Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SULFONAMIDES AS INHIBITORS OF BCL-2 FAMILY PROTEINS FOR THE TREATMENT OF CANCER
Document Type and Number:
WIPO Patent Application WO/2011/029842
Kind Code:
A1
Abstract:
The present invention includes novel compound and methods of treating a disease or disorder by antagonizing Bcl-2 family proteins, particularly compounds of Formula (I) or pharmaceutically acceptable salt thereof, as well as methods of treating a disease, disorder, or syndrome associated with Bcl-2 inhibition, particularly hyperproliferative diseases. The present invention also includes pharmaceutical compositions including compounds of formula (I) and pharmaceutically acceptable salts thereof.

Inventors:
MILLER-MOSLIN KAREN (US)
TOURE BAKARY-BARRY (US)
VISSER MICHAEL SCOTT (US)
YUSUFF NAEEM (US)
Application Number:
PCT/EP2010/063169
Publication Date:
March 17, 2011
Filing Date:
September 08, 2010
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NOVARTIS AG (CH)
MILLER-MOSLIN KAREN (US)
TOURE BAKARY-BARRY (US)
VISSER MICHAEL SCOTT (US)
YUSUFF NAEEM (US)
International Classes:
C07D471/04; A61K31/519; A61P35/00; C07C311/20
Domestic Patent References:
WO2008061208A22008-05-22
WO2004078163A22004-09-16
Other References:
RAFFELD ET AL., CANCER RESEARCH, vol. 47, no. 10, 1987, pages 2537 - 42
OLTERSDORF ET AL., NATURE, vol. 435, 2005, pages 677 - 681
DENG ET AL., CANCER CELL, vol. 12, no. 2, 2007, pages 171 - 185
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-19, 1967, WILEY
T. W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
"Remington's Pharmaceutical Sciences, 20th ed.,", 1985, MACK PUBLISHING COMPANY
STAHL; WERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
WENDT, M. D. ET AL., J. MED. CHEM., vol. 49, 2006, pages 1165 - 1181
WENDT, M. D. ET AL., J MED. CHEM., vol. 49, 2006, pages 1165 - 1181
PARK, C. ET AL., J MED. CHEM., vol. 51, 2008, pages 6902 - 6915
WENDT, M. D. ET AL., J. MED. CHEM, vol. 49, 2006, pages 1165 - 1181
Attorney, Agent or Firm:
STRANG, Andrea (Patent Department, Basel, CH)
Download PDF:
Claims:
What is Claimed is:

1. A compound of formula I:

(I)

wherein

A is a divalent bicyclic radical comprising a saturated cyclic structure and an unsaturated cyclic structure, wherein said unsaturated cyclic structure is attached to NH, and said saturated cyclic structure is attached to R1, wherein A is unsubstituted or substituted with one or more substituents each independently selected from halogen, OH, (Ci-C6)alkyl, halo-substituted (C C6)alkyl, CN, or NR10Rn;

R1 is 3- to 8-membered cycloheteroalkyl, (C3-C8)cycloalkyl, or (C6-Ci )aryl group which is unsubstituted or substituted with (Ci-C6)alkyl, halogen, OR57, NR58R59, or deuterium;

L1 is (Ci-C3)alkylene, (C,-C4)alkenylene, -C(O)-, -C(0)0-, C(0)N-, -(Cr

C3)alkyleneC(0)-, -(C|-C3)alkylene-C(0)0-, or a bond, wherein L1 is unsubstituted or substituted by one or more substituents each independently selected from (C C4)alkyl, halo-substituted (Ci-C4)alkyl, or (Cj-C8)cycloalkyl;

L2 is (Q-C^alkylene, NR9, -0-, or -S-;

R2 is (C6-Ci )aryl, 5- to 14-membered heteroaryl, 3- to 8-membered

cycloheteroalkyl, or (C3-Ci4)cycloalkyl, each of which is unsubstituted or substituted with one or more of halogen, OH, (Ci-C6)alkyl, halo-substituted (Ci-C6)alkyl, or CN;

R8 is (C C6)alkyl, (C6-C14) aryl, C3-CM)cycloalkyl, halogen, or 3- to 14- membered cycloheteroalkyl, in which any of the aforementioned hydrocarbon groups is optionally substituted by one or more substituents each independently selected from halogen, (Ci-C6)alkyl, halo-substituted (Ci-C6)alkyl, OH, or NR R45; R3 and R4 are each independently H, (Ci-C6)alk l, (Ci-C6)alkylene-(C6-Ci4)aryl, (C!-C6)alkylene-(5- to 14-membered heteroaryl), (C C6)alkylene-CONRI6R17, (Cr C6)alkylene-0-R15, (C,-C6)alkylene-NR13R14, (d-C6)alkylene-(3- to 14-membered cycloheteroalkyl), (C|-C3)alkylene-S-(C6-C,4)aryl, (Ci-C6)alkylene-COR18, (Ci- C6)alkylene-C(0)0-R19, (CrC6)alkylene-0-C(0)-R20, (C C3)alkylene-S-R21, (C,- C3)alkylene-SOR22, or (CrC3)alkylene-S02R23;

R6 and R7 are each independently H, N02, S02CF3, S02(Ci-C6)alkyl, halo- substituted (Ci-C6)alkyl, halogen, (C3-C]4)cycloalkyl, or CN;

R9 is H, (C,-C6)alkyl, -(C,-C3)alkylene-(C6-C,4)aryl, -(Ci-C3)alkylene-(5- to 14- membered heteroaryl), -(C,-C3)alkylene-CONR46R47, -(Ci-C3)alkylene-0-R4S, -(C,- C3)alkylene-NR49R50, -(C C3)alkylene-(3- to 14-membered cycloheteroalkyl), -(d-

-(CrC3)alkylene-0-C(0)-R53, -(C C3)alkylene-S-R54, -(C|-C3)alkylene-SOR55, -(C,- C3)alkylene-S02R56, -C(O) (Ci-C6)alkyl, -C(0)(C1-C3)alkylene-(C6-Ci4)aryl, -C(O) (d- C3)alkylene-(5- to 14-membered heteroaryl), -C(O) (Ci-C3)alkylene-(3- to 14-membered cycloheteroalky), -C(O) (Ci-C3)alkylene-CONR46R47, -C(O) (C C3)alkylene-0-R48, -C(O) (CrC3)alkylene-NR49R50, -C(O) (CrC3)alkyl-S-(C6-C14)aryl, -C(O) (C,- C3)alkylene-C(0)R51, -C(O) (CrC3)alkylene-C(0)0-R52, -C(O) (d-C3)alkylene-0- C(0)-R53, -C(O) (C,-C3)alkylene-S-R54, -C(O) (CrC3)alkylene-SOR55, or -C(O) (C,- C3)alkylS02R56;

R]0, R1 1, R13, R14 , R16, R17 R44, R45, R46, R47, R49, R50 , R57, R58, and R59 are each independently H, (C,-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, OH, -C(0)(Ci-C6)alkyl, (Ci-C6)alkoxy, halogen, (C3-Ci4)cycloalkyl, (C6-C14)aryl, 4- to 14-membered cycloheteroalkyl, or 5- to 14-membered heteroaryl, wherein each of the aforementioned hydrocarbon groups is optionally substituted by one or more substituents each independently selected from halogen, hydroxyl, (C[-C6)alkoxy, amino, (Cr

C6)alkylamino, di((C C6)alkyl)amino or cyano;

or R13 and R14 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or 5- to 14 membered-heteroaryl, each of which is substituted or unsubstituted; or R16 and R17 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R58 and R59 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R44 and R45 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R46 and R47 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R49 and R50 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

R15, R18, R19, R20, R22, R23, R48, R51, R52, R53, R55, and R56 are each independently -(CrC^alkylene-iQ-C^aryl, or -(CrC6)alkylene-(5- to 14-membered heteroaryl), each of which is unsubstituted or substituted with one or more substituents each independently selected from halogen, OH, (Ci-Cejalkyl, halo-substituted (Ci-C6)alkyl, or CN;

R21 and R54 are each independently (C6-C14)aryl, 5- to 14-membered heteroaryl,

-(Ci-C6)alkylene-(C6-Ci4)aryl, or -(Ci-C6)alkylene-(5- to 14-membered heteroaryl), each of which is unsubstituted or substituted with one or more substituents each independently selected form halogen, OH, (Ci-C6)alkyl, halo-substituted (d- Jalkyl, or CN;

or a pharmaceutically acceptable salt thereof.

2. The compound of formula (I) according to claim 1 wherein A is:

where G1, G2, and G3, are each independently CR29 or N;

Y1 and Y2 are each independently -CR30R31-, -CR32R33-CR34R35-, -NR36-, -CR37R38-NR39-, -0-, -S-, -CR40R41-O-, or -CR42R43-S-;

X is CH or ; and

R29, R30, R31, R32, R33, R34, R35, R36, R37, R38, R39, R40, R41, R42, and R43 are each independently H, (CrC6)alkyl, halogen, halo-substituted (CrC6)alkyl, or (C3- Ci4)cycloalkyI;

or a pharmaceutically acceptable salt thereof.

3. The compound of formula (1) according to claim 2 wherein A is

wherein

X is CH or N; G is CR^, or N; G4 is O, S, or NR0U; and

R29, R30, R31 , R32, R33, R34, R35, and R60 are each independently H, methyl, ethyl, propyl, CI, F, or CF3;

or a pharmaceutically acceptable salt thereof. The compound of formula (I) according to Claims 2 wherein A

where at least one of G', G2, and G3 are N;

Y1 and Y2 are each independently -CR30R31 -, or -CR32R3 -CR34R35-;

X is N, and

R30, R31, R32, R33, R34, and R35 are each independently H, methyl, ethyl, propyl, CI, F, or CF3

or a pharmaceutically acceptable salt thereof.

5. The compound of formula (I) according to claim 4 wherein two of G!, G2, and G3 are N, and the remaining G is CR29;

Y1 is-CR32R33-CR34R35-;

Y2 is-CR30R31-;

R29 is H, methyl, CI, F, CF3;

R32, R33, R34, and R35 are H;

R30 and R31 are H; and

X is N;

or a pharmaceutically acceptable salt thereof.

6. The compound of formula (I) according to claims 4 or 5 wherein G1 and G3 are N, and G2 is CR29;

Y1 is -CR32R33-CR34R35-;

Y2 is -CR30R31-;

R29 is H, methyl, CI, F, or CF3;

R32, R33, R34, and R35 are H; and

X is N;

or a pharmaceutically acceptable salt thereof.

7. The compound of formula (I) according to any one of the proceding claims wherein R1 is piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or phenyl each of which is

unsubstituted or substituted with one or more substituents each independently selected from methyl, ethyl, propyl, CI, F, Br, I, or methoxy; or a pharmaceutically acceptable salt thereof. 8. The compound of formula (I) according to any one of the proceding claims wherein R1 is piperidinyl, piperazinyl, morpholinyl, cyclopentyl, or cyclohexyl, each of which is unsubstitued or substituted with methyl, methoxy, CI, or F; or a pharmaceutically acceptable salt thereof. 9. The compound of formula (I) according to any of the preceding claims wherein L1 is -CH2-, -CH2CH2-, or a bond; and L2 is -CH2- or NH; or a pharmaceutically acceptable salt thereof.

10. The compound of formula (I) according to any one of the proceding claims wherein R2 is phenyl, naphthyl, pyridinyl, pyrimidinyl, or pyridazinyl; each of which is unsubstituted or substituted with one or more substituents each independently selected from methyl, dimethyl, ethyl, propyl, butyl, pentyl, hexyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, CF3, or CH2C1; or a pharmaceutically acceptable salt thereof. 1 1. The compound of formula (I) according to any of claims 1 -10 wherein R and R4 are independently (Ci-C6)alkyl, (C| -C3)alkylene-(C6-Ci4)aryl, (Ci-C3)alkylene-(5- to 14-membered heteroaryl), (C C3)alkylene-CONR16R17, (Ci-C3)alkylene-0-R15, (d- C3)alkylene-NR13R14, (CrC3)alkylene-(3- to 14-membered cycloheteroalkyl), or (C, - C3)alkylene-S-(C6-Ci4)aryl; or a pharmaceutically acceptable salt thereof.

12. The compound of formula (I) according to any one of the preceding claims wherein

R3 is -CH2-phenyl, -CH2CH2-phenyl, -CH2CH2CH2-phenyl, -CH2-pyridinyl, -CH2CH2-pyridinyl, -CH2-pyrimidinyl, -CH2CH2-pyrimidinyl, -CH2C(0)NCH3CH3, -CH2C(0)NHCH3, -CH2CH2C(0)NCH3CH3, -CH2-0-CH3, -CH2-0-CH2CH3, -CH2-S- phenyl, -CH2CH2-S-phenyl, or -CH2CH2CH2-S-phenyl; and

R4 is -CH2C(0)CH3, -CH2C(0)CH2CH3, -CH2CH2C(0)CH3, -CH2-S-CH3, CH2- S-CH2CH3,-CH2-N(CH3)(CH3), -CH2-NHCH3, -CH2-NH2, -CH2-NH-phenyl, -CH2-CH2- N(CH3)(CH3), -CH2CH2-NHCH3, -CH2CH2-NH2, or -CH2CH2-NH-phenyl;

or a pharmaceutically acceptable salt thereof.

13. A compound of formula (lb) :

(lb)

wherein

Rlb is H, N02, S02CF3, S02(C|-C6)alkyl, halo-substituted (Ci-C6)alkyl, halogen, (C3-Ci4)cycloalkyl, or CN;

R2b is a divalent bicyclic radical comprising a saturated cyclic structure and an unsaturated cyclic structure, wherein said unsaturated cyclic structure is attached to NH, and said saturated cyclic structure is attached to R5b, wherein R2b may unsubstituted or substituted with one or more of halogen, OH, (Ci-C6)alkyl, halo-substituted (Ci-C6)alkyl, CN or NH2;

R3b and R4b are independently H, (C,-C6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, (C6-C|4)aryl, 5- to 14-membered heteroaryl, (C3-Ci )cycloalkyl, halogen, or 3- to 14- membered cycloheteroalkyl, each of which when not H is unsubstituted or substituted with one or more substituents each independently selected from hydroxyl, cyano, nitro, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C,-C8)alkoxy,(C2-C8)alkenyloxy, (C2- C8)alkynyloxy, halogen, (Ci-C8)alkylcarbonyl, carboxy, (C|-C8)alkoxycarbonyl, amino, (Ci-C8)alkylamino, di((C]-C8)alkyl)amino, (C]-C8)alkylaminocarbonyl, di((Ci- C8)alkyl)aminocarbonyl, (Ci-C8)alkylcarbonylamino, (CrC8)alkylcarbonyl(Ci-

C8)aIkyl)amino, (Ci-C8)alkylsulfonylamino,(C,-C8)alkylthio, (Ci-C8)alkylsulfinyl, (Cr C8)alkylsulfonyl, aminosulfonyl, (Ci-C8)alkylaminosulfonyl or di((Cr

Cg)alkyl)aminosulfonyl, where each of the afore- mentioned hydrocarbon groups is optionally substituted by one or more substituents each independently selected from halogen, hydroxyl or (C|-C8)alkoxy;

R5 is a 3- to 8-membered cycloheteroalkyl, (C3-C8)cycloalkyl, or (C6-Ci4)aryl which is unsubstituted or substituted with (Ci-C6)alkyl, halogen, OH, (Ci-C3)alkoxy, NH2, or deuterium;

R6b is Lb-R8b;

R7b is hydroxyl, cyano, nitro, (C,-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C2-

C8)alkoxy, (C2-C8)alkenyloxy, (C2-C8)alkynyloxy, halogen, (Ci-C8)alkylcarbonyl, carboxy, (Ci-Cg)alkoxycarbonyl, amino, (C]-C8)alkylamino, di(CrC8)alkylamino, (Cj- C8)alkylaminocarbonyl, di((C|-Cg)alkyl)aminocarbonyl, (C C8)alkylcarbonyIamino, (Ci- C8)alkylcarbonyl((Ci-C8)alkyl)amino, (Ci-C8)alkylsulfonylamino, (C]-C8)alkylthio, (Cj- C8)alkylsulfinyl, (C|-C8)alkylsulfonyl, aminosulfonyl, (Ci-C8)alkylam'tnosulfonyl or di((Ci-C8)alkyl)aminosulfonyl, where each of the afore-mentioned hydrocarbon groups is optionally substituted by one or more substituents each independently selected from halogen, OH or (CrC8)alkoxy;

Lb is -(C, -C3)alkylene-, -(C2-C4)alkenylene-, -C(O)-, -C(0)0-, -C(0)N(H)-, -(d- C3)alkylC(0)-, -(Ci-C3)alkyl-C(0)0-, or a bond, wherein Lb is unsubstituted or substituted by one or more (Ci-C4)alkyl, halo-substituted(Ci-C4)alkyl, or (C3- C8)cycloalkyl;

R8b is (C6-Ci4)aryl, a 5- to 14-membered heteroaryl, a 3- to 8-membered cycloheteroalkyl, or a (C3.Ci4)cycloalkyl, each of which is unsubstituted or substituted with one or more substituents each independently selected form halogen, OH, (Ci - C6)alkyl, halo-substituted(Ci-C6)alkyl, or CN; and n is 0, 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof.

14. A method of inhibiting Bcl-2 activity comprising the step of administering to a subject in need thereof (i) a therapeutically effective amount of a compound according to any one of claims 1-13, or a pharmaceutically acceptable salt thereof, or (ii) a pharmaceutical composition comprising a compound according to any one of claims 1- 13, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.

15. A method of treating a proliferative disease comprising the step of administering to a subject in need thereof (i) a therapeutically effective amount of a compound of claims 1-13, or a pharmaceutically acceptable salt thereof, or (ii) a pharmaceutical composition comprising a compound of claims 1-13, or a

pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.

16. The method of treating as claimed in claim 15 wherein the disease, disorder, or syndrome is hyperproliferative in a subject, wherein said subject is an animal and said disease, disorder or syndrome is cancer or inflammation.

17. The compound according to any one of claims 1 - 13 for use in therapy.

18. A pharmaceutical composition comprising a compound of formula I according to any one of claims 1-13, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.

Description:
SULFONAMIDES AS INHIBITORS OF BCL-2 FAMILY PROTEINS FOR THE

TREATMENT OF CANCER

FIELD OF THE INVENTION

The present invention relates to sulfonamide compounds that bind to and inhibit Bcl-2 family proteins, and uses thereof for the treatment of diseases associated with such inhibition. Consequently, the present invention includes sulfonamide compounds, compositions thereof, methods of their use, and methods of their manufacture, where such compounds are generally pharmacologically useful in therapies whose mechanism of action rely on the inhibition of Bcl-2 family proteins, and more particularly in therapies for the treatment of proliferative diseases, including cancer.

BACKGROUND

Bcl-2 family proteins are anti-apoptotic proteins associated with cancer and other diseases. Bcl-2 family proteins under investigation as therapeutic targets include Bcl-2 (BCL2), Bcl-xL (BCL2L I ), Bcl-w (BCL2L2), A 1 (BCL2A 1 ), and MCL 1 (MCL 1 ).

Numerous cancers express one or more Bcl-2 family proteins leading to cancer cell survival and resistance to chemotherapeutics. For example, chromosomal translocation of Bcl-2, t(14;18), is a transforming event in some cancer cells (Raffeld, et al. Cancer Research 1987, 47(10):2537-42), and these cancer cells demonstrate dependence on Bel- 2 for survival based on RNAi (figure 1), and sensitivity to recently described small molecule inhibitors of Bcl-2 family proteins (Oltersdorf, et al. Nature 2005, 435, 677- 681 : Deng et al. Cancer Cell 2007, 12(2), 171-185). This invention is directed to a series of compounds that inhibit Bcl-2 family proteins and promote apoptosis of cancer cells, alone or in combination with other chemotherapeutics.

In addition to B-cell lymphomas, Bcl-2 family antagonists have been shown to be useful for treating cancers that express Bcl-2 family members and/or have deregulated apoptosis such as chronic lymphocytic leukemia, diffuse large B-cell lymphomas, follicular lymphomas, chronic or acute leukemia, chronic myeloid leukemia, lymphoid malignancies of T-cell or B-cell origin, small cell lung cancer, non-small cell lung cancer, melanoma or other skin cancers, multiple myeloma, ovarian cancer, breast cancer, colon cancer, gastrointestinal cancer (gastric, colorectal, and duodenal), prostate cancer, bladder cancer, uterine cancer, cervical cancer, sarcoma of soft tissue origin, pancreatic cancer,

l kidney cancer, brain tumors, hepatocellular cancer, head and neck cancer, cervical cancer, fibrosarcoma, and other cancers.

SUMMARY

The present invention provides compounds and pharmaceutical formulations thereof that are useful in the treatment of diseases and/or disorders modulated by the inhibition of BcI-2.

Compounds of formula (I) are provided herein

(I)

wherein

A is a divalent bicyclic radical comprising a saturated cyclic structure and an unsaturated cyclic structure, wherein said unsaturated cyclic structure is attached to NH, and said saturated cyclic structure is attached to R 1 , wherein A is unsubstituted or substituted with one or more of halogen, OH, (Ci.C 6 )aIkyl, halo-substituted(Ci-C 6 )aIkyI, CN or NR'V 1 ;

R 1 is a 3- to 8-membered cycloheteroalkyl group, a (C 3 -C 8 )cycIoaIkyI group, or a (C 6- Ci4)aryl group which is unsubstituted or substituted with (Ci-C 6 )alkyl, halogen, OR 57 , NR 58 R 59 , or deuterium;

L 1 is (Ci-C 3 )alkylene, (C)-C 4 )alkenylene, -C(O)-, -C(0)0~, C(0)N-, -(d- C 3 )alkylene-C(0)-, -(Ci-C 3 )alkylene-C(0)0-, or a bond, wherein L 1 is unsubstituted or substituted by one or more (Ci-C 4 )alkyl, halo-substituted (Ci-C 4 )haloaIkyl, or (C 3 - C 8 )cycloalk l;

L 2 is (Ci-C 3 )alkylene, NR 9 , -0-, or -S-;

R is (C 6 -Ci 4 )aryl group, 5- to 14-membered heteroaryl, 3- to 8-membered cycloheteroalkyl group, or (C 3 .Ci 4 )cycIoaIkyl group, each of which is unsubstituted or substituted with one or more of halogen, OH, (Ci-C 6 )alkyl, halo-substituted(Ci-C 6 )alkyl, or CN;

R 8 is (C|-C 6 )alkyl, (C 6 -C 14 ) aryl, (C 3- C 14 )cycloalkyl, halogen, or 3-to 14- membered cycloheteroalkyl, in which any the aforementioned hydrocarbon groups (e.g., (C]-C 6 )alkyl, (C 6 -C| 4 ) aryl, and (C 3- C 14 )cycloalkyl) is optionally substituted with one or more substituents each independently selected from halogen, (C|-C 6 )alkyl, halo- substituted (C,-C 6 )alkyl, OH, or NR 44 R 45 ;

R 3 and R 4 are each independently H, (C|-C 6 )alkyl, -(C Ceialkylene-fCe-CuJaryl, -(C,-C 6 )alkylene-(5-to 14-membered heteroaryl), -(C,-C 6 )alkylene-CONR 16 R 17 , -(d- C 6 )alkylene-0-R 15 , -(C,-C 6 )alkylene-NR I 3 R 14 , -(C,-C 3 )alkylene-(3- to 14-membered cycloheteroalkyl), group, -(C|-C 6 )alkylene-COR 18 , -(C,- C 6 )alkylene-C(0)0-R 19 , -(C,-C 6 )alkylene-0-C(0)-R 20 , -(Ci-C 3 )alkylene-S-R 21 , -(Ci- C 3 )alkylene-SOR 22 , or -(C,-C 3 )alkylene-S0 2 R 23 ;

R 6 and R 7 are each independently H, N0 2 , -S0 2 CF 3 , -S0 2 (C,-C 6 )alkyl, halo- substituted (CrC 6 )alkyl, halogen, (C 3 .C i4 )cycloalkyl, or CN;

R 9 is H, (Ci-C 6 )alkyl, -(C,-C 6 )alkylene-(C 6 -C, 4 )aryl, -(C r C 3 )alkylene-(5- to 14- membered heteroaryl), -(C,-C 3 )alkylene-CONR 46 R 47 , -(C r C 3 )alkylene-0-R 48 , -(C r C 3 )alkyene-NR 49 R 50 , -(C C 3 )alkylene-(3- to 14-membered cycloheteroalkyl), -(C C 3 )alkyene-S-(C 6 -C l4 )aryl group, -(Ci-C 3 )alkylene-C(0)0- R 52 , -(C,-C 3 )alkylene-0-C(0)-R 53 , -(C,-C 3 )alkylene-S-R 54 , -(C,-C 3 )alkylene-SOR 5S , -

(C,-C 3 )alkylene-S0 2 R 56 , -C(0)-(C,-C 6 )alkyl, -C(0)-(C 1 -C 3 )alkylene-(C 6 -C 14 )aryl, -C(O)- (Ci-C 6 )alkylene-(5- to 14-membered heteroaryl), -C(0)-(Ci-C 3 )alkylene-(3- to 14- membered cycloheteroalkyl), -C(0)-(C 1 -C 3 )alkylene-C(0)NR 46 R 47 , -C(0)-(C

C 3 )alkylene-0-R 48 , C(O)-(Ci-C 3 )alkylene-NR 49 R 50 , -C(O)-(C 1 -C 3 )alkylene-S-(C 6 - C, 4 )aryl, -C(0)-(C,-C 3 )alkylene-C(0)R 51 , -C(0)-(C C 3 )alkylene-C(0)0-R 52 , -C(0)-(C]- C 3 )alkylene-0-C(0)-R 53 , -C(0)-(C 1 -C 3 )alkylene-S-R 54 , -C(0)-(Ci-C 3 )alkylene-SOR 55 , or C(0)-(C! -C 3 )alkylene-S0 2 R 56 ;

R 10 , R", R 13 , R 14 , R 16 , R 17 R 44 , R 45 , R 46 , R 47 , R 49 , R 50 , R 57 , R 58 , and R 59 are each independently H, (C t -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, OH, -C(0)-(C,-C 6 )alkyl, (C|-C 6 )alkoxy, halogen, (C 3- Ci 4 )cycloalkyl, (C 6 -Ci 4 ) aryl, 4- to 14-membered

cycloheteroalkyl, or 5- to 14-membered heteroaryl, wherein each of the aforementioned hydrocarbon groups (e.g., (Ci-C6)alkyl, (C 2 -C 6 )alkenyl, (C2-C 6 )alkynyl, (C 3 .

