Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HAT INHIBITORS AND METHODS FOR THEIR USE
Document Type and Number:
WIPO Patent Application WO/2016/044777
Kind Code:
A1
Abstract:
Compounds having a structure of Formula I or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein R1, R2a, R2b, R3a, R3b, R3c, R4a, R4b, R5, R6, Z and X are as defined herein are provided. Pharmaceutical compositions comprising such compounds and methods for treating various HAT-related conditions or diseases, including cancer, by administration of such compounds are also provided.

Inventors:
KESICKI EDWARD A (US)
WANG CE (CN)
PATANE MICHAEL A (US)
KLUGE ARTHUR F (US)
VAN DRIE JR JOHN H (US)
Application Number:
PCT/US2015/051040
Publication Date:
March 24, 2016
Filing Date:
September 18, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ABBVIE INC (US)
International Classes:
C07D405/14; A61K31/4166; A61K31/4178; A61K31/421; A61K31/422; A61P3/00; A61P25/28; A61P29/00; A61P35/00; C07D233/74; C07D233/76; C07D233/78; C07D263/44; C07D401/04; C07D403/04; C07D403/06; C07D403/14; C07D405/04; C07D405/12; C07D409/04; C07D413/04
Foreign References:
US20130142887A12013-06-06
Other References:
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 9 November 2010 (2010-11-09), XP002748595, Database accession no. 1252058-93-7
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 10 April 2013 (2013-04-10), XP002748596, Database accession no. 1427770-67-9
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 9 November 2010 (2010-11-09), XP002748597, Database accession no. 1252080-55-9
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 9 November 2011 (2011-11-09), XP002748598, Database accession no. 1252080-38-8
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 9 November 2010 (2010-11-09), XP002748599, Database accession no. 1252057-38-7
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 16 September 2009 (2009-09-16), XP002748600, Database accession no. 1185172-60-4
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 6 February 2009 (2009-02-06), XP002748601, Database accession no. 1101847-27-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 27 January 2009 (2009-01-27), XP002748602, Database accession no. 1096690-49-1
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 26 January 2009 (2009-01-26), XP002748603, Database accession no. 1096155-09-7
DATABASE Pubchem compound [online] NCBI; 15 July 2005 (2005-07-15), XP002748604, Database accession no. CID 2462866
DATABASE Pubchem Compound [online] NCBI; 17 September 2005 (2005-09-17), XP002748605, Database accession no. CID 4842105
DATABASE Pubchem Compound [online] NCBI; 20 May 2011 (2011-05-20), XP002748608, Database accession no. CID 51556301
DATABASE Pubchem Compound [online] NCBI; 29 May 2009 (2009-05-29), XP002748607, Database accession no. CID 34507151
BOWERS E M ET AL: "Virtual Ligand Screening of the p300/CBP Histone Acetyltransferase: Identification of a Selective Small Molecule Inhibitor", CHEMISTRY AND BIOLOGY, CURRENT BIOLOGY, LONDON, GB, vol. 17, no. 5, 28 May 2010 (2010-05-28), pages 471 - 482, XP002697557, ISSN: 1074-5521, DOI: 10.1016/J.CHEMBIOL.2010.03.006
DATABASE pubchem Compound [online] NCBI; 19 October 2014 (2014-10-19), XP002748606, Database accession no. CID 78812744
LOPES DA ROSA JESSICA ET AL: "A small molecule inhibitor of fungal histone acetyltransferase Rtt109", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, AMSTERDAM, NL, vol. 23, no. 10, 4 April 2013 (2013-04-04), pages 2853 - 2859, XP028582276, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2013.03.112
Attorney, Agent or Firm:
BAUDE, Eric J. et al. (1 North Waukegan RoadAP34-2/V37, North Chicago Illinois, US)
Download PDF:
Claims:
CLAIMS at is claimed is: 1. A com ound havin the followin structure (I):

(I)

stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:

X is–NH- or -O-;

Z is a direct bond or–C(R7a)(R7b)-;

R1 is carbocyclyl or heterocyclyl;

R2a and R2b are each independently H, D or C1-C6 alkyl;

R3a is carbocyclyl or heterocyclyl and R3b is C1-C6 alkyl or carbocyclyl; or R3a R3b are each independently C1-C6 alkyl; or R3a and R3b are taken together with the carbon toch they are bound to form a carbocyclic or heterocyclic ring;

R3c is H or D;

R4a and R4b are each independently H, D or C1-C6 alkyl;

R5 is carbocyclyl or heterocyclyl;

R6 is H or D when Z is a direct bond; or R6 is H, D or C1-C6 alkyl when ZC(R7a)(R7b)-;

R7a and R7b are each independently H, D or C1-C6 alkyl;

with the proviso that R3a, R3b and R3c are not unsubstituted cyclopropyl, methyl and H, ectively, when Z is–CH2-, R1 is unsubstituted phenyl and R5 is unsubstituted indolyl.

2. The compound of claim 1, wherein X is -NH-. 3. The compound of claim 1, wherein X is–O-. 4. The compound of any one of claims 1-3, wherein Z is a direct bond and R6 or D. 5. The compound of any one of claims 1-3, wherein Z is–C(R7a)(R7b)-. 6. The compound of claim 5, wherein Z is–CH2-. 7. The compound of claim 5, wherein R7a is H and R7b is C1-C6 alkyl. 8. The compound of claim 5, wherein R7a and R7b are each independently C1-lkyl. 9. The compound of claim 5, wherein at least one of R7a or R7b is D. 10. The compound of any one of claims 1-9, wherein R1 is unsubstituted. 11. The compound of any one of claims 1-9, wherein R1 is substituted with or more substituents. 12. The compound of claim 11, wherein the substituents are selected from, nitrilyl and C1-C6 alkyl. 13. The compound of claim 12, wherein the substituents are selected fromro, nitrilyl and methyl.

14. The compound of any one of claims 1-13, wherein R1 is carbocyclyl. 15. The compound of claim 14, wherein R1 is aryl. 16. The compound of claim 15, wherein R1 is phenyl. 17. The compound of claim 16, wherein R1 has one of the followingctures:

. 18. The compound of any one of claims 1-13, wherein R1 is heterocyclyl. 19. The compound of claim 18, wherein R1 is tetrahydropyranyl. 20. The compound of claim 18, wherein R1 is heteroaryl. 21. The compound of claim 20, wherein R1 is furanyl, benzodioxazolyl ordazolyl. 22. The compound of claim 14, wherein R1 has one of the followingctures: . 23. The compound of any one of claims 1-22, wherein at least one of R2a or is H.

24. The compound of any one of claims 1-22, wherein each of R2a and R2b is

25. The compound of any one of claims 1-23, wherein at least one of R2a or is C1-C6 alkyl. 26. The compound of any one of claims 1-23, wherein at least one of R2a or is D. 27. The compound of any one of claims 1-26, wherein R3a is carbocyclyl and is C1-C6 alkyl. 28. The compound of claim 27, wherein R3a is cycloalkyl. 29. The compound of claim 28, wherein R3a is cyclopropyl or cyclobutyl. 30. The compounds of any one of claims 27-29, wherein R3a is substitutedh C1-C6 alkyl. 31. The compound of any one of claims 27-30, wherein R3b is methyl. 32. The compound of any one of claims 1-26, wherein R3a and R3b are each pendently carbocyclyl. 33. The compound of claim 32, wherein R3a and R3b are each independently oalkyl. 34. The compound of claim 33, wherein cycloalkyl is cyclopropyl or obutyl.

35. The compound of any one of 1-26, wherein R3a and R3b are each pendently C1-C6 alkyl. 36. The compound of claim 35, wherein R3a and R3b are each ethyl. 37. The compound of any one of claims 1-26, wherein R3a and R3b are taken ther with the carbon to which they are bound to form a carbocyclic or heterocyclic ring. 38. The compound of claim 37, wherein R3a and R3b are taken together with carbon to which they are bound to form a heterocyclic ring. 39. The compound of claim 38, wherein the heterocyclic ring isahydropyranyl. 40. The compound of any one of claims 1-39, wherein R3c is H. 41. The compound of any one of claims 1-39, wherein R3c is D. 42. The compound of any one of claims 1-41, wherein at least one of R4a or is H. 43. The compound of any one of claims 1-41, wherein each of R4a and R4b is

44. The compound of any one of claims 1-42, wherein at least one of R4a or is C1-C6 alkyl. 45. The compound of any one of claims 1-42, wherein at least one of R4a or is D.

46. The compound of any one of claims 1-45, wherein R5 is unsubstituted. 47. The compound of any one of claims 1-45, wherein R5 is substituted with or more substituents. 48. The compound of claim 47, wherein the substituents are selected from , C1-C6 alkyl and alkoxy. 49. The compound of claim 48, wherein the substituents are selected fromro, chloro, methyl and methoxy. 50. The compound of any one of claims 1-49, wherein R5 is carbocyclyl. 51. The compound of claim 50, wherein R5 is aryl. 52. The compound of claim 51, wherein R5 is phenyl. 53. The compound of claim 51, wherein R5 is napthyl. 54. The compound of any one of claims 50-53, wherein R5 has one of theowin structures:

55. The compound of any one of claims 1-49, wherein wherein R5 is rocyclyl. 56. The compound of claim 55, wherein R5 is heteroaryl. 57. The compound of claim 56, wherein R5 is furanyl, indolyl, indazolyl, dinyl, pyridinyl oxide, imidazolyl, pyrazolyl, benzofuranyl or benzothiophenyl. 58. The compound of any one of claims 55-57, wherein the R5 has one of theowing structures:

;

The compound of claim 1, wherein the compound is selected from the group consisting of:

-(1H-indol-3-yl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1-cyclopropylethyl)acetamide; enzyl-N-(1-cyclopropylethyl)-2-(4-(2-methoxyphenyl)-4-methyl-2,5-dioxoimidazolidin-1- yl)acetamide; enzyl-N-(1-cyclopropylethyl)-2-(4-methyl-2,5-dioxo-4-phenylimidazolidin-1-yl)acetamide;enzyl-N-(1-cyclopropylethyl)-2-(4-(furan-2-yl)-2,5-dioxoimidazolidin-1-yl)acetamide;

enzyl-N-(1-cyclopropylethyl)-2-(2,5-dioxo-4-(pyridin-3-yl)imidazolidin-1-yl)acetamide;enzyl-N-(1-cyclopropylethyl)-2-(4-(1-methyl-1H-imidazol-4-yl)-2,5-dioxoimidazolidin-1- yl)acetamide;

-(2-(benzyl(1-cyclopropylethyl)amino)-2-oxoethyl)-2,5-dioxoimidazolidin-4-yl)pyridine 1- oxide;

enzyl-N-(1-cyclopropylethyl)-2-(4-(naphthalen-1-yl)-2,5-dioxoimidazolidin-1-yl)acetamide;enzyl-N-(1-cyclopropylethyl)-2-(2,5-dioxo-4-(1H-pyrazol-4-yl)imidazolidin-1-yl)acetamide;enzyl-N-(1-cyclopropylethyl)-2-(4-(1-methyl-1H-indol-3-yl)-2,5-dioxoimidazolidin-1- yl)acetamide;

-(1H-indazol-3-yl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1-cyclopropylethyl)acetamide;enzyl-N-(1-cyclopropylethyl)-2-(4-(6-fluoro-1H-indol-3-yl)-2,5-dioxoimidazolidin-1- yl)acetamide;

enzyl-N-(1-cyclopropylethyl)-2-(4-(naphthalen-2-yl)-2,5-dioxoimidazolidin-1-yl)acetamide;enzyl-N-(1-cyclopropylethyl)-2-(4-(5-fluoro-1H-indol-3-yl)-2,5-dioxoimidazolidin-1- yl)acetamide;

enzyl-2-(4-(4-chloro-1H-indol-3-yl)-2,5-dioxoimidazolidin-1-yl)-N-(1- cyclopropylethyl)acetamide;

enzyl-2-(4-(7-chloro-1H-indol-3-yl)-2,5-dioxoimidazolidin-1-yl)-N-(1- cyclopropylethyl)acetamide;

enzyl-2-(4-(4-chloro-1H-indol-3-yl)-2,5-dioxoimidazolidin-1-yl)-N-((S)-1- cyclobutylethyl)acetamide;

enzyl-N-((S)-1-cyclopropylethyl)-2-(4-(2,6-dichlorophenyl)-2,5-dioxoimidazolidin-1- yl)acetamide;

enzyl-N-((S)-1-cyclopropylethyl)-2-(2,5-dioxo-4-o-tolylimidazolidin-1-yl)acetamide;

enzyl-N-((S)-1-cyclopropylethyl)-2-(2,5-dioxo-4-phenylimidazolidin-1-yl)acetamide;

enzyl-N-((S)-1-cyclopropylethyl)-2-(4-(2,6-dimethylphenyl)-2,5-dioxoimidazolidin-1- yl)acetamide;

-(1H-indol-3-yl)-4-methyl-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-((S)-1- cyclopropylethyl)acetamide;

enzyl-2-(4-(2-chlorophenyl)-2,5-dioxoimidazolidin-1-yl)-N-((S)-1- cyclopropylethyl)acetamide;

enzyl-N-((S)-1-cyclopropylethyl)-2-(5-(2,6-dichlorophenyl)-2,4-dioxooxazolidin-3- yl)acetamide;

-(1H-indol-3-yl)-2,4-dioxooxazolidin-3-yl)-N-benzyl-N-((S)-1-cyclopropylethyl)acetamide; enzyl-2-(5-(4-chloro-1H-indol-3-yl)-2,4-dioxooxazolidin-3-yl)-N-((S)-1- cyclopropylethyl)acetamide;

-(benzo[b]thiophen-3-yl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1- cyclopropylethyl)acetamide;

R)-4-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-((S)-1- cyclopropylethyl)acetamide;

S)-4-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-((R)-1- cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1- cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N- (dicyclopropylmethyl)acetamide;

S)-4-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-((S)-1- cyclopropylethyl)acetamide;

R)-4-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-((R)-1- cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(1-cyclopropylethyl)-N-(3- methylbenzyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(benzo[d][1,3]dioxol-5-ylmethyl)- N-(1-cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(1-cyclopropylethyl)-N-(furan-2- ylmethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1-(1- methylcyclopropyl)ethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(4-cyanobenzyl)-N-(1- cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(1-cyclopropylethyl)-N- ((tetrahydro-2H-pyran-4-yl)methyl)acetamide;

1H-imidazol-2-yl)methyl)-2-(4-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(1- cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1- cyclobutylethyl)acetamide;

R)-4-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1- cyclopropylethyl)acetamide;

-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(pentan-3-yl)acetamide;-((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(tetrahydro-2H-pyran-4- yl)acetamide; -((1H-indol-3-yl)methyl)-2,5-dioxoimidazolidin-1-yl)-N-(1-cyclobutylethyl)-N-(3- fluorobenzyl)acetamide; and

-(benzofuran-2-yl)-4-methyl-2,5-dioxoimidazolidin-1-yl)-N-benzyl-N-(1- cyclopropylethyl)acetamide. 60. A pharmaceutical composition comprising a pharmaceutically acceptableier or excipient and a com ound havin the followin structure (I):

(I)

stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:

X is–NH- or -O-;

Z is a direct bond or–C(R7a)(R7b)-;

R1 is carbocyclyl or heterocyclyl;

R2a and R2b are each independently H, D or C1-C6 alkyl;

R3a is carbocyclyl or heterocyclyl and R3b is C1-C6 alkyl or carbocyclyl; or R3a R3b are each independently C1-C6 alkyl; or R3a and R3b are taken together with the carbon toch they are bound to form a carbocyclic or heterocyclic ring;

R3c is H or D;

R4a and R4b are each independently H, D or C1-C6 alkyl;

R5 is carbocyclyl or heterocyclyl;

R6 is H, D or C1-C6 alkyl; and

R7a and R7b are each independently H, D or C1-C6 alkyl. 61. The composition of claim 60, wherein the compound is a compoundording to any one of claims 1-59.

62. A method for treating a HAT dependent condition in a mammal in needeof, the method comprising administering an effective amount of the pharmaceuticalposition of any one of claims 60 or 61 to the mammal. 63. The method of claim 62, wherein the condition is cancer, metabolicase, neurodegenerative disorders or inflammation. 64. The method of claim 63, wherein the condition is cancer.

Description:
HAT INHIBITORS AND

METHODS FOR THEIR USE RELATED APPLICATION INFORMATION

This application claims the benefit of PCT Patent Application PCT/CN2014/086798, filed on September 18, 2014, the contents of which are herein incorporated by reference. BACKGROUND Technical Field

The present invention generally relates to compounds having activity as inhibitors of HAT enzymes. The compounds find utility in any number of therapeutic applications, including treatment of cancer. Background of the Invention

Within the eukaryotic cell nucleus, DNA is condensed and packaged into chromatin. The structural unit of chromatin is a nucleosome, which consists of 147 base pairs of DNA wrapped 1.6 times around a histone core of two H2A-H2B dimers and a H3- H4 tetramer (Kornberg et al., 1999, Cell 98:285-294). Histones undergo extensive post- translational modification, which determines whether a gene is transcriptionally active or inactive (Goll and Bestor, 2002, Genes Dev.16:1739-1742; Grant, 2001, Genome Biol.2:). The reversible acetylation of histones and other proteins is one of the most abundant post- translational modifications in eukaryotic cells and is a major mechanism of cellular regulation. Histone acetyltransferases (HATs) catalyze the acetylation (transfer of an acetyl group) on a ε-amino group of a target lysine side chain within a substrate histone, and histone deacetylases (HDACs) catalyze the removal of acetyl groups from lysine residues. HATs are categorized into four major families based on primary sequence homology, shared structural features, and functional roles: Gcn5/PCAF (General control nonrepressed protein 5 and p300 and CBP associated factor); MYST (named for the founding members MOZ, Ybf2/Sas3, Sas2, and Tip60); p300/CBP (protein of 300kDa and CREB Binding Protein); and Rtt109 (Regulator of Ty1 Transposition gene production 109).

