Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
1,4-PYRIDONE COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2014/100646
Kind Code:
A1
Abstract:
The present invention relates to 1,4-pyridone compounds. The present invention also relates to pharmaceutical compositions containing these compounds and methods of treating cancer by administering these compounds and pharmaceutical compositions to subjects in need thereof. The present invention also relates to the use of such compounds for research or other non-therapeutic purposes.

Inventors:
KUNTZ KEVIN W (US)
CAMPBELL JOHN EMMERSON (US)
Application Number:
PCT/US2013/077048
Publication Date:
June 26, 2014
Filing Date:
December 20, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
EPIZYME INC (US)
International Classes:
C07D213/68; A61K31/4422; A61P35/00; C07D401/04; C07D401/12; C07D401/14; C07D409/12; C07D413/12; C07D413/14; C07D471/04; C07D498/04
Domestic Patent References:
WO2012142513A12012-10-18
WO2012068589A22012-05-24
WO2012142504A12012-10-18
WO2012142513A12012-10-18
WO2012118812A22012-09-07
Foreign References:
US20090197834A12009-08-06
US20130040906A12013-02-14
US4522811A1985-06-11
US5763263A1998-06-09
Other References:
See also references of EP 2935214A4
SNEERINGER ET AL., PROC NATL ACAD SCI USA, vol. 107, no. 49, 2010, pages 20980 - 5
CAHN ET AL., ANGEW. CHEM. INTER. EDIT., vol. 5, 1966, pages 385
CAHN ET AL., ANGEW. CHEM., vol. 78, 1966, pages 413
CAHN; INGOLD, J. CHEM. SOC., 1951, pages 612
CAHN ET AL., EXPERIENTIA, vol. 12, 1956, pages 81
CAHN, J. CHEM. EDUC., vol. 41, 1964, pages 116
PATANI; LAVOIE, CHEM. REV., vol. 96, 1996, pages 3147 - 3176
SMITH, M. B.; MARCH, J.: "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2001, JOHN WILEY & SONS
GREENE, T.W.; WUTS, P.G. M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
L. FIESER; M. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 2005, JOHN WILEY AND SONS, INC.
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 2000, COLD SPRING HARBOR PRESS
COLIGAN ET AL.: "Current Protocols in Immunology", JOHN WILEY & SONS
ENNA ET AL.: "Current Protocols in Pharmacology", JOHN WILEY & SONS
FINGL ET AL.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO., article "The Pharmacological Basis of Therapeutics"
"Remington: the Science and Practice of Pharmacy", 1995, MACK PUBLISHING CO.
DEVLIN: "High Throughput Screening", 1998, MARCEL DEKKER
Attorney, Agent or Firm:
ELRIFI, Ivor R. et al. (P.C.One Financial Cente, Boston MA, US)
Download PDF:
Claims:
What is claimed is:

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

wherein

Z is NR7R8, OR7, S(0)aR7, or CR7R8R14, in which a is 0, 1, or 2;

each of Ri, R5, Rg, and Rio, independently, is H or Ci-C6 alkyl optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-Ci-C6 alkyl, cyano, Ci-C6 alkoxyl, amino, mono-Ci-C6 alkylamino, di-Ci- C alkylamino, C3-C8 cycloalkyl, C6-Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

each of R2, R3, and R4, independently, is -Q1-T1, in which Qj is a bond or C1-C3 alkyl linker optionally substituted with halo, cyano, hydroxyl or Ci-C6 alkoxy, and Ti is H, halo, hydroxyl, C(0)OH, cyano, azido, or Rsl, in which RSi is C1-C3 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Ci-C6 alkoxyl, Ci-C6 thioalkyl, C(0)0-Ci-C6 alkyl, CONH2, S02NH2, - C(0)-NH(Ci-C6 alkyl), -C(0)-N(Ci-C6 alkyl)2, -S02-NH(Ci-C6 alkyl), - S02-N(Ci-C6 alkyl)2, C3-C8 cycloalkyl, C6-Cio aryl, C6-Cio aryloxy, amino, mono-Ci-C6 alkylamino, di- Ci-C6 alkylamino, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and Rsi is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C(0)OH, C(0)0-Ci-C6 alkyl, cyano, Ci-C6 alkoxyl, amino, mono-Ci-C6 alkylamino, di-Ci-C6 alkylamino, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl; or Ri and R2, together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or Ri and R4, together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12- membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R3 and R4, together with the atoms to which they are attached, form Cs-Cg cycloalkyl, C6-C10 aryl, or a 5- or 6-membered heteroaryl having 1 to 3 heteroatoms, or a 5 to 12-membered

heterocycloalkyl ring having 1 to 3 heteroatoms; in which each of the ring structures formed by Ri and R2, by R and R4, or by R3 and R4, independently is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C -C alkyl, C(0)OH, C(0)0-C1-C6 alkyl, cyano, C -C alkoxyl, amino, mono-C Ce alkylamino, di-C C6 alkylamino, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R6 is H, halo, cyano, azido, ORa, -NRaRb, -C(0)Ra, -C(0)ORa, -C(0)NRaRb, -NRbC(0)Ra, -S(0)bRa, -S(0)bNRaRb, or RS2, in which RS2 is Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Cg cycloalkyl, C6-Cio aryl, 5- or 6-membered heteroaryl, or 4 to 12- membered heterocycloalkyl, b is 0, 1, or 2, each of Ra and Rb, independently is H or Rs3, and Rs3 is Q-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12- membered heterocycloalkyl, or 5- or 6-membered heteroaryl; or Ra and Rb, together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 or 1 additional heteroatom; and each of Rs2, Rs3, and the 4 to 12-membered heterocycloalkyl ring formed by Ra and Rb, is optionally substituted with one or more -Q2- T2, wherein Q2 is a bond or C C3 alkyl linker each optionally substituted with halo, cyano, hydroxyl or Ci-C6 alkoxy, and T2 is H, halo, cyano, -ORc, -NRcRd, -C(0)Rc, -C(0)ORc, - C(0)NRcRd, -NRdC(0)Rc, -NRdC(0)ORc, -S(0)2Rc, -S(0)2NRcRd, or RS4, in which each of Rc and Rd, independently is H or Rss, each of Rs4 and Rss, independently, is C C6 alkyl, C3-Cg cycloalkyl, C6-Cio aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, or Rc and Rd, together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 or 1 additional heteroatom, and each of Rs4, Rss, and the 4 to 12-membered heterocycloalkyl ring formed by Rc and Rd, is optionally substituted with one or more -Q3-T , wherein Q is a bond or Ci-C3 alkyl linker each optionally substituted with halo, cyano, hydroxyl or Ci-C6 alkoxy, and T is selected from the group consisting of halo, cyano, C C6 alkyl, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12- membered heterocycloalkyl, 5- or 6-membered heteroaryl, ORe, C(0)ORe, -S(0)2Re, - NReRf, and -C(0)NReRf, each of Re and Rf independently being H or Q-C6 alkyl optionally substituted with OH, 0-C -C alkyl, or NH-Q-C6 alkyl, or -Q3-T is oxo; or-Q2-T2 is oxo; or any two neighboring -Q2-T2, when R6 is C6-C10 aryl or 5- or 6-membered heteroaryl, together with the atoms to which they are attached form a 5- or 6-membered ring optionally containing 1-4 heteroatoms selected from N, O and S and optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0- C -C alkyl, cyano, Ci-C6 alkoxyl, amino, mono-C Ce alkylamino, di-C^Ce alkylamino, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R7 is -Q4-T4, in which Q4 is a bond, C C4 alkyl linker, or C2-C4 alkenyl linker, each linker optionally substituted with halo, cyano, hydroxyl or Ci-C6 alkoxy, and T4 is H, halo, cyano, NRgRh, -ORg, -C(0)Rg, -C(0)ORg, -C(0)NRgRh, -C(0)NRgORh, -NRgC(0)Rh, -S(0)2Rg, or Rs6, in which each of Rg and Rh, independently is H or Rs7, each of Rs6 and Rs7, independently is C C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 14-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and each of Rs6 and RS7 is optionally substituted with one or more -Q5-T5, wherein (¾ is a bond, C(O), C(0)NRk, NRkC(0), NRk, S(0)2, NRkS(0)2, or Q-C3 alkyl linker, Rk being H or Ci-C6 alkyl, and T5 is H, halo, CrC6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, hydroxyl, cyano, CrC6 alkoxyl, amino, mono-Q-Ce alkylamino, di-Q-Ce alkylamino, CrCg cycloalkyl, C6-Cio aryl, 4 to 12-membered heterocycloalkyl, 5- or 6-membered heteroaryl, or S(0)qRq in which q is 0, 1, or 2 and Rq is CrC6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and T5 is optionally substituted with one or more substituents selected from the group consisting of halo, C -C alkyl, hydroxyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, di-Q- C6 alkylamino, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl except when T5 is H, halo, hydroxyl, or cyano; or -Q5-T5 is oxo; each of R8, and R12, independently, is H, halo, hydroxyl, C(0)OH, cyano, RS8, ORsg, or C(0)ORS8, in which RS8 is Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 4 to 12-membered heterocycloalkyl, amino, mono-Q-Ce alkylamino, or di-CrC6 alkylamino, and Rs8 is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-C1-C6 alkyl, cyano, C -C alkoxyl, amino, mono-Ci-Ce alkylamino, and di-C^Ce alkylamino; or R7 and R8, together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms, or R7 and Rg, together with the atom to which they are attached, form C3-C8 cycloalkyl or a 4 to 12-membered heterocycloalkyl ring having 1 to 3 heteroatoms, and each of the 4 to 12-membered heterocycloalkyl rings or C3-C8 cycloalkyl formed by R7 and Rg is optionally substituted with one or more -Q6-T6, wherein Q6 is a bond, C(O), C(0)NRm, NRmC(0), S(0)2, or C1-C3 alkyl linker, Rm being H or Ci-C6 alkyl, and T6 is H, halo, CrC6 alkyl, hydroxyl, cyano, CrC6 alkoxyl, amino, mono-Ci-C6 alkylamino, di-Q-Ce alkylamino, C3-C8 cycloalkyl, C6-Cio aryl, 4 to 12-membered heterocycloalkyl, 5- or 6-membered heteroaryl, or S(0)pRp in which p is 0, 1, or 2 and Rp is CrC6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and T6 is optionally substituted with one or more substituents selected from the group consisting of halo, C -C alkyl, hydroxyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, di-Q-Ce alkylamino, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl except when T6 is H, halo, hydroxyl, or cyano; or -Q6-T6 is oxo; and

R14 is absent, H, or CrC6 alkyl optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-C1-C6 alkyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, di-Q-Ce alkylamino, C3-C8 cycloalkyl, C6- Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl.

2. The compound of claim 1 wherein the compound is of Formula (la):

3. The compound of claim 1, wherein the compound is of Formula (lb):

wherein R4 and R12 are each, independently Ci_6 alkyl, and R2 is Ci_6 alkyl or halo.

4. The compound of any of claims 1-3, wherein R6 is C6-C10 aryl or 5- or 6-membered heteroaryl, each of which is optionally, independently substituted with one or more -Q2-T2, wherein Q2 is a bond or Q-C3 alkyl linker, and T2 is H, halo, cyano, -ORc, -NRcRd, - C(0)NRcRd, -NRdC(0)Rc, -S(0)2Rc, -S(0)2NRcRd, or RS4, in which each of Rc and Rd, independently is H or Rss, each of Rs4 and Rss, independently, is C C6 alkyl, or Rc and Rd, together with the N atom to which they are attached, form a 4 to 7-membered

heterocycloalkyl ring having 0 or 1 additional heteroatom, and each of RS4, Rss, and the 4 to 7-membered heterocycloalkyl ring formed by Rc and Rd, is optionally, independently substituted with one or more -Q3-T3, wherein Q3 is a bond or CrC3 alkyl linker and T3 is selected from the group consisting of halo, CrC6 alkyl, 4 to 7-membered heterocycloalkyl, ORe, -S(0)2Re, and -NReRf, each of Re and Rf independently being H or C -C alkyl optionally substituted with OH, 0-C -C alkyl, or NH-C Ce alkyl, or -Q3-T is oxo; or any two neighboring -Q2-T2, together with the atoms to which they are attached form a 5- or 6- membered ring optionally containing 1-4 heteroatoms selected from N, O and S.

5. The compound of any of claims 1-4, wherein the compound is of Formula (II):

wherein Q2 is a bond or methyl linker, and T2 is H, halo, -ORc, -NRcRd, or -S(0)2NRcRd.

6. The compound of claim 5, wherein the compound is of Formula (Ila):

7. The compound of claim 6, wherein Rc and Rd, together with the N atom to which they are attached, form a 4 to 7-membered heterocycloalkyl ring having 0 or 1 additional heteroatoms to the N atom and the ring is optionally substituted with one or more -Q3-T3> wherein the heterocycloalkyl is azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6- tetrahydropyridinyl, piperazinyl, or morpholinyl.

8. The compound of any of claims 1-3, wherein R6 is halo, CrC3 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, C(0)H, or -C(0)Ra, in which Ra is Ci-Ce alkyl or 4 to 12-membered heterocyclo alkyl .

9. The compound of any of claims 1-3, wherein R6 is F, Br, or CI.

10. The compound of any of claims 1-3, wherein R6 is CI.

11. The compound of any of claims 1-10, wherein R7 is C3-C8 cycloalkyl or 4 to 7- membered heterocycloalkyl, each optionally substituted with one or more -Q5-T5.

12. The compound of any of claims 1-11, wherein R7 is piperidinyl, tetrahydropyran, tetrahydro-2H-thiopyranyl, piperazinyl, cyclopentyl, cyclohexyl, pyrrolidinyl, or cycloheptyl, each optionally substituted with one or more -Q5-T5.

13. The compound of any of claims 1-12, wherein Rg is H or CrC6 alkyl which is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-C1-C6 alkyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, and di-Q-Ce alkylamino.

14. The compound of any of claims 1-13, wherein R7 is piperidinyl, tetrahydropyran, cyclopentyl, or cyclohexyl, each optionally substituted with one -Q5-T5 and R8 is ethyl.

pound of any of claims 1-14, wherein R7 is tetrahydropyran, 5 or

16. The compound of any of claims 1-15, wherein R7 is

17. The compound of claim 1, wherein the compound is of Formula (IV):

wherein R7 is -Q4-T4, wherein Q4 is a bond or methyl linker, T4 is optionally substituted C3- Cg cycloalkyl or optionally substituted 4- to 14-membered heterocycloalkyl.

18. The compound of claim 1, wherein the compound is of Formula (IVc):

(IVc)

Wherein:

R/U1 is H or Ci-4 alkyl;

702

R is Ci_6 alkoxyl or C6-Cio aryloxy, each optionally substituted with one or more halo;

R703 is H, halo, or CM alkyl;

R704 is halo, C1-4 alkyl, or C1-6 alkoxyl,where each CrC4 alkyl or CrC6 alkoxy is optionally substituted with one or more halo; or R 701 and R 702 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R and R , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocyclo alkyl ring having 0 to 2 additional heteroatoms; or R703 and R704, together with the atoms to which they are attached, form Cs-Cg cycloalkyl, C6-10 aryl, or a 5- or 6- membered heteroaryl having 1 to 3 heteroatoms, or a 5 to 12-membered heterocyclo alkyl

701 ring having 1 to 3 heteroatoms; in which each of the ring structures formed by R and R702, by R701 and R704, or by R703 and R704, independently is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C -C alkyl, C(0)OH, C(0)0-Ci-C6 alkyl, cyano, CrC6 alkoxyl, amino, mono-Ci-C6 alkylamino, di-Cr C6 alkylamino, C3-C8 cycloalkyl, C6-C10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R705 is Ci_4 alkyl; and

R706 is tetrahydropyranyl, piperidine substituted by 1, 2, or 3 Ci_4 alkyl groups, or cyclohexyl substituted by N(C1-4 alkyl)2 wherein one or both of the C1-4 alkyl is optionally substituted with C1-6 alkoxyl.

19. The compound of claim 18, wherein R/L,D is or

The compound of claim 18 or 19, wherein R6 or halo.

21. A pharmaceutical composition comprising a compound of any of claims 1-20 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.

22. A method of treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a compound of any of claims 1-20 or a pharmaceutically acceptable salt thereof.

23. The method of claim 22, wherein the cancer is lymphoma, leukemia or melanoma.

Description:
1,4-PYRIDONE COMPOUNDS

RELATED APPLICATIONS

[001] This application claims priority to, and the benefit of, U.S. provisional application No. 61/745,182, filed December 21, 2012 and U.S. provisional application No. 61/785,827, filed March 14, 2013. The entire contents of each of these applications are incorporated herein by reference in their entireties.

BACKGROUND OF THE INVENTION

[002] There is an ongoing need for new agents as inhibitors of EZH2 mutants, which can be used for treating an EZH2-mediated disorder (e.g., cancer).

SUMMARY OF THE INVENTION

[003] In one aspect, the present invention features a 1,4-pyridone compound of Formula (I) below or a pharmaceuticall acceptable salt thereof.

(I).

In this Formula,

Z is NR 7 R 8 , OR 7 , S(O) a R 7 , or CR 7 R 8 R 14 , in which a is 0, 1, or 2;

each of R 1 ; R5, R 9 , and R 10 , independently, is H or Ci-C 6 alkyl optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(O)OH, C(O)O-Ci-C 6 alkyl, cyano, C C 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Cr C alkylamino, C3-C8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl; each of R 2 , R 3 , and R 4 , independently, is -Q T^ in which Qi is a bond or CrC 3 alkyl linker optionally substituted with halo, cyano, hydroxyl or CrC 6 alkoxy, and Ύι is H, halo, hydroxyl, C(0)OH, cyano, azido, or R sl , in which R sl is Q-C3 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, Ci-C 6 alkoxyl, Ci-C 6 thioalkyl, C(0)0-Ci-C 6 alkyl, CONH 2 , S0 2 NH 2 , -CO- NH(Ci-C 6 alkyl), -CO-N(Ci-C 6 alkyl) 2 , -S0 2 -NH(Ci-C 6 alkyl), - S0 2 -N(Ci-C 6 alkyl) 2 , C 3 - C 8 cycloalkyl, C 6 -C 10 aryl, C 6 -C 10 aryloxy, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and R sl is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Q- C 6 alkylamino, di-CrC 6 alkylamino, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl; or R and R 2 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R and R^, together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R and R 4 , together with the C atoms to which they are attached, form Cs-Cg cycloalkyl, C 6 -Cio aryl, or a 5- or 6- membered heteroaryl having 1 to 3 heteroatoms, or a 5 to 12-membered heterocycloalkyl ring having 1 to 3 heteroatoms; in which each of the ring structures formed by R and R 2 , by Ri and R^, or by R and R^, independently is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C -C alkyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q- C 6 alkylamino, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R 6 is H, halo, cyano, azido, OR a , -NR a R b , -C(0)R a , -C(0)OR a , -C(0)NR a R b , -NR b C(0)R a , -S(0) b R a , -S(0) b NR a Rb, or R S2 , in which R S2 is Ci-Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 5- or 6-membered heteroaryl, or 4 to 12- membered heterocycloalkyl, b is 0, 1, or 2, each of R a and R , independently is H or R S , and R S3 is C C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12- membered heterocycloalkyl, or 5- or 6-membered heteroaryl; or R a and R b , together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 or 1 additional heteroatom; and each of R S2 , R S , and the 4 to 12-membered heterocycloalkyl ring formed by R a and R b , is optionally substituted with one or more -Q 2 - T 2 , wherein Q 2 is a bond or Q-C3 alkyl linker each optionally substituted with halo, cyano, hydroxyl or Ci-Ce alkoxy, and T 2 is H, halo, cyano, -OR c , -NR c R d , -C(0)R c , -C(0)OR c , - C(0)NR c R d , -NR d C(0)R c , -NR d C(0)OR c , -S(0) 2 R c , -S(0) 2 NR c R d , or R S4 , in which each of R c and R d , independently is H or R S 5, each of R S4 and R S 5, independently, is C -C alkyl, C3-C 8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, or R c and R d , together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 or 1 additional heteroatom, and each of Rs 4 , R S 5, and the 4 to 12-membered heterocycloalkyl ring formed by R c and R d , is optionally substituted with one or more -Q3-T3, wherein Q 3 is a bond or Q-C3 alkyl linker each optionally substituted with halo, cyano, hydroxyl or CrC 6 alkoxy, and T 3 is selected from the group consisting of halo, cyano, CrC 6 alkyl, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12- membered heterocycloalkyl, 5- or 6-membered heteroaryl, OR e , C(0)OR e ,

-S(0) 2 R e , -NR e R f , and -C(0)NR e R f , each of R e and R f independently being H or Ci-C 6 alkyl optionally substituted with OH, 0-C -C alkyl, or NH-Ci-C 6 alkyl, or -Q3-T3 is oxo; or-Q 2 - T 2 is oxo; or any two neighboring -Q 2 -T 2 , when R 6 is C 6 -C 10 aryl or 5- or 6-membered heteroaryl, together with the atoms to which they are attached form a 5- or 6-membered ring optionally containing 1-4 heteroatoms selected from N, O and S and optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q- C 6 alkylamino, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R 7 is -Q 4 -T 4 , in which Q 4 is a bond, C C 4 alkyl linker, or C 2 -C 4 alkenyl linker, each linker optionally substituted with halo, cyano, hydroxyl or CrC 6 alkoxy, and T 4 is H, halo, cyano, NR g R h , -OR g , -C(0)R g , -C(0)OR g , -C(0)NR g R h , -C(0)NR g OR h , -NR g C(0)R h , -S(0) 2 R g , or R S 6, in which each of R g and R h , independently is H or R S7 , each of R s6 and Rs 7 , independently is C -C alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 6 -Cio aryl, 4 to 14-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and each of Rs 6 and Rs 7 is optionally substituted with one or more -Q5-T5, wherein Q5 is a bond, C(O), C(0)NR k , NR k C(0), NR k , S(0) 2 , NR k S(0) 2 , or C 1 -C3 alkyl linker, R k being H or Ci-C 6 alkyl, and T5 is H, halo, C -C alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, hydroxyl, cyano, C -C alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, C 3 -C 8 cycloalkyl, CrC 6 alkylene-C3-C8 cycloalkyl, C6-C 10 aryl, CrC 6 alkylene-Ce-Qo aryl, 4 to 12-membered heterocycloalkyl, C -C alkylene-4 to 12-membered heterocycloalkyl, 5- or 6-membered heteroaryl, C -C alkylene-5- or 6-membered heteroaryl, or S(0) q R q in which q is 0, 1, or 2 and R q is CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12- membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and T 5 is optionally substituted with one or more substituents selected from the group consisting of halo, CrC 6 alkyl, hydroxyl, cyano, C -C alkoxyl, O-Q-Gi alkylene-C 1 -C 4 alkoxy, amino, mono-C Ce alkylamino, di-C^Ce alkylamino, C3-C8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl except when T5 is H, halo, hydroxyl, or cyano; or -Q5-T5 is oxo;

each of R 8 , and R 12 , independently, is H, halo, hydroxyl, C(0)OH, cyano, R S8 , ORs 8 , or C(0)OR S8 , in which R S8 is Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, 4 to 12-membered heterocycloalkyl, amino, mono-Ci-C 6 alkylamino, or di-C C 6 alkylamino, and Rs 8 is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-Ce alkylamino, and di-C^Ce alkylamino; or R 7 and R 8 , together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms, or R 7 and Rg, together with the atom to which they are attached, form C 3 -C8 cycloalkyl or a 4 to 12-membered heterocycloalkyl ring having 1 to 3 heteroatoms, and each of the 4 to 12-membered heterocycloalkyl rings or C 3 -Cg cycloalkyl formed by R 7 and R 8 is optionally substituted with one or more -Q6-T 6 , wherein Q 6 is a bond, C(O), C(0)NR m , NR m C(0), S(0) 2 , or C 1 -C3 alkyl linker, R m being H or Ci-C 6 alkyl, and T 6 is H, halo, CrC 6 alkyl, hydroxyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, C 3 -Cg cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, 5- or 6-membered heteroaryl, or S(0) p R p in which p is 0, 1, or 2 and R p is CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Cs cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and T 6 is optionally substituted with one or more substituents selected from the group consisting of halo, C -C alkyl, hydroxyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, di-Q-Ce alkylamino, C 3 -Cg cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl except when T 6 is H, halo, hydroxyl, or cyano; or -Q6-T 6 is oxo; and

R 14 is absent, H, or C -C alkyl optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-C 1 -C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-CrC 6 alkylamino, C 3 -Cs cycloalkyl, C 6 - Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl.

[004] One subset of the compounds of Formula (I) includes those of Formula (la):

[005] Another subset of the com ounds of Formula (I) includes those of Formula (lb):

wherein R 4 and R 12 are each, independently Ci_6 alkyl, and R 2 is Ci_6 alkyl or halo.

[006] Still another subset of the compounds of Formula (I) includes those of Formula (II) or (Ila):

(Π) (Ila), wherein Q 2 is a bond or methyl linker, T 2 is H, halo, -OR c , -NR c Rd, or -S(0) 2 NR c Rd.

[007] The compounds of Formulae (I), (la), (lb), (II), and (Ila) can include one or more of the following features when applicable:

[008] Z is NR 7 R 8 .

[009] Z is CR 7 R 8 R 14 .

[010] Z is OR 7 .

[011] Z is S(0) a R 7 , in which a is 0, 1, or 2.

[012] Z is SR 7 .

[013] R 6 is C 6 -C 10 aryl or 5- or 6-membered heteroaryl, each of which is optionally, independently substituted with one or more -Q 2 -T 2 , wherein Q 2 is a bond or C C 3 alkyl linker, and T 2 is H, halo, cyano, -OR c , -NR c R d , -C(0)NR c R d , -NR d C(0)R c , -S(0) 2 R c , - S(0) 2 NR c R d , or R S4 , in which each of R c and R d , independently is H or R S 5, each of R S4 and Rss, independently, is Ci-C 6 alkyl, or R c and R d , together with the N atom to which they are attached, form a 4 to 7-membered heterocycloalkyl ring having 0 or 1 additional heteroatom, and each of Rs 4 , Rss, and the 4 to 7-membered heterocycloalkyl ring formed by R c and R d , is optionally, independently substituted with one or more -Q3-T3, wherein Q 3 is a bond or C C 3 alkyl linker and T 3 is selected from the group consisting of halo, Ci-C 6 alkyl, 4 to 7-membered heterocycloalkyl, OR e , -S(0) 2 R e , and -NR e R f , each of R e and R f independently being H or C C 6 alkyl optionally substituted with OH, 0-C C 6 alkyl, or NH-Ci-Ce alkyl, or -Q3-T 3 is oxo; or any two neighboring -Q 2 -T 2 , together with the atoms to which they are attached form a 5- or 6-membered ring optionally containing 1-4 heteroatoms selected from N, O and S.

[014] R c and R d , together with the N atom to which they are attached, form a 4 to 7- membered heterocycloalkyl ring having 0 or 1 additional heteroatoms to the N atom and the ring is optionally substituted with one or more -Q3-T 3 wherein the heterocycloalkyl is azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, or morpholinyl.

[015] R 6 is phenyl or 5- or 6-membered heteroaryl substituted with O-Ci-6 alkyl or NH- Ci-6 alkyl, each of which is optionally substituted with hydroxyl, O-Ci-3 alkyl or NH-Ci-3 alkyl, each of the O-Ci-3 alkyl and NH-Ci-3 alkyl being optionally further substituted with 0-Ci_ 3 alkyl or NH-Ci_ 3 alkyl.

[017] R 6 is halo, C C 3 alkyl, C 2 -C 6 alkenyl, C 3 -C 6 cycloalkyl, C(0)H, or -C(0)R a , in which R a is CrC 6 alkyl or 4 to 12-membered (e.g., 4 to 7-membered) heterocycloalkyl.

[018] R 6 is F, Br, or CI.

[019] Re is CI.

[020] R 7 is C 3 -C8 cycloalkyl or 4 to 7-membered heterocycloalkyl, each optionally substituted with one or more -Q5-T5.

[021] R 7 is piperidinyl, tetrahydropyran, tetrahydro-2H-thiopyranyl, piperazinyl, cyclopentyl, cyclohexyl, pyrrolidinyl, or cycloheptyl, each optionally substituted with one

[022] Rg is H or CrC 6 alkyl which is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-C 1 -C 6 alkyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, and di-Q-Ce alkylamino.

[023] R 7 is piperidinyl, tetrahydropyran, cyclopentyl, or cyclohexyl, each optionally substituted with one -Q5-T5 and Rg is ethyl.

[024] R 7 is tetrahydropyran

[030] R 7 is Rioo is phenyl, 5- or 6- membered heteroaryl, or 4 to 12-membered heterocycloalkyl, each optionally substituted with one or more T 5a in which each T 5a is independently Ci-C 6 alkoxyl or O-C G* alkylene- Q-C 4 alkoxy, and R 10 i is H or Q-C 4 alkyl.

wherein each T 5a is independently C C? alkoxyl or 0-C C 3 alkylene-CrCi alkoxy.

[032] The present invention also provides pharmaceutical compositions comprising one or more pharmaceutically acceptable carriers and one or more compounds selected from those of any of the Formulae described herein.

[033] Another aspect of this invention is a method of treating or preventing an EZH2- mediated disorder. The method includes administering to a subject in need thereof a therapeutically effective amount of one or more compounds selected from those of any of the Formulae described herein. The EZH2-mediated disorder is a disease, disorder, or condition that is mediated at least in part by the activity of EZH2. In one embodiment, the EZH2-mediated disorder is related to an increased EZH2 activity. In one embodiment, the EZH2-mediated disorder is a cancer. The EZH2-mediated cancer may be lymphoma, leukemia or melanoma, for example, diffuse large B-cell lymphoma (DLBCL), non- Hodgkin's lymphoma (NHL), follicular lymphoma, chronic myelogenous leukemia (CML), acute myeloid leukemia, acute lymphocytic leukemia, mixed lineage leukemia, or myelodysplasia syndromes (MDS). In one embodiment the EZH2-mediated cancer may be a malignant rhabdoid tumor or INIl-defecient tumor. The histologic diagnosis of malignant rhabdoid tumor depends on identification of characteristic rhabdoid cells (large cells with eccentrically located nuclei and abundant, eosinophilic cytoplasm) and

immunohistochemistry with antibodies to vimentin, keratin and epithelial membrane antigen. In most malignant rhabdoid tumors, the SMARCB1/INI1 gene, located in chromosome band 22ql 1.2, is inactivated by deletions and/or mutations. In one

embodiment, the malignant rhabdoid tumors may be INIl-defecient tumor. [034] Unless otherwise stated, any description of a method of treatment includes uses of the compounds to provide such treatment or prophylaxis as is described in the specification, as well as uses of the compounds to prepare a medicament to treat or prevent such condition. The treatment includes treatment of human or non-human animals including rodents and other disease models. Methods described herein may be used to identify suitable candidates for treating or preventing EZH2-mediated disorders. For example, the invention also provides methods of identifying an inhibitor of a wild-type EZH2, a mutant EZH2 (e.g., a Y641, A677, and/or A687 mutant EZH2), or both.

[035] For example, the method comprises the step of administering to a subject having a cancer with aberrant H3-K27 methylation an effective amount of one or more compounds of Formulae described herein, wherein the compound(s) inhibits histone methyltransferase activity of EZH2, thereby treating the cancer. Examples of aberrant H3-K27 methylation may include a global increase in and/or altered distribution of H3-K27 di or tri-methylation within the cancer cell chromatin.

[036] For example, the cancer is selected from the group consisting of cancers that overexpress EZH2 or other PRC2 subunits, contain loss-of-function mutations in H3-K27 demethylases such as UTX, or overexpress accessory proteins such as PHF19/PCL3 capable of increasing and or mislocalizing EZH2 activity (see references in Sneeringer et al. Proc Natl Acad Sci USA 107(49):20980-5, 2010).

[037] For example, the method comprises the step of administering to a subject having a cancer overexpressing EZH2 a therapeutically effective amount of one or more compounds of Formulae described herein, wherein the compound(s) inhibits histone methyltransferase activity of EZH2, thereby treating the cancer.

[038] For example, the method comprises the step of administering to a subject having a cancer with a loss-of-function mutation in the H3-K27 demethylase UTX a therapeutically effective amount of one or more compounds of Formulae described herein, wherein the compound(s) inhibits histone methyltransferase activity of EZH2, thereby treating the cancer

[039] For example, the method comprises the step of administering to a subject having a cancer overexpressing an accessory component(s) of the PRC2, such as PHF19/PCL3, a therapeutically effective amount of one or more compounds of Formulae described herein, wherein the compound(s) inhibits histone methyltransferase activity of EZH2, thereby treating the cancer [040] In still another aspect, this invention relates to a method of modulating the activity of the wild-type EZH2, the catalytic subunit of the PRC2 complex which catalyzes the mono- through tri-methylation of lysine 27 on histone H3 (H3-K27). For example, the present invention relates to a method of inhibiting the activity of EZH2 in a cell. This method can be conducted either in vitro or in vivo.

[041] In yet another aspect, this invention features to a method of inhibiting in a subject conversion of H3-K27 to trimethylated H3-K27. The method comprises administering to a subject a therapeutically effective amount of one or more of the compounds of Formulae described herein to inhibit histone methyltransferase activity of EZH2, thereby inhibiting conversion of H3-K27 to trimethylated H3-K27 in the subject.

[042] For example, the method comprises the step of administering to a subject having a cancer expressing a mutant EZH2 a therapeutically effective amount of one or more compounds of Formulae described herein, wherein the compound(s) inhibits histone methyltransferase activity of EZH2, thereby treating the cancer.

[043] For example, the cancer is selected from the group consisting of follicular lymphoma and diffuse large B-cell lymphoma (DLBCL) of germinal center B cell-like (GCB) subtype. For example, the cancer is lymphoma, leukemia or melanoma. Preferably, the lymphoma is non-Hodgkin's lymphoma (NHL), follicular lymphoma or diffuse large B- cell lymphoma. Alternatively, the leukemia is chronic myelogenous leukemia (CML), acute myeloid leukemia, acute lymphocytic leukemia or mixed lineage leukemia.