C 14 )cycloalkyl), (C 6 -C 14 ) aryl, and the (C C 6 )alkyl moieties of -C(0)-(Ci-C 6 )alkyl, and (Ci-C6)alkoxy) is optionally substituted by one or more halogen, hydroxyl, (Cj- C 6 )alkoxy, amino, (C]-C6)alkylamino, d((Ci-C6)alkyl)amino or cyano;

or R 13 and R 14 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or a 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R 16 and R 17 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or a 5-14 membered heteroaryl, each of which is substituted or unsubstituted;

or R 58 and R 59 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or a 5-14 membered heteroaryl, each of which is substituted or unsubstituted;

or R 44 and R 45 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or a 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R 46 and R 47 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or a 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

or R 49 and R 50 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl, or a 5- to 14-membered heteroaryl, each of which is substituted or unsubstituted;

R 15 , R 18 , R 19 , R 20 , R 22 , R 23 , R 48 , R 51 , R 52 , R 53 , R 55 , and R 56 are each independently -(Ci-C 6 )alkylene-(C6-Ci4)aryl, or -(C]-C 6 )alkylene-(5- to 14-membered heteroaryl), each of which is unsubstituted or substituted with one or more of halogen, OH, (C]-C 6 )alkyl, halo-substituted (Ci-C 6 )alkyl, or CN; and

R 21 and R 54 are each independently (C6-Ci 4 )aryl, 5- to 14-membered heteroaryl, -(C]-C 6 )alkylene-(C 6 -Ci4)aryl, or -(Ci-C 6 )alkylene-(5- to 14-membered heteroaryl), each of which is unsubstituted or substituted with one or more of halogen, OH, (Ci-C 6 )alkyl, halo-substituted (C C 6 )alkyl, or CN;

or a pharmaceutically acceptable salt thereof. In one embodiment, the present invention includes compounds of formula (I) where A is a divalent bicyclic radical comprising a saturated cyclic structure and an unsaturated cyclic structure, wherein said unsaturated cyclic structure is attached to NH, and said saturated cyclic structure is attached to R 1 , wherein A is unsubstituted or substituted with one or more of halogen, OH, (Ci-C6)alk l (e.g., methyl, ethyl, propyl, or butyl), halo-substituted (d-C 6 )alkyl (e.g., CF 3 , CH 2 CF 3 ), or CN.

In a pre

, or where

G 1 , G 2 , and G 3 are independently CR 29 or N; Y 1 and Y 2 are each independently -CR 30 R 31 -, -CR 32 R 33 -CR 34 R 35 -, -NR 36 -, -CR 37 R 38 -NR 39 -, -0-, -S-, -CR 40 R 4, -O-, or -CR 42 R 43 -S-;

X is CH or N; and

R 29 , R 30 , R 31 , R 32 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , and R 43 are each independently H, (Ci-C 6 )alkyl, halogen, halo-substituted (Ci-C 6 )alkyl, or (C 3- C 14 )cycloalkyl. Preferably, R 29 , R 30 , R 31 , R 32 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , and R 43 are each independently H, methyl, ethyl, propyl, F, CI, CF 3 , cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl; or a pharmaceutically acceptable salt thereof.

In another embodiment, the present invention includes compounds of formula (I) where A is:

and at least one of G 1 , G 2 , and G 3 is N;

Y 1 and Y 2 are independently -CR 30 R 31 -, -CR 32 R 33 -CR 34 R 35 -; and

X is N, and R 30 , R 31 , R 32 , R 33 , R 34 , and R 35 are each independently H, methyl, ethyl, propyl, CI, F, or CF 3 ; or a pharmaceutically acceptable salt thereof.

In a preferred embodiment, G , G , and G are each independently CH, C-CH 3 , C-CN, C-NR ,0 R n , C-CF 3 , C-F, C-Cl, or N.

1 3

In another preferred embodiment, G and G are N.

In another embodiment, Y 1 and Y 2 are each independently -CH 2 -, -CH 2 -CH 2 -, -NH-, -NCH 3 -, -CH 2 NH-, -0-, -S-, -CH 2 -0-, or -CH 2 -S-.

In yet another preferred embodiment, Y is -CH 2 CH 2 - and Y is -CH 2 -. In another embodiment, X is CH. In another preferred embodiment, X is N.

In another embodiment, X is N, and N is attached to a carbon atom of a heteroaryl or cycloheteroalkyl of R 1 , and L 1 is a bond.

In another embodiment where X is CH, X is attached to a heteroatom of a heteroaryl or cycloheteroalkyl of R 1 , and L 1 is a bond.

In another embodiment, the present invention includes compounds of formula (I) where two of G 1 , G 2 , and G 3 are N, and the remaining G is CR 29 ;

Y 1 is-CR 32 R 33 -CR 34 R 35 -;

Y 2 is-CR 30 R 31 -;

R 29 is H, methyl, CI, F, CF 3 ;

R 32 , R 33 , R 34 , and R 35 are H;

R 30 and R 31 are H; and X is N.

In another embodiment, the present invention includes compounds of formula (I) where G 1 and G 3 are N, and G 2 is CR 29 ; Y 1 is -CR 32 R 33 -CR 34 R 35 -; Y 2 is -CR 0 R 31 -; R 29 is H, methyl, CI, F, or CF 3 ; R 32 , R 33 , R 34 , and R 35 are H; and X is N.

In yet another embodiment of the present invention, compounds of formula (I) are included wherein A is:

where X is CH or N; G 2 is CR 29 , or N; G 4 is O, S, or NR 60 ; and

R 29 , R 30 , R 31 , R 32 , R 33 , R 34 , R 35 , and R 60 are each independently H, methyl, ethyl, propyl, CI, F, or CF 3 ; or a pharmaceutically acceptable salt thereof.

In another embodiment, A is

each of which is unsubstituted or substituted with one or more substituents each independently selected from halogen, OH, (Ci-C 6 )alkyl (e.g., methyl, ethyl, propyl, or butyl), halo-substituted (Ci-Csjalkyl (e.g., CF 3 ), CN, or NR 10 R u .

In an embodiment, R 1 is 3-to 8-membered cycloheteroalkyl (e.g., piperidinyl, piperazinyl, morpholinyl, or tetrahydrofuranyl), (C3-Cg)cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or (C 6 -Ci 4 )aryl (e.g., phenyl or naphthyl), each of which is unsubstituted or substituted with one or more substituents each independently selected from (Ci-C6)alkyl (e.g., methyl, ethyl, propyl, butyl, or pentyl), halogen, NR 58 R 59 (e.g., NH 2 , NHCH 3 , or NCH 3 CH 3 ), deuterium, or OR 57 , where R 57 is (d- C 6 )alkyl or H.

In yet another embodiment, the present invention includes compounds of formula

(I) wherein R 1 is piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or phenyl which is unsubstituted or substituted with methyl, ethyl, propyl, CI, F, Br, I, or methoxy. In another embodiment, R 1 is piperidinyl, piperazinyl, morpholinyl, cyclopentyl, or cyclohexyl, each of which is unsubstituted or substituted with methyl, methoxy, CI, or F. In another embodiment, R 1 is piperidinyl, piperazinyl, or cyclohexyl, each of which is unsubstituted or substituted with methyl, ethyl, F, CI, or Br. In another embodiment, R 1 is piperidinyl which is unsubstituted or substituted with methyl or F.

In another embodiment, L 1 is (Ci-C )alkylene (e.g., methylene, ethylene or propylene), (C r C 4 )aIkenylene (e.g., =C-, -C=C-, -C=C-C-, or -C-C=C-; -C(O)-, -C(0)0- , -C(0)N-, -((Ci-C 3 )aIkyl)-C(0)- (e.g., -CH 2 C(0)-, -CH 2 CH 2 C(0)- or CH 2 -CH 2 -CH 2 - C(O)-), ((Ci-C 3 alkyl)-C(0)0- (e.g., -CH 2 C(0)0-, -CH 2 CH 2 C(0)0- or CH 2 -CH 2 -CH 2 - C(O)O-), or a bond, wherein L 1 is unsubstituted or substituted by one or more substituents each independently selected from (Ci-C 4 )alkyl (e.g., methyl, ethyl, propyl, or butyl), halo-substituted (Ci-C 4 )alkyl (e.g., CF 3 or CH 2 CF 3 ) or (C 3 -C 8 )cycloaIkyI (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl).

In another embodiment, L 1 is methylene, ethylene, propylene, =C-, -C=C-, -C=C-C-, -C(0 , -C(0)0-, C(0)N-, -CH 2 C(0)-, -CH 2 C(0)0-, or a bond, wherein L 1 is unsubstituted or substituted by one or more substituents each independently selected from methyl, ethyl, or CF 3 . In another embodiment, L 1 is methylene ethylene, or a bond. In another embodiment, L 1 is methylene.

In another embodiment, L 2 is (Ci-C 3 )alkylene (e.g., methylene, ethylene, or propylene), NR 9 , -0-, or -S-, where R 9 is H, (Ci-C6)alkyl (e.g., methyl, ethyl, propyl, or butyl), -(Ci-C 3 )alkylene-(C 6 -Ci )aryl (e.g., -CH 2 -phenyI, -CH 2 CH 2 -phenyI, or

CH 2 CH 2 CH 2 -phenyl), -(Ci-C 3 )alkyIene-(5- to 14-membered heteroaryl) (e.g., -CH 2 - pyridinyl, -CH 2 CH 2 -pyridinyI, -CH 2 -pyrimidinyl, or -CH 2 CH 2 -pyrimidinyl), -(Ci- C 3 )alkylene-CONR 46 R 47 (e.g., -CH 2 C(0)NCH 3 CH 3 , -CH 2 C(0)NHCH 3 , - CH 2 CH 2 C(0)NCH 3 CH 3 , or -CH 2 CH 2 C(0)NHCH 3 ), -(C,-C 3 )alkylene-0-R 48 (e.g., -CH 2 - 0-CH 3 , -CH 2 -0-CH 2 CH 3 , -CH 2 CH 2 -0-CH 3 , or -CH 2 CH 2 -0-CH 2 CH 3 ), -(Ci-C 3 )alkylene- NR 49 R 50 , -(Ci-C 3 )alkylene-(3- to 14-membered cycloheteroalkyl) (e.g., -CH 2 - piperidinyl, -CH 2 CH 2 - piperidinyl, or -CH 2 CH 2 CH 2 -piperidinyl), -(Ci-C 3 )alkylene-S- (C 6 -Ci 4 )aryl (e.g., -CH 2 -S-phenyl, -CH 2 CH 2 -S-phenyl, or -CH 2 CH 2 CH 2 -S-phenyl), (C C 3 )alkylene-C(0)R sl (e.g., -CH 2 C(0)CH 3 , -CH 2 C(0)CH 2 CH 3 , -CH 2 CH 2 C(0)CH 3 , or - CH 2 CH 2 C(0)CH 2 CH 3 ), -(C r C 3 )alkylene-C(0)0-R 52 (e.g., -CH 2 C(0)OCH 3 , - CH 2 C(0)OCH 2 CH 3 , -CH 2 CH 2 C(0)OCH 3 , or -CH 2 CH 2 C(0)OCH 2 CH 3 ), -(C

C 3 )alkylene-0-C(0)-R 53 (e.g., -CH 2 -0-C(0)CH 3 , -CH 2 -0-C(0)CH 2 CH 3 , -CH 2 CH 2 -0- C(0)CH 3 , or -CH 2 CH 2 -0-C(0)CH 2 CH 3 ), -(Ci-C 3 )alkylene-S-R 54 (e.g., -CH 2 -S-CH 3 , - CH 2 -S-CH 2 CH 3 , -CH 2 CH 2 -S-CH 3 , or -CH 2 CH 2 -S-CH 2 CH 3 ), -(C r C 3 )alkylene-SOR 55 (e.g., -CH 2 S(0)CH 3 , -CH 2 S(0)CH 2 CH 3 , -CH 2 CH 2 S(0)CH 3 , or -CH 2 CH 2 S(0)CH 2 CH 3 ), - (d-C 3 )alkylene-S0 2 R 56 (e.g., -CH 2 S(0)OCH 3 , -CH 2 S(0)OCH 2 CH 3 ,

-CH 2 CH 2 S(0)OCH 3 , or -CH 2 CH 2 S(0)OCH 2 CH 3 ), -C(0)-(Ci-C 6 )alkyl (e.g., -C(0)CH 3 or -C(0)CH 2 CH 3 ), -C(0)-(CrC 3 )alkylene-(C 6 -C H )aryl (e.g., -C(0)CH 2 -phenyl or - C(0)CH 2 CH 2 -phenyl), -C(0)-(C r C 3 )alkylene-(5-to 14-membered heteroaryl) (e.g., -C(0)CH 2 -pyridinyl, -C(0)CH 2 CH 2 -pyridinyl, -C(0)CH 2 -pyrimidinyl, or -C(0)CH 2 CH 2 - pyrimidinyl), or -C(0)-(Ci-C 3 )alkylene-(3- to 14-membered cycloheteroalkyl) (e.g., -C(0)CH 2 - piperidinyl, -C(0)CH 2 CH 2 - piperidinyl, or -C(0)CH 2 CH 2 CH2-piperidinyl).

In another embodiment, L 2 is -CH2- or NR 9 where R 9 is H, methyl, or ethyl. In another embodiment, , L 2 is -CH 2 - or -NH-. In another embodiment, L 2 in -NH-.

In yet another embodiment, the present invention includes compounds of formula (I) where L 1 is ~CH 2 -, -CH 2 CH 2 -, or a bond; and L 2 is ~CH 2 - or NH.

In an embodiment, R 2 is a (C6-Ci4)aryl (e.g., phenyl or naphthyl), 5- to 14- membered heteroaryl (e.g., pyridinyl, pyrimidinyl, or pyridazinyl), 3- to 8-membered cycloheteroalkyl (e.g., piperidinyl, piperazinyl, morpholinyl, or tetrahydrofuranyl) or (C 3- Ci 4 )cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or a partially saturated cyclohexyl (cyclohexenyl or cyclohexadienyl)) each of which is unsubstituted or substituted with one or more of halogen, OH, -(CrC 6 )alkyl, halo-substituted -(C)- C 3 )alkyl (preferably, CF 3 ); or CN. In another embodiment, the present invention includes compounds of formula (1) where R 2 is phenyl, naphthyl, pyridinyl, pyrimidinyl, or pyridazinyl; each of which is unsubstituted or substituted with one or more substituents each independently selected from methyl, dimethyl, ethyl, propyl, butyl, pentyl, hexyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, CF 3 , or CH 2 C1. In another embodiment, R 2 is phenyl, cyclohexyl, or a partially saturated cyclohexyl, where R 2 is unsubstituted or substituted with one or more substituents selected from methyl, dimethyl, OH, F, CI, or Br. In another embodiment, R 2 is phenyl.

In another embodiment, R 3 and R 4 are each independently H, (Ci-C 6 )alkyl, -(Ci- C 6 )alkylene-(C 6 -C 14 )aryl (e.g., -CH 2 -phenyl, -CH 2 CH 2 -phenyl, -CH 2 CH 2 CH 2 -phenyl,

-CH 2 CH 2 CH 2 CH 2 -phenyl, or -CH 2 CH 2 CH 2 CH 2 CH 2 -phenyl), -(Ci-C 6 )alkylene-(5- to 14- membered heteroaryl) (e.g., -CH 2 CH 2 CH 2 CH 2 -pyridinyl or -CH 2 CH 2 CH 2 CH 2 - pyrimidinyl), -(C C 6 )alkylene-C(0)NR 16 R 17 (e.g., -CH 2 CH 2 CH 2 CH 2 C(0)NCH 3 CH 3 , -CH 2 CH 2 CH 2 CH 2 C(0)NHCH 3 , or -CH 2 CH 2 CH 2 CH 2 C(0)NH 2 ), -(C,-C 6 )alkylene-0-R 15 (e.g., -butyl-0-CH 3 or -butyl-0-CH 2 CH 3 ), -(Ci-C 6 )alkylene-NR 13 R 14 (e.g., -butyl-

N(CH 3 )(CH 3 ), -butyl-NHCH 3 , -butyl-NH 2 , -butyl-NH-phenyl, or -butyl-N(CH 3 )(CH 3 )), -(Ci-C 6 )alkylene-(3-tol 4-membered cycloheteroalkyl) (e.g., -butyl-piperidinyl), -(C r C 3 )alkylene-S-(C 6 -Ci 4 )aryl group, -(Ci-C 6 )alkylene-COR 18 , -(Ci-C 6 )alkylene-C(0)0-R 19 , -(Ci-C 6 )alkylene-0-C(0)-R 2 °, (Ci-C 3 )alkylene-S-R 21 , -(Ci-C 3 )alkylene-SOR 22 , or -(d- C 3 )alkylene-S0 2 R 23 .

In a preferred embodiment, R 3 and R 4 are each independently H, (Ci-C 6 )alkyl (preferably, methyl, ethyl, propyl, or butyl), -(Ci-C 3 )alkylene-(C 6 -Ci 4 )aryl (preferably, -CH 2 -phenyl, -CH 2 CH 2 -phenyl, or -CH 2 CH 2 CH 2 -phenyl), -(Ci-C 3 )alkylene-(5- to 14- membered heteroaryl) (preferably, -CH 2 -pyridinyl, -CH 2 CH 2 -pyridinyl, -CH 2 - pyrimidinyl, or -CH 2 CH 2 -pyrimidinyl), -(C,-C 3 )alkylene-CONR 16 R 17 (preferably,

-CH 2 C(0)NCH 3 CH 3 , -CH 2 C(0)NHCH 3 , -CH 2 CH 2 C(0)NCH 3 CH 3 , or

-CH 2 CH 2 C(0)NHCH 3 ), -(C,-C 3 )-0-R 15 (preferably, -CH 2 -0-CH 3 , -CH 2 -0-CH 2 CH 3 , -CH 2 CH 2 -0-CH 3 , or -CH 2 CH 2 -0-CH 2 CH 3 ), -(C r C 3 )alkylene-NR 1 R 14 (preferably, -CH 2 -N(CH 3 )(CH 3 ), -CH 2 -NHCH 3 , -CH 2 -NH 2 , -CH 2 -NH-phenyl, -CH 2 -CH 2 - N(CH 3 )(CH 3 ), -CH 2 CH 2 -NHCH 3 , -CH 2 CH 2 -NH 2 , or -CH 2 CH 2 -NH-phenyl), -(d- C 3 )alkylene-(3- to 14-membered cycloheteroalkyl (preferably, -CH 2 - piperidinyl, -CH 2 CH 2 - piperidinyl, or -CH 2 CH 2 CH 2 -piperidinyl), -(Ci-C 3 )alkylene-S-(C 6 -Ci 4 )aryl (preferably, -CH 2 -S-phenyl, -CH 2 CH 2 -S-phenyl, or -CH 2 CH2CH2-S-phenyl), -(Ci- C 3 )alkylene-COR 18 (preferably, -CH 2 C(0)CH 3 , -CH 2 C(0)CH 2 CH 3 , -CH 2 CH 2 C(0)CH 3 , or -CH 2 CH 2 C(0)CH 2 CH 3 ), -(Ci-C 3 )alkylene-C(0)0-R 19 (preferably, -CH 2 C(0)OCH 3 , -CH 2 C(0)OCH 2 CH 3 , -CH 2 CH 2 C(0)OCH 3 , or -CH 2 CH 2 C(0)OCH 2 CH 3 ), -(Ci-C 3 )alkyl- 0-C(0)-R 20 (preferably, -CH 2 -0-C(0)CH 3 , -CH 2 -0-C(0)CH 2 CH 3 , -CH 2 CH 2 -0- C(0)CH 3 , or -CH 2 CH 2 -0-C(0)CH 2 CH 3 ), -(Ci-C 3 )alkylene-S-R 21 (preferably, -CH 2 -S- CH 3 , -CH 2 -S-CH 2 CH 3 , -CH 2 CH 2 -S-CH 3 , or -CH 2 CH 2 -S-CH 2 CH 3 ), -(Ci-C 3 )alkylene- S(0)R 22 (preferably, -CH 2 S(0)CH 3 , -CH 2 S(0)CH 2 CH 3 , -CH 2 CH 2 S(0)CH 3 , or

-CH 2 CH 2 S(0)CH 2 CH 3 ), or -(C,-C 3 )alkylene-S0 2 R 23 (preferably, -CH 2 S(0)OCH 3 , -CH 2 S(0)OCH 2 CH 3 , -CH 2 CH 2 S(0)OCH 3 , or -CH 2 CH 2 S(0)OCH 2 CH 3 ).

In another embodiment, R 3 and R 4 are independently (Ci-Ce)alkyl, (CpC 3 )alkyl- (C6-Ci 4 )aryl, -(Ci-C 3 )alkylene-(5-tol4-membered) heteroaryl, -(Ci-C 3 )alkylene- C(0)NR l6 R 17 , -(C 1 -C 3 )alkylene-0-R 15 , -(Ci-C 3 )alkylene-NR 13 R 14 , -(Ci-C 3 )alkyl-(3- to 14-membered cycloheteroalkyl, or -(Ci-C 3 )alkyl-S-(C6-Ci 4 )aryl.