Paralog HATs p300 (KATB) and CBP (referred to as p300/CBP) have >90% sequence identity and are conserved in metazoans. In addition to the enzymatic HAT domain, p300/CBP has multiple domains including three cysteine-histidine rich domains (CH1, CH2, and CH3), a KIX domain, a bromodomain, and a steroid receptor coactivator interaction domain (SRC-1 interaction domain). P300 and CBP were originally discovered as binding partners of E1A adenoviral protein and cAMP-regulated enhancer binding proteins, respectively (Yee and Branton, 1985, Virology 147:142-153; Harlow et al., 1986, Mol. Cell Biol.6:1579-1589; Chrivia et al., 1993, Nature 365:855-859).

P300/CBP was later found to have intrinsic HAT activity (Ogryzko et al., 1996, Cell 87:953-959; Bannister and Kouzarides, 1996, Nature 384:641-643). In addition to acetylating multiple lysines on all four core histones (H2A, H2B, H3 and H4), P300/CBP has been shown to have promiscuous acetyltransferase activity towards > 70 substrates (Wang et al., 2008, Curr. Opin. Struct. Biol.18:741-747), including, for example, p53 (Gu et al., 1997, Cell 90:595-606), MyoD (Polesskaya et al., 2002, J. Biol. Chem.275:34359- 64), STAT3 (Yuan et al., 2005, Science 307:269-73) and NFκβ (Chen et al., 2002, EMBO J.21:6539-48). Besides acting as an acetyltransferase, p300 also acts as a scaffold for transcription factors or a bridge to connect the transcription factors and the basal transcriptional machinery to activate transcription (Chan and Thangue, 2001, J. Cell Sci. 114:2363-2373; Chen and Li, 2011, Epigenetics 6:957-961). P300/CBP proteins are involved in many cellular processes, including cell growth, proliferation, and

differentiation (Chan and Thangue, supra).

Aberrant p300/CBP activity or mutations has been associated with various diseases. High p300 expression has been observed in prostate cancer (Heemers et al., 2008, Adv. Exp. Med. Biol.617:535-40; Isharwal et al., 2008, Prostate 68:1097-104), liver cancer (Yokomizo et al., 2011, Cancer Lett.310:1407; Li et al., 2011, J. Transl. Med.9:5), and breast cancer (Fermento et al., 2010, Exp. Mol. Pathol.88:256-64). Mutations in p300/CBP genes have been found in human tumors (Petrij et al., 1995, Nature 376:348-51; Muraoka et al, 1996, Oncogene 12:1565-69; Sobulo et al., 1997, Proc. Natl. Acad. Sci. USA 94:8732-37; Gayther et al., 2000, Nat. Genet.24:300-304). P300 missense mutations and truncations have been found in solid tumors and B-cell lymphoma, suggesting a role as a tumor suppressor (Iyer et al., 2004, Oncogene 23:4225-31; Pasqualucci et al., 2011, nature 471:189-95). Inhibition of p300/CBP has therapeutic potential in cancer (Iyer et al., 2004, Proc. Natl. Acad. Sci. USA 101:7386-7391; Stimson et al., 2005, Mol. Cancer Ther. 4:1521-1532; Zheng et al., 2004, Methods Enzymol.376:188-199), cardiac disease (Davidson et al., 2005, Chembiochem.6:162-170); diabetes mellitus (Zhou et al., 2004, Nat. Med.10:633-637), and HIV (Varier and Kundu, 2006, Curr. Pharm. Des.12:1975- 1993). Heterozygous germline mutations in CBP or p300 have also been described in Rubinstein-Taybi syndrome, an autosomal dominant disease characterized by mental retardation, skeletal abnormalities, and a high incidence of neoplasia (Petrij et al., 1995, Nature 376:348-51; Petrij et al., 2000, Am. J. Med. Genet.92:47-52). P300/CBP is also involved in regulating inflammatory mediators (Deng et al., 2004, Blood 103:2135-42; Turner-Brannen et al., 2011, J. Immunol.186:7127-7135). P300/CBP has also been linked to other diseases, such as fibrosis (Ghosh and Varga, 2007, J. Cell. Physiol.213:663-671), metabolic syndrome (Bricambert et al., 2010, J. Clin. Invest.120:4316-4331), and progressive neurodegenerative diseases, such as Huntington Disease (Cong et al., 2005, Mol. Cell. Neurosci.30:12-23), Kennedy’s disease (Lieberman et al., 2002, Hum. Mol. Genet.11:1967-76), and Alzheimer’s disease (Francis et al., 2007, Neurosci. Lett.413:137- 140).

The association of p300/CBP activity in disease pathogenesis suggests potential utility of p300/CBP as a therapeutic target. However, the identification of potent, specific histone acetyltransferase inhibitors has been challenging (Cole, 2008, Nat. Chem. Biol.4:590-97). P300 HAT inhibitors derived from natural compounds have moderate potency but lack specificity (Dekker and Haisma, 2009, Drug Disc. Today 14:942-8). Lys- CoA, converted to a cell-permeable form with a Tat peptide attachment, is more selective, but has limited use in pharmacological studies due to its complexity. Recently, a selective p300 inhibitor C646 was identified using the Lys-CoA/p300 HAT structure in a virtual ligand screening approach (Bowers et al., 2010, Chemistry & Biology 17:471-482).

Accordingly, while progress has been made in this field, there remains a need in the art for improved HAT inhibitors. The present invention fulfills this need and provides further related advantages. BRIEF SUMMARY

In brief, the present invention is directed to compounds having activity as HAT inhibitors, including stereoisomers, tautomers, pharmaceutically acceptable salts and prodrugs thereof, and the use of such compounds to treat HAT-related conditions or diseases, such as cancer.

In accordance with one embodiment, there is provided a compound having the following structure (I):

or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein R 1 , R 2a , R 2b , R 3a , R 3b , R 3c , R 4a , R 4b , R 5 , R 6 , Z and X are as defined herein.

In another embodiment, pharmaceutical compositions comprising a pharmaceutically acceptable carrier or excipient and a compound of structure (I) are provided.

In other embodiments, a method for treating a HAT dependent condition in a mammal in need thereof is provided. The method comprises administering an effective amount of a pharmaceutical composition comprising a compound of structure (I) to the mammal. Exemplary conditions which can be treated with the disclosed compounds and compositions include, but are not limited to, cancer, metabolic disease, neurodegenerative disorders and inflammation.

These and other aspects of the invention will be apparent upon reference to the following detailed description. To this end, various references are set forth herein which describe in more detail certain background information, procedures, compounds and/or compositions, and are each hereby incorporated by reference in their entirety. DETAILED DESCRIPTION

In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments of the invention. However, one skilled in the art will understand that the invention may be practiced without these details.

Unless the context requires otherwise, throughout the present specification and claims, the word“comprise” and variations thereof, such as,“comprises” and “comprising” are to be construed in an open, inclusive sense, that is as“including, but not limited to”.

Reference throughout this specification to“one embodiment” or“an embodiment” means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, the appearances of the phrases“in one embodiment” or“in an embodiment” in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.

“Amino” refers to the -NH 2 radical.

“Carboxyl” refers to the–CO 2 H radical.

“Cyano” or“nitrilyl” refers to the -CN radical.

“Ester” refers to a structure of the formula R a C(=O)OR b where R a and R b are each independently non-hydrogen substitutents (e.g., alkyl or aryl and the like).

“Hydrazone” refers to =N-NH 2 substituent. “Hydroxy” or“hydroxyl” refers to the -OH radical.

“Imino” refers to the =NH substituent.

“Nitro” refers to the -NO 2 radical.

“Oxo” refers to the =O substituent.

“Oxime” refers to =N-OH substituent.

“Thioxo” refers to the =S substituent.

“Alkyl” refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C 1 -C 12 alkyl), from one to eight carbon atoms (C 1 -C 8 alkyl) or from one to six carbon atoms (C 1 - C 6 alkyl), and which is attached to the rest of the molecule by a single bond. Alkyls include alkenyls and alkynyls. Representative alkyls include, but are not limited to, methyl, ethyl, n-propyl, 1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-1,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted.

“Alkylene” or“alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, ethenylene, propenylene, n-butenylene, propynylene, n-butynylene, and the like. The alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted.

“Alkenyl” refers to an alkyl group which comprises one or more double bonds and has from one to twelve carbon atoms (C 1 -C 12 alkenyl), from one to eight carbon atoms (C 1 -C 8 alkenyl) or from one to six carbon atoms (C 1 -C 6 alkenyl), and which is attached to the rest of the molecule by a single bond, e.g., ethenyl, propenyl, butenyl, pentenyl, hexenyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group may be optionally substituted.

“Alkynyl” refers to an alkyl group which comprises one or more triple bonds and has from one to twelve carbon atoms (C 2 -C 12 alkynyl), from one to eight carbon atoms (C 2 -C 8 alkynyl) or from one to six carbon atoms (C 2 -C 6 alkynyl), and which is attached to the rest of the molecule by a single bond, e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group may be optionally substituted.

“Alkoxy” refers to a radical of the formula -OR a where R a is an alkyl radical as defined above. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.

“Alkylaminyl” refers to a radical of the formula -NHR a or -NR a R a where each R a is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylaminyl group may be optionally substituted.

“Aryl” refers to a hydrocarbon ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include spiro, fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as-indacene, s-indacene, indane, indene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term“aryl” or the prefix“ar-“ (such as in“aralkyl”) is meant to include aryl radicals that are optionally substituted.

“Aralkyl” refers to a radical of the formula -R b -R c where R b is an alkylene chain as defined above and R c is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated otherwise specifically in the

specification, an aralkyl group may be optionally substituted. “Carbocyclyl” or“carbocyclic ring” refers to a stable monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Carbocycles include cycloalkyls and aryls as defined herein. Unless stated otherwise specifically in the specification, a carbocyclyl group may be optionally substituted.

“Cycloalkyl” refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include spiro, fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic radicals include, for example, adamantyl, norbornyl, decalinyl,

7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted.

“Cycloalkylalkyl” refers to a radical of the formula -R b R d where R b is an alkylene chain as defined above and R d is a cycloalkyl radical as defined above. In certain embodiments, R b is substituted with a further cycloalkyl group, such that the

cycloalkylalkly comprises two cycloalkyl moieties. Cyclopropylalkyl and cyclobutylalkyl are exemplary cycloalkylalkyl groups, comprising at least one cyclopropyl or at least one cyclobutyl group, respectively. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted.

“Fused” refers to any ring structure described herein which is fused to an existing ring structure in the compounds of the invention. When the fused ring is a heterocyclyl ring or a heteroaryl ring, any carbon atom on the existing ring structure which becomes part of the fused heterocyclyl ring or the fused heteroaryl ring may be replaced with a nitrogen atom.

“Halo” or“halogen” refers to bromo, chloro, fluoro or iodo. “Haloalkyl” refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl,

1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the specification, a haloalkyl group may be optionally substituted.

“Heterocyclyl” or“heterocyclic ring” refers to a stable 3- to 18-membered non-aromatic or aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Heterocyclyl includes heteroaryl as defined herein. Unless stated otherwise specifically in the specification, the heterocyclyl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include spiro, fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl,

octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl,

thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocyclyl group may be optionally substituted.

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

“Heterocyclylalkyl” refers to a radical of the formula -R b R e where R b is an alkylene chain as defined above and R e is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. Unless stated otherwise specifically in the specification, a heterocyclylalkyl group may be optionally substituted.

“Heteroaryl” refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include spiro, fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. The heteroatom may be a member of an aromatic or non-aromatic ring, provided at least one ring in the heteroaryl is aromatic. Examples include, but are not limited to, azepinyl, acridinyl, benzimidazolyl,

benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotriazolyl,

benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, dibenzofuranyl,

dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl,

1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1-oxidopyridazinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinazolinyl, quinoxalinyl, quinolinyl, quinuclidinyl, isoquinolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteroaryl group may be optionally substituted.

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

“Heteroarylalkyl” refers to a radical of the formula -R b R f where R b is an alkylene chain as defined above and R f is a heteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted.

“Thioalkyl” refers to a radical of the formula -SR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted.

The term“substituted” used herein means any of the above groups (i.e., alkyl, alkylene, alkenyl, alkynyl, alkoxy, aryl, carbocyclyl, cycloalkyl, heterocyclyl and/or heteroaryl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups.

“Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example,“substituted” includes any of the above groups in which one or more hydrogen atoms are replaced

with -NR g R h , -NR g C(=O)R h , -NR g C(=O)NR g R h , -NR g C(=O)OR h , -NR g SO 2 R h , -OC(=O)N R g R h , -OR g , -SR g , -SOR g , -SO 2 R g , -OSO 2 R g , -SO 2 OR g , =NSO 2 R g , and -SO 2 NR g R h .

“Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=O)R g , -C(=O)OR g , -C(=O)NR g R h , -CH 2 SO 2 R g , -CH 2 SO 2 NR g R h . In the foregoing, R g and R h are the same or different and independently hydrogen, alkyl, alkoxy, alkylaminyl, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N- heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or heteroarylalkyl.

“Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyl, heteroaryl, N-heteroaryl and/or

heteroarylalkyl group. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.

“Prodrug” is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term“prodrug” refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when

administered to a subject in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a

mammalian organism (see, Bundgard, H., Design of Prodrugs (1985), pp.7-9, 21-24 (Elsevier, Amsterdam)). A discussion of prodrugs is provided in Higuchi, T., et al., A.C.S. Symposium Series, Vol.14, and in Bioreversible Carriers in Drug Design, Ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.

The term“prodrug” is also meant to include any covalently bonded carriers, which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amide derivatives of amine functional groups in the compounds of the invention and the like.

Certain embodiments of the invention disclosed herein are meant to encompass all pharmaceutically acceptable compounds of structure (I) being isotopically- labeled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively. These radiolabelled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labeled compounds of structure (I), 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 particularly 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 11 C, 18 F, 15 O and 13 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.

Embodiments of the invention disclosed herein are also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabelled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.

“Stable compound” and“stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.

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

“Optional” or“optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example,“optionally substituted aryl” means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.

“Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.

“Pharmaceutically acceptable salt” includes both acid and base addition salts.

“Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2- dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2- hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo- glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.

“Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.

Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine,

ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol,

dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine,

N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.

Often crystallizations produce a solvate of the compound of the invention. As used herein, the term“solvate” refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.

A“pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.

“Effective amount” or“therapeutically effective amount” refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a HAT related condition or disease in the mammal, preferably a human. The amount of a compound of the invention which constitutes a“therapeutically effective amount” will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.

“Treating” or“treatment” as used herein covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes: (i) preventing the disease or condition from occurring in a mammal, in particular, when such mammal is predisposed to the condition but has not yet been diagnosed as having it;

(ii) inhibiting the disease or condition, i.e., arresting its development; (iii) relieving the disease or condition, i.e., causing regression of the disease or condition; or

(iv) relieving the symptoms resulting from the disease or condition, i.e., relieving pain without addressing the underlying disease or condition. As used herein, the terms“disease” and“condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.

The compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefinic double bonds or other centres of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. A“stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes“enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.

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

The chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C. nomenclature system, using the ACD/Name Version 9.07 software program and/or ChemDraw Ultra Version 11.0.1 software naming program (CambridgeSoft). For complex chemical names employed herein, a substituent group is generally named before the group to which it attaches. For example, cyclopropylethyl comprises an ethyl backbone with cyclopropyl substituent. Unless specifically stated otherwise, all bonds are identified in the chemical structure diagrams herein, except for some carbon atoms, which are assumed to be bonded to sufficient hydrogen atoms to complete the valency. I. Compounds

As noted above, the present disclosure provides a compound having the followin structure I :

X is–NH- or -O-;

Z is a direct bond or–C(R 7a )(R 7b )-;

R 1 is carbocyclyl or heterocyclyl;

R 2a and R 2b are each independently H, D or C 1 -C 6 alkyl;

R 3a is carbocyclyl or heterocyclyl and R 3b is C 1 -C 6 alkyl or carbocyclyl; or R 3a and R 3b are each independently C 1 -C 6 alkyl; or R 3a and R 3b are taken together with the carbon to which they are bound to form a carbocyclic or heterocyclic ring;

R 3c is H or D;

R 4a and R 4b are each independently H, D or C 1 -C 6 alkyl;

R 5 is carbocyclyl or heterocyclyl;

R 6 is H or D when Z is a direct bond; or R 6 is H, D or C 1 -C 6 alkyl when Z is -C(R 7a )(R 7b )-;

R 7a and R 7b are each independently H, D or C 1 -C 6 alkyl;

with the proviso that R 3a , R 3b and R 3c are not unsubstituted cyclopropyl, methyl and H, respectively, when Z is–CH 2 -, R 1 is unsubstituted phenyl and R 5 is unsubstituted indolyl.

In some embodiments, X is -NH-. In other embodiments, X is–O-.

In some more embodiments of any of the foregoing, Z is a direct bond and R 6 is H or D. In some of these embodiments, R 6 is H. In other of these embodiments, R 6 is D.

In different embodiments of the foregoing, Z is–C(R 7a )(R 7b )- and R 6 is H, D or C 1 -C 6 alkyl, for example in some of these embodiments Z is–CH 2 -. In other embodiments, R 7a is H and R 7b is C 1 -C 6 alkyl. For example, in some embodiments R 7a and R 7b are each independently C 1 -C 6 alkyl. In still other exemplary embodiments at least one of R 7a or R 7b is D. In other of the foregoing embodiments, R 6 is H. In other embodiments, R 6 is D. In still other embodiments, R 6 is C 1 -C 6 alkyl, such as methyl.