[044] For example, the precancerous condition is myelodysplasia syndromes (MDS, formerly known as preleukemia).

[045] For example, the cancer is a hematological cancer.

[046] For example, the cancer is selected from the group consisting of brain and central nervous system (CNS) cancer, head and neck cancer, kidney cancer, ovarian cancer, pancreatic cancer, leukemia, lung cancer, lymphoma, myeloma, sarcoma, breast cancer, and prostate cancer. Preferably, a subject in need thereof is one who had, is having or is predisposed to developing brain and CNS cancer, kidney cancer, ovarian cancer, pancreatic cancer, leukemia, lymphoma, myeloma, and/or sarcoma. Exemplary brain and central CNS cancer includes medulloblastoma, oligodendroglioma, atypical teratoid/rhabdoid tumor, choroid plexus carcinoma, choroid plexus papilloma, ependymoma, glioblastoma, meningioma, neuroglial tumor, oligoastrocytoma, oligodendroglioma, and pineoblastoma. Exemplary ovarian cancer includes ovarian clear cell adenocarcinoma, ovarian

endomethrioid adenocarcinoma, and ovarian serous adenocarcinoma. Exemplary pancreatic cancer includes pancreatic ductal adenocarcinoma and pancreatic endocrine tumor.

Exemplary sarcoma includes chondrosarcoma, clear cell sarcoma of soft tissue, ewing sarcoma, gastrointestinal stromal tumor, osteosarcoma, rhabdomyosarcoma, and not otherwise specified (NOS) sarcoma. Alternatively, cancers to be treated by the compounds of the present invention are non NHL cancers.

[047] For example, the cancer is selected from the group consisting of medulloblastoma, oligodendroglioma, ovarian clear cell adenocarcinoma, ovarian endomethrioid

adenocarcinoma, ovarian serous adenocarcinoma, pancreatic ductal adenocarcinoma, pancreatic endocrine tumor, malignant rhabdoid tumor, astrocytoma, atypical

teratoid/rhabdoid tumor, choroid plexus carcinoma, choroid plexus papilloma,

ependymoma, glioblastoma, meningioma, neuroglial tumor, oligoastrocytoma,

oligodendroglioma, pineoblastoma, carcinosarcoma, chordoma, extragonadal germ cell tumor, extrarenal rhabdoid tumor, schwannoma, skin squamous cell carcinoma,

chondrosarcoma, clear cell sarcoma of soft tissue, ewing sarcoma, gastrointestinal stromal tumor, osteosarcoma, rhabdomyosarcoma, and not otherwise specified (NOS) sarcoma. Preferably, the cancer is medulloblastoma, ovarian clear cell adenocarcinoma, ovarian endomethrioid adenocarcinoma, pancreatic ductal adenocarcinoma, malignant rhabdoid tumor, atypical teratoid/rhabdoid tumor, choroid plexus carcinoma, choroid plexus papilloma, glioblastoma, meningioma, pineoblastoma, carcinosarcoma, extrarenal rhabdoid tumor, schwannoma, skin squamous cell carcinoma, chondrosarcoma, ewing sarcoma, epithelioid sarcoma, renal medullary carcinoma, diffuse large B-cell lymphoma, follicular lymphoma and/or NOS sarcoma. More preferably, the cancer is malignant rhabdoid tumor, medulloblastoma and/or atypical teratoid/rhabdoid tumor.

[048] For example, the method comprises the step of administering to a subject having a cancer expressing a mutant EZH2 a therapeutically effective amount of one or more compounds of Formulae described herein, wherein the compound(s) inhibits activity (e.g., histone methyltransferase activity) of the mutant EZH2, the wild-type EZH2, or both, thereby treating the cancer.

[049] For example, the method further comprises the steps of performing an assay to detect a mutant EZH2 in a sample comprising cancer cells from a subject in need thereof.

[050] In another aspect, the invention features a method of selecting a therapy for a patient having a disease associated with EZH2-mediated protein methylation. The method includes the steps of determining the presence of gene mutation in the EZH2 gene of the subject; and selecting, based on the presence of a gene mutation in the EZH2 gene a therapy for treating the disease. In one embodiment, the therapy includes the administration of one or more of the compounds of the invention. In one embodiment, the method further includes administrating one or more of the compounds of the invention to the subject. In one embodiment, the disease is cancer and the mutation is a Y641 mutation.

[051] In yet another aspect, a method of treatment is provided for a patient in need thereof, the method comprising the steps of determining the presence of gene mutation in the EZH2 gene and treating the patient in need thereof, based on the presence of a gene mutation in the EZH2 gene, with a therapy that includes the administration of the compounds of the invention. In one embodiment, the patient is a cancer patient and the mutation is a Y641 mutation.

[052] In still another aspect, this invention relates to a method of modulating the activity of the wild-type and mutant histone methyltransferase EZH2, the catalytic subunit of the PRC2 complex which catalyzes the mono- through tri-methylation of lysine 27 on histone H3 (H3-K27). For example, the present invention relates to a method of inhibiting the activity of certain mutant forms of EZH2 in a cell. The mutant forms of EZH2 include a substitution of another amino acid residue for tyrosine 641 (Y641, also Tyr641) of wild- type EZH2. The method includes contacting the cell with an effective amount of one or more of the compounds of any Formula described herein. This method can be conducted either in vitro or in vivo.

[053] In yet another aspect, this invention features to a method of inhibiting in a subject conversion of H3-K27 to trimethylated H3-K27. The method comprises administering to a subject expressing a mutant EZH2 a therapeutically effective amount of one or more of the compounds of any Formula described herein to inhibit histone methyltransferase activity of EZH2, thereby inhibiting conversion of H3-K27 to trimethylated H3-K27 in the subject. For example, the histone methyltransferase activity inhibited is that of the Y641 mutant of EZH2. For example, the compound of this invention selectively inhibits histone

methyltransferase activity of the Y641 mutant of EZH2. For example, the Y641 mutant of EZH2 is selected from the group consisting of Y641C, Y641F, Y641H, Y641N, and Y641S.

[054] The method of inhibiting in a subject conversion of H3-K27 to trimethylated H3- K27 may also comprise performing an assay to detect a mutant EZH2 in a sample from a subject before administering to the subject expressing a mutant EZH2 a therapeutically effective amount of one or more of the compounds of any Formula described herein. For example, performing the assay to detect the mutant EZH2 includes whole-genome resequencing or target region resequencing that detects a nucleic acid encoding the mutant EZH2. For example, performing the assay to detect the mutant EZH2 includes contacting the sample with an antibody that binds specifically to a polypeptide or fragment thereof characteristic of the mutant EZH2. For example, performing the assay to detect the mutant EZH2 includes contacting the sample under highly stringent conditions with a nucleic acid probe that hybridizes to a nucleic acid encoding a polypeptide or fragment thereof characteristic of the mutant EZH2.

[055] Further, the invention also relates to a method of identifying an inhibitor of a mutant EZH2, wild-type EZH2, or both. The method comprises the steps of combining an isolated EZH2 with a histone substrate, a methyl group donor, and a test compound, wherein the histone substrate comprises a form of H3-K27 selected from the group consisting of unmethylated H3-K27, monomethylated H3-K27, dimethylated H3-K27, and any combination thereof; and performing an assay to detect methylation of H3-K27 (e.g., formation of trimethylated H3-K27) in the histone substrate, thereby identifying the test compound as an inhibitor of the EZH2 when methylation of H3-K27 (e.g., formation of trimethylated H3-K27) in the presence of the test compound is less than methylation of H3- K27 (e.g., formation of trimethylated H3-K27) in the absence of the test compound.

[056] In one embodiment, performing the assay to detect methylation of H3-K27 in the histone substrate comprises measuring incorporation of labeled methyl groups.

[057] In one embodiment, the labeled methyl groups are isotopically labeled methyl groups.

[058] In one embodiment, performing the assay to detect methylation of H3-K27 in the histone substrate comprises contacting the histone substrate with an antibody that binds specifically to trimethylated H3-K27.

[059] Also within the scope of the invention is a method of identifying a selective inhibitor of a mutant EZH2. The method comprises the steps of combining an isolated mutant EZH2 with a histone substrate, a methyl group donor, and a test compound, wherein the histone substrate comprises a form of H3-K27 selected from the group consisting of monomethylated H3-K27, dimethylated H3-K27, and a combination of monomethylated H3-K27 and dimethylated H3-K27, thereby forming a test mixture; combining an isolated wild-type EZH2 with a histone substrate, a methyl group donor, and a test compound, wherein the histone substrate comprises a form of H3-K27 selected from the group consisting of monomethylated H3-K27, dimethylated H3-K27, and a combination of monomethylated H3-K27 and dimethylated H3-K27, thereby forming a control mixture; performing an assay to detect trimethylation of the histone substrate in each of the test mixture and the control mixture; calculating the ratio of (a) trimethylation with the mutant EZH2 and the test compound (M+) to (b) trimethylation with the mutant EZH2 without the test compound (M-); calculating the ratio of (c) trimethylation with wild-type EZH2 and the test compound (WT+) to (d) trimethylation with wild-type EZH2 without the test compound (WT-); comparing the ratio (a)/(b) with the ratio (c)/(d); and identifying the test compound as a selective inhibitor of the mutant EZH2 when the ratio (a)/(b) is less than the ratio (c)/(d).

[060] The present invention further provides a method of identifying a subject as a candidate for treatment with one or more compounds of the invention. The method comprises the steps of performing an assay to detect a mutant EZH2 in a sample from a subject; and identifying a subject expressing a mutant EZH2 as a candidate for treatment with one or more compounds of the invention, wherein the compound(s) inhibits histone methyltransferase activity of EZH2.

[061] Still another aspect of the invention is a method of inhibiting conversion of H3-K27 to trimethylated H3-K27. The method comprises the step of contacting wild- type EZH2, a mutant EZH2, or both with a histone substrate comprising H3-K27 and an effective amount of a compound of the present invention, wherein the compound inhibits histone

methyltransferase activity of EZH2, thereby inhibiting conversion of H3-K27 to

trimethylated H3-K27.

[062] Further, the compounds or methods described herein can be used for research (e.g., studying epigenetic enzymes) and other non-therapeutic purposes.

[063] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In the specification, the singular forms also include the plural unless the context clearly dictates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents and other references mentioned herein are incorporated by reference. The references cited herein are not admitted to be prior art to the claimed invention. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods and examples are illustrative only and are not intended to be limiting. In the case of conflict between the chemical structures and names of the compounds disclosed herein, the chemical structures will control. [064] Other features and advantages of the invention will be apparent from the following detailed description and claims.

DETAILED DESCRIPTION OF THE INVENTION

[065] The present invention provides novel 1,4-pyridone compounds, synthetic methods for making the compounds, pharmaceutical compositions containing them and various uses of the compounds.

[066] The present invention rovides the compounds of Formula (I):

In this formula:

Z is NR 7 R 8 , OR 7 , S(0) a R 7 , or CR 7 R 8 R 14 , in which a is 0, 1, or 2;

each of R 1 ; R5, R 9 , and R 10 , independently, is H or Ci-C 6 alkyl optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, C C 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Cr C alkylamino, C 3 -C8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

each of R 2 , R3, and R 4 , independently, is -Qj-Ti, in which Qj is a bond or C C 3 alkyl linker optionally substituted with halo, cyano, hydroxyl or C C 6 alkoxy, and T is H, halo, hydroxyl, C(0)OH, cyano, azido, or R sl , in which R sl is C C 3 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, Ci-C 6 alkoxyl, Ci-C 6 thioalkyl, C(0)0-Ci-C 6 alkyl, CONH 2 , S0 2 NH 2 , -CO- NH(Ci-C 6 alkyl), -CO-N(Ci-C 6 alkyl) 2 , -S0 2 -NH(Ci-C 6 alkyl), - S0 2 -N(Ci-C 6 alkyl) 2 , C 3 - C8 cycloalkyl, C 6 -C 10 aryl, C 6 -C 10 aryloxy, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and R s l is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Cr C alkylamino, di-Ci-C 6 alkylamino, C3-C 8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl; or Ri and R 2 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or Ri and R 4 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R 3 and R 4 , together with the C atoms to which they are attached, form C5-C 8 cycloalkyl, C 6 -C 10 aryl, or a 5- or 6- membered heteroaryl having 1 to 3 heteroatoms, or a 5 to 12-membered heterocycloalkyl ring having 1 to 3 heteroatoms; in which each of the ring structures formed by Ri and R 2 , by Ri and R 4 , or by R 3 and R 4 , independently is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, CrC 6 alkyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Cr C alkylamino, C 3 -Cg cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R 6 is H, halo, cyano, azido, OR a , -NR a R b , -C(0)R a , -C(0)OR a , -C(0)NR a R b , -NR b C(0)R a , -S(0) b R a , -S(0) b NR a Rb, or R S2 , in which R S2 is Ci-Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Cg cycloalkyl, C 6 -Cio aryl, 5- or 6-membered heteroaryl, or 4 to 12- membered heterocycloalkyl, b is 0, 1, or 2, each of R a and R , independently is H or R S , and Rs 3 is CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 6 -Cio aryl, 4 to 12- membered heterocycloalkyl, or 5- or 6-membered heteroaryl; or R a and R b , together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 or 1 additional heteroatom; and each of R S2 , Rs 3 , and the 4 to 12-membered heterocycloalkyl ring formed by R a and R b , is optionally substituted with one or more -Q 2 - T 2 , wherein Q 2 is a bond or CrC 3 alkyl linker each optionally substituted with halo, cyano, hydroxyl or Ci-C 6 alkoxy, and T 2 is H, halo, cyano, -OR c , -NR c R d , -C(0)R c , -C(0)OR c , - C(0)NR c R d , -NR d C(0)R c , -NR d C(0)OR c , -S(0) 2 R c , -S(0) 2 NR c R d , or R S4 , in which each of R c and R d , independently is H or Rss, each of Rs 4 and Rss, independently, is CrC 6 alkyl, C 3 -C 8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, or R c and R d , together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 or 1 additional heteroatom, and each of R S4 , Rss, and the 4 to 12-membered heterocycloalkyl ring formed by R c and R d , is optionally substituted with one or more -Q3-T3, wherein Q 3 is a bond or CrC 3 alkyl linker each optionally substituted with halo, cyano, hydroxyl or C C 6 alkoxy, and T 3 is selected from the group consisting of halo, cyano, Ci-C 6 alkyl, C 3 -Cg cycloalkyl, C 6 -Cio aryl, 4 to 12- membered heterocycloalkyl, 5- or 6-membered heteroaryl, OR e , C(0)OR e , -S(0) 2 R e , - NR e R f , and -C(0)NR e R f , each of R e and R f independently being H or C C 6 alkyl optionally substituted with OH, 0-C C 6 alkyl, or NH-C C 6 alkyl, or -Q 3 -T 3 is oxo; or-Q 2 -T 2 is oxo; or any two neighboring -Q 2 -T 2 , when R 6 is C 6 -Cio aryl or 5- or 6-membered heteroaryl, together with the atoms to which they are attached form a 5- or 6-membered ring optionally containing 1-4 heteroatoms selected from N, O and S and optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0- C C 6 alkyl, cyano, C C 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-C C 6 alkylamino, C 3 -Cg cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl;

R 7 is -Q4-T4, in which Q 4 is a bond, CrC 4 alkyl linker, or C 2 -C 4 alkenyl linker, each linker optionally substituted with halo, cyano, hydroxyl or Q-C 6 alkoxy, and T 4 is H, halo, cyano, NR g R h , -OR g , -C(0)R g , -C(0)OR g , -C(0)NR g R h , -C(0)NR g OR h , -NR g C(0)R h , -S(0) 2 R g , or R S 6, in which each of R g and R h , independently is H or R S7 , each of R s6 and Rs 7 , independently is Q-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 14-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and each of Rs 6 and R S7 is optionally substituted with one or more -Q5-T5, wherein (¾ is a bond, C(O), C(0)NR k , NR k C(0), NR k , S(0) 2 , NR k S(0) 2 , or Ci-C 3 alkyl linker, R k being H or Ci-C 6 alkyl, and T5 is H, halo, CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, hydroxyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, C 3 -C 8 cycloalkyl, CrC 6 alkylene-C 3 -Cg cycloalkyl, C 6 -C 10 aryl, C -C alkylene-Ce-Qo aryl, 4 to 12-membered heterocycloalkyl, C -C alkylene-4 to 12-membered heterocycloalkyl, 5- or 6-membered heteroaryl, CrC 6 alkylene-5- or 6-membered heteroaryl, or S(0) q R q in which q is 0, 1, or 2 and R q is CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12- membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and T5 is optionally substituted with one or more substituents selected from the group consisting of halo, C -C alkyl, hydroxyl, cyano, CrC 6 alkoxyl, 0-CrC 4 alkylene-C 1 -C 4 alkoxy, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, C 3 -C 8 cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl except when T5 is H, halo, hydroxyl, or cyano; or -Q5-T5 is oxo; each of Rg, and R 12 , independently, is H, halo, hydroxyl, C(0)OH, cyano, Rs 8 , ORs 8 , or C(0)OR S8 , in which R S8 is CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, 4 to 12-membered heterocycloalkyl, amino, mono-C Ce alkylamino, or di-C^Ce alkylamino, and R S8 is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, and di-C C 6 alkylamino; or R 7 and Rg, together with the N atom to which they are attached, form a 4 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms, or R 7 and R 8 , together with the atom to which they are attached, form C 3 -C8 cycloalkyl or a 4 to 12-membered heterocycloalkyl ring having 1 to 3 heteroatoms, and each of the 4 to 12-membered heterocycloalkyl rings or C 3 -C8 cycloalkyl formed by R 7 and Rg is optionally substituted with one or more -Q6-T 6 , wherein Q 6 is a bond, C(O), C(0)NR m , NR m C(0), S(0) 2 , or C 1 -C3 alkyl linker, R m being H or Ci-C 6 alkyl, and T 6 is H, halo, C -C alkyl, hydroxyl, cyano, C -C alkoxyl, amino, mono-Q-Ce alkylamino, di-C C 6 alkylamino, C 3 -Cg cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, 5- or 6-membered heteroaryl, or S(0) p R p in which p is 0, 1, or 2 and R p is C -C alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Cg cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, or 5- or 6-membered heteroaryl, and T 6 is optionally substituted with one or more substituents selected from the group consisting of halo, CrC 6 alkyl, hydroxyl, cyano, CrC 6 alkoxyl, amino, mono-Q-Ce alkylamino, di-CrC 6 alkylamino, C 3 -Cg cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl except when T 6 is H, halo, hydroxyl, or cyano; or -Q6-T 6 is oxo; and

R 14 is absent, H, or CrC 6 alkyl optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, C -C alkoxyl, amino, mono-C Ce alkylamino, di-Q-Ce alkylamino, C 3 -Cg cycloalkyl, C 6 - Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl.

[067] The compounds of Formula (I) can have one or more of the following features:

[068] For example, Z is NR 7 R 8 .

[069] For example, Z is CR 7 R 8 R 14 .

[070] For example, Z is OR 7 .

[071] For example, Z is S(0) a R 7 , in which a is 0, 1, or 2 .

[072] For example, Z is SR 7.

[073] For example, R 6 is unsubstituted C 6 -Cio aryl or unsubstituted 5- or 6-membered heteroaryl. [074] For example, R 6 is substituted C 6 -C 10 aryl or substituted 5- or 6-membered heteroaryl.

[075] For example, R 6 is C 6 -C 10 aryl substituted with one or more -Q 2 -T 2 or 5- or 6- membered heteroaryl substituted with one or more -Q 2 -T 2 .

[076] For example, R 6 is unsubstituted or substituted phenyl.

[077] For example, R 6 is phenyl substituted with one or more -Q 2 -T 2 .

[078] For example, R 6 is 5 to 6-membered heteroaryl containing 1-3 additional heteroatoms selected from N, O, and S and optionally substituted with one or more -Q 2 -T 2 .

[079] For example, R 6 is pyridinyl, pyrazolyl, pyrimidinyl, quinolinyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl, or thienyl, each of which is optionally substituted with one or more -Q 2 -T 2 .

[080] For example, R 6 is C 6 -Cio aryl or 5- or 6-membered heteroaryl, each of which is optionally, independently substituted with one or more -Q 2 -T 2 , wherein Q 2 is a bond or C - C 3 alkyl linker, and T 2 is H, halo, cyano, -OR c , -NR c R d , -C(0)NR c R d , -NR d C(0)R c , - S(0) 2 R c , -S(0) 2 NR c R d , or Rs 4 , in which each of R c and R d , independently is H or Rss, each of R S4 and R S s, independently, is C -C alkyl, or R c and R d , together with the N atom to which they are attached, form a 4 to 7-membered heterocycloalkyl ring having 0 or 1 additional heteroatom, and each of Rs 4 , Rss, and the 4 to 7-membered heterocycloalkyl ring formed by R c and R d , is optionally, independently substituted with one or more -Q3-T3, wherein Q is a bond or Q-C3 alkyl linker and T 3 is selected from the group consisting of halo, C -C alkyl, 4 to 7-membered heterocycloalkyl, OR e , -S(0) 2 R e , and -NR e R f , each of R e and R f independently being H or C C 6 alkyl optionally substituted with OH, 0-C C 6 alkyl, or NH-C C 6 alkyl, or -Q3-T3 is oxo; or any two neighboring -Q 2 -T 2 , together with the atoms to which they are attached form a 5- or 6-membered ring optionally containing 1- 4 heteroatoms selected from N, O and S.

[081] For example, R 6 is phenyl or 5- or 6-membered heteroaryl substituted with O-Ci-6 alkyl or NH-Ci-6 alkyl, each of which is optionally substituted hydroxyl, O-Ci-3 alkyl or NH-Ci-3 alkyl, each of the O-C^ alkyl and NH-Ci-3 alkyl being optionally further substituted with 0-C 1 3 alkyl or NH-Ci_ 3 alkyl.

[082]

[083] For example, R 6 is H.

[084] For example, R 6 is halo (e.g. , fluorine, chlorine, bromine, and iodine). [085] For example, R 6 is CI.

[086] For example, R 6 is Q-C 3 alkyl optionally substituted with one or more -Q 2 -T 2 .

[087] For example, R 6 is CF 3 .

[088] For example, R 6 is C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, or C 3 -C 6 cycloalkyl each optionally substituted with one or more -Q 2 -T 2 .

[089] For example, R 6 is ethenyl.

[090] For example, R 6 is ethynyl.

[091] For example, R 6 is ethynyl substituted with one or more -Q 2 -T 2 , in which Q 2 is a bond or Q-C 3 alkyl linker and T 2 is CrC 6 alkyl, C 3 -C 6 cycloalkyl, or 4 to 7-membered heterocyclo alkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5- azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, and morpholinyl, and the like) optionally substituted with one or more -Q3-T3.

[092] For example, R 6 is azido.

[093] For example, R 6 is cyano.

[094] For example, R 6 is C(0)H.

[095] For example, R 6 is OR a or -C(0)R a .

[096] For example, R a is C -C alkyl or 4 to 7-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5- diazabicyclo[2.2. l]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like), which is optionally substituted with one or more -Q 2 -T 2 .

[097] For example, R 6 is 4 to 7-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa- 6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like) optionally substituted with one or more -Q 2 -T 2 . [098] For example, R 6 is piperidinyl, 2,2,6,6-tetramethyl-piperidinyl, 1,2,3,6- tetrahydropyridinyl, 2,2,6,6-tetramethyl- 1 ,2,3,6-tetrahydropyridinyl, piperazinyl, morpholinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, or pyrrolidinyl, each of which is optionally substituted with one or more -Q 2 -T 2 .

[099] For example, R 6 is 4 to 7-membered heterocycloalkyl optionally substituted with one or more -Q 2 -T 2 , and -Q 2 -T 2 is oxo or Q 2 is a bond and T 2 is -OR c , -NR c R d , -C(0)R c , -C(0)OR c , -S(0) 2 R c , C -C alkyl, or 4 to 7-membered heterocycloalkyl, each of which is optionally substituted with one or more -Q3-T3 when R c or Ra is not H.

[0100] For example, R 6 is -NR a R b , -C(0)R a , -C(0)OR a , -C(0)NR a R b , -NR b C(0)R a , -SR a , - S(0) 2 R a , or -S(0) 2 NR a R b .

[0101] For example, each of R a and R b , independently is H, CrC 6 alkyl or C 3 -C8 cycloalkyl optionally substituted with one or more -Q 2 -T 2

[0102] For example, one of R a and R b is H.

[0103] For example, R a and R b , together with the N atom to which they are attached, form a 4 to 7-membered heterocycloalkyl ring having 0 or 1 additional heteroatoms to the N atom (e.g., azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, 1,4-diazepanyl, 1,4- oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa-6- azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like) and the ring is optionally substituted with one or more -Q 2 -T 2

[0104] For example, -Q 2 -T 2 is not H.

[0105] For example, -Q 2 -T 2 is oxo.

[0106] For example, Q 2 is a bond.

[0107] For example, Q 2 is an unsubstituted Q-C 3 alkyl linker.

[0108] For example, T 2 is C -C alkyl or C6-Cio aryl, each optionally substituted with one

[0109] For example, T 2 is an unsubstituted substituted straight chain CrC 6 or branched C3- C alkyl, including but not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, s-pentyl and n-hexyl.

[0110] For example, T 2 is phenyl.

[0111] For example, T 2 is halo (e.g. , fluorine, chlorine, bromine, and iodine).

[0112] For example, T 2 is 4 to 7-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa- 6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like) optionally substituted with one or more -Q 3 -T3.

[0113] For example, T 2 is -OR c , -NR c R d , -C(0)R c , -C(0)OR c , or -S(0) 2 R c .

[0114] For example, R c is CrC 6 alkyl or 4 to 7-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5- diazabicyclo[2.2. l]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like), which is optionally substituted with one or more -Q 3 -T3.

[0115] For example, each of R c and R d , independently is H or C -C alkyl optionally substituted with one or more -Q 3 -T3.

[0116] For example, R c is H.

[0117] For example, R d is H.

[0118] For example, R c and R d , together with the N atom to which they are attached, form a 4 to 7-membered heterocycloalkyl ring having 0 or 1 additional heteroatoms to the N atom (e.g., azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, 1,4- diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5- diazabicyclo[2.2. l]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like) and the ring is optionally substituted with one or more -Q 3 - T 3 .

[0119] For example, Q 2 is a bond and T 2 is -OR c , -NR c R d , -C(0)R c , -C(0)OR c , -S(0) 2 R c , CrC 6 alkyl, or 4 to 7-membered heterocycloalkyl, each of which is optionally substituted with one or more -Q 3 -T3 when R c or R d is not H.

[0120] For example,-Q 3 -T 3 is oxo.

[0121] For example, T 2 is 4 to 7-membered heterocycloalkyl or C3-C8 cycloalkyl and one

[0122] For example, (¾ is a bond or unsubstituted or substituted Q-C 3 alkyl linker.

[0123] For example, T 3 is H, halo, 4 to 7-membered heterocycloalkyl, Q-C 3 alkyl, OR e , C(0)ORe,-S(0) 2 R e ,-NR e Rf, or -C(0)NR e R f .

[0124] For example, one of R d and R e is H. [0125] For example, Q 3 is a bond or CrC 3 alkyl linker and T 3 is selected from the group consisting of C C 3 alkyl, halo, OR e , -S(0) 2 R e , -NR e Rf, and-C(0)NR e R f .

[0126] For example, Q is a bond or C C 3 alkyl linker and T is selected from the group consisting of C C 3 alkyl, OR e , -S(0) 2 R e , or -NR e R f .

[0127] For example, R e is H.

[0128] For example, R f is H.

[0129] For example, R 7 is not H.

[0130] For example, R 7 is -C(0)R g .

[0131] For example, R 7 is -C(0)R g , in which R g is C 3 -C 8 cycloalkyl, or 4 to 7-membered heterocycloalkyl, C 3 -C 8 cycloalkyl.

[0132] For example, R 7 is C 6 -C 10 aryl substituted with one or more -Q 5 -T5.

[0133] For example, R 7 is phenyl optionally substituted with one or more -Q 5 -T5.

[0134] For example, R 7 is Q-C 6 alkyl optionally substituted with one or more -Q 5 -T5.

[0135] For example, R 7 is C 3 -C 8 cycloalkyl optionally substituted with one or more

[0136] For example, R 7 is 4 to 7-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa- 6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like) optionally substituted with one or more

[0137] For example, R 7 is 8 to 14-membered heterocycloalkyl such as 1,4- dioxaspiro[4.5]decanyl (e.g., l,4-dioxaspiro[4.5]decan-8-yl), l,4-dioxa-8- azaspiro[4.5]decanyl (e.g., l,4-dioxa-8-azaspiro[4.5]decan-8-yl), l-oxaspiro[4.5]decanyl (e.g., l-oxaspiro[4.5]decan-8-yl or l-oxaspiro[4.5]decan-2-one-8-yl), 1- azaspiro[4.5]decanyl (e.g., l-azaspiro[4.5]decan-8-yl or l-azaspiro[4.5]decan-2-one-8-yl), 3'H-spiro[cyclohexane-l,l'-isobenzofuran]-yl (e.g., 3'H-spiro[cyclohexane-l,l'- isobenzofuran]-4-yl or 3'H-spiro[cyclohexane-l, -isobenzofuran]-3'-one-4-yl), 7Ή- spiro[cyclohexane-l,5'-furo[3,4-b]pyridin]-yl (e.g., 7'H-spiro[cyclohexane-l,5'-furo[3,4- b]pyridin]-4-yl or 7'H-spiro[cyclohexane-l,5'-furo[3,4-b]pyridin]-7'-one-4-yl), or 3Ή- spiro [cyclohexane- 1 , 1 '-furo [3 ,4-c]pyridin] -yl (e.g . , 3Ή- spiro [cyclohexane- 1 , 1 '-furo [3 ,4- c]pyridin]-4-yl or 3'H-spiiO[cyclohexane- l,r-furo[3,4-c]pyridin]-3'-one-4-yl), each optionally substituted with one or more -Q5-T5.

[0138] For example, R 7 is 5 to 6-membered heterocycloalkyl optionally substituted with

[0139] For example, R 7 is isopropyl.

[0140] For example, R 7 is piperidinyl, tetrahydropyran, tetrahydro-2H-thiopyranyl, piperazinyl, cyclopentyl, cyclohexyl, pyrrolidinyl, or cycloheptyl, each optionally substituted with one or more -Q 5 -T 5 .

[0141] For example, R 7 is piperidinyl, tetrahydropyran, cyclopentyl, or cyclohexyl, each optionally substituted with one -Q5-T5 and ethyl.

[0142] For example, R 7 is tetrahydropyran

[0143] For example, R 7 is

[0144] For example, R 7 is

[0145] For example, R 7 is

[0146] For example, R 7 is [0147] For example, R 7 is

[0148] For example, R 7 is Rioo is phenyl, 5- or 6-membered heteroaryl, or 4 to 12-membered heterocycloalkyl, each optionally substituted with one or more Ts a in which each Ts a is independently C -C alkoxyl or O-C G* alkylene-C 1 -C 4 alkoxy, and Rioi is H or CrC 4 alkyl.

, wherein each T 5a is independently C C ? alkoxyl or 0-C C 3 alkylene-Cr^ alkoxy.

[0150] For example, R 7 is cyclopentyl or cyclohexyl, each optionally substituted with one

[0151] For example, Q 5 is NHC(O) and T 5 is Ci-C 6 alkyl or Ci-C 6 alkoxy.

[0152] For example, -Q5-T5 is oxo.

[0153] For example, T 4 is 4 to 7-membered heterocycloalkyl or C 3 -C 8 cycloalkyl or C 6 -C 10 aryl, and one or more -Q5-T5 are oxo.

[0154] For example, R 7 is l-oxide-tetrahydro-2H-thiopyranyl or 1, 1-dioxide-tetrahydro- 2H-thiopyranyl.

[0155] For example, R 7 is cyclohexanonyl, e.g., cyclohexanon-4-yl.

[0156] For example, T5 is H, halo, C -C alkyl, C -C alkoxyl, C 3 -C 8 cycloalkyl, C 6 -Cio aryl, or 4 to 7-membered heterocycloalkyl.

[0157] For example, Q5 is a bond and T5 is CrC 6 alkyl, C 3 -C 8 cycloalkyl, or 4 to 7- membered heterocycloalkyl.

[0158] For example, Q5 is a bond and T5 is 5- or 6-membered heteroaryl, amino, mono-Q- C alkylamino, di-C^Ce alkylamino, T5 being optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C -C alkoxyl, or C 3 -C 8 cycloalkyl.

[0159] For example, Q 5 is CO, S(0) 2 , or NHC(O); and T 5 is Ci-Ce alkyl, Ci-Ce alkoxyl, C 3 -C 8 cycloalkyl, or 4 to 7-membered heterocycloalkyl.

[0160] For example, T5 is C -C alkyl or C -C alkoxyl, each optionally substituted with halo, hydroxyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, or C 3 -C 8 cycloalkyl.

[0161] For example, (¾ is Q-C 3 alkyl linker and T5 is H or C 6 -C 10 aryl.

[0162] For example, (¾ is Q-C 3 alkyl linker and T5 is C 3 -C 8 cycloalkyl, 4 to 7-membered heterocycloalkyl, or S(0) q R q .

[0163] For example, R 6 is halo (e.g. , fluorine, chlorine, bromine, and iodine) and Z is S(0) a R 7 , in which a is 0, 1, or 2 and R 7 is C -C alkyl (e.g., methyl, ethyl, n-propyl, i- propyl, butyl, or t-butyl), C 3 -C8 cycloalkyl (e.g., cyclopentyl, cyclohexyl, or cycloheptyl) or 4 to 14-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro- 2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5- azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, morpholinyl, 1,4- dioxaspiro[4.5]decanyl, l,4-dioxa-8-azaspiro[4.5]decanyl, l-oxaspiro[4.5]decanyl, 1- azaspiro[4.5]decanyl, 3'H-spiro[cyclohexane-l, l'-isobenzofuran]-yl, 7Ή- spiro[cyclohexane-l,5'-furo[3,4-b]pyridin]-yl, or 3'H-spiro[cyclohexane-l, l'-furo[3,4- c]pyridin]-yl, and the like) and R 7 is optionally substituted with one or more -Q5-T5.