In an embodiment, R 3 and R 4 are each independently -CH 2 -phenyl, -CH 2 CH 2 - phenyl, -CH 2 CH 2 CH 2 -phenyl, -CH 2 -pyridinyl, -CH 2 CH 2 -pyridinyl, -CH 2 -pyrimidinyl, -CH 2 CH 2 -pyrimidinyl, -CH 2 C(0)NCH 3 CH 3 , -CH 2 C(0)NHCH 3 ,

-CH 2 CH 2 C(0)NCH 3 CH 3 , -CH 2 -0-CH 3 , -CH 2 -0-CH 2 CH 3 , -CH 2 -S-phenyl, -CH 2 CH 2 -S- phenyl, -CH 2 CH 2 CH 2 -S-phenyl;-CH 2 C(0)CH 3 , -CH 2 C(0)CH 2 CH 3 , -CH 2 CH 2 C(0)CH 3 , -CH 2 -S-CH 3 , -CH 2 -S-CH 2 CH 3 , -CH 2 -N(CH 3 )(CH 3 ), -CH 2 -NHCH 3 , -CH 2 -NH 2 , CH 2 -NH- phenyl, -CH 2 -CH2-N(CH 3 )(CH 3 ), -CH 2 CH 2 -NHCH 3 , -CH 2 CH 2 -NH 2 , or -CH 2 CH 2 -NH- phenyl.

In yet another embodiment R 3 is -CH 2 -phenyl, -CH 2 CH 2 -phenyl, -CH 2 CH 2 CH 2 - phenyl, -CH 2 -pyridinyl, -CH 2 CH 2 -pyridinyl, -CH 2 -pyrimidinyl, -CH 2 CH 2 -pyrimidinyl, -CH 2 C(0)NCH 3 CH 3 , -CH 2 C(0)NHCH 3 , -CH 2 CH 2 C(0)NCH 3 CH 3 , -CH 2 -0-CH 3 , -CH 2 - 0-CH 2 CH 3 , -CH 2 -S-phenyl, -CH 2 CH 2 -S-phenyl, -CH 2 CH 2 CH 2 -S-phenyl;

and R 4 is -CH 2 C(0)CH 3 , -CH 2 C(0)CH 2 CH 3 , -CH 2 CH 2 C(0)CH 3 , -CH 2 -S-CH 3 , -CH 2 -S- CH 2 CH 3j -CH2-N(CH 3 )(CH 3 ), -CH 2 -NHCH 3 , -CH 2 -NH 2 , CH 2 -NH-phenyl, -CH 2 -CH 2 - N(CH 3 )(CH 3 ), -CH 2 CH 2 -NHCH 3 , -CH 2 CH 2 -NH 2 , or -CH 2 CH 2 -NH-phenyl. In another embodiment, R 3 is -CH 2 -phenyl, -CH 2 CH 2 -phenyl, -CH 2 CH CH - phenyl, -CH 2 -S-phenyl, -CH 2 CH 2 -S-phenyl, or -CH 2 CH 2 CH 2 -S-phenyl. In another embodiment, R 4 is -CH 2 -N(CH 3 )(CH 3 ), -CH 2 -NHCH 3 , -CH 2 -NH 2 , -CH 2 -NH-phenyl, -CH 2 -CH 2 -N(CH 3 )(CH 3 ), -CH 2 CH 2 -NHCH 3 , -CH 2 CH 2 -NH 2 , or -CH 2 CH 2 -NH-phenyl. In another embodiment R 3 is -CH 2 -S-phenyl, or -CH 2 CH 2 -S-phenyl. In another

embodiment R 4 is -CH 2 -N(CH 3 )(CH 3 ), -CH 2 -NHCH 3 , -CH 2 -NH 2 , -CH 2 -NH-phenyl, or -CH 2 -CH 2 -N(CH 3 )(CH 3 ).

In an embodiment, R 8 is (C]-C 6 )alkyl (preferably, methyl, ethyl, propyl, butyl, pentyl, or hexyl), (C 6 -C )4 ) aryl (preferably, naphthyl or phenyl), (C 3 -Ci 4 )cycloalkyl (preferably, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), halogen, or 3- to 14- membered cycloheteroalkyl (preferably, piperidinyl, piperazinyl, morpholinyl, or tetrahydrofuranyl) in which any the aforementioned hydrocarbon groups (e.g., (Ci- C 6 )alkyl, (C 6 -C) 4 ) aryl, and (C 3 -Ci 4 )cycloalkyl) is optionally substituted with halogen, (Ci-C 6 )alkyl (preferably, methyl, ethyl, propyl, or butyl), halo -substituted (Ci-C6)alkyl (preferably, CF 3 ), OH, or NR 44 R 45 .

In another embodiment, R 8 is methyl, ethyl, propyl, butyl, naphthyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, F, Br, piperidinyl, or piperazinyl, in which any the aforementioned hydrocarbon groups (e.g., methyl, ethyl, propyl, butyl, naphthyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl) is substituted with F, CI, Br, methyl, ethyl, propyl, or CF 3 . In another embodiment, R 8 is phenyl which is unsubstituted or substituted with F, CI, Br, methyl, ethyl, propyl, or CF 3 .

In an embodiment, R 6 and R 7 are each independently H, N0 2 , S0 2 CF 3 , S0 2 -(Ci- C 6 )alkyl (preferably, S0 2 CH 3 , or S0 2 CH 2 CH 3 ), halo-substituted (C r C 6 )alkyl

(preferably, CF 3 ), halogen, (C 3 -C ] )cycloalkyl (preferably, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or CN. In another embodiment, one of R 6 and R 7 is H, and the other is selected from N0 2 , S0 2 CF 3 , S0 2 CH 3 , S0 2 CH 2 CH 3 , CF 3 , CI, or F. In another embodiment, one of R 6 and R 7 is H, and the other is selected from N0 2 , or S0 2 CF 3 . In another embodiment, both of R 6 and R 7 are H.

In an embodiment, R 10 , R 1 1 , R 13 , R 14 , R 16 , R 17 R 46 , R 47 , R 49 , and R 50 are each independently H, (C]-C6)alkyl (preferably, methyl, ethyl, propyl or butyl), (C 2 -C 6 )alkenyl (preferably, ethenyl or propenyl), (C -C 6 )alkynyl, OH, -C(0)-(Ci-C 6 )alkyl (preferably, -C(0)methyl, -C(0)ethyl, or -C(O)propyl), (Cj-Cejalkoxy (preferably, methoxy, ethoxy, or propoxy), halogen, (C 3 -Cj 4 )cycloalkyl (preferably, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), (C CH) aryl (preferably, phenyl), 4- to 14-membered cycloheteroalkyl (preferably, piperidinyl, piperazinyl, morpholinyl, or tetrahydrofuranyl), or 5-to 14-membered heteroaryl (preferably, pyridinyl, pyrimidinyl, or pyridazinyl) wherein each of the aforementioned hydrocarbon groups (e.g., Ci-C 6 )alkyl, (C 2 - C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 3 -C 14 )cycloalkyl, and (C 6 -C 14 ) aryl, and the (d-Q alkyl moiety of -CiOMQ-CsJalkyl and (C ! -C 6 )alkoxy) is optionally substituted by one or more substituents selected from halogen, hydroxyl, (Ci-C 6 )alkoxy, amino, (C CeJalkylamino, di((Ci-C6)alkyl)amino or cyano;

In a further embodiment, R 13 and R 14 together with the N to which they are attached form a 4- to 8-membered cycloheteroalkyl group (preferably, piperidinyl or piperazinyl), or a 5- to 14-membered heteroaryl (preferably, pyridinyl or pyrimidinyl) each of which is substituted or unsubstituted.

or R 16 and R 17 together with the N to which they are attached form a 4- to 8- membered cycloheteroalkyl (preferably, piperidinyl or piperazinyl), or a 5- to 14- membered heteroaryl (preferably, pyridinyl or pyrimidinyl) each of which is substituted or unsubstituted.

In an embodiment, R 15 , R 18 , R 19 , R 20 , R 22 , R 23 , R 44 , R 45 , R 48 , R 51 , R 52 , R 53 , R 55 , and R 56 are each independently -(Q-CeJalkylene-fCs-CnJaryl (preferably, -CH 2 -phenyl, - CH 2 CH 2 -phenyl, or- CH 2 CH 2 CH 2 -phenyl), or -(C 1 -C 6 )alkylene-(5-to 14-membered heteroaryl), (preferably, -CH 2 -pyridinyl, -CH 2 CH 2 -pyridinyl, -CH 2 -pyrimidinyl, or - CH 2 CH 2 -pyrimidinyl) each of which is unsubstituted or substituted with one or more substituents each independently selected from halogen, OH, (Ci-C 6 )alkyl (preferably, methyl, ethyl, propyl, or butyl), halo-substituted (Ci- )alkyl (preferably,CF 3 ), or CN.

R 21 and R 54 are each independently (C 6 -Ci 4 )aryl (preferably, phenyl or naphthyl), 5- to 14-membered heteroaryl, -(Ci-C 6 )alkyl-(C 6 -Ci 4 )aryl (preferably, -CH 2 -phenyl, - CH 2 CH 2 -phenyl, or -CH 2 CH 2 CH 2 -phenyl), or (d-C 6 )alkylene-(5 -to 14-membered heteroaryl) (preferably, -CH 2 -pyridinyl, -CH 2 CH 2 -pyridinyl, -CH 2 -pyrimidinyl, or - CH 2 CH 2 -pyrimidinyl) each of which is unsubstituted or substituted with one or more substituents each independently selected from halogen, OH, (Ci-C 6 )alkyl (preferably, methyl, ethyl, or propyl), halo-substituted (C r C 6 )alkyl (preferably,CF 3 ), or CN.

In another embodiment, the present invention includes compounds of formula

(lb):

(lb)

or a pharmaceutically acceptable salt thereof, wherein

R lb is H, N0 2 , S0 2 CF 3 , S0 2 (Ci-C 6 )alkyl, halo-substituted (d-C 6 )alkyl, halogen, a (C 3- Ci 4 )cycloalkyl group, or CN;

R is a divalent bicyclic radical comprising a saturated cyclic structure and an unsaturated cyclic structure, wherein said unsaturated cyclic structure is attached to NH, and said saturated cyclic structure is attached to R 5b , wherein R 2b is unsubstituted or substituted with one or more substituents each independently selected from halogen, OH, (C]-C 6 )alkyl, halo-substituted(C C 6 )alkyl, CN or NH 2 ;

R 3b and R 4b are each independently H, (C C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -

C 6 )alkynyl, a (C 6 -C) 4 )aryl group, a 5- to 14-membered heteroaryl group, a (C 3 .

Ci4)cycloalkyl group, halogen, or a 3- to 14-membered cycloheteroalkyl group, each of which when not H may be unsubstituted or substituted with one or more of hydroxyl, cyano, nitro, (C,-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, (d -Q alkoxy, (C 2 - C 8 )alkenyloxy, (C 2 -C 8 )alkynyloxy, halogen, (Ci-C 8 )alkylcarbonyl, carboxy, (Ci- C 8 )alkoxycarbonyl, amino, (C C 8 )alkylamino, di((Ci-C 8 )alkyl)amino, (C|- C 8 )alkylaminocarbonyl, di((Ci-C 8 )alkyl)aminocarbonyl, (C|-C 8 )alkylcarbonylamino, (d- C 8 )alkylcarbonyl((C] -Cs)alkyl)amino, (Ci-C 8 )alkylsulfonylamino, (C| -C 8 )alkylthio, (Cj- C 8 )alkylsulfinyl, (C]-C 8 )alkylsulfonyl, aminosulfonyl, (Ci-C 8 )alkylaminosulfonyl or di(Ci-C 8 )alkylaminosulfonyl, where each of the afore-mentioned hydrocarbon groups (e.g., (C,-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, and the (C C 8 )alkyl, (C 2 -C 8 )alkenyl and (C 2 -C 8 )alkynyl moieties of (C 2 -C 8 )alkoxy, (C 2 -Cs)alkenyloxy, (C 2 -C8)alkynyloxy, (Cj-C 8 )alkylcarbonyl, (Ci-Cg)alkoxycarbonyl, (CrC 8 )alkylamino, di(Ci-C 8 )alkylamino, (C i -C 8 )al kylaminocarbony 1 , di((C i -C 8 )alk l)aminocarbon 1, (C i -C 8 )alk lcarbon lamino, (Ci-C 8 )alkylcarbonyl((C]-Cg)alkyl)amino, (Ci-C 8 )alkylsulfonylamino, (Ci-Cg)alkylthio, (C C 8 )alkylsulfinyl, (Q-Cgjalkylsulfonyl, (C,-C 8 )alkylaminosulfonyl and di((C r C 8 )alkyl)aminosulfonyl) is optionally substituted by one or more substituents each independently selected from halogen, OH or (Cj-Cg)alkoxy;

R 5b is a 3- to 8-membered cycloheteroalkyl, (C3-C 8 )cycloalkyl, or

(C 6 -Ci4)aryl which is unsubstituted or substituted with (C t -C6)alkyl, halogen, OH, (Ci- C 3 )alkoxy, NH 2 , or deuterium;

R 6b is L b -R 8b ;

R 7b is hydroxyl, cyano, nitro, (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, (C 2 - C 8 )alkoxy, (C 2 -C 8 )alkenyloxy, (C 2 -C 8 )alkynyloxy, halogen, (Ci-Cg)alkylcarbonyl, carboxy, (Ci-Cg)alkoxycarbonyl, amino, (CrC 8 )alky lamino, diiCpCgJalkylamino, (C Cg)alkylaminocarbonyl, diiiC CgJalky aminocarbonyl, (C|-Cg)alkylcarbonylamino, (C|- C 8 )alkylcarbonyl((Ci-C 8 )alkyl)amino, (Cj-C 8 )alkylsulfonylamino, (C Cg)alkylthio, (C r Cg)alkylsulfinyl, (C 5 -C 8 )alkylsulfonyl, aminosulfonyl, (CrQJalkylaminosulfonyl or di((C C8)alkyl)aminosulfonyl, where each of the afore-mentioned hydrocarbon groups (e.g., (d-CgJalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, and the (Ci-C 8 )alkyl, (C 2 -C 8 )alkenyl and (C 2 -C 8 )alkynyl moieties of (C 2 -Cg)alkoxy, (C 2 -C 8 )alkenyloxy, (C 2 -C 8 )alkynyloxy, (Ci-C 8 )alkylcarbonyl, (Ci-Cg)alkoxycarbonyl, (Ci-C 8 )alkylamino, di(Ci-C 8 )alky lamino, (C]-C 8 )alkylaminocarbonyl, di((C] -C8)alkyl)aminocarbonyl, (CpCsialkylcarbony lamino, (C i -C 8 )alkylcarbonyl((C t -C 8 )alkyl)amino, (C i -C 8 )alkylsulfonylamino, (Ci -Cg)alkylthio, (C r C 8 )alkylsulfinyl, (C C 8 )alkylsulfonyl, (C,-C 8 )alkylaminosulfonyl and di((C

C 8 )alkyl)aminosulfonyl) is optionally substituted by one or more substituents each independently selected from halogen, OH or (Ci-C 8 )alkoxy;

L b is -(Ci-C 3 )alkylene- f -(C 2 -C 4 )alkenylene-, -C(O)-, -C(0)0-, -C(0)N(H)-, -(Q- C 3 )alkylC(0)-, -(Ci-C 3 )alkyl-C(0)0-, or a bond, wherein L b is unsubstituted or substituted by one or more (C|-C 4 )alkyl, halo-substituted(Ci-C 4 )alkyl, or (C 3 - Cg)cycloalkyl; is (C 6 -Ci 4 )aryl, a 5- to 14-membered heteroaryl, a 3- to 8-membered cycloheteroalkyl, or a (C3.C 14 )cycIoalkyl, each of which is unsubstituted or substituted with one or more substituents each independently selected form halogen, OH, (d - C 6 )alkyl, halo-substituted(C] -C 6 )alkyl, or CN; and

n is 0, 1 , 2, 3, or 4.

In an embodiment, the present invention further includes compounds of formula (lb) where R is defined as A in formula I. In another embodiment, the present invention includes compounds of formula (lb) where n is 0, 1 , or 2. In another embodiment, n is 0.

In another embodiment, the present invention includes compounds of formula (lb) where R 3b and R 4b are each independently H, (Cj -C6)alkyl (preferably, methyl, ethyl or propyl), (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 6 -Ci 4 )aryl (preferably, phenyl), a 5- to 14- membered heteroaryl group (preferably, pyridinyl or pyrimidinyl), (C 3 .Ci 4 )cycloalkyl (preferably, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or halogen (preferably, F, CI, or Br).

Compounds of particular interest include:

(R)-N-(7-(l -((2-(4-chlorophenyl)-5,5-dimethylcyclohex- l -enyl)methyl)piperidin- 4-yl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(d imethylamino)- l - (phenylthio)butan-2-ylamino)-3-nitrobenzenesulfonamide;

N-(7-((2S)-l -((4'-chlorobiphenyl-2-yl)methyl)-2-methylpiperidin-4-yl)-5, 6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylami no)- 1 -(phenylthio)butan-2- ylamino)-3-(trifluoromethyl-sulfonyl)benzenesulfonamide;

(R)-4-(4-(dimethylamino)-l -(phenylthio)butan-2-ylamino)-N-(7-(l -((4'- fluorobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7,8-tetrahydr opyrido[3,4-d]pyrimidin-4- yl)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l -((4'-bromobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l -(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l -((2-(4-chlorophenyl)-5,5-dimethylcyclohex- l -enyl)methyl)piperidin- 4-yl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(d imethylamino)-l - (phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benz enesulfonamide; (R)-N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)- 5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l-(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-( 1 -((2-(4-chlorophenyl)-4,4-dimethylcyclohex-l -enyl)methyl)piperidin- 4-yl)-5,6,7,8-tetrahydΓopyrido[3,4-d]pyrimidin-4-yl)-4-(4-m oφholino-l-

(phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfonyl)b enzenesulfonamide;

(R)-N-(7-( 1 -((2-(4-chlorophenyl)-4,4-dimethylcyclohex-l -enyl)methyl)piperidin- 4-y 1) -5 ,6, 7 , 8-tetrahydropyrido[3 ,4-d]py rimidin-4-y 1) -4-(4-(di methy lamino)- 1 - (phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benz enesulfonamide;

(R)-N-(7-(l-((4'-chloro-5-fluorobiphenyl-2-yl)methyl)piperid in-4-yl)-5,6,7,8- tetrahydropyrido [3 ,4-d]pyri midin-4-y l)-4-(4-(dimethy lam ino)- 1 -{phen ylthio)butan-2- y 1 am ino)-3 -(tri fluoromethy lsul fonyl)benzenesul fonamide;

(R)-N-(7-(l-((4'-chloro-4-fluorobiphenyl-2-yl)methyl)piperid in-4-yl)-5,6,7,8- tetrahydropyrido [3 ,4-d]pyr i mi din-4-y l)-4-(4-(di methy lam ino)- 1 -(phen ylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l-((4'-chloro-3-fluorobiphenyl-2-yl)methyl)piperid in-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l-(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

N-{7-[l-(4'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6, 7,8-tetrahydro- pyrido[3,4-d]pyrimidin-4-yl}-4[(R)-3-(4-ethyl-piperazin-l-yl )-l-phenylsulfanylmethyl- propylamino]-3-trifluoromethanesulfonyl-benzenesulfonamide;

N-{7-[l-(4'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6, 7 s 8-tetrahydro- pyrido [3 ,4-d]pyrim idin-4-yl } -4- [(R)- 1 -(3-chloro-phenylsul fanyl methy l)-3 - dimethylamino-propylamino]-3-trifluoromethanesulfonyl-benzen esulfonamide;

(R)-N-(7-(l -((2-(4-chlorophenyl)-4,4-dimethylcyclohex- 1 -enyl)methyl)piperidin-

4-yl) - 5,6,7, 8-tetrahydropyrido[ 3 ,4-d]pyri mi din-4-yl)-4-( 1 -(2-chlorophenylthio)-4- (dimethylamino)butan-2-ylamino)-3-(trifluoromethylsulfonyl)b enzenesulfonamide;

(R)-N-(7-(l-((4'-chloro-4-methoxybiphenyl-2-yl)methyl)piperi din-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l-(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide; (R)-N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)- 5,6,7,8- tetrahydropyrido [3,4-d]pyrimidin-4-yl)-4-(l-(2,6-dichlorophenylthio)-4- (dimethylamino) butan-2-ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonami de;

N-{7-[l -(4'-ChIoro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6,7,8-tet rahydro- pyrido[3,4-d]pyrimidin-4-yl}-4-[(R)- 1 -(3,4-dichloro-phenylsulfanylmethyl)-3- dimethylamino-propylamino]-3-trifluoromethanesulfonyl-benzen e-sulfonamide;

(R)-N-(7-( l -((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7,8- tetrahydropyridot3,4-d]pyrimidin-4-yl)-4-(l-(2-chlorophenyft hio)-4-(dimethylamino) butan-2-ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonami de;

N-{7-t l -(4'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6,7,8-tet rahydro- pyrido[3,4-d]pyrimidin-4-yl}-4-[(R)-3-dimethylamino- l-(2-fluoro- phenylsulfanylmethyl)-propylamino]-3-trifluoromethanesulfony l-benzenesulfonamide;

N-{(R)-7-[l -(4'-Chloro-biphenyN2-ylmethyl)-piperidin-4-yl]-6-methyl-5,6 ,7,8- tetrahydro-pyrido[3,4-d]pyrimidin-4-yl}-4-((R)-3-dimethylami no-l - phenylsulfanylmethyl-propylamino)-3-trifluoromethanesulfonyl -benzene sulfonamide;

(R)-N-(2-chloro-7-( l-((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l-(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

N-(7-( 1 -( 1 -(4'-chlorobiphenyl-2-yl)ethyl)piperidin-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylami no)- 1 -(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-( 1 -((2-(4-chlorophenyl)-4,4-dimethylcyclohex- 1 -enyl)methyl)piperidin- 4-yl)-5,6 f 7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(l-(2-flu orophenylthio)-4-(4- methy 1 piperazi n- 1 -y I )butan-2-y I amino)-3 - (trifluorom ethy 1 su 1 fony l)benzene sul fonam ide ;

(R)-N-(7-(l -((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7,8- tetrahydropyridot3,4-d]pyrimidin-4-yl)-4-(l -(2-fluorophenylthio)-4-(4-methylpiperazin- l-yl)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benzenesulf onamide;

N-(7-(l -((4'-chlorobiphenyl-2-yl)methyl)-3-fluoropiperidin-4-yI)-5, 6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylami no)-l -(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide; (R)-N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)- 2-(trifluoromethyl)- 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethy lamino)-l- (phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benz enesulfonamide;

N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)-3-fluoropiperidin-4 -yl)-2- (trifluoromethyl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4 -yl)-4-((R)-4- (dimeth ylamino)- 1 -(phen ylthio)butan-2-ylam ino)-3-(tri fluoromethyl sul fonyl - benzenesulfonamide;

(R)-N-(7-(l-((2-(4-chlorophenyl)-5,5-dimethylcyclohex-l-enyl )methyl)piperidin- 4-yl)-2-(trifluoromethyl)-5,6,7,8-tetrahydropyrido[3,4-d]pyr imidin-4-yl)-4-(4- (dimethy lamino)- 1 -(phen yl thio)butan-2-y 1 am ino)-3-(tri fluoromethyl sul fony 1)- benzenesulfonamide;

(R)-N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)- 2-(trifluoromethyl)- 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethy lamino)-l- (phenylthio)butan-2-ylamino)-3-nitrobenzenesulfonamide;

(R)-N-(7-( 1 -((4'-chloro-4-fluorobiphenyl-2-yl)methyl)-4-deuteropiperidi n-4-yl)-

5,6 ) 7 ) 8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethy lamino)-l- (phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benz enesulfonamide;

N-(7-( 1 -((S)- 1 -(4'-chlorobiphenyl-2-yl)ethyl)-4-deuteropiperidin-4-yl)-2- (trifluoromethyl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4 -yl)-4-((R)-l-(2- fluorophenylthio)-4-(4-methylpiperazin- 1 -yl)butan-2-ylamino)-3- (trifluoromethylsulfonyl)benzenesulfonamide;

N-(7-{ l-[2-(4-Chloro-phenyl)-4,4-dimethyl-cyclohex~l-enylmethyl]-4 -deutero- piperidin-4-yl}-5,6,7,8-tetrahydro-pyridot3,4-d]pyrimidin-4- yl)-4-[(R)-3-(isopropyl- methyl-amino)-l-phenylsulfanylmethyl-propylamino]-3-trifluor omethanesulfonyl- benzenesulfonamide;

(R)-N-(7-( 1 -((2-(4-chlorophenyl)-4,4-dimethylcyclohex- 1 -enyl)methyl)-4- deuteropiper i din-4-yl)-5 ,6, 7, 8-tetrahy dropyrido [3,4-d]pyrimidin-4-yl)-4-(l-(2- fluoropheny lthio)-4-(4-methy lpiperazin- 1 -yl)butan-2-y lam ino)-3- (trifluoromethylsulfonyl)benzenesulfonamide; (R)-N-(7-(l -((2-(4-chlorophenyl)-4,4-dimethylcyclohex-l -enyl)methyl)-4- deuteropiperidin-4-yl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimi din-4-yl)-4-(l -(phenylthio)- 4-(pyrroHdin-l-yl)butan-2-ylamino)-3-(trifluoromethylsulfony l)benzenesulfonarnide;

N-(7-{ l-[(R)- l -(4'-Chloro-biphenyl-2-yl)-ethyl]-4-deuteropiperidin-4-yl}-5 ,6,7,8- tetrahydro-pyrido[3,4-d]pyrimidin-4-yl)-4-((R)-3-moφholin-4 -yl- l - phenylsulfanylmethyl-propylamino)-3-trifluoromethanesulfonyl -benzenesulfonamide;

(R)-N-(7-(l-((4'-chloro-4-fluorobiphenyl-2-yl)methyl)-4-deut eropiperidin-4-yl)- 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-( l-(2-fluorophenylthio)-4-(4- methylpiperazin- l -yl)butan-2-ylamino)-3-(trifluoromethylsulfonyl)-benzenesulf onamide;

(R)-N-(7-(l -((4'-chloro-4-fluorobiphenyl-2-yl)methyl)-4-deuteropiperidi n-4-yl)- 5,6,7,8-tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(4-ethyl piperazin-I -yl)-l- (phenylthio)butan-2-ylarnino)-3-(trifluoromethylsulfonyl)ben zenesulfonamide;

(R)-N-(7-(l -((2-(4-chlorophenyl)-4,4-dimethylcyclohex-l -enyl)methyl)-4- deuteropiperidin-4-yI)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimi din-4-yl)-4-(4-morpholino- l -(phenyIthio)butan-2-ylamino)-3-(trifluoromethylsuIfonyl)ben zenesulfonamide;

N-{6-[l -(4'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6,7,8-tet rahydro-4H- l ,2,3a,6-tetraaza-azuIen-3-yl}-4-((R)-3-dimethylamino- l-phenyl-sulfanylmethyl- propylamino)-3-trifluoromethanesulfonyl-benzenesulfonamide;

N-{7-[ l -(3'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6,7,8-tet rahydro- pyrido[3,4-d]pyrimidin-4-yl}-4-((R)-3-dimethylamino- l-phenylsulfanyImethyl- propylamino)-3-trifluoromethanesulfonyl-benzenesulfonamide;

N-(7-( l -((2'-chlorobiphenyl-2-yI)methyl)piperiditi-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylami no)- l -(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide; and

(R)-4-(4-(dimethyIamino)-l -(phenylthio)butan-2-ylamino)-N-(7-(l -((4'- (trifluoromethyl)biphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7, 8-tetrahydropyrido[3,4- d]pyrimidin-4-yl)-3-(trifluoromethylsulfonyI)benzenesulfonam ide;

or a pharmaceutically acceptable salt thereof.

More particularly, compounds such as: N-(7-((2S)-l-((4'-chlorobiphenyl-2-yl)methyl)-2-methylpiperi din-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylami no)-l-(phenylthio)butan-2- ylamino)-3-(trifluoromethyl-sulfonyl)benzenesulfonamide;

(R)-4-(4-(dimethylamino)- 1 -(phenylthio)butan-2-ylamino)-N-(7-( 1 -((4'- fluorobiphen yl-2-y l)methy l)piperid in-4-y l)-5 ,6, 7 , 8 -tetrah ydropyrido [3 ,4-d]pyr imidin-4- yl)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l-((4 , -bromobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6 ; 7,8- tetrah ydropyrido [3 ,4-d]pyr imi din-4-y l)-4-(4-(di meth y lamino)- 1 -(phenyl thio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l-((4 , -chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5 f 6,7,8- tetrahydropyridot3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l-(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l-((2-(4-chlorophenyl)-4,4-dimethylcyclohex-l-enyl )methyl)piperidin- 4- y l)-5 ,6, 7, 8-tetrahydropyr ido[3,4-d]p yrimid in-4-y 1 )-4-(4-(dimethy lamino)- 1 - (phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benz enesulfonamide;

(R)-N-(7-(l-((4'-chloro-4-fluorobiphenyl-2-yl)rnethyl)piperi din-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamino)- l-(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide;

N-{7-[ l-(4 , -Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6,7,8-t etrahydro- pyr i do [3 ,4-d]p yrimidin-4-y 1 }-4-[(R)-3-dimethy lamino- 1 -(2-fluoro- phenylsulfanylmethyl)-propylamino]-3-trifluoromethanesulfony l-benzenesulfonamide;

(R)-N-(7-(l-((4 , -chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(l-(2-fluorophenylt hio)-4-(4-methylpiperazin- l-yl)butan-2-ylamino)-3-(trifluoromethylsulfonyl)benzenesulf onamide;

N-(7-(l-(l-(4'-chlorobiphenyl-2-yl)ethyl)piperidin-4-yl)-5,6 ,7,8- tetrahydropyridot3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylami no)-l-(phenylthio)butan-2- ylam ino)-3 -(trifluoromethy lsul fony l)benzenesul fo namide;

(R)-N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)- 2-(trifluoromethyl)- 5,6, 7,8-tetrahy dropyr ido [3 ,4-d] pyrimi din-4-y 1 )-4-(4-( dimeth y lamino)- 1 - (phen ylth io) butan-2-ylamino)-3-nitrobenzenesu lfo namide; (R)-N-(7-(l -((4'-chloro-4-fluorobiphenyl-2-yl)methyl)-4-deuteropiperidi n-4-yl)- 5 , 6, 7, 8-tetrahydropyrido[3 ,4-d] pyr i mid in-4-yl)-4-(4-(dimethy lamino)- 1 - (phenylthio) butan-2-y lamino)-3-(tri fluoromethy 1 sulfony 1 )benzenesulfonam ide ;

(R)-N-(7-( 1 -((2-(4-chlorophenyl)-4,4-dimethylcyclohex- 1 -enyl)methyl)-4- deuteropiperidin-4-yl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimi din-4-yl)-4-( 1 -(2- fluorophenylthio)-4-(4-methylpiperazin-l -yl)butan-2-ylamino)-3- (trifluoromethylsulfonyl)benzenesulfonamide;

(R)-N-(7-(l -((2-(4-chlorophenyl)-4,4-dimethylcyclohex- 1 -enyl)methyl)-4- deuteropiperidin-4-yl)-5,6,7,8-tetrahydropyrido[3,4-d]pyrimi din-4-yl)-4-(l-(phenylthio)- 4-(pyrrolidin-l -yl)butan-2-ylamino)-3-(tr ' ifluoromethylsulfonyl)benzenesulfonamide; and N-{6-[l -(4'-Chloro-biphenyl-2-ylmethyl)-p ' iperidin-4-yl]-5,6,7,8-tetrahydro- 4H- 1 ,2,3a,6-tetraaza-azulen-3-yl}-4-((R)-3-dimethylamino- 1 -phenyl-sulfanylmethyl- propylamino)-3-trifluoromethanesulfonyl-benzenesulfonamide; or a pharmaceutically acceptable salt thereof.

In another aspect of the present invention a pharmaceutical composition is provided which comprises a compound of the present invention and a pharmaceutically acceptable carrier or excipient. The pharmaceutical composition optionally comprises at least one additional pharmaceutical agent (suitable pharmaceutical agents are described herein below).

In yet another aspect of the present invention, a method of inhibiting Bcl-2 activity is provided comprising the step of administering to a subject in need thereof (i) a therapeutically effective amount of a compound or the present invention, or (ii) a pharmaceutical composition comprising a compound of the present invention and at least one pharmaceutically acceptable excipient.

Still further, the present invention includes a method of treating a proliferative disease comprising the step of administering to a subject in need thereof (i) a

therapeutically effective amount of a compound of the present invention, or (ii) a pharmaceutical composition comprising a compound of the present invention, and at least one pharmaceutically acceptable excipient.

Alternatively, the method for treating a proliferative disease or inhibiting Bcl-2 activity may include a combination therapy which comprises the step(s) of administering (i) a first composition comprising a compound of the present invention and a pharmaceutically acceptable carrier or excipient; and

(ii) a second composition comprising at least one additional pharmaceutical agent and a pharmaceutically acceptable carrier or excipient;

wherein said at least one additional pharmaceutical agent is an anticancer agent, chemotherapy agent, or antiproliferative compound.

The first composition and the second composition may be administered simultaneously or sequentially in any order.

Also included herein is administering a compound of the present invention or a pharmaceutical composition including a compound of the present invention for use in therapy.

Preferably, the disease, disorder, or syndrome is hyperproliferative in a subject, wherein said subject is an animal including humans, and is selected from the group consisting of cancer and inflammation.

The present invention further includes a pharmaceutical composition comprising

(i) a compound of the present invention, and (ii) a pharmaceutically acceptable carrier or excipient. Still further, the present invention includes a pharmaceutical composition comprising a compound of the present invention in combination with a second active agent, and a pharmaceutically acceptable carrier or excipient.

Definitions

As used herein, "alkyl" refers to a straight chain or branched hydrocarbon (C n H 2n+ i). Alkyl moieties having from 1 to 5 carbons are referred to as "lower alkyl" and examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec- butyl, t-butyl, isobutyl, n-pentyl, iso-pentyl, and neopentyl). The term "alkylene" refers to an alkyl moiety where the moiety contains two binding sites. The alkylene group may be straight (e.g., -(CH 2 )-, -(CH 2 ) 2 -, -(CH 2 ) 3 -, -(CH 2 ) 4 -, etc.) or branched (e.g., -CH(CH 3 )-, -C(CH 3 ) 2 -, -CH 2 CH(CH 3 >, -CH(CH 3 )-CH 2 -, -C(CH 3 ) 2 -CH 2 -, etc.). Suitable alkylene moieties are the same as those described above for alkyl except with two binding sites instead of just one. "Halo-substituted alkyi" refers to an alkyi group substituted with halogen groups, e.g. fluoro groups. For example, where the substituent is fluoro, common haloalkyl groups are trifluoroalkyl, 2, 2, 2-trifluoroethyl or 2, 2, 2, 1 , I -pentafluoroethyl groups. Generally, a halo-substituted (Ci-C 6 ) alkyi is substituted with up to seven halogen atoms, which may be the same or different. A perhalo alkyi refers to an alkyi group where each of the hydrogen atoms is replaced with a halogen (e.g., trifluoromethyl).

The term "alkenyl" refers to a monovalent group derived from a hydrocarbon having at least one carbon-carbon double bond. For example, vinyl, prop- l -enyl, prop-2- enyl, 2-methylprop-2-enyl, 3-methylbut-2-enyl, and the like. The term "alkenylene" refers to an alkenyl moiety containing two binding sites. For example, -CH 2 -CH=CH- CH 2 -. Suitable alkenylene moieties are the same as those described above for alkenyl except with two binding sites instead of just one.

The term "alkynyl" refers to a monovalent group derived from a hydrocarbon having at least one carbon-carbon triple bond. The term "C2-C6-alkynyl" refers to a monovalent group derived from a hydrocarbon having two to six carbon atoms and comprising at least one carbon-carbon triple bond.

The term "alkoxy" refers to a group in which an alkyi group is attached to oxygen, wherein alkyi is as previously defined.

The term "cycloalkyl" refers to a monocyclic, bicyclic, or spiral, fully or partially saturated carbocyclic ring. The cycloalkyl may be attached using any of the ring members. Suitable cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. A partially monocyclic saturated ring includes moieties such as cyclohexenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl,

cyclohexadienyl, etc. The term "cycloalkylene" refers to a fully saturated carbocyclic ring(s) having two points of attachment. The carbocyclic ring may be a single ring, a bicyclic ring, or a spiral ring where the two binding sites on the bicyclic ring and spiral ring may be on the same ring or different rings. See, e.g., the illustration below.

Same ring Different ring

The term "aryl" refers to aromatic moieties having a single (e.g., phenyl) or a fused ring system (e.g., naphthalene, anthracene, phenanthrene, etc.). A typical aryl group is a 6- to 14-membered aromatic carbocyclic ring(s). A fused aromatic ring system may also include a phenyl fused to a partially or fully saturated cycloalkyl. For example, 2,3- dihydroindenyl, 1,2,3,4-tetrahydronaphthalenyl, 1,2-dihydronaphthalenyl, 2,3- dihydronaphthalenyl, 9,10-dihydroanthracenyl, fluorenyl, and the like.

The term "arylene" refers to a carbocyclic aromatic moiety having two binding sites. Suitable arylenes include those groups described above for an aryl moiety except with two binding sites rather than one. For example, 1,2-phenylene, 1 ,3-phenylene, 1,4- phenylene, 1,3- naphthylene, 1,4- naphthylene, 1,5-naphthylene, 1,6-naphthylene, 1,7- naphthylene, 2,3- naphthylene, 2,4-napthylene, 2,5-naphthylene, 2,6- naphthylene, 2,7- naphthylene, 3,4-naphthylene, 3, 5 -naphthylene, 3,6-naphthylene, 3,7-naphthylene, etc. The two binding sites on the fused arylene system may be on the same ring or different rings.

The term "heteroaryl" refers to a monocyclic or fused ring system, wherein the monocyclic and at least one of the bicyclic fused rings is an aromatic ring comprising either (a) 1 to 4 nitrogen atoms, (b) one oxygen or one sulphur atom or (c) 1 oxygen atom or 1 sulphur atom and 1 or 2 nitrogen atoms, and the fused ring may be an aryl group, another heteroaryl, a saturated or partially unsaturated cycloalkyl, or a saturated or partially unsaturated heterocycle. The heteroaryl may optionally include one to three ring members selected from the group consisting of O, S or N. The heteroaryl may be attached using any of the ring members. Suitable monocyclic heteroaryl groups include pyridyl, thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl and tetrazolyl. Suitable fused heteroaryl groups include indolyl, benzofuranyl, quinolyl, isoquinolyl indazolyl, indolinyl, isoindolyl, indolizinyl, benzamidazolyl, and quinolinyl. The term "heteroarylene" refers to a biradical heteroaryl monocyclic or fused ring system wherein the ring(s) have two points of attachment which may be on the same ring or different rings in the case of a fused ring system. The term "Cycloheteroalkyl" or "heterocycle" refers to a nonaromatic ring(s) that are either partially or fully saturated and may exist as a single ring, bicyclic ring or a spiral ring comprising one or two ring members selected from the group consisting of N(R 27 ), O or S(0) r . The cycloheteroalkyl may optionally include one to three ring members each independently selected from C(=0), N(R 28 )q, O or S(0)r where R 27 or R 28 is H or (Ci-C6)alkyl, q is 0-1 and r is 0-2. The cycloheteroalkyl may be attached using any of the ring members. Suitable heterocycloalkyi groups include [ 1 , 3] dioxolane, [1 , 4] dioxane, oxiranyl, aziridinyl, oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, morpholino, thiomorpholinyl, piperazinyl, azepinyl, oxapinyl, oxazepinyl and diazepinyl. The term "cycloheteroalkylene" refers to a biradical heterocyle having two points of attachment. The heterocyclene ring may be a single ring, a bicyclic ring, or a spiral ring where the two binding sites on the bicyclic ring and spiral ring may be on the same ring or different rings. See, e.g., the illustration below.

Same ring Different ring

"Halogen" or "halo" may be fluorine, chlorine, bromine or iodine.

It is to be understood that the terminology C (O) refers to a -C=0 group, whether it be ketone, aldehyde or acid or acid derivative. Similarly, S (O) refers to a -S=0 group.

The phrase "therapeutically effective amount" means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.

The term "animal" refers to humans (male or female), companion animals (e.g., dogs, cats and horses), food-source animals, zoo animals, marine animals, birds and other similar animal species. "Edible animals" refers to food-source animals such as cows, pigs, sheep and poultry. A preferred animal is human. The phrase "pharmaceutically acceptable" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.

The terms "treating", "treat", or "treatment" embrace both preventative, i.e., prophylactic, and palliative treatment.

The term "compounds of the present invention" (unless specifically identified otherwise) refer to compounds of Formula (I) and (lb), and salts thereof, as well as all stereoisomers (including diastereoisomers and enantiomers), tautomers and isotopically labeled compounds (including deuterium substitutions), as well as inherently formed moieties (e.g., polymorphs, solvates and/or hydrates). For purposes of this invention, hydrates and solvates are considered compositions comprising a compound of the present invention and an excipient (e.g, water or a solvent).

DETAILED DESCRIPTION

Compounds of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-19, Wiley, New York (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer-Verlag, Berlin, including supplements (also available via the Beil stein online database)).

For illustrative purposes, the reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key

intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.

In the preparation of compounds of the present invention, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.

Scheme 1 below illustrates how one could make compounds of Formula (I) or (lb). Those of skill in the art will know how to modify the conditions and/or starting materials to make other useful derivatives.

Scheme 1

Compounds of Formula I can be prepared by first performing a metal-catalyzed cross-coupling of sulfonamides of Type II with heteroaryl halides of Type III (Hal is typically CI or Br and Pg is a protecting group) to afford heteroaryl sulfonamides of Type IV. For example, the cross-coupling can be accomplished using

tris(dibenzylideneacetone)dipalladium(0)(also referred to as Pd 2 (dba)3), 2-(2- dicyclohexyl-phosphanylphenyl)-N,N-dimethylaniline (also know as DavePhos), cesium carbonate in dioxane at elevated temperatures (e.g., about 180°C). Alternatively, the cross-coupling can be accomplished using copper iodide, cesium carbonate, and N,N- dimethylcyclohexane-l,2-diamine in toluene at about 90°C.

The nitrogen protection group may then be removed using conditions appropriate for the particular protecting group used to produce intermediates of Type V. Intermediates of Type V can then be subjected to reductive amination, either with ketones of Type VI, to afford, following removal of the protecting group, intermediates of Type VII, or with ketones of Type IX, to afford compounds of Formula I. A second reductive amination of amines of Type VII with carbonyl compounds of Type VIII also affords compounds of Formula I. For example, the reductive amination may be accomplished using sodium cyanoborohydride or sodium triacetoxyborohydride using conditions well- known to those of skill in the art.

It is recognized that in order to illustrate the general scheme, variable A as

described in the claims is shown as: , and variable R 1 as described

in the claims is shown as

The compounds may be isolated and used as the compound per se or as its salt. As used herein, the terms "salt" or "salts" refers to an acid addition or base addition salt of a compound of the invention. "Salts" include in particular "pharmaceutical acceptable salts". The term "pharmaceutically acceptable salts" refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable. In many cases, the compounds of the present invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.

Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate,

bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfornate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate,

methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polygalacturonate, propionate, stearate, succinate, sulfosalicylate, tartrate, tosylate and trifluoroacetate salts.

Inorganic acids from which salts can be derived include, for example,

hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.

Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fiimaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.

Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.

Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns 1 to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.

Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.

The pharmaceutically acceptable salts of the present invention can be synthesized from a parent compound, a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, Or in a mixture of the two.

Generally, use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable. Lists of additional suitable salts can be found, e.g., in "Remington's Pharmaceutical Sciences", 20th ed., Mack Publishing Company, Easton, Pa., (1 85); and in "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" by Stahl and Wermuth (Wiley- VCH, Weinheim, Germany, 2002).

The present invention includes all pharmaceutically acceptable isotopically- labeled compounds of the invention, i.e. compounds of formula (I) and (lb), wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.

Examples of isotopes suitable for inclusion in the compounds of the invention comprises isotopes of hydrogen, such as 2 H and 3 H, carbon, such as U C, 13 C and l C, chlorine, such as CI, fluorine, such as F, iodine, such as I and I, nitrogen, such as 13 N and l5 N, oxygen, such as l 5 0, 17 0 and l8 0, phosphorus, such as 32 P, and sulphur, such as 3S S.

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

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

Substitution with positron emitting isotopes, such as U C, 1 F, 15 0 and 13 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.

lsotopically-labeled compounds of formula (I) and (lb) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.

It will be recognized by those skilled in the art that the compounds of the present invention may contain chiral centers and as such may exist in different isomeric forms. As used herein, the term "isomers" refers to different compounds that have the same molecular formula but differ in arrangement and configuration of the atoms. Also as used herein, the term "an optical isomer" or "a stereoisomer" refers to any of the various stereo isomeric configurations which may exist for a given compound of the present invention and includes geometric isomers. It is understood that a substituent may be attached at a chiral center of a carbon atom. Therefore, the invention includes enantiomers, diastereomers or racemates of the compound.

"Enantiomers" are a pair of stereoisomers that are non- superimposable mirror images of each other. A 1 : 1 mixture of a pair of enantiomers is a "racemic" mixture. The term is used to designate a racemic mixture where appropriate.

"Diastereoisomers" are stereoisomers that have at least two asymmetric atoms, but which are not mirror-images of each other. The absolute stereochemistry is specified according to the Cahn- Ingold- Prelog R-S system. When a compound is a pure enantiomer the stereochemistry at each chiral carbon may be specified by either R or S. Resolved compounds whose absolute configuration is unknown can be designated (+) or (-) depending on the direction (dextro- or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line. Certain of the compounds described herein contain one or more asymmetric centers or axes and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (5)-.

Unless specified otherwise, the compounds of the present invention are meant to include all such possible isomers, including racemic mixtures, optically pure forms and intermediate mixtures. Optically active (R)- and (5)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans-configuration. All tautomeric forms are also intended to be included.

Compounds of the invention that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers. These co-crystals may be prepared from compounds of the present invention by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of the present invention with the co-crystal former under crystallization conditions and isolating co- crystals thereby formed. Suitable co-crystal formers include those described in WO 2004/078163. Hence the invention further provides co-crystals comprising a compound of the present invention.

A compound of the formula (I) or (lb) may also be used to advantage in combination with other antiproliferative compounds. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase 1 inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors, compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors, antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti -androgens; methionine aminopeptidase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3, Hsp90 inhibitors, kinesin spindle protein inhibitors, P13K inhibitors, RAF inhibitors, EDG binders, antileukemia compounds, ribonucleotide reductase inhibitors, S-adenosylmethionine decarboxylase inhibitors, antiproliferative anti-bodies or other chemotherapeutic compounds. Further, alternatively or in addition they may be used in combination with other tumor treatment approaches, including surgery, ionizing radiation, photodynamic therapy, implants, e.g. with corticosteroids, hormones, or they may be used as radiosensitizers. Also, in anti-inflammatory and/or antiproliferative treatment, combination with anti-inflammatory drugs is included. Combination is also possible with antihistamine drug substances, bronchodilatatory drugs, NSAID or antagonists of chemokine receptors. More particularly, some therapeutic agents with which a compound of the formula (I) or (lb) may be used include doxorubicin, docetaxel, 5FU, Camptothecin, Erlotinib, Paclitaxel, Carboplatin, Etoposide, and Gemcitabine.

The compounds of the present invention are useful as both prophylactic and therapeutic treatments for diseases or conditions related to the hyperactivity of Bcl-2. Thus, as a further aspect, the invention relates to a method for treating a disease or condition related to the hyperactivity of Bcl-2, or a disease or condition modulated by the Bcl-2, comprising administration of an effective therapeutic amount of a compound of formula (I) or (lb) or a pharmaceutically acceptable salt thereof. As a further aspect, the invention relates to a method for treating proliferative diseases, such as cancer, comprising administration of an effective amount of a compound of formula (I), (lb), or a pharmaceutically acceptable salt thereof. Examples of cancers include but are not limited to: breast cancer, colorectal cancer, ovarian cancer, pancreatic cancer, or prostate cancer, chronic lymphocytic leukemia, diffuse large B-cell lymphomas, follicular lymphomas, chronic or acute leukemia, chronic myeloid leukemia, lymphoid malignancies of T-cell or B-cell origin, lung cancers, such as small cell lung cancer and non-small cell lung cancer, melanoma or other skin cancers, multiple myeloma, ovarian cancer, gastrointestinal cancer (gastric, colorectal, and duodenal), bladder cancer, uterine cancer, cervical cancer, sarcoma of soft tissue origin, kidney cancer, brain tumors, hepatocellular cancer, head and neck cancer, cervical cancer, fibrosarcoma, and other cancers.

The present invention is further exemplified, but not limited, by the following representative examples, which are intended to illustrate the invention and are not to be construed as being limitations thereof.

EXAMPLES

The structure of final products described herein can be confirmed by standard analytical methods, e.g., spectrometric and spectroscopic methods (e.g., MS, NMR, HPLC). Retention times provided in the Examples below were observed on an Agilent 1100 HPLC system; Inertsil ODS3 100 x 3mm C 18 column; flow rate of 1.0 mL/minute; gradient of 5-95% acetonitrile/water with 0.1% formic acid. Compounds are purified by standard methods, e.g., crystallization, flash chromatography, or reversed phase HPLC.

The following abbreviations have the corresponding meaning in the Examples below.

aq. Aqueous

DCE 1 ,2-Dichloroethane

DIPEA Di isopropy lethylami ne

DMA Dimethylacetamide DME 1 ,2-Dimethoxyethane

DMF Dimethylformamide

eq. equivalents

HPLC High-performance liquid chromatography

HR-MS High-resolution mass spectrometry

MS Mass spectrometry

NMR Nuclear magnetic resonance

sat. saturated

SPE Solid-phase extraction

TOF Time-of-Flight mass spectrometry

Preparation of Key Intermediates

Synthesis of Type II Intermediates:

ntermediate 1 : fji Nl.Nl-Dimethyl^-phenylsu fany -butane-l .3-diamine.

Intermediate 1 was prepared as described by Wendt, M. D., et al., in J. Med. Chem. 2006, 49, 1 165- 1 181. Intermediate 2: f/ -3-Morpholin-4-yl-l-phenylsulfanylmethyl-propylamine.

Intermediate 2 was prepared as described by Wendt, M. D., et al., in J. Med. Chem. 2006, 49, 1 165-1 181.

Intermediate 3 : 4-Fluoro-3-(trifluoromethylsulfonyl)benzenesulfonamide.

Intermediate 3 was prepared as described by Park, C, et al., in J. Med. Chem.

2008, 51, 6902-6915.

Intermediate 4: 4-f(-?V3-Dimethylamino- 1 -phenylsulfanylmethyl-propylamino)-3- tr i fluoromethanesul fonyl -benzen lfonam ide.

A solution of 4-fluoro-3-(trifluoromethylsulfonyl)benzenesulfonamide (685 mg, 2.3 mmol), (if)- l , l-dimethyl-4-phenylsulfanyl-butane-l,3-diamine (500 mg, 2.3 mmol), and DIPEA (0.78 mL, 4.5 mmol) in DMA (18.8 mL) was heated to 100°C for 6 hours. The reaction was cooled to room temperature and diluted with EtOAc/ heptanes (1 : 1, 50 mL). This solution was washed with water, the layers were separated, and the aqueous layer was extracted with EtOAc/ heptanes (1 : 1) (50 mL x 3). The combined organic layers were washed with water and brine, dried over MgS0 4 , and concentrated to afford the title compound as a yellow solid (965 mg, 82% yield). MS (ESI) m/e (M+H + ):

512.2 Intermediate 5: 4-f( ?)-3-Morpholin-4-yl- l-phenylsulfanylmethyl-propylamino -3- trifluoromethanesulfonyl-benzenesulfonamide.

The title compound was prepared from 4-fluoro-3-(trifluoromethylsulfonyl)- benzenesulfonamide and ( ^)-3-mo holin-4-yl-l -phenylsulfanylmethyl-propylamine using the procedure described for Intermediate 4. MS (ESI) m/e (M+H + ): 553.6.

Intermediate 6: (R)-4-(4-fdimethylamino ' )-l -(phenylthio')butan-2-ylamino)-3- (trifluoromethvQbenzenesulfonamide.

The title compound was prepared from 4-fluoro-3-(trifluoromethyl)-benzenesulfonamide and (if)-N l ,N l -dimethyl-4-phenylsulfanyl-butane- l ,3-diamine using the procedure described for Intermediate 4. Intermediate 7: (R)-3-cyano-4-(4-(dimethylamino l-(phenylthio)butan-2- ylamino^benzenesulfonami

The title compound was prepared from 4-fluoro-3-(cyano)-benzenesulfonamide and (R)- Nl,Nl-dimethyl-4-phenylsulfanyl-butane-l,3-diamine using the procedure described for Intermediate 4.

Intermediate 8 : ( RV 4-(4-(dimethy lamino)- 1 -(phenyl thio)butan-2-ylaminoV3.5 - difluorobenzenesulfonamide.

The title compound was prepared from 3,4,5-trifluorobenzenesulfonamide and (R)~ Nl,Nl-dimethyl-4-phenylsulfanyl-butane-l,3-diamine using the procedure described for Intermediate 4.

Intermediate 9: 4-((j?)-3-Dimethylamino- 1 -phenyl sulfanylmethyl-propyl amino)-3-nitro- benzenesulfonamide.

Intermediate 9 was prepared as described by Wendt, M. D., et al., in J. Med.

Chem. 2006, 49, 1 165-1 181.

Synthesis of Type III Intermediates:

Intermediate 10: 4-Ch loro-5 , 8-d ih ydro-6H-p yr ido [3 ,4-dlpyrimidine- 7-carboxyl ic acid fert-butyl ester.

STEP A: l -Benzyl-3-oxo-piperidine-4-carboxylic acid ethyl ester (5.0 g, 16.8 mmol) was stirred in dry ethanol (55 mL). The flask was evacuated and filled with nitrogen prior to the addition of 10% Pd/C ( 1 .0 g). The resulting suspension was stirred for 16 hours under an atmosphere of hydrogen. The reaction mixture was filtered through Celite and concentrated to afford a yellow solid (2.9 g). This was immediately dissolved in CH 2 C1 2 (100 mL), treated with Boc-anhydride (4.4 g, 20.2 mmol) and DIPEA (4.3 g, 33.6 mmol), and stirred for 16 hours at room temperature. The organics were washed sequentially with HCI (IN), water and brine, dried over magnesium sulfate, filtered, and concentrated to afford 3-oxo-piperidine-l ,4-dicarboxylic acid 1 -te -butyl ester 4-ethyl ester as a clear oil (4.8 g, 100% yield). MS (ESI) m/e (M+H + ): 271.36

STEP B: 3-Oxo-piperidine- l ,4-dicarboxylic acid 1 -te -butyl ester 4-ethyl ester (4.6 g, 16.8 mmol) was dissolved in EtOH (90 mL), and sodium ethoxide (2.3 g, 33.6 mmol) and formamidine hydrochloride (2.0 g, 25.2 mmol) were sequentially added. The resulting suspension was heated to 70°C and stirred for 5 hours, and then the reaction mixture was cooled to room temperature and concentrated. The residue was dissolved in saturated ammonium chloride and extracted with ethyl acetate. The aqueous phase was adjusted to pH -5.5 using concentrated AcOH, and extracted several more times with ethyl acetate. The combined organic layers were sequentially washed with water and brine, dried, and concentrated to afford 4-hydroxy-5,8-dihydro-6H-pyrido[3,4- d]pyrimidine-7-carboxylic acid /er/-butyl ester as a brown solid (1.8 g, 42% yield). MS (ESl) m/e (M+H + ) = 251.28

STEP C: A mixture of 4-hydroxy-5,8-dihydro-6H-pyrido[3,4-d]pyrimidine-7- carboxylic acid ter/-butyl ester ( 1 .4 g, 5.6 mmol) and triphenylphosphine (2.9 g, 1 1.1 mmol) was stirred in DCE (41 mL) until the solution became clear, and then carbon tetrachloride ( 1.6 mL, 16.7 mmol) was added. The reaction mixture was heated to 70°C and stirred for 2.5 hours. The volatiles were removed in vacuo and the crude material was directly purified by flash chromatography on silica gel (0-50% EtOAc/ heptanes) to afford the title compound as an off-white solid (1.31 g, 87% yield). MS (ESI) m/e (M+H + ): 269.73 Intermediate 11 : /ert-Butyl 4-chloro-2-(trifluoromethyD-5.6-dihydropyrido[3.4- d]pyr im id ine-7 (8H)-carboxylate.

STEP A: Sodium ethoxide was prepared by combining sodium metal (0.5 g, 21.8 mmol) and ethanol (33 mL). At ambient temperature, trifluoroacetamidine (1.35 g, 12.0 mmol) was added, followed immediately by addition of ethyl 1 -benzyl- 3 -oxo-4- piperidinecarboxylate hydrochloride (2.92 g, 9.81 mmol) in portions over 15 minutes. The reaction was stirred at ambient temperature for 1 hour and then heated to 80 °C for 16 hours. The reaction was then cooled to ambient temperature, and the solvent was removed in vacuo. The resulting dark red foamy solid was taken up in diethyl ether (40 mL) and IN aqueous NaOH (40 mL). The layers were separated, and the aqueous layer was washed with Et 2 0 (40 mL). The combined organic layers were extracted with IN NaOH ( 10 mL). The combined aqueous layers were cooled to 0 °C and acidified to pH 7 with concentrated HC1. A tan solid precipitated which was isolated by slow filtration. Drying overnight over the filter afforded 7-benzyl-2-(trifluoromethyl)-5 ,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-ol (2.35 g, 78% yield). MS (ESI) m/e (M+H) + = 310.4.

STEP B: To 7-benzyl-2-(trifluoromethyl)-5,6,7,8-tetrahydropyrido[3,4- d]pyrimidin-4-ol (0.47 g, 1.52 mmol) partially dissolved in methanol (20 mL) was added palladium hydroxide on carbon (70 mg, wet, 20% by weight dry basis). The reaction was stirred for 20 hours in a Parr shaker under a pressure of 50 psi hydrogen. The reaction was then filtered through a Celite plug, eluting with methanol (400 mL). The filtrate was concentrated in vacuo to yield 2-(trifluoromethyl)-5,6,7,8-tetrahydropyrido[3,4- d]pyrimidin-4-ol as a light beige solid (255 mg, 77% yield). Ή NMR (400 MHz, MeOD) δ ppm 4.09 (s, 2H), 3.48 (t, J= 6.32 Hz, 2H), 2.78 (t, J= 6.32 Hz, 2H).

STEP C: To a stirring mixture of 2-(trifluoromethyl)-5,6,7,8- tetrahydropyrido[3,4-d]pyrimidin-4-ol (0.246 g, 1.122 mmol) in tetrahydrofuran (11 mL) was added a solution of di-/er/-butyl dicarbonate (0.269 g, 1.235 mmol) in

tetrahydrofuran (11 mL). The mixture was stirred at ambient temperature for 16 h and was then concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (eluent: 0 to 7% methanol / dichloromethane) to afford teri-butyl 4-hydroxy- 2-(trifluoromethyl)-5,6-dihydropyrido[3,4-d]pyrimidine-7(8H) -carboxylate as a light yellow foamy solid (0.306 g, 85% yield). MS (ESI) m/e (M-H) " = 318.3.

STEP D: fe -Butyl 4-hydroxy-2-(trifluoromethyl)-5,6-dihydropyrido[3,4- d]pyrimidine-7(8H)-carboxylate (5.12 g, 16.05 mmol) was combined with

triphenylphosphine (8.42 g, 32.1 mmol) and stirred in dichloroethane (1 0 mL) at ambient temperature for 15 minutes, and then carbon tetrachloride (7.41 g, 48.1 mmol) was added. The reaction was stirred at 70°C for 3 hours, cooled to room temperature, and concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (elution gradient from 0% to 3% methanol / DCM) to afford the title compound as a clear oil (5.22 g, 86% yield). MS (ESI) m/e (M-H) " = 336.3. lntermed iate 12 : 3 -B romo-4.5.7.8-tetrah vdro- 1.2.3a.6-tetraaza-azulene-6-carbox yli c acid /grf-butyl ester

STEP A: To a stirred solution of ter/-butyl 5-oxo-l,4-diazepane-l-carboxylate (1000 mg, 4.67 mmol) in dichloromethane (10 mL) was added trimethyloxonium tetrafluoroborate (690 mg, 4.67 mmol) under nitrogen and the reaction mixture was stirred for 16 hours. At this point, formic hydrazine (280 mg, 4.67 mmol) in

dichloromethane (8 mL) was added, and the reaction was stirred for an additional 16 hours. The reaction mixture was then concentrated under reduced pressure, resuspended in methanol (10 mL), and heated to reflux for 16 hours. The reaction mixture was concentrated and purified via flash chromatography on silica gel (0-50% 2N NH 3 in methanol in CH 2 C1 2 ) to afford 4,5,7,8-tetrahydro-l,2,3a,6-tetraaza-azulene-6-carboxylic acid tert-butyl ester (680 mg, 61% yield). MS [m/z; (M+l) + ]: 319.3

STEP B: To a suspension of 4,5,7, 8-tetrahydro-l ,2,3a,6-tetraaza-azulene-6- carboxylic acid tert-bxAy] ester (200 mg, 0.839 mmol) in water was added 10N NaOH until a clear solution was formed. Br 2 (0.432 mL, 8.39 mmol) was then added slowly while maintaining a pH of 12 by addition of cone. NaOH. The reaction was stirred at ambient temperature for 16 hours. The reaction mixture was then acidified to pH ~4 with 6 M HC1. The organics were extracted three times with CH 2 C1 2 . The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to afford the title compound as a white solid (185 mg, 70% yield).

Ή NMR (400 MHz, DMSO-i/ 6 ) δ ppm 1.43 (s, 9H), 3.05 (br. s., 2H), 3.54 (br. s., 2H), 3.63 - 3.71 (m, 2H), 4.10 (br. s., 2H). General Procedure for the Pd-catalyzed Cross-coupling of Sulfonamides II with Heteroaryl Chlorid

A 20 mL oven-dried vial equipped with a teflon cap was charged with Pd 2 (dba) 3 (0.01 eq), cesium carbonate (1.4 eq), and 2'-(dicyclohexylphosphino)-N,N- di methyl biphenyl-2-amine (DavePhos) (0.03 eq). The vial was then capped and purged with nitrogen. Dioxane (1.5 mL) was added, and the resulting suspension was stirred for 15 minutes under an atmosphere of nitrogen. A solution of sulfonamide (1 eq) and heteroaryl chloride (1 eq) in dioxane (1.0 mL/ mmol II) was added. After 5 minutes, the mixture was placed in a microwave and heated at 180°C for 30 minutes. The solvent was then filtered through a pad of magnesium sulfate, rinsing with CH 2 C1 2 . The filtrate was concentrated to provide an orange solid. The solid was redissolved in CH 2 C1 2 and the resulting solution was washed with saturated aqueous NaHC0 3 followed by brine. The combined aqueous layers were extracted twice with CH 2 C1 2 and once with EtOAc. The combined organic layers were dried over MgS0 4 , concentrated, and the crude residue was purified by flash chromatography on silica gel (gradient: 0 to 40% methanol/

C¾CI 2 ). Intermediate 13: 4-f4- 4-((Jί)-3-lyloφholin-4-yl- l -phenylsulfanylmeth l-propylamino - 3-trifluoromethanesulfonyl-benzenesulfonylamino1-5.8-dihvdro -6H- pyridof3,4dlpyrimidin-7-yl }-piperidine-l -carboxylic acid tert-butyl ester.

Following the general procedure, 4-(( ^)-3-moφholin-4-yl- l-phenylsulfanylmethyl- propylamino)-3-trifluoromethanesulfonyl-benzenesuIfonamide (616 mg, 1.1 mmol) and 4-chloro-5,8-dihydro-6H-pyrido[3,4-d]pyrimidine-7-carboxylic acid tert-butyl ester (300 mg, 1.1 mmol) afforded the title compound (600 mg, 68% yield). MS (ESI) m/e (M+H + ): 787.4 Intermediates 14-18 listed below were prepared by cross-coupling of

sulfonamides II with heteroaryl chlorides III following the general procedure described above.

Intermediate 14: (R)-tert-butyl 4-(4-(4-(dimethylamino)- l-(phenylthio butan-2- ylamino)-3-(tr ' ifluoromethylsulfonyl)phenylsulfonamido)-5.6-dihvdropy ridoi3.4- d1pyrimidine-7(8I-D-carboxylate.

MS [m/z; M+l] = 745

Intermediate 15: fRVtert-butyl 4-(4-(4-(dimethylamino)-l-fphenylthio')butan-2- ylamino)-3-nitrophenylsulfonamido')-5.6-dihvdropyridof3.4-d1 pyrimidine-7f8H)- carboxylate.

MS [m/z; M+l] = 658 Intermediate 16: (R)-tert-butyl 4-f4-f4-fdimethylamino)-l-fphenylthio)butan-2- ylaminoV3-(trifluoromemylsulfonynphenylsulfonamidoV2-ftriflu oromemyl)-5.6- dihydropyridor3,4-dlpyrimidine-7(8H)-carboxylate.

MS [m/z; M+l] = 813

Intermediate 17: (R)-tert-butyl 4-f4-f4-morpholino-l-(phenylthio ' )butan-2-ylaminoV3- (trifluoromethylsulfonynphenylsulfonamido)-2-(trifluoromethy l)-5,6-dihydropyri

MS [m/z; M+l ] = 855.

Intermediate 18: (RV/e -butyl 4-(4-(4-fdimethylamino)-l-fphenylthio butan-2- viamino)-3-nitrophenylsulfonamido -2-ftrifluoromethyl)-5.6-dihydropyrido[3.4- dlpyrimidine-7(8H)-carboxyiate.

MS [m/z; M+l] = 727.

Intermediate 19: 3- [4-( (R)-3 -Morpholin-4- yl- 1 -pheny lsulfany 1 methyl-propylamino)-3 - trifluoromethanesulfonyl-benzenesulfonylaminol-5,6-dihvdro-8 H-[l,2.41triazolo 4,3- alpyrazine-7-carboxylic acid ferf-butyl ester

3-Bromo-5,6-dihydro-8H-[l,2,4]triazolo[4,3-a]pyrazine-7-carb oxylic acid tert- butyl ester (54.8 mg, 0.181 mmol), 4-(( ^)-3-mo holin-4-yl-l-phenylsulfanyl-methyl- propylamino)-3-trifluoromethanesulfonyl-benzenesulfonamide ( 100 mg, 0.181 mmol), (IS, 25)-N,N'-dimethyl-cyclohexane-l,2-diamine (07.71 mg, 0.054 mmol), cesium carbonate (100 mg, 0.307 mmol), and copper (I) iodide (5.16 mg, 0.027 mmol) were added to a microwave vial followed by toluene (2 mL). The reaction mixture was degassed for 15 minutes under nitrogen and then heated to 90°C for 14 hours. The crude material was directly purified via flash chromatography on silica gel (0-25% 7 N ammonia in methanol in CH 2 C1 2 ) to afford the title compound (130 mg, 92% yield). MS [m/z; (M+l) + ]: 776.5 Intermediate 20: 3-[4-( (j?)-3-Morpholin-4-yl- 1 -phenylsulfanylmethyl-propylaminol-S- trifluoromethanesulfonyl-benzenesulfonylamino1-4.5.7.8-tetra hvdro-l,2,3a,6-tetraaza- azulene-6-carboxylic acid tert-butyl ester

3-Bromo-4,5,7,8-tetrahydro- 1 ,2,3a,6-tetraaza-azulene-6-carboxylic acid tert-b ty\ ester (154 mg, 0.486 mmol), 4-((-¾)-3-mo holin-4-yl-l -phenylsulfanylmethyl- propylamino)-3-trifluoromethane sulfonyl-benzenesulfonamide (269 mg, 0.486 mmol), (15, 25)-N,N'-dimethyl-cyclohexane-l,2-diamine (0.01 1 mL, 0.146 mmol), cesium carbonate (269 mg, 0.825 mmol), and copper (I) iodide (13.87 mg, 0.073 mmol) were added to a microwave vial followed by toluene (8 mL). The reaction mixture was degassed for 15 minutes under nitrogen, sealed, and then heated conventionally to 90°C for 14 hours. The crude material was directly purified via flash chromatography on silica gel (0-15% 2N NH 3 in methanol in CH 2 C1 2 ) to afford the title compound (170 mg, 44% yield). MS [m/z; (M+l ) + ]: 790.5

Intermediate 21 : (R)-tert -Butyl 4-(4-(4-morpholino-l -(phenylthio)butan-2-ylamino)-3- ftr ' ifluoromethylsulfonyl)phenylsulfonamido)-5H-pyrrolo 3.4-d1pyrimidine-6(7H)- carboxylate.

STEP A: fert-Butyl 4-hydroxy-5H-pyrrolo[3,4-d]pyrimidine-6(7H)-carboxylate (300 mg, 1.264 mmol) and (lH-benzo[d][l,2,3]triazol-l-yloxy)tris(dimethylamino)- Ophosphonium hexafluorophosphate(V) (671 mg, 1.517 mmol) were stirred in

acetonitrile ( 10 mL). To the slurry was added 2,3,4,6,7,8,9, 10-octahydropyrimido-[ 1 ,2- a]azepine (289 mg, 1.897 mmol). The reaction mixture was stirred 1.5 hours. The solvents were removed in vacuo to afford tert-butyl 4-(lH-benzo[d][l,2,3]triazol-l- yloxy)-5H-pyrrolo[3,4-d]pyrimidine-6(7H)-carboxylate which was carried on to the next step without further purification.

STEP B: tert-Butyl 4-(lH-benzo[d][l,2,3]triazol-l-yloxy)-5H-pyrrolo[3,4- d]pyrimidine-6(7H)-carboxylate (448 mg, 1.264 mmol) , (ii)-4-(4-morpholino-l- (phenylthio)butan-2-ylamino)-3(trifluoromethylsulfonyl)benze nesulfonamide (910 mg, 1.643 mmol), and potassium carbonate (524 mg, 3.79 mmol) were combined in N,N- dimethylformamide (6.3 mL) and stirred for 1.5 hours at 85°C and then for 2.5 hours at 95°C. The reaction was then cooled to room temperature and stirred for 16 hours. The reaction was treated with water (-30 mL) and sonicated. A dark brown solid stuck to the flask. The solid was washed several times with water and then several times with diethyl ether. This residue was then treated with methanol (~15 mL), and the off-white precipitate formed was removed by filtration. The dark brown filtrate was concentrated in vacuo to afford a brown solid. The water and ether washes were combined and diluted with more water and ether. The layers were separated, and the organic layer was dried over sodium sulfate, filtered and concentrated down. This residue was combined with the brown solid and purified by flash chromatography on silica gel (0 to 10% methanol/ ethyl acetate followed by 0 to 30% methanol/ dichloromethane) to afford the title compound (1 9 mg, 13% yield). MS (ESI) m/e (M+H) + = 773.5.

General Procedure for Removal of the Boc Protecting Group from Amines IV.

Amine ( 1 eq) was dissolved in CH 2 Ci 2 (5 mL/ mmol) and the flask was cooled to 0°C. Hydrochloric acid (2N in diethyl ether, 4 eq) was added slowly, and the reaction mixture was then stirred for 3 hours at room temperature. The solvent was removed in vacuo, and CH 2 C1 2 (20 mL) was added to the residue. The resulting solution was washed with saturated aqueous NaHC0 3 and brine. The combined aqueous layers were extracted twice with CH 2 Ci 2 and once with EtOAc. The organic layers were combined, dried over MgS0 4 , and concentrated. No additional purification was performed.

Intermediate 22: (J?)-4-(4-Morpholino- l -(phenylthio)butan-2-ylamino)-N-(5.6.7.8- tetrahvdropyrido[3,4-dlDyrimidin-4- l)-3-ftrifluoromethylsulfonyl)-benzenesulfonamide.

Following the general procedure, 4-{4-[4-((/i)-3-Morpholin-4-yl-l - phenylsulfanylmethyl-propyiamino)-3-trifluoromethanesulfonyI -benzene- sulfonylamino]-5,8-dihydro-6H-pyrido[3,4-d]pyrimidin-7-yl}-p iperidine- l -carboxylic acid tert-butyl ester (300 mg, 0.38 mmol) afforded the title compound (170 mg, 50%). MS (ESI) m/e (Μ+Η*): 687.3

Intermediates 23-28 were prepared by deprotection of amines IV following the general procedure described above.

Intermediate 23: (R)-4-f4-fdimethylamino)-l-(phenylthio)butan-2-ylamino ' )-N-f5.6.7.8- tetrahvdropyrido[3.4-d1pyrimidin-4-yl)-3-(trifluoromethylsul fonylVbenzenesulfonamide.

MS [m/z; M+l] = 645.

Intermediate 24: (R)-4-(4-(dimethylamino)-l -(phenylthio)butan-2-ylaminoV3-nitro-N- (5.6.7,8-tetrahvdropyrido[3.4-dl yrimidin-4-yl benzenesulfonamide.

MS [m/z; M+l] = 558. Intermediate 25: fR -f4-fdimethylamino)-l -(phenylthio butan-2-viamino)-N- 2- ftrifluoromethyl)-5.6,7,8-tetrahydropyrido[3.4-d]pyrimidin-4 -yl)-3- ftrifluoromethylsulfonyDbenzenesulfonamide.

MS [m/z; M+ l] = 713.

Intermediate 26: (RV4-(4-morpholino- 1 -fphenylthio)butan-2-ylamino)-N-r2- ftrifluoromethyl)-5,6.7.8-tetrahydropyrido[3,4-dlpyrimidin-4 -yl)-3- ftrifluoromethylsulfonyDbenzenesulfonamide.

MS [m/z; M+ l ] = 755.

Intermediate 27: (R)-4-(4-(dimethylamino)- l -(phenylthio butan-2-ylamino)-3-nitro-N- (2-(trifluoromethvn-5,6,7,8-tetrahydropyrido[3.4-d]pyrimidin -4-yl)-benzenesulfonamide.

MS [m/z; M+l] = 627.

Intermediate 28: (ΚνΝ-(6.7-άΐ1ΐν(ΐΓθ-5Η-ρν η ΌΐοΓ3.4-ά1ρνΓΪηΐΐί1ϊη-4-ν1)-4-(4-ηι φ1ΐοΗηο- 1 -Cpheny lthi o)fautan-2-yl am ino)-3-(trifluoromethyl sulfon P-benzenesu lfonam ide

MS [m/z; M+l] = 673.

Intermediate 29: 4-((R)-3-Morpholin-4-yl-l-phenylsulfanylmethyl-piOpylamino)- N- f5.6J.8-tetrahydro-n,2^1triazolo 4,3-alpyrazin-3-yl)-3-trifluoromethanesulfonyl- benzenesu lfonam ide .

To a solution of 3- 4-(( f)-3-mo holin-4-yl-l -phenylsulfanylmethyl- propy I amino)-3 -tri fluoromethanesu 1 fony 1 -benzenesu 1 fony lamino] -5 , 6-di hy dro-8 H- [ l ,2,4]triazolo[4,3-a]pyrazine-7-carboxylic acid tert-butyl ester (130 mg, 0.168 mmol) in dichloromethane (3 mL) under nitrogen was added trifluoroacetic acid (0.323 mL, 4.19 mmol), and the reaction was stirred for 2.5 hours. The reaction mixture was then concentrated and diluted with water and CH 2 C1 2 . It was then basified to pH

approximately 8 with saturated Na 2 C0 3 . The organics were extracted three times with CH 2 C1 2 . The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to afford the title compound (1 12 mg, 99% yield). MS [m/z; (M+ l ) + ]: 676.5

Intermediate 30: 4-((j?)-3-Morpholin-4-yl-l -phenylsulfanylmethyl-propylamino)-N- (5,6,7,8-tetrahvdro-4H- 1.2.3a.6-tetraaza-azulen-3-yl)-3-trifluoromethanesulfonyl- benzenesul fonamide

To a solution of 3-[4-(( ^)-3-mo holin-4-yl- l -phenylsulfanylmethyl- propylamino)-3 trifluoromethanesulfonyl-benzenesulfonylamino]-4,5,7,8-tetra hydro- l,2,3a,6-tetraaza-azulene-6-carboxylic acid ter/-butyl ester (170 mg, 0.215 mmol) in dichloromethane (4 mL) under nitrogen was added trifluoroacetic acid (0.415 mL, 5.38 mmol), and the reaction was stirred for 2 hours. The reaction mixture was concentrated and diluted with water and dichloromethane. It was then basified to pH approximately 8 with saturated Na 2 C0 3 . The aqueous layer was extracted three times with

dichloromethane. The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to afford the title compound (155 mg, 100% yield). MS [m/z; (M+l) + ]: 690.4 Synthesis of Types XIII and IX Intermediates.

Intermediate 31 : 4'-Chloro-biphenyl-2-carbaldehyde.

To a solution of 2-bromobenzaldehyde (18.0 g, 97.0 mmol) and 4-chloro- phenylboronic acid (19.0 g, 122 mmol) in a 1:1 mixture of ethanol/DME (400 mL) is added a solution of potassium carbonate (26.9 g, 195 mmol) in water (100 mL), and the reaction mixture is purged with nitrogen for 30 minutes. Tetrakis(triphenyl- phosphine)palladium (5.62 g, 4.86 mmol) is then added, and the reaction is purged again with nitrogen for 20 minutes. The pale yellow-green solution is heated to 75 °C and stirred for 20 hours. The reaction is then cooled to room temperature, diluted with ethyl acetate (200 mL), and filtered through a ZAPCAP filter. The filtrate is washed with brine (400 mL) and the aqueous layer is back-extracted with ethyl acetate (2 X 200 mL). The combined organic layers are dried over MgS0 4 and concentrated to afford an orange oily solid. This crude material is purified via flash chromatography on silica gel (0-20% EtOAc/ heptanes) to afford the title compound (14.9 g, 71% yield). Ή NMR (400 MHz, CDC1 3 ) δ 10.0 (s, 1H), 8.05 (d, J= 7.5 Hz, 1H), 7.68 (t, J= 6.2 Hz, 1H), 7.54 (t, J= 7.4 Hz, I H), 7.49 (d, J= 9.4 Hz, 2H), 7.45 (d, J= 9.1 Hz, 1 H), 7.34 (d, J= 8.5 Hz, 2H). Intermediate 32: 4'-Chloro-2-(iodomethyl)biphenyl

STEP A: 4'-Chloro-biphenyl-2-carbaldehyde (6.50 g, 30.0 mmol) was dissolved in CH2CI2 (75 mL) and methanol (75 mL). The solution was cooled to 0°C in an ice bath, and then sodium borohydride ( 1.36 g, 36.0 mmol) was added slowly portionwise. The reaction was allowed to warm gradually to room temperature and stir for 1 hours. The reaction was quenched by addition of 2M NaOH, and the organics were extracted with CH 2 CI 2 (3 x 75 mL). The combined organic layers were dried over MgS0 4 and concentrated to afford (4'-chloro-biphenyl-2-yl)-methanol (6.42 g, 98% yield), which was carried on to the next step without further purification. MS (ESI) m/e (M-H) ~ : 217.4 STEP B: Triphenylphosphine (8.47 g, 32.3 mmol), iodine (8.94 g, 35.2 mmol), imidazole (2.40 g, 35.2 mmol), and CH 2 C1 2 (1 10 mL) were combined in a round-bottom flask, and the mixture was stirred for 1 5 minutes. A solution of (4'-chloro-biphenyl-2- yl)-methanol in CH 2 C1 2 (40 mL) was then added, and the reaction was stirred at room temperature for 16 hours (flask was wrapped in aluminum foil to shield it from the light). A saturated aqueous Na 2 S 2 0 was added. The layers were separated, and the aqueous layer was extracted with CH 2 C1 2 (2 x 100 mL). The organic layers were combined, dried over MgS0 4 , and concentrated. The crude residue was purified by flash chromatography on silica gel (0-10% EtOAc/ heptanes) to afford the title compound as an off-white solid (5.33 g, 55% yield). 1 H NMR (400 MHz, CDC1 3 ) δ ppm 7.51 - 7.53 (m, 1 H), 7.38 - 7.46 (m, 4H), 7.28 - 7.37 (m, 2H); 7.15 - 7.17 (m, 1H).

Intermediate 33: l -(4'-Chloro-biphenyl-2-ylmethyn-p ' iperidin-4-one.

STEP A: 4'-Chloro-biphenyl-2-carbaldehvde (5.00 g, 23.08 mmol) and 1,4-dioxa- 8-azaspiro[4.5]decane (4.30 g, 30.0 mmol) were combined in DCE (90 mL) and methanol (68 mL). MgS0 4 (~5 g) was added, and the resulting mixture was stirred for 1 hour at room temperature. Sodium triacetoxyborohydride (14.67 g, 69.2 mmol) was then added, and the reaction was stirred for 16 hours at room temperature. The solvent was removed in vacuo, and the residue was resuspended in CH C1 / H 2 0. The layers were separated, and the aqueous layer was extracted with CH C1 2 (2 x 100 mL). The organic layers were combined, dried over MgS04, and concentrated. Flash chromatography on silica gel (0- 10% NH 3 in methanol (~2M)/ CH C1 ) afforded 8-((4'-chlorobiphenyl-2-yl)methyl)- 1 ,4- dioxa-8-azaspiro[4.5]decane as a brown oil (4.73 g, 60% yield). MS (ESI) m/e (M+H) + : 344.5

STEP B: 8-((4'-Chlorobiphenyl-2-yl)methyl)-l,4-dioxa-8-azaspiro[4.5] decane (4.73 g, 13.76 mmol) was dissolved in dioxane (50 mL) and water (30 mL).

Hydrochloric acid (37%, 14.3 mL, 174 mmol) was added, and the reaction was heated to 85°C for 26 hours. After cooling to room temperature, the volatiles are removed in vacuo. The remaining aqueous phase is basified to pH ~9 with 2M NaOH, and then extracted with CH 2 C1 2 . The organic layers are dried over MgS0 4 and concentrated to afford the title compound (4.13 g, 100% yield). MS (ESI) m/e (M+H) + : 300.4

Intermed iate 34: ( S)- 1 -( 4' -Chloro-b iphenyl -2-y lmethyl)-2-methy 1-piper i din-4-one.

(5}-2-Methyl-piperidin-4-one hydrochloride (100 mg, 0.67 mmol) was combined with 2CO3 (554 mg, 4.0 mmol) in acetonitrile (5.0 mL) and stirred for 15 minutes. 4'- Chloro-2-(iodomethyl)biphenyl (220 mg, 0.668 mmol) was added, and the resulting mixture was heated to 70°C and stirred for 16 hours. The reaction was cooled to room temperature, filtered, and concentrated. The crude residue was purified by flash chromatography on silica gel (heptanes/ EtOAc gradient) to afford the title compound as an off-white solid (178 mg, 85% yield). MS (ESI) m/e (M+H + ): 313.8 Intermediate 35: I -(2-Bromo-benzv0-piperidin-4-one,

According to the procedure described for Intermediate 31 , alkylation of 4- piperidone hydrochloride (1.0 g, 4.0 mmol) with l-bromo-2-bromomethylbenzene (543 mg, 4.0 mmol) afforded the title compound (882 mg, 82% yield). HR-MS (m/z, MH+): measured 268.15

Intermediate 36: l-(4'-FIuoro-biphenyl-2-ylmethylVpiperidin-4-one

To a 5 mL microwave vial equipped with a stir bar was added sodium carbonate (79 mg, 0.75 mmol). The vial was dried in an oven for 16 hours, then cooled to room temperature under nitrogen. l-(2-Bromo-benzyl)-piperidin-4-one (100 mg, 0.37 mmol), 4-fluorophenyl boronic acid (52.2 mg, 0.37 mmol) and Pd(PPh 3 ) 4 (43 mg, 0.037 mmol) were added, followed by dioxane (5 mL) water (1 mL). The resulting mixture was degassed thoroughly with nitrogen and then heated in a microwave to 100°C for 10 minutes. The reaction was filtered and the filtrate was concentrated. The crude residue was purified via flash chromatography (heptanes/EtOAc gradient) to afford the desired product as a yellow solid (58 mg, 40% yield). MS (ESI) m/e (M+H + ): 283.3.

Intermediate 37 : 1 -[2-(4-Chloro-phenyl)-5.5-dimethyl-cyclohex-l -enylmethyl]- piperidin-

STEP A: A suspension of 2-bromo-5,5-dimethylcyclohex- 1 -enecarbaldehyde

(250 mg, 1.15 mmol), 4-chlorophenyl boronic acid (270 mg, 1.727 mmol) and Pd(Ph 3 P) 4 (66.5 mg, 0.058 mmol) in dioxane (2.5 mL) was stirred at room temperature in a 5 mL microwave vial under an atmosphere of nitrogen. A solution of potassium carbonate (318 mg, 2.3 mmol) in water (0.3 mL) was added and the solution became clear. The vial was capped and heated to 100 °C for 12 minutes in a microwave. The reaction was cooled to room temperature, diluted with CH2CI2, filtered through a plug of Celite, and

concentrated. The crude residue was purified by flash chromatography on silica gel (0- 25% EtOAc/ heptanes) to afford 2-(4-chlorophenyl)-5,5-dimethyIcyclohex-l - enecarbaldehyde as a clear colorless oil (220mg, 77% yield).

STEP B: A suspension of the hydrochloride salt of 4-oxopiperidine (273 mg, 2.01 mmol) and D1PEA (351 μί, 2.01 mmol) was stirred in DCE (9.0 mL) at room temperature for 10 minutes. 2-(4-Chlorophenyl)-5,5-dimethylcyclohex- l- enecarbaldehyde (500 mg, 2.01 mmol) was then added, followed by NaBH(OAc) 3 (426 mg, 2.01 mmol) and activated molecular sieves (~1 g). The resulting suspension was stirred at room temperature for 16 hours. The reaction mixture was then filtered through a plug of Celite, rinsing with EtOAc. The filtrate was washed sequentially with saturated aqueous NaHC0 3 , water, and brine. The organic layer was dried over MgS0 4 and concentrated. The crude residue was purified by flash chromatography on silica gel to afford the title compound (262 mg, 40% yield). MS (ESI) m/e (M+H + ): 33 1.88

Intermediate 38: 1 -(^'-Chlorobiphenyl^-vDmethvD-S-fluoropiperidin^-one

STEP A: 4-Oxo-piperidine- l -carboxylic acid /er/-butyl ester (10-Og, 50.2 mmol) was dissolved in DMF (20 mL). Chlorotrimethylsilane (6.6 g, 60.6 mmol) was added, followed by Et 3 N ( 12.3 g, 122 mmol). The mixture was stirred at 80°C for 16 hours, cooled to ambient temperature, diluted with Et 2 0, and then washed with water, followed by brine. The organic layer was separated, dried over Na 2 S0 4 , filtered and concentrated. Purification by flash chromatography on silica gel (EtOAc/ heptanes 0-50%) afforded 4- trimethylsilanyloxy-3,6-dihydro-2H-pyridine-l-carboxylic acid ieri-butyl ester (4.3g, 32% yield). 1H NMR (400 MHz, CHLOROFORM-ύΤ) δ ppm 4.80 (br. s., 1H), 3.88 (br. s., 2H), 3.53 (t, J=5.81 Hz, 2H), 2.1 1 (br. s., 2H), 1.49 (s, 9H), 0.20 (s, 9H).

STEP B: 4-Trimethylsilanyloxy-3,6-dihydro-2H-pyridine-l-carboxylic acid ten¬s ' butyl ester (0.60 g, 2.2 mmol) was dissolved in MeCN and Selectfluor (0.86 g, 2.4 mmol) is added. The reaction was stirred at ambient temperature for 75 minutes, and then poured into EtOAc (100 mL). The organics were washed with dilute brine and saturated brine, then dried (Na 2 S0 4 ), filtered and concentrated. Purification by flash

chromatography on silica gel (0-50% EtOAc/Hept) afforded 3-fluoro-4-oxo-piperidine-l-0 carboxylic acid tert-butyl ester (0.40 g, 83% yield). 1H NMR (400 MHz,

CHLOROFORM-*/) δ ppm 4.64 - 4.91 (m, 1H), 4.39 (d, 1H), 4.1 1 (td, J=6.57, 3.03 Hz, 1H), 3.10 - 3.24 (m, 2H), 2.40 - 2.61 (m, 2H), 1.43 (s, 9H).

STEP C: 3-Fluoro-4-oxo-piperidine-l -carboxylic acid tert-butyl ester (2.7 g, 12.4 mmol) was dissolved in dioxane/HCl (100 mL, 0.12 M) and stirred at ambient

5 temperature for 2 hours. Volatiles were then removed in vacuo to afford 3-fluoro- piperidin-4-one as an HC1 salt, which was carried on without further purification (1.7 g, 92% yield). 1H NMR (400 MHz, DMSO-i/6) δ ppm 9.77 (br. s., lH), 8.77 (br. s., 1H), 6.36 (br. s., 2H), 4.41-4.53 (d, J=48 Hz, 1H), 3.37 (t, j=9.85 Hz, 1H), 3.21 (m, 1H), 3.08 (m, 1H), 2.93 (m, 1H), 1.94 (m, 1H), 1.77 (m, 1H).

0 STEP D: 3-Fluoro-piperidin-4-one (0.32 g, 2.7 mmol) was dissolved in DMF (13 mL) and 4'-chloro-2-(iodomethyl)biphenyl (1.03 g, 3.13 mmol) was added followed by DIPEA (1.06 g, 8.2 mmol). The reaction was stirred at ambient temperature for 16 hours, and then poured into EtOAc (100 mL). The organics were washed with water followed by brine, dried (Na 2 S0 4 ), filtered and concentrated. Purification by flash chromatography5 on silica gel afforded the title compound (0.75 g, 86% yield). Ή NMR (400 MHz,

CDC1 3 ) 6 ppm 7.47 - 7.59 (m, 1H), 7.21 - 7.47 (m, 7H), 4.72 - 5.00 (m, 1H), 3.62 - 3.65 (d, J = 12 Hz, 1H), 3.56 - 3.59 (d, J = 12 Hz, 1H), 3.25 - 3.42 (m, 1H), 2.88 - 3.06 (m, 1H), 2.28 - 2.63 (m, 4H). 0 Intermediate 39: 1 -[ 1 -(4'-Chloro-biphenyl-2-yl)-ethyl1-piperidin-4-one

Step A: Same experimental as intermediate 31 substituting l -(2-Bromo-phenyl)- ethanone to afford l-(4'-Chloro-biphenyl-2-yl)-ethanone ( 1.31 g, 74 %). Ή NMR (400 MHz, CDC1 3 ) δ 7.57 (dd, 1 H, J = 7.6, 1.0 Hz ), 7.54 (m, 1 H), 7.46 (m, 1 H), 7.43 (2H, d, J = 8.6 Hz), 7.39 (1 H, dd, J = 7.6, 1 .0 Hz), 7.30 (2H, d, J = 8.6 Hz), 2.1 1 (3H, s).

Step B: To a mixture of l-(4'-Chloro-biphenyl-2-yl)-ethanone (2.42g, 10.5 mmol) and 4-piperidone ethylene ketal (1.50g, 10.5mmol) and titanium (IV) isopropoxide (5.96 g, 21 .0 mmol) is heated up to 75°C for 3 hours. The reaction is then cooled to room temperature and is added 50 mL of ethanol followed by sodium borohydride (1.19 g, 31.5 mmol) the reaction is then stirred for 18 hours at room temperature and then quench with 50 mL of methanol. The crude reaction is then dissolved in ethyl acetate and then washed with a saturated solution of sodium bicarbonate. The aqueous layer is then extracted 2 times with ethyl acetate. The combined organic layer are then dried on MgS0 4 and the solvent is removed under vacuum. The residue is chromatographed on silica gel gel (gradient: heptane/EtOAc; 0 to 60% EtOAc over 15 minutes) to afford 8-[ 1 -(4'-Chloro- biphenyl-2-yl)-ethyl]- l ,4-dioxa-8-aza-spiro[4.5]decane (3.6 g, 96 %). Found m/z ES+ = 358.5 (M+H); Ή NMR (400 MHz, CD 2 C1 2 ) δ 7.64 (1 H, d, J = 8. 1 Hz), 7.41 (2H, d, J = 8.6 Hz), 7.40 ( 1 H, t), 7.30 (2H, J = 8.6 Hz), 7.29 ( 1 H, t), 7.18 ( 1 H, d, J = 7.6 Hz), 3.90 (4H, s), 3.51 ( 1 H, m), 2.48 (2H, m), 2.36 (2H, m), 1.62 (4H, m), 1.27 (3H, d, J =6.6 Hz).

Step C: To a solution of 8-[l -(4'-Chloro-biphenyl-2-yl)-ethyl]-l ,4-dioxa-8-aza- spiro[4.5]decane (3.1 g, 8.66 mmol) in dioxane-water ( 1 : 1 60 mL) is added HC1 37% (10.5 mL, 346 mmol). The reaction is then heated up to 85°C for 26 hours. The solvent is then removed under vacuum and the acqueous phase is basified to pH=9 using sodium hydroxide 4N and then extracted with DCM. The organic layer is then washed with brine. The combined organic phase is then dried on MgS0 4 and concentrated to afford l -[l -(4'- Chloro-biphenyl-2-yl)-ethyl]-piperidin-4-one. The crude material is used without further purification. Found m/z ES+ = 314.2 (M+H); Ή NMR (400 MHz, CDC1 3 ) 6 7.57 (1H, d, J = 7.1 Hz), 7.32 (1H, t), 7.30 (2H, d, J = 8.6 Hz), 7.22 (1H, t), 7.15 (2H, d, J - 8.6 Hz), 7.09 (1H, d, J = 6.1 Hz), 3.58 (1H, m), 2.63 (2H, m), 2.52 (2H, m), 2.25 (4H, t, J = 6.1 Hz), 1.24 (3H, d, J = 6.6 Hz).

Intermediate 40: 4-Benzylidenecyclohexanone

To a solution of 8-benzylidene-l,4-dioxaspiro[4.5]decane (5.79 g, 25.1 mmol) in

THF (126 mL) a solution of H 2 S0 4 (10% in water, 67 mL) was added and the result mixture was stirred at room temperature for 3 days. THF was removed under reduce pressure and water was added. The aqueous phase was extracted with DCM and the organic layers were combined, washed with brine, dried with Na 2 S0 4 , filtered and concentrated to give 4-Benzylidenecyclohexanone (4.35 g 84%) as a yellowish oil: Ή NMR (400 MHz, CHLOROFORM-*/) 6 ppm 7.23-7.33 (m, 2 H), 7.13-7.20 (m, 3 H), 6.42 (s, 1 H), 2.67 - 2.77 (m, 2 H), 2.62 (t, J=7.0 Hz, 2 H), 2.46 (t, J=7.0 Hz, 2 H), 2.37 (t, J=7.0 Hz, 2 H). Intermediate 41 : 4-((4'-Chlorobiphenyl-2-yl)methylene1cvclohexanone

Step A: To a solution of 4'-chloro-2-(iodomethyl)biphenyl in toluene (11 mL), triphenylphosphine (1.20 g, 4.57 mmol) was added. The result mixture was refluxed overnight, allowed to cool to room temperature and filtered. The solid was washed with ether and dried under vacuum to afford ((4'-Chlorobiphenyl-2- yl)methyl)triphenylphosphonium iodide (2.50 g, 93%): Ή NMR (400 MHz, chloroform- d) δ ppm 6.35 - 8.06 (m, 23 H), 5.12 - 5.63 (m, 2 H).

Step B: A suspension of sodium hydride (60% in mineral oil, 77 mg, 1.92 mmol) in DMSO (5.8 mL) was heated for 1 hour at 80°C and then cooled to rt. ((4'- Chlorobiphenyl-2-yl)methyl)triphenylphosphonium iodide (1.04 g, 1.76 mmol) was added to the suspension and then, after stirring 10 minutes at room temperature, 1 ,4- cyclohexanedione monoethylene acetal (0.25 g, 1.60 mmol) was added. The reaction mixture was heated at 80°C for 3 hours. After standing at rt overnight, the reaction mixture was poured into a saturated solution of NaHS0 4 and extracted with EtOAc. The organic layers were combined, washed with brine, dried with Na 2 S0 , filtered and concentrated. The crude was purified by flash chromatography (silica gel, 0% to 30% EtOAc in heptane) to give ((4'-Chlorobiphenyl-2-yl)methylene)-l ,4- dioxaspiro[4.5]decane (0.28 g, 52%) as a white solid: Ή NMR (400 MHz,

CHLOROFORM-i/) δ ppm 7.12 - 7.33 (m, 8 H), 5.98 (s, 1 H), 3.92 (s, 4 H), 2.30 - 2.36 (m, 2 H), 2.22 - 2.29 (m, 2 H), 1.62 - 1.71 (m, 2 H), 1.52 - 1.58 (m, 2 H).

Step C: To a solution of the ketal ((4'-Chlorobiphenyl-2-yl)methylene)- 1 ,4 dioxaspiro[4.5]decane (180 mg, 0.53 mmol) in a mixture of dioxane (3 mL) and water (2.3 mL) a solution of aqueous HC1 (12 M, 0.80 ml, 9.6 mmol) was added. The result mixture was heated to 80°C and stirred overnight. THF was removed under reduce pressure and water was added. The aqueous phase was extracted with EtOAc and the organic layers were combined, washed with brine, dried with Na 2 S0 , filtered and concentrated to afford 4-((4'-Chlorobiphenyl-2-yl)methylene)cyclohexanone (157 mg, 100%) of the pure ketone as a colorless oil: Ή NMR (400 MHz, CHLOROFORM-i/) δ ppm 7.16 - 7.32 (m, 8 H), 6.21 (s, 1 H), 2.43 - 2.51 (m, 4 H), 2.35 (t, J=7.0 Hz, 2 H), 2.17 (t, J=7.0 Hz, 2 H).

EXAMPLES 1-99 Synthesis of Examples 1-19 by reductive ani ation of ketones IX with amines V.

General Reductive Amination Procedure 1.

A solution of amine V (1 eq), ketone IX (1 eq), acetic acid (0.2 eq), and powdered molecular sieves (approximately 330 mg mmol V) in 1: 1 CH 2 C1 2 : methanol (6 mL) was stirred at 0°C for 1 hour. Sodium cyanoborohydride (2 eq) was added, and the reaction was allowed to warm to room temperature and stir for 16 hours. The reaction was filtered, and the filtrate was concentrated in vacuo. The crude residue was purified by flash chromatography on silica gel (0-100% methanol in CH 2 C1 2 ).

Example 1

Preparation of N- ί 7- Γ 1 -( 4'-Chloro-biphenyl-2-yl methyll-piper idin-4- yll-5.6.7.8- tetrahydro-pyridor3.4-d1pyrimidin-4-yl|-4-((j-)-3-dimethylam ino-l- phenylsulfanylmethyl-propylamino)-3-nitro-benzenesulfonamide .

Following General Reductive Amination Procedure 1, 4-((ii)-3-dimethylamino-l- phenylsulfanylmethyl-propylamino)-3-nitro-N-(5,6,7,8-tetrahy dro-pyridot3,4- d]pyrimidin-4-yl)-benzenesulfonamide (85 mg, 0.15 mmol) and 1 -(4'-chloro-biphenyl-2- ylmethyl)-piperidin-4-one (45.7 mg, 0.15 mmol) afforded the title compound as a yellow solid (40 mg, 31% yield). 'HNMR (400 MHz, MeOD) δ (ppm): 8.66 (d,J=2.01 Hz, 1H), 8.14 (s, 1H), 7.86 (dd,J= 9.03, 2.01 Hz, 1H),7.48 (d,J=7.03 Hz, 1H), 7.17-7.41 (m, 9H), 7.06-7.10 (m, 3H), 6.80 (d,J= 9.03 Hz, 1H), 3.96-4.06 (m, 1H), 3.51 (s, 2H) , 3.38 (s, 2H), 3.25- 3.30 (m, 1H), 3.11-3.17 (dd,J= 4.0, 16.0 Hz, 1H), 2.76-2.85 (m, 4H), 2.62-2.68 (m, 2H), 2.30-2.45 (m, 3H), 2.20 (s, 6H), 1.98-2.10 (m, 1H), 1.78-1.91 (m, 5H), 1.45-1.55 (m, 2H).

HR-MS (m/z, MH + ): 841.50

HPLC retention time = 3.13 minutes (Agilent 1100 HPLC system; Inertsil ODS3 100 x 3mm C18 column; flow rate of 1.0 mL/minute; gradient: 5-95% acetonitrile/water with 0.1% FA over 7.75 minutes).

Examples 2-7 were prepared by reductive amination of ketones IX with amines V following General Procedure 1 above. Example 2

(R)-N-(7-fl-((2-(4-chloropheml)-5,5-dimethylcyclohex-l-eml)m ethy

5,6, 7,8-tetrahvdropyridof3A-d]pyrimidin-4-yl)-4-(4-(dimethylamin o)-l- fphenylthio)buian-2-ylamino)-3- trobertzertesulfort mide:

'HNMR(400MHz, MeOD) δ (ppm): 8.55 (d, J =2.01 Hz, 1H), 8.04 (s, 1H),

7.77 (dd,J=2.29, 9.29 Hz, 1H), 7.24 (d,J= 8.53 Hz, 2H), 7.12-7.16 (m, 2H), 6.96-7.00 (m, 5 H), 6.72 (d,J= 9.54 Hz, 1H), 3.91-4.00 (m, 1H), 3.44 (s, 2H), 3.23-3.26 (m, 1H), 3.09 (dd,J= 5.5, 14.5 Hz, 1H), 2.93-3.05 (m, 4H), 2.66 -2.77 (m, 4H), 2.53-2.58 (m, 2H), 2.37-2.47 (m, 7H), 2.17-2.26 (m, 2H), 1.77-2.11 (m, 8H), 1.47-1.62 (m, 2H), 1.41 (t, J= 6.27 Hz, 2H), 0.92 (s, 6H); HR-MS (m/z, MH+): 873.57.

Example 3

N-(7-(QS)-l-((4'-chlorobipheml-2-yl)methyl)-2-methylpiper id^^

tetrahvdropyridof3A-d1pyrimidin-4-yl)-4-((R)-4-(dimethyla m

ylamino)~3-nitrobenzenesulfonamide:

Isolated as a 2: 1 mixture of diastereomers. Ή NMR of major diastereomer (400 MHz, MeOD) δ (ppm): 8.64 (d, J= 2.01 Hz, 1H), 8.12 (s, 1 H), 7.83-7.87 (m, 1H), 7.52 (d, J= 7.53 Hz, 1H), 7.22-7.42 (m, 9H), 7.07-7.11 (m, 3H), 6.79 (d, J= 9.03 Hz, 1H), 4.08 (d, J= 13.05 Hz, 1H), 3.98-4.08 (m, 1H), 3.50 (s, 2H), 3.28 (m, 1H), 3.13-3.18 (dd, J= 5.5, 16.0 Hz, 1H), 2.96 (d, J= 13.05 Hz, 1H), 2.74-2.81 (m, 3H), 2.63-2.68 (m, 2H), 2.33-2.50 (m, 3 H), 2.21 (s, 6H), 2.00-2.15 (m, 2H), 1.81-1.92 (m, 2H), 1.68-1.77 (m, 2H), 1.25-1.43 (m, 2H), 1.01 (d, J = 6.02 Hz, 3H); HR-MS (m/z, MH+): measured 855.51.

Example 4

N-(7-((2S)-l-((4'-chlorobiphenyl-2-vttmethyl)-2-methylDi^

tetrahvdropyridof 3,4-d ]pyrimidin-4-yl) ~4-((R)-4-( dimethylam mo)-l- (phenylthio)b utan-2- ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide.

Isolated as a 2.5: 1 mixture of diastereomers. Ή NMR of the major diastereomer (400 MHz, MeOD) 5 (ppm): 8.34 (d, J= 2.01 Hz, IH), 8.1 1-8.25 (s, I H), 8.0 (d, J- 9.54 Hz, I H), 7.66-7.70 (m, I H), 7.30-7.58 (m, 9H), 7.12-7.25 (m, 3H), 6.88 (d, J= 9.54 Hz, I H), 4.60-4.79 (m, I H), 3.90-4.12 (m, 2H), 3.58-3.70 (m, 2H), 3.42-3.57 (m, I H), 3.04- 3.27 (m, 4H), 2.68-2.99 (m, 10H), 2.44-2.64 (m, 3H), 2.01-2.32 (m, 3H), 1.55-2.00 (m, 3H), 1.38 (d, J= 6.02 Hz, 3H); HR-MS (m/z, MH+): 942.57.

Example 5

(R)-4-(4-(dimet ylammo)-i-(phenylthio)buta^

yl)methyl)piperidin-4-yl)-5, 6, 7,8-tetrahydropyrido[3, 4-d]pyri ' midin-4-yl)-3- (trifluoromethylsulfonyl)benzenesulfonamide.

Ή NMR (400 MHz, MeOD) δ ppm: 8.32 (d, J= 12.05 Hz, 2H), 8.01 (d, J= 8.53 Hz, I H), 7.76 (s, I H), 7.49-7.59 (m, 2H), 7.1 1 -7.45 (m, 9H), 6.93 (t, J= 8.03 Hz, 2H), 4.36 (s, 2H), 4.04-4.10 (m, I H), 3.66-3.86 (m, 2H), 3.29-3.42 (m, 2H), 3.08-3.24 (m, 4H), 2.69-3.04 (m, 5H), 2.85 (s, 6H), 2.54-2.66 (m, 2H), 2.19-2.31 (m, IH), 1.99-2.16 (m, 3H), 1.73-1.87 (m, 2H); HR-MS (m/z, MH+): 912.09.

Example 6

(R)-N-(7~(l-((4 '-bromobiohenyl-2-yl)methyl)piperidin-4-yl)'5.6.7.8- tetrahvdropyrido[3.4-dlwrimidin-4-yl)-4-(4-(dimethylamiw

ylamim)-3-(trijluoromethylsulfonyl)benzenesulfonamide.

! H NMR (600 MHz, Acetone-i/6) 6 ppm: 8.37 (s, IH), 8.30 (s, IH), 7.94-7.99 (m, IH), 7.61 (d, J= 8.31 Hz, 2H), 7.51 (d, J= 7.18 Hz, IH), 7.41-7.44 (m, 4H), 7.32-7.38 (m, 2H), 7.30 (t, J= 7.55 Hz, 2H), 7.21-7.26 (m, 2H), 6.87-6.90 (m, IH), 4.10-4.16 (m, IH), 3.59 (s, 2H), 3.36 (s, 2H), 3.26-3.35 (m, 2H), 3.04-3.08 (m, IH), 2.84 (d, J= 1 1.71 Hz, 2H), 2.78 (t, J= 5.67 Hz, 2H), 2.55-2.61 (m, IH), 2.53 (t, J= 5.29 Hz, 2H), 2.40-2.46 (m, IH), 2.31-2.37 (m, IH), 2.18-2.25 (s, 6H), 1.86-1.97 (m, 3H), 1.78 (d, J= 1 1.33 Hz, 2H), 1.49-1.58 (m, 2H); HR-MS (m/z, MH+): 973.01.

Example 7

(R)-N-(7-(l-(2-bromoben∑yl)piperidin-4-yl)-5 .8-tetrg}wdro

yl)-4-(4-(dimethylamino)-l-(phenylthio)butan-2-ylammo)-3- (trifluoromethylsulfonyl)benzenesulfonamide.

'H NMR (400 MHZ, MeOD) MeOD) δ (ppm): 8.34 (d, J = 1.51 Hz, 1H), 8.12 (s, 1 H), 7.95 (dd, J= 2.01 , 9.03 Hz, 1 H), 7.57 (d, J= 7.53 Hz, 1H), 7.50 (d, J = 7.53 Hz, 1H), 7.31 -7.36 (m, 3H), 7.15-7.24 (m, 4H), 6.70 (d, J= 9.03 Hz, 1 H), 3.87- 3.96 (m, 1 H), 3.68 (s, 2H), 3.62 (s, 2H), 3.1 1 -3.26 (m, 3H), 3.05 (m, 2H), 2.91 (t, J = 5.77 Hz, 2H), 2.54-2.79 (m, 5H), 2.46 (s, 6H), 1.82-2.32 (m, 5H), 1.62 - 1.74 (m, 2H); HR-MS (m/z, MH+): 896.90.

General Reductive Animation Procedure 2.

A solution of amine V (1 eq), ketone IX (1 eq), acetic acid (1 eq), sodium triacetoxyborohydride ( 1 .5 eq) and molecular sieves (-400 mg/ mmol V) in DCE (12.4 mL/ mmol V) was stirred at 45°C for 12 hours. The mixture was cooled to room temperature, filtered through Celite, and concentrated. The crude residue was purified by flash chromatography on silica gel (0-100% methanol in CH 2 C1 2 ).

Example 8

N-{7-[2-(4-Chloro-phenyl)-5,5-dimethyl-cyclohex-l-enylmethyl ]-5, 6, 7,8-tetrahydro- pyridof 3, 4-d]pyrimidin-4-yl}-4-((R)-3-morpholin-4-yl- 1 -phenylsulfanylmethyl- propylamim)-3-trifluoromethanesulfonylbenzenesulfommide.

Following General Reductive Amination Procedure 2 above, (Λ)-4-(4- morphol ino- 1 -(pheny lth io)butan-2-y lamino)-N-(5 ,6,7, 8-tetrahydropy rido [3,4- d]pyrimidin-4-yl)-3-(trifluoromethylsulfonyl)benzenesulfonam ide (83 mg, 0.121 mmol) and l-[2-(4-chloro-phenyl)-5,5-dimethyl-cycIohex-l-enyImethyl]-p iperidin-4-one (40 mg, 0.121 mmol), afford the title compound (1 1.4 mg, 9% yield).

Ή NMR (400 MHz, MeOD) δ ppm: 8.34 (bs, 1H), 8.25 (bs, 1H), 7.97 (d, J= 8.4 Hz, 1H), 7.37 (m, 4H), 7.09-7.27 (m, 5H), 6.79-6.89 (d, J = 8.4 Hz, 1H), 3.96-4.19 (m, 1H), 3.1 1-3.84 (m, 9H), 2.49-2.96 (m, 10H), 2.38 (bs, 2H), 1.62-2.22 (m, 8H), 1.55 (bs, 2H.), 1.29 (bs, 4H), 1.05 (bs, 6H), 0.9 (s, 2H)

TOF MS ES+ (M+H ): 1002.47

HPLC retention time: 1.84 minutes (Agilent 1100 HPLC system; Inertsil ODS3 100 x 3mm C18 column; flow rate of 1.0 mL/minute; gradient of 5-95%

acetonitrile/ water with 0.1% FA).

Examples 9-64 were prepared by reductive amination of ketones IX with

V following General Procedure 2 above.

Example 9

(R)'N-(7-(l-((4' hlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5

tetrahvdropyridof3 -d]pyrimidin-4-yl)-4-(4-morpholino-l-(phe

ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide

'H NMR (400 MHz, MeOD) 5 ppm: 8.47 (d, J= 2.1 Hz, 1 H), 8.31 (s,l H), 7.95 (d, J = 8.46 Hz, 1 H), 7.10 - 7.50 (m, 13 H), 6.90 (d, J= 9.35 Hz, 1 H), 4.30-4.15 (m, 1 H), 3.86-3.92 (m, 2H), 3.83-3.86 (m, 2H), 3.69-3.80 (m, 4H), 3.23-3.42 (m, 2 H), 3.1 1-3.21 (m, 4H) 2.80-2.87 (m, 2H), 2.04-2.63 (m, 13H), 1.67- 1.96 (m, 2 H); TOF MS ES+ (M+H + ): 970.29.

Example 10

(R)-N-(7-(l-((2-(4-chlorophem>l)-5, 5-dimethylcvclohex-l-enyl)methyl)piperidin-4-yl)- 5, 6, 7, 8'tetrahydropyridof 3, 4-d]pyrimidin-4-yl) -4-(4-(dimethylam ino)-l - (phenylthio)butan-2-ylamino)-3-(trifluoromethylsulf^

'H NMR (400 MHz, MeOD) δ ppm: 8.30 (d, J= 2.1 Hz, 1 H), 8.1 1 (s,l H), 7.97 2.1, 8.1 Hz, 1 H), 7.39-7.30 (m, 4H), 7.25-7.13 (m, 3H), 7.09 (d, J= 8.2 Hz, 2H), 6.70 (d, J= 8.0 Hz, 1H), 3.87-3.98 (m, 1H), 3.54 (s, 2 H), 3.23 (dd, J= 4.8, 14.2 Hz, 1H), 3.06-3.18 (m, 5H) 2.78-2.88 (m, 2H), 2.50-2.75 (m, 5H) 2.45 (s, 6H), 2.26-2.37 ( m, 2H), 2.05-2.24 (m, 3H), 2.01 (bs, 2H), 1.82-1.98 (m, 3H), 1.59-1.75 (m,2H), 1.50 (t, J = 7.6 Hz, 2H), 1.02 (s, 6H); TOF MS ES+ (M+tT): 960.34.

Example 11

(R)-N-n-(l-((4'-chlorobiphenyl-2-\l)meth\l)piwrid^

tetrahvdropyrido[3 -dlmrimidin-4-yl)-4-(4-(dimethylamm^

Ή NMR (400 MHz, MeOD) δ ppm: 8.31 (d, J= 2.05 Ηζ,ΙΗ), 8.10 (s, 1H), 7.90- 7.95 (m, 1H), 7.10-7.52 (m, 13H), 6.65 (d, J- 8.2 Ηζ,ΙΗ), 3.81-3.96 (m s lH), 3.52 (s, 2H), 3.40 (s, 2H), 3.20 (dd, J= 5.1, 16.0 Hz, 1H), 3.1 1 (dd, J= 5.1, 16.0 Hz, 1H), 2.75- 2.89 (m, 4H), 2.68-2.79 (m, 2H), 2.26-2.45 (m, 3H), 2.19 (s, 6H,), 1.96-2.08 (m, 1H), 1.79-1.88 (m, 4H), 1.68-1.79 (m, 1H), 1.44-1.59 (m, 2H); TOF MS ES+ (M+H + ): 928.28.

Example 12

(R)-N-n-(l-((4'-chlorobxyhenyl-2-yl)metM)piperidM

tetmhvdropyridof3A-dlpyrimidin-4-yl)-4-(4-(dimethylamino) -l-(phem^

ylamino)-3,5-difluorobenzenesulfonamide

1H NMR (400 MHz, chloroform-i ) δ ppm 1.38 - 1.55 (m, 2 H) 1.62 - 2.00 (m, 7 H) 2.20 (s, 6 H) 2.25 - 2.56 (m, 5 H) 2.67 (t, J=5.56 Hz, 2 H) 2.77 (d, J=l 1.12 Hz, 2 H) 2.98 (dd, J=l 3.39, 7.33 Hz, 1 H) 3.13 (dd, J= 13.14, 4.55 Hz, 1 H) 3.26 (s, 2 H) 3.46 - 3.61 (m, 2 H) 4.01 (br. s., 1 H) 5.47 (br. s., 1 H) 7.01 - 7.50 (m, 15 H) 8.05 (s, 1 H) TOF MS ES+ (M+H + ): 832.31 Retention time = 3.09 minutes.

Example 13

N-(7-n- < / 2-{4-chlorophenyl)-5 -dimethylcvclohex-l-enyl)methyl)piperidin-4-y!)-5, 6, 7,8- tetrahydmpyndo[3, 4'd]pyrimidin-4-yl)-4-(l^henylthio)butan-2-ylamirio)-3- (trifluoromeihylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H ): 885.24; Retention time = 4.86 minutes. Example 14

N-(7~(l-((2-( 4-chlorophe nyl )-4, 4-dimethylcyclohex- 1 -enyl)methyl)piperid in- 4-yl) -5,6, 7,8- tetrahvdropyridof3, 4-d]pynmidin-4-yl)-4-(l-(phenylthio)butan-2-ylamino)-3-

(triftuoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H*): 917.29; Retention time = 5.19 minutes

Example 15

N-(4-(N-(7-(l-((4 '-chIoro-4-fluorobiphenyl-2-yl)methyl)piperidin-4-yl)-5.6. 7, 8- tetrahvdropyrido[3A-dltM"imidin-4-yl)sulfamoyl)-2-(trifl^

(2-(phenylthio)ethyl)acetamide

TOF MS ES+ (M+FT): 917.20; HPLC Retention time = 4.55 minutes.

Example 16

(R)-N-(7-(l-((2-(4-chloropheml)-4 -dimethylcyclohex-l-eriyl)methyl}pipe

5.6. 7.8-tetrahvdropyrido[3, 4-d Jpyrim idin-4-yl ) - 4-1 ' 4-morpholm ' o-l ' - (phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)bemenesulfonamide

TOF MS ES+ (M+H + ): 1002.35; HPLC Retention time = 3.75 minutes.

Example 17

(R)-N-(7-(l-((2-(4-chloropheml)-4A-dimethylcyclohex-l-enyl}m etM

5.6, 7,8-tetrahydropyrido[3, 4-dlpyrimidin-4-yl)-4-(4'(dimethylamino)-l-

TOF MS ES+ (M+H + ): 960.35; HPLC retention time = 3.68 minutes.

Example 18

N-(7-(l-((2-(4-chlorophenyl)~4A-dimethylcyclohex-l-enyl)meth yl)pipe tetrahydropyrido[3, 4-d]pyrimidin-4-yl)-4-( l-(phenylthio)pentan-2-ylamino)-3- (trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H + ): 931.31 ; HPLC Retention time = 5.33 minutes.

Example 19

(R)-N-(7-(l-((4'-chloro-5-nuorobipheml-2-yl)metM)pip^^ 6. 7.8- tetrahydropwido[3A-d]pyrimidinA-yl)-4-(4-(dimethylamim

ylamino)-3-(tri oromethylsulfonyl)benzemsulfonamide

Example 20

(R)-N-(7-(l-((4'-chloro^-fluorobipheml-2-yl)methyl)piperid^ 6, 7, 8- tetrahvdropyrido f 3 , 4-dlpyrimidin-4-yl) -4-(4-(dimethylamino) - 1 -(phenylthio)butan-2- ylamino)-3-(trifluoromethylsulfonyl)ben∑enesulfonamide

TOF MS ES+ (M+H + ): 946.26; HPLC Retention time = 3.46 minutes.

Example 21

(R) -N- (7 -(!-(( 4 '-chloro-3 -jluorob iphenyl-2~yl)methyl)piperidin- 4-yl)-5, 6, 7,8- tetrahydropyrido[3,4-d]pyrimidin-4-yl)-4-(4-(dimethylamim)-l -(pheny

ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H + ): 946.26; HPLC Retention time = 3.37 minutes.

Example 22

(R)-3-(4-(N-a-(l-((4'-chlorobiphenyl-2-yl)methyl)piperid^

tetrahvdropyrido f 3 , 4-dJpyrimidin- 4-yl)sulfamoyl) -2-nitrophenylam ino)-N, N-dimethyl-4- (phenylthio)butanamide

TOF MS ES+ (M+H 4 : 855.29; HPLC Retention time - 4.03 minutes.

Example 23

N-{7-[l-(4 '-Chloro-biphenyl-2-ylmethyl) -viveridin-4-yl 1-5.6.7, 8-tetrahvdro-pyridoF3.4- d]pyrimid\n-4-\l)-4l(R)-3-(4-ethyl-piperazin-l-yl)-l-phem^

propylamino -3'trifluoromethanesulfonyl-benzenesulfonamide

TOF MS ES+ (M+H 4 ): 997.33; HPLC Retention time = 3.46 minutes.

Example 24

N-(7-(l-(l-(4'-chlorobiphenyl-2-yl)ethyl)piperidin-4 )-5,6, 7,8-tetrah^

d1pyrimidin-4-yl)-4-( (R)-4- morpholino- 1 - (phenylthio)b utan-2-ylamino)-3- ( trifluoromethylsulfonyl) benzene sidfonam ide

TOF MS ES+ (M+H + ): 1016.36; HPLC retention time = 3.69 minutes.

Example 25

4-((R)-l-Benzyloxymethyl-3-dimethylamino-propylamino)-N-{7-f l-(4'-chloro-bip^ ylmethyl)-piperidin-4-yll-5, 6, 7 ,8-tetrahydro-pyrido[3, 4-d]pyrimidin-4-yl)-3- trifluoromethanesulfonyl-benzenesulfonamide

TOF MS ES+ (M+H*): 926.31 ; HPLC Retention time = 3.42 minutes.

Example 26

N-{7-fl-(4'~ Chloro-biphenyl- 2-ylmethyl)-piperidin- 4-yl 1-5,6.7.8-tetrahvdro-pyrido[3, 4- d]pyrim\din-4-yl}-4-f(R)-l-(3-chloro-phenylsulfarrylmeth^

propylamino ]- -trifl uoromethanesulfonyl- benzene sulfonamide

TOF MS ES+ (M+H : 962.24; HPLC Retention time = 3.47 minutes.

Example 27

N-(7-(l-((2-(4-chlorophenyl)-4, 4-dimethylcyclohex-l-enyl)methyl)-2- methylpiperidin-4-yl) -5.6, 7.8-tetrahvdropyridof 3, 4-dlpwimidin-4-yl)-4-{ (R)-4- morpholino-l-(phenylthio) butan-2-ylamino)-3- (trifluoromethylsulfonv benzenesulfonamide

TOF MS ES+ (M+H ): 1016.36; HPLC Retention time = 3.76 minutes.

Example 28

(R)~N-(7-(l-((2-(4-chlomphenyl)-4A-dimethylcvclobe

5, 6, 7, 8-tetrahvdropyridof3, 4-d]pyrimidin-4-yl)-4-{l-(2~chlorophenylthio)-4- (dimethylamim)butm-2-ylamxno)-3-(trifluorometM

1 H NMR (400 MHz, MeOD) δ ppm 1.00 (s, 6 H) 1.44 - 1.58 (m, 2 H) 1.58 - 1.75 (m, 2 H) 1.82 - 1.98 (m, 3 H) 2.01 - 2.20 (m, 5 H) 2.26 (br. s., 2 H) 2.43 (s, 6 H) 2.52 (t, J=l 1.12 Hz, 1 H) 2.57 - 2.74 (m, 4 H) 2.74 - 2.86 (m, 2 H) 3.02 - 3.25 (m, 5 H) 3.25 - 3.37 (m, 1 H) 3.54 (s, 2 H) 3.99 (dd, J=8.34, 4.80 Hz, 1 H) 6.78 (d, J=9.60 Hz, 1 H) 7.06 (d, J=8.59 Hz, 2 H) 7.09 - 7.18 (m, 2 H) 7.27 - 7.37 (m, 3 H) 7.37 - 7.49 (m, 1 H) 7.99 (dd, J=9.09, 2.02 Hz, 1 H) 8.13 (s, 1 H) 8.36 (d, J=2.02 Hz, 1 H) TOF MS ES+ (M+H + ): 994.29; HPLC Retention time = 3.69 minutes.

Example 29

N-(7-(l-(l-(4 '-chlorobiphenyl-2-yl)ethvI)piperidin- 4-yl) -5.6.7.8-tetrahvdropyridof 3, 4- dlOyrimidin-4-yl)-4-((R)-l-(2-chlorophem thio)-4-(dimethylammo

(trifluoromethylsulfonvDbenzenesulfonamide

1 H NMR (400 MHz, MeOD) 6 ppm 1.44 (d, J=6.57 Hz, 3 H) 1.47 - 1.57 (m, 1 H) 1.57 - 1.71 (m, 1 H) 1.77 - 2.21 (m, 6 H) 2.42 - 2.56 (m, 7 H) 2.58 - 2.86 (m, 6 H) 2.90 (d, J=10.1 1 Hz, 1 H) 3.13 - 3.27 (m, 2 H) 3.27 - 3.37 (m, 1 H) 3.56 (s, 2 H) 3.73 (q, J=6.57 Hz, 1 H) 3.94 - 4.06 (m, 1 H) 6.78 (d, J=9.60 Hz, 1 H) 7.07 - 7.17 (m, 2 H) 7.17 7.38 (m, 5 H) 7.38 - 7.50 (m, 4 H) 7.63 (d, J=6.57 Hz, 1 H) 7.99 (dd, J=9.09, 2.53 Hz, 1 H) 8.12 (s, 1 H) 8.36 (d, J-2.53 Hz, 1 H) TOF MS ES+ (M+H 4 ): 976.25; HPLC retention time - 3.49 minutes.

Example 30

(R)-N-n-a-((4'-chlorobipheml-2-yl)methyl)piperidin^^

tetra vdrop\ridoi3A~dlpwim\dm-4-yl)-4-(l-(phenylthio)-4-(pipe

yl mino)- -(trifl oromethylsulfonyl)benzemsulfonamide

TOF MS ES+ (M+H^): 968.31 ; HPLC Retention time = 3.48 minutes.

Example 31

N-(7-(l-(l-(4'-chlom-4-fluorobiphenyl-2-yttethyl)piperidi n-4^ 6.7.8- tetrahydropyrido f3 -d]pyrimidin-4-yl)-4-((R)-4-morpholino-l-(phenylthio)butan-2 - ylamim)-3-(trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H + ): 1002.29; HPLC Retention time = 4.26 minutes.

Example 32

(R)-N-(7-(l-((2-(4-chlorophewl)-4A-dimethylcvclohex-l-enyl)m eth^

(trifluoromethyD-S, 6, 7,8-tetrahvdrovyrido[3, 4-dlpyrimidin-4-yl)-4-(l-(2- chlorophenylthio)-4-(dimethylammo)butan-2-ylaminoi-3- (trifluoromethylsulfonyl)be enesulfonamide

TOF MS ES+ (M+H + ): 1062.28; HPLC Retention time = 4.09 minutes.

Example 33

(R) -N-(7- (l-((4 '-chlorobiphenyl-2-yl) methyl)piperidin-4'yl) ~2-(trifluoromethyl) -5, 6, 7, 8- tetrahydropyrido[3, 4-d Jpyrimidin- 4-yl )-4-( l-(2-chlorophenyl thio )-4- (dimethylammo)butan'2'Vlamino)-3-(trifluoromethylsulfonyl)be nzenesulfy

TOF MS ES+ (M+H 4 ): 1030.22; HPLC Retention time = 3.83 minutes.

Example 34

N-(7-(l-(l-(4'-chlorobiphenyl-2-yl)ethyl)piperidin-4-yl)-2-( M

tetrahydropyridof 3, 4-d]pyrimidin-4-yl)-4-( (R)-l- (2-chlorophenylthio)- 4- (dimethylam ino) butan-2-ylam ino)-3-(triiluoro methylsulfonyl) benzenesulfonamide

TOF MS ES+ (M+H 4 ): 1044.24; HPLC Retention time = 4.58 minutes.

Example 35

N-f7-(l-(l-(4'-chloro-4- uorobiphenyl-2-yl)ethyl)piperidin-4-yl)-5,6, 7,8- tetrahydropyri(fol3A-dlpyrimidin-4-yl)-4-((R)-4-(4-rmthy

(phenylthio)butc -2-ylamino)-3-(trifluoromethylsulfonyl)be

TOF MS ES+ (M+H + : 1033.32; HPLC Retention time = 4.15 minutes.

Example 36

(R)'N-(7-(l-( f 4 '-chloro-4-methoxybiphenyl-2-yl)methyl)piperidin-4-yl)-5.6, 7.8- tetrahvdropyrido[3, 4-d]pyrimidin- 4-yl) -4-(4-( dimethylam ino) -I- (phenylihio)butan-2- ylamino -3-(trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+Ff): 958.28; HPLC Retention time = 3.57 minutes.

Example 37

N-{7-[l-( ' 4'-Chloro-bipheml-2-ylmethyl)-piperidin~4-ylJ~5,6 ,8~te^

d]pyrimidin-4 l-4-{(R)-$-dimethylamino-l-(4-fluoro-pfa

propylamino]-3'trifluoromethanesulfoml-benzenesidfonamide

TOF MS ES+ (M+FT): 946.26; HPLC 3.39 Retention time = minutes.

Example 38

(R) -N- (7-(l-{(4 '-chlorobiphenyl-2-yl)methyl)piperidiri-4-yl)-5, 6. 7.8-tetrahvdropyrido f3.4-dJpyrimidm-4-v -4-fl- 2.6-dichlorophenylthio)-4-fdimethyl m o) butan-2~ ylamino -3-(trifluoromethylsulfonyl)bertzenes lfonamide

'H MR (400 MHz, MeOD) 6 ppm 1.49 - 1.65 (m, 2 H) 1.82 - 2.00 (m, 3 H) 2.05 (t, J=l 1.12 Hz, 2 H) 2. I I - 2.25 (m, 1 H) 2.46 - 2.60 (m, 7 H) 2.66 (t, J=4.80 Hz, 2 H) 2.68 - 2.84 (m, 2 H) 2.84 - 2.98 (m, 4 H) 3.09 - 3.20 (m, 1 H) 3.20 - 3.29 (m, 1 H) 3.57 (s, 2 H) 3.61 (s, 2 H) 3.81 (dd, J=8.08, 5.05 Hz, 1 H) 6.59 (d, J=9.60 Hz, 1 H) 7.20 - 7.29 (m, 2 H) 7.31 - 7.46 (m, 8 H) 7.49 - 7.56 (m, 1 H) 7.95 (dd, J=9.09, 2.02 Hz, 1 H) 8.13 (s, 1 H) 8.34 (d, J=2.02 Hz, 1 H) TOF MS ES+ (M+H + ): 996.20; HPLC Retention time = 3.55 minutes. Example 39

N-{7-[l-(4'-Chloro-b iphenyl-2-ylmethyl)-piperidin- 4-yl 1-5.6.7.8-tetrahydro-pyrido[3 , 4- d]pyrimidm-4-yl}-4-[(R)-l-(3A-dichloro-phemlsulfaw

propyl mino]-3-trifluoromethanesulfonyl-benzenesulfonamide

TOF MS ES+ (M+H + ): 996.19; HPLC Retention time = 3.55 minutes.

Example 40

(R)-N-{7-(l-(f4'-chlorobiphe l-2-yl)methyl)piperidin-4-yl)-5.6.7,8- tetrahvdropyridof3.4-d]pyrimidin-4-yl)-4-(l-(2-chlorophenyUh io)-4-(dimethylam b utan-2-yl amino) -3 -(trifl uoromethylsulfonyl) benzenes lfonamide

' H NMR (400 MHZ, CHLOROFORM-d) 6 ppm 1 .46 (t, J=10.36 Hz, 2 H) 1 .58 - 1 .88 (m, 4 H) 1 .89 - 2.01 (m, 1 H) 2.1 1 (s, 6 H) 2. 1 5 - 2.44 (m, 4 H) 2.49 (br. s., 2 H) 2.67 (br. s., 2 H) 2.71 - 2.84 (m, 2 H) 2.89 - 3. 1 1 (m, 2 H) 3.25 (s, 2 H) 3.45 - 3.57 (m, 2 H) 3.57 - 3.66 (m, 1 H) 3.75 - 3.92 (m, 1 H) 6.53 (d, J=9.60 Hz, 1 H) 7.04 - 7.44 (m, 12 H) 7.62 (d, J=8.08 Hz, 1 H) 7.81 (d, J=9.09 Hz, 1 H) 8.08 (br. s., 1 H) 8.22 (s, 1 H) TOF MS ES+ (M+H*): 962.24; HPLC retention time = 3.45 minutes.

Example 41

(R)-N-(7-(1 (4'-chlorobiDhenyl'2~yl)methyl)Dweridin-^

tetrahvdropyrido[3 -d]pyrimidin-4-yl)-4-(l-(3,5-dichloroDfa

buttm-2-ylamino)-3-(trifluoromethylsulfonyl)benzenesulfon amid^

Ή NMR (400 MHz, MeOD) δ ppm 1.47 - 1.65 (m, 2 H) 1.82 - 1.96 (m, 3 H) 1.96

- 2.14 (m, 3 H) 2.40 - 2.57 (m, 7 H) 2.57 - 2.77 (m, 4 H) 2.81 - 2.96 (m, 4 H) 3.19 - 3.29 (m, 1 H) 3.35 - 3.44 (m, 1 H) 3.54 (s, 2 H) 3.59 (s, 2 H) 4.04 (dd, J=8.34, 4.80 Hz, 1 H) 6.92 (d, J=9.60 Hz, 1 H) 7.13 (s, 1 H) 7.19 - 7.28 (m, 3 H) 7.30 - 7.46 (m, 6 H) 7.52 (d, J=6.57 Hz, 1 H) 8.08 (dd, J=9.09, 2.53 Hz, 1 H) 8.14 (s, 1 H) 8.41 (d, J=2.02 Hz, 1 H) TOF MS ES+ (M+H : 996.19; HPLC retention time = 3.60 minutes.

Example 42

N-(7-(l-(l-(4 '-chlorobiphenyl-2-yl)ethyl)piperidin- 4-yl) -5.6, 7.8-tetrahvdropyridof 3, 4- dJpyrimidin-4-yl) -4-( (R)-4-morpholino- 1 -(phenylthio)butan-2-ylamino) -3- (trifluoromethylsulfonvDbenzenesulfonamide

TOF MS ES+ (M+H + ): 984.30; HPLC Retention time = 6.97 minutes.

Example 43

(R)-N 7-(1 (4'-chlorobwhenyl-2'yl)methyl)Dweridin-4-yl)-5.6.7.8- tetr hvdropyrido[3,4-d] pyrimidin-4-yl)-4-(l-(phenylthio)pentan-2-ylamino)-3- (trifluoromethylsulfonyl) benzenesulfonamide

TOF MS ES+ (M+H ): 899.25; HPLC Retention time = 5.02 minutes.

Example 44

N-(7-(l-((4 '-chlorobiphenyl-2-yl) methvDpiperidin- 4-yl )-5, 6, 7.8- tetrahydropyrido[3, 4- d]pyrm ' idin-4-yl)-4-(l-(phenvhhio)propan-2-ylamino)-3-(trifluo

benzenesulfonamide

TOF MS ES+ (M+H : 871.21; HPLC Retention time = 4.75 minutes.

Example 45

N-(7-(l-((2-(4-chlorophenyl)-4.4-dimethylcvclohex-l-ewl)meth yl)piperidin-4-yl)-5, 6, 7,8- tetrahvdropwido[3 -dlDwimidin-4-yl)-4-(l-(yhenylthw^

(trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H 4 ): 903.28; HPLC retention time - 5.07 minutes.

Example 46

(R)-N-(7-(l-(2-(but-2-vmloxy)benzyl)piperidm-4-yl)-5,6J -tetrahy

dJpyri idin-4-yl)-4-f4- di ethylamino)-]-(phenylthio)butan-2-y^

(trifluoromethylsulfonvDbenzenesulfonamide

TOF MS ES+ (M+H 4 ): 886.31 ; HPLC retention time = 3.07 minutes.

Example 47

d]pyrimidin-4-vU-4-[(R)-3-dimethylamino-l-(2-fluoro^heny

propylamino 1-3-trifluoromethanesulfonyl-benzene sulfonamide

TOF MS ES+ (M+H + ): 946.26; HPLC retention time = 3.36 minutes.

Example 48

N-{(S)'7-[l-(4'~Chloro-biphenyl-2-ylmethyl)^iperidin-4-yll-6 -methyl-5,6, 7,8-tetrahydro- pyndof3.4-d]pyrimidin~4~vU-4-((R)~3-dim€thylammo-l-Dhenyls ulfanylmethyl- propylamino)-3 trifluoromethanesulfonyl-benzene sulfonamide

TOF MS ES+ (M+H*): 942.29; HPLC retention time - 3.42 minutes.

Example 49

N-{(R)-7-fl-(4 '-Chloro-bipheml-2 metM)-piperidm-4-yl 6~methyl~5.6.7, 8- tetrahydro-pyridof 3, 4-d]pyrimidin-4-vU -4-((R) -3-dimethylamino- 1 -

TOF MS ES+ (M+H 4 : 942.29; HPLC retention time = 3.42 minutes.

Example SO

(R)-N-(2~chloro- 7- (l- ( r 4'-chlorobiphe >l-2-yl)meihyl)piperidin-4'yl)-5.6.7, 8- tetrahvdropyrido[3 -dlpyrimidin-4-yl)-4-(4-(dimethylamino

ylammo)-3-(trifluoromethyl^lfonyl)benzenesulfonamide

Example 51

N-(7-( l-(l-(4'-chlorobiphenyl-2-yl)ethyl)piperidin-4-yl)-5, 6, 7, 8-tetrahydropyridof3, 4- d]pyrimidin-4-yl)-4-((R)-4-(dimethylamino)-l-(phenylthio)b

(trifluoromethvhulfonvDbenzenesulfonamide

Ή NMR (400 MHz, DMSO) δ 8.26 (d, J = 2.02 Hz, 1H), 8.01 (s, 1 H), 7.89 (dd, i = 7.07, 2.02 Hz, 1 H), 7.56 (d, J = 8.08 Hz, 1H), 7.49 (d, J = 8.59 Hz, 1H), 7.40 (t, J = 8.59 Hz, 1 H), 7.36-7.26 (m, 7H), 7.21 (d, J = 7.07 Hz, 1H), 7.15 (d, J = 6.57 Hz, 1H), 6.91 (d, J = 8.59 Hz, 1H), 6.77 (d, J = 9.60 Hz, 1H), 3.96 (m, 2H), 3.52 (m, 2H), 2.98 (m, 2H), 2.79 (m, 2H), 2.72-2.62 (m, 3H), 2.47-2.33 (m, 8H), 1.96 (m, 1H), 1.82 (m, 1H), 1.70 (m, I H), 1.45 (m, 1H), 1.35 (m, I H), 1.21 (d, J = 6.57 Hz, 3H); TOF MS ES+ (M+H + ): 942.28; HPLC retention time = 3.41 minutes. Example 52

N-(7-(l-((4 '-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5, 6, 7, 8-tetrahydropyridof 3, 4- dlpyrimidin-4-yl)-4-(2-(phenylthio)ethylamino)~3^

benzene sulfonamide

TOF MS ES+ (M+tT): 857.20; HPLC retention time - 4.62 minutes.

Example 53

(R)-N-(7-a-((4'-chlowbiphe l-2-yl)methyl)Di rid^

tetrahydmpyrido[3A-d]pyrimidin-4-yl}-4-(4-(dimetM

ylamin -3-(triiluoromethyl)benzenesulfonamide

TOF MS ES+ (M+FT): 864.31 ; HPLC retention time = 3.26 minutes.

Example 54 (R)-N-(7-(l-((4'-chlorobiphenyl-2-yl)methyl)DiDeridin-^

tetrahydropyrido[3,4-d]pyrimidin-4-yl)-3-cyano-4-(4-{dime tto

(phenylthio)butan-2-yl amino) bemenesulfonamide

TOF MS ES+ (M+H : 821.32; HPLC retention time = 3.05 minutes.

Example 55

(R)-N-(7-(l-((2-(4-chlorovhenyl)-4 -dimethylcyclo ex-l-enyl)methyl^

5,6, 7,8-tetrahvdropyridoiS -d]pyrimidm-4 )-4-(l-(2-fluorophenylthio)-4'(4-

'HNMR(400 MHz, MeOD) δ ppm 1.01 (s, 6 H) 1.46- 1.58 (m, 2 H) 1.60- 1.82 (m,3H) 1.87 - 1.99 (m, 2 H) 1.99 - 2.13 (m, 3 H) 2.13 - 2.31 (m,4H) 2.31 - 2.76 (m, 16 H) 2.81 (t,J=5.81 Hz, 2 H) 3.06 - 3.27 (m, 6 H) 3.55 (s, 2 H) 3.98 (dd,J=8.59, 4.55 Hz, 1 H) 6.70 - 6.84 (m, 1 H) 6.96 - 7.13 (m, 4 H) 7.17 - 7.29 (m, 1 H) 7.29 - 7.43 (m, 3 H) 7.97 (dd, J=9.09, 2.02 Hz, 1 H) 8.13 (s, 1 H) 8.34 (d, J=2.53 Hz, 1 H) TOF MS ES+ (M+H + ): 1033.37; HPLC retention time = 3.74 minutes.

Example 56

(R)-N-(7-a-«4'-chlorobiphenyl-2-yl)metM)piperid^

tetrahvdropwidof 3, 4-dlpyrimidin-4-yl)-4-(l-(2-iluorophenylthio)-4-(4-methylpip erazin- l-yl)b tan~2-ylamino)-3-(trifluoromethylsulfo l)benzenesulfonamide

Ή NMR (400 MHz, MeOD) δ ppm 1.47 - 1.67 (m, 2 H) 1.73 (d, J=6.06 Hz, 1 H) 1.87 (br. s., 2 H) 2.04 (t, J=10.86 Hz, 3 H) 2.28 - 2.82 (m, 16 H) 2.83 - 3.01 (m, 4 H) 3.04 - 3.26 (m, 2 H) 3.58 (d, J=17.68 Hz, 4 H) 3.89 - 4.05 (m, 1 H) 6.70 - 6.85 (m, 2 H) 6.95 - 7.1 1 (m, 2 H) 7.17 - 7.30 (m, 2 H) 7.30 - 7.47 (m, 6 H) 7.47 - 7.59 (m, 1 H) 7.97 (dd, J=9.35, 2.27 Hz, 1 H) 8.13 (s, 1 H) 8.33 (d, J=2.02 Hz, 1 H) TOF MS ES+ (M+F^): 1001.31; HPLC retention time = 3.43 minutes.

Example 57

N-{7-[l-(4'-Fluoro-biphenyl-2~ylmethyl)-piperidm-4-yl J-5, 6, 7, 8-tetrahvdro-pyrido[3,4- d]pyrimidin-4-yl}-4-f(R)-l'(3-chloro^henylsulfaml ethyl)-3-dime

propylamim]-3-trifluoromethanesulfoml-benzenesulfