contains from 3 to 10 atoms in the ring, preferably from 5 to 7.

The term“silyl” refers to a silicon moiety with three hydrocarbyl moieties attached thereto.

The term“substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that“substitution” or“substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by

rearrangement, cyclization, elimination, etc. As used herein, the term“substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as“unsubstituted,” references to chemical moieties herein are understood to include substituted variants. For example, reference to an“aryl” group or moiety implicitly includes both substituted and unsubstituted variants.

The term“sulfate” is art-recognized and refers to the group -OSCbH, or a

pharmaceutically acceptable salt thereof.

The term“sulfonamide” is art-recognized and refers to the group represented by the general formulae

wherein R 9 and R 10 independently represents hydrogen or hydrocarbyl, such as alkyl, or R 9 and R 10 taken together with the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure.

The term“sulfoxide” is art-recognized and refers to the group -S(0)-R 10 , wherein R 10 represents a hydrocarbyl.

The term“sulfonate” is art-recognized and refers to the group SCbH, or a

pharmaceutically acceptable salt thereof.

The term“sulfone” is art-recognized and refers to the group -S(0)2-R 10 , wherein R 10 represents a hydrocarbyl.

The term“thioalkyl”, as used herein, refers to an alkyl group substituted with a thiol group.

The term“thioester”, as used herein, refers to a group -C(0)SR 10 or -SC(0)R 10 wherein R 10 represents a hydrocarbyl.

The term“thioether”, as used herein, is equivalent to an ether, wherein the oxygen is replaced with a sulfur. The term“urea” is art-recognized and may be represented by the general Formula

wherein R 9 and R 10 independently represent hydrogen or a hydrocarbyl, such as alkyl, or either occurrence of R 9 taken together with R 10 and the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure.

The term“protecting group” refers to a group of atoms that, when attached to a reactive functional group in a molecule, mask, reduce or prevent the reactivity of the functional group. Typically, a protecting group may be selectively removed as desired during the course of a synthesis. Examples of protecting groups can be found in Greene and Wuts, Protective Groups in Organic Chemistry, 3 rd Ed., 1999, John Wiley & Sons, NY and Harrison et ah, Compendium of Synthetic Organic Methods, Vols. 1-8, 1971-1996, John Wiley & Sons, NY. Representative nitrogen protecting groups include, but are not limited to, formyl, acetyl, trifluoroacetyl, benzyl, benzyloxycarbonyl (“CBZ”), tert-butoxycarbonyl (“Boc”), trimethyl silyl (“TMS”), 2-trimethylsilyl-ethanesulfonyl (“TES”), trityl and substituted trityl groups, allyloxycarbonyl, 9-fluorenylmethyloxycarbonyl (“FMOC”), nitro- veratryloxycarbonyl (“NVOC”) and the like. Representative hydroxyl protecting groups include, but are not limited to, those where the hydroxyl group is either acylated (esterified) or alkylated such as benzyl and trityl ethers, as well as alkyl ethers, tetrahydropyranyl ethers, trialkylsilyl ethers (e.g., TMS or TIPS groups), glycol ethers, such as ethylene glycol and propylene glycol derivatives and allyl ethers.

The term“prodrug” is intended to encompass compounds which, under physiologic conditions, are converted into the therapeutically active agents of the present invention (e.g., a compound of Formula (I) or Formula (II)). A common method for making a prodrug is to include one or more selected moieties which are hydrolyzed under physiologic conditions to reveal the desired molecule. In other embodiments, the prodrug is converted by an enzymatic activity of the subject. For example, esters or carbonates (e.g., esters or carbonates of alcohols or carboxylic acids) are preferred prodrugs of the present invention. In certain embodiments, some or all of the compounds of Formula (I) or Formula (II) in a formulation represented above can be replaced with the corresponding suitable prodrug, e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate or carboxylic acid present in the parent compound is presented as an ester. The present invention includes all pharmaceutically acceptable isotopically-labelled compounds as described herein wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. In certain embodiments, compounds of the invention are enriched in such isotopically labeled substances (e.g., compounds wherein the distribution of isotopes in the compounds in the composition differ from a natural or typical distribution of isotopes).

Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H carbon, such as U C, 13 C and 14 C, chlorine, such as 36 C1, fluorine, such as 18 F, iodine, such as 123 I and 125 I, nitrogen, such as 13 N and 15 N, oxygen, such as 15 0, 17 0 and 18 0, phosphorus, such as 32 P, and sulphur, such as 35 S.

Certain isotopically-labelled compounds as disclosed herein, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are useful for this purpose in view of their ease of incorporation and ready means of detection.

Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.

Substitution with positron-emitting isotopes, such as U C, 18 F, 15 0 and 13 N, can be useful in Positron Emission Tomography (PET) studies for examining substrate receptor occupancy.

Compounds of the invention can have one or more asymmetric carbon atoms and can exist in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric race mates or mixtures of diastereoisomeric racemates. The optically active forms can be obtained for example by resolution of the racemates, by asymmetric synthesis or asymmetric chromatography (chromatography with a chiral adsorbents or eluant). That is, certain of the disclosed compounds may exist in various stereoisomeric forms.

Stereoisomers are compounds that differ only in their spatial arrangement.

Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. "Enantiomer" means one of a pair of molecules that are mirror images of each other and are not superimposable.“Diastereomers” are stereoisomers that are not related as mirror images, most commonly because they contain two or more asymmetrically substituted carbon atoms and represent the configuration of substituents around one or more chiral carbon atoms. Enantiomers of a compound can be prepared, for example, by separating an enantiomer from a racemate using one or more well-known techniques and methods, such as, for example, chiral chromatography and separation methods based thereon. The appropriate technique and/or method for separating an enantiomer of a compound described herein from a racemic mixture can be readily determined by those of skill in the art.

“Geometric isomer" means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon- carbon double bond may be in an E (substituents are on opposite sides of the carbon- carbon double bond) or Z (substituents are oriented on the same side) configuration. "R," "S," "S*," "R*," "E," "Z," "cis," and "trans," indicate configurations relative to the core molecule. Certain of the disclosed compounds may exist in atropisomeric forms. Atropisomers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers. The compounds of the invention may be prepared as individual isomers by either isomer-specific synthesis or resolved from an isomeric mixture. Conventional resolution techniques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide of each of the isomers of an isomeric pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chromatographic methods.

Diastereomeric purity by weight is the ratio of the weight of one diastereomer or over the weight of all the diastereomers. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by weight relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by weight optically pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by weight pure. Percent optical purity is the ratio of the weight of the enantiomer or over the weight of the enantiomer plus the weight of its optical isomer.

Percent purity by mole fraction is the ratio of the moles of the enantiomer (or diastereomer) or over the moles of the enantiomer (or diastereomer) plus the moles of its optical isomer. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure relative to the other

stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure.

When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has at least one chiral center, it is to be understood that the name or structure encompasses either enantiomer of the compound free from the corresponding optical isomer, a racemic mixture of the compound or mixtures enriched in one enantiomer relative to its corresponding optical isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry and has two or more chiral centers, it is to be understood that the name or structure encompasses a diastereomer free of other diastereomers, a number of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other diastereomer(s) or mixtures of diastereomers in which one or more diastereomer is enriched relative to the other

diastereomers. The invention embraces all of these forms.

As used herein, the term "pharmaceutically acceptable salt" means any

pharmaceutically acceptable salt of the compound of Formula (I). For example,

pharmaceutically acceptable salts of any of the compounds described herein include those that are within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66: 1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting a free base group with a suitable organic acid.

The compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases and methods for preparation of the appropriate salts are well-known in the art. Salts may be prepared from pharmaceutically acceptable non-toxic acids and bases including inorganic and organic acids and bases.

Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate,

camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate,

ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, and valerate salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.

The term "subject" to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys); mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, goats, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, and/or turkeys. Preferred subjects are humans. As used herein, a therapeutic that“prevents” a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset or reduces the severity of one or more symptoms of the disorder or condition relative to the untreated control sample.

In“treatment,” the object is to prevent or slow down (lessen) an undesired

physiological condition, disorder, or disease, or obtain beneficial or desired clinical results. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.