[0164] For example, R 6 is halo (e.g. , fluorine, chlorine, bromine, and iodine) and Z is OR 7 in which R 7 is 4 to 14-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H- pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4- oxazepanyl, 2-oxa-5-azabicyclo[2.2. l]heptanyl, 2,5-diazabicyclo[2.2. l]heptanyl, morpholinyl, l,4-dioxaspiro[4.5]decanyl, l,4-dioxa-8-azaspiro[4.5]decanyl, 1- oxaspiro[4.5]decanyl, l-azaspiro[4.5]decanyl, 3'H-spiro[cyclohexane-l, l'-isobenzofuran]- yl, 7'H-spiro[cyclohexane-l,5'-furo[3,4-b]pyridin]-yl, or 3'H-spiro[cyclohexane-l, l'- furo[3,4-c]pyridin]-yl, and the like) and R 7 is optionally substituted with one or more -Q5- T 5 .

[0165] For example, each of R 2 and R 4 , independently, is H, halo, or CrC 6 alkyl optionally substituted with amino, azido, halo, mono-Ci-C 6 alkylamino, di-C C 6 alkylamino, or C 6 - C10 aryl.

[0166] For example, each of R 2 and R 4 , independently is Q-C 3 alkyl optionally substituted withC C6 alkoxyl.

[0167] For example, each of R 2 and R 4 is methyl.

[0168] For example, each of R 2 and R 4 , independently is halo, e.g., F, CI, or Br.

[0169] For example, each of R 2 and R 4 , independently, is CN, mono-Q-Ce alkylamino, or di-C C6 alkylamino.

[0170] For example, each of R 2 and R 4 , independently, is optionally substituted phenyl.

[0171] For example, each of R 2 and R 4 , independently, is optionally substituted 5- or 6- membered heteroaryl (e.g., pyrrolyl, pyrazolyl, imidazoly, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, and the like). [0172] For example, each of R 2 and R 4 , independently, is optionally substituted 4 to 12- membered heterocycloalkyl (e.g., pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, 1,4- diazepanyl, 1,4-oxazepanyl, and morpholinyl, and the like).

[0173] For example, each of R 2 and R 4 , independently, is C 1-6 alkoxyl or C 6 -C 10 aryloxy, each optionally substituted with one or more halo.

[0174] For example, R 2 is C 1-6 alkoxyl or C 6 -Cio aryloxy, each optionally substituted with one or more halo.

[0175] For example, R 4 is halo, or C 1-4 alkyl or C 1-6 alkoxyl, each optionally substituted with one or more halo.

[0176] For example, R 3 is H, halo, or C 1-4 alkyl;

[0177] For example, Ri is H or C 1-4 alkyl.

[0178] For example, Ri and R 2 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms (e.g., pyrrolyl, pyrazolyl, imidazoly, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, and the like), or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms (e.g., pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, 1,4- diazepanyl, 1,4-oxazepanyl, and morpholinyl, and the like). Further,each of the ring structures formed by Ri and R 2 mentioned above, independently, is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C - C 6 alkyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, Ci-C 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Ci-C 6 alkylamino, C 3 -C8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl.

[0179] For example, Ri and R 4 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms (e.g., pyrrolyl, pyrazolyl, imidazoly, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, and the like), or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms (e.g., pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, 1,4- diazepanyl, 1,4-oxazepanyl, and morpholinyl, and the like). Further,each of the ring structures formed by Ri and R 4 mentioned above, independently, is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C - C 6 alkyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, Ci-C 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Ci-C 6 alkylamino, C3-C8 cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl.

[0180] For example, R 3 and R 4 , together with the C atoms to which they are attached, form Cs-Cg cycloalkyl (e.g., C5-C 6 cycloalkyl), C 6 -Cio aryl (e.g., phenyl), or a 5- or 6-membered heteroaryl having 1 to 3 heteroatoms (e.g., pyrrolyl, pyrazolyl, imidazoly, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, and the like), or a 5 to 12-membered heterocycloalkyl ring having 1 to 3 heteroatoms(e.g., pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, 1,4-diazepanyl, 1,4-oxazepanyl, and morpholinyl, and the like). Further, each of the above-mentioned ring structures formed by R 3 and R 4 , independently, is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, C -C alkyl, C(0)OH, C(0)0-C 1 -C 6 alkyl, cyano, C -C alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Ci-C 6 alkylamino, C 3 -Cg cycloalkyl, C 6 -C 10 aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6-membered heteroaryl.

[0181] For example, Ri is H.

[0182] For example, Ri is Q-C 6 alkyl optionally substituted with azido, halo, amino, mono-Ci-C6 alkylamino, di-Ci-C6 alkylamino, or C6-Cio aryl.

[0183] For example, R 12 is H, methyl, ethyl, ethenyl, or halo.

[0184] For example, R 12 is methyl.

[0185] For example, R 12 is ethyl or propenyl.

[0186] For example, R 12 is methoxyl.

[0187] For example, R 12 is ethenyl.

[0188] For example, R 8 is H, methyl, ethyl, or ethenyl.

[0189] For example, R 8 is methyl.

[0190] For example, Rg is ethyl.

[0191] For example, Rg is propyl.

[0192] For example, Rg is ethenyl or propenyl.

[0193] For example, Rg is Q-C 6 alkyl substituted with one or more substituents selected from the group consisting of halo (e.g., F, CI, or Br), hydroxyl, or C C 6 alkoxyl.

[0194] For example, Rg is 4 to 7-membered optionally substituted heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5- diazabicyclo[2.2. l]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, and morpholinyl, and the like).

[0195] For example, R 8 is piperidinyl.

[0196] For example, Rg is 4 to 7-membered optionally substituted heterocycloalkyl and R 7 is

-Q4-T4, in which Q 4 is a bond or C C 4 alkyl linker and T 4 is H, Ci-C 6 alkyl, C3-C 8 cycloalkyl or 4 to 7-membered heterocycloalkyl.

[0197] For example, Z is NR 7 Rg or CR 7 RgR 14 wherein R 7 and Rg, together with the atom to which they are attached, form a 4 to 11-membered heterocycloalkyl ring having 1 to 3 heteroatoms (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6- tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5- azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, morpholinyl, l,4-dioxa-8-azaspiro[4.5]decanyl, and the like) or C3-C 8 cycloalkyl, each optionally substituted with one or more -(¾-Τ 6 .

[0198] For example, the ring formed by R 7 and Rg is selected from the group consisting of azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, l,4-dioxa-8- azaspiro[4.5]decanyl, and cyclohexenyl, each optionally substituted with one -Q6-T 6 .

[0199] For example, Z is l,4-dioxa-8-azaspiro[4.5]decan-8-yl, pyrrolidine-2,5-dione-l-yl, or piperidine-2,6-dione- l-yl.

[0200] For example, one or more -Q6-T 6 is oxo.

[0201] For example, T 6 is H, halo, Ci-C 6 alkyl, Ci-C 6 alkoxyl, C 3 -Cg cycloalkyl, C 6 -Cio aryl, or 4 to 7-membered heterocycloalkyl.

[0202] For example, Q 6 is a bond and T 6 is C C 6 alkyl, C 3 -Cg cycloalkyl, or 4 to 7- membered heterocycloalkyl.

[0203] For example, Q 6 is CO, S(0) 2 , or NHC(O); and T 6 is Ci-Ce alkyl, Ci-C 6 alkoxyl, C 3 -Cg cycloalkyl, or 4 to 7-membered heterocycloalkyl.

[0204] For example, T 6 is CrC 6 alkyl or CrC 6 alkoxyl, each optionally substituted with halo, hydroxyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-Q-Ce alkylamino, or C 3 -Cg cycloalkyl.

[0205] For example, Q 6 is Q-C3 alkyl linker and T 6 is H or C 6 -C 10 aryl. [0206] For example, Q 6 is Q-C 3 alkyl linker and T 6 is C 3 -C 8 cycloalkyl, 4 to 7-membered heterocycloalkyl, or S(0) p R p .

[0207] For example, each of R p and R q , independently, is C -C alkyl.

[0208] For example, R 6 is -S(0) b R a or azido, in which b is 0, 1, or 2 and R a is C -C alkyl or C 3 -C 8 cycloalkyl; and Z is NR 7 R 8 , in which R 7 is C 3 -C 8 cycloalkyl (e.g., cyclopentyl, cyclohexyl, or cycloheptyl) or 4 to 14-membered heterocycloalkyl (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, morpholinyl, l,4-dioxaspiro[4.5]decanyl, l,4-dioxa-8-azaspiro[4.5]decanyl, 1- oxaspiro[4.5]decanyl, l-azaspiro[4.5]decanyl, 3'H-spiro[cyclohexane-l, l'-isobenzofuran]- yl, 7'H-spiro[cyclohexane-l,5'-furo[3,4-b]pyridin]-yl, or 3'H-spiro[cyclohexane-l, l'- furo[3,4-c]pyridin]-yl, and the like), each optionally substituted with one or more -Q5-T5; and Rs is H or CrC 6 alkyl (e.g., methyl, ethyl, n-propyl, i-propyl, butyl, or t-butyl).

[0209] For example, R 6 is halo (e.g. , fluorine, chlorine, bromine, and iodine) and Z is NR 7 Rg or CR 7 RgR 14 wherein R 7 and R 8 , together with the atom to which they are attached, form a 4 to 11-membered heterocycloalkyl ring having 1 to 3 heteroatoms (e.g., azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, piperidinyl, 1,2,3,6-tetrahydropyridinyl, piperazinyl, tetrahydro-2H-pyranyl, 3,6-dihydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, 1,4-diazepanyl, 1,4-oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, morpholinyl, 2-oxa-6-azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, l,4-dioxa-8- azaspiro[4.5]decanyl, l,4-dioxa-8-azaspiro[4.5]decan-8-yl, pyrrolidine-2,5-dione- l-yl, piperidine-2,6-dione- 1-yl,

and the like) or C3-C8 cycloalkyl, each optionally substituted with one or more -Q6-T 6 .

[0210] For example, R3 is H.

[0211] For example, each of R5, R 9 , and R 10 is H.

[0212] For example, one subset of compounds of Formula (I) includes those of Formula (la)

or a pharmaceutically acceptable salt thereof, wherein Z, R 2 , R 4 , R 6 , and R 12 , are as deflined herein for Formula (I).

[0213] Another subset of the compounds of Formula (I) includes those of Formula (lb), (Ic),

(lb) (Ic) (Id)

or pharmaceutically acceptable salts thereof, wherein, R 2 , R 4 , R 5 , R 6 , R 7 , Rs, and R 12 are as defined herein for Formula (I).

[0214] Another subset of the compounds of Formula (I) includes those of Formula (II):

or pharmaceutically acceptable salts thereof, wherein Q 2 is a bond or methyl linker, T 2 is H, halo, -OR c , -NR c R d , or -S(0) 2 NR c R d and R c , R d , R 2 , R 7 , and R 8 are defined herein for Formula (I).

[0215] Another subset of the compounds of Formula (I) includes those of Formula (Ila):

or pharmaceutically acceptable salts thereof, wherein R c , R d , R 2 , R 7 , and Rg are defined herein for Formula (I).

[0216] Yet another subset of the compounds of Formula (I) includes those of Formula (lib):

or pharmaceutically acceptable salts thereof, wherein n is 0, 1, or 2; U is O, S, N-Q5-T5, or CH-Q5-T5; R 12 is CI, Br, or methyl; and R 6 , R 7 , Rg, Q5, and T5 are defined herein for Formula (I).

[0217] In addition to the above-described features of the compounds of this invention where applicable, the compounds of Formula (II), (Ila), or (lib) can include one or more of the following features: [0218] For example, Q 5 is a bond and T 5 is H, CrC 6 alkyl, C 3 -C 8 cycloalkyl, 4 to 12- membered heterocycloalkyl, 5- or 6-membered heteroaryl, amino, mono-Ci-C 6 alkylamino, or di-Ci-C 6 alkylamino, T5 being optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, C -C alkoxyl, or C 3 -C 8 cycloalkyl.

[0219] For example, Q 5 is CO, S(0) 2 , or NHC(O); and T 5 is Ci-C 6 alkyl, Ci-C 6 alkoxyl,

C 3 -C 8 cycloalkyl, or 4 to 12-membered heterocycloalkyl.

[0220] For example, (¾ is Q-C 3 alkyl linker and T5 is H or C 6 -Cio aryl.

[0221] For example, (¾ is Q-C 3 alkyl linker and T5 is C 3 -C 8 cycloalkyl, 4 to 12-membered heterocycloalkyl, or S(0) q R q .

[0222] For example, Q 5 is NHC(O) and T 5 is Q-Ce alkyl or Ci-C 6 alkoxy.

[0223] For example, one or more -Q5-T5 are oxo.

[0224] For example, U is CH-Q5-T5 and n is 0.

[0225] For example, one or more -Q6-T 6 are oxo.

[0226] For example, Q 6 is a bond or C(O) and T 6 is CrC 6 alkyl or CrC 6 alkoxy.

[0227] Still another subset of the compounds of Formula (I) includes those of Formula

(HI):

or pharmaceutically acceptable salts thereof, wherein R 7 is optionally substituted C 3 -C 8 cycloalkyl or a 4- to 6-membered heterocycloalkyl, and R 2 , R3, and R 6 are defined herein for Formula (I).

[0228] Still another subset of the compounds of Formula (I) includes those of Formula

(IV):

or pharmaceutically acceptable salts thereof, wherein R 7 is -Q4-T4, wherein Q 4 is a bond or methyl linker, T 4 is optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 14-membered heterocycloalkyl, and R 1; R 2 , R3, R 4 , R 6 , and R 12 , are as defined herein for Formula (I).

[0229] In addition to the above-described features of the compounds of this invention where applicable, the compounds of Formula (IV) can include one or more of the following features:

[0231] For example, T 4 is

in which R'" is T5, -C(0)T 5 , or S(0) 2 T 5 , T5 being as defined herein for Formula (I).

[0232] For example, the compounds of Formula (IV) include those of Formula (IV a):

or a pharmaceutically acceptable salt thereof; wherein

¾ is 0, 1, or 2;

R 501 is C(H) or N;

R 504 is CM alkyl;

R 506 is Ci-C alkyl, piperidine substituted by 1, 2, or 3 Ci_4 alkyl groups, or cyclohexyl substituted by N(Ci_4 alkyl) 2 wherein one or both of the C 1-4 alkyl is optionally substituted with Ci_6 alkoxyl;

507

R is morpholine, piperazine, piperidine, diazepane, pyrrolidine, azetidine, O-Ci-6 alkyl, NH-Ci-6 alkyl, or O-heterocycle, wherein the heterocycle is a 4-7 membered heterocycle containing an oxygen or nitrogen, or both, and wherein the nitrogen can optionally be substituted with Ci_3 alkyl; wherein the piperazine, piperidine, diazepane, pyrrolidine or azetidine groups can be optionally further substituted with OH, Ci_6 alkyl, or O-C^ alkyl; wherein each of the 0-Ci_6 alkyl and NH-Ci-6 alkyl is optionally substituted with hydroxyl, 0-Ci_3 alkyl or NH-C^ alkyl, each of the O-Ci-3 alkyl and NH-Ci-3 alkyl being optionally further substituted with O-Ci-3 alkyl or NH-C 1-3 alkyl; and

each of Ri and R 2 is as defined herein for Formula (I).

[0233] In addition to the above-described features of the compounds of this invention where applicable, the compounds of Formula (IVa) can include one or more of the following features.

[0234] In certain compounds of Formula (IVa), R 501 is C(H), and R 507 is piperidine;

diazepane; pyrrolidine; azetidine; O-C^ alkyl; or O-heterocycle, wherein the heterocycle is a 4-7 membered heterocycle containing an oxygen or nitrogen, or both, and wherein the nitrogen can optionally be substituted with Ci_3 alkyl; wherein the piperidine, diazepane, pyrrolidine or azetidine groups can be optionally further substituted with OH, C 1-6 alkyl, or O-Ci-3 alkyl.

[0235] In certain compounds of Formula (IVa), R 501 is C(H) and R 507 is piperidine, diazepane, pyrrolidine, azetidine or 0-Ci_6 alkyl, wherein the piperidine, diazepane, pyrrolidine or azetidine groups can be optionally further substituted with OH or C 1-6 alkyl.

[0236] In certain compounds of Formula (IVa), R 501 is C(H), R 507 is piperazine optionally further substituted with Ci_6 alkyl, and R 506 is piperidine substituted by 1, 2, or 3 Ci_ 4 alkyl groups.

[0237] In certain compounds of Formula (IVa), R 501 is N, and R 507 is morpholine, piperidine, piperazine, diazepane, pyrrolidine, azetidine or O-Ci-6 alkyl, wherein the piperidine, piperazine, diazepane, pyrrolidine or azetidine groups can be optionally further substituted with OH or C^ alkyl.

[0238] In certain compounds of Formula (IVa), Ri is H, methyl, or ethyl, and R 2 is halo, cyano, CrC 6 alkoxyl optionally substituted with CrC 6 alkoxyl, Q-C3 alkyl optionally substituted with CrC 6 alkoxyl, amino, mono-Ci-C6 alkylamino, di-CrC 6 alkylamino, optionally substituted 4 to 6-membered heterocycloalkyl (e.g., pyrrolidinyl, pipendinyl, piperazinyl, or morpholinyl), optionally substituted phenyl, or optionally substituted 5- or 6- membered heteroaryl (e.g., pyridinyl, pyrazolyl, pyrimidinyl, quinolinyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furyl, or thienyl).

[0239] In certain compounds of Formula (IVa), Ri and R 2 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms (e.g., pyrazolyl) or a 5 to 12-membered heterocycloalkyl ring having 0 to 2 additional heteroatoms (pyrrolidinyl).

[0240] In certain compounds of Formula (IVa), R 504 is methyl.

[0241] In certain compounds of Formula (IVa), R is

[0242] In certain compounds of Formula (IVa), R is [0243] In certain compounds of Formula (IVa), R is

[0244] In certain compounds of Formula (IVa), R 506 is

[0245] In certain compounds of Formula (IVa), R is

[0246] In certain compounds of Formula (IVa), R is

[0247] In certain compounds of Formula (IVa), R is

R 10 o is phenyl, 5- or 6-membered heteroaryl, or 4 to 12-membered heterocycloalkyl, each optionally substituted with one or more T 5a in which each T 5a is independently Ci-C 6 alkoxyl or O-C1-C4 alkylene-Q^ alkoxy, and R 101 is H or C1-C 4 alkyl. , wherein each Ts a is independently Q-C 3 alkoxyl or 0-Ci-C 3 alkylene-Ci-Ci alkoxy.

[0249] In certain compounds of Formula (IVa), when R 501 is C(H), R 507 is piperidine or diazepane, which are substituted with OH or C 1-6 alkyl, or when R 501 is N, R 507 is piperidine, piperazine, or diazepane, which are optionally further substituted with OH or C . 6 alkyl.

[0250] In certain compounds of Formula (IVa), when R 501 is C(H), R 507 is piperidine substituted with C 1-6 alkyl, or when R 501 is N, R 507 is piperidine substituted with OH or piperazine substituted with C 1-6 alkyl.

[0251] In certain compounds of Formula (IVa), when R 501 is N, R 507 is unsubstituted piperazine.

[0252] In certain compounds of Formula (IVa), ¾ is 0 or 1.

[0253] In certain compounds of Formula (IVa), when R 501 is C(H) or N, R 507 is 0-C 1-6 alkyl or O-heterocycle, and n 5 is 1.

[0254] In certain compounds of Formula (IVa), when R 501 is C(H), R 507 is unsubstituted piperazine and R 506 is piperidine substituted by 1, 2, or 3 C 1-4 alkyl groups.

507

[0255] In certain compounds of Formula (IVa), R is O-C2-3 alkyl substituted with 0-C 1-2 alkyl, e.g., -OCH 2 CH 2 OCH 3 .

[0256] For example, the compounds of Formula (IV) include those of Formula (IVb):

or a pharmaceutically acceptable salt thereof; wherein

n 6 is 0, 1 or 2;

R 604 is Ci_4 alkyl;

R 606 is Ci-C alkyl, tetrahydropyranyl, piperidine substituted by 1, 2, or 3 Q. 4 alkyl groups, or cyclohexyl substituted by N(C 1-4 alkyl) 2 wherein one or both of the Ci-4 alkyl is optionally substituted with C 1-6 alkoxyl;

R 607 is morpholine, piperidine, piperazine, pyrrolidine, diazepane, oxetane, azetidine or O-C^ alkyl, wherein the piperidine, diazepane, oxetane or azetidine groups can be optionally further substituted with one or more C 1-6 alkyl, C 1-6 haloalkyl, C _8 cycloalkyl, or 4 to 6-membered heterocyclo alkyl; and

each of Ri and R 2 is as defined herein for Formula (I).

[0257] In addition to the above-described features of the compounds of this invention where applicable, the compounds of Formula (IVb) can include one or more of the following features:

[0258] In certain compounds of Formula (IVb), R 604 is methyl.

[0259] In certain compounds of Formula (IVb),

[0260] In certain compounds of Formula (IVb),

[0261] In certain compounds of Formula (IVb),

[0262] In certain compounds of Formula (IVb), R

[0263] In certain compounds of Formula (IVb), R

[0264] In certain compounds of Formula (IVb), R is mpounds of Formula (IVb), R is , or

R 10 o is phenyl, 5- or 6-membered heteroaryl, or 4 to 12-membered heterocycloalkyl, each optionally substituted with one or more Ts a in which each Ts a is independently Ci-C6 alkoxyl or O-C Gi alkylene-C 1 -C 4 alkoxy, and R 10 i is H or C C 4 alkyl.

wherein each T 5a is independently Q-C 3 alkoxyl or 0-C C 3 alkylene-Ci-Ci alkoxy.

[0267] In certain compounds of Formula (IVb), R 607 is piperidine or oxetane, each of which is substituted with Ci_ 6 alkyl.

[0268] In certain compounds of Formula (IVb), R 607 is piperidine substituted with CH 2 CF 3 , cyclopropyl, cyclobutyl, or oxetane.

[0269] In certain compounds of Formula (IVb), n 6 is 0 or 1.

[0270] For example, the compounds of Formula (IV) include those of Formula (IVc):

(IVc)

or a pharmaceutically acceptable salt thereof; wherein

R /U1 is H or CM alkyl;

702

R is Ci- 6 alkoxyl or C6-C 10 aryloxy, each optionally substituted with one or more halo;

R 703 is H, halo, or C 1-4 alkyl; R is halo, C 1-4 alkyl or C 1-6 alkoxyl, where each C 1-4 alkyl or C 1-6 alkoxyl is optionally substituted with one or more halo;

701 702

or R and R , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-

701 membered heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R and R 704 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl having 0 to 2 additional heteroatoms or a 5 to 12-membered

heterocycloalkyl ring having 0 to 2 additional heteroatoms; or R 703 and R 704 , together with the C atoms to which they are attached, form C5-C 8 cycloalkyl, C 6 -io aryl, or a 5- or 6-membered heteroaryl having 1 to 3 heteroatoms, or a 5 to 12- membered heterocycloalkyl ring having 1 to 3 heteroatoms; in which each of the ring structures formed by R 701 and R 702 , by R 701 and R 704 , or by R 703 and R 704 , independently is optionally substituted with one or more substituents selected from the group consisting of halo, hydroxyl, oxo, CrC 6 alkyl, C(0)OH, C(0)0-Ci-C 6 alkyl, cyano, CrC 6 alkoxyl, amino, mono-Ci-C 6 alkylamino, di-CrC 6 alkylamino, C3-C 8 cycloalkyl, C 6 -Cio aryl, 4 to 12-membered heterocycloalkyl, and 5- or 6- membered heteroaryl;

R 705 is CM alkyl;

706 707

R is tetrahydropyranyl, piperidine substituted by 1, 2, or 3 R groups, or

707 707

cyclohexyl substituted by N(R ) 2 , wherein each R is independently C 1-4 alkyl that is optionally substituted with Ci_6 alkoxyl, 4 to 12-membered heterocycloalkyl, C6-Q0 aryl that is optionally further substituted with CrC 6 alkoxyl or 0-CrC 4 alkylene-C 1 -C 4 alkoxy, or 5- or 6-membered heteroaryl that is optionally further substituted with C -C alkoxyl or 0-Ci-C 4 alkylene-C 1 -C 4 alkoxy; and

R 6 is as defined herein for Formula (I).

[0271] In addition to the above-described features of the compounds of this invention where applicable, the compounds of Formula (IVc) can include one or more of the following features:

[0274] For example, R /uo is

[0275] For example, R 706 is

[0276] For example, R is

[0277] For example, R /u& is

[0278] For example, R /uo is

[0279] For example, R /uo is

[0280] For example, R 706 is wherein R 00 is phenyl, 5- or 6-membered heteroaryl, or 4 to 12-membered heterocycloalkyl, each optionally substituted with one or more T 5a in which each T 5a is independently CrC 6 alkoxyl or O-C G* alkylene-C 1 -C 4 alkoxy, and R 01 is H or CrC 4 alkyl.

[0283] For example, R /U1 is H.

[0284] For example, R 702 is methoxy, ethoxy, -OCF 3 , or phenoxy.

[0285] For example, R 703 is H.

[0286] For example, R 703 is F.

[0287] For example, R 704 is methyl, ethyl or CF 3 .

[0288] For example, R 704 is methoxy or ethoxy.

[0289] For example, R 704 is F or CI.

[0290] For example, R 701 and R 702 , together with the atoms to which they are attached, form a 5 to 6-membered heterocycloalkyl ring having an oxygen atom.

[0291] For example, R 703 and R 704 , together with the C atoms to which they are attached, form C 5 -C6 cycloalkyl. [0292] For example, R and R , together with the C atoms to which they are attached, form a 5- or 6-membered heteroaryl having 1 to 3 heteroatoms, and the heteroaryl is optionally substituted with Ci_4 alkyl.

[0293] For example, R 703 and R 704 , together with the C atoms to which they are attached, form a 5 to 7-membered heterocycloalkyl ring having 1 to 3 heteroatoms, and the heterocyclo alkyl ring is optionally substituted with C 1-4 alkyl.

[0294] For example, R 701 and R 702 , together with the atoms to which they are attached, form a 5- or 6-membered heteroaryl or a 5 to 6-membered heterocycloalkyl, and R 703 and R 704 , together with the C atoms to which they are attached, form C5-C6 cycloalkyl, a 5- or 6- membered heteroaryl, or a 5 to 12-membered heterocycloalkyl ring.

[0295] For example, R 6 is halo, e.g., F, CI, or Br.

[0296] For example, R 6 is CI.

[0297] For example, R 6 is unsubstituted or substituted phenyl or 5- or 6-membered heteroaryl.

[0298] For example, R 6 is phenyl or 5- or 6-membered heteroaryl substituted with O-Ci-6 alkyl or NH-Ci-6 alkyl, each of which is optionally substituted with hydroxyl, O-C^ alkyl or NH-Ci-3 alkyl, each of the O-C^ alkyl and NH-C 1-3 alkyl being optionally further substituted with 0-Ci- 3 alkyl or NH-C 1-3 alkyl.

[0299] For example, R 6 is

[0300] Representative compounds of the present invention include compounds listed in Tables 1-3. In Table 3, R'" is T5, -C(0)Ts, or S(0) 2 T5, and the other variables such as R 1; R 2 , R 6 , T5 and Ts a are as defined herein for Formula (I).

Table 1

50

51

52

53

55



Table 2B Table 3

[0301] As used herein, "alkyl", "Ci, C 2 , C 3 , C 4 , C 5 or C 6 alkyl" or "C C 6 alkyl" is intended to include C 1 ; C 2 , C 3 , C 4 , C 5 or C 6 straight chain (linear) saturated aliphatic hydrocarbon groups and C 3 , C 4 , C 5 or C 6 branched saturated aliphatic hydrocarbon groups. For example, C1-C6 alkyl is intended to include Cl, C2, C3, C4, C5 and Cfr alkyl groups. Examples of alkyl include, moieties having from one to six carbon atoms, such as, but not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, t-butyl, n-pentyl, s-pentyl or n- hexyl.

[0302] In certain embodiments, a straight chain or branched alkyl has six or fewer carbon atoms (e.g. , Ci-C6 for straight chain, C 3 -C 6 for branched chain), and in another embodiment, a straight chain or branched alkyl has four or fewer carbon atoms.

[0303] As used herein, the term "cycloalkyl" refers to a saturated or unsaturated

nonaromatic hydrocarbon mono-or multi-ring (e.g., fused, bridged, or spiro rings) system having 3 to 30 carbon atoms (e.g., C 3 -Cio). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, and adamantyl. The term "heterocycloalkyl" refers to a saturated or unsaturated nonaromatic 3-8 membered monocyclic, 7-12 membered bicyclic (fused, bridged, or spiro rings), or 11- 14 membered tricyclic ring system (fused, bridged, or spiro rings) having one or more heteroatoms (such as O, N, S, or Se), unless specified otherwise. Examples of heterocycloalkyl groups include, but are not limited to, piperidinyl, piperazinyl, pyrrolidinyl, dioxanyl, tetrahydrofuranyl, isoindolinyl, indolinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl, isoxazolidinyl, triazolidinyl, tetrahyrofuranyl, oxiranyl, azetidinyl, oxetanyl, thietanyl, 1,2,3,6-tetrahydropyridinyl, tetrahydropyranyl, dihydropyranyl, pyranyl, morpholinyl, tetrahydrothiopyranyl, 1,4-diazepanyl, 1,4- oxazepanyl, 2-oxa-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2-oxa-6- azaspiro[3.3]heptanyl, 2,6-diazaspiro[3.3]heptanyl, l,4-dioxa-8-azaspiro[4.5]decanyl, 1,4- dioxaspiro[4.5]decanyl, l-oxaspiro[4.5]decanyl, l-azaspiro[4.5]decanyl, 3Ή- spiro [cyclohexane- 1 , 1 '-isobenzofuran] -yl, 7Ή- spiro [cyclohexane- 1 ,5 '-furo [3 ,4-b]pyridin] - yl, 3'H-spiro[cyclohexane-l, l'-furo[3,4-c]pyridin]-yl, and the like.

[0304] The term "optionally substituted alkyl" refers to unsubstituted alkyl or alkyl having designated substituents replacing one or more hydrogen atoms on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl,

aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.

[0305] An "arylalkyl" or an "aralkyl" moiety is an alkyl substituted with an aryl (e.g. , phenylmethyl (benzyl)). An "alkylaryl" moiety is an aryl substituted with an alkyl (e.g. , methylphenyl).

[0306] As used herein, "alkyl linker" is intended to include C 1 ; C 2 , C 3 , C 4 , C 5 or C 6 straight chain (linear) saturated divalent aliphatic hydrocarbon groups and C 3 , C 4 , C 5 or C 6 branched saturated aliphatic hydrocarbon groups. For example, Cl-C6 alkyl linker is intended to include Ci, C2, C3, C4, C5 and Cfr alkyl linker groups. Examples of alkyl linker include, moieties having from one to six carbon atoms, such as, but not limited to, methyl (-CH 2 -), ethyl (-CH 2 CH 2 -), n-propyl (-CH 2 CH 2 CH 2 -), i-propyl (-CHCH 3 CH 2 -), n-butyl (- CH 2 CH 2 CH 2 CH 2 -), s-butyl (-CHCH 3 CH 2 CH 2 -), i-butyl (-C(CH 3 ) 2 CH 2 -), n-pentyl (- CH 2 CH 2 CH 2 CH 2 CH 2 -), s-pentyl (-CHCH 3 CH 2 CH 2 CH 2 -) or n-hexyl (- CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) .

[0307] "Alkenyl" includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double bond. For example, the term "alkenyl" includes straight chain alkenyl groups (e.g. , ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl), and branched alkenyl groups. In certain embodiments, a straight chain or branched alkenyl group has six or fewer carbon atoms in its backbone (e.g. , C 2 -C 6 for straight chain, C3-C 6 for branched chain). The term "C 2 -C 6 " includes alkenyl groups containing two to six carbon atoms. The term "C 3 - C 6 " includes alkenyl groups containing three to six carbon atoms.

[0308] The term "optionally substituted alkenyl" refers to unsubstituted alkenyl or alkenyl having designated substituents replacing one or more hydrogen atoms on one or more hydrocarbon backbone carbon atoms. Such substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy,

alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl,

alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino

(including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.

[0309] "Alkynyl" includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond. For example, "alkynyl" includes straight chain alkynyl groups (e.g. , ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl), and branched alkynyl groups. In certain embodiments, a straight chain or branched alkynyl group has six or fewer carbon atoms in its backbone (e.g. , C 2 -C 6 for straight chain, C 3 -C 6 for branched chain). The term "C 2 -C 6 " includes alkynyl groups containing two to six carbon atoms. The term "C 3 -C 6 " includes alkynyl groups containing three to six carbon atoms.

[0310] The term "optionally substituted alkynyl" refers to unsubstituted alkynyl or alkynyl having designated substituents replacing one or more hydrogen atoms on one or more hydrocarbon backbone carbon atoms. Such substituents can include, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy,

alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl,

alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino

(including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.

[0311] Other optionally substituted moieties (such as optionally substituted cycloalkyl, heterocycloalkyl, aryl, or heteroaryl) include both the unsubstituted moieties and the moieties having one or more of the designated substituents. For example, substituted heterocycloalkyl includes those substituted with one or more alkyl groups, such as 2,2,6,6- tetramethyl-piperidinyl and 2,2,6,6-tetramethyl- 1 ,2,3,6-tetrahydropyridinyl.