In other of the foregoing embodiments, R 1 is unsubstituted. In still other of the foregoing embodiments, R 1 is substituted with one or more substituents, for example in some embodiments the substituents are selected from halo, nitrilyl and C 1 -C 6 alkyl. In other more specific embodiments, the substituents on R 1 are selected from fluoro, nitrilyl and methyl. Other embodiments include compounds wherein R 1 is substituted with one or more deuterium atoms.

In more embodiments of any of the foregoing, R 1 is carbocyclyl. For example, in some embodiments R 1 is aryl, such as phenyl. In certain embodiments, R 1 has one of the followin structures:

.

In still other embodiments of the foregoing, R 1 is heterocyclyl. For example, in certain embodiments R 1 is tetrahydropyranyl.

In other embodiments of the foregoing, R 1 is heteroaryl. In certain of these embodiments, the heteroaryl is furanyl, benzodioxazolyl or imidazolyl.

In some other embodiments of the foregoing, R 1 has one of the following structures: . In some of any of the foregoing embodiments, at least one of R 2a or R 2b is H. In some embodiments, each of R 2a and R 2b is H.

In other of the foregoing embodiments, at least one of R 2a or R 2b is C 1 -C 6 alkyl.

In still other of the foregoing embodiments, at least one of R 2a or R 2b is D. In other of any of the foregoing embodiments, R 3a is carbocyclyl and R 3b is C 1 -C 6 alkyl. For example, in some embodiments R 3a is cycloalkyl, such as cyclopropyl or cyclobutyl. In still more of these embodiments, R 3a is substituted with C 1 -C 6 alkyl, such as methyl.

In other of any of the foregoing embodiments, R 3a and R 3b are each independently carbocyclyl. In some of these embodiments, R 3a and R 3b are each independently cycloalkyl. For example, in some embodiments the cycloalkyl is cyclopropyl or cyclobutyl.

In other of the foregoing embodiments, R 3a and R 3b are each independently C 1 -C 6 alkyl. For example, in some embodiments R 3a and R 3b are each ethyl.

In still more of the foregoing embodiments, R 3a and R 3b are taken together with the carbon to which they are bound to form a carbocyclic or heterocyclic ring. In some more specific embodiments, R 3a and R 3b are taken together with the carbon to which they are bound to form a heterocyclic ring. In some of these embodiments, the heterocyclic ring is tetrahydropyranyl.

In still more embodiments of any of the foregoing, R 3c is H. In other emboldens, R 3c is D.

In yet other embodiments of any of the foregoing, at least one of R 4a or R 4b is H. For example, in some embodiments each of R 4a and R 4b is H.

In still other embodiments of any of the foregoing, at least one of R 4a or R 4b is C 1 -C 6 alkyl.

In more embodiments of the foregoing, at least one of R 4a or R 4b is D.

Still more embodiments of any of the foregoing embodiments include compounds wherein R 5 is unsubstituted.

In other embodiments, R 5 is substituted with one or more substituents. In some of these embodiments, the substituents are selected from halo, C 1 -C 6 alkyl and alkoxy. For example, certain specific embodiments provide compounds wherein R 5 is substituted with substituents selected from fluoro, chloro, methyl and methoxy.

In still more embodiments of the foregoing embodiments, R 5 is carbocyclyl. For example, in some embodiments, R 5 is aryl. In other embodiments the aryl is phenyl, and in other embodiments the aryl is naphthyl.

In still other embodiments of the foregoing, R 5 has one of the following structures:

In still other aspects, R 5 is heterocyclyl. For example, in some embodiments R 5 is heteroaryl. More specific embodiments include those wherein R 5 is furanyl, indolyl, indazolyl, pyridinyl, pyridinyl oxide, imidazolyl, pyrazolyl, benzofuranyl or benzothiophenyl.

Some embodiments of the invention also include deuterium substituted compounds of structure (I). The deuterium may be included at various positions in the compound, for example in some embodiments one or more of R 2a , R 2b , R 4a and/or R 4b are deuterium. In other embodiments, substituents, such as R 1 , are substituted with one or more deuterium atoms. While not wishing to be bound by theory, such deuterium substitutions may contribute to advantageous metabolism of the compounds.

In other particular embodiments of the compounds as described anywhere herein, the compound is a compound selected from one of the compounds in Table 1. Table 1

Representative Compounds

H N O cycopropyety acetam e O

It is understood that any embodiment of the compounds of structure (I), as set forth above, and any specific substituent or value set forth herein for R 1 , R 2a , R 2b , R 3a , R 3b , R 3c , R 4a , R 4b , R 5 , R 6 , Z and/or X in the compounds of structure (I), as set forth above, may be independently combined with other embodiments and/or substituents and/or values of the above variables of compounds of structure (I) to form embodiments of the inventions not specifically set forth above. In addition, in the event that a list of choices is listed for any particular R group or other variable in a particular embodiment and/or claim, it is understood that each individual choice may be deleted from the particular embodiment and/or claim and that the remaining list of choices will be considered to be within the scope of the invention.

It is understood that in the present description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.

The compounds of the present invention (i.e., compounds of Structure I) may contain one or more asymmetric centers. Compounds as described herein include all stereoisomers. Accordingly, the compounds include racemic mixtures, enantiomers and diastereomers of any of the compounds described herein. Tautomers of any of the compounds described herein are also included within the scope of the invention.

Accordingly, in some embodiments the compounds are mixtures of different enantiomers (e.g., R and S) or different diastereomers. In other embodiments, the compounds are pure (or enriched) enantiomers or diastereomers. For purpose of clarity, the chiral carbons are not always depicted in the compounds; however, the present invention includes all stereoisomers (pure and mixtures) of all compounds of Structure I.

By way of example, certain embodiments of the compounds of Structure I, contain at least one stereocenter. For example, in some embodiments the compounds have one of the following structures (Ia) or (Ib):

(Ia) (Ib)

In other embodiments, the compounds have one of the following structures

(Ic) (Id)

In still other embodiments, the compounds comprise at least two stereocenters. For example, in some embodiments the compounds have one of the following structures (Ie), (If), (Ig) or (Ih):

(Ie) (If)

(Ig) (Ih) In an analogous fashion, the invention includes all possible stereoisomers of all the foregoing compounds, including the compounds provided in Table 1. One of ordinary skill in the art will readily understand how to derive all possible stereoisomers, especially in reference to the above exemplary compounds of Structure I.

General Reaction Scheme I illustrates an exemplary method of making compounds of this invention, i.e., compound of structure (I). Variations of General Reaction Scheme I and alternative methods of making the compounds of the invention are described in more detail in the Examples.

General Reaction Scheme I

Referring to General Reaction Scheme I, compounds of structure A and B are reacted under reductive amination conditions to yield compounds of structure C.

Compounds of structures A and B can be purchased or prepared according to methods known in the art. Compounds wherein R 2b is C 1 -C 6 alkyl, can be prepared using appropriate alkylation procedures known in the art. Furthermore, enantiomerically pure or racemic compound of structure B can be used depending on the desired product.

In a parallel reaction pathway, compounds of structure D can be purchased or prepared according to methods well-known to those of ordinary skill in the art. Examples of such methods are provided in the Examples. Reaction of D with an appropriate reagent results in E. In embodiments where A is–NH-, appropriate reagents include ammonium carbonate and potassium cyanide. Reaction of E with an appropriate reagent (e.g.

bromoacetate F), followed by deprotection results in compounds of structure G. Reaction of C with G under appropriate amide forming conditions (e.g., HATU and the like) results in compounds of structure I.

Various different compounds of structure (I) can be prepared according to the above general description. For example, compounds of structure (I), wherein X is– O- can be prepared according to the above General Reaction Scheme by modifying the scheme to include the preparation of intermediate 39 as described in Example 1. Various other alternative preparations of the compounds of structure (I), including those described in the Examples, are well-known to those of ordinary skill in the art

It is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other compounds of structure (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. For example, in various embodiments the substituents R 6 and/or R 7 are installed after the spiro cyclic ring structure is assembled) In these embodiments, it may be useful to include a bromide or other suitable leaving group in the compound of structure A so that the compounds can be further functionalized. Single stereoisomers at the spiro cyclic ring juncture can be prepared by chiral (e.g., enzymatic) hydrolysis of compounds of structure B. Recyclization and preparation of compounds of structure I using similar conditions is then possible. Such methods are provided in more detail in the Examples.

In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and

Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared as described in this invention.

It will also be appreciated by those skilled in the art that in the processes described herein the functional groups of intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkylsilyl or diarylalkylsilyl (for example, t-butyldimethylsilyl, t-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butoxycarbonyl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(O)-R” (where R” is alkyl, aryl or arylalkyl),

p-methoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or arylalkyl esters. Protecting groups may be added or removed in accordance with standard techniques, which are known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and P.G.M. Wutz, Protective Groups in Organic Synthesis (1999), 3rd Ed., Wiley. As one of skill in the art would appreciate, the protecting group may also be a polymer resin such as a Wang resin, Rink resin or a 2-chlorotrityl-chloride resin.

It will also be appreciated by those skilled in the art, although such protected derivatives of compounds of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the body to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as“prodrugs”. All prodrugs of compounds of this invention are included within the scope of the invention.

Furthermore, all compounds of the invention which exist in free base or acid form can be converted to their pharmaceutically acceptable salts by treatment with the appropriate inorganic or organic base or acid by methods known to one skilled in the art. Salts of the compounds of the invention can be converted to their free base or acid form by standard techniques. II. Pharmaceutical Compositions and Administration

For the purposes of administration, the compounds of the present invention may be administered as a raw chemical or may be formulated as pharmaceutical compositions. In some embodiments, the present disclosure provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a compound having the following structure (I):

or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, wherein:

X is–NH- or -O-;

Z is a direct bond or–C(R 7a )(R 7b )-;

R 1 is carbocyclyl or heterocyclyl;

R 2a and R 2b are each independently H, D or C 1 -C 6 alkyl;

R 3a is carbocyclyl or heterocyclyl and R 3b is C 1 -C 6 alkyl or carbocyclyl; or R 3a and R 3b are each independently C 1 -C 6 alkyl; or R 3a and R 3b are taken together with the carbon to which they are bound to form a carbocyclic or heterocyclic ring;

R 3c is H or D;

R 4a and R 4b are each independently H, D or C 1 -C 6 alkyl;

R 5 is carbocyclyl or heterocyclyl;

R 6 is H, D or C 1 -C 6 alkyl; and

R 7a and R 7b are each independently H, D or C 1 -C 6 alkyl.

In more specific embodiments, the compound is any one of the compounds described in the foregoing section entitled“compounds.”

The compound of structure (I) is present in the composition in an amount which is effective to treat a particular disease or condition of interest - that is, in an amount sufficient to inhibit HAT activity, and preferably with acceptable toxicity to the patient. HAT activity of compounds of structure (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art.

Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. The term parenteral as used herein includes subcutaneous injections, intravenous,

intramuscular, intrasternal injection or infusion techniques. Pharmaceutical compositions of the invention are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition

(Philadelphia College of Pharmacy and Science, 2000). The composition to be

administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this invention.

A pharmaceutical composition of the invention may be in the form of a solid or liquid. In one aspect, the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form. The carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inhalatory administration.

When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.

As a solid composition for oral administration, the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition will typically contain one or more inert diluents or edible carriers. In addition, one or more of the following may be present: binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide;

sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.

When the pharmaceutical composition is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.

The pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension. The liquid may be for oral administration or for delivery by injection, as two examples. When intended for oral administration, preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer. In a composition intended to be administered by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.

The liquid pharmaceutical compositions of the invention, whether they be solutions, suspensions or other like form, may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer’s solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Physiological saline is a preferred adjuvant. An injectable pharmaceutical composition is preferably sterile.

A liquid pharmaceutical composition of the invention intended for either parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage will be obtained.

The pharmaceutical composition of the invention may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base. The base, for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers. Thickening agents may be present in a pharmaceutical composition for topical administration. If intended for transdermal administration, the composition may include a transdermal patch or iontophoresis device.

The pharmaceutical composition of the invention may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug. The composition for rectal administration may contain an oleaginous base as a suitable non-irritating excipient. Such bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.

The pharmaceutical composition of the invention may include various materials, which modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating shell around the active ingredients. The materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents. Alternatively, the active ingredients may be encased in a gelatin capsule.

The pharmaceutical composition of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.

The pharmaceutical composition of the invention may consist of dosage units that can be administered as an aerosol. The term aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the invention may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols.

The pharmaceutical compositions of the invention may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution. A surfactant may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.

The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.

Compounds of the invention, or pharmaceutically acceptable derivatives thereof, may also be administered simultaneously with, prior to, or after administration of one or more other therapeutic agents. Such combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of the invention and one or more additional active agents, as well as administration of the compound of the invention and each active agent in its own separate pharmaceutical dosage formulation. For example, a compound of the invention and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations. Where separate dosage formulations are used, the compounds of the invention and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens. III. Methods

Diseases, Disorders, and Conditions

The compounds for inhibiting the activity of p300/CBP disclosed herein can be useful in analyzing p300/CBP signaling activity in model systems and for preventing, treating, or ameliorating of a symptom associated with a disease, disorder, or pathological condition involving p300/CBP, preferably one afflicting humans. A compound which inhibits the activity of p300/CBP will be useful in preventing, treating, ameliorating, or reducing the symptoms or progression of cancer, cardiac disease, metabolic disease, fibrotic disease, inflammatory disease, or viral infections. The present invention provides methods for inhibiting p300/CBP comprising administering the compounds described herein in a therapeutically effective amount to a subject in need thereof. A subject may be a human, non-human primate, rodent, canine, feline, ungulate, bovine, equine, or other species. A wide variety of cancers, including solid tumors and leukemias are amenable to the compositions and methods disclosed herein. Types of cancer that may be treated include, but are not limited to: adenocarcinoma of the breast, prostate, and colon; all forms of bronchogenic carcinoma of the lung; myeloid; melanoma; hepatoma;

neuroblastoma; papilloma; apudoma; choristoma; branchioma; malignant carcinoid syndrome; carcinoid heart disease; and carcinoma (e.g., Walker, basal cell, basosquamous, Brown-Pearce, ductal, Ehrlich tumor, Krebs 2, merkel cell, mucinous, non-small cell lung, oat cell, papillary, scirrhous, bronchiolar, bronchogenic, squamous cell, and transitional cell). Additional types of cancers that may be treated include: histiocytic disorders;

leukemia; histiocytosis malignant; Hodgkin's disease; immunoproliferative small; non- Hodgkin's lymphoma; plasmacytoma; reticuloendotheliosis; melanoma; chondroblastoma; chondroma; chondrosarcoma; fibroma; fibrosarcoma; giant cell tumors; histiocytoma; lipoma; liposarcoma; mesothelioma; myxoma; myxosarcoma; osteoma; osteosarcoma; chordoma; craniopharyngioma; dysgerminoma; hamartoma; mesenchymoma;

mesonephroma; myosarcoma; ameloblastoma; cementoma; odontoma; teratoma; thymoma; trophoblastic tumor. Further, the following types of cancers are also contemplated as amenable to treatment: adenoma; cholangioma; cholesteatoma; cyclindroma;

cystadenocarcinoma; cystadenoma; granulosa cell tumor; gynandroblastoma; hepatoma; hidradenoma; islet cell tumor; Leydig cell tumor; papilloma; sertoli cell tumor; theca cell tumor; leimyoma; leiomyosarcoma; myoblastoma; myomma; myosarcoma; rhabdomyoma; rhabdomyosarcoma; ependymoma; ganglioneuroma; glioma; medulloblastoma;

meningioma; neurilemmoma; neuroblastoma; neuroepithelioma; neurofibroma; neuroma; paraganglioma; paraganglioma nonchromaffin. The types of cancers that may be treated also include, but are not limited to, angiokeratoma; angiolymphoid hyperplasia with eosinophilia; angioma sclerosing; angiomatosis; glomangioma; hemangioendothelioma; hemangioma; hemangiopericytoma; hemangiosarcoma; lymphangioma;

lymphangiomyoma; lymphangiosarcoma; pinealoma; carcinosarcoma; chondrosarcoma; cystosarcoma phyllodes; fibrosarcoma; hemangiosarcoma; leiomyosarcoma; leukosarcoma; liposarcoma; lymphangiosarcoma; myosarcoma; myxosarcoma; ovarian carcinoma;

rhabdomyosarcoma; sarcoma; neoplasms; nerofibromatosis; and cervical dysplasia.

In a particular embodiment, the present disclosure provides for methods of treating colon cancer, gastric cancer, thyroid cancer, lung cancer, leukemia, pancreatic cancer, melanoma, multiple melanoma, brain cancer, CNS cancer, renal cancer, prostate cancer, ovarian cancer, leukemia, or breast cancer.

Another aspect of the present disclosure provides for using the p300/CBP inhibitory compositions disclosed herein to treat, prevent, or ameliorate a symptom associated with a chronic inflammatory disorder or condition, including but not limited to asthma, inflammatory bowel disease (Crohn’s disease or ulcerative colitis), chronic obstructive pulmonary disease, rheumatoid arthritis, and psoriasis.

Another aspect of the present disclosure provides for methods of treating, preventing, or ameliorating a symptom associated with a viral infection, including, but not limited to human immunodeficiency virus, hepatitis C virus, and human papilloma virus.

Yet another aspect of the present disclosure provides for methods of treating, preventing, or ameliorating a symptom associated with metabolic disease, including but not limited to: obesity, hepatic steatosis, dyslipidemia, hypertension, coronary heart disease, hepatic inflammation, and diabetes mellitus type 2.