Pharmaceutical Compositions

The compositions and methods of the present invention may be utilized to treat a subject in need thereof. In certain embodiments, the subject is a mammal such as a human, or a non-human mammal. When administered to subject, such as a human, the composition or the compound is preferably administered as a pharmaceutical composition comprising, for example, a compound of the invention and a pharmaceutically acceptable carrier.

Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, oils such as olive oil, or injectable organic esters. In a preferred embodiment, when such pharmaceutical compositions are for human

administration, particularly for invasive routes of administration (i.e., routes, such as injection or implantation, that circumvent transport or diffusion through an epithelial barrier), the aqueous solution is pyrogen-free, or substantially pyrogen-free. The excipients can be chosen, for example, to effect delayed release of an agent or to selectively target one or more cells, tissues or organs. The pharmaceutical composition can be in dosage unit form such as tablet, capsule (including sprinkle capsule and gelatin capsule), granule, lyophile for reconstitution, powder, solution, syrup, suppository, injection or the like. The composition can also be present in a transdermal delivery system, e.g., a skin patch. The composition can also be present in a solution suitable for topical administration, such as an eye drop.

A pharmaceutically acceptable excipient can contain physiologically acceptable agents that act, for example, to stabilize, increase solubility or to increase the absorption of a compound such as a compound of the invention. Such physiologically acceptable agents include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients. The choice of a pharmaceutically acceptable excipient, including a physiologically acceptable agent, depends, for example, on the route of administration of the composition. The preparation or pharmaceutical composition can be a self-emulsifying drug delivery system or a self-microemulsifying drug delivery system. The pharmaceutical composition (preparation) also can be a liposome or other polymer matrix, which can have incorporated therein, for example, a compound of the invention. Liposomes, for example, which comprise phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject without excessive toxicity, irritation, allergic response, or other problem or complication,

commensurate with a reasonable benefit/risk ratio.

The phrase "pharmaceutically acceptable excipient" as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each excipient must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.

A pharmaceutical composition (preparation) can be administered to a subject by any of a number of routes of administration including, for example, orally (for example, drenches as in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including sprinkle capsules and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (for example, as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (for example as a patch applied to the skin); and topically (for example, as a cream, ointment or spray applied to the skin, or as an eye drop). The compound may also be Formulated for inhalation. In certain embodiments, a compound may be simply dissolved or suspended in sterile water. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973, 5,763,493, 5,731,000, 5,541,231, 5,427,798, 5,358,970 and 4,172,896, as well as in patents cited therein.

The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, the particular mode of administration. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.

Methods of preparing these formulations or compositions include the step of bringing into association an active compound, such as a compound of the invention, with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. formulations of the invention suitable for oral administration may be in the form of capsules (including sprinkle capsules and gelatin capsules), cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), lyophile, powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. Compositions or compounds may also be administered as a bolus, electuary or paste.

To prepare solid dosage forms for oral administration (capsules (including sprinkle capsules and gelatin capsules), tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; (10) complexing agents, such as, modified and unmodified cyclodextrins; and (11) coloring agents. In the case of capsules (including sprinkle capsules and gelatin capsules), tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.

A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets, and other solid dosage forms of the pharmaceutical compositions, such as dragees, capsules (including sprinkle capsules and gelatin capsules), pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-Formulating art. They may also be Formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient can also be in micro- encapsulated form, if appropriate, with one or more of the above-described excipients.

Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.

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

Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

formulations of the pharmaceutical compositions for rectal, vaginal, or urethral administration may be presented as a suppository, which may be prepared by mixing one or more active compounds with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.

formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, or an oral spray, or an oral ointment.

Alternatively or additionally, compositions can be Formulated for delivery via a catheter, stent, wire, or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum, or intestine.

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

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

The ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

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

Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056, 2005/0059744, 2005/0031697 and 2005/004074 and U.S. Patent No. 6,583,124, the contents of which are incorporated herein by reference. If desired, liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatible with such fluids. A preferred route of administration is local administration (e.g., topical administration, such as eye drops, or administration via an implant).

The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. Pharmaceutical compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

Examples of suitable aqueous and non-aqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin. In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be

accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly (anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue.

For use in the methods of this invention, active compounds can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.