[0312] "Aryl" includes groups with aromaticity, including "conjugated," or multicyclic systems with at least one aromatic ring and do not contain any heteroatom in the ring structure. Examples include phenyl, benzyl, 1,2,3,4-tetrahydronaphthalenyl, etc.

[0313] "Heteroaryl" groups are aryl groups, as defined above, except having from one to four heteroatoms in the ring structure, and may also be referred to as "aryl heterocycles" or "heteroaromatics." As used herein, the term "heteroaryl" is intended to include a stable 5-, 6-, or 7-membered monocyclic or 7-, 8-, 9-, 10-, 11- or 12-membered bicyclic aromatic heterocyclic ring which consists of carbon atoms and one or more heteroatoms, e.g. , 1 or 1- 2 or 1-3 or 1-4 or 1-5 or 1-6 heteroatoms, or e.g. ^ l, 2, 3, 4, 5, or 6 heteroatoms,

independently selected from the group consisting of nitrogen, oxygen and sulfur. The nitrogen atom may be substituted or unsubstituted (i.e. , N or NR wherein R is H or other substituents, as defined). The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e. , N— >0 and S(0) p , where p = 1 or 2). It is to be noted that total number of S and O atoms in the aromatic heterocycle is not more than 1.

[0314] Examples of heteroaryl groups include pyrrole, furan, thiophene, thiazole, isothiazole, imidazole, triazole, tetrazole, pyrazole, oxazole, isoxazole, pyridine, pyrazine, pyridazine, pyrimidine, and the like.

[0315] Furthermore, the terms "aryl" and "heteroaryl" include multicyclic aryl and heteroaryl groups, e.g. , tricyclic, bicyclic, e.g. , naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, quinoline, isoquinoline, naphthrydine, indole, benzofuran, purine, benzofuran, deazapurine, indolizine.

[0316] In the case of multicyclic aromatic rings, only one of the rings needs to be aromatic (e.g. , 2,3-dihydroindole), although all of the rings may be aromatic (e.g. , quinoline). The second ring can also be fused or bridged.

[0317] The cycloalkyl, heterocycloalkyl, aryl, or heteroaryl ring can be substituted at one or more ring positions (e.g., the ring-forming carbon or heteroatom such as N) with such substituents as described above, for example, alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminocarbonyl, aralkylaminocarbonyl,

alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or hetero aromatic moiety. Aryl and heteroaryl groups can also be fused or bridged with alicyclic or heterocyclic rings, which are not aromatic so as to form a multicyclic system (e.g. , tetralin, methylenedioxyphenyl such as benzo[J][l,3]dioxole-5-yl).

[0318] As used herein, "carbocycle" or "carbocyclic ring" is intended to include any stable monocyclic, bicyclic or tricyclic ring having the specified number of carbons, any of which may be saturated, unsaturated, or aromatic. Carbocycle includes cycloalkyl and aryl. For example, a C3-C 14 carbocycle is intended to include a monocyclic, bicyclic or tricyclic ring having 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 carbon atoms. Examples of carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, fluorenyl, phenyl, naphthyl, indanyl, adamantyl and tetrahydronaphthyl. Bridged rings are also included in the definition of carbocycle, including, for example, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, and [4.4.0]

bicyclodecane and [2.2.2] bicyclooctane. A bridged ring occurs when one or more carbon atoms link two non-adjacent carbon atoms. In one embodiment, bridge rings are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge. Fused (e.g. , naphthyl, tetrahydronaphthyl) and spiro rings are also included.

[0319] As used herein, "heterocycle" or "heterocyclic group" includes any ring structure (saturated, unsaturated, or aromatic) which contains at least one ring heteroatom (e.g. , N, O or S). Heterocycle includes heterocycloalkyl and heteroaryl. Examples of heterocycles include, but are not limited to, morpholine, pyrrolidine, tetrahydrothiophene, piperidine, piperazine, oxetane, pyran, tetrahydropyran, azetidine, and tetrahydrofuran.

[0320] Examples of heterocyclic groups include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-l,5,2-dithiazinyl,

dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl (e.g., benzo[J][l,3]dioxole-5-yl), morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4- oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, l,2,4-oxadiazol5(4H)-one, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl,

phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl,

tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-l,2,5-thiadiazinyl, 1,2,3- thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3- triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl and xanthenyl.

[0321] The term "substituted," as used herein, means that any one or more hydrogen atoms on the designated atom is replaced with a selection from the indicated groups, provided that the designated atom' s normal valency is not exceeded, and that the substitution results in a stable compound. When a substituent is oxo or keto (i.e. , =0), then 2 hydrogen atoms on the atom are replaced. Keto substituents are not present on aromatic moieties. Ring double bonds, as used herein, are double bonds that are formed between two adjacent ring atoms (e.g. , C=C, C=N or N=N). "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.

[0322] When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom in the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such formula. Combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds. [0323] When any variable (e.g. , R) occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-2 R moieties, then the group may optionally be substituted with up to two R moieties and R at each occurrence is selected independently from the definition of R. Also, combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds.

[0324] The term "hydroxy" or "hydroxyl" includes groups with an -OH or -O " .

[0325] As used herein, "halo" or "halogen" refers to fluoro, chloro, bromo and iodo. The term "perhalogenated" generally refers to a moiety wherein all hydrogen atoms are replaced by halogen atoms. The term "haloalkyl" or "haloalkoxyl" refers to an alkyl or alkoxyl substituted with one or more halogen atoms.

[0326] The term "carbonyl" includes compounds and moieties which contain a carbon connected with a double bond to an oxygen atom. Examples of moieties containing a carbonyl include, but are not limited to, aldehydes, ketones, carboxylic acids, amides, esters, anhydrides, etc.

[0327] The term "carboxyl" refers to -C(0)OH or its Ci-C 6 alkyl ester.

[0328] "Acyl" includes moieties that contain the acyl radical (R-C(O)-) or a carbonyl group. "Substituted acyl" includes acyl groups where one or more of the hydrogen atoms are replaced by, for example, alkyl groups, alkynyl groups, halogen, hydroxyl,

alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or hetero aromatic moiety.

[0329] "Aroyl" includes moieties with an aryl or heteroaromatic moiety bound to a carbonyl group. Examples of aroyl groups include phenylcarboxy, naphthyl carboxy, etc.

[0330] "Alkoxyalkyl," "alkylaminoalkyl," and "thioalkoxyalkyl" include alkyl groups, as described above, wherein oxygen, nitrogen, or sulfur atoms replace one or more hydrocarbon backbone carbon atoms. [0331] The term "alkoxy" or "alkoxyl" includes substituted and unsubstituted alkyl, alkenyl and alkynyl groups covalently linked to an oxygen atom. Examples of alkoxy groups or alkoxyl radicals include, but are not limited to, methoxy, ethoxy, isopropyloxy, propoxy, butoxy and pentoxy groups. Examples of substituted alkoxy groups include halogenated alkoxy groups. The alkoxy groups can be substituted with groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy,

alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl,

alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkylamino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino

(including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moieties. Examples of halogen substituted alkoxy groups include, but are not limited to, fluoromethoxy, difluoromethoxy, trifluoromethoxy, chloromethoxy, dichloromethoxy and trichloromethoxy.

[0332] The term "ether" or "alkoxy" includes compounds or moieties which contain an oxygen bonded to two carbon atoms or heteroatoms. For example, the term includes "alkoxyalkyl," which refers to an alkyl, alkenyl, or alkynyl group covalently bonded to an oxygen atom which is covalently bonded to an alkyl group.

[0333] The term "ester" includes compounds or moieties which contain a carbon or a heteroatom bound to an oxygen atom which is bonded to the carbon of a carbonyl group. The term "ester" includes alkoxycarboxy groups such as methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, etc.

[0334] The term "thioalkyl" includes compounds or moieties which contain an alkyl group connected with a sulfur atom. The thioalkyl groups can be substituted with groups such as alkyl, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy,

alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, carboxyacid, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, amino (including alkylamino, dialkylamino, arylamino, diarylamino and alkylarylamino), acylamino (including alkylcarbonylamino,

arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moieties.

[0335] The term "thiocarbonyl" or "thiocarboxy" includes compounds and moieties which contain a carbon connected with a double bond to a sulfur atom.

[0336] The term "thioether" includes moieties which contain a sulfur atom bonded to two carbon atoms or heteroatoms. Examples of thioethers include, but are not limited to alkthioalkyls, alkthioalkenyls, and alkthioalkynyls. The term "alkthioalkyls" include moieties with an alkyl, alkenyl, or alkynyl group bonded to a sulfur atom which is bonded to an alkyl group. Similarly, the term "alkthioalkenyls" refers to moieties wherein an alkyl, alkenyl or alkynyl group is bonded to a sulfur atom which is covalently bonded to an alkenyl group; and alkthioalkynyls" refers to moieties wherein an alkyl, alkenyl or alkynyl group is bonded to a sulfur atom which is covalently bonded to an alkynyl group.

[0337] As used herein, "amine" or "amino" refers to -NH 2 . "Alkylamino" includes groups of compounds wherein the nitrogen of -NH 2 is bound to at least one alkyl group. Examples of alkylamino groups include benzylamino, methylamino, ethylamino, phenethylamino, etc. "Dialkylamino" includes groups wherein the nitrogen of -NH 2 is bound to two alkyl groups. Examples of dialkylamino groups include, but are not limited to, dimethylamino and diethylamino. "Arylamino" and "diarylamino" include groups wherein the nitrogen is bound to at least one or two aryl groups, respectively. "Aminoaryl" and "aminoaryloxy" refer to aryl and aryloxy substituted with amino. "Alkylarylamino," "alkylaminoaryl" or "arylaminoalkyl" refers to an amino group which is bound to at least one alkyl group and at least one aryl group. "Alkaminoalkyl" refers to an alkyl, alkenyl, or alkynyl group bound to a nitrogen atom which is also bound to an alkyl group. "Acylamino" includes groups wherein nitrogen is bound to an acyl group. Examples of acylamino include, but are not limited to, alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido groups.

[0338] The term "amide" or "aminocarboxy" includes compounds or moieties that contain a nitrogen atom that is bound to the carbon of a carbonyl or a thiocarbonyl group. The term includes "alkaminocarboxy" groups that include alkyl, alkenyl or alkynyl groups bound to an amino group which is bound to the carbon of a carbonyl or thiocarbonyl group. It also includes "arylaminocarboxy" groups that include aryl or heteroaryl moieties bound to an amino group that is bound to the carbon of a carbonyl or thiocarbonyl group. The terms "alkylaminocarboxy", "alkenylaminocarboxy", "alkynylaminocarboxy" and

"arylaminocarboxy" include moieties wherein alkyl, alkenyl, alkynyl and aryl moieties, respectively, are bound to a nitrogen atom which is in turn bound to the carbon of a carbonyl group. Amides can be substituted with substituents such as straight chain alkyl, branched alkyl, cycloalkyl, aryl, heteroaryl or heterocycle. Substituents on amide groups may be further substituted.

[0339] Compounds of the present invention that contain nitrogens can be converted to N- oxides by treatment with an oxidizing agent (e.g. , 3-chloroperoxybenzoic acid (mCPBA) and/or hydrogen peroxides) to afford other compounds of the present invention. Thus, all shown and claimed nitrogen-containing compounds are considered, when allowed by valency and structure, to include both the compound as shown and its N-oxide derivative (which can be designated as N— >0 or N + -0 " ). Furthermore, in other instances, the nitrogens in the compounds of the present invention can be converted to N-hydroxy or N-alkoxy compounds. For example, N-hydroxy compounds can be prepared by oxidation of the parent amine by an oxidizing agent such as m-CPBA. All shown and claimed nitrogen- containing compounds are also considered, when allowed by valency and structure, to cover both the compound as shown and its N-hydroxy (i.e. , N-OH) and N-alkoxy (i.e. , N-OR, wherein R is substituted or unsubstituted C C 6 alkyl, C C 6 alkenyl, C C 6 alkynyl, 3- 14- membered carbocycle or 3- 14-membered heterocycle) derivatives.

[0340] In the present specification, the structural formula of the compound represents a certain isomer for convenience in some cases, but the present invention includes all isomers, such as geometrical isomers, optical isomers based on an asymmetrical carbon,

stereoisomers, tautomers, and the like, it being understood that not all isomers may have the same level of activity. In addition, a crystal polymorphism may be present for the compounds represented by the formula. It is noted that any crystal form, crystal form mixture, or anhydride or hydrate thereof is included in the scope of the present invention.

[0341] "Isomerism" means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or in the arrangement of their atoms in space.

Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers." Stereoisomers that are not mirror images of one another are termed "diastereoisomers," and stereoisomers that are non-superimposable mirror images of each other are termed

"enantiomers" or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a "racemic mixture."

[0342] A carbon atom bonded to four nonidentical substituents is termed a "chiral center." [0343] "Chiral isomer" means a compound with at least one chiral center. Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed "diastereomeric mixture." When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Rule of Cahn, Ingold and Prelog. (Cahn et al., Angew. Chem. Inter. Edit. 1966, 5, 385; errata 511; Cahn et al., Angew. Chem. 1966, 78, 413; Cahn and Ingold, J. Chem. Soc. 1951 (London), 612; Cahn et al., Experientia 1956, 12, 81; Cahn, J. Chem. Educ. 1964, 41, 116).

[0344] "Geometric isomer" means the diastereomers that owe their existence to hindered rotation about double bonds or a cycloalkyl linker (e.g., 1,3-cylcobutyl). These

configurations are differentiated in their names by the prefixes cis and trans, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingold-Prelog rules.

[0345] It is to be understood that the compounds of the present invention may be depicted as different chiral isomers or geometric isomers. It should also be understood that when compounds have chiral isomeric or geometric isomeric forms, all isomeric forms are intended to be included in the scope of the present invention, and the naming of the compounds does not exclude any isomeric forms, it being understood that not all isomers may have the same level of activity.

[0346] Furthermore, the structures and other compounds discussed in this invention include all atropic isomers thereof, it being understood that not all atropic isomers may have the same level of activity. "Atropic isomers" are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques, it has been possible to separate mixtures of two atropic isomers in select cases.

[0347] "Tautomer" is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solutions where tautomerization is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertable by tautomerizations is called tautomerism.

[0348] Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs. Ring-chain tautomerism arises as a result of the aldehyde group (-CHO) in a sugar chain molecule reacting with one of the hydroxy groups (-OH) in the same molecule to give it a cyclic (ring-shaped) form as exhibited by glucose.

[0349] Common tautomeric pairs are: ketone-enol, amide-nitrile, lactam-lactim, amide- imidic acid tautomerism in heterocyclic rings (e.g. , in nucleobases such as guanine, thymine and cytosine), imine-enamine and enamine-enamine. An example of keto-enol equilibria is between pyridin-4(lH)-ones and the corresponding pyridin-4-ols, as shown below.

pyridin-4(1H)-one pyridin-4-ol

[0350] It is to be understood that the compounds of the present invention may be depicted as different tautomers. It should also be understood that when compounds have tautomeric forms, all tautomeric forms are intended to be included in the scope of the present invention, and the naming of the compounds does not exclude any tautomer form. It will be understood that certain tautomers may have a higher level of activity than others.

[0351] The term "crystal polymorphs", "polymorphs" or "crystal forms" means crystal structures in which a compound (or a salt or solvate thereof) can crystallize in different crystal packing arrangements, all of which have the same elemental composition. Different crystal forms usually have different X-ray diffraction patterns, infrared spectral, melting points, density hardness, crystal shape, optical and electrical properties, stability and solubility. Recrystallization solvent, rate of crystallization, storage temperature, and other factors may cause one crystal form to dominate. Crystal polymorphs of the compounds can be prepared by crystallization under different conditions.

[0352] The compounds of any Formula described herein include the compounds themselves, as well as their salts, and their solvates, if applicable. A salt, for example, can be formed between an anion and a positively charged group (e.g., amino) on a 1,4-pyridone compound. Suitable anions include chloride, bromide, iodide, sulfate, bisulfate, sulfamate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, glutamate, glucuronate, glutarate, malate, maleate, succinate, fumarate, tartrate, tosylate, salicylate, lactate, naphthalenesulfonate, and acetate (e.g., trifluoroacetate). The term "pharmaceutically acceptable anion" refers to an anion suitable for forming a pharmaceutically acceptable salt. Likewise, a salt can also be formed between a cation and a negatively charged group (e.g., carboxylate) on a 1,4-pyridone compound. Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion. The 1,4-pyridone compounds also include those salts containing quaternary nitrogen atoms.

[0353] Additionally, the compounds of the present invention, for example, the salts of the compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules. Nonlimiting examples of hydrates include monohydrates, dihydrates, etc. Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.

[0354] "Solvate" means solvent addition forms that contain either stoichiometric or non stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate; and if the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one molecule of the substance in which the water retains its molecular state as H 2 0.

[0355] As used herein, the term "analog" refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the replacement of one functional group by another functional group). Thus, an analog is a compound that is similar or comparable in function and appearance, but not in structure or origin to the reference compound.

[0356] As defined herein, the term "derivative" refers to compounds that have a common core structure, and are substituted with various groups as described herein. For example, all of the compounds represented by Formula (I) are 1,4-pyridone compounds, and have Formula (I) as a common core.

[0357] The term "bioisostere" refers to a compound resulting from the exchange of an atom or of a group of atoms with another, broadly similar, atom or group of atoms. The objective of a bioisosteric replacement is to create a new compound with similar biological properties to the parent compound. The bioisosteric replacement may be physicochemically or topologically based. Examples of carboxylic acid bioisosteres include, but are not limited to, acyl sulfonimides, tetrazoles, sulfonates and phosphonates. See, e.g., Patani and LaVoie, Chem. Rev. 96, 3147-3176, 1996.

[0358] The present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium, and isotopes of carbon include C-13 and C-14.

[0359] The present invention provides methods for the synthesis of the compounds of any of the Formulae described herein. The present invention also provides detailed methods for the synthesis of various disclosed compounds of the present invention according to the following schemes as shown in the Examples.

[0360] Throughout the description, where compositions are described as having, including, or comprising specific components, it is contemplated that compositions also consist essentially of, or consist of, the recited components. Similarly, where methods or processes are described as having, including, or comprising specific process steps, the processes also consist essentially of, or consist of, the recited processing steps. Further, it should be understood that the order of steps or order for performing certain actions is immaterial so long as the invention remains operable. Moreover, two or more steps or actions can be conducted simultaneously.

[0361] The synthetic processes of the invention can tolerate a wide variety of functional groups, therefore various substituted starting materials can be used. The processes generally provide the desired final compound at or near the end of the overall process, although it may be desirable in certain instances to further convert the compound to a pharmaceutically acceptable salt thereof.

[0362] Compounds of the present invention can be prepared in a variety of ways using commercially available starting materials, compounds known in the literature, or from readily prepared intermediates, by employing standard synthetic methods and procedures either known to those skilled in the art, or which will be apparent to the skilled artisan in light of the teachings herein. Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be obtained from the relevant scientific literature or from standard textbooks in the field.

Although not limited to any one or several sources, classic texts such as Smith, M. B., March, J., March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure , 5 th edition, John Wiley & Sons: New York, 2001; Greene, T.W., Wuts, P.G. M., Protective Groups in Organic Synthesis, 3 r edition, John Wiley & Sons: New York, 1999; R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); L. Fieser and M. Fieser, Fieser and Fieser' s Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), incorporated by reference herein, are useful and recognized reference textbooks of organic synthesis known to those in the art. The following descriptions of synthetic methods are designed to illustrate, but not to limit, general procedures for the preparation of compounds of the present invention.

[0363] Compounds of the present invention can be conveniently prepared by a variety of methods familiar to those skilled in the art or those described in W 2012/142504, WO 2012/142513 and WO 2012/118812, which are incorporated herein by reference. The compounds of this invention with any of the Formulae described herein may be prepared according to the procedures illustrated in Schemes 1-4 below, from commercially available starting materials or starting materials which can be prepared using literature procedures. The R groups (such as R 1; R 2 , R 3 , R4, R 6 , R7, Rs, and R 12 ) in Schemes 1-4 are as defined in any Formula described herein, unless otherwise specified.

[0364] One of ordinary skill in the art will note that, during the reaction sequences and synthetic schemes described herein, the order of certain steps may be changed, such as the introduction and removal of protecting groups.

[0365] One of ordinary skill in the art will recognize that certain groups may require protection from the reaction conditions via the use of protecting groups. Protecting groups may also be used to differentiate similar functional groups in molecules. A list of protecting groups and how to introduce and remove these groups can be found in Greene, T.W., Wuts, P.G. M., Protective Groups in Organic Synthesis, 3 rd edition, John Wiley & Sons: New York, 1999.

[0366] Preferred protecting groups include, but are not limited to:

[0367] For a hydroxyl moiety: TBS, benzyl, THP, Ac

[0368] For carboxylic acids: benzyl ester, methyl ester, ethyl ester, allyl ester

[0369] For amines: Cbz, BOC, DMB

[0370] For diols: Ac (x2) TBS (x2), or when taken together acetonides

[0371] For thiols: Ac

[0372] For benzimidazoles: SEM, benzyl, PMB, DMB

[0373] For aldehydes: di-alkyl acetals such as dimethoxy acetal or diethyl acetyl. [0374] In the reaction schemes described herein, multiple stereoisomers may be produced. When no particular stereoisomer is indicated, it is understood to mean all possible stereoisomers that could be produced from the reaction. A person of ordinary skill in the art will recognize that the reactions can be optimized to give one isomer preferentially, or new schemes may be devised to produce a single isomer. If mixtures are produced, techniques such as preparative thin layer chromatography, preparative HPLC, preparative chiral HPLC, or preparative SFC may be used to separate the isomers.

[0375] The following abbreviations are used throughout the specification and are defined

AA ammonium acetate

ACN acetonitrile

Ac acetyl

AcOH acetic acid

atm atmosphere

aq. Aqueous

BID or b.i.d. bis in die (twice a day)

tBuOK potassium t-butoxide

Bn benzyl

BOC tert-butoxy carbonyl

BOP (benzotriazol- 1 -yloxy)tris(dimethylamino)- pho sphoniumhexafluoropho sphate

Cbz benzyloxy carbonyl

CDC1 3 deuterated chloroform

CH 2 C1 2 dichloromethane

COMU (l-Cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethyl- amino-morpholino-carbenium hexafluorophosphate d days

DBU l,8-diazabicyclo[5.4.0]undec-7-ene

DCE 1,2 dichloroethane

DCM dichloromethane

DEAD Diethyl azodicarboxylate

DIAD Diisopropyl azodicarboxylate

DiBAL-H diisobutyl aluminium hydride

DIPEA Ν,Ν-diisopropylethylamine (Hunig's base) [0399] DMA Dimethylacetamide

[0400] DMAP N, N dimethyl-4-aminopyridine

[0401] DMB 2,4 dimethoxy benzyl

[0402] DMF N,N-Dimethylformamide

[0403] DMSO Dimethyl sulfoxide

[0404] DPPA Diphenylphosphonic azide

[0405] EA or EtOAc Ethyl acetate

[0406] EDC or EDCI N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide

[0407] Et 2 0 diethyl ether

[0408] ELS Evaporative Light Scattering

[0409] ESI- Electrospray negative mode

[0410] ESI+ Electrospray positive mode

[0411] Et 3 N or TEA triethylamine

[0412] EtOH ethanol

[0413] FA formic acid

[0414] FC or FCC Flash chromatogrpahy

[0415] h hours

[0416] H 2 0 water

[0417] HATU 0- (7-Azabenzotriazol-l-yl)-N,N,N',N'- tetramethyluronium hexafluorophosphate

[0418] HOAT 1- Hydroxy-7-azabenzotriazole

[0419] HOBt 1 -Hydroxybenzotriazole

[0420] HO-Su N-Hydroxysuccinimide

[0421] HC1 hydrogen chloride or hydrochloric acid

[0422] HPLC High performance liquid chromatography

[0423] K 2 C0 3 potassium carbonate

[0424] KHMDs Potassium hexamethyldisilazide

[0425] LC/MS or LC-MS Liquid chromatography mass spectrum

[0426] LDA Lithium diisopropylamide

[0427] LiHMDs Lithium hexamethyldisilazide

[0428] LG leaving group

[0429] M Molar

[0430] m/z mass/charge ratio

[0431] m-CPBA meta-chloroperbenzoic acid [0432] MeCN Acetonitrile

[0433] MeOD d4-methanol

[0434] Mel Methyl iodide

[0435] MS3A 3A molecular sieves

[0436] MgS0 4 Magnesium Sulfate

[0437] min minutes

[0438] Ms Mesyl

[0439] MsCl Mesyl chloride

[0440] MsO Mesylate

[0441] MS Mass Spectrum

[0442] MWI microwave irradiation

[0443] Na 2 C0 3 sodium carbonate

[0444] Na 2 S0 4 sodium sulfate

[0445] NaHC0 3 sodium bicarbonate

[0446] NaHMDs Sodium hexamethyldisilazide

[0447] NaOH sodium hydroxide

[0448] NaHC0 3 sodium bicarbonate

[0449] Na 2 S0 4 sodium sulfate

[0450] NIS N-iodo succinimide

[0451] NMR Nuclear Magnetic Resonance

[0452] o/n or O/N overnight

[0453] Pd/C Palladium on carbon

[0454] Pd(dppf)Cl 2 .DCM [1,1 '-Bis(diphenylphosphino)ferrocene]

dichloropalladium(II),complex with dichloromethane

[0455] PPAA 1-Propanephosphonic acid cyclic anhydride

[0456] Pd(OH) 2 Palladium dihydroxide

[0457] PE Petroleum Ether

[0458] PG protecting group

[0459] PMB para methoxybenzyl

[0460] ppm parts per million

[0461] p.o. per os (oral adinsitration)

[0462] prep HPLC preparative High Performance Liquid Chromatography

[0463] prep TLC preparative thin layer chromatography

[0464] p-TsOH para-toluenesulfonic acid [0465] PYBOP (Benzotriazol- 1 -yloxy)tripyrrolidinophosphonium

Hexafluoropho sphate

[0466] QD or q.d. quaque die (once a day)

[0467] RBF round bottom flask

[0468] RP-HPLC Reverse phase High Perfomance liquid chromatography

[0469] Rt or RT Room temperature

[0470] SEM (Trimethylsilyl)ethoxymethyl

[0471] SEMC1 (Trimethylsilyl)ethoxymethyl chloride

[0472] SFC Super critical chromatography

[0473] SGC silica gel chromatography

[0474] STAB Sodium triacetoxy borohydride

[0475] TBAF tetra-n-butylammonium fluoride

[0476] TBME ie/t-Butyl methyl ether

[0477] TEA Triethylamine

[0478] TFA trif uoroacetic acid

[0479] TfO trif ate

[0480] THF tetrahydrofuran

[0481] THP tetrahydropyran

[0482] TID or t.i.d ter in die (three times a day)

[0483] TLC thin layer chromatography

[0484] TMSC1 Trimethylsilyl chloride

[0485] Ts tosyl

[0486] TsOH to sic acid

[0487] UV ultraviolet

Scheme 1

[0488] Scheme 1 shows the synthesis of modified aryl analogs following a general route that utilizes well-established chemistry. Substituted nitrobenzoic acids, many of which are commercially available or can be made by nitration of the appropriate substituted benzoic acids or other chemistry known to one skilled in the art, can be converted to their methyl esters by treatment with methyliodide in a polar solvent, such as DMF, in the presence of an appropriate base, such as sodium carbonate, at an appropriate temperature, such as 60 °C (Step 1). The nitro group can be reduced to an amine using an appropriate reducing agent, such as iron, in the presence of an acid, such as ammonium chloride, in a protic solvent, such as ethanol, at an appropriate temperature, such as 80 °C (Step 2). Introduction of the R 8 can be done using a reductive amination with an appropriate ketone or aldehyde in the presence of an appropriate reducing agent, such as sodium cyanoborohydride, and catalytic acid, such as acetic acid, in an appropriate solvent, such as methanol. A variety of R 7 groups can be introduced by alkylation using R 7 -LG, where LG is a leaving group, such as iodine, in the presence of a mild base, such as cesium carbonate, in an appropriate polar solvent, such as acetonitrile, at an appropriate temperature, such as 80 °C (Step 4).

Alternatively, R 7 groups can be introduced by reductive amination with R 7 -ketone or R 7 - aldehyde in the presence of an appropriate reducing agent, such as sodium

cyanoborohydride, and catalytic acid, such as acetic acid, in an appropriate solvent, such as methanol. The ester moiety can be converted to an amide using a standard two step protocol. The ester can be hydrolyzed to the corresponding acid using a suitable base, such as sodium hydroxide, in a polar solvent, such as ethanol (Step 5).

Scheme 2

[0489] Scheme 2 shows the synthesis of modified pyridone analogs following a general route that utilizes well-established chemistry. Substituted l,3-dioxin-4-ones, many of which are commercially available or can be made via rearrangement of acylated Meldrum's acid or via halogenation of l,3-dioxin-4-one followed by aliphatic, aromatic or

heteroaromatic coupling reactions, or other chemistry known to one skilled in the art, can be converted to the substituted 1,4-pyridones by reaction with appropriately substituted (E)-3- aminoacrylonitriles (Step 1) at, e.g., an elevated temperature. The resulting nitrile group can be reduced to an amine using an appropriate reducing agent, such as Raney-Nickel in the presence of hydrogen, in a protic solvent, such as methanol containing ammonia, at an appropriate temperature, such as 22 °C (Step 2). The resulting amine would then be subjected to a standard amide coupling reaction whereupon the appropriate acid (see, e.g., Scheme 1, WO 2012/142504 and WO 2012/142513, which are incorporated herein by reference) would be added along with a suitable amide coupling reagent, such as PYBOP, in a suitable solvent, such as DMSO, to give the desired amide (Step 3).

Scheme 3

[0490] In certain embodiments the 1,4-pyridone can be modified via alkylation or acylation protocols known to ones skilled in the art, prior to condensation with the elaborated acid coupling partner. A variety of Ri groups can be introduced by alkylation using Ri-LG, where LG is a leaving group such as iodine, in the presence of a strong base such as NaH in an appropriate polar aprotic solvent such as DMF at an appropriate temperature such as 80 °C (Step 2).

Scheme 4

R 2 =OR a or NHR ;

[0491] In another protocol as depicted in Scheme 4, substituted 2,4-dichloronicotinonitriles can be synthesized in two steps from substituted 2-amino-4H-pyran-4-ones. Rearrangement to the 4-hydroxypyridin-2(lH)-one is affected by the addition of aqueous inorganic acid and the resulting pyridone is converted into the 2,4-dichloronicotinonitrile by dehydrative chlorination using an appropriate reagent such as POCl 3 in an aprotic solvent such as DMF (Steps 1-2). The nitrile group can then be reduced to an amine using an appropriate reducing agent such as Raney-Nickel in the presence of hydrogen in a protic solvent such as methanol containing ammonia at an appropriate temperature such as 22 °C (Step 3). The resulting amine would then be subjected to a standard amide coupling reaction whereupon the appropriate acid (see, e.g., Scheme 1, WO 2012/142504 and WO 2012/142513, which are incorporated herein by reference) would be added along with a suitable amide coupling reagent such as PYBOP in a suitable solvent such as DMSO to give the desired amide (Step 4). In Steps 5 and 6, the 2,4-dichloropyridine can be converted into a mixture of 1,2 and 1,4-pyridones by heating in the presence of NaOH in p-methoxy benzyl alcohol as solvent followed by deprotection of the resulting PMB ethers with an appropriate reagent such as TFA in an appropriate solvent such as dichlorome thane. Purification of the resulting pyridone mixture can be accomplished using standard protocols known to ones skilled in the art including, but not limited to, RP-HPLC and preparative TLC. In Step 7, the remaining chlorine is replaced with R 2 by reacting the purified 1,4-pyridone with an alcohol or amine.

[0492] A person of ordinary skill in the art will recognize that in the above schemes the order of many of the steps are interchangeable.

[0493] Compounds of the present invention inhibit the histone methyltransferase activity of EZH2 or a mutant thereof and, accordingly, in one aspect of the invention certain compounds disclosed herein are candidates for treating, or preventing, certain conditions and diseases in which EZH2 plays a role. The present invention provides methods for treating conditions and diseases the course of which can be influenced by modulating the methylation status of histones or other proteins, wherein said methylation status is mediated at least in part by the activity of EZH2. Modulation of the methylation status of histones can in turn influence the level of expression of target genes activated by methylation, and/or target genes suppressed by methylation. The method includes administering to a subject in need of such treatment, a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, polymorph, solvate, or stereoisomeror thereof.

[0494] Unless otherwise stated, any description of a method of treatment includes uses of the compounds to provide such treatment or prophylaxis as is described in the specification, as well as uses of the compounds to prepare a medicament to treat or prevent such condition. The treatment includes treatment of human or non-human animals including rodents and other disease models.

[0495] In still another aspect, this invention relates to a method of modulating the activity of the EZH2, the catalytic subunit of the PRC2 complex which catalyzes the mono- through tri-methylation of lysine 27 on histone H3 (H3-K27) in a subject in need thereof. For example, the method comprises the step of administering to a subject having a cancer expressing a mutant EZH2 a therapeutically effective amount of a compound described herein, wherein the compound(s) inhibits histone methyltransferase activity of EZH2, thereby treating the cancer.

[0496] For example, the EZH2-mediated cancer is selected from the group consisting of follicular lymphoma and diffuse large B-cell lymphoma (DLBCL) of germinal center B cell-like (GCB) subtype. For example, the cancer is lymphoma, leukemia or melanoma. Preferably, the lymphoma is non-Hodgkin's lymphoma (NHL), follicular lymphoma or diffuse large B-cell lymphoma. Alternatively, the leukemia is chronic myelogenous leukemia (CML), acute myeloid leukemia, acute lymphocytic leukemia or mixed lineage leukemia.

[0497] For example, the EZH2-mediated precancerous condition is myelodysplasia syndromes (MDS, formerly known as preleukemia).

[0498] For example, the EZH2-mediated cancer is a hematological cancer.

[0499] The compound(s) of the present invention inhibit the histone methyltransferase activity of EZH2 or a mutant thereof and, accordingly, the present invention also provides methods for treating conditions and diseases the course of which can be influenced by modulating the methylation status of histones or other proteins, wherein said methylation status is mediated at least in part by the activity of EZH2. In one aspect of the invention, certain compounds disclosed herein are candidates for treating, or preventing certain conditions and diseases. Modulation of the methylation status of histones can in turn influence the level of expression of target genes activated by methylation, and/or target genes suppressed by methylation. The method includes administering to a subject in need of such treatment, a therapeutically effective amount of a compound of the present invention.

[0500] As used herein, a "subject" is interchangeable with a "subject in need thereof, both of which refer to a subject having a disorder in which EZH2-mediated protein methylation plays a part, or a subject having an increased risk of developing such disorder relative to the population at large. A "subject" includes a mammal. The mammal can be e.g. , a human or appropriate non-human mammal, such as primate, mouse, rat, dog, cat, cow, horse, goat, camel, sheep or a pig. The subject can also be a bird or fowl. In one embodiment, the mammal is a human. A subject in need thereof can be one who has been previously diagnosed or identified as having cancer or a precancerous condition. A subject in need thereof can also be one who has (e.g., is suffering from) cancer or a precancerous condition. Alternatively, a subject in need thereof can be one who has an increased risk of developing such disorder relative to the population at large (i.e. , a subject who is predisposed to developing such disorder relative to the population at large). A subject in need thereof can have a precancerous condition. A subject in need thereof can have refractory or resistant cancer (i.e., cancer that doesn't respond or hasn't yet responded to treatment). The subject may be resistant at start of treatment or may become resistant during treatment. In some embodiments, the subject in need thereof has cancer recurrence following remission on most recent therapy. In some embodiments, the subject in need thereof received and failed all known effective therapies for cancer treatment. In some embodiments, the subject in need thereof received at least one prior therapy. In a preferred embodiment, the subject has cancer or a cancerous condition. For example, the cancer is lymphoma, leukemia, melanoma, or rhabdomyosarcoma. Preferably, the lymphoma is non- Hodgkin' s lymphoma, follicular lymphoma or diffuse large B-cell lymphoma.

Alternatively, the leukemia is chronic myelogenous leukemia (CML). The precancerous condition is myelodysplasia syndromes (MDS, formerly known as preleukemia).

[0501] As used herein, "treating" or "treat" describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of a compound of the present invention, or a pharmaceutically acceptable salt, polymorph or solvate thereof, to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the disease, condition or disorder. The term "treat" can also include treatment of a cell in vitro or an animal model.

[0502] A compound of the present invention, or a pharmaceutically acceptable salt, polymorph or solvate thereof, can or may also be used to prevent a relevant disease, condition or disorder, or used to identify suitable candidates for such purposes. As used herein, "preventing," "prevent," or "protecting against" describes reducing or eliminating the onset of the symptoms or complications of such disease, condition or disorder.

[0503] Point mutations of the EZH2 gene at a single amino acid residue (e.g., Y641, A677, and A687) of EZH2 have been reported to be linked to lymphoma. More examples of EZH2 mutants and methods of detection of mutation and methods treatment of mutation- associated disorders are described in, e.g., U.S. Patent Application Publication No. US 20130040906, the entire content of which is incorporated herein by reference in its entirety.

[0504] One skilled in the art may refer to general reference texts for detailed descriptions of known techniques discussed herein or equivalent techniques. These texts include Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Inc. (2005);

Sambrook et al, Molecular Cloning, A Laboratory Manual (3 rd edition), Cold Spring Harbor Press, Cold Spring Harbor, New York (2000); Coligan et al., Current Protocols in Immunology, John Wiley & Sons, N.Y.; Enna et al., Current Protocols in Pharmacology, John Wiley & Sons, N.Y.; Fingl et al, The Pharmacological Basis of Therapeutics (1975), Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA, 18 th edition (1990). These texts can, of course, also be referred to in making or using an aspect of the invention.

[0505] As used herein, "combination therapy" or "co-therapy" includes the administration of a compound of the present invention, or a pharmaceutically acceptable salt, polymorph or solvate thereof, and at least a second agent as part of a specific treatment regimen intended to provide the beneficial effect from the co-action of these therapeutic agents. The beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination of therapeutic agents.

[0506] The present invention also provides pharmaceutical compositions comprising a compound of any of the Formulae described herein in combination with at least one pharmaceutically acceptable excipient or carrier. [0507] A "pharmaceutical composition" is a formulation containing the compounds of the present invention in a form suitable for administration to a subject. In one embodiment, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial. The quantity of active ingredient (e.g. , a formulation of the disclosed compound or salt, hydrate, solvate or isomer thereof) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the patient. The dosage will also depend on the route of administration. A variety of routes are

contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In one embodiment, the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that are required.

[0508] As used herein, the phrase "pharmaceutically acceptable" refers to those compounds, anions, cations, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[0509] "Pharmaceutically acceptable excipient" means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use. A "pharmaceutically acceptable excipient" as used in the specification and claims includes both one and more than one such excipient.

[0510] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g. , inhalation), transdermal (topical), and transmucosal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; 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 pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.

[0511] A compound or pharmaceutical composition of the invention can be administered to a subject in many of the well-known methods currently used for chemotherapeutic treatment. For example, for treatment of cancers, a compound of the invention may be injected directly into tumors, injected into the blood stream or body cavities or taken orally or applied through the skin with patches. The dose chosen should be sufficient to constitute effective treatment but not so high as to cause unacceptable side effects. The state of the disease condition (e.g. , cancer, precancer, and the like) and the health of the patient should preferably be closely monitored during and for a reasonable period after treatment.

[0512] The term "therapeutically effective amount", as used herein, refers to an amount of a pharmaceutical agent to treat, ameliorate, or prevent an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect. The effect can be detected by any assay method known in the art. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician. In a preferred aspect, the disease or condition to be treated is cancer. In another aspect, the disease or condition to be treated is a cell proliferative disorder.

[0513] For any compound, the therapeutically effective amount can be estimated initially either in cell culture assays, e.g. , of neoplastic cells, or in animal models, usually rats, mice, rabbits, dogs, or pigs. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. Therapeutic/prophylactic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED 50 (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD 50 /ED 50 . Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.

[0514] Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.

[0515] The pharmaceutical compositions containing active compounds of the present invention may be manufactured in a manner that is generally known, e.g. , by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. Pharmaceutical compositions may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and/or auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically. Of course, the appropriate formulation is dependent upon the route of administration chosen.

[0516] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens,

chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol and sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.

[0517] Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

[0518] Oral compositions generally include an inert diluent or an edible pharmaceutically acceptable carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.

[0519] For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g. , a gas such as carbon dioxide, or a nebulizer.

[0520] Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.

[0521] The active compounds can be prepared with pharmaceutically acceptable carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.

Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova

Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.

[0522] It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.

[0523] In therapeutic applications, the dosages of the pharmaceutical compositions used in accordance with the invention vary depending on the agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy, among other factors affecting the selected dosage. Generally, the dose should be sufficient to result in slowing, and preferably regressing, the growth of the tumors and also preferably causing complete regression of the cancer.

Dosages can range from about 0.01 mg/kg per day to about 5000 mg/kg per day. In preferred aspects, dosages can range from about 1 mg/kg per day to about 1000 mg/kg per day. In an aspect, the dose will be in the range of about 0.1 mg/day to about 50 g/day; about 0.1 mg/day to about 25 g/day; about 0.1 mg/day to about 10 g/day; about 0.1 mg to about 3 g/day; or about 0.1 mg to about 1 g/day, in single, divided, or continuous doses (which dose may be adjusted for the patient's weight in kg, body surface area in m , and age in years). An effective amount of a pharmaceutical agent is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer. For example, regression of a tumor in a patient may be measured with reference to the diameter of a tumor. Decrease in the diameter of a tumor indicates regression. Regression is also indicated by failure of tumors to reoccur after treatment has stopped. As used herein, the term "dosage effective manner" refers to amount of an active compound to produce the desired biological effect in a subject or cell.

[0524] The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.

[0525] The compounds of the present invention are capable of further forming salts. All of these forms are also contemplated within the scope of the claimed invention.

[0526] As used herein, "pharmaceutically acceptable salts" refer to derivatives of the compounds of the present invention wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2-acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, 1,2-ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic, hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxymaleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic,

phenylacetic, phosphoric, polygalacturonic, propionic, salicyclic, stearic, subacetic, succinic, sulfamic, sulfanilic, sulfuric, tannic, tartaric, toluene sulfonic, and the commonly occurring amine acids, e.g. , glycine, alanine, phenylalanine, arginine, etc.

[0527] Other examples of pharmaceutically acceptable salts include hexanoic acid, cyclopentane propionic acid, pyruvic acid, malonic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo- [2.2.2] -oct-2-ene- l -carboxylic acid, 3- phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, muconic acid, and the like. The present invention also encompasses salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g. , an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like. In the salt form, it is understood that the ratio of the compound to the cation or anion of the salt can be 1: 1, or any ration other than 1: 1, e.g., 3: 1, 2: 1, 1:2, or 1:3.

[0528] It should be understood that all references to pharmaceutically acceptable salts include solvent addition forms (solvates) or crystal forms (polymorphs) as defined herein, of the same salt.

[0529] The compounds of the present invention can also be prepared as esters, for example, pharmaceutically acceptable esters. For example, a carboxylic acid function group in a compound can be converted to its corresponding ester, e.g., a methyl, ethyl or other ester. Also, an alcohol group in a compound can be converted to its corresponding ester, e.g., acetate, propionate or other ester.

[0530] The compounds, or pharmaceutically acceptable salts thereof, are administered orally, nasally, transdermally, pulmonary, inhalationally, buccally, sublingually, intraperintoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally and parenterally. In one embodiment, the compound is administered orally. One skilled in the art will recognize the advantages of certain routes of administration.

[0531] The dosage regimen utilizing the compounds is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress of the condition.

[0532] Techniques for formulation and administration of the disclosed compounds of the invention can be found in Remington: the Science and Practice of Pharmacy, 19 th edition, Mack Publishing Co., Easton, PA (1995). In an embodiment, the compounds described herein, and the pharmaceutically acceptable salts thereof, are used in pharmaceutical preparations in combination with a pharmaceutically acceptable carrier or diluent. Suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions. The compounds will be present in such pharmaceutical compositions in amounts sufficient to provide the desired dosage amount in the range described herein.

[0533] All percentages and ratios used herein, unless otherwise indicated, are by weight. Other features and advantages of the present invention are apparent from the different examples. The provided examples illustrate different components and methodology useful in practicing the present invention. The examples do not limit the claimed invention.

Based on the present disclosure the skilled artisan can identify and employ other

components and methodology useful for practicing the present invention.

[0534] In the synthetic schemes described herein, compounds may be drawn with one particular configuration for simplicity. Such particular configurations are not to be construed as limiting the invention to one or another isomer, tautomer, regioisomer or stereoisomer, nor does it exclude mixtures of isomers, tautomers, regioisomers or stereoisomers; however, it will be understood that a given isomer, tautomer, regioisomer or stereoisomer may have a higher level of activity than another isomer, tautomer, regioisomer or stereoisomer.

[0535] Compounds designed, selected and/or optimized by methods described above, once produced, can be characterized using a variety of assays known to those skilled in the art to determine whether the compounds have biological activity. For example, the molecules can be characterized by conventional assays, including but not limited to those assays described below, to determine whether they have a predicted activity, binding activity and/or binding specificity.

[0536] Furthermore, high-throughput screening can be used to speed up analysis using such assays. As a result, it can be possible to rapidly screen the molecules described herein for activity, using techniques known in the art. General methodologies for performing high- throughput screening are described, for example, in Devlin (1998) High Throughput Screening, Marcel Dekker; and U.S. Patent No. 5,763,263. High-throughput assays can use one or more different assay techniques including, but not limited to, those described below.

[0537] All publications and patent documents cited herein are incorporated herein by reference as if each such publication or document was specifically and individually indicated to be incorporated herein by reference. Citation of publications and patent documents is not intended as an admission that any is pertinent prior art, nor does it constitute any admission as to the contents or date of the same. The invention having now been described by way of written description, those of skill in the art will recognize that the invention can be practiced in a variety of embodiments and that the foregoing description and examples below are for purposes of illustration and not limitation of the claims that follow. [0538] Example 1: Synthesis of Compound 1: 5-chloro-N-((2,6-dimethyl-4-oxo-l,4- dihydropyridin-3-yl)methyl)-3-(((trans)-4-(dimethylamino)cyc lohexyl)(ethyl)amino)-2- methylbenzamide

Compound 1

[0539] Step 1: Synthesis of 2,6-dimethyl-4-oxo-l,4-dihydropyridine-3-carbonitrile:

[0540] A mixture of 2,2,6-trimethyl-4H-l,3-dioxin-4-one (5 g, 38.46 mmol) and (E)-3- aminobut-2-enenitrile (4.73 g, 57.69 mmol) was heated at 120 °C for 1 h. On completion, the reaction mixture was cooled to room temperature and ethyl acetate was added, the solid precipitated was filtered and dried under vacuum giving 2,6-dimethyl-4-oxo-l,4- dihydropyridine-3-carbonitrile (1.7g, 30.4%).

[0541] Step 2: Synthesis of 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one:

[0542] To a solution of 2,6-dimethyl-4-oxo-l,4-dihydropyridine-3-carbonitrile (1.7g, 11.48 mmol) in methanol and aq. ammonia solution (9: 1), was added a catalytic amount of Raney Nickel. The combined reaction mixture was stirred at room temperature under hydrogen (balloon pressure) for 2-5 h. On completion of reaction, the mixture was filtered through celite bed and the filtrate was concentrated under reduce pressure to afford the title amine (1.8 g, 31%).

[0543] Step 3: Synthesis of 5-chloro-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methylbenzamide:

[0544] To 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amin o)-2- methylbenzoic acid (WO 2012142513, 0.3g, 0.88 mmol) dissolved in DMSO (3 mL) was added 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one (0.27g, 1.77 mmol). The reaction mixture was stirred at room temperature for 15 min at which time PYBOP (0.69 g, 1.33) and triethylamine (0.18g, 1.77 mmol) were added and the resulting reaction mixture was stirred overnight. After completion of the reaction, the solution was poured into ice and extracted with 10 % MeOH/DCM. The combined organic layers were dried and concentrated to obtain crude; which then purified by prep. HPLC to afford the title compound as the trifluoroacetic acid salt (0.07 g, 16.7%).

[0545] LCMS: 473.35 (M+l) + ; HPLC: 98.48% (@ 210 nm-370 nm) 1H NMR (DMSO-d 6 , 400 MHz) δ 13.85 (brs, 1H), 9.50 (brs, 1H), 8.65 (t, 1H), 7.23 (s, 1H), 6.99 (s, 1H), 6.95 (s, 1H), 4.39 (d, 2H, J=4.8 Hz), 3.17-3.12 (m, 1H), 3.03-3.02 (m, 2H), 2.69-2.68 (m, 6H), 2.67 (m, 4H), 2.53 (s, 3H), 2.14 (s, 3H), 1.95-1.83 (m, 4H), 1.45-1.40 (m, 4H) 0.79 (t, 3H, J=6.8 Hz).

[0546] Example 2: Synthesis of Compound 2: N-((2,6-dimethyl-4-oxo-l,4- dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-y l)amino)-4-methyl-4'- (morpholinomethyl)-[ 1 , 1 '-b

Compound 2

[0547] Step 1: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-5-

(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morph olinomethyl)-[l,l'-biphenyl]-3- carboxamide

[0548] To 5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpho linomethyl)- [l,r-biphenyl]-3-carboxylic acid (WO 2012142504, 0.3 g, 0.684 mmol) dissolved in DMSO (3 mL) was added 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one (Compound 1, Step 2, 0.208 g, 1.36 mmol) and triethylamine (0.138 g, 1.36 mmol). The reaction mixture was stirred at room temperature for 15 min before PYBOP (0.534 g, 1.02 mmol) was added to it and stirring was continued overnight. After completion of the reaction, the solution was poured into ice and extracted with 10 % MeOH/DCM. The combined organic layers were dried and concentrated to obtain crude material which was then purified by prep. HPLC to afford the title compound as the trifluoroacetic acid salt (0.07 g, 17.9 %).

[0549] LCMS: 573.55 (M+l) + ; HPLC: 89.28% (@ 210 nm-370 nm) 1H NMR (DMSO-J 6 , 400 MHz) δ 13.94 (brs, 1H), 10.17 (brs, 1H), 8.70 (t, 1H), 7.75 (d, 2H, J=7.6 Hz), 7.57 (d, 2H, J=8.0 Hz), 7.48 (s, 1H), 7.30 (s, 1H), 6.99 (s, 1H), 4.43-4.39 (m, 4H), 3.96-3.83 (m, 4H), 3.64 (m, 2H), 3.27-3.22 (m, 4H), 3.13 (m, 4H), 3.02-3.01 (m, 1H), 2.71 (s, 3H), 2.54 (s, 3H), 2.23 (s, 3H), 1.73-1.65 (m, 2H), 1.55-1.53 (m, 2H), 0.83 (t, 3H, J=7.2 Hz).

[0550] Example 3: Synthesis of Compound 3: 5-chloro-N-((4-chloro-6-methyl-2-oxo-l,2- dihydropyridin-3-yl)methyl)-3-(((trans)-4-(dimethylamino)cyc lohexyl)(ethyl)amino)-2- methylbenzamide

Compound 3

[0551] Step 1: Synthesis of 4-hydroxy-6-methyl-2-oxo-l,2-dihydropyridine-3-carbonitrile:

[0552] A suspension of 2-amino-6-methyl-4-oxo-4H-pyran-3-carbonitrile (3 g, 20 mmol) in 10% HC1 (45 mL) was heated under reflux for 4 h. The precipitate was collected by filtration and washed with water and then recrystallized from MeOH to give the desired product (2.1 g, 70 %).

[0553] Step 2: Synthesis of 2,4-dichloro-6-methylnicotinonitrile:

[0554] A stirring mixture of 4-hydroxy-6-methyl-2-oxo-l,2-dihydropyridine-3-carbonitrile (lg, 6.66 mmol) and POCl 3 (2.04 g, 13.33 mmol) in DMF was refluxed for 6 h. After completion, the solution was concentrated to dryness. The residue was then basified with sat. sodium bicarbonate solution to pH 8 and extracted with 10% MeOH/DCM. The combined organic phases were dried over Na 2 S0 4 and the solvent was removed under reduced pressure to afford the crude product that was used in the next step without further purification (0.85 g, 68.5%).

[0555] Step 3: Synthesis of (2,4-dichloro-6-methylpyridin-3-yl)methanamine:

[0556] To a stirred solution of 2,4-dichloro-6-methylnicotinonitrile (0.85 g, 4.54 mmol) in DCM (10 mL) at -78 °C was added DIBAL solution (23 mL, 22.72 mmol) and the resulting mixture was stirred at room temperature for 12 h. On completion, reaction was quenched with sat. NH 4 C1 solution and extracted with 10% MeOH/DCM, Combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to give desired compound that was used in the subsequent step without further purification (l.lg, 90.9 %).

[0557] Step 4: Synthesis of 5-chloro-N-((2,4-dichloro-6-methylpyridin-3-yl)methyl)-3- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl benzamide:

[0558] To 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amin o)-2- methylbenzoic acid (WO 2012142513, 0.3 g, 0.89 mmol) dissolved in DMSO was added (2,4-dichloro-6-methylpyridin-3-yl)methanamine (0.339 g, 1.78 mmol). The reaction mixture was stirred at room temperature for 15 min at which time PYBOP (0.695 g, 1.33 mmol) and triethyl amine (0.089 g, 0.881 mmol) were added and stirring was continued overnight. After completion of the reaction, the solution was poured into ice and extracted with 10 % MeOH/DCM. The combined organic layers were dried and concentrated to obtain crude product, which was purified by column chromatography to afford the title compound (0.25 g, 55.1%).

[0559] LCMS: 511.35 (M+l) + ; HPLC: 90.27% (@ 210 nm-370 nm) 1H NMR (DMSO-J 6 , 400 MHz) δ 8.66 (t, 1H), 7.53 (s, 1H), 7.22 (s, 1H), 7.00 (s, 1H), 4.61 (d, 2H, J=4.0 Hz), 3.03-2.99 (m, 3H), 2.66 (s, 6H), 2.54 (m, 1H), 2.45 (s, 3H), 2.18 (s, 3H), 1.94-1.83 (m, 4H), 1.42 (m, 4H), 0.79 (t, 3H, J=6.8 Hz).

[0560] Step 5: Synthesis of 5-chloro-N-((4-chloro-6-methyl-2-oxo-l,2-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methylbenzamide:

[0561] A mixture of 5-chloro-N-((2,4-dichloro-6-methylpyridin-3-yl)methyl)-3-((( trans)-4- (dimethylamino)cyclohexyl)(ethyl)amino)-2-methylbenzamide (0.22 g, 0.431 mmol) and NaOH (0.086g, 2.16 mmol) in p-methoxy benzyl alcohol (3 mL) were heated at 80 °C for 12 h. On completion, the solution was concentrated to dryness and the crude material obtained was purified by column chromatography to give the PMB protected compound (0.15 g, 56.7%).

[0562] To a stirred solution of PMB protected compound (0.15 g, 0.24 mmol) in DCM (4 mL) at 0 °C was added TFA (2 mL) and the resulting reaction mixture was stirred for 12 h at room temperature. After completion, the solution was concentrated to dryness. The residue was then basified with aqueous sat. bicarbonate solution to pH 8 and the aqueous layer was extracted with 20% methanol in DCM. The combined organic layers were dried over Na 2 S0 4 and solvent was removed under reduced pressure, the crude material obtained was purified by prep. HPLC to give the desired product (0.03 g, 24.9%).

[0563] LCMS: 493.30 (M+l) + ; HPLC: 93.49% (@ 210 nm-370 nm) 1H NMR (DMSO-J 6 , 400 MHz) δ 11.11 (brs, 1H), 9.33 (brs, 1H), 8.51 (t, 1H), 7.21 (s, 1H), 6.99 (s, 1H), 6.69 (s, 1H), 4.43 (d, 2H, J=4.0 Hz), 3.17-3.11 (m, 1H), 3.03-3.02 (m, 2H), 2.69-2.68 (m, 6H), 2.31 (s, 3H), 2.17 (s, 3H), 1.95-1.84 (m, 4H), 1.52-1.38 (m, 4H), 0.79 (t, 3H, J=6.8 Hz), 1H merged in solvent peak.

[0564] Example 4: Synthesis of Compound 4: N-((2-chloro-6-methyl-4-oxo-l,4- dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-y l)amino)-4-methyl-4'- (morpholinomethyl)-[ 1 , 1 '-b

Compound 4

[0565] Step 1: Synthesis of N-((2,4-dichloro-6-methylpyridin-3-yl)methyl)-5-

(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morph olinomethyl)-[l,l'-biphenyl]-3- carboxamide:

[0566] To 5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpho linomethyl)- [l,l'-biphenyl]-3-carboxylic acid (WO 2012142504, 0.3 g, 0.684 mmol) dissolved in DMSO was added (2,4-dichloro-6-methylpyridin-3-yl)methanamine (Compound 3, Step 3, 0.261 g, 1.369 mmol. The reaction mixture was stirred at room temperature for 15 min at which time PYBOP (0.534 g, 1.03 mmol) and triethylamine (0.138 g, 1.37 mmol) were added and stirring was continued overnight. After completion of the reaction, the solution was poured into ice, extracted with 10 % MeOH/DCM. The combined organic layers were dried and concentrated to afford the crude compound that was used in the subsequent step without further purification (0.4 g, 95.0%).

[0567] LCMS: 611.45 (M+l) + ; HPLC: 93.03% (@ 210 nm-370 nm) 1H NMR (DMSO-J 6 , 400 MHz) δ 10.02 (bs, 1H), 8.65 (t, 1H), 7.74 (m, 2H), 7.59-7.47 (m, 4H), 7.31 (s, 1H), 4.65 (d, 2H, J=4.4 Hz), 4.40 (m, 2H), 3.85-3.83 (m, 2H), 3.65-3.54 (m, 4H), 3.28-3.22 (m, 4H), 3.17-3.13 (m, 5H), 2.44 (s, 3H), 2.29 (s, 3H ), 1.66-1.54 (m, 4H), 0.84 (t, 3H, J=6.8 Hz).

[0568] Step 2: Synthesis of N-((4-chloro-6-methyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)- 5-(ethyl-(tetrahydro-2H-pyran-4-yl)-amino)-4-methyl-4'-(morp holinomethyl)-[l,l'- biphenyl] -3-carboxamide. [0569] A mixture of N-((2,4-dichloro-6-methylpyridin-3-yl)methyl)-5-(ethyl(tetra hydro- 2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[l,l'-bi phenyl]-3-carboxamide (0.2 g, 0.327 mmol) and NaOH (0.053 g, 1.31 mmol) in p-methoxy benzyl chloride (2 mL) was heated at 80 °C for 12 h. Upon completion, the solution was concentrated to dryness and the crude material obtained was purified by column chromatography to give the title PMB protected compound (0.2 g, 83.7%).

[0570] To a stirred solution of PMB protected compound (0.2 g, 0.274 mmol) in DCM (4 mL) at 0 °C was added TFA (2 mL) and the resulting reaction mixture was stirred for 12 h at room temperature. After completion, the reaction was concentrated to dryness and the residue was then basified with aqueous sat. bicarbonate solution to pH 8 and the aqueous layer was extracted with 20% methanol in DCM. The combined organic layers were dried over Na 2 S0 4 and solvent was removed under reduced pressure, the crude material obtained was purified by prep. HPLC to give N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl -4'-(morpholinomethyl)- [l,r-biphenyl]-3-carboxamide (0.09 g, 55.6%).

[0571] LCMS: 297.15 (M/2+l) + ; HPLC: 94.76% (@ 210 nm-370 nm) 1H NMR (DMSO- d 6 , 400 MHz) δ 11.14 (bs, 1H), 9.85 (bs, 1H), 8.84 (t, 1H), 7.80-7.75 (m, 2H), 7.59-7.57 (m, 2H), 7.47-7.46 (m, 1H), 7.32-7.29 (m, 1H), 5.72 (s, 1H), 4.46 (d, 2H, J=4.4 Hz), 4.39 (m, 2H), 4.24-4.22 (m, 2H), 3.66-3.56 (m, 4H), 3.31-3.23 (m, 4H), 3.17-3.03 (m, 5H), 2.31 (s, 3H), 2.09 (s, 3H), 1.65 (m, 2H), 1.56-1.53 (m, 2H), 0.84 (t, 3H, J=6.8 Hz).

[0572] Example 5: Synthesis of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl) amino)-2-methyl-N-(( 1 ,2,6-trimethyl-4-oxo- 1 ,4-dihydropyridin-3-yl)methyl)benzamide:

Compound 86

[0573] Step 1: Synthesis of 3-(aminomethyl)-l,2,6-trimethylpyridin-4(lH)-one: [0574] To a solution of 2,6-dimethyl-4-oxo-l,4-dihydropyridine-3-carbonitrile (0.4 g, 2.46 mmol) in methanol (10 mL) was added a catalytic amount of Raney Nickel in ammonia solution (3 mL). The reaction mixture was stirred at room temperature under hydrogen (1 atm) for 3 h. On completion, the reaction mixture was filtered through celite and washed with methanol and the filtrate was concentrated under reduced pressure to afford the title compound (0.3 g, 73.2%).

[0575] Step 2: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-N-((l,2, 6-trimethyl-4-oxo-l,4- dihydropyridin- 3 - yl)methyl)benzamide :

[0576] To a solution of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amin o)-2- methylbenzoic acid (0.3 g, 0.887 mmol) in DMSO (3 mL), 3-(aminomethyl)- 1,2,6- trimethylpyridin-4(lH)-one (0.295 g, 1.77 mmol) and triethyl amine (0.180 g, 1.78 mmol) were added. The reaction mixture was stirred at room temperature for 15 min before PyBOP (0.692 g, 1.33 mmol) was added at 0 °C and then the resulting reaction mixture was stirred at room temperature overnight. After completion, the reaction was quenched with ice and extracted with 10% MeOH/DCM. The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure to obtain crude material which was then purified by preparative HPLC to afford desired compound (0.11 g, 25.5%).

[0577] LCMS: 487.30 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 9.54 (brs, 1H), 8.46 (t, 1H, J=4.8 Hz), 7.22 (s, 1H), 7.00 (s, 1H), 6.17 (s, 1H), 4.28 (d, 2H, J=4.8 Hz), 3.43 (s, 3H), 3.12 (m, 1H), 3.03 (q, 2H, J=7.2 Hz), 2.69-2.68 (m, 7H), 2.39 (s, 3H), 2.21 (s, 3H), 2.16 (s, 3H), 1.95 (m, 2H), 1.83 (m, 2H), 1.48-1.41 (m, 4H), 0.79 (t, 3H, J=6.8 Hz).

[0578] Example 6: Synthesis of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl) amino)-N-((l-ethyl-2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-y l)methyl)-2- methylbenzamide:

Compound 87

[0579] Step 1: Synthesis of 3-(aminomethyl)-l-ethyl-2,6-dimethylpyridin-4(lH)-one:

[0580] To a solution of l-ethyl-2,6-dimethyl-4-oxo-l,4-dihydropyridine-3-carbonitril e (0.4 g, 2.27 mmol) in methanol (10 mL) was added a catalytic amount of Raney Nickel and ammonia solution (3 mL). The reaction mixture was stirred at room temperature under hydrogen (1 atm) for 3 h. Upon completion, the reaction mixture was filtered through celite, washed with methanol and the filtrate was concentrated under reduced pressure to afford the title compound (0.3 g, 73.3%).

[0581] Step 2: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((l-ethyl-2,6-d imethyl-4-oxo-l,4- dihydropyridin- 3 - yl)methyl) -2-methylbenzamide :

[0582] To a solution of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amin o)-2- methylbenzoic acid (0.28 g, 0.83 mmol) in DMSO (3 mL), 3-(aminomethyl)-l-ethyl-2,6- dimethylpyridin-4(lH)-one (0.298 g, 1.66 mmol) and triethyl amine (0.167 g, 1.65 mmol) were added. The reaction mixture was stirred at room temperature for 15 min before PyBOP (0.646 g, 1.24 mmol) was added at 0 °C and stirring was continued at room temperature overnight. After completion, the reaction was quenched with ice and extracted with 10% MeOH/DCM. The combined organic layers were dried over sodium sulphate and

concentrated under reduced pressure to obtain crude material which was then purified by preparative HPLC to afford the title compound (0.04 g, 9.7 %).

[0583] LCMS: 501.30 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 9.47 (brs, IH), 8.64 (t, IH, J=5.2 Hz), 7.45 (s, IH), 6.23 (s, IH), 6.98 (s, IH), 4.42 (d, 2H, J=4.8 Hz), 4.37 (q, 2H, J=7.2 Hz), 3.11 (m, IH), 3.03 (q, 2H, J=6.8 Hz), 2.72 (s, 3H), 2.69-2.68 (m, 7H), 2.63 (s, 3H), 2.13 (s, 3H), 1.95 (m, 2H), 1.82 (m, 2H), 1.44-1.40 (m, 7H), 0.78 (t, 3H, J=6.8 Hz). [0584] Example 7: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-N-((6-me thyl-2-(methylamino)-4-oxo- l,4-dihydropyridin-3-yl)methyl)benzamide:

Compound 18

[0585] Step 1: Synthesis of 4-((4-methoxybenzyl)oxy)-6-methyl-2- (methylamino)nicotinonitrile:

[0586] To a stirred solution of 2-chloro-4-((4-methoxybenzyl)oxy)-6-methylnicotinonitrile (0.4 g, 1.38 mmol)) in DIPEA (0.358 g, 2.77 mmol) was added methyl amine (4 mL). The resulting reaction mass was stirred at 120 °C for 12h. The progress of the reaction was monitored by TLC & LCMS. After consumption of the starting material, the reaction mixture was concentrated to dryness. The residue obtained was diluted with water and precipitated solid was collected by filtration and dried under reduced pressure to obtain crude material which then purified by column chromatography to afford unseparable mixture of the title compound and 6-methyl-2,4-bis(methylamino)nicotinonitrile (0.35 g).

[0587] Step 2: Synthesis 3-(aminomethyl)-4-((4-methoxybenzyl)oxy)-N,6- dimethylpyridin-2- amine :

[0588] To a solution of crude 4-((4-methoxybenzyl)oxy)-6-methyl-2- (methylamino)nicotinonitrile (0.35 g, 1.98 mmol) in methanol (5 mL), was added a catalytic amount of Raney Nickel in ammonia solution (2 mL). The reaction mass was stirred at room temperature under hydrogen (1 atm) for 3-4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was filtered through celite, washed with methanol and the filtrate was concentrated under reduced pressure to afford the crude title compound that was used in the subsequent reaction without further purification (0.32 g).

Ill [0589] Step 3: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((4-((4-methoxy benzyl)oxy)-6-methyl-2-

(methylamino)pyridin-3-yl)methyl)-2-methylbenzamide:

[0590] To a stirred solution of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methylbenzoic acid (0.3 g, 0.88 mmol) in DMSO (3 mL), was added 3-(aminomethyl)-4-((4-methoxybenzyl)oxy)-N,6- dimethylpyridin-2-amine (0.32 g, 1.77 mmol) and triethyl amine (0.18 g, 1.77 mmol). The reaction mixture was stirred at room temperature for 10 min before PyBOP (0.692 g, 1.33 mmol) was added at 0 °C and stirring was continued for 12 h at room temperature. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was diluted with ice water and extracted with 10% MeOH/DCM. The combined organic layers were washed with brine, dried over sodium sulphate and concentrated under reduced pressure to obtain crude material which then purified by column chromatography to afford the crude title compound which was used in the subsequent reaction without further purification (0.35 g).

[0591] Step 4: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-N-((6-me thyl-2-(methylamino)-4-oxo- l,4-dihydropyridin-3-yl)methyl)benzamide:

[0592] To a stirred solution of crude 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((4-((4-methoxy benzyl)oxy)-6-methyl-2- (methylamino)pyridin-3-yl)methyl)-2-methylbenzamide (0.35 g, 0.575 mmol) in DCM (5 mL) cooled to 0 °C was added TFA (2 mL). The reaction mass was stirred at room temperature for 2 h. Upon completion, the reaction was concentrated to dryness. The residue was basified with aqueous sodium bicarbonate to pH 8 and the aqueous layer was extracted with 20% MeOH/DCM. The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to afford crude material which was then purified by prep HPLC to afford the title compound (0.025 g, 8.9%).

[0593] LCMS: 488.30 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 12.37 (brs, IH), 11.68 (s, IH), 9.50 (brs, IH), 8.86 (t, IH), 8.07 (brs, IH), 7.25 (s, IH), 7.06 (s, IH), 6.35 (s, IH), 4.32 (d, 2H, J=5.6 Hz), 3.12 (m, IH), 3.06 (m, 3H), 3.03 (d, 2H, J=4.0 Hz), 2.69-2.68 (m, 7H), 2.42 (s, 3H), 2.16 (s, 3H), 1.95 (m, 2H), 1.83 (m, 2H), 1.48-1.41 (m, 4H), 0.79 (t, 3H, J=6.8 Hz). [0594] Example 8: Synthesis of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl) amino)-N-((2-(ethylamino)-6-methyl-4-oxo-l,4-dihydropyridin- 3-yl)methyl)-2- methylbenzamide:

Compound 19

[0595] Step 1: Synthesis of 2-(ethylamino)-4-((4-methoxybenzyl)oxy)-6- methylnicotinonitrile :

[0596] To a stirred solution of 2-chloro-4-((4-methoxybenzyl)oxy)-6-methylnicotinonitrile (0.3 g, 1.04 mmol) in DIPEA (0.269 g, 2.08 mmol), ethyl amine (2M, 0.235 g, 5.20 mmol) was added. Resulting reaction mass was stirred at 80 °C for 12h. Upon completion, the reaction mixture was concentrated to dryness. The residue obtained was diluted with water and extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude material was purified by silica gel column chromatography to afford the title compound (0.06 g, 19.4%)

[0597] Step 2: Synthesis of 3-(aminomethyl)-N-ethyl-4-((4-methoxybenzyl)oxy)-6- methylpyridin-2- amine :

[0598] To a solution of 2-(ethylamino)-4-((4-methoxybenzyl) oxy)-6-methylnicotinonitrile (0.35 g, 1.17 mmol) in methanol (5 mL), was added a catalytic amount of Raney Nickel in ammonia solution (2 mL). The reaction mass was stirred at room temperature under (1 atm) for 3-4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was filtered through celite, washed with methanol and the filtrate was concentrated under reduced pressure to afford the title compound (0.325 g, 91.5%).

[0599] Step 3: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((2-(ethylamino )-4-((4-methoxybenzyl)oxy)- 6-methylpyridin-3-yl)-methyl)-2-methylbenzamide: [0600] To a stirred solution of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methylbenzoic acid (0.3 g, 0.88 mmol) in DMSO (3 mL), 3-(aminomethyl)-N-ethyl-4-((4-methoxybenzyl)oxy)-6-methylpyr idin-2- amine (0.32 g, 1.06 mmol) and triethyl amine (0.18 g, 1.77 mmol) were added. The reaction mixture was stirred at room temperature for 10 min before PyBOP (0.692 g, 1.33 mmol) was added to it at 0 °C and stirring was continued for 12 h at room temperature. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was diluted with ice water and extracted with 10% MeOH/DCM. The combined organic layers were washed with brine, dried over sodium sulphate and concentrated under reduced pressure to obtain crude material which then purified by column chromatography to afford the title compound (0.3 g, 54.4%).

[0601] Step 4: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((2-(ethylamino )-6-methyl-4-oxo-l,4- dihydropyridin- 3 - yl)methyl) -2-methylbenzamide :

[0602] A solution of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amin o)-N- ((2-(ethylamino)-4-((4-methoxybenzyl)oxy)-6-methylpyridin-3- yl)-methyl)-2- methylbenzamide (0.3 g, 0.48 mmol) in DCM (5 mL) was cooled to 0 °C and TFA (2 mL) was added to it. The reaction mass was stirred at room temperature for 2 h. Upon completion, the reaction was concentrated to dryness. The residue obtained was treated with aqueous sodium bicarbonate solution and extracted with 20% MeOH/DCM. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to afford crude material which then purified by preparative HPLC to afford the title compound (0.125 g, 51.7%).

[0603] LCMS: 502.35 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 12.47 (brs, 1H), 11.72 (brs, 1H), 9.56 (brs, 1H), 9.04 (t, 1H), 8.05 (brs, 1H), 7.25 (s, 1H), 7.07 (s, 1H), 6.35 (s, 1H), 4.35 (d, 2H, J=5.6 Hz), 3.45 (q, 2H, J=6.8 Hz), 3.17-3.12 (m, 2H), 3.04-3.01 (m, 2H), 2.69-2.68 (m, 6H), 2.41 (s, 3H), 2.17 (s, 3H), 1.95 (m, 2H), 1.83 (m, 2H), 1.46-1.41 (m, 4H), 1.26 (t, 3H, J=7.6 Hz), 0.78 (t, 3H, J=6.8 Hz).

[0604] Example 9: Synthesis of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl) amino)-N-((2-methoxy-6-methyl-4-oxo-l,4-dihydropyridin-3-yl) methyl)-2- methylbenzamide

Compound 15

[0605] Step 1: Synthesis of 2-methoxy-4-((4-methoxybenzyl)oxy)-6-methylnicotinonitrile [0606] To a stirred solution of 2-chloro-4-((4-methoxybenzyl)oxy)-6-methylnicotinonitrile (0.4 g, 1.38 mmol)) in methanol (5 mL) at 0 °C, sodium methoxide (0.226 g, 4.16 mmol) was added and stirred at room temperature for 12h and the further heated at 60 °C for 2h. The progress of the reaction was monitored by TLC & LCMS. After consumption of the starting material, the reaction mixture was concentrated to dryness. The residue obtained was diluted with water and extracted with 10% MeOH/DCM. The combined organic layers were washed brine, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford an unseparable mixture of the title compound and 2,4-dimethoxy-6-methylnicotinonitrile (0.245 g)

[0607] Step 2: Synthesis of 2-methoxy-4-((4-methoxybenzyl)oxy)-6-methylpyridin-3- yl)methanamine

[0608] To a solution of crude 2-methoxy-4-((4-methoxybenzyl)oxy)-6- methylnicotinonitrile (0.35 g, 1.96 mmol) in methanol (5 mL), was added a catalytic amount of Raney Nickel in ammonia solution (2 mL). The reaction mass was stirred at room temperature under hydrogen (1 atm) for 4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was filtered through celite, washed with methanol and the filtrate was concentrated under reduced pressure to afford the crude title compound that was used without further purification (0.3 g).

[0609] Step 3: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((2-methoxy-4-( (4-methoxybenzyl)oxy)-6- methylpyridin-3-yl)methyl)-2-methylbenzamide [0610] To a stirred solution of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methylbenzoic acid (0.3 g, 0.88 mmol) in DMSO (3 mL), was added 2-methoxy-4-((4-methoxybenzyl)oxy)-6-methylpyridin-3- yl)methanamine (0.3 g, 1.64 mmol) and triethyl amine (0.18 g, 1.77 mmol). The reaction mixture was stirred at room temperature for 15 min before PyBOP (0.692 g, 1.33 mmol) was added at 0 °C and stirring was continued at room temperature overnight. After completion, the reaction was diluted with ice water and extracted with 10% MeOH/DCM. The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure to obtain crude material which was then purified by column

chromatography to afford the crude title compound that was used in the next reaction without further purification (0.3 g).

[0611] Step 4: Synthesis of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl) amino)-N-((2-methoxy-6-methyl-4-oxo-l,4-dihydropyridin-3-yl) methyl)-2- methylbenzamide

[0612] To a stirred solution of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-N-((2-methoxy-4-( (4-methoxybenzyl)oxy)-6- methylpyridin-3-yl)methyl)-2-methylbenzamide (0.3 g, 0.49 mmol) in DCM (5 mL) cooled to 0 °C was added TFA (2 mL). The reaction mass was stirred at room temperature for 2 h. Upon completion, the reaction was concentrated to dryness. The obtained residue was basified with aqueous sodium bicarbonate to pH 8 and the aqueous layer was extracted with 20% MeOH/DCM. The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to afford crude material which was then purified by prep HPLC to afford desired compound (0.02 g, 8.29%).

[0613] LCMS: 489.20 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 10.50 (brs, 1H), 9.46 (brs, 1H), 8.42 (t, 1H), 7.20 (s, 1H), 6.97 (s, 1H), 6.37 (s, 1H), 4.30 (d, 2H, J=4.4 Hz), 3.82 (s, 3H), 3.11-3.05 (m, 1H), 3.03 (q, 2H, J=7.2 Hz), 2.69-2.68 (m, 7H), 2.26 (s, 3H), 2.15 (s, 3H), 1.95 (m, 2H), 1.84 (m, 2H), 1.44-1.40 (m, 4H), 0.79 (t, 3H, J=6.8 Hz).

[0614] Example 10: Synthesis of 5-chloro-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methyl-N-((6-methyl-4-oxo-2-(pyrrolidin-l-yl )-l,4-dihydropyridin-3- yl)methyl)benzamide:

Compound 26

[0615] Step 1: Synthesis of 4-methoxy-6-methyl-2-(pyrrolidin-l-yl)nicotinonitrile

[0616] To a stirred solution of 2-chloro-4-methoxy-6-methylnicotinonitrile (0.66 g, 3.63 mmol) in acetonitrile (10 mL), DIPEA (0.936 g, 7.26 mmol) and pyrrolidine (0.515 g, 7.26 mmol) were added. The reaction mixture was stirred at 80 °C for 5 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was concentrated to dryness and the residue obtained was diluted with water. The precipitated solid was collected by filtration and dried under reduced pressure to afford the title compound (0.75 g,

95%).

[0617] Step 2: Synthesis of 4-hydroxy-6-methyl-2-(pyrrolidin-l-yl)nicotinonitrile

To a stirred solution of 4-methoxy-6-methyl-2-(pyrrolidin-l-yl)nicotinonitrile (0.75 g, 3.45 mmol), HBr in acetic acid (10 mL) was added and the reaction mixture was stirred at 100 °C for 14 h. The progress of the reaction was monitored by TLC. Upon completion, the precipitated solid was collected by filtration, washed with water and dried under reduced pressure to afford the title compound (0.6 g., 85%).

[0618] Step 3: Synthesis of 3-(amino methyl)-6-methyl-2-(pyrrolidin-l-yl)pyridin-4-ol [0619] To a stirred solution of 4-hydroxy-6-methyl-2-(pyrrolidin-l-yl)nicotinonitrile (0.5 g, 2.46 mmol) in methanol (5 mL), catalytic amount of Raney Nickel and ammonia solution (2 mL) were added. The reaction mass was stirred at rt under hydrogen (1 atm) for 4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was filtered through a bed of celite and washed with methanol. The filtrate was concentrated under reduced pressure to afford the title compound (0.4 g) which was used in the subsequent step without further purification. [0620] Step 4: Synthesis of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-N-((6-me thyl-4-oxo-2-(pyrrolidin-l- yl)- 1 ,4-dihydropyridin-3-yl)methyl)benzamide

[0621] To a stirred solution of 5-chloro-3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methylbenzoic acid (0.325 g, 0.961 mmol) in DMSO (3 mL), 3-(aminomethyl)-6-methyl-2-(pyrrolidin-l-yl)pyridin-4-ol (0.398 g, 1.92 mmol) and triethyl amine (0.2 mL, 1.44 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (0.75 g, 1.44 mmol) was added to it and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was diluted with water and extracted with 10%

MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by prep HPLC to afford the title compound (0.025 g, 4.9%).

[0622] LCMS: 528.50 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 11.23 (brs, 1H), 9.71 (brs, 1H), 8.50 (t, 1H), 7.24 (s, 1H), 6.99 (s, 1H), 6.45 (s, 1H), 4.44 (d, 2H), 3.71-3.68 (m, 4H), 3.17-3.03 (m, 4H), 2.69-2.67 (m, 6H), 2.43 (s, 3H), 2.17 (s, 3H), 1.98-1.95 (m, 4H), 1.85- 1.82 (m, 4H), 1.45-1.42 (m, 4H), 0.79 (t, J=6.8 Hz, 3H).

[0623] Example 11: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3-yl) methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino )-5-(6-

(methoxymethyl)pyridin-3-yl)- -methylbenzamide:

Compound 88

[0624] Step 1: Synthesis of 5-bromo-2-(methoxymethyl)pyridine: [0625] To a stirred solution of (5-bromopyridin-2-yl)methanol (2 g, 10.63 mmol) in dry THF (10 mL) at 0 °C, was added NaH (60%, 0.851 g, 21.27 mmol) and the resulting solution was stirred at 0 °C for 15 min. Methyliodide (1.52 g, 10.63 mmol) was added at 0 °C and the resulting reaction mixture was stirred at rt for 1 h. The progress of the reaction was monitored by TLC. After complete consumption of starting material, the reaction mixture was quenched with ice and extracted with ethyl acetate. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (1 g, 47.2%).

[0626] Step 2: Synthesis of (6-(methoxymethyl)-pyridin-3-yl)boronic acid:

To a stirred solution of 5-bromo-2-(methoxymethyl) pyridine (1 g, 5 mmol) in 1,4-dioxane (10 mL), bis(pinacolato)diboron (1.52 g, 6 mmol) and potassium acetate (0.735 g, 7.5 mmol) were added and the reaction flask was purged with argon for 10 min. Pd 2 (dba) 3 (0.228 g, 0.25 mmol) and tricyclohexyl phosphine (0.182 g, 0.65 mmol) were added and the reaction flask was again purged with argon for 10 min. The reaction mass was heated at 80 °C for 8 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (0.6 g, 71.9%).

[0627] Step 3: Synthesis of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-5-(6-(methoxymeth yl)pyridin-3-yl)-2- methylbenzoate:

[0628] To a stirred solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methylbenzoate (1 g, 2.51 mmol) and (6-(methoxymethyl)-pyridin-3- yl)boronic acid (0.462 g, 2.77 mmol) in dioxane/water mixture (9/3 mL), Na 2 C0 3 (0.961 g, 9.06 mmol) was added and the solution was purged with argon for 15 min. Pd (PPh 3 ) 4 (0.29 g, 0.251 mmol) was added and the reaction flask was again purged with argon for 15 min. The reaction mass was heated at 100 °C for 4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and

concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.8 g, 72.7%). [0629] Step 4: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-

3- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-5-(6-(me thoxymethyl)pyridin-3-yl)- 2-methylbenzamide :

[0630] Aqueous NaOH (0.109 g, 2.73 mmol) was added to a solution of methyl 3-(((trans)-

4- (dimethylamino)cyclohexyl)(ethyl)amino)-5-(6-(methoxymethyl) pyridin-3-yl)-2- methylbenzoate (0.8 g, 1.82 mmol) in EtOH (8 mL) and stirred at 60 °C for 3 h. After completion of the reaction, the solvent was removed under reduced pressure and the resulting oil was acidified first using dilute HC1 and then adjusted to pH 4 with citric acid. Extraction was carried out using MeOH:DCM (10:90). The combined organic layers were dried concentrated giving respective acid (0.5 g, 64.6%).

[0631] To the acid (0.3 g, 0.705 mmol) dissolved in DMSO (3 mL) was added 3- (aminomethyl)-2-chloro-6-methylpyridin-4-ol (0.242 g, 1.41 mmol) and triethyl amine (0.213 g, 2.11 mmol). The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (0.55 g, 1.05 mmol) was added, and stirring was continued for overnight at room temperature. Upon completion, the reaction mixture was poured on ice cold water and extracted with 10% methanol in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by preparative HPLC to afford the title compound (0.012 g, 2.9%).

[0632] LCMS: 580.40 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 9.41 (brs, 1H), 8.84 (s, 1H), 8.57 (t, 1H), 8.16-8.14 (m, 1H), 7.56-7.52 (m, 2H), 7.35 (s, 1H), 6.70 (s, 1H), 5.00 (brs, 2H), 4.57 (s, 2H), 4.47 (d, 2H, J=4.0 Hz), 3.39 (s, 3H), 3.17-3.13 (m, 3H), 2.75 (m, 1H), 2.69-2.68 (m, 6H), 2.31 (s, 3H), 2.27 (s, 3H), 1.94 (m, 4H), 1.45-1.38 (m, 4H), 0.84 (t, 3H, J=6.8 Hz).

[0633] Example 12: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methyl-5-(l-methyl- lH-pyrazol-4-yl)benzamide:

Compound 89

[0634] Step 1: Synthesis of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-(l-met hyl-lH-pyrazol-4-yl)benzoate [0635] To a stirring solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methylbenzoate (1 eq) and l-methyl-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (1.2 eq) in dioxane/water mixture, was added Na 2 C0 3 (3.6 eq) and the solution was purged with argon for 10 min. Then Pd(PPh 3 ) 4 (0.1 eq) was added and argon was purged again for 10 min. The resulting reaction mass was heated at 100 °C for 4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford the title compound. (69% yield)

[0636] Aqueous NaOH (1.5 eq) was added to the solution of methyl 3-(((trans)-4- (dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-(l-methyl -lH-pyrazol-4-yl)benzoate (1 eq) in EtOH and stirred at 60 °C for 1 h. Upon completion, the solvent was removed under reduced pressure and the resulting oil was acidified first using dilute HC1 and then adjusted to pH 4 with citric acid. Extraction was carried out using 10% MeOH/DCM. The combined organic layers were dried and concentrated to give the corresponding acid.

[001] To a solution of above acid (1 equiv) in DMSO, 3-(amino methyl)-2-chloro-6- methylpyridin-4(lH)-one (1.2 equiv) and triethylamine (3 equiv) was added. The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (1.5 eq) was added and stirring was continued at room temperature for 16 h. Upon completion, the reaction mass was poured into ice cold water and extraction was carried out using 10% MeOH/DCM The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by preparative HPLC to afford the title compound. (3% yield)

[002] LCMS: 539.40 (M+l); 1H NMR DMSO-d 6 , 400 MHz) δ 9.30 (brs, 1H), 8.55 (t, 1H), 8.13 (s, 1H), 7.82 (s, 1H), 7.34 (s, 1H), 7.16 (s, 1H), 6.69 (s, 1H), 4.44 (d, 2H, J=4.4 Hz), 3.84 (s, 3H), 3.08 (m, 3H), 2.69-2.68 (m, 7H), 2.31 (s, 3H), 2.18 (s, 3H), 1.95-1.88 (m, 4H), 1.42 (m, 4H), 1.23 (m, 1H), 0.81 (t, 3H, J=6.8 Hz).

[003] Example 13: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methyl-5-(6- methylpyridin-3-yl)benzamide:

Compound 90

[004] Step 1: Synthesis of methyl 3-(((trans)-4-(dimethylamino)cyclohexyl)

(ethyl)amino)-2-methyl-5-(6-methylpyridin-3-yl)benzoate

[005] To a stirring solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methylbenzoate (1 eq) and 2-methyl-5-(4,4,5,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)pyridine (1.2 eq) in dioxane/water mixture, was added Na 2 C0 3 (3.6 eq) and the solution was purged with argon for 10 min. Then Pd(PPh 3 ) 4 (0.1 eq) was added and argon was purged again for 10 min. The resulting reaction mass was heated at 100 °C for 4 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by silica gel column chromatography to afford the title compound. (66% yield)

[006] Aqueous NaOH (1.5 eq) was added to the solution of 3-(((trans)-4- (dimethylamino)cyclohexyl) (ethyl)amino)-2-methyl-5-(6-methylpyridin-3-yl)benzoate ( 1 eq) in EtOH and stirred at 60 °C for 1 h. Upon completion, the solvent was removed under reduced pressure and the resulting oil was acidified first using dilute HCl and then adjusted to pH 4 with citric acid. Extraction was carried out using 10% MeOH/DCM. The combined organic layers were dried and concentrated to give the corresponding acid.

[007] To a solution of above acid (1 eq) in DMSO, 3-(amino methyl)-2-chloro-6- methylpyridin-4(lH)-one (1.2 eq) and triethyl amine (3 eq) was added. The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (1.5 eq) was added and stirring was continued at room temperature for 16 h. Upon completion, the reaction mass was poured into ice cold water and extraction was carried out using 10% MeOH/DCM The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by preparative HPLC to afford the title compound. (4% yield)

[008] LCMS: 550.35 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 9.45 (brs, 1H), 8.93 (s, 1H), 8.56 (t, 1H), 8.38 (d, 1H, J=6.4 Hz), 7.68 (d, 1H, J=7.6 Hz), 7.54 (s, 1H), 7.39 (s, 1H), 6.70 (s, 1H), 4.47 (d, 2H, J=4.0 Hz), 3.17-3.13 (m, 3H), 2.75 (m, 1H), 2.69-2.68 (m, 6H), 2.64 (s, 3H), 2.31 (s, 3H), 2.27 (s, 3H), 1.97-1.90 (m, 4H), 1.51-1.41 (m, 4H), 0.83 (t, 3H, J=6.4 Hz).

[009] Example 14: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methyl-5-(5- methylpyrazin-2-yl)benzamide:

Compound 91

[010] Step 1: Synthesis of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-(4,4,5 ,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)benzoate:

[Oi l] To a stirred solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methylbenzoate (0.8 g, 2.01 mmol) in 1,4-dioxane (10 mL), bis

(pinacolato)diboron (0.614 g, 2.41 mmol) and potassium acetate (0.789 g, 8.06 mmol) were added and solution was purged with argon for 10 min. PdCl 2 (dppf) (0.147 g, 0.201 mmol) was added and the reaction flask was again purged with argon for 10 min. The resulting reaction mass was heated at 80 °C for 8 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (0.7 g) which was used in the next step without further purification.

[012] Step 2: Synthesis of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-(5-met hylpyrazin-2-yl)benzoate:

[013] To a stirred solution of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-(4,4,5 ,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)benzoate (0.4 g, 0.9 mmol) in 1,4-dioxane/water (6/2 mL), were added 2- bromo-5-methylpyrazine (0.311 g, 1.80 mmol) and K 3 P0 4 (0.572 g, 2.70 mmol) and the reaction flask was purged with argon for 10 min. PdCl 2 (dppf) (0.073 g, 0.09 mmol) was added and the reaction flask was again purged with argon for 10 min. The resulting reaction mass was heated at 80 °C for 8 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford crude material which then purified by silica gel column chromatography to afford the title compound (0.27 g, 73.2%).

[014] Step 3: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-

3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-m ethyl-5-(5-methylpyrazin-2- yl)benzamide:

[015] Aqueous NaOH (0.039 g, 0.987 mmol) was added to a solution of methyl 3- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl -5-(5-methylpyrazin-2- yl)benzoate (0.27 g, 0.658 mmol) in EtOH (3 mL) and stirred at 60 °C for 1 h. After completion of the reaction, ethanol was removed under reduced pressure and the resulting oil was acidified first using dilute HCl and then adjusted to pH 4 with citric acid. Extraction was carried out using MeOH:DCM (10:90). The combined organic layers were dried and concentrated to afford the respective acid (0.25 g, 96.2%).

[016] To the acid (0.2 g, 0.505 mmol) dissolved in DMSO (2 mL) were added 3- (aminomethyl)-2-chloro-6-methylpyridin-4(lH)-one (0.13 g, 0.757 mmol) and triethyl amine (0.153 g, 1.51 mmol). The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (0.393 g, 0.757 mmol) was added, and stirring was continued at room temperature for 16 h. Upon completion, the reaction mixture was poured into ice cold water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford crude material which then purified by preparative HPLC to afford the title compound (0.03 g, 10.8%).

[017] LCMS: 551.35 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 9.36 (brs, 1H), 9.10 (s, 1H), 8.59 (s, 2H), 7.87 (s, 1H), 7.71 (s, 1H), 6.70 (s, 1H), 4.48 (d, 2H, J=4.4 Hz), 3.12 (m, 3H), 2.75 (m, 1H), 2.69-2.68 (m, 6H), 2.53 (s, 3H), 2.31 (s, 3H), 2.28 (s, 3H), 1.95-1.90 (m, 4H), 1.46 (m, 4H), 0.83 (t, 3H, J=6.8 Hz).

[018] Example 15: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-4'-(2-methoxyethoxy)-4- methyl- [1,1 '-biphenyl] -3-carboxamide:

Compound 92

[019] Step 1: Synthesis of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)- (ethyl) amino)-4'-hydroxy-4-methyl-[l,l'-biphenyl]-3-carboxylate: [020] To a stirred solution of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)- amino)-cyclohexyl)-(ethyl)-amino)-2-methylbenzoate (189 g, 402 mmol) and (4- hydroxyphenyl) boronic acid (66.73 g, 484 mmol) in dioxane/ water mixture (3.5/1.26 L), Na 2 C0 3 (153.7 g, 1450 mmol) was added and the solution was purged with argon for 40 min. Then Pd (PPh 3 ) 4 (46.54 g, 40.29 mmol) was added and argon was purged again for 30 min. The reaction mass was heated at 100°C for 5 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was diluted with 10% MeOH/ DCM and filtered. The filtrate was concentrated, diluted with water and extracted with 10% MeOH/ DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (179 g, 92 %).

[021] Step 2: Synthesis of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)- (ethyl) amino)-4'-(2-methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carbo xylate:

[022] To stirred solution of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)-(ethyl)amino)-4'-hydroxy-4-methyl-[l,l'-biphenyl ]-3-carboxylate (180 g, 373 mmol) in acetonitrile (2 L), cesium carbonate (364 g, 1120 mmol) and l-bromo-2- methoxyethane (62.29 g, 448 mmol) were added. The reaction mass was heated at 80 °C for 12 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture diluted with water and extracted with 10% MeOH/ DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (170 g, 84%).

[023] Step 3: Synthesis of methyl 5-(((trans)-4-aminocyclohexyl) (ethyl) amino)-4'-(2- methoxyethoxy)-4-methyl-[ 1 , 1 ' -biphenyl] -3-carboxylate:

[024] To a stirred solution of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)-(ethyl)amino)-4'-(2-methoxyethoxy)-4-methyl-[l,l '-biphenyl]-3-carboxylate (5 g, 9.25 mmol) in DCM (50 mL) at 0°C, TFA (5 mL) was added and reaction was stirred at rt for 2 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was concentrated to dryness. The residue obtained was basified with aqueous sat. bicarbonate solution till pH 8 and aqueous layer extracted with 10% MeOH/DCM The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (4 g) which was used in subsequent step without further purification. [025] Step 4: Synthesis of methyl 5-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-4'-(2-methoxyetho xy)-4-methyl- [ 1 , 1 ' -biphenyl] - 3-carboxylate:

[026] To a stirred solution of methyl 5-(((trans)-4-aminocyclohexyl)(ethyl)amino)-4'-(2- methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carboxylate (4 g, 9.07 mmol) in DCM (40 mL) at 0°C, aq. 35% formaldehyde solution (0.952 g, 31.74 mmol) was added and solution was stirred for 20 min. Then sodium triacetoxyborohydride (4.82 g, 22.73 mmol) was added. The reaction mixture was stirred at 0°C for 2 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was diluted with water and extracted with 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (2 g, 47%).

[027] Step 5: Synthesis of 5-(((trans)-4-(dimethylamino)-cyclohexyl)-(ethyl)amino)-4'-( 2- methoxyethoxy)-4-methyl-[ 1 , 1 '-biphenyl] -3-carboxylic acid:

[028] To a solution of methyl 5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-4'- (2-methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carboxylate (2 g, 4.26 mmol) in EtOH (20 mL), IN NaOH (8 mL) was added and the reaction was stirred at 60°C for 1 h. The progress of the reaction was monitored by TLC. Upon completion ethanol was removed under reduced pressure and the reaction mass was acidified with IN HC1 and extracted with 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (1.8 g) which was used in subsequent step without further purification.

[029] Step 6: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)- 5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-4'-(2- methoxyethoxy)-4-methyl- [1,1 '-biphenyl] -3-carboxamide:

[030] To a stirred solution of 5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-4'- (2-methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carboxylic acid (0.2 g, 0.44 mmol) in DMSO (2 mL), 3-(aminomethyl)-2-chloro-6-methylpyridin-4(lH)-one (0.091 g, 0.53 mmol) and triethyl amine (0.18 mL, 1.32 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (0.343 g, 0.66 mmol) was added to it at 0 °C and stirring was continued at rt for 12 h. The progress of the reaction was monitored by TLC. Upon completion reaction mixture was diluted with water and extracted with 10%

MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by prep HPLC to afford the title compound (0.017 g, 6.3 %).

[031] LCMS: 609.50 (M+l); 1H NMR (400 MHz, DMSO-d6) δ 8.48 (t, IH), 7.53 (d, J = 8.8 Hz, 2H), 7.31 (s, IH), 7.17 (s, IH), 7.01 (d, J =9.2 Hz, 2H), 6.62 (s, IH), 6.30 (brs, 2H), 4.42 (d, J = 2.4 Hz, 2H), 4.12 (t, J =4.4 Hz, 2H), 3.67 (t, J = 4.4 Hz, 2H), 3.31 (s, 3H), 3.08 (q, J = 7.2 Hz, 2H), 2.23 -2.18 (m, 12H), 1.84-1.78 (m, 4H), 1.42-1.13 (m, 4H), 0.83 (t, J = 7.2 Hz, 3H), 2H merged in solvent peak.

[032] Example 16: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-4'-(2-methoxyethoxy)-4- methyl- [1,1 '-biphenyl] -3-carboxamide:

Compound 93

[033] Step 1: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-5- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-4'-(2-me thoxyethoxy)-4-methyl-[l,l'- biphenyl] -3-carboxamide:

[034] To a stirred solution of 5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-4'- (2-methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carboxylic acid (0.15 g, 0.329 mmol) in DMSO (2 mL), 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one (0.075 g, 0.494 mmol) and triethyl amine (0.14 mL, 0.989 mmol) were added. The reaction mixture was stirred at room temperature for 15 min at which time the mixture was cooled to 0 °C, PyBOP (0.257 g, 0.494 mmol) was added and stirring was continued at room temperature for 16 h. Upon completion, the reaction mixture was poured into ice cold water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and the solvent removed under reduced pressure. The crude compound was purified by column

chromatography to afford the title compound (0.08 g, 41.45 %). [035] LCMS: 589.65 (M+l); 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 8.14 (s, 1H), 7.53 (d, J =9.2 Hz, 2H), 7.30 (d, J = 2.0 Hz, 1H), 7.12 (d, J = 1.9 Hz, 1H), 7.01 (d, J = 8.8 Hz, 2H), 5.86 (s, 1H), 4.23 (d, J = 5.2 Hz, 2H), 4.15-4.07 (m, 2H), 3.71-3.63 (m, 2H), 3.32 (s, 3H), 3.03 (q, J = 6.4 Hz, 2H), 2.67-2.62 (m, 2H), 2.33 (s, 3H), 2.19 (s, 3H), 2.15 (s, 3H), 2.11 (s, 6H), 1.83-1.75 (m, 4H), 1.41-1.08 (m, 4H), 0.82 (t, J = 6.4 Hz, 3H).

[036] Example 17: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(((trans)-4-(dimethylamino)cyclohexyl)(propyl)a mino)-4'-(2-methoxyethoxy)- 4-methyl-[ 1 , 1 '-biphenyl] -3-carboxamide

Compound 94

[037] Step 1: Synthesis of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)-(propyl)-amino)-2-methylbenzoate:

[038] To a stirred solution of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)- amino)cyclohexyl)amino)-2-methylbenzoate (8 g, 18.18 mmol) and propionaldehyde (5.28 g, 90.90 mmol) in DCE (80 mL), acetic acid (6.54 g, 109.08 mmol) was added and the reaction was stirred at rt for 30 min. Then sodium triacetoxyborohydride (11.56 g, 54.54 mmol) was added at 0 °C and the reaction was stirred at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was quenched with aqueous sodium bicarbonate, the organic layer was separated and the aqueous layer was extracted with dichloromethane. The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (6.5 g, 74%).

[039] Step 2: Synthesis of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)-(propyl)-amino)-4'-hydroxy-4-methyl-[l,l'-biphen yl]-3-carboxylate: [040] To a stirred solution of methyl 5-bromo-3-(((trans)-4-((tert-butoxycarbonyl)-amino) cyclohexyl)(propyl)amino)-2-methylbenzoate (6.5 g, 13.48 mmol) and (4-hydroxyphenyl) boronic acid (2.23 g, 16.18 mmol) in dioxane/ water mixture (50/20 mL), Na 2 C0 3 (5.15 g, 48.54 mmol) was added and the solution was purged with argon for 20 min. Then Pd(PPh ) 4 (1.55 g, 1.35 mmol) was added and argon was purged again for 20 min. The reaction mass was heated at 100 °C for 5 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10%

MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (6.55 g, 98%).

[041] Step 3: Synthesis of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)-(propyl)-amino)-4'-(2-methoxyethoxy)-4-methyl-[l ,l'-biphenyl]-3-carboxylate:

[042] To a stirred solution of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)- cyclohexyl)(propyl)amino)-4'-hydroxy-4-methyl-[l,l'-biphenyl ]-3-carboxylate (6.55 g, 13.20 mmol) in ACN (60 mL), cesium carbonate (12.87 g, 39.61 mmol) and l-bromo-2- methoxyethane (2.20 g, 15.84 mmol) were added. The resulting reaction mass was heated at 80 °C for 12 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was diluted with water and extracted with DCM. Combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (4.2 g, 57%).

[043] Step 4: Synthesis of tert-butyl ((trans)-4-((5-(((2,6-dimethyl-4-oxo-l,4- dihydropyridin-3-yl)methyl)carbamoyl)-4'-(2-methoxyethoxy)-4 -methyl- [ 1 , 1 '-biphenyl] -3- yl)(propyl)amino)cyclohexyl)carbamate:

[044] To a solution of methyl 5-(((trans)-4-((tert-butoxycarbonyl)-amino)-cyclohexyl)- (propyl)-amino)-4'-(2-methoxyethoxy)-4-methyl-[l,l'-biphenyl ]-3-carboxylate (4.2 g, 7.58 mmol) in EtOH (40 mL), aq. NaOH (0.455 g, 11.37 mmol) was added and the reaction mixture was stirred at 60 °C for 1 h. The progress of the reaction was monitored by TLC. Upon completion, the solvent was removed under reduced pressure and the reaction mass was acidified with IN HC1 and extracted with 10% MeOH/DCM. The combined organic layers were dried and concentrated to afford the respective acid (3 g) which was used in the subsequent step without further purification.

[045] To a solution of above acid (1 g, 1.85 mmol) in DMSO (10 mL), 3-(amino methyl)- 2,6-dimethylpyridin-4(lH)-one (0.422 g, 2.77 mmol) and triethyl amine (1.03 mL, 7.40 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (1.44 g, 2.77 mmol) was added to it at 0 °C and stirring was continued at rt for 12 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.5 g, 40%).

[046] Step 5: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-5- (((trans)-4-(dimethylamino)cyclohexyl)(propyl)amino)-4'-(2-m ethoxyethoxy)-4-methyl- [1,1 '-biphenyl] -3-carboxamide:

[047] To a stirred solution of tert-butyl ((trans)-4-((5-(((2,6-dimethyl-4-oxo-l,4- dihydropyridin-3-yl)methyl)carbamoyl)-4'-(2-methoxyethoxy)-4 -methyl- [ 1 , 1 '-biphenyl] -3- yl)(propyl)amino)cyclohexyl)carbamate (0.5 g, 0.74 mmol) in DCM (10 mL) at 0 °C, TFA (2 mL) was added and the reaction mixture was stirred at rt for 2 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was concentrated to dryness. The residue obtained was basified with aqueous sat. bicarbonate solution till pH 8 and the aqueous layer was extracted with 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the crude amine (0.29 g) which was used in the subsequent step without further purification.

[048] To a stirred solution of above crude amine (0.29 g, 0.505 mmol) in

dichloromethane (10 mL) at 0 °C, aq. 35% formaldehyde solution (0.053 g, 1.77 mmol) was added and the solution was stirred for 20 min. Then sodium triacetoxyborohydride (0.267 g, 1.26 mmol) was added and the reaction mixture was stirred at 0 °C for 2 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted water and extracted with 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.11 g, 36%).

[049] LCMS: 603.65 (M+l); 1H NMR (400 MHz, Methanol-d4) δ 7.48 (d, J = 8.8 Hz, 2H), 7.36 (d, J = 1.6 Hz, 1H), 7.20 (d, J = 1.6 Hz, 1H), 6.99 (d, J = 8.4 Hz, 2H), 6.23 (s, 1H), 4.45 (s, 2H), 4.15 (t, J = 4.4 Hz, 2H), 3.76 (t, J = 4.8 Hz, 2H), 3.44 (s, 3H), 3.08 (t, J = 6.4 Hz, 2H), 2.75-2.69 (m, 1H), 2.57 (s, 3H), 2.31 (s, 3H), 2.28 (s, 3H), 2.26 (s, 6H), 1.95- 1.92 (m, 4H), 1.51-1.45 (m, 2H), 1.32-1.21 (m, 4H), 0.82 (t, J = 6.8 Hz, 3H). 1H merged in solvent peak. [050] Example 18: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methyl-5-(3- morpholinoprop-l-yn-l-yl)benzamide:

Compound 95

[051] Step 1 : Synthesis of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-(3-mor pholinoprop-l-yn-l-yl) benzoate:

[052] The solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl)

(ethyl)amino)-2-methylbenzoate (0.5 g, 1.26 mmol), 4-(prop-2-yn-l-yl)morpholine (0.315 g, 2.52 mmol) and triethylamine (0.35 mL, 2.52 mmol) in DMF (10 mL) was bubbled through argon for 20 min. Then Cul (0.048 g, 0.252 mmol) and Pd(PPh 3 ) 4 (0.145 g, 0.126 mmol) were added and argon was bubbled through for a further 20 min. The reaction mixture was heated at 80 °C for 12 h and then cooled to room temperature. The reaction was quenched with water and extracted with 10% MeOH/DCM. The combined organic extracts were dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by column chromatography to give the title compound (0.375 g, 67.4%).

[053] Step 2: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-3-

(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-met hyl-5-(3-morpholinoprop-l-yn- l-yl)benzamide:

[054] Aqueous NaOH (0.047 g, 1.18 mmol in 2 mL H 2 0) was added to the solution of methyl 3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-meth yl-5-(3- morpholinoprop-l-yn-l-yl)benzoate (0.35 g, 0.79 mmol) in EtOH (10 mL) and stirred at 60 °C for 1 h. After completion of the reaction, ethanol was removed under reduced pressure and the resulting oil was acidified first using dilute HC1 and then adjusted to pH 4 with citric acid. Extraction was carried out using 10 % MeOH/DCM. The combined organic layers were dried and concentrated giving respective acid (0.3 g, 88.75 %).

[055] To the solution of the above acid (0.2 g, 0.468 mmol) in DMSO (2 mL), were added 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one (0.106 g, 0.702 mmol) and triethyl amine (0.2 mL, 1.40 mmol). The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (0.365 g, 0.702 mmol) was added, and stirring was continued at room temperature for 16 h. Upon completion, the reaction mass was poured into ice water and extracted with 10 % MeOH/DCM. The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.025 g, 9.5%).

[056] LCMS: 562.60 (M+l); 1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H), 8.15 (t, J = 5.1 Hz, 1H), 7.13 (d, J = 1.7 Hz, 1H), 6.93 (d, J = 1.6 Hz, 1H), 5.86 (s, 1H), 4.19 (d, J = 5.1 Hz, 2H), 3.60 (t, J = 4.1 Hz, 4H), 3.48 (s, 2H), 3.33-3.24 (m, 1H), 3.00 (q, J = 1.4 Hz, 2H), 2.64-2.51 (m, 1H), 2.32 (s, 3H), 2.22-2.05 (m, 14H), 1.91-1.75 (m, 6H), 1.34-1.29 (m, 2H), 1.18-1.04 (m, 2H), 0.77 (t, J = 6.8 Hz, 3H).

[057] Example 19: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methyl-5-(3- morpholinoprop-l-yn-l-yl)benzamide:

Compound 96

[058] To the solution of 3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2- methyl-5-(3-morpholinoprop-l-yn-l-yl)benzoic acid (0.3 g, 0.7 mmol) in DMSO (3 mL), were added 3-(aminomethyl)-2-chloro-6-methylpyridin-4(lH)-one (0.241 g, 1.40 mmol) and triethyl amine (0.1 mL, 0.7 mmol). The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (0.546 g, 1.05 mmol) was added, and stirring was continued at room temperature for 16 h. Upon completion, the reaction mass was poured into ice water and extracted with 10 % MeOH/DCM. The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.025 g, 6.1%).

[059] LCMS: 582.40 (M+l); 1H NMR (DMSO-J 6 , 400 MHz) δ 9.43 (brs, 1H), 8.50 (t, 1H), 7.27 (s, 1H), 7.13 (s, 1H), 6.69 (s, 1H), 4.43 (d, 2H, J=4.4 Hz), 4.32 (m, 2H), 3.79 (brs, 2H), 3.25 (brs, 2H), 3.03 (q, 2H), 2.69-2.68 (m, 8H), 2.31 (s, 3H), 2.22 (s, 3H), 1.97- 1.92 (m, 2H), 1.82 (m, 2H), 1.43-1.41 (m, 4H), 0.78 (t, 3H, J=6.8 Hz), 7H merged in solvent peak.

[060] Example 20: Synthesis of N-((2,6-dimethyl-4-oxo- 1 ,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methyl-5-((3- methyloxetan-3 - yl)ethynyl)benzamide :

Compound 97

[061] Step 1: Synthesis of methyl 3-(((trans)-4-

(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl-5-((3-me thyloxetan-3-yl)ethynyl) benzoate:

[062] To a solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methylbenzoate (0.5 g, 1.26 mmol) in DMF (10 mL), was added 3-ethynyl- 3-methyloxetane (0.242 g, 2.52 mmol) and triethylamine (0.35 mL, 2.52 mmol). Argon was bubbled through the reaction mixture for 20 min at which time Cul (0.048 g, 0.252 mmol) and Pd(PPh 3 ) 4 (0.145 g, 0.126 mmol) were added and argon was bubbled through for a further 20 min. The reaction mixture was heated at 80 °C for 12 h and then cooled to room temperature. The reaction was quenched with water and extracted with 10% MeOH/DCM. The combined organic extracts were dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by column chromatography to give the title compound (0.3 g, 57.69%).

[063] Step 2: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-3- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl -5-((3-methyloxetan-3- yl)ethynyl)benzamide:

[064] Aqueous NaOH (0.027 g, 0.679 mmol) was added to a solution of methyl 3- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-2-methyl -5-((3-methyloxetan-3- yl)ethynyl)benzoate (0.14 g, 0.33 mmol) in EtOH (10 mL) and the resulting mixture was stirred at 60 °C for 1 h. After completion of the reaction, ethanol was removed under reduced pressure and the resulting oil was acidified first using dilute HC1 and then adjusted to pH 4 with citric acid. Extraction was carried out using 10% MeOH/DCM. The combined organic layers were dried and concentrated giving respective acid (0.11 g, 81.48 %).

[065] To a solution of above acid (0.11 g, 0.27 mmol) in DMSO (1 mL), was added 3- (aminomethyl)-2,6-dimethylpyridin-4(lH)-one (0.084 g, 0.55 mmol) and triethyl amine (0.11 mL, 0.82 mmol). The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (0.211 g, 0.414 mmol) was added, and stirring was continued at room temperature for 16 h. Upon completion, the reaction mass was poured into ice water and extracted with 10% MeOH/DCM. The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by preparative HPLC to afford the title compound (0.005 g, 3.4%).

[066] LCMS: 533.50 (M+l); 1H NMR (400 MHz, MeOD-d4) δ 7.28 (s, 1H), 7.08 (s, 1H), 6.91 (s, 1H), 4.55 (s, 2H), 4.48 (d, J = 4.8 Hz, 2H), 3.11-3.09 (m, 3H), 2.82 (s, 6H), 2.78 (s, 3H), 2.56 (s, 3H), 2.26 (s, 3H), 2.08-2.02 (m, 4H), 1.67 (s, 3H), 1.55-1.48 (m, 4H), 0.87 (t, J = 6.0 Hz, 3H), 3H merged in solvent peak.

[067] Example 21: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-4-methyl-4'- (morpholinomethyl)-[ 1 , 1 '-biphenyl] -3-carboxamide:

Compound 98

[068] Step 1: Synthesis of 5-bromo-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methylbenzamide:

[069] To a solution of methyl 5-bromo-3-(((trans)-4-(dimethylamino)cyclohexyl) (ethyl)amino)-2-methylbenzoate (1 g, 2.52 mmol) in EtOH (10 mL), aq. NaOH (0.202 g, 5.05 mmol) was added and reaction was stirred at 60 °C for 1 h. Upon completion, the solvent was removed under reduced pressure and acidified to pH 6 with IN HCl. Extraction was carried out using 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the corresponding acid (0.9 g, 93.36%).

[070] To a stirred solution of the above acid (0.9 g, 2.35 mmol) in DMSO (5 mL) was added 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one (0.716 g, 4.71 mmol) and triethyl amine (0.9 mL, 7.06 mmol). The reaction mixture was stirred at room temperature for 15 min at which time the reaction mixture was cooled to 0 °C, PyBOP (1.83 g, 3.53 mmol) was added, and stirring was continued at room temperature for 16 h. Upon completion, the reaction mass was poured into ice water and extracted with 10% MeOH/DCM. The combined organic layers were dried over sodium sulphate and concentrated under reduced pressure. The crude compound was purified by preparative HPLC to afford the title compound (0.26 g, 21.5%).

[071] Step 2: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-5- (((trans)-4-(dimethylamino)cyclohexyl)(ethyl)amino)-4-methyl -4'-(morpholinomethyl)- [1,1 '-biphenyl] -3-carboxamide:

[072] To a stirred solution of 5-bromo-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(((trans)-4-(dimethylamino)cyclohexyl)(ethyl)am ino)-2-methylbenzamide (0.2 g, 0.387 mmol) and 4-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzyl)mor pholine (0.141 g , 0.465 mmol) in dioxane/water mixture (2/1 mL), Na 2 C0 3 (0.147 g, 1.39 mmol) was added and solution was purged with argon for 20 min. Then Pd(PPh 3 ) 4 (0.045 g, 0.038 mmol) was added and argon was purged again for 10 min. The reaction mass was heated at 100 °C for 5 h. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH in DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by preparative HPLC to afford the title compound (0.045 g, 19.0 %).

[073] LCMS: 614.65 (M+l); 1H NMR (400 MHz, DMSO-d6) δ 14.09 (s, 1H), 9.81 (s, 1H), 8.68 (t, J = 4.8 Hz, 1H), 7.74 (d, J = 8.4 Hz, 2H), 7.58 (d, J = 8.0 Hz, 2H), 7.46 (s, 1H), 7.29 (s, 1H), 7.01 (s, 1H), 4.43 (d, J = 4.8 Hz, 2H), 4.39 (s, 2H), 4.00-3.94 (m, 2H), 3.70-3.63 (m, 2H), 3.26-3.13 (m, 7H), 2.75-2.67 (m, 10H), 2.53 (s, 3H), 2.22 (s, 3H), 1.97- 1.87 (m, 4H), 1.45-1.43 (m, 4H), 0.83 (t, J = 6.4 Hz, 3H).

[074] Example 22: Synthesis of 5-chloro-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(ethyl((trans)-4-((3-methoxybenzyl)(methyl)amin o)cyclohexyl)amino)-2- methylbenzamide

Compound 99

[075] Step 1: Synthesis of methyl 3-(((trans)-4-((tert-butoxycarbonyl)-(methyl)-amino)- cyclohexyl)(ethyl)amino)-5-chloro-2-methylbenzoate:

[076] To a stirred solution of methyl 3-(((trans)-4-((tert- butoxycarbonyl)amino)cyclohexyl) (ethyl) amino)-5-chloro-2-methylbenzoate (10 g, 23.58 mmol) in dry DMF (100 mL) at 0 °C, NaH (60%, 2.35 g, 58.95 mmol) was added and the solution was stirred at same temperature for 20 min. Then methyl iodide (20 g, 141.5 mmol) was added at 0 °C and the reaction was stirred at rt for 12 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction was quenched with water and extracted with DCM. The combined organic layers were washed with water, dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (9.1 g, 88%).

[077] Step 2: Synthesis of methyl 5-chloro-3-(ethyl ((trans)-4-(methylamino)cyclohexyl) amino)-2-methylbenzoate:

[078] To a stirred solution of methyl 3-(((trans)-4-((tert-butoxycarbonyl)-(methyl)- amino)-cyclohexyl)(ethyl)amino)-5-chloro-2-methylbenzoate (4.5 g, 10.27 mmol) in DCM (50 mL) at 0 °C, TFA (10 mL) was added and the reaction was stirred at rt for 2 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was concentrated to dryness. The residue obtained was basified with sat. sodium bicarbonate solution to pH 8 and aqueous layer was extracted with 10%MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (3 g) which was used in the subsequent step without further purification.

[079] Step 3: Synthesis of methyl 5-chloro-3-(ethyl((trans)-4-((3-methoxybenzyl)- (methyl)-amino)-cyclohexyl)amino)-2-methylbenzoate:

[080] To a stirred solution of methyl 5-chloro-3-(ethyl((trans)-4-(methylamino)- cyclohexyl)-amino)-2-methylbenzoate (1 g, 2.96 mmol) and 3-methoxybenzaldehyde (0.482 g, 3.55 mmol) in dichloroethane (10 mL), titanium isopropoxide (1.68 g, 5.91 mmol) was added and the solution was stirred at rt for 15 min. Then sodium cyanoborohydride (0.372 g, 5.91 mmol) was added and the reaction was stirred at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction was quenched with water and filtered through a bed of celite. The filtrate was diluted with 10%MeOH/DCM. The organic layer was separated and aqueous layer was extracted with 10%MeOH/DCM. The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (1 g, 74%).

[081] Step 4: Synthesis of 5-chloro-3-(ethyl((trans)-4-((3-methoxybenzyl)-(methyl)- amino)-cyclohexyl)-amino)-2-methylbenzoic acid:

[082] To a stirred solution of methyl 5-chloro-3-(ethyl((transJ-4-((3-methoxybenzyl)- (methyl)-amino)cyclohexyl)amino)-2-methylbenzoate (1 g, 2.18 mmol) in EtOH (10 mL), aq. NaOH (0.13 g, 3.26 mmol) was added and the reaction was stirred at 60 °C for 1 h. The progress of the reaction was monitored by TLC. Upon completion ethanol was removed under reduced pressure and the reaction mass was acidified with dil. HC1 up to pH 6 and extracted with 10%MeOH/DCM. The combined organic layers were dried over anhydrous sodium sulphate and concentrated giving the respective acid (0.6 g) which was used in the subsequent step without further purification.

[083] Step 5: Synthesis of 5-chloro-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(ethyl((trans)-4-((3-methoxybenzyl)(methyl)amin o)cyclohexyl)amino)-2- methylbenzamide:

[084] To a stirred solution of 5-chloro-3-(ethyl((trans)-4-((3-methoxybenzyl)-(methyl)- amino)-cyclohexyl)amino)-2-methylbenzoic acid (0.25 g, 0.563 mmol) in DMSO (3 mL), 3- (amino methyl)-2,6-dimethylpyridin-4(lH)-one (0.128 g, 0.84 mmol) and triethyl amine (0.23 mL, 1.69 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (0.439 g, 0.84 mmol) was added to it at 0 °C and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mixture was diluted with water and extracted with 10 MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.08 g,

25%).

[085] LCMS: 579.35 (M+l); 1H NMR (400 MHz, DMSO- d 6 ) δ 10.99 (s, IH), 8.19 (t, J = 5.2 Hz, IH), 7.19 (t, J = 1.6 Hz, IH), 7.13 (s, IH), 6.91 (s, IH), 6.85-6.76 (m, 3H), 5.86 (s, IH), 4.19 (d, J = 5.2 Hz, 2H), 3.72 (s, 3H), 3.47 (s, 2H), 3.01 (q, J = 7.6 Hz, 2H), 2.67-2.60 (m, IH), 2.42-2.37 (m, IH), 2.32 (s, 3H), 2.15 (s, 3H), 2.13 (s, 3H), 2.06 (s, 3H), 1.78-1.76 (m, 4H), 1.38- 1.23 (m, 4H), 0.77 (t, J = 6.8 Hz, 3H).

[086] Example 23: Synthesis of 5-chloro-N-((2-chloro-6-methyl-4-oxo- l,4- dihydropyridin-3-yl)methyl)-3-(ethyl((trans)-4-((3- methoxybenzyl)(methyl)amino)cyclohexyl)amino)-2-methylbenzam ide

Compound 101 [087] Step 1: Synthesis of 5-chloro-N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(ethyl((trans)-4-((3-methoxybenzyl)(methyl)amin o)cyclohexyl)amino)-2- methylbenzamide:

[088] To a stirred solution of 5-chloro-3-(ethyl((trans)-4-((3-methoxybenzyl)-(methyl)- amino)-cyclohexyl)amino)-2-methylbenzoic acid (0.2 g, 0.45 mmol) in DMSO (2 mL), 3- (amino methyl)-2-chloro-6-methylpyridin-4(lH)-one (0.116 g, 0.675 mmol) and triethylamine (0.2 mL, 1.35 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (0.351 g, 0.675 mmol) was added to it at 0 °C and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10% MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound

(0.014 g, 5.2%).

[089] LCMS: 599.50 (M+l); 1H NMR (400 MHz, DMSO-J 6 ) δ 11.21 (s, 1H), 8.51 (t, 1H), 7.22-7.17 (m, 2H), 6.96 (s, 1H), 6.88-6.82 (m, 2H), 6.69 (s, 1H), 6.55 (s, 1H), 4.42 (d, J = 4.0 Hz, 2H), 3.73 (s, 3H), 3.62-3.55 (m, 2H), 3.02 (q, J = 7.2 Hz, 2H), 2.67-2.64 (m, 1H), 2.33-2.31 (m, 1H), 2.30 (s, 3H), 2.16 (s, 6H), 1.85-1.78 (m, 4H), 1.40-1.37 (m, 4H), 0.78 (t, J =7.2 Hz, 3H).

[090] Example 24: Synthesis of 5-chloro-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(ethyl(l-(2-methoxyethyl)piperidin-4-yl)amino)- 2-methylbenzamide

Compound 103

[091] Step 1: Synthesis of tert-butyl 4-((5-chloro-3-(methoxycarbonyl)-2-methylphenyl)- amino)-piperidine- 1 -carboxylate: [092] To a stirred solution of methyl 3-amino-5-chloro-2-methylbenzoate (10 g, 50.25 mmol) and tert-butyl 4-oxopiperidine-l-carboxylate (19.9 g, 100.5 mmol) in dichloroethane (100 mL), acetic acid (18.09 g, 301.5 mmol) was added and the reaction was stirred at rt for 20 min. Then sodium triacetoxyborohydride (31.95 g, 150.7 mmol) was added at 0 °C and the reaction was stirred at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction was quenched with aqueous sodium bicarbonate solution, the organic layer was separated and the aqueous layer was extracted with dichloromethane. The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (12.5 g, 65%).

[093] Step 2: Synthesis of tert-butyl 4-((5-chloro-3-(methoxycarbonyl)-2-methylphenyl)- (ethyl)-amino)piperidine- 1 -carboxylate:

[094] To a stirred solution of tert-butyl 4-((5-chloro-3-(methoxycarbonyl)-2- methylphenyl)-amino)-piperidine-l -carboxylate (12 g, 31.41 mmol) and acetaldehyde (3.46 g, 78.53 mmol) in dichloroethane (150 mL), acetic acid (11.30 g, 188.4 mmol) was added and the reaction was stirred at rt for 20 min. Then sodium triacetoxyborohydride (19.9 g, 93.86 mmol) was added and the reaction was stirred at rt for 12 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction was quenched with aqueous sodium bicarbonate solution, the organic layer was separated and the aqueous layer was extracted with dichloromethane. The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (10.2 g, 79%).

[095] Step 3: Synthesis of methyl 5-chloro-3-(ethyl(piperidin-4-yl)amino)-2- methylbenzoate:

[096] To a stirred solution of tert-butyl 4-((5-chloro-3-(methoxycarbonyl)-2- methylphenyl)-(ethyl)-amino) piperidine-1 -carboxylate (10.2 g, 24.87 mmol) in DCM (100 mL) at 0 °C, TFA (10 mL) was added and the reaction was stirred at rt for 2 h. The progress of the reaction was monitored by TLC. Upon completion the reaction mass was

concentrated to dryness. The residue obtained was treated with sat. sodium bicarbonate solution to pH 8 and the aqueous layer extracted with 10%MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (7.5 g) which was used in the subsequent step without further purification. [097] Step 4: Synthesis of methyl 5-chloro-3-(ethyl(l-(2-methoxyethyl)piperidin-4- yl)amino)-2-methylbenzoate:

[098] To a stirred solution of methyl 5-chloro-3-(ethyl(piperidin-4-yl)amino)-2- methylbenzoate (4 g, 12.90 mmol) and l-bromo-2-methoxyethane (2.69 g, 19.35 mmol) in ACN (40 mL), K 2 C0 3 (3.56 g, 25.8 mmol) and KI (1.28 g, 7.71 mmol) were added. The reaction mixture was stirred at 60 °C for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mass was diluted with water and extracted with 10%MeOH/DCM. The combined organic layers were dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (3 g, 63%).

[099] Step 5: Synthesis of 5-chloro-3-(ethyl (l-(2-methoxyethyl) piperidin-4-yl) amino)- 2-methylbenzoic acid:

[0100] To a stirred solution of methyl 5-chloro-3-(ethyl(l-(2-methoxyethyl)piperidin-4- yl)amino)-2-methylbenzoate (3 g, 8.15 mmol) in EtOH (30 mL), aq. NaOH (0.489 g, 12.22 mmol) was added and the reaction was stirred at 60 °C for 1 h. The progress of the reaction was monitored by TLC. Upon completion, the ethanol was removed under reduced pressure and the reaction mass was acidified using dil. HC1 up to pH 6 and extracted with

10%MeOH/DCM. The combined organic layers were dried over anhydrous sodium sulphate and concentrated giving the respective acid (2.5 g) which was used in the subsequent step without further purification.

[0101] Step 6: Synthesis of 5-chloro-N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(ethyl(l-(2-methoxyethyl)piperidin-4-yl)amino)- 2-methylbenzamide:

[0102] To a stirred solution of 5-chloro-3-(ethyl(l-(2-methoxyethyl)piperidin-4-yl)amino)- 2-methylbenzoic acid (0.5 g, 1.41 mmol) in DMSO (5 mL), 3-(amino methyl)-2,6- dimethylpyridin-4(lH)-one (0.429 g, 2.82 mmol) and triethylamine (0.58 mL, 4.23 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (1.1 g, 2.11 mmol) was added to it at 0 °C and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10%MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography over basic alumina to afford the title compound (0.02 g, 2.9%).

[0103] LCMS: 489.45 (M+l); 1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H), 8.19 (t, J = 5.0 Hz, 1H), 7.15 (d, J = 2.3 Hz, 1H), 6.93 (d, J = 2.2 Hz, 1H), 5.86 (s, 1H), 4.19 (d, J = 4.9 Hz, 2H), 3.37 (t, J = 5.9 Hz, 2H), 3.20 (s, 3H), 3.00 (q, J = 6.9 Hz, 2H), 2.82 (d, J = 11.0 Hz, 2H), 2.72-2.62 (m, 1H), 2.39 (t, J =5.6 Hz, 2H), 2.32 (s, 3H), 2.14 (d, J = 9.0

1.95- 1.84 (m, 2H), 1.65- 1.42 (m, 4H), 0.77 (t, J = 6.9 Hz, 3H).

[0104] Example 25: Synthesis of 5-chloro-N-((2-chloro-6-methyl-4-oxo- l,4- dihydropyridin-3-yl)methyl)-3-(ethyl(l-(2-methoxyethyl)piper idin-4-yl)amino)-2- methylbenzamide

Compound 104

[0105] Step 1 : Synthesis of 5-chloro-N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-3-(ethyl(l-(2-methoxyethyl)piperidin-4-yl)amino)- 2-methylbenzamide:

[0106] To a stirred solution of 5-chloro-3-(ethyl(l-(2-methoxyethyl)piperidin-4-yl)amino)- 2-methylbenzoic acid (0.5 g, 1.41 mmol) in DMSO (5 mL), 3-(aminomethyl)-2,6- dimethylpyridin-4(lH)-one (0.485 g, 2.82 mmol) and triethylamine (0.58 mL, 4.23 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (1.1 g, 2.11 mmol) was added to it at 0 °C and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10 MeOH/DCM. The combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography over basic to afford the title compound (0.02 g, 2.7%).

[0107] LCMS: 509.45 (M+l); 1H NMR (400 MHz, DMSO-d6) δ 11.36 (brs, 1H), 8.52 (t, J = 4.5 Hz, 1H), 7.18 (d, J = 2.2 Hz, 1H), 6.97 (d, J = 2.1 Hz, 1H), 6.64 (s, 1H), 4.42 (d, J = 4.3 Hz, 2H), 3.37 (t, J = 5.9 Hz, 2H), 3.20 (s, 3H), 3.01 (q, J = 6.8 Hz, 2H), 2.82 (d, J = 11.2 Hz, 2H), 2.74-2.61 (m, 1H), 2.40 (t, J = 5.9 Hz, 2H), 2.29 (s, 3H), 2.16 (s, 3H), 1.95-1.84 (m, 2H), 1.65- 1.43 (m, 4H), 0.78 (t, J = 7.2 Hz, 3H). [0108] Example 26: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(ethyl((trans)-4-((2-methoxyethyl)(methyl)amino )cyclohexyl)amino)-4'-(2- methoxyethoxy)-4-methyl-[l,l'-bi henyl]-3-carboxamide:

Compound 100

[0109] Step 1: Synthesis of N-((2,6-dimethyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-5- (ethyl((trans)-4-((2-methoxyethyl)(methyl)amino)cyclohexyl)a mino)-4'-(2-methoxyethoxy)- 4-methyl-[ 1 , Γ-biphenyl] -3-carboxamide:

[0110] To a stirred solution of 5-(ethyl((trans)-4-((2-methoxyethyl)-(methyl)-amino)- cyclohexyl)-amino)-4'-(2-methoxyethoxy)-4-methyl-[l,l'-biphe nyl]-3-carboxylic acid (0.25 g, 0.501 mmol) in DMSO (3 mL), 3-(aminomethyl)-2,6-dimethylpyridin-4(lH)-one (0.076 g, 0.501 mmol) and triethylamine (0.28 mL, 2 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (0.391 g, 0.75 mmol) was added to it at 0 °C and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with

10 MeOH/DCM. Combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.035 g, 11%).

[0111] LCMS: 633.50 (M+1); 1H NMR (400 MHz, DMSO-J 6 ) δ 10.98 (s, 1H), 8.12 (t, J = 4.8 Hz, 1H), 7.53 (d, J = 8.8 Hz, 2H), 7.30 (s, 1H), 7.13 (s, 1H), 7.01 (d, J = 8.8 Hz, 2H), 5.86 (s, 1H), 4.23 (d, J = 4.8 Hz, 2H), 4.12 (t, J =4.4 Hz, 2H), 3.67 (t, J =4.4 Hz, 2H), 3.20 (s, 3H), 3.08 (q, J = 6.8 Hz, 2H), 2.67-2.65 (m, 1H), 2.33 (s, 3H), 2.30-2.28 (m, 1H), 2.20 (s, 3H), 2.15 (s, 3H), 2.14 (s, 3H), 1.83-1.80 (m, 2H), 1.71-1.68 (m, 2H), 1.38- 1.36 (m, 2H), 1.24-1.17 (m, 2H), 0.82 (t, J =6.8 Hz, 3H), 7H merged in solvent peak. [0112] Example 27: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3- yl)methyl)-5-(ethyl((trans)-4-((2-methoxyethyl)(methyl)amino )cyclohexyl)amino)-4'-(2- methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carboxamide:

Compound 102

[0113] Step 1: Synthesis of N-((2-chloro-6-methyl-4-oxo-l,4-dihydropyridin-3-yl)methyl)-

5-(ethyl((trans)-4-((2-methoxyethyl)(methyl)amino)cyclohe xyl)amino)-4'-(2- methoxyethoxy)-4-methyl-[l,l'-biphenyl]-3-carboxamide:

[0114] To a stirred solution of 5-(ethyl((trans)-4-((2-methoxyethyl)-(methyl)-amino)- cyclohexyl)-amino)-4'-(2-methoxyethoxy)-4-methyl-[l, -biphenyl]-3-carboxylic acid (0.5 g, 1.00 mmol) in DMSO (5 mL), 3-(aminomethyl)-2-chloro-6-methylpyridin-4(lH)-one (0.259 g, 1.50 mmol) and triethylamine (0.55 mL, 4.01 mmol) were added. The reaction mixture was stirred at rt for 15 min before PyBOP (0.78 g, 1.50 mmol) was added to it at 0 °C and stirring was continued at rt for 16 h. The progress of the reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water and extracted with 10 MeOH/DCM. Combined organic layers were dried over Na 2 S0 4 and concentrated under reduced pressure. The crude compound was purified by column chromatography to afford the title compound (0.11 g, 16%).

[0115] LCMS: 653.70 (M+1); 1H NMR (400 MHz, DMSO-J 6 ) δ 11.24 (s, 1H), 8.56 (t, J = 4.8 Hz, 1H), 7.54 (d, J = 8.8 Hz, 2H), 7.35 (s, 1H), 7.20 (s, 1H), 7.02 (d, J = 8.8 Hz, 2H), 6.69 (s, 1H), 4.45 (d, J = 4.4 Hz, 2H), 4.11 (t, J =4.4 Hz, 2H), 3.67 (t, J =4.4 Hz, 2H), 3.63- 3.60 (m, 2H), 3.39-3.32 (m, 7H), 3.31-3.11 (m, 4H), 3.09-2.69 (m, 4H), 2.31 (s, 3H), 2.24 (s, 3H), 1.97-1.91 (m, 4H), 1.50-1.47 (m, 4H), 0.83 (t, J =7.2 Hz, 3H).

[0116] Example 28: Bioassay protocol and General Methods

Protocol for Wild-Type and Mutant PRC2 Enzyme Assays [0117] General Materials. S-adenosylmethionine (SAM), S-adenosylhomocyteine (SAH), bicine, KC1, Tween20, dimethylsulfoxide (DMSO) and bovine skin gelatin (BSG) were purchased from Sigma- Aldrich at the highest level of purity possible. Dithiothreitol (DTT) was purchased from EMD. H-SAM was purchased from American Radiolabeled Chemicals with a specific activity of 80 Ci/mmol. 384-well streptavidin Flashplates were purchased from PerkinElmer.

[0118] Substrates. Peptides representative of human histone H3 residues 21 - 44 containing either an unmodified lysine 27 (H3K27meO) or dimethylated lysine 27

(H3K27me2) were synthesized with a C-terminal G(K-biotin) linker- affinity tag motif and a C-terminal amide cap by 21 st Century Biochemicals. The peptides were high-performance liquid chromatography (HPLC) purified to greater than 95% purity and confirmed by liquid chromatography mass spectrometry (LC-MS). The sequences are listed below.

H3K27meO: ATKAARKSAPATGGVKKPHRYRPGGK(biotin)-amide (SEQ ID

NO: 101)

H3K27me2: ATKAARK(me2)SAPATGGVKKPHRYRPGGK(biotin)-amide (SEQ ID NO: 102)

[0119] Chicken erythrocyte oligonucleosomes were purified from chicken blood according to established procedures.

[0120] Recombinant PRC2 Enzymes. Human PRC2 enzymes were purified as 4- component enzyme complexes co-expressed in Spodoptera frugiperda (sf9) cells using a baculovirus expression system. The subunits expressed were wild-type EZH2

(NM_004456) or EZH2 Y641F, N, H, S or C mutants generated from the wild-type EZH2 construct, EED (NM_003797), Suzl2 (NM_015355) and RbAp48 (NM_005610). The EED subunit contained an N-terminal FLAG tag that was used to purify the entire 4- component complex from sf9 cell lysates. The purity of the complexes met or exceeded 95% as determined by SDS-PAGE and Agilent Bioanalyzer analysis. Concentrations of enzyme stock concentrations (generally 0.3 - 1.0 mg/mL) was determined using a Bradford assay against a bovine serum albumin (BSA) standard.

[0121] General Procedure for PRC2 Enzyme Assays on Peptide Substrates. The assays were all performed in a buffer consisting of 20 mM bicine (pH = 7.6), 0.5 mM DTT, 0.005% BSG and 0.002% Tween20, prepared on the day of use. Compounds in 100% DMSO (1 were spotted into polypropylene 384-well V-bottom plates (Greiner) using a Platemate 2 X 3 outfitted with a 384-channel pipet head (Thermo). DMSO (1 was added to columns 11, 12, 23, 24, rows A - H for the maximum signal control, and SAH, a known product and inhibitor of PRC2 (1 μΐ ^ ) was added to columns 11,12, 23, 24, rows I - P for the minimum signal control. A cocktail (40 μί) containing the wild-type PRC2 enzyme and H3K27meO peptide or any of the Y641 mutant enzymes and H3K27me2 peptide was added by Multidrop Combi (Thermo). The compounds were allowed to incubate with PRC2 for 30 min at 25 °C, then a cocktail (10 μί) containing a mixture of non-radioactive and H-SAM was added to initiate the reaction (final volume = 51 μί). In all cases, the final concentrations were as follows: wild- type or mutant PRC2 enzyme was 4 nM, SAH in the minimum signal control wells was 1 mM and the DMSO concentration was 1%. The final concentrations of the rest of the components are indicated in Table 4, below. The assays were stopped by the addition of non-radioactive SAM (10 μί) to a final concentration of 600 μΜ, which dilutes the H-SAM to a level where its incorporation into the peptide substrate is no longer detectable. 50 μΐ ^ of the reaction in the 384- well polypropylene plate was then transferred to a 384- well Flashplate and the biotinylated peptides were allowed to bind to the streptavidin surface for at least lh before being washed three times with 0.1% Tween20 in a Biotek ELx405 plate washer. The plates were then read in a PerkinElmer TopCount platereader to measure the quantity of H-labeled peptide bound to the Flashplate surface, measured as disintegrations per minute (dpm) or alternatively, referred to as counts per minute (cpm).

Table 4: Final concentrations of components for each assay variation based upon EZH2 identity (wild-type or Y641 mutant EZH2)

[0122] General Procedure for Wild-Type PRC2 Enzyme Assay on Oligonucleosome Substrate. The assays was performed in a buffer consisting of 20 mM bicine (pH = 7.6), 0.5 mM DTT, 0.005% BSG, 100 mM KC1 and 0.002% Tween20, prepared on the day of use. Compounds in 100% DMSO (1 μί) were spotted into polypropylene 384-well V- bottom plates (Greiner) using a Platemate 2 X 3 outfitted with a 384-channel pipet head (Thermo). DMSO (1 μί) was added to columns 11, 12, 23, 24, rows A - H for the maximum signal control, and SAH, a known product and inhibitor of PRC2 (1 μΐ ^ ) was added to columns 11,12, 23, 24, rows I - P for the minimum signal control. A cocktail (40 μν> containing the wild-type PRC2 enzyme and chicken erythrocyte oligonucleosome was added by Multidrop Combi (Thermo). The compounds were allowed to incubate with PRC2 for 30 min at 25 °C, then a cocktail (10 μί) containing a mixture of non-radioactive and H-SAM was added to initiate the reaction (final volume = 51 μί). The final concentrations were as follows: wild-type PRC2 enzyme was 4 nM, non-radioactive SAM was 430 nM, H-SAM was 120 nM, chicken erythrocyte olignonucleosome was 120 nM, SAH in the minimum signal control wells was 1 mM and the DMSO concentration was 1%. The assay was stopped by the addition of non-radioactive SAM (10 μί) to a final concentration of 600 μΜ, which dilutes the H-SAM to a level where its incorporation into the chicken erythrocyte olignonucleosome substrate is no longer detectable. 50 μΐ ^ of the reaction in the 384- well polypropylene plate was then transferred to a 384- well Flashplate and the chicken erythrocyte nucleosomes were immobilized to the surface of the plate, which was then washed three times with 0.1% Tween20 in a Biotek ELx405 plate washer. The plates were then read in a PerkinElmer TopCount platereader to measure the quantity of H-labeled chicken erythrocyte oligonucleosome bound to the Flashplate surface, measured as disintegrations per minute (dpm) or alternatively, referred to as counts per minute (cpm).

[0123] % Inhibition Calculation

V dpm max -dpm min /

[0124] Where dpm = disintegrations per minute, cmpd = signal in assay well, and min and max are the respective minimum and maximum signal controls.

[0125] Four-parameter IC 50 fit

(Top-Bottom)

Y=Bottom+

j Coefficient

[0126] Where top and bottom are the normally allowed to float, but may be fixed at 100 or 0 respectively in a 3-parameter fit. The Hill Coefficient normally allowed to float but may also be fixed at 1 in a 3-parameter fit. Y is the % inhibition and X is the compound concentration.

[0127] IC 50 values for the PRC2 enzyme assays on peptide substrates (e.g., EZH2 wild type andY641F) are presented in Table 5 below.

[0128] WSU-DLCL2 Methylation Assay

[0129] WSU-DLCL2 suspension cells were purchased from DSMZ (German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany). RPMI/Glutamax Medium, Penicillin-Streptomycin, Heat Inactivated Fetal Bovine Serum, and D-PBS were purchased from Life Technologies, Grand Island, NY, USA. Extraction Buffer and Neutralization Buffer(5X) were purchased from Active Motif, Carlsbad, CA, USA. Rabbit anti-Histone H3 antibody was purchased from Abeam, Cambridge, MA, USA. Rabbit anti-H3K27me3 and HRP-conjugated anti-rabbit- IgG were purchased from Cell Signaling Technology, Danvers, MA, USA. TMB "Super Sensitive" substrate was sourced from BioFX

Laboratories, Owings Mills, MD, USA. IgG-free Bovine Serum Albumin was purchased from Jackson ImmunoResearch, West Grove, PA, USA. PBS with Tween (10X PBST) was purchased from KPL, Gaithersburg, MD, USA. Sulfuric Acid was purchased from Ricca Chemical, Arlington, TX, USA. Immulon ELISA plates were purchased from Thermo, Rochester, NY, USA. V-bottom cell culture plates were purchased from Corning Inc., Corning, NY, USA. V-bottom polypropylene plates were purchased from Greiner Bio-One, Monroe, NC, USA.

[0130] WSU-DLCL2 suspension cells were maintained in growth medium (RPMI 1640 supplemented with 10% v/v heat inactivated fetal bovine serum and 100 units/mL penicillin- streptomycin) and cultured at 37 °C under 5% C0 2. Under assay conditions, cells were incubated in Assay Medium (RPMI 1640 supplemented with 20% v/v heat inactivated fetal bovine serum and 100 units/mL penicillin-streptomycin) at 37 °C under 5% C0 2 on a plate shaker.

[0131] WSU-DLCL2 cells were seeded in assay medium at a concentration of 50,000 cells per mL to a 96- well V-bottom cell culture plate with 200 μί ρεΓ well. Compound (Ιμί) from 96 well source plates was added directly to V-bottom cell plate. Plates were incubated on a titer-plate shaker at 37 °C, 5% C0 2 for 96 hours. After four days of incubation, plates were spun at 241 x g for five minutes and medium was aspirated gently from each well of cell plate without disturbing cell pellet. Pellet was resuspended in 200 μΐ ^ DPBS and plates were spun again at 241 x g for five minutes. The supernatant was aspirated and cold (4 °C) Extraction buffer (100 μί) was added per well. Plates were incubated at 4 °C on orbital shaker for two hours. Plates were spun at 3427 x g x 10 minutes. Supernatant (80 μί ρεΓ well) was transferred to its respective well in 96 well V-bottom polypropylene plate.

Neutralization Buffer 5X (20 μΐ ^ per well) was added to V-bottom polypropylene plate containing supernatant. V-bottom polypropylene plates containing crude histone preparation (CHP) were incubated on orbital shaker x five minutes. Crude Histone Preparations were added (2μΙ. per well) to each respective well into duplicate 96 well ELIS A plates containing 100 μΐ. Coating Buffer (IX PBS + BSA 0.05% w/v). Plates were sealed and incubated overnight at 4 °C. The following day, plates were washed three times with 300 μΐ ^ per well IX PBST. Wells were blocked for two hours with 300 μΐ. per well ELISA Diluent ((PBS (IX) BSA (2% w/v) and Tween20 (0.05% v/v)). Plates were washed three times with IX PBST. For the Histone H3 detection plate, 100 μί ρεΓ well were added of anti-Histone-H3 antibody (Abeam, abl791) diluted 1: 10,000 in ELISA Diluent. For H3K27 trimethylation detection plate, 100 μί ρεΓ well were added of anti-H3K27me3 diluted 1:2000 in ELISA diluent. Plates were incubated for 90 minutes at room temperature. Plates were washed three times with 300 μΐ, IX PBST per well. For Histone H3 detection, 100 μΐ, of HRP- conjugated anti-rabbit IgG antibody diluted to 1:6000 in ELISA diluent was added per well. For H3K27me3 detection, 100 μΐ ^ of HRP conjugated anti-rabbit IgG antibody diluted to 1:4000 in ELISA diluent was added per well. Plates were incubated at room temperature for 90 minutes. Plates were washed four times with IX PBST 300 μΐ ^ per well. TMB substrate 100 μΐ ^ was added per well. Histone H3 plates were incubated for five minutes at room temperature. H3K27me3 plates were incubated for 10 minutes at room temperature. The reaction was stopped with sulfuric acid IN (100 μί ρεΓ well). Absorbance for each plate was read at 450 nm.

f!T3K27me3 OD450 vat.ue\

[0132] First, the ratio for each well was determined by Histone H3 OLHbO value}

[0133] Each plate included eight control wells of DMSO only treatment (Minimum

Inhibition) as well as eight control wells for maximum inhibition (Background wells).

[0134] The average of the ratio values for each control type was calculated and used to determine the percent inhibition for each test well in the plate. Test compound was serially diluted three-fold in DMSO for a total of ten test concentrations, beginning at 25 μΜ.

Percent inhibition was determined and IC 50 curves were generated using duplicate wells per concentration of compound. IC 50 values for this assay are presented in Table 5 below.

[0135] Percent Inhibition = 100-

[0136] Cell proliferation analysis [0137] WSU-DLCL2 suspension cells were purchased from DSMZ (German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany). RPMI/Glutamax Medium, Penicillin-Streptomycin, Heat Inactivated Fetal Bovine Serum were purchased from Life Technologies, Grand Island, NY, USA. V-bottom polypropylene 384-well plates were purchased from Greiner Bio-One, Monroe, NC, USA. Cell culture 384-well white opaque plates were purchased from Perkin Elmer, Waltham, MA, USA. Cell-Titer Glo® was purchased from Promega Corporation, Madison, WI, USA. SpectraMax M5 plate reader was purchased from Molecular Devices LLC, Sunnyvale, CA, USA.

[0138] WSU-DLCL2 suspension cells were maintained in growth medium (RPMI 1640 supplemented with 10% v/v heat inactivated fetal bovine serum and cultured at 37 °C under 5% C0 2 . Under assay conditions, cells were incubated in Assay Medium (RPMI 1640 supplemented with 20% v/v heat inactivated fetal bovine serum and 100 units/mL penicillin- streptomycin) at 37 °C under 5% C0 2 .

[0139] For the assessment of the effect of compounds on the proliferation of the WSU- DLCL2 cell line, exponentially growing cells were plated in 384-well white opaque plates at a density of 1250 cell/ml in a final volume of 50 μΐ of assay medium. A compound source plate was prepared by performing triplicate nine-point 3-fold serial dilutions in DMSO, beginning at 10 mM (final top concentration of compound in the assay was 20 μΜ and the DMSO was 0.2%). A 100 nL aliquot from the compound stock plate was added to its respective well in the cell plate. The 100% inhibition control consisted of cells treated with 200 nM final concentration of staurosporine and the 0% inhibition control consisted of DMSO treated cells. After addition of compounds, assay plates were incubated for 6 days at 37 °C, 5% C0 2 , relative humidity > 90% for 6 days. Cell viability was measured by quantization of ATP present in the cell cultures, adding 35 μΐ of Cell Titer Glo® reagent to the cell plates. Luminescence was read in the SpectraMax M5. The concentration inhibiting cell viability by 50% was determined using a 4-parametric fit of the normalized dose response curves. IC 50 values for this assay are also presented in Table 5 below.

Table 5

Cpd # WT Mutant H3K27Me3 wsu Salt EZH2 Y641F ICso ELISA ICso proliferation ICso (μΜ) (μΜ) (μΜ) ICso (μΜ)

86 >50 > 50 >50 >20 TFA

87 35.4 37.4 >50 3 TFA

18 0.125 0.108 8.8 14 TFA

19 0.78 0.480 >50 6.8 TFA

15 0.02 0.038 0.65 0.48 TFA

26 0.2 0.572 nd nd TFA

88 0.04 0.046 2.4 5.1 TFA

89 0.27 0.229 5.9 11.5 TFA

90 0.05 0.040 2.7 5.3 TFA

91 0.14 0.049 3.3 5.4 TFA

92 0.038 0.023 3.9 4.4 HC02H

93 0.005 0.005 1.7 1.2 None

94 0.025 0.007 5.1 2.9 None

95 0.017 0.012 11.4 nd None

96 0.13 0.118 4.3 nd TFA

97 0.005 0.008 9.2 nd TFA

98 0.005 0.005 3.1 4.4 TFA

99 0.001 0.005 0.56 nd None

100 0.027 0.018 7.5 4.2 None

101 0.015 0.010 3 nd None

102 0.15 0.093 10.8 nd None

103 0.36 0.341 41.3 nd None

104 0.87 0.598 49.5 nd None

[0140] Example 29: Derivation of the Lowest Cytotoxic Concentration (LCC)

[0141] It is well established that cellular proliferation proceeds through cell division that results in a doubling of the number of cells after division, relative to the number of cells prior to division. Under a fixed set of environmental conditions (e.g., pH, ionic strength, temperature, cell density, medium content of proteins and growth factors, and the like) cells will proliferate by consecutive doubling (i.e., division) according to the following equation, provided that sufficient nutrients and other required factors are available.

t

[0142] N t = N Q X 2' D (A. l)

where N t is the cell number at a time point (t) after initiation of the observation period, No is the cell number at the initiation of the observation period, t is the time after initiation of the observation period and t D is the time interval required for cell doubling, also referred to as the doubling time. Equation A.1 can be converted into the more convenient form of an exponential equation in base e, taking advantage of the equality, 0.693 = ln(2).

0.693

[0143] N t = N Q e tD (A.2)

[0144] The rate constant for cell proliferation (k p ) is inversely related to the doubling time as follows.

_ 0.693

[0145] - (A 3)

[0146] Combining equation A.2 and A.3 yields,

[0147] i N t = i N 0 p^ kpt (A.4)

[0148] Thus, according to equation A.4 cell number is expected to increase exponentially with time during the early period of cell growth referred to as log-phase growth.

Exponential equations like equation A.4 can be linearized by taking the natural logarithm of each side.

[0149] ln(N,) = ln(N 0 )+k t (A.5)

[0150] Thus a plot of ln(N t ) as a function of time is expected to yield an ascending straight line with slope equal to k p and y- intercept equal to In (No).

[0151] Changes in environmental conditions can result in a change in the rate of cellular proliferation that is quantifiable as changes in the proliferation rate constant k p . Among conditions that may result in a change in proliferation rate is the introduction to the system of an antiproliferative compound at the initiation of the observation period (i.e., at t = 0). When an antiproliferative compound has an immediate impact on cell proliferation, one expects that plots of ln(N t ) as a function of time will continue to be linear at all compound concentrations, with diminishing values of k p at increasing concentrations of compound.

[0152] Depending on the mechanistic basis of antiproliferative action, some

compounds may not immediately effect a change in proliferation rate. Instead, there may be a period of latency before the impact of the compound is realized. In such cases a plot of ln(N t ) as a function of time will appear biphasic, and a time point at which the impact of the compound begins can be identified as the breakpoint between phases. Regardless of whether a compound's impact on proliferation is immediate or begins after a latency period, the rate constant for proliferation at each compound concentration is best defined by the slope of the ln(N t ) vs. time curve from the time point at which compound impact begins to the end of the observation period of the experiment.

[0153] A compound applied to growing cells may affect the observed proliferation in one of two general ways: by inhibiting further cell division (cytostasis) or by cell killing (cytotoxicity). If a compound is cytostatic, increasing concentration of compound will reduce the value of k p until there is no further cell division. At this point, the rate of cell growth, and therefore the value of k p , will be zero. If, on the other hand, the compound is cytotoxic, then the value of k p will be composed of two rate constants: a rate constant for continued cell growth in the presence of the compound (k g ) and a rate constant for cell killing by the compound (k d ). The overall rate constant for proliferation at a fixed concentration of compound will thus be the difference between the absolute values of these o osing rate constants.

[0155] At compound concentrations for which the rate of cell growth exceeds that of cell killing, the value of k p will have a positive value (i.e., k p > 0). At compound concentrations for which the rate of cell growth is less than that for cell killing, the value of k p will have a negative value (i.e., k p < 0) and the cell number will decrease with time, indicative of robust cytotoxicity. When k g exactly matches k d then the overall proliferation rate constant, k p , will have a value of zero. We can thus define the lowest cytotoxic concentration (LCC) as that concentration of compound that results in a value of k p equal to zero, because any concentration greater than this will result in clearly observable cytotoxicity. Nota bene: at concentrations below the LCC there is likely to be cell killing occurring, but at a rate that is less than that of residual cell proliferation. The treatment here is not intended to define the biological details of compound action. Rather, the goal here is to merely define a practical parameter with which to objectively quantify the concentration of compound at which the rate of cell killing exceeds new cell growth. Indeed, the LCC represents a breakpoint or critical concentration above which frank cytotoxicity is observed, rather than a cytotoxic concentration per se. In this regard, the LCC can be viewed similar to other physical breakpoint metrics, such as the critical micelle concentration (CMC) used to define the concentration of lipid, detergent or other surfactant species above which all molecules incorporate into micellar structures.

[0156] Traditionally, the impact of antiproliferative compounds on cell growth has been most commonly quantified by the IC 50 value, which is defined as that concentration of compound that reduces the rate of cell proliferation to one half that observed in the absence of compound (i.e., for the vehicle or solvent control sample). The IC 50 , however, does not allow the investigator to differentiate between cytostatic and cytotoxic compounds. The LCC, in contrast, readily allows one to make such a differentiation and to further quantify the concentration at which the transition to robust cytotoxic behavior occurs.

[0157] If one limits the observation time window to between the start of impact and the end of the experiment, then the data will generally fit well to a linear equation when plotted as ln(N t ) as a function of time (vide supra). From fits of this type, the value of k p can be determined at each concentration of compound tested. A replot of the value of k p as a function of compound concentration ([I]) will have the form of a descending isotherm, with a maximum value at [I] = 0 of k max (defined by the vehicle or solvent control sample) and a minimum value at infinite com ound concentration of k m i n .

mid

where I mi( j is the concentration of compound yielding a value of k p that is midway between the values of k max and k min (note that the value of I mi( j is not the same as the IC 50 , except in the case of a complete and purely cytostatic compound). Thus, fitting the replot data to equation A.7 provides estimates of k max , k m i n and I m i d - If a compound is cytostatic (as defined here), the value of k min cannot be less than zero. For cytotoxic compounds, k min will be less than zero and the absolute value of k min will relate directly to the effectiveness of the compound in killing cells. [0159] The fitted values derived from equation A.7 can also be used to determine the value of the LCC. By definition, when [I] = LCC, k p = 0. Thus, under these conditions equation A.7 becomes.

Q ( ^rnax ^rnin )

[0160] „ LCC (A.8)

1 +

mid

[0161] Algebraic rearrangement of equation A.8 yields an equation for the LCC.

[0163] This analysis is simple to implement with nonlinear curve fitting software and may be applied during cellular assays of compound activity throughout the drug discovery and development process. In this manner, the LCC may provide a valuable metric for the assessment of compound SAR (structure- activity relationship).

[0164] Example 30: In vivo Assays

Mice

[0165] Female Fox Chase SCID ® Mice (CB17/Icr-PrMc s «y/IcrIcoCrl, Charles River Laboratories) or athymic nude mice (Crl:NU(Ncr)-i¾jOTim<, Charles River Laboratories) are 8 weeks old and had a body-weight (BW) range of 16.0-21.1 g on Dl of the study. The animals are fed ad libitum water (reverse osmosis 1 ppm CI) and NIH 31 Modified and Irradiated Lab Diet ® consisting of 18.0% crude protein, 5.0% crude fat, and 5.0% crude fiber. The mice are housed on irradiated Enrich- o' cobs™ bedding in static microisolators on a 12-hour light cycle at 20-22 °C (68-72 °F) and 40-60% humidity. All procedures comply with the recommendations of the Guide for Care and Use of Laboratory Animals with respect to restraint, husbandry, surgical procedures, feed and fluid regulation, and veterinary care.

Tumor Cell Culture

[0166] Human lymphoma cell lines line are obtained from different sources (ATCC, DSMZ), e.g., WSU-DLCL2 obtained from DSMZ. The cell lines are maintained at Piedmont as suspension cultures in RPMI-1640 medium containing 100 units/mL penicillin G sodium salt, 100 g/mL streptomycin, and 25 g/mL gentamicin. The medium is supplemented with 10% fetal bovine serum and 2 mM glutamine. The cells are cultured in tissue culture flasks in a humidified incubator at 37 °C, in an atmosphere of 5% CO2 and 95% air.

In Vivo Tumor Implantation

[0167] Human lymphoma cell lines, e.g., WSU-DLCL2 cells, are harvested during mid-log phase growth, and re-suspended in PBS with 50% Matrigel™ (BD Biosciences). Each mouse receives 1 x 10 cells (0.2 mL cell suspension) subcutaneously in the right flank. Tumors are calipered in two dimensions to monitor growth as the mean volume approached the desired 80-120 mm 3 range. Tumor size, in mm 3 , is calculated from:

1 utnor vol ,ume ~— w " ^ x I

where w = width and / = length, in mm, of the tumor. Tumor weight can be estimated with the assumption that 1 mg is equivalent to 1 mm? of tumor volume. After 10-30 days mice with 108-126 mm tumors are sorted into treatment groups with mean tumor volumes of 117-119 mm 3 .

Test Articles

[0168] Test compounds are stored at room temperature and protected from light. On each treatment day, fresh compound formulations are prepared by suspending the powders in 0.5% sodium carboxymethylcellulose (NaCMC) and 0.1% Tween® 80 in deionized water. Compound 141 (free base) is dissolved in sterile saline and the pH is adjusted to 4.5 with HC1 fresh every day. The vehicles, 0.5% NaCMC and 0.1% Tween ® 80 in deionized water or sterile saline pH 4.5, are used to treat the control groups at the same schedules.

Formulations are stored away from light at 4 °C prior to administration. Unless otherwise specified, compounds referered to and tested in this experiment are in their specific salt forms mentioned in this paragraph.

Treatment Plan

[0169] Mice are treated at compound doses ranging from 12.5 - 600 mg/kg and at TID (three time a day every 8h), BID (2 times a day every 12 h) or QD (once a day) schedules for various amounts of days by oral gavage or injections via the intraperitoneal route. Each dose is delivered in a volume of 0.2 mL/20 g mouse (10 mL/kg), and adjusted for the last recorded weight of individual animals. The maximal treatment length is 28 days. Median Tumor Volume (MTV) and Tumor Growth Inhibition (TGI) Analysis

[0170] Treatment efficacy is determined on the last treatment day. MTV(n), the median tumor volume for the number of animals, n, evaluable on the last day, is determined for each group. Percent tumor growth inhibition (%TGI) can be defined several ways. First, the difference between the MTV(n) of the designated control group and the MTV(n) of the drug-treated group is expressed as a percentage of the MTV(n) of the control group: a / Trj M7T( ) e0ntr0 j - ΜΤν( η )„ βαίβ ^ nn

' J} 01 = 1 M¾ 1 * 100

[0171] Another way of calculating %TGI is taking the change of the tumor size from day 1 to day n into account with n being the last treatment day.

■ AMW con ,,. pi - M1TV trprt ,

= ( ^ ) X 1M

AMTV C0!ltrd Λ?7¥: .- ·„, , , - MTV(r) C01ltrd

ΑΜΤν Μά = MTVin) treated - MTV(l) treated

Toxicity

[0172] Animals are weighed daily on Days 1-5, and then twice weekly until the completion of the study. The mice are examined frequently for overt signs of any adverse, treatment related side effects, which are documented. Acceptable toxicity for the maximum tolerated dose (MTD) is defined as a group mean BW loss of less than 20% during the test, and not more than 10% mortality due to TR deaths. A death is to be classified as TR if it is attributable to treatment side effects as evidenced by clinical signs and/or necropsy, or due to unknown causes during the dosing period. A death is to be classified as NTR if there is evidence that the death is unrelated to treatment side effects. NTR deaths during the dosing interval would typically be categorized as NTRa (due to an accident or human error) or NTRm (due to necropsy-confirmed tumor dissemination by invasion and/or metastasis). Orally treated animals that die from unknown causes during the dosing period may be classified as NTRu when group performance does not support a TR classification and necropsy, to rule out a dosing error, is not feasible. Sampling

[0173] On days 7 or 28 during the studies mice are sampled in a pre-specified fashion to assess target inhibition in tumors. Tumors are harvested from specified mice under RNAse free conditions and bisected. Frozen tumor tissue from each animal is snap frozen in liquid N 2 and pulverized with a mortar and pestle.

Statistical and Graphical Analyses

[0174] All statistical and graphical analyses are performed with Prism 3.03 (GraphPad) for Windows. To test statistical significance between the control and treated groups over the whole treatment time course a repeated measures ANOVA test followed by Dunnets multiple comparison post test or a 2 way ANOVA test are employed. Prism reports results as non- significant (ns) at P > 0.05, significant (symbolized by "*") at 0.01 < P < 0.05, very significant ("**") at 0.001 < P < 0.01 and extremely significant ("***") at P < 0.001. Histone Extraction

[0175] For isolation of histones, 60-90 mg tumor tissue is homogenized in 1.5 ml nuclear extraction buffer (10 mM Tris-HCl, 10 mM MgCl 2 , 25 mM KC1, 1% Triton X-100, 8.6% Sucrose, plus a Roche protease inhibitor tablet 1836145) and incubated on ice for 5 minutes. Nuclei are collected by centrifugation at 600 g for 5 minutes at 4 °C and washed once in PBS. Supernatant is removed and histones extracted for one hour, with vortexing every 15 minutes, with 0.4 N cold sulfuric acid. Extracts are clarified by centrifugation at 10,000 g for 10 minutes at 4 °C and transferred to a fresh microcentrifuge tube containing lOx volume of ice cold acetone. Histones are precipitated at -20 °C for 2 hours- overnight, pelleted by centrifugation at 10,000 g for 10 minutes, and resuspended in water.

ELISA

[0176] Histones are prepared in equivalent concentrations in coating buffer

(PBS+0.05%BSA) yielding 0.5 ng/ul of sample, and 100 ul of sample or standard is added in duplicate to 2 96-well ELISA plates (Thermo Labsystems, Immulon 4HBX #3885). The plates are sealed and incubated overnight at 4 °C. The following day, plates are washed 3x with 300 ul/well PBST (PBS+0.05% Tween 20; 10X PBST, KPL #51-14-02) on a Bio Tek plate washer. Plates are blocked with 300 ul/well of diluent (PBS+2%BSA+0.05% Tween 20), incubated at RT for 2 hours, and washed 3x with PBST. All antibodies are diluted in diluent. 100 ul/well of anti-H3K27me3 (CST #9733, 50% glycerol stock 1 : 1,000) or anti- total H3 (Abeam abl791, 50% glycerol 1: 10,000) is added to each plate. Plates are incubated for 90 min at RT and washed 3x with PBST. 100 ul/well of anti-Rb-IgG-HRP (Cell Signaling Technology, 7074) is added 1:2,000 to the H3K27Me3 plate and 1:6,000 to the H3 plate and incubated for 90 min at RT. Plates are washed 4X with PBST. For detection, 100 ul/well of TMB substrate (BioFx Laboratories, #TMBS) is added and plates incubated in the dark at RT for 5 min. Reaction is stopped with 100 ul/well IN H 2 SO 4. Absorbance at 450 nm is read on SpectaMax M5 Microplate reader.

7 day PD study

[0177] In order to test whether a compound can modulate the H3K27me3 histone mark in tumors in vivo, WSU-DLCL2 xenograft tumor bearing mice are treated with the compound at either 200 mg/kg BID or 400 mg/kg QD or vehicle (BID schedule) for 7 days. There are 4 animals per group. Animals are euthanized 3 h after the last dose and tumor is preserved in a frozen state as described above. Following histone extraction the samples are applied to ELISA assays using antibodies directed against the trimethylated state of histone H3K27 (H3K27me3) or total histone H3. Based on these data the ratio of globally methylated to total H3K27 is calculated. The mean global methylation ratios for all groups as measured by ELISA indicate target inhibition range compared to vehicle.

28 day efficacy study in WSU-DLCL2 xenograft model

[0178] In order to test whether a compound could induce a tumor growth inhibition in vivo WSU-DLCL2 xenograft tumor bearing mice are treated with the compound at 12.5, 25 or 50 mg/kg QD for 28 days via intraperitoneal injection. Tumor volume and body weights are determined twice a week. A parallel cohort of mice (n=4 per group) is treated at the same doses for 7 days, and mice are euthanized on day 7, 3 h after the last dose for tumor sampling and assessment of target inhibition. The result of the ELISA measuring global methylation of H3K27me3 normalized to total H3 is determined.

Efficacy study with increasing doses in WSU-DLCL2 xenograft model

[0179] In order to test whether a compound could induce an anti-tumor effect in vivo, WSU-DLCL2 xenograft tumor bearing mice are treated with a compound at, e.g., 37.5, 75 or 150 mg/kg TID for 28 days. There are 12 mice per group for the efficacy arm of the experiment. A parallel cohort is dosed for 7 days at the same doses and schedules for assessment of target inhibition after 7 days (n=6 per group). The tumor growth over the treatment course of 28 days for vehicle and compound treated groups is measured. [0180] Histones are extracted from tumors collected after 7 days of dosing (parallel PD cohort) and at the end of the study on day 28 for the efficacy cohort (3h after the last dose for both cohorts). The H3K27me3 methyl mark is assessed for modulation with treatment in a dose dependent matter.

Efficacy study at different dose schedules

[0181] To assess whether a compound would lead to tumor growth inhibition at other dosing schedules but TID a WSU-DLCL2 xenograft efficacy study is performed where TID, BID and QD schedules are compared side by side. There are 12 animals per group, and mice are treated for 28 days. The tumor growth over the treatment course of 28 days for vehicle and compound treated groups is measured.

[0182] On day 28 mice are euthanized and tumors are collected 3h after the last dose for assessment of target inhibition.

[0183] Example 31: Anti-cancer effect on the KARPAS-422 human diffused large B-Cell lymphoma mouse xenograft model

[0184] A test compound is analyzed for its anti-cancer activity in KARPAS-422 mouse xenograft model, which is a human diffused large B-Cell lymphoma xenograft model. 45 female of CAnN.Cg-Foxnlnu/CrlCrlj mice (Charles River Laboratories Japan) with KARPAS-422 tumors whose mean tumor volume (TV) reaches approximately 150 mm are selected based on their TVs, and are randomly divided into five groups. The oral administration of compound (e.g., 80.5, 161, 322, and 644 mg/kg) or vehicle is started on day 1. Compound is given once daily on day 1 and day 29 and twice daily everyday from day 2 to day 28. The administration volume (0.1 mL/10 g body weight) is calculated from the body weight before administration. The TV and body weight are measured twice a week. The design for this experiment is shown in Table 6.

Table 6 Dosing Scheme

[0185] TV is calculated from caliper measurements by the formula for the volume of a prolate ellipsoid (LxW )/2 where L and W are the respective orthogonal length and width measurements (mm).

[0186] Data are expressed as the mean + standard deviation (SD). The differences in TV between the vehicle-treated and compound -treated groups are analyzed by a repeated measures analysis of variance (ANOVA) followed by the Dunnett-type multiple comparison test. A value of P < 0.05 (two sided) is considered statistically significant. Statistical analyses are performed using the Prism 5 software package version 5.04 (GraphPad Software, Inc., CA, USA).

[0187] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.

[0188] The invention can be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.