Another aspect of the present disclosure provides for methods of treating, preventing, or ameliorating a symptom associated with a fibrotic disease or disorder. Fibrotic diseases and disorders include, for example, radiation-induced pneumonitis, radiation fibrosis, acute respiratory distress syndrome, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, interstitial lung disease, myocardial infarction, ischemic stroke, ischemic kidney disease, transplant rejection, Leishmaniasis, type I diabetes, rheumatoid arthritis, chronic hepatitis, cirrhosis, inflammatory bowel disease, Crohn’s disease, scleroderma, keloid, post-operative fibrosis, chemotherapy induced fibrosis (e.g., chemotherapy induced pulmonary fibrosis or ovarian cortical fibrosis), nephrogenic systemic fibrosis, retroperitoneal fibrosis, myelofibrosis, mediastinal fibrosis, cystic fibrosis, asbestosis, asthma, and pulmonary hypertension. Another aspect of the present disclosure provides for methods of treating, preventing, or ameliorating a symptom associated with cardiac disease, including but not limited to cardiac hypertrophy and heart failure.

The present disclosure also relates to methods of treating, preventing, or ameliorating a symptom associated with a disease, disorder, or pathological condition involving p300/CBP comprising administering the compounds described herein in a therapeutically effective amount to a subject in need thereof as part of a combination therapy. It is apparent to a person of skill in the medical arts that agent(s) administered with the p300/CBP compounds disclosed herein are selected based upon the subject’s disease, disorder, or pathological condition.

Accordingly, various embodiments of the present disclosure are directed to a method for treating a HAT dependent condition in a mammal in need thereof, the method comprising administering an effective amount of the pharmaceutical composition described herein (i.e., as described in the foregoing section entitled“Pharmaceutical Compositions and Administration”). In some embodiments, the condition is cancer, metabolic disease, neurodegenerative disorders or inflammation. In some more specific embodiments, the condition is cancer (e.g., the various cancers described herein above). Assays for Detecting HAT Activity

Methods for detecting HAT activity are well known in the art, and a variety of HAT assay kits are commercially available. For example, filter-binding assays measure the transfer of radiolabeled acetate from acetyl-CoA to protein, and continuous, spectroscopic enzyme coupled assays link the HAT reaction to the reduction of NAD+ by pyruvate or α-ketoglutarate dehydrogenase (Berndsen and Denu, 2005, Methods 36:321- 333).

Scintillation proximity assays (SPA) using low energy radioisotopes permit rapid and sensitive analysis of a wide range of biological processes and is well suited for high-throughput screening of HAT inhibitors (see, e.g., Aherne et al., 2002, Methods 26:245-253; Turlais et al., 2001, Anal. Biochem.298:62-68). In brief, beads or plates coated with capture molecule (target) is incubated with radiolabeled ligand. When the radiolabeled ligand is attached or in proximity to bead or plate surface, light emission is stimulated and measured by a photometer.

In a particular embodiment, compounds disclosed herein may be screened by using a radiolabel that is incorporated into a biotinylated form of the substrate as a result of an enzymatic reaction (i.e., acetylation). The reaction contents are then incubated on a specially manufactured multi-well plate (Perkin-Elmer), where the wells have been precoated with avidin and a scintillant. Alternatively, beads precoated with avidin and scintillant may be used instead of plates. The tight interaction of biotin-avidin complexes brings the radiolabel on the reaction product in close proximity to the scintillant, resulting in emission of a light signal. The need for the proximity of the radiolabel to the scintillant to generate a signal enables a rapid readout without elaborate post-assay work up. The interference from unreacted (hence free) radiolabel is minimal.

Particular methods for testing the activity of the compounds of the invention are described in more detail in the examples.

The following examples are provided for purpose of illustration and not limitation.

EXAMPLE 1

PREPARATION OF SYNTHETIC INTERMEDIATES

The following intermediates are useful for preparation of the disclosed compounds. Intermediate 11

N-benzyl-1-cyclobutylethanamine (Int-11):

To a stirring solution of phenylmethanamine SM1 (3 g, 27.6 mmol) and 1- cyclobutylethanone (2.7 g, 27.6) in dry THF (30 mL) was added tetraisopropoxytitanium (9.4 g, 33.1 mmol) at room temperature. The reaction mixture was stirred at room temperature for 2h then NaBH 4 (2.1 g, 55.2 mmol) was added. The reaction mixture was stirred at room temperature overnight then quenched with water (50 ml). The mixture was filtrated and washed with EtOAc, the filtrate was concentrated and purified by silica gel column chromatography eluting with 25% EtOAc/Hexane to afford compound 11 (1.5 g, 30%).

LC-MS: m/z = 190.1[(M + +1)] Intermediate 13

To a stirring solution of N-benzyl-1-cyclopropylethanamine Int-14 (10.0 g,57.14 mmol) and 2-bromoacetyl bromide (11.6 g, 57.14 mmol) in DCM (150 mL) was added dropwise pyridine (7.63 g, 85.71 mmol) and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 30% EtOAc /PE to afford Intermediate 13 (8.8 g, 52.4%) as an off-white solid. Intermediate 14

Int-14

To a solution of phenylmethanamine (10.7 g, 100 mmol) and 1- cyclopropylethanone 2 (8.4 g, 100 mmol) in MeOH (200 mL) was added Titanium tetraisopropanolate (31.24 g, 110 mmol) and stirred at room temperature for 4 h. Then NaBH 4 (7.6 g, 200 mmol) was added and stirred at room temperature for 2 h. Diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous MgSO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting (EtOAc/Hexane 1: 1) to afford intermediate 14 ( 11.7 g, 66%) as oil. Intermediate 16

(E)-N-(cyclobutylmethylene)-2-methylpropane-2-sulfinamide (1):

To a stirring solution of SM1 (9.7 g, 115 mmol) in DCM (100 mL) was (S)- 2-methylpropane-2-sulfinamide (11.1 g, 92 mmol) and PPTS (0.723 g, 2.88 mmol) followed by MgSO 4 (34.5 g, 288 mmol) at room temperature. The reaction mixture was stirred at room temperature overnight. After consumption of the starting material (by TLC), the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 14% EtOAc / PE to afford compound 1 (14.32 g, 83%) as a clear oil. LC-MS: m/z = 188[(M + +1)]

To a stirring solution of compound 1 (14.32 g, 76.6 mmol) in DCM (150 mL) was added 3M methylmagnesium bromide in Et 2 O (61.3 mL, 183.8 mmol) dropwise at -50 ^ under nitrogen atmosphere. The reaction mixture was stirred at -48 ^ for 3 h and stirred at room temperature overnight. The resulting mixture was quenched by sat. NH 4 Cl solution, extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 50% EtOAc / PE to afford Intermediate 16 (14.8 g, 95%) as a clear oil. Intermediate 17

Intermediate 17

(E)-N-(cyclobutylmethylene)-2-methylpropane-2-sulfinamide (1):

To a stirring solution of SM1 (2 g, 23.8 mmol) in DCM (100 mL) was (R)- 2-methylpropane-2-sulfinamide (1.44 g, 11.9 mmol) and PPTS (0.149 g, 0.595 mmol) followed by MgSO 4 (7.14 g, 59.5 mmol) at room temperature. The reaction mixture was stirred at room temperature overnight. After consumption of the starting material (by TLC), the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 14% EtOAc / PE to afford compound 1 (2.27 g, 51%) as a clear oil.

LC-MS: m/z = 188[(M + +1)] To a stirring solution of compound 1 (2.27 g, 12.1 mmol) in DCM (20 mL) was added 3M methylmagnesium bromide in Et 2 O (9.68 mL, 29.04 mmol) dropwise at -50 ^ under nitrogen atmosphere. The reaction mixture was stirred at -48 ^ for 4 h and stirred at room temperature overnight. The resulting mixture was quenched by sat. NH 4 Cl solution, extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 50% EtOAc / PE to afford Intermediate 17 (1.71 g, 70%) as a clear oil. Intermediate 23 and 24

(S)-N-benzyl-1-cyclopropylethanamine (Int-24):

A mixture of (S)-1-cyclopropylethanamine SM1 (850 mg, 10.0 mmol) and benzaldehyde SM2 (1060 mg, 1.0 mmol) in methanol (8 mL) and 1,2-dichloroethane (8 mL) was stirred at rt for 1 h. NaBH 3 CN (2.6 g, 12 mmol) was added and the mixture was stirred at 40 °C for 1h. The reaction mixture was poured to NaHCO 3 (sat, aq, 10 mL) and then extracted with chloroform (20 mL*2). The organic phase was combined and concentrated under reduced pressure to obtain crude product, which was purified by column chromatography (dichloromethane-methanol=100:1) to afford (S)-N-benzyl-1- cyclopropylethanamine Int-24 (1.3 g, 74%). 1 H NMR (300 MHz, CDCl 3 ): δ 7.46-7.17 (m, 5H), 3.84 (s, 2H), 1.98-1.81 (m, 1H), 1.64 (brs, 1H), 1.19 (d, J = 6.3 Hz, 3H), 0.86-0.71 (m, 1H), 0.60-0.37 (m, 2H), 0.23-0.02 (m, 2H).

(S)-N-benzyl-2-bromo-N-(1-cyclopropylethyl)acetamide (Int-23): To a stirring solution of (S)-N-benzyl-1-cyclopropylethanamine Int-24 (200 mg, 1.1 mmol) in dichloromethane (6 mL) was added a solution of 2-bromoacetyl bromide (303 mg, 1.5 mmol) in dichloromethane (1 mL) at 0 °C by dropwise. The mixture was stirred at room temperature for 1 h. The reaction mixture was poured to water (10 mL) and then extracted with chloroform (20 mL*2). The organic phase was combined and concentrated under reduced pressure to obtain crude product, which was purified by column chromatography (dichloromethane-methanol=100:1) to afford (S)-N-benzyl-2- bromo-N-(1-cyclopropylethyl)acetamide Int-23 (225 mg, 70%) as oil. 1 H NMR (300 MHz, CDCl 3 ): δ 7.35-7.25 (m, 6H), 4.68 (s, 2H), 4.05-3.80 (m, 2H), 3.30-3.20 (m, 1H), 1.28- 1.20 (m, 3H), 0.87-0.79 (m, 1H), 0.63-0.48 (m, 1H), 0.45-0.15 (m, 3H). Intermediate 29

To a stirring solution of Intermediate 16 (500 mg, 2.46 mmol) in MeOH (2 mL) was added 2 M HCl/dioxane (3 mL). The reaction mixture was stirred at room temperature for 0.5 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude Intermediate 29 (300 mg, 90%) as a white solid. Intermediate 39

To a stirring solution of 4-chloro-1H-indole SM1 (1.5 g, 10mmol) in EtOH (40ml), was added compound 1 (1.42g, 10mmol), followed by 1N HCl (10ml) at room temperature. The reaction mixture was heated to reflux for 15 min. After consumption of the starting material (by TLC), the reaction mixture concentrated under reduced pressure to obtain crude product, which washed with water to afford compound 2 (2 g, 68%). TLC: 50% EA/PE (R f : 0.2). LC-MS: m/z = 294 [(M + +1)]. 5-(4-chloro-1H-indol-3- yl)oxazolidine-2,4-dione (3): (LNB No: b120629-091-1):

A solution of 5-(4-chloro-1H-indol-3-yl)-5-hydroxypyrimidine- 2,4,6(1H,3H,5H)-trione compound 1 (2g, 6.8 mmol), in 1N NaOH (40 mL) was stirred for 15min at reflux . Adjust the PH<7 with 1N HCl, extracted with EA, Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 30% EA/PE to afford compound 3 (1g, 58.8%). TLC: 30% EA/PE (R f : 0.3). LC-MS: m/z = 251 [(M + +1)]. EXAMPLE 2

To a suspension of 1 H-indole-3-carbaldehyde 1 (7.26 g, 50 mmol) in EtOH (50 mL) and H 2 O (50 mL) was added KCN (4.7g, 75 mmol) and (NH 3 ) 2 CO 3 (14.4g, 150 mmol), the reaction mixture was stirred in steel tube at 80 °C for 72 h. Cooled to room temperature and the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting (EtOAc/Hexane 3: 1) to afford compound 2 (6.8 g, 63%) as brown solid. LC-MS: m/z = 216.1 [(M + +1)] (95% purity).

To a mixture of 5-( 1 H-indol-3-yl)imidazolidine-2,4-dione 2 (2.15 g, 10 mmol) and tert-butyl 2-bromoacetate 3 (1.95 g, 10 mmol) in MeCN (50 mL) was added DIPEA (2.58 g, 20 mmol) and stirred at room temperature for overnight. Concentrated under reduced pressure to obtain crude product, which was purified by combiflsah eluting (EtOAc/Hexane 2: 1) to afford compound 4 (1.68 g, 51%) as brown solid. LC-MS: m/z = 273.9[(M + -55)] (95% purity).

To a solution of tert-butyl 2-(4-( 1 H-indol-3-yl)-2,5-dioxoimidazolidin-1- yl)acetate 4 (1.68 g, 5.1 mmol) in DCM (20 mL) was added TFA (20 mL) and stirred at room temperature for 2 h. After consumption of the starting material (by TLC) and concentrated under reduced pressure to obtain crude compound 5 (1.5 g, 108%) as brown solid. LC-MS: m/z = 274.1[(M + +1)] (85% purity).

To a solution of 2-(4-( 1 H-indol-3-yl)-2,5-dioxoimidazolidin-1-yl)acetic acid 5 (273 mg, 1 mmol) and intermediate 14 (175 mg, 1 mmol) in DMF (10 mL) was added DIPEA (516 mg, 4 mmol) under N2 and stirred at room temperature for 15 min. Then HATU (570 mg, 1.5 mmol) was added and stirred at room temperature for 1 h. After consumption of the starting material (by TLC) diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by combiflash eluting (EtOAc/Hexane 1: 1) to afford A-0001947 (280 mg, 65%) as white solid. The compound 2- 1 (150mg) was purified by Chiral-HPLC to give P1 (14 mg) and P2 (14 mg). P1: LC-MS: m/z = 431.2[(M++1)] at room temperature 6.67 (89.47% purity); P2: LC-MS: m/z = 431.2[(M++1)] at room temperature 7.37 (99.47% purity). P1: 1 H NMR (400MHz, CD3OD): δ 7.64 (d, J = 8.0 Hz, 1 H), 7.48 (d, J = 7.6 Hz, 1 H), 7.46– 7.36 (m, 3H), 7.35– 7.25 (m, 2H), 7.16 (s, 1 H), 7.13 (d, J = 8.0 Hz, 1 H), 7.05 (t, J = 7.5 Hz, 1 H), 5.55 (d, J = 16.2 Hz, 1 H), 4.83 (s, 1 H), 4.77 (d, J = 9.7 Hz, 1 H), 4.56 (d, J = 3.6 Hz, 1 H), 4.45– 4.36 (m, 1 H), 3.85 (dd, J = 9.7, 7.0 Hz, 1 H), 1.28 (dt, J = 34.2, 5.5 Hz, 3H), 0.96 (s, 1 H), 0.66– 0.49 (m, 1 H), 0.45– 0.16 (m, 3H); P2: 1 H NMR (400MHz, CD3OD): δ 7.64 (d, J = 7.9 Hz, 1 H), 7.48 (d, J = 7.6 Hz, 1 H), 7.45– 7.36 (m, 3H), 7.36– 7.25 (m, 2H), 7.25– 7.10 (m, 2H), 7.04 (t, J = 7.5 Hz, 1 H), 5.55 (d, J = 15.5 Hz, 1 H), 4.80 (d, J = 19.6 Hz, 2H), 4.64 (d, J = 16.6 Hz, 1 H), 4.54– 4.31 (m, 2H), 3.93– 3.79 (m, 1 H), 1.28 (dd, J = 33.7, 6.7 Hz, 3H), 0.98 (d, J = 8.3 Hz, 1 H), 0.66– 0.53 (m, 1 H), 0.33 (tdd, J = 14.5, 10.2, 5.8 Hz, 3H). EXAMPLE 3

3-1 To a stirring solution of SM1 (3 g, 20 mmol) in EtOH (30 mL), H 2 O (30 mL) was added (NH 4 ) 2 CO 3 (5.76 g, 60 mmol) followed by potassium cyanide (2 g, 30.8 mmol) at room temperature. The reaction mixture was heated to 60 °C overnight. After consumption of the starting material (by TLC), the reaction mixture was cooled to room temperature, concentrated under reduced pressure to obtain crude product, which was washed by water, dried to afford compound 1 (3.82 g, 87%) as a white solid. LC-MS: m/z = 221[(M + +1)]

To a stirring solution of compound 1 (0.88 g, 4 mmol) in DMF (5 mL) was added K 2 CO 3 (1.104 g, 8 mmol) followed by tert-butyl 2-bromoacetate (0.858 g, 4.4 mmol) at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was washed by water, dried to afford compound 2 (1.082 g, 81%) as a white solid. LC- MS: m/z = 279[(M + -55)]

To a stirring solution of compound 2 (1.051 g, 3.15 mmol) in DCM (3 mL) was added TFA (3 mL) at room temperature and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude compound 3 (0.874 g, 99%) as a white solid.

To a stirring solution of compound 3 (200 mg, 0.719 mmol) in DMF (2 mL) was added intermediate 14 (126 mg, 0.719 mmol) and DIPEA (463.8 mg, 3.595 mmol) followed by HATU (546.4 mg, 1.438 mmol)at room temperature and stirred at room temperature overnight. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 50% EtOAc/PE to afford 3-1 (148 mg, 47%) as a white solid. LC-MS: m/z = 436.0[(M++1)]. 1 H NMR (400MHz, DMSO-d6): δ 8.30 (d, J = 9.7 Hz, 1 H), 7.53– 7.16 (m, 7H), 7.10– 6.91 (m, 2H), 4.65 (dd, J = 46.4, 34.3 Hz, 2H), 4.47– 4.19 (m, 2H), 3.68 (d, J = 31.9 Hz, 3H), 3.42 (d, J = 8.5 Hz, 1 H), 1.78– 1.59 (m, 3H), 1.16 (dd, J = 36.4, 0.5 Hz, 3H), 0.94 (s, 1 H), 0.56– 0.27 (m, 2H), 0.21 (d, J = 3.0 Hz, 2H). EXAMPLE 4

To a stirring solution of compound 1 (5.0 g, 41.6 mmol) in 60% EtOH/H 2 O (80 mL) was added (NH 4 ) 2 CO 3 (20.0 g, 0.21 mol) followed by potassium cyanide (4.05 g, 62.5 mmol) at room temperature. The reaction mixture was heated to 70 °C for 16 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/DCM to afford compound 2 (2.7 g, 57%). LC-MS: m/z = 191.07[(M + +1)]

To a stirring solution of compound 2 (0.95 g, 5 mmol) in DMF (20 mL) was added 2-bromoacetyl bromide (0.98 g, 5 mmol) and K 2 CO 3 (1.38 g, 10 mmol) and stirred at room temperature overnight. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product 3 (1.27 g, 87%), which was used to the next step without any other purification LC-MS: m/z = 305.1[(M + +1)]

To a stirring solution of compound 3 (0.56 g, 1.84 mmol) in DCM (4 mL) was added trifluoroacetic acid (4 ml) at 0 °C and stirred at room temperature for 0.5 h. The reaction mixture was concentrated under reduced pressure to obtain crude product, which was used to the next step without any other purification as compound 4 (0.4 g, 90%) as an off-white solid. LC-MS: m/z = 249.1[(M + +1)]

To a stirring solution of intermediate 14 (0.11 g, 0.6 mmol) in DMF (10 mL) was added compd-4 (150 mg, 0.6 mmol) and DIPEA (100 mg, 0.73 mmol) at room temperature and stirred for 2 mins, then HATU (0.25 g, 0.66 mmol) was added and the reaction mixture was stirred at room temperature for 1 h. The solvent from the reaction was concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford 4-1 (0.13 g, 67%) as an off-white solid. LC- MS: m/z = 406.1[(M++1)] at room temperature 1.62 (100% purity). 1 H NMR (400 MHz, DMSO) δ 8.94 (d, J = 19.8 Hz, 1 H), 7.53 (t, J = 6.7 Hz, 2H), 7.37 (dq, J = 22.1, 7.3 Hz, 5H), 7.24 (dt, J = 28.0, 8.8 Hz, 3H), 4.66 (d, J = 53.1 Hz, 2H), 4.46– 4.27 (m, 1 H), 4.24 (dd, J = 16.9, 6.3 Hz, 1 H), 4.14 (dd, J = 13.4, 10.8 Hz, 1 H), 3.70 (d, J = 6.6 Hz, 1 H), 1.71 (d, J = 6.4 Hz, 3H), 1.28– 1.03 (m, 3H), 1.01– 0.85 (m, 1 H), 0.46 (d, J = 7.7 Hz, 1 H), 0.40– 0.08 (m, 3H). EXAMPLE 5

To a stirring solution of furan-2-carbaldehyde SM1 (10 g, 10.4 mmol) in 50% EtOH/H 2 O (300 mL) was added (NH 4 ) 2 CO 3 (50 g, 52.1 mmol) followed by potassium cyanide (10.5 g, 15.6 mmol) at room temperature. The reaction mixture was heated to 75 °C for 18 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10% MeOH/DCM to afford compound 1 (9 g, 52 %) as an off-white solid. LC-MS: m/z = 167.0 [(M + +1)].

To a stirring solution of compound 1 (332 mg, 2 mmol) and tert-butyl 2- bromoacetate SM2 (429 mg, 2.2 mmol) in DMF (6 mL) was added K 2 CO 3 (553 mg, 4 mmol) and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc.

Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/DCM to afford compound 2 (450 mg, 80%) as an off-white solid. LC-MS: m/z = 281.1 [(M + +1)].

To a stirring solution of compound 2 (450 mg, 1.61 mmol) in DCM (6 mL) was added TFA (3 mL) and stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure to obtain compound 3 (360 mg, 100%) as an off-white solid used for next step directly. LC-MS: m/z = 225.0 [(M + +1)].

To a stirring solution of compound 3 (250 mg, 1.12 mmol) and intermediate 14 (215 mg, 1.23 mmol) in DMF (5 mL) was added DIPEA (289 mg, 2.24 mmol) followed by HATU (638 mg,1.68 mmol) and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 20% PE/ EtOAc to afford compound 5-1 (300 mg, 71%) as an off-white solid. LC-MS: m/z =382. 2[(M++1)] at room temperature 8.53 (99.78% purity). 1 H NMR (400 MHz, DMSO-d6) δ 8.75 (d, J = 14.9 Hz, 0H), 7.68 (s, 0H), 7.39 (s, 1 H), 7.33– 7.10 (m, 1 H), 6.49 (d, J = 11.0 Hz, 0H), 5.46 (d, J = 18.6 Hz, 0H), 4.73 (s, 0H), 4.59 (s, 0H), 4.41 (d, J = 16.6 Hz, 0H), 4.27 (d, J = 19.1 Hz, 0H), 4.20 (d, J = 17.2 Hz, 0H), 3.67 (d, J = 7.4 Hz, 0H), 1.34– 1.04 (m, 1 H), 0.94 (s, 0H), 0.63– 0.05 (m, 1 H). EXAMPLE 6

To a stirring solution of nicotinaldehyde SM1 (5.5 g, 51 mmol) in water (50 mL) and EtOH (50 ml) was added (NH 4 ) 2 CO 3 (25 g, 255 mmol) and KCN (5 g, 76 mmol) at room temperature. The reaction mixture was heated at 70 0 C for 3 h. After being cooled to room temperature and concentrated to afford crude compound 1 (1.5 g). LC-MS: m/z = 178.2[(M + +1)].

To a stirring solution of crude compound 1 (1.5 g, 8.5 mmol) in dry DMF (10 ml) was added K 2 CO 3 (2.3 g, 17 mmol) and tert-butyl 2-bromoacetate (1.65 g, 8.5 mmol) at room temperature. The reaction mixture was heated at 45 0 C for 0.5h. After being cooled to room temperature the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 75% EtOAc/Hexane to afford compound 2 (0.5 g, 20%). TLC: 75% EtOAc/Hexane (R f : 0.2).

To a stirring solution of compound 2 (0.5 g, 1.7 mmol) in dry DCM (2 ml) was added TFA (2 g, 17 mmol) at room temperature. The reaction mixture was stirred at room temperature for 2h then concentrated under reduced pressure to afford compound 3 (0.5 g, crude). LC-MS: m/z = 236.2[(M + +1)].

To a stirring solution of compound 3280 mg, 1.2 mmol) in DMF (5 mL) was added compound intermediate 14 (200 mg, 1.2 mmol) and DIPEA (460 mg, 3.6 mmol) at room temperature. To this added HATU (910 mg, 2.4 mmol) at room temperature and the reaction mixture was stirred at room temperature for 1 h. the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 3% MeOH/DCM to afford compound 6-1 (10 mg, 2%).as an off-white solid. TLC: 10% MeOH/DCM (Rf: 0.5). LC- MS: m/z = 393[(M++1)] (98% purity). 1 H NMR (400 MHz, DMSO-d6) δ 8.83 (d, J = 16.8 Hz, 1 H), 8.61 (d, J = 26.0 Hz, 2H), 7.84 (d, J = 8.0 Hz, 1 H), 7.50– 7.15 (m, 6H), 5.42 (d, J = 20.4 Hz, 1 H), 4.75 (s, 1 H), 4.33 (m, 4H), 1.21– 1.06 (m, 3H), 0.94 (s, 1 H), 0.21 (m, 4H). EXAMPLE 7

To a stirring solution of NaH (1.25 g, 52.1 mmol) in DMF (65 mL) was added SM1 (5 g, 52.1 mmol) followed by iodomethane (8.13 g, 57.3 mmol) at 0 °C. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/ DCM to compound 1 (1.95 g, 34%) as a yellow solid. LC-MS: m/z = 111[(M + +1)].

To a stirring solution of compound 1 (1.93 g, 17.5 mmol) in MeOH (30 mL), H 2 O (30 mL) was added (NH 4 ) 2 CO 3 (13.4 g, 140 mmol) followed by potassium cyanide (6 g, 92.3 mmol) at room temperature. The reaction mixture was heated to 60 °C for 1 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude product, which was dissolved in 20% MeOH/ DCM solution, filtered, the filtrate was concentrated to afford compound 2 (3.12 g, 99%) as an orange solid. LC-MS: m/z = 181[(M + +1)].

To a stirring solution of compound 2 (3.12 g, 17.3 mmol) in DMF (30 mL) was added K 2 CO 3 (3.58 g, 25.95 mmol) followed by tert-butyl 2-bromoacetate (3.37 g, 17.3 mmol) at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain compound 3 (2.72 g, 54%) as a brown solid. LC-MS: m/z = 295[(M + +1)].

To a stirring solution of compound 3 (2.72 g, 9.26 mmol) in DCM (10 mL) was added TFA (10 mL) at room temperature and stirred at room temperature for 3 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude compound 4 (2.2 g, 99%) as a yellow solid. LC- MS: m/z = 239[(M + +1)].

To a stirring solution of compound 4 (200 mg, 0.84 mmol) in DMF (2 mL) was added intermediate 14 (147 mg, 0.84 mmol) and DIPEA (325 mg, 2.52 mmol) followed by HATU (638 mg, 1.68 mmol)at room temperature and stirred at room

temperature for 1 h. The reaction mixture was diluted with water and extracted with

EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by prep-HPLC to afford 7-1 (22 mg, 7%) as a white solid. LC-MS: m/z = 396.0[(M++1)]. 1 H NMR

(400MHz, MeOD): δ 8.90 (s, 1 H), 7.72 (s, 1 H), 7.49– 7.18 (m, 5H), 5.49 (s, 1 H), 4.86– 4.62 (m, 2H), 4.41 (t, J = 43.9 Hz, 2H), 3.94 (d, J = 4.8 Hz, 3H), 3.81 (s, 1 H), 1.35– 1.20 (m, 3H), 0.98 (s, 1 H), 0.68– 0.53 (m, 1 H), 0.46– 0.19 (m, 3H). EXAMPLE 8

 

To a stirring solution of isonicotinaldehyde SM1 (4.2 g, 39.3 mmol) in water (20 mL) and MeOH (20 ml) was added (NH 4 ) 2 CO 3 (18.8 g, 196.5 mmol) and KCN (3.8 g, 58.9 mmol) at room temperature. The reaction mixture was heated at 60 0 C overnight. After being cooled to room temperature, the reaction was filtered to afford compound 1 (2.4 g, 35%). LC-MS: m/z = 178.1[(M + +1)]. To a solution of intermediate 13 (300 mg, 1mmol) in DMF (5ml) was added Compd-1 ( 177 mg, 1 mmol) and K 2 CO 3 (276 mg, 2 mmol). The reaction mixture was then stirred at room temperature for 1 H. the mixture was filtered, the filtrate was concentrated in vacuo, purified by Prep-HPLC to afford 10 mg of 8-1. yield : 2.5%. LC-MS: m/z = 409.9[(M++1)] at room temperature 1.31 (100.00% purity). 1 H NMR (400 MHz, MeOD) δ 8.80 (d, J = 20.7 Hz, 2H), 8.19 (s, 2H), 7.30 (dt, J = 18.0, 8.4 Hz, 5H), 4.87– 4.61 (m, 3H), 3.77 (dd, J = 9.6, 6.9 Hz, 0.5H), 3.53 (d, J = 14.8 Hz, 2H), 3.08 (d, J = 7.7 Hz, 0.5H), 1.29 – 1.14 (m, 3H), 0.94 (d, J = 5.0 Hz, 1 H), 0.58 (dd, J = 11.6, 6.5 Hz, 1 H), 0.41– 0.15 (m, 3H). EXAMPLE 9

To a stirring solution of 1-naphthaldehyde SM1 (1.56 g, 10 mmol) in 50% EtOH/H 2 O (40 mL) was added (NH 4 ) 2 CO 3 (20 g, 120 mmol) followed by potassium cyanide (2 g, 30 mmol) at room temperature. The reaction mixture was heated to 75 °C for 18 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10% MeOH/DCM to afford compound 1 (1.2 g, 53%) as a yellow solid. LC-MS: m/z = 227.2 [(M + +1)].

To a stirring solution of compound 1 (452 mg, 2 mmol) and tert-butyl 2- bromoacetate SM2 (429 mg, 2.2 mmol) in DMF (6 mL) was added K 2 CO 3 (553 mg, 4 mmol) and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc.

Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/DCM to afford compound 2 (0.41 g, 60%) as a yellow solid. LC-MS: m/z = 285.1 [(M + +1)].

To a stirring solution of compound 2 (0.41 g, 1.44 mmol) in DCM (10 mL) was added TFA (5 mL) and stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure to obtain compound 3 (340 mg, 100%) as a light yellow solid used for next step directly. LC-MS: m/z = 285.1 [(M + +1)].

To a stirring solution of compound 3 (200 mg, 0.7 mmol) and N-benzyl-1- cyclopropylethanamine intermediate-14(122 mg, 0.7 mmol) in DCM (40 mL) was added Et3N (142 mg, 1.4 mmol) followed by T3P (50% in EtOAc) (890 mg, 1.4 mmol) and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/DCM to afford compound 9-1 (85 mg, 27%) as an off-white solid. LC-MS: m/z = 442.0 [(M++1)] at room temperature 4.81 (99.22% purity). 1 H NMR (300 MHz, DMSO-d6) δ 8.81 (d, J = 11.5 Hz, 1 H), 8.28 (d, J = 3.9 Hz, 1 H), 7.97 (t, J = 9.2 Hz, 2H), 7.73– 7.47 (m, 4H), 7.45– 7.07 (m, 5H), 6.16 (d, J = 12.3 Hz, 1 H), 4.74 (s, 1 H), 4.61 (d, J = 5.9 Hz, 1 H), 4.53– 4.17 (m, 2H), 3.73-3.66 (m, 0.5H), 3.39-3.36 (m, 0.5H), 1.22– 1.04 (m, 3H), 0.99-0.89 (m, 1 H), 0.61– 0.04 (m, 4H).

EXAMPLE 10

To a stirring solution of 1 H-pyrazole-4-carbaldehyde SM1 (500 mg, 5.2mmol) in DMF (10mL) was added NaH (248mg, 6.2mmol), followed by SEMCl (1.03g, 6.2mmol) at 0 °C and stirred at room temperature for 1 H. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford compound 1 (800mg,57%). TLC: 20% EtOAc/Hexane (R f : 0.5). LC-MS: m/z = 227[(M + +1)].

To a stirring solution of 1-((2-(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazole-4- carbaldehyde compound 1 (800 mg, 3.5mmol) in MeOH (10ml),was added TMSCN (525mg, 5.3mmol), NH 3 (gas) was charged into the mixture at 0°C. The reaction mixture stirred at 45°C for 3 h. After consumption of the starting material (by TLC), the reaction mixture concentrated under reduced pressure to obtain crude product, which washed with ether to afford compound 2 (700mg, 79%). TLC: 30% EA/PE (R f : 0.4). LC-MS: m/z = 253[(M + +1)]

To a stirring solution of 2-amino-2-(1-((2-(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-4-yl)acetonitrile compound 2 (700 mg, 2.7mmol) in MeOH (10mL) and DCM (10mL) was added DIEA (1.07g, 8.3mmol) followed by dry ice at 0 °C and stirred at room temperature for overnight. The reaction mixture concentrated under reduced pressure to obtain crude product, which washed with ether to afford compound 3 (550mg,68.8%). TLC: 30% EtOAc/Hexane (R f : 0.5). LC-MS: m/z = 297[(M + +1)] To a stirring solution of 5-(1-((2-(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol- 4-yl)imidazolidine-2,4-dione compound 3 (50 mg, 0.17mmol) in DMF (10 mL) was added N-benzyl-2-bromo-N-(1-cyclopropylethyl)acetamide Intermediate 13 (50mg, 0.17mmol), followed by K 2 CO 3 (46.6mg, 0.34mmol ) at room temperature and stirred at room temperature for 2 h. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford compound 5 (50 mg, 58%). TLC: 30% EtOAc/Hexane (R f : 0.6). LC-MS: m/z = 512[(M + +1)]

A stirring solution of N-benzyl-N-(1-cyclopropylethyl)-2-(2,5-dioxo-4-(1- ((2-(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-4-yl)imidazolidin-1-yl)acetamide compound 5 (50 mg, 0.1mmol) in HCl/MeOH (10 mL) stirred at reflux for 1 h. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford 10-1 (30 mg, 81%). LC-MS: m/z = 382 [(M++1)] at room temperature 1.70 (100% purity). 1 H NMR (400 MHz, DMSO) δ 9.33– 9.03 (m, 1 H), 7.80– 7.07 (m, 7H), 4.80– 4.00 (m, 4H), 3.78– 3.27 (m, 1 H), 1.25– 1.02 (m, 3H), 0.98– 0.77 (m, 1 H), 0.21 (s, 4H). EXAMPLE 11

To a stirring solution of 1-methyl- 1 H-indole-3-carbaldehyde SM1 (2.0 g, 12.6 mmol) in 50% EtOH/H 2 O (40 mL) was added (NH 4 ) 2 CO 3 (6.04 g, 62.9 mmol) followed by potassium cyanide (1.23 g, 18.86 mmol) at room temperature. The reaction mixture was heated to 75 °C for 18 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10% MeOH/DCM to afford compound 1 (2 g, 70 %) as a colorless solid. LC-MS: m/z = 229.1 [(M + +1)].

To a stirring solution of compound 1 (464 mg, 2 mmol) and tert-butyl 2- bromoacetate SM2 (429 mg, 2.2 mmol) in DMF (6 mL) was added K 2 CO 3 (553 mg, 4 mmol) and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc.

Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/DCM to afford compound 2 (400 mg, 58%) as an off-white solid. LC-MS: m/z = 344.2 [(M + +1)].

To a stirring solution of compound 2 (400 mg, 1.17 mmol) in DCM (6 mL) was added TFA (3 mL) and stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure to obtain compound 3 (260 mg, 77%) as an off-white solid used for next step directly. LC-MS: m/z = 288.1 [(M + +1)].

To a stirring solution of compound 3 (260 mg, 0.91 mmol) and intermediate 14 (159 mg, 0.90 mmol) in DMF (5 mL) was added DIPEA (292 mg, 2.27 mmol) followed by HATU (689 mg,1.81 mmol) and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 20% PE/ EtOAc to afford 11-1 (300 mg, 71%) as an off-white solid. LC-MS: m/z =445.2 [(M++1)] at room temperature 8.90 (99.65% purity). 1 H NMR ((400 MHz, DMSO-d6) δ 8.59 (d, J = 14.8 Hz, 1 H), 7.64 (d, J = 7.8 Hz, 1 H), 7.41 (td, J = 15.7, 8.1 Hz, 4H), 7.33– 7.23 (m, 2H), 7.19 (dd, J = 13.4, 5.9 Hz, 1 H), 7.02 (td, J = 7.4, 4.1 Hz, 1 H), 5.57– 5.43 (m, 1 H), 4.76 (s, 1 H), 4.69– 4.58 (m, 1 H), 4.55– 4.14 (m, 2H), 3.75– 3.67 (m, 1 H), 3.40 (d, J = 6.6 Hz, 0H), 1.16 (ddd, J = 11.6, 6.4, 4.5 Hz, 3H), 1.04– 0.87 (m, 1 H), 0.58– 0.26 (m, 2H), 0.26– 0.11 (m, 2H). EXAMPLE 12

To a stirring solution of SM1 (700 mg, 5.3 mmol) in acetonitrile (35 mL) was 3,4-dihydro-2H-pyran (532 mg, 6.36 mmol) followed by 4-methylbenzenesulfonic acid (91 mg, 0.53 mmol) at room temperature. After the reaction mixture was stirred at room temperature for 3 h, the solvent was removed under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 20% EtOAc / PE to afford compound 1 (850 mg, 70%) as a white solid. LC-MS: m/z = 231[(M + +1)].

To a stirring solution of compound 1 (816 mg, 3.55 mmol) in formamide (25 mL) was added (NH 4 ) 2 CO 3 (1.704 g, 17.75 mmol) followed by potassium cyanide (0.347 g, 5.32 mmol) at room temperature in a sealed tube. The reaction mixture was heated to 100 °C overnight. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% methanol/DCM to afford compound 2 (113 mg, 11%) as a yellow solid. LC-MS: m/z = 301[(M + +1)].

To a stirring solution of compound 2 (113 mg, 0.377 mmol) in DMF (5 mL) was added K 2 CO 3 (104 mg, 0.753 mmol) followed by intermediate 13 (112 mg, 0.377 mmol) at room temperature and stirred at room temperature overnight. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to afford compound 3 (216 mg, 99%) as a yellow solid. LC-MS: m/z = 516[(M + +1)].

To a stirring solution of compound 3 (216 mg, 0.377 mmol) in DCM (1.5 mL) was added TFA (1.5 mL) at room temperature and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude product, which was purified by prep-HPLC eluting with ACN/H 2 O containing 5% TFA to afford 12-1 (35 mg,19%) as a white solid. LC-MS: m/z = 432.2[(M++1)] at room temperature 7.222 (98.530% purity). 1 H NMR (400 MHz, DMSO): δ 13.17 (d, J = 5.2 Hz, 1 H), 8.80 (d, J = 15.4 Hz, 1 H), 7.89 (s, 1 H), 7.65– 6.85 (m, 8H), 5.76– 5.53 (m, 1 H), 4.86– 4.11 (m, 4H), 3.78– 3.66 (m, 0.6H), 3.52– 3.45 (m, 0.4H), 1.17 (dt, J = 14.7, 10.2 Hz, 3H), 0.96 (s, 1 H), 0.62– 0.03 (m, 4H). EXAMPLE 13

To a stirring solution of 6-fluoro- 1 H-indole-3-carbaldehyde SM1 (1.0 g, 6 mmol) in 50% EtOH/H 2 O (20 mL) was added (NH 4 ) 2 CO 3 (3.0 g, 30 mmol) followed by potassium cyanide (0.6 g, 9 mmol) at room temperature. The reaction mixture was heated to 75 °C for 18 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10%

MeOH/DCM to afford compound 1 (0.67 g, 47 %) as a white solid. LC-MS: m/z = 234.1 [(M + +1)].

To a stirring solution of compound 1 (233 mg, 1.0 mmol) and intermediate- 13 (295 mg, 1.0 mmol) in DMF (5 mL) was added K 2 CO 3 (207 mg,1.5 mmol) and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by prep-hplc to afford compound 13-1 (110 mg, 25%) as an white solid. LC-MS: m/z =449.2 [(M++1)] at room temperature 4.42 (93.05% purity). 1 H NMR ((400 MHz, DMSO-d6) δ 11.22 (s, 1 H), 8.59 (d, J = 11.3 Hz, 1 H), 7.63 (d, J = 2.9 Hz, 1 H), 7.38 (dd, J = 16.4, 8.6 Hz, 2H), 7.21 (dd, J = 25.1, 7.1 Hz, 3H), 6.83 (dd, J = 15.5, 7.3 Hz, 1 H), 5.48 (d, J = 13.4 Hz, 1 H), 4.68 (d, J = 40.3 Hz, 2H), 4.55– 4.05 (m, 2H), 3.78– 3.56 (m, 1 H), 3.38 (s, 2H), 1.33– 1.01 (m, 2H), 0.95 (s, 1 H), 0.33 (dd, J = 53.5, 28.2 Hz, 3H). EXAMPLE 14

To a stirring solution of 2-naphthaldehyde SM1 (500 mg, 3.2mmol) in MeOH (10ml),was added TMSCN (317mg, 3.2mmol), NH 3 (gas) was charged into the mixture at 0°C. The reaction mixture stirred at 45°C for 3 h. After consumption of the starting material (by TLC), the reaction mixture concentrated under reduced pressure to obtain crude product, which washed with ether to afford compound 1 (400mg, 68%). TLC: 30% EA/PE (R f : 0.4). LC-MS: m/z = 183[(M + +1)]

To a stirring solution of 2-amino-2-(naphthalen-2-yl)acetonitrile compound 1 (400 mg, 2.2mmol) in MeOH (10mL) and DCM (10mL) was added DIEA (851mg, 6.6mmol) followed by dry ice at 0 °C and stirred at room temperature for overnight. The reaction mixture concentrated under reduced pressure to obtain crude product, which washed with ether to afford compound 2 (300mg,60%). TLC: 30% EtOAc/Hexane (R f : 0.5). LC-MS: m/z = 227[(M + +1)].

To a stirring solution of 5-(naphthalen-2-yl)imidazolidine-2,4-dione compound 2 (50 mg, 0.22mmol) in DMF (10 mL) was added N-benzyl-2-bromo-N-(1- cyclopropylethyl)acetamide intermediate 13 (65mg, 0.22mmol), followed by K 2 CO 3 (60.7mg, 0.44mmol ) at room temperature and stirred at room temperature for 2 h. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford 14-1 (20 mg, 20.6%) as an off-white solid. LC-MS: m/z = 442 [(M++1)] at room temperature 1.68 (100% purity). 1 H NMR (300 MHz, DMSO) δ 8.86 (d, J = 11.8 Hz, 1 H), 8.01– 7.83 (m, 4H), 7.62– 7.13 (m, 9H), 5.46 (d, J = 14.7 Hz, 1 H), 4.86– 4.07 (m, 5H), 3.79– 3.56 (m, 1 H), 1.36– 1.03 (m, 4H), 1.02– 0.82 (m, 1 H), 0.66– 0.05 (m, 4H). EXAMPLE 15

 

To a stirring solution of 5-fluoro- 1 H-indole-3-carbaldehyde SM1 (1.0 g, 6.13 mmol) in water (10 mL) and MeOH (10 ml) was added (NH 4 ) 2 CO 3 (3.0 g, 30.8 mmol) and KCN (0.59 g, 9.2 mmol) at room temperature. The reaction mixture was heated at 50 0 C overnight. After being cooled to room temperature and concentrated, the residue was purified by silica gel column chromatography eluting with 50% EtOAc/Hexane to afford compound 1 (0.55 g, 39%). LC-MS: m/z = 234.2[(M + +1)].

To a stirring solution of compound 1 (0.5 g, 2.1 mmol) in dry DMF (6 ml) was added K 2 CO 3 (0.29 g, 2.1 mmol) and tert-butyl 2-bromoacetate (0.41 g, 2.1 mmol) at room temperature. The reaction mixture was heated at 40 0 C for 2.5 h. After being cooled to room temperature the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 30% EtOAc/Hexane to afford compound 2 (0.45 g, 62%). TLC: 40% EtOAc/Hexane (R f : 0.3).

To a stirring solution of compound 3 (0.45 g, 1.29 mmol) in dry DCM (5 ml) was added TFA (2.7 g, 23.7 mmol) at room temperature. The reaction mixture was stirred at room temperature for 2h then concentrated under reduced pressure to afford compound 3 (0.30g, 80%). LC-MS: m/z = 292.2[(M + +1)].

To a stirring solution of Compound-3 (150 mg, 0.52 mmol) and intermediate 14 (91.1 mg, 0.52 mmol) was added HATU (1.2eq) and DIPEA (2 ml). The resulted mixture was stirred at room temperature for 1 h. The reaction mixture washed with water and extracted with EtOAc, the organic phase separated and dried with Na 2 SO 4 , concentrated in vacuum and purified by Prep HPLC to afford 100 mg of desired product 15-1. yield: 43.0%. LC-MS: m/z = 449.0[(M++1)] at room temperature 1.58 (100.00% purity). 1 H NMR (400 MHz, DMSO) δ 11.27 (s, 1 H), 8.58 (d, J = 16.8 Hz, 1 H), 7.60– 6.87 (m, 9H), 5.50 (d, J = 18.0 Hz, 1 H), 4.76 (s, 1 H), 4.63 (d, J = 5.7 Hz, 1 H), 4.53– 4.16 (m, 2H), 3.75– 3.65 (m, 0.5H), 3.44– 3.36 (m, 0.5H), 1.28– 1.06 (m, 3H), 0.95 (s, 1 H), 0.59– 0.13 (m, 4H). EXAMPLE 16

intermediate 13 To a stirring DMF (10.63 mL) was added dropwise POCl 3 (3.55 mL) at 10- 20 °C followed by addition of a solution of SM1 (2 g,13.19 mmol) at 20-30 °C. The reaction mixture was stirred at 35-37 °C for 45 minutes and finally poured into stirred ice (28 g) and water (21 mL). Sodium hydroxide (6.86 g) in water (36 mL) was added at 20-30 °C and made the solution pH=8. The mixture was boiled for 5 minutes and cooled to room temperature, extracted with EtOAc, washed with brine. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 60% PE/EtOAc to afford compound 1 (1.037 g, 44%) as an off-white solid. LC-MS: m/z = 180[(M + +1)].

To a stirring solution of compound 1 (0.5 g, 2.78 mmol) in EtOH (6 mL), H 2 O (6 mL) was added (NH 4 ) 2 CO 3 (1.334 g, 13.9 mmol) followed by potassium cyanide (0.272 g, 4.17 mmol) at room temperature in a sealed tube. The reaction mixture was heated to 95 °C overnight. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 66% PE/EtOAc to afford compound 2 (0.344 g, 50%) as a yellow solid. LC-MS: m/z = 250[(M + +1)].

To a stirring solution of compound 2 (80 mg, 0.32 mmol) in DMF (3 mL) was added intermediate 13 (95 mg, 0.32 mmol) followed by K 2 CO 3 (88 mg, 0.64 mmol) at room temperature and stirred at room temperature overnight. EtOAc was added and then filtered. The filtrate was concentrated under reduced pressure to obtain crude product, which was purified by prep-HPLC eluting with ACN/H 2 O containing 5% TFA to afford 16-1 (69 mg, 46%) as a white solid. LC-MS: m/z = 465.1[(M++1)] at room temperature 7.807 (99.125% purity). 1 H NMR (400 MHz, DMSO): δ 11.62 (s, 1 H), 8.62 (d, J = 13.2 Hz, 1 H), 7.71– 6.95 (m, 9H), 5.86 (d, J = 6.8 Hz, 1 H), 4.45 (dd, J = 196.6, 29.3 Hz, 4H), 3.71 (dd, J = 16.4, 7.0 Hz, 0.6H), 3.40 (d, J = 6.9 Hz, 0.4H), 1.32– 1.05 (m, 3H), 0.95 (s, 1 H), 0.29 (ddd, J = 27.0, 26.3, 6.1 Hz, 4H). EXAMPLE 17

To a stirring solution of 7-chloro- 1 H-indole-3-carbaldehyde SM1 (200 mg, 1.1mmol) in MeOH (10ml),was added TMSCN (168mg, 1.5mmol), NH 3 (gas) was charged into the mixture at 0°C. The reaction mixture stirred at 45°C for 3 h. After consumption of the starting material (by TLC), the reaction mixture concentrated under reduced pressure to obtain crude product, which washed with ether to afford compound 1 (200mg, 88%). TLC: 30% EA/PE (R f : 0.3). LC-MS: m/z = 206[(M + +1)]

To a stirring solution of 2-amino-2-(7-chloro- 1 H-indol-3-yl)acetonitrile compound 1 (200 mg, 0.97mmol) in MeOH (5mL) and DCM (5mL) was added DIEA (374mg, 2.9mmol) followed by dry ice at 0 °C and stirred at room temperature for overnight. The reaction mixture concentrated under reduced pressure to obtain crude product, which washed with ether to afford compound 2 (150mg,62.2%). TLC: 30% EtOAc/Hexane (R f : 0.4). LC-MS: m/z = 250[(M + +1)]

To a stirring solution of 5-(7-chloro- 1 H-indol-3-yl)imidazolidine-2,4-dione compound 2 (50 mg, 0.2mmol) in DMF (10 mL) was added N-benzyl-2-bromo-N-(1- cyclopropylethyl)acetamide intermediate 13 (60mg, 0.2mmol), followed by K 2 CO 3 (55.2mg, 0.4mmol ) at room temperature and stirred at room temperature for 2 h. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford 17-1 (10 mg, 10.7%) as an off-white solid. LC-MS: m/z = 465 [(M++1)] at room temperature 1.67 (100% purity). 1 H NMR (400 MHz, DMSO) δ 11.56 (s, 1 H), 8.73– 8.44 (m, 1 H), 7.90– 6.79 (m, 9H), 5.64– 5.35 (m, 1 H), 4.95– 4.05 (m, 4H), 3.79– 3.37 (m, 1 H), 1.29– 0.82 (m, 5H), 0.60– 0.06 (m, 4H). EXAMPLE 18

To a stirring solution of intermediate 39 (40 mg, 0.16 mmol) in DMF (1 mL) was added (S)-N-benzyl-2-bromo-N-(1-cyclobutylethyl)acetamide (50 mg, 0.16 mmol) followed by K 2 CO 3 (44 mg, 0.32 mmol) at room temperature and stirred at room temperature overnight. EtOAc was added and then filtered. The filtrate was concentrated under reduced pressure to obtain crude product, which was purified by prep-HPLC eluting with ACN/H 2 O containing 5% TFA to afford 18-1 (22 mg, 29%) as a white solid. LC-MS: m/z = 479.1[(M++1)] at room temperature 4.822 (99.695% purity). 1 H NMR (400 MHz, DMSO): δ 11.62 (s, 1 H), 8.63 (d, J = 18.0 Hz, 1 H), 7.74– 6.94 (m, 9H), 5.88 (d, J = 10.2 Hz, 1 H), 4.83– 3.94 (m, 5H), 2.43 (s, 1 H), 1.93 (s, 1 H), 1.59 (d, J = 89.0 Hz, 5H), 1.16– 0.76 (m, 3H). EXAMPLE 19

To a stirring solution of 2,6-dichlorobenzaldehyde SM (0.35 g, 2 mmol) in 50% EtOH/H 2 O (20 mL) was added (NH 4 ) 2 CO 3 (2.3 g, 24 mmol) followed by potassium cyanide (0.39 g, 6 mmol) at room temperature. The reaction mixture was heated to 75 °C for 16 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10% MeOH/DCM to afford compound 1 (0.18 g, 36%). LC-MS: m/z = 245 [(M + +1)].

To a stirring solution of compound 1 (29 mg, 0.12 mmol) and compound 2 (35 mg, 0.12 mmol) in DMF (1 mL) was added K 2 CO 3 (33 mg, 0.24 mmol) and stirred at room temperature for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by prep-hplc to afford compound 19-1 (3 mg, 5%) as an off- white solid. LC-MS: m/z = 460.1[(M++1)] at room temperature 4.65 (95.95% purity). 1 H NMR (300 MHz, CD3OD-d6) δ 7.75– 6.98 (m, 8H), 6.15 (dd, J = 15.2, 2.0 Hz, 1 H), 4.78 (d, J = 17.5 Hz, 2H), 4.58– 4.35 (m, 2H), 3.84-3.78 (m, 0.5H), 3.41– 3.35 (m, 0.5H), 1.37 – 1.18 (m, 3H), 0.98-0.91 (m, 1 H), 0.59-0.23 (m, 4H).

EXAMPLE 20

To a stirring solution of SM1 (1.2 g, 10 mmol) in MeOH (10 mL), H 2 O (10 mL) was added (NH 4 ) 2 CO 3 (4.8 g, 50 mmol) followed by potassium cyanide (2.5 g, 38.5 mmol) at room temperature. The reaction mixture was heated to 60 °C overnight. After consumption of the starting material (by TLC), the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10% MeOH / DCM to afford compound 1 (0.58 g, 30%) as a yellow solid. LC-MS: m/z = 191[(M + +1)].

To a stirring solution of compound 1 (571 mg, 3 mmol) in DMF (5 mL) was added K 2 CO 3 (621 mg, 4.5 mmol) followed by tert-butyl 2-bromoacetate (614.3 mg, 3.15 mmol) at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH / DCM to afford compound 2 (510 mg, 56%) as a yellow oil. LC-MS: m/z = 249[(M + -55)].

To a stirring solution of compound 2 (235 mg, 0.773 mmol) in DCM (2 mL) was added TFA (2 mL) at room temperature and stirred at room temperature for 2 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude compound 3 (190 mg, 99%) as a yellow solid. LC- MS: m/z = 249[(M + +1)].

To a stirring solution of compound 3 (85.1 mg, 0.343 mmol) in DMF (1 mL) was added intermediate 24 (60 mg, 0.343 mmol) and DIPEA (97.4 mg, 0.755 mmol) followed by HATU (143.4 mg, 0.377 mmol)at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by Prep-HPLC to afford 20-1 (40 mg, 29%) as a yellow solid. LC-MS: m/z = 406.0[(M++1)]. 1 H NMR (400MHz, DMSO-d6): δ 8.64 (d, J = 16.5 Hz, 1 H), 7.43– 7.14 (m, 9H), 5.52 (d, J = 18.2 Hz, 1 H), 4.77– 4.55 (m, 2H), 4.45– 4.15 (m, 2H), 3.70 (d, J = 6.5 Hz, 1 H), 2.42 (d, J = 4.1 Hz, 3H), 1.14 (ddd, J = 36.0, 6.6, 3.2 Hz, 3H), 0.94 (s, 1 H), 0.47 (d, J = 5.0 Hz, 1 H), 0.40– 0.09 (m, 3H). EXAMPLE 21

To a stirring solution of SM1 (1.06 g, 10 mmol) in MeOH (10 mL), H 2 O (10 mL) was added (NH 4 ) 2 CO 3 (4.8 g, 50 mmol) followed by potassium cyanide (2.5 g, 38.5 mmol) at room temperature. The reaction mixture was heated to 60 °C overnight. After consumption of the starting material (by TLC), the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 10% MeOH / DCM to afford compound 1 (1.2 g, 68%) as a yellow solid. LC-MS: m/z = 177[(M + +1)].

To a stirring solution of compound 1 (535 mg, 3.02 mmol) in DMF (5 mL) was added K 2 CO 3 (625.1 mg, 4.53 mmol) followed by tert-butyl 2-bromoacetate (618.3 mg, 3.171 mmol) at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH / DCM to afford compound 2 (534 mg, 61%) as a clear oil. LC-MS: m/z = 235[(M + -55)].

To a stirring solution of compound 2 (534 mg, 1.84 mmol) in DCM (5 mL) was added TFA (5 mL) at room temperature and stirred at room temperature for 2 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude compound 3 (429 mg, 99%) as a yellow solid. LC- MS: m/z = 235[(M + +1)].

To a stirring solution of compound 3 (30 mg, 0.128 mmol) in DMF (1 mL) was added intermediate 24 (22.4 mg, 0.128 mmol) and DIPEA (36.4 mg, 0.282 mmol) followed by HATU (53.6 mg, 0.141 mmol)at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by Prep-HPLC to afford 21-1 (4 mg, 8%) as a yellow solid. LC-MS: m/z = 392.0[(M++1)]. 1 H NMR (400MHz, MeOD): δ 7.52– 7.20 (m, 10H), 5.24 (d, J = 16.2 Hz, 1 H), 4.76 (dd, J = 17.5, 12.2 Hz, 2H), 4.51– 4.33 (m, 2H), 3.87– 3.78 (m, 1 H), 1.27 (dd, J = 31.2, 5.6 Hz, 3H), 0.92 (s, 1 H), 0.58 (s, 1 H), 0.36– 0.22 (m, 3H). EXAMPLE 22

To a suspense of 2,6-dimethylbenzaldehyde 1 (0.536 g, 4 mmol) in EtOH (20 mL) and H 2 O (20 mL) was added KCN (0.39 g, 6 mmol) and (NH 3 ) 2 CO 3 (1.53 g, 16 mmol), the reaction mixture was stirred in steel tube at 85 °C for 16 h. Cooled to room temperature and the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by combiflash eluting (DCM/MeOH 20: 1) to afford compound 2 (0.5 g, 62%) as white solid. 1 H NMR (300 MHz, DMSO-d 6 ) δ 10.89 (s, 1 H), 8.10 (s, 1 H), 7.08 (ddd, J = 21.6, 14.5, 7.4 Hz, 3H), 5.56 (d, J = 1.3 Hz, 1 H), 2.36 (s, 3H), 2.14 (s, 3H).

To a mixture of 5-(2,6-dimethylphenyl)imidazolidine-2,4-dione 2 (0.5 g, 2.45 mmol) and tert-butyl 2-bromoacetate 3 (0.478 g, 2.45 mmol) in DMF (15 mL) was added K 2 CO 3 (0.69 g, 5 mmol) and stirred at room temperature for 2 h. The reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by combiflash eluting (DCM/MeOH 40: 1) to afford compound 4 (0.23 g, 29%) as brown solid. TLC: 5% MeOH/DCM (R f : 0.8).

To a solution of tert-butyl 2-(4-(2,6-dimethylphenyl)-2,5- dioxoimidazolidin-1-yl)acetate 4 (0.23 g, 0.73 mmol) in DCM (5 mL) was added TFA (5 mL) and stirred at room temperature for 2 h. After consumption of the starting material (by TLC) and concentrated under reduced pressure to obtain crude compound 5 (0.2 g, 108%) as brown solid. LC-MS: m/z = 263.1[(M + +1)] (80% purity). To a solution of intermediate 24 (40 mg, 0.23 mmol) in DCM (10 mL) was added TEA (50 mg, 0.46 mmol) and 2,4,6-tripropyl-1,3,5,2,4,6-trioxatriphosphorinane- 2,4,6-trioxide (0.292mg, 0.46 mmol) under N2 at 0 °C, then stirred at room temperature for 1 h. After consumption of the starting material (by TLC) diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous MgSO4 and

concentrated under reduced pressure to obtain crude product, which was purified by Prep- HPLC (MeCN/H 2 O 7: 3) to afford 22-1 (55 mg, 58%) as light yellow solid. LC-MS: m/z = 420.2(M++1)] at room temperature 4.56 (96.75% purity). 1 H NMR (300 MHz, DMSO-d6) δ 8.48 (d, J = 14.3 Hz, 1 H), 7.48– 6.95 (m, 8H), 5.61 (d, J = 10.7 Hz, 1 H), 4.67 (d, J = 43.9 Hz, 2H), 4.32 (dt, J = 22.6, 10.1 Hz, 2H), 3.76– 3.62 (m, 1 H), 2.46– 2.33 (m, 3H), 2.20 (s, 3H), 1.25– 1.03 (m, 3H), 0.94 (s, 1 H), 0.34 (ddd, J = 46.6, 27.4, 4.4 Hz, 4H). EXAMPLE 23

To a suspension of 1-( 1 H-indol-3-yl)ethanone 1 (1.59 g, 10 mmol) in EtOH (20 mL) and H 2 O (20 mL) was added KCN (0.975 g, 15 mmol) and (NH 3 ) 2 CO 3 (3.84 g, 40 mmol), the reaction mixture was stirred in steel tube at 85 °C for 16 h. Cooled to room temperature and the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by combiflash eluting (EtOAc/ Hexane 1: 1) to afford compound 2 (0.09 g, 4%) as white solid. 1 H NMR (300 MHz, DMSO-d 6 ) δ 11.16 (s, 1 H), 10.79 (s, 1 H), 8.40 (s, 1 H), 7.47– 7.31 (m, 3H), 7.15– 6.93 (m, 2H), 1.73 (s, 3H).

To a mixture of 5-( 1 H-indol-3-yl)-5-methylimidazolidine-2,4-dione 2 (0.09 g, 0.4 mmol) and tert-butyl 2-bromoacetate 3 (0.077 g, 0.4 mmol) in MeCN (15 mL) was added K 2 CO 3 (0.69 g, 5 mmol) and stirred at room temperature for 16 h. The reaction mixture was concentrated under reduced pressure to obtain crude product, which was purified by combiflash eluting (EtOAc/Hexane 1: 2) to afford compound 4 (0.12 g, 87%) as brown solid. LC-MS: m/z = 288.0 [(M + -55)] (95% purity).

To a solution of tert-butyl 2-(4-( 1 H-indol-3-yl)-4-methyl-2,5- dioxoimidazolidin-1-yl)acetate 4 (0.12 g, 0.35 mmol) in DCM (5 mL) was added TFA (5 mL) and stirred at room temperature for 2 h. After consumption of the starting material (by TLC) and concentrated under reduced pressure to obtain crude compound 5 (0.1 g, 100%) as white solid. LC-MS: m/z = 287.9 [(M + +1)] (90% purity).

To a solution of 2-(4-( 1 H-indol-3-yl)-4-methyl-2,5-dioxoimidazolidin-1- yl)acetic acid 5 (58 mg, 0.2 mmol) and intermediate 24 (35 mg, 0.2 mmol) in DCM (15 mL) was added TEA (40 mg, 0.6 mmol) and 2,4,6-tripropyl-1,3,5,2,4,6- trioxatriphosphorinane-2,4,6-trioxide (0.25mg, 0.4 mmol) under N2 at 0 °C, then stirred at room temperature for 1 h. After consumption of the starting material (by TLC) diluted with water and extracted with DCM. Combined organic extracts were dried over anhydrous MgSO4 and concentrated under reduced pressure to obtain crude product, which was purified by Prep-HPLC (MeCN/H 2 O 7: 3) to afford 23-1 (30 mg, 35%) as light yellow solid. LC-MS: m/z = 445.2(M++1)] at room temperature 4.46 (99.79% purity). 1 H NMR (300 MHz, DMSO-d6) δ 11.15 (s, 1 H), 8.71 (d, J = 8.8 Hz, 1 H), 7.71– 6.71 (m, 10H), 4.66 (d, J = 40.8 Hz, 2H), 4.46– 4.13 (m, 2H), 3.68 (s, 1 H), 1.77 (s, 3H), 1.12 (d, J = 25.0 Hz, 3H), 0.92 (s, 1 H), 0.73– 0.00 (m, 4H). EXAMPLE 24

To a stirring solution of SM1 (1.68 g, 11.95 mmol) in MeOH (10 mL), H 2 O (10 mL) was added (NH 4 ) 2 CO 3 (5.74 g, 59.75 mmol) followed by potassium cyanide (2.33 g, 35.85 mmol) at room temperature. The reaction mixture was heated to 60 °C for 1 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with cold water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 50% EtOAc / PE to afford compound 1 (1.18 g, 47%) as a yellow solid. LC-MS: m/z = 211[(M + +1)].

To a stirring solution of compound 1 (550 mg, 2.62 mmol) in DMF (5 mL) was added K 2 CO 3 (542.3 mg, 3.93 mmol) followed by tert-butyl 2-bromoacetate (536.4 mg, 2.75 mmol) at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 50% EtOAc / PE to afford compound 2 (429 mg, 51%) as a yellow solid. LC-MS: m/z = 269[(M + -55)].

To a stirring solution of compound 2 (336 mg, 1.03 mmol) in DCM (3 mL) was added TFA (3 mL) at room temperature and stirred at room temperature for 0.5 h. After consumption of the starting material (by TLC), the reaction mixture was concentrated under reduced pressure to obtain crude compound 3 (260 mg, 94%) as a yellow solid. LC- MS: m/z = 269[(M + +1)].

To a stirring solution of compound 3 (67 mg, 0.249 mmol) in DMF (1 mL) was added intermediate 24 (43.5 mg, 0.249 mmol) and DIPEA (70.7 mg, 0.548 mmol) followed by HATU (104.1 mg, 0.274 mmol)at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 5% MeOH/DCM to afford 24-1 (150 mg, 94%) as a yellow solid. LC-MS: m/z = 426.1[(M++1)]. 1 H NMR (400MHz, DMSO-d6): δ 8.74 (d, J = 15.1 Hz, 1 H), 7.61– 7.13 (m, 9H), 5.63 (d, J = 15.8 Hz, 1 H), 4.83– 4.54 (m, 2H), 4.51– 4.14 (m, 2H), 3.70 (s, 1 H), 3.39 (s, 1 H), 1.16 (dd, J = 45.9, 13.7 Hz, 3H), 0.94 (s, 1 H), 0.56– 0.07 (m, 4H). EXAMPLE 25

To a stirring solution of 2,6-dichlorobenzaldehyde SM1 (500 mg,

2.87mmol) in CH3CN (20ml),was added TMSCN (369.3mg, 3.73mmol), followed by ZnI2 (27.3mg, 0.086mmol) at room temperature. The reaction mixture stirred at rt for 16 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with aq NaHCO3 and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 20% EA/PE to afford compound 1 (500mg, 63.8%). TLC: 20% EA/PE (R f : 0.3). A solution of 2-(2,6-dichlorophenyl)-2-(trimethylsilyloxy)acetonitrile compound 1 (500 mg, 1.83 mmol), in ethanol (10 mL) was cooled to 0°C. HCl (gas) was charged for 3h. After consumption of the starting material (by TLC), the solvent from reaction mixture was removed under reduced pressure. Obtained residue was washed with ether to afford compound 2 (400 mg, 77%). LC-MS: m/z = 248[(M + +1)].

To a stirring solution of ethyl 2-(2,6-dichlorophenyl)-2-hydroxyacetimidate hydrochloride compound 2 (100 mg, 0.35mmol) in dry THF (10 mL) was added Et 3 N (107mg, 1.06 mmol) followed by phosgene (103.6mg, 0.35mmol) at 0 °C and stirred at room temperature for 1 h. The reaction mixture was diluted with 1N HCl, stirred at room temperature for 0.5 h, and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 30% EA/PE to afford compound 3 (50mg,58%). TLC: 30% EtOAc/Hexane (R f : 0.4). LC-MS: m/z =

246[(M + +1)].

To a stirring solution of 5-(2,6-dichlorophenyl)oxazolidine-2,4-dione compound 3 (50 mg, 0.2mmol) in DMF (10 mL) was added intermediate 23 (60mg, 0.2mmol), followed by K 2 CO 3 (55.2mg, 0.4mmol ) at room temperature and stirred at room temperature for 1 h. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography to afford 25-1 (20 mg, 21.7%) as an off-white solid. LC-MS: m/z = 461 [(M++1)] at room temperature 1.76 (100% purity). 1 H NMR (300 MHz, CD3OD) δ 7.63– 7.12 (m,8H), 6.79 (d, J = 1.2 Hz, 1 H), 4.54 (dd, J = 56.7, 54.0 Hz, 4H), 3.92– 3.73 (m, 1 H), 1.40– 1.15 (m, 4H), 1.06– 0.49 (m, 1 H), 0.29 (d, J = 5.2 Hz, 3H). EXAMPLE 26

SM2 (1.6 g, 10 mmol) was dissolved in ethanol (40 ml) by heating. SM1 (1.17 g, 10 mmol) was added and heating near reflux continued for 15 minutes. After consumption of the starting material (by TLC), 1N Hydrochloric acid (10 ml) was then added while maintaining the reaction reflux. After 10 minutes, the reaction was concentrated to wet solids. Trituration of these wet solids with water give Compound 1 (3.2 g, 100%) as a white solid.

Compound 1 (1.0 g, 3.86 mmol)was heated on a steam bath with 20 ml of 1N sodium hydroxide for 15 minutes, then cooled and crude product precipitated by acidification with conc. Hydrochloric acid, which was purified by silica gel column chromatography eluting with 50% EtOAc /PE to afford Compound 2 (130 mg, 16%) as a yellow solid.

To a stirring solution of compound 1 (110 mg, 0.5 mmol) and intermediate- 23 (148 mg, 0.5 mmol) in DCM (5 mL) was added DIPEA (97 mg,0.75 mmol) and stirred at room temperature overnight. After consumption of the starting material (by TLC), the reaction mixture was diluted with water and extracted with EtOAc. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by prep-hplc to afford 26-1 (12 mg, 6%) as an off white solid. LC-MS: m/z =432.2[(M++1)] at room temperature 5.18 (99.55% purity). 1 H NMR ((400 MHz, DMSO-d6) δ 11.48 (s, 1 H), 7.69 (m, 2H), 7.53– 7.35 (m, 3H), 7.28 (m, 2H), 7.17 (m, 1 H), 7.03 (m, 1 H), 6.51 (d, J = 24.8 Hz, 1 H), 4.88– 4.25 (m, 4H), 3.77 (s, 0.5H), 3.40 (s, 0.5H), 1.19 (dd, J = 18.8, 6.7 Hz, 3H), 0.98 (s, 1 H), 0.62– 0.11 (m, 4H). EXAMPLE 27

To a stirring solution of 4-chloro- 1 H-indole SM1 (1.5 g, 10mmol) in EtOH (40ml), was added compound 1 (1.42g, 10mmol), followed by 1N HCl (10ml) at room temperature. The reaction mixture was heated to reflux for 15 min. After consumption of the starting material (by TLC), the reaction mixture concentrated under reduced pressure to obtain crude product, which washed with water to afford compound 2 (2 g, 68%). TLC: 50% EA/PE (R f : 0.2). LC-MS: m/z = 294 [(M + +1)]

A solution of 5-(4-chloro- 1 H-indol-3-yl)-5-hydroxypyrimidine- 2,4,6( 1 H,3H,5H)-trione compound 1 (2g, 6.8 mmol), in 1N NaOH (40 mL) was stirred for 15min at reflux . Adjust the PH<7 with 1N HCl, extracted with EA, Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 30% EA/PE to afford compound 3 (1g, 58.8%). TLC: 30% EA/PE (R f : 0.3). LC-MS: m/z = 251 [(M + +1)].

To a stirring solution of compound 3 (100 mg, 0.4mmol) in dry DCM (10 mL) was added intermediate 23 (118mg , 0.4 mmol) followed by DIEA (103.2mg, 0.8mmol) at room temperature and stirred at room temperature for overnight. The reaction mixture was diluted with water and extracted with EA. Combined organic extracts were dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure to obtain crude product, which was purified by silica gel column chromatography eluting with 30% EA/PE to afford 27-1 (20mg, 10.7%). LC-MS: m/z = 466 [(M++1)] at room temperature 1.70 (100% purity). 1 H NMR (300 MHz, DMSO) δ 11.91 (s, 1 H), 7.81– 7.06 (m, 9H), 6.79 (d, J = 15.0 Hz, 1 H), 4.97– 4.05 (m, 4H), 3.84– 3.62 (m, 1 H), 1.32– 1.05 (m, 4H), 0.93 (d, J = 7.4 Hz, 1 H), 0.23 (dd, J = 12.2, 6.1 Hz, 4H). EXAMPLE 28

28-1

Compound 28-1 was prepared in a manner analogous to those described above. EXAMPLE 29

29-1

Compound 29-1 was prepared in a manner analogous to those described above.

EXAMPLE 30

30-1

Compound 30-1 was prepared in a manner analogous to those described above.

31-1

Compound 31-1 was prepared in a manner analogous to those described above.

32-1

Compound 32-1 was prepared in a manner analogous to those described above. EXAMPLE 33

33-1

Compound 33-1 was prepared in a manner analogous to those described above. EXAMPLE 34

34-1

Compound 34-1 was prepared in a manner analogous to those described above. EXAMPLE 35

35-1

Compound 35-1 was prepared in a manner analogous to those described above. EXAMPLE 36

36-1

Compound 36-1 was prepared in a manner analogous to those described above. EXAMPLE 37

37-1

Compound 37-1 was prepared in a manner analogous to those described above. EXAMPLE 38

Compound 38-1 was prepared in a manner analogous to those described above. EXAMPLE 39

39-1

Compound 39-1 was prepared in a manner analogous to those described above.

40-1

Compound 40-1 was prepared in a manner analogous to those described above. EXAMPLE 41

41-1 Compound 41-1 was prepared in a manner analogous to those described above. EXAMPLE 42

42-1

Compound 42-1 was prepared in a manner analogous to those described above. EXAMPLE 43

43-1

Compound 43-1 was prepared in a manner analogous to those described above. EXAMPLE 44

44-1 Compound 44-1 was prepared in a manner analogous to those described above. EXAMPLE 45

45-1

Compound 45-1 was prepared in a manner analogous to those described above.

46-1

Compound 46-1 was prepared in a manner analogous to those described above.

EXAMPLE 47

47-1

Compound 47-1 was prepared in a manner analogous to those described above. EXAMPLE 49

TESTING OF REPRESENTATIVE COMPOUNDS

p300 HAT Biochemical Assay

p300 HAT can acetylate all four core histones (H1A, H1B, H3, and H4), with acetylation occurring predominantly on the N-terminal amino acid residues. A p300 HAT assay in SPA format was designed using a substrate (Biotin-C6- GRGKGGKGLGKGGAK) comprising a synthetic peptide of 15 amino acids derived from the N-terminus of human histone 4 (GRGKGGKGLGKGGAK (SEQ ID NO:1)) that is chemically attached to biotin with an amino hexanoic acid linker (C6). The synthetic peptide was re-suspended in nanopure water; adjusted to pH 7.0 with concentrated NaOH; and subjected to amino acid analysis to estimate purity and concentration. Tritiated acetylCoA was diluted with cold acetylCoA (in water) as needed and used in the assay. A truncated variant of the HAT domain of the p300 enzyme was used in the assay. This variant consisted of residues 1287-1666 of p300 and was missing residues 1529-1560 of the autoacetylation loop. In addition, it contained the M1652G mutation as a result of the "expressed protein ligation" method that was used in its preparation, as previously described in the literature (Thompson et.al.2004, Nature Structural & Molecular Biology, 11, 308-315). The test compounds were dissolved in DMSO to make 10mM stocks and diluted further to make 4X stocks in 10mM HEPES, pH 7.8 with 20% DMSO. The final concentration of DMSO in assays was kept at 5%. Compounds were tested at 11 concentrations ranging from 120 μM to 2 nM in 3-fold dilutions.

Assays were performed in a volume of 40μl in a polypropylene 96-well plate. A typical reaction (40μl) contained: 100mM HEPES, pH 7.9, 100mM KCl, 1mM DTT, 50 μg/ml BSA, p300 HAT (~5 nM), 0.01% Triton-X-100, and 5% DMSO. In addition, two combinations of substrate concentrations were used as follows: Biochemical Assay Condition #1: 100 μM biotinylated substrate peptide, 1 μM acetyl CoA.

Biochemical Assay Condition #2: 12.5μM biotinylated substrate peptide, 0.6 μM acetyl CoA.

Briefly, 20μl of 2X reaction mixture (buffer ion, salt, substrate peptide, DTT, BSA, detergent, and enzyme) was incubated with 10 μl of 4X compound stock in 20% DMSO for 30 min. The reaction was initiated by the addition of 10 μl of 4X acetyl CoA and quenched with 120μl of 0.5N HCl at required time intervals. The contents (~160μl) were delivered to the SPA plate coated with avidin and scintillant, incubated for 1 h, and light emission was counted in a TOP COUNT Microplate scintillation and luminescence counter (Perkin Elmer). IC 50 values were calculated based on percent inhibition calculated from these readings.

The compounds in Table 1 were tested according to the above procedures. All of the compounds in Table 1 were found to have an IC 50 against HAT p300 of 120 ^m or less. Table 2 summarizes the activity data of the exemplary compounds from Table 1.

Table 2

Activity Data*

+++ indicates IC 50 of 5 ^m or less

++ indicates IC 50 between 5 ^m and 25 ^m

+ indicates IC 50 from 25 ^m to 120 ^m

*Compounds 2-1 through 34-1 were tested with Biochemical Assay Condition #2; Compounds 35-1 through 47-1 were tested with Biochemical Assay Condition #1 Western Blot

To assess the efficacy of p300 inhibition in vitro, western blot analyses of histone, NF-κB, and p53 acetylation were performed in HeLa and HEK293 cell lines. p300/CBP dependent acetylation of lysines 18 and 27 of H3 histone (see Di Cerbo and Schneider., Briefings in Functional Genomics Advance Access, January 15.2013,pp 1-13), lysine 310 of the NF-kB p65 subunit, and lysine 382 of p53 (in HEK293 only, HeLa do not express p53) was monitored. The involvement of p300/CBP in acetylation at these residues was confirmed in preliminary experiments utilizing p300/CBP siRNA prior to experiments with inhibitors.

Cell treatment

HeLa and HEK 293 were maintained and passaged in EMEM (ATCC, Manassas, VA) supplemented with 10% heat inactivated FBS, and were kept in a humidified incubator at 37 0 C and 5% CO 2 . For experiments, cells were seeded into BD BioCoat TM 6-well poly-D-lysine coated plates (3 x 10 5 cells per well for HeLa and 6 x 10 5 cells for HEK 293) in 2 ml complete media and incubated overnight at 37 0 C and 5% CO 2 . For siRNA experiments, transfections were done the day after seeding. Cells were treated with a mixture of DharmaFECT formulation 1 and 25 pg/mL of p300 siRNA alone, both p300 and CBP siRNAs, or nontargeting siRNA (Thermo Scientific, Pittsburgh, PA) in complete media overnight. Treatments were aspirated off the following day and replaced with fresh media, and cells were incubated an additional 24 h. Cells were then treated with HDAC inhibitors and TNF-α as described below.

For inhibitor studies, compound dilutions were prepared in complete media from 10mM stock solutions in DMSO. 2 mL of media containing the appropriate concentration of inhibitor was added to each well. The final DMSO concentration in all wells was kept at 0.3%. Compounds were allowed to incubate in the presence of compound for 1 h.

Cells were then treated with the HDAC inhibitors nicotinamide (3.3mM) and Trichostatin A (1.65uM) for 1 h. Cells were then treated with TNF-α (10ng/ml) for 1 h prior to lysis. Upon harvest, the media was discarded, and cells were lysed in a lithium dodecyl sulfate buffer containing protease and phophatase inhibitors and TurboDNase. Lysates were boiled for 5 minutes and then frozen for subsequent protein quantitation, electrophoresis, and blotting.

Protein Measurement

Protein of the samples is measured using BCA (Bicinchoninic Acid) Protein assay kit (Thermo Scientific, product # 23227). Protein level is normalized across samples and prepared with sample buffer for loading into gels for electrophoresis.

Western Blot

Prepared samples were loaded on to SDS-polyacrylamide gels using Novex® NuPAGE® gels and transferred to nitrocellulose. After 30 min block with PBS containing Tween and 5% nonfat dry milk at room temperature with constant agitation, overnight incubation with primary antibody was done. Primary antibodies included: anti-pan acetyl- lysine , anti-acetyl NF-ƙB (Ac K310), anti-total NF-ƙB, anti-acetyl histone 3 (Ac K18), anti-acetyl histone 3 (Ac K27), anti-total Histone 3, and anti-β-actin as load control (Cell Signaling Technology). The anti-p300 was obtained from Abcam, and anti-CBP was obtained from Santa Cruz. Anti-p53 acetyl-Lysine 382 was obtained from Cell Signaling Technology, and anti-total p53 antibody was obtained from Santa Cruz. On the following day, membranes were washed 5x for 5min each time with PBS-T milk and incubated with secondary antibody for 3 h and washed 5x for 5 min each time. Secondary antibodies linked to horseradish peroxidase , including anti-rabbit IgG and anti-mouse IgG, were obtained from Life Technologies. Detection was done by chemiluminescence and measured with FluorChem imager (Protein Simple).

CRE luciferase gene reporter assay

CRE luciferase reporter system has tandem repeats of the CRE transcriptional response element and basic promoter elements to drive the transcription of a downstream reporter gene (luciferase). P300, a transcriptional coactivator, binds CRE and drives the transcription of the downstream genes. Cignal CRE luciferase reporter assay kit (Qiagen (CCS-002L)) was used to establish a cell based assay for p300 function. HEK293 cells were purchased from ATCC and cultured in EMEM with 10% HI FBS (ATCC ® 30- 2003). To transfect the reporter in HEK 293 cells, Attractene, a transfection reagent from Qiagen (301005) was used.

HEK-293 cells, were grown in EMEM with 10% FBS to 70% confluency. The cells were harvested and plated in a 96 well Poly D-Lys plate (BIOCOAT ® Becton and Dickinson) with a cell density of 60000 cells/well in 100 µL of EMEM with 10% HI FBS. In parallel, the transfection cocktail (provided by the kit) was prepared in plain EMEM with no FBS. Positive control (GFP) and negative control from the kit were included. The plate was treated with 25-50 µL of transfection cocktail and incubated at 37˚C for 18-20 h. The next day, the plate was gently tapped over an absorbent pad to remove media. 100 µL aliquots of compounds at various concentrations (in EMEM with 10% HI FBS and 0.4% DMSO) were delivered to the appropriate wells and incubated for 1 h at 37˚C. The control wells received media (EMEM with 10% HI FBS and 0.4% DMSO) with no compound. The plate was then treated with Forskolin (final conc: 2 µM) in EMEM (with 0.4% DMSO and 10% HI FBS) and incubated for 3 h at 37˚C. After 3 h, the plate was gently tapped over an absorbent pad, treated with 50 µL of passive lysis buffer (from Qiagen, Dual Luciferase Reporter Kit), and incubated at room temperature for 20 min. The plate was subjected to a freeze-thaw cycle, and a 20 µL aliquot was assayed for luciferase activity using DLR Kit with a GloMAX® luminometer. The luminescence from“unstimulated” control wells was subtracted from the rest, and the EC 50 was calculated.

NF-ƙB luciferase gene reporter assay

The protocol is identical to CRE luciferase gene reporter assay except Cignal NF-ƙB reporter assay kit from Qiagen (CCS-013L) was used, and human TNF-α (final conc: 5 ng/mL in EMEM with 0.4% DMSO and 10% FBS) was used for induction. EXAMPLE 50

CELL PROLIFERATION ASSAY 3 H-thymidine incorporation assay to measure cell proliferation

The protocol for the methyl- 3 H-thymidine incorporation assay to measure and rank the effect of our p300/CBP Histone Acetyl Transferase (HAT) inhibitors on the proliferation of a collection n of cancer cell lines was adapted from Griffiths, M and Sundaram, H. Drug Design and Testing: Profiling of Antiproliferative Agents for Cancer Therapy Using a Cell-Based Methyl-[3H]-Thymidine Incorporation Assay. In: Cancer Cell Culture: Methods and Protocols, Second Edition, Methods in Molecular Biology, vol.731, 451-465, 2011. Ian A. Cree (ed.), Springer Science+Business Media, LLC.

Determination optimal cell seeding density

As a first step the optimal number of cells per well to be seeded for each cancer cell line has to be determined. For scientific and logistical reasons the optimal seeding density was determined for a 72 h assay. Most tumor cell lines have an approximate doubling time of 24-48 h, thus to be able to observe an effect on cell proliferation 72 h seems an appropriate time period because the cells will undergo one or more divisions. Cells were counted using an inverted microscope, hemocytometer and trypan blue. Solid tumor cell lines were first treated with trypsin to prepare a single-cell suspension. For hematopoietic tumor cell lines a suspension containing 4 x10 5 viable cells/mL and for solid tumors a suspension containing 2x10 5 viable cells/mL were prepared.

Cell suspensions are added to wells in row A of a 96-well sterile polystyrene surface treated culture plate and serial two-fold dilutions are performed across the plate. The plates are incubated at a temperature of 37 o C and an atmosphere of 5% CO 2 in air for 16-20 h after which 100 µL of fresh culture medium was added to each well. The plates were then returned to the incubator for an additional 72 h. During the last 6 h the cells are incubated in the presence of 0.1 mCi/well.

After 6 h the plates were harvested using a PerkinElmerFilterMate cell harvester and the contents of the wells collected on 96-well Unifilter plates. After drying the plates at 37 o C for greater than 16 h the plates, scintillation fluid was added to the wells and the radioactivity in each well determined using a TopCount NXT scintillation counter (PerkinElmer).

The log cell density was plotted vs. the calculated average cpm to determine the optimal cell density (exponential growth phase/rising part of the curve). The results are shown in the Table 3 below. Table 3

ll lin n h ir im l in n i 2 h

Determination effect of HAT inhibitors and chemotherapeutic agents on cell proliferation

Cells at their predetermined optimal cell density (see Table 3) were added to wells of 96-well sterile polystyrene surface treated culture plates in a volume of 150 µL/well. The plates were incubated for 16-20 h in an incubator set to maintain a temperature of 37 o C and an atmosphere of 5% CO 2 in air.

The next day serial dilutions of HAT inhibitors and selected chemotherapeutics (cytarabine, doxorubicin and paclitaxel) were prepared as follows. HAT inhibitors were diluted to 5 mM in DMSO from a 10 mM stock solution. The 5 mM solution was further diluted 166.7-fold in the appropriate culture medium such that each cell line received a 30 mM solution in culture medium + 0.6% DMSO. This solution was added to wells in row A of a 96-well plate and serial three-fold dilutions were performed across the plate.

Chemotherapeutics dilutions were made in a similar fashion, except that the mM solution was diluted 125-fold in the appropriate culture medium for each cell line to get a 40 mM solution in culture medium + 0.8% DMSO. The 40 mM solution was then diluted 2.5-fold in the appropriate culture medium supplemented with 0.6% DMSO to get a 16 mM solution in 0.68% DMSO. This solution was added to wells in row A of a 96-well plate and serial four-fold dilutions awere performed across the plate.

HAT inhibitor and chemotherapeutic agent dilutions were then added to corresponding wells of a 96-well plate containing cells seeded the day before, resulting in another 4-fold dilution and a final DMSO concentration in the wells of 0.2%. The final concentrations of the HAT inhibitors and chemotherapeutic agents in the assay are shown in Table 4. Table 4

HAT inhi i r r h m h r i n n n r i n in 3 H-Th mi in

The plates were then incubated at 37 o C, 5% CO 2 for an additional 72 h. During the last 6 h the cells were incubated in the presence of 0.1 mCi/well (20 mL of a 1:200 dilution of a 1 mCi/mL stock solution) and processed as described above for determination of the optimal cell density.

The mean cpm for each triplicate condition and the percent inhibition were calculated for each HAT inhibitor and chemotherapeutic concentration as follows: mean cpm in treatment group– mean cpm medium control (no cells)/mean cpm cells only- medium cpm medium control x 100% (normalized response). The data was entered in GraphPad Prism and the IC 50 value for each inhibitor was determined using the log inhibitor concentration vs. normalized response-variable slope function.

All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification or Application Data sheet are incorporated herein by reference, in their entirety to the extent not inconsistent with the present description.

From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.