Methods of introduction may also be provided by rechargeable or biodegradable devices. Various slow release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinaceous biopharmaceuticals. A variety of biocompatible polymers (including hydrogels), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a compound at a particular target site.

Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular subject, composition, and mode of

administration, without being toxic to the subject.

The selected dosage level will depend upon a variety of factors including the activity of the particular compound or combination of compounds employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound(s) being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound(s) employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the pharmaceutical composition or compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. By “therapeutically effective amount” is meant the concentration of a compound that is sufficient to elicit the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary according to the weight, sex, age, and medical history of the subject. Other factors which influence the effective amount may include, but are not limited to, the severity of the subject's condition, the disorder being treated, the stability of the compound, and, if desired, another type of therapeutic agent being administered with the compound of the invention. A larger total dose can be delivered by multiple administrations of the agent. Methods to determine efficacy and dosage are known to those skilled in the art (Isselbacher et al. (1996) Harrison’s Principles of Internal Medicine 13 ed., 1814-1882, herein incorporated by reference).

In general, a suitable daily dose of an active compound used in the compositions and methods of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.

If desired, the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain embodiments of the present invention, the active compound may be administered two or three times daily. In preferred embodiments, the active compound will be administered once daily.

Effective dosage amounts of the disclosed compounds, when used for the indicated effects, range from about 0.5 mg to about 5000 mg of the disclosed compound as needed to treat the condition. Compositions for in vivo or in vitro use can contain about 0.5, about 5, about 20, about 50, about 75, about 100, about 150, about 250, about 500, about 750, about 1000, about 1250, about 2500, about 3500, or about 5000 mg of the disclosed compound, or, in a range of from one amount to another amount in the list of doses

In certain embodiments, compounds of the invention may be used alone or conjointly administered with another type of therapeutic agent. As used herein, the phrase“conjoint administration” refers to any form of administration of two or more different therapeutic compounds such that the second compound is administered while the previously administered therapeutic compound is still effective in the body (e.g., the two compounds are

simultaneously effective in the subject, which may include synergistic effects of the two compounds). For example, the different therapeutic compounds can be administered either in the same formulation or in a separate formulation, either concomitantly or sequentially. In certain embodiments, the different therapeutic compounds can be administered within one hour, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, or a week of one another. Thus, a subject who receives such treatment can benefit from a combined effect of different therapeutic compounds.

In certain embodiments, conjoint administration of compounds of the invention with one or more additional therapeutic agent(s) provides improved efficacy relative to each individual administration of the compound of the invention (e.g., compound of Formula (I) or Formula (II)) or the one or more additional therapeutic agent(s). In certain such

embodiments, the conjoint administration provides an additive effect, wherein an additive effect refers to the sum of each of the effects of individual administration of the compound of the invention and the one or more additional therapeutic agent(s).

This invention includes the use of pharmaceutically acceptable salts of compounds of the invention in the compositions and methods of the present invention. In certain

embodiments, contemplated salts of the invention include, but are not limited to, alkyl, dialkyl, trialkyl or tetra-alkyl ammonium salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, lH-imidazole, lithium, L- lysine, magnesium, 4-(2-hydroxyethyl)morpholine, piperazine, potassium, l-(2- hydroxyethyl)pyrrolidine, sodium, triethanolamine, tromethamine, and zinc salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, Na, Ca, K, Mg, Zn or other metal salts.

The pharmaceutically acceptable acid addition salts can also exist as various solvates, such as with water, methanol, ethanol, dimethylformamide, and the like. Mixtures of such solvates can also be prepared. The source of such solvate can be from the solvent of crystallization, inherent in the solvent of preparation or crystallization, or adventitious to such solvent.

Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.

Examples of pharmaceutically acceptable antioxidants include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabi sulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal-chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like

EXAMPLES

Example 1: Synthesis of Exemplary Compounds of the Disclosure

The compounds of Formula (I) and Formula (II) may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthetic schemes. The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes. NMR data was obtained using a 500 MHz NMR with CDCb or d6-DMSO as solvent.

Synthesis of Intermediate II

Compound A1 (8.5g, 0.042 mol) and compound A2 (6.3 g, 0.042 mol) and AcOH (100 ml) were heated at reflux for 16 h, then concentrated to dryness at reduced pressure and stirred for 30 min in ethanol (150 ml). The precipitate was collected and dried to afford 10 g of the target product as a white solid. MS: m/z (M+l)+: 330.0, 332.0 Compound A3 (4.5g, 0.009 mol), compound A4 (2.97g, 0.01 mol), BINAP (0.1 eq.), Pd2(dba)3 (0.05 eq.), and CS2CO3 (1.4 eq.) were suspended in 300 ml of toluene and then stirred at 90°C under N2 for 16h. After the reaction was completed, the mixture was concentrated and purified by SGC (PE / EA=5 / 1 ~ EA) to afford 2.5 g of the target product as a yellow solid. MS: m/z (M+l)+: 448.2

Compound A5 (2.5g, 5.59mmol) was dissolved in 200 ml of ethanol and then 98% NH2NH2.H2O (5 eq.) was added. The mixture was stirred at 60°C for 4h, and then filtered. The filtrate was concentrated to afford 1.7g of the crude product which was used in the next step without further purification. MS: m/z (M+l)+: 318.3

Compound A6 (1.7 g, 5.36mmol) was dissolved in 300 ml of DCM, and then DIPEA (3 eq.) and FmocCl (1.2 eq.) was added. The mixture was stirred at room temperature for 16 h. The mixture was washed with H2O (50 ml) and saturated NaCl solution (50 ml) and then dried and concentrated to afford the crude product 2.5 g, which was used in the next step without further purification. MS: m/z (M+l)+: 540.3

Compound A7 (2.5 g, 4.63mmol) was dissolved in 100 ml of DCM and then 30 ml of the TFA was added. The mixture was stirred at room temperature for 4 h, and then

concentrated to afford the crude product which was adjusted pH = 7 with NaHCCb solution and then concentrated to afford the crude product 2.0 g which was used in the next step without further purification. MS: m/z (M+l)+: 440.2

Compound A8 (2.0 g, 4.55 mmol) and compound 9 (2 eq.) was dissolved in 70 ml of ethanol and stirred at 85°C for 72 h. The mixture was purified by SGC (PE / EA = 5 / 1 to EA) to afford the 500 mg of the target product as a yellow solid. MS: m/z (M+l)+: 586.3 Compound A10 (500mg, 0.85mmol) was dissolved in 20 ml of CH3CN and then Et2 H (10 eq.) was added. The mixture was stirred at room temperature for 16 h and then concentrated and purified by pre-HPLC (NH4HCO3) to afford the II 150 mg as a yellow solid. MS: m/z (M+l)+: 364.2. ¾ NMR (DMSO-d6, 500 MHz): d 1.99-2.08 (m, 2H), 2.68- 2.70 (t, 2H, J=7.0), 3.02-3.04 (d, 2H, J=9.0), 3.62-3.64 (d, 2H, J=7.5), 4.65 (s, 1H), 4.73 (s, 1H), 5.81-5.83 (d, 1H, J=12.0), 6.56-6.58 (d, 2H, J=8.5), 6.77-6.79 (m, 2H), 7.01-7.03 (d, 2H, J=8.0), 7.32-7.35 (t, 1H, J=8.0), 7.83-7.84 (d, 1H, J=7.5), 8.21-8.23 (d, 1H, J=12.0), 14.45 (s, 1H). Synthesis of Intermediate 12

Compound B1 (2.2 g, 14.42 mmol), B2 (2.86 g, 14.42 mmol) and Et3N (2.0 eq.) were dissolved in n-BuOH and then stirred at 180°C for 2 h under microwave condition. The mixture was concentrated and purified by SGC (PE / EA = 4 / 1) to afford 800 mg of the target product as a white solid. MS: m/z (M+l)+: 315.1

To the suspension of Compound B3 (800 mg, 2.54 mmol), and Ra-Ni (160 mg) in 60 ml of methanol was added NaBEE (120 mg) in 20 ml of 8N NaOH solution at 50°C. The reaction mixture was stirred at 60°C for 3 h. After the reaction was completed, the mixture was filtered and the filtrate was concentrated to get a dark red oil which was stirred for 1 h with 10 g of KOH and extracted with DCM to afford 700 mg of the crude product which was used in the next step directly without further purification. MS: m/z (M+l)+: 319.2

Compound B4 (700 mg, 2.20mmol) was dissolved in 150 ml of DCM, and then DIPEA (3 eq.) and FmocCl (1.2 eq.) was added. The mixture was stirred at room temperature for 16 h. The mixture was washed with EbO (20 ml) and saturated NaCl solution (20 ml) and then dried and concentrated to afford the crude product 1.10 g, which was used in the next step without further purification. MS: m/z (M+l)+: 541.3

Compound B5 (1.10 g, 2.03mmol) was dissolved in 50 ml of DCM and then 15 ml of the TFA was added. The mixture was stirred at room temperature for 16 h, and then concentrated to afford the crude product which was adjusted pH = 7 with Et3N and then concentrated to afford 800 mg of the crude product which was used in the next step without further purification. MS: m/z (M+l)+: 441.2 Compound B6 (800 mg, 1.82 mmol) and compound 7 (3 eq.) was dissolved in 70 ml of ethanol and stirred at 85°C for 72 h. The mixture was purified by SGC (PE / EA = 3 / 1 to EA) to afford the 200 mg of the target product as a yellow oil. MS: m/z (M+l)+: 587.2

Compound B8 (200mg, 0.34mmol) was dissolved in 20 ml of CEECN and then Et2NH (10 eq.) was added. The mixture was stirred at room temperature for 16 h and then concentrated and purified by pre-HPLC (NH4HCO3) to afford the pure product 111 mg as a yellow solid. MS: m/z (M+l)+: 365.1. ¾ NMR (DMSO-d6, 500 MHz): d 1.99-2.07 (m, 2H), 2.67-2.70 (t, 2H, J=7.0), 3.07-3.10 (m, 1H), 3.30-3.37 (m, 2H), 3.48-3.51 (d, 1H, J=11.0), 3.57-3.60 (m, 1H), 4.77 (s, 1H), 4.91 (s, 1H), 5.82-5.85 (d, 1H, J=12.0), 6.50-6.52 (d, 2H, J=8.5), 6.76-6.79 (m, 2H), 7.31-7.40 (m, 2H), 7.84-7.86 (d, 1H, J=8.0), 7.93 (s, 1H), 8.23- 8.25 (d, 1H, J=12.0), 14.44 (s, 1H).

Synthesis of LI, L2 and L3

2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione:

A mixture of 4-fluoroisobenzofuran-l,3-dione (498.33 mg, 3.00 mmol), 3- aminopiperidine-2,6-dione hydrogen chloride (493.77 mg, 3.00 mmol) and NaOAc (246.09 mg, 3.00 mmol) in HOAc (10 mL) was stirred at 135 °C overnight, cooled and concentrated in vacuo. The residue was suspended in H2O (100 mL), and stirred at room temperature for 4 hours. The solid was collected via filtration and dried in vacuo to afford Cl as a white solid (751.43 mg, 92% yield). MS: m/z (M+l) + : 277.25. Tert-butyl 3-(2-(2-(2-( (2-(2, 6-dioxopiperidin-3-yl)-l, 3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)ethoxy)propanoate

To a solution of Cl (320 mg, 1.16 mmol) and /er/-butyl 3-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)propanoate (321.60 mg, 1.16 mmol) in NMP (6 mL, 0.2 M) was added DIPEA (299.28 mg, 2.32 mmol). The mixture was stirred at 90 °C overnight, cooled to room temperature, diluted with EA (60 mL), and washed with LbO (3 x 20 mL). the organic phase was washed with brine (30 mL), dried over anhydrous Na2SC>4, and filtered. The filtrate was concentrated in vacuo , and the residue was purified via silica gel column chromatography (EA/PE: 2/1, Rf = 0.4) to afford C2 (276.10 mg, 45% yield) as a yellow solid. MS: m/z (M+l) + : 534.62.

3-(2-(2-(2-((2-(2, 6-Dioxopiperidin-3-yl)-l , 3-dioxoisoindolin-4

yl)amino)ethoxy)ethoxy)ethoxy)propanoic acid

To a solution of C2 (54 mg, 0.10 mmol) in DCM (0.2 mL) was added TFA (50 pL). The reaction mixture was stirred at room temperature for 2 hours until complete, concentrated and dried in vacuo to afford LI (50 mg) as a yellow solid. MS: m/z (M+l) + : 478.51.

Tert-butyl 12-aminododecanoate

To 12-aminododecanoic acid (960 mg, 6 mmol) was added SOCb (10 mL) slowly at 0 °C. The solid was dissolved, turned into a light yellow solution with the completion of addition. The reaction mixture was kept stirring for 2 hours at room temperature, and concentrated in vacuo to remove the extra SOCb. The residue was dissolved in a solution of NaHC03 (3.57 g, 30 mmol) in’BuOH (15 mL) at 0 °C and stirred at room temperature overnight. The mixture was concentrated in vacuo to afford C3 as a yellow oil (700 mg).

Tert-butyl 12-( (2-(2, 6-dioxopiperidin-3-yl)-l, 3-dioxoisoindolin-4-yl)amino)dodecanoate To a solution of Cl (150 mg, 0.54 mmol) in NMP (3 mL) were added C3 (150 mg, 0.54 mmol) and DIPEA (130 mg, 1.08 mmol). The reaction mixture was stirred at 90 °C for 15 hours, cooled down to room temperature, diluted with ethyl acetate (50 mL), and washed with water (20 mL x 2). The organic phase was dried over anhydrous MgSCri and filtered.

The filtrate was concentrated in vacuo , and the residue was purified via silica gel column chromatography (hexane/EA = 4/1) to afford C4 as a yellow oil (75 mg, 26%). MS: m/z (M+H) + : 528.23. 12-((2-(2,6-Dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)am ino)dodecanoic acid

To a solution of C4 (75 mg, 0.14 mmol) in DCM (5 mL) was added TFA (1 mL). The reaction mixture was stirred at room temperature for 24 hours, concentrated and dried in vacuo to afford L2 as a yellow oil (60 mg). MS: m/z (M+H) + : 472.34.

2-(2, 6-Dioxopiperidin-3-yl)-5-fluoroisoindoline-l, 3-dione

A mixture of 5-fluoroisobenzofuran-l, 3-dione (498.33 mg, 3.00 mmol), 3- aminopiperidine-2,6-dione hydrogen chloride (493.77 mg, 3.00 mmol) and NaOAc (246.09 mg, 3.00 mmol) in HOAc (10 mL) was stirred at 135 °C overnight, cooled and concentrated in vacuo. The residue was suspended in H2O (100 mL), and stirred at room temperature for 4 hours. The solid was collected via filtration and dried in vacuo to afford C5 as a white solid (751.43 mg, 92% yield). MS: m/z (M+l) + : 240.55.

Tert-butyl 12-( (2-(2, 6-dioxopiperidin-3-yl)-l , 3-dioxoisoindolin-5-yl)amino)dodecanoate To a solution of C5 (150 mg, 0.54 mmol) in NMP (3 mL) were added C3 (150 mg, 0.54 mmol) and DIPEA (130 mg, 1.08 mmol). The reaction mixture was stirred at 90 °C for 15 hours, cooled down to room temperature, diluted with ethyl acetate (50 mL), and washed with water (20 mL x 2). The organic phase was dried over anhydrous MgSCri and filtered. The filtrate was concentrated in vacuo, and the residue was purified via silica gel column chromatography (hexane/EA = 4/1) to afford C6 as a yellow oil (51 mg, 18%). MS: m/z (M+H) + : 528.23.

12-((2-(2,6-Dioxopiperidin-3-yl)-l,3-dioxoisoindolin-5-yl)am ino)dodecanoic acid

To a solution of C6 (51 mg, 0.14 mmol) in DCM (4 mL) was added TFA (0.75 mL). The reaction mixture was stirred at room temperature for 24 hours, concentrated and dried in vacuo to afford L3 as a yellow oil (43 mg). MS: m/z (M+H) + : 472.45.

3-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l, 3-dioxoisoindolin-4- yl)amino)ethoxy)ethoxy)ethoxy)-N-(4-( (IS, 4S)-5-( (E)-3-(2-hydroxyphenyl)-3-oxoprop-l -en-1 - yl)-2, 5-diazabicyclo[ 2.2.1 ]heptan-2-yl)phenethyl)propanamide:

To a solution of LI (2.62 mg, 0.0055 mmol) and HATU (2.90 mg, 0.0076 nnol) in DMF (0.2 mL) was added DIPEA (7.10 mg, 0.055 mmol) at room temperature. (E)-3- ((l S , ,4,S)-5-(4-(2-aminoethyl)phenyl)-2,5-diazabicyclo[2.2. 1]heptan-2-yl)-l-(2- hydroxyphenyl)prop-2-en-l-one (1.92 mg, 0.0053 mmol) was added 2 minutes later, and the mixture was stirred at room temperature for another 15 minutes. Monitored via LCMS, the desired product was major, and the reaction mixture was purified via HPLC (0.1%

TFA/MeCN). MS: m/z (M+l) + : 823.92.

12-((2-(2, 6-dioxopiperidin-3-yl)-l , 3-dioxoisoindolin-5-yl)amino)-N-( 4-( (IS, 4S)-5-( (E)-3-(2- hydroxyphenyl)-3-oxoprop-l-en-l-yl)-2, 5-diazabicyclo[ 2.2.1 ]heptan-2- yl)phenethyl)dodecanamide

To a solution of L2 (2.30 mg, 0.0049 mmol) and HATU (2.23 mg, 0.0058 nnol) in DCM (0.2 mL) was added DIPEA (6.32 mg, 0.049 mmol) at room temperature. (E)- 3- ((l S , ,4,S)-5-(4-(2-aminoethyl)phenyl)-2,5-diazabicyclo[2.2. 1]heptan-2-yl)-l-(2- hydroxyphenyl)prop-2-en-l-one (1.80 mg, 0.0049 mmol) was added 2 minutes later, and the mixture was stirred at room temperature for another 15 minutes. Monitored via LCMS, the desired product was major, and the reaction mixture was purified via silica gel column chromatography (EA) to afford E3 (5.14 mg, 60-70% purity) as a yellow solid. MS: m/z (M+l) + : 818.02.

12-((2-(2, 6-dioxopiperidin-3-yl)-l , 3-dioxoisoindolin-4-yl)amino)-N-( 4-( (IS, 4S)-5-( (E)-3-(2- hydroxyphenyl)-3-oxoprop-l-en-l-yl)-2, 5-diazabicyclo[ 2.2.1 ]heptan-2- yl)phenethyl)dodecanamide

To a solution of L2 (2.20 mg, 0.0047 mmol) and HATU (2.23 mg, 0.0058 nnol) in DCM (0.2 mL) was added DIPEA (6.32 mg, 0.049 mmol) at room temperature. (E)- 3- ((l S , ,4,S)-5-(4-(2-aminoethyl)phenyl)-2,5-diazabicyclo[2.2. 1]heptan-2-yl)-l-(2- hydroxyphenyl)prop-2-en-l-one (1.70 mg, 0.0047 mmol) was added 2 minutes later, and the mixture was stirred at room temperature for another 15 minutes. Monitored via LCMS, the desired product was major, and the reaction mixture was purified via silica gel column chromatography (EA) to afford E2 (60-70% purity) as a yellow solid (neutral MeCN). MS: m/z (M+l) + : 818.05. Compounds E4-E13 were synthesized in an analogous manner to compounds E1-E3.

Example 2: Biochemical Assays

We incubated molecules with 0.05uM of His-tag SMARCA4 protein and 0.015uM of biotinylated probe in alpha assay buffer (50mM HEPES, 150mM NaCl, 0.01% Tween-20,

0.1% BSA, pH 7.5) for 30 min at room temperature. After incubation, 0.02 mg/ml of Streptavidin donor beads (Perkin, Cat. #6760002B) and 0.02 mg/ml of Nickel chelate acceptor beads (Perkin, Cat. #AL 108L) were added for additional 30 min. Luminescence signals were subsequently read and quantified for protein-molecule interactions.

INCORPORATION BY REFERENCE

All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.

EQUIVALENTS

While specific embodiments of the subject invention have been discussed, the above specification is illustrative and not restrictive. Many variations of the invention will become apparent to those skilled in the art upon review of this specification and the claims below.

The full scope of the invention should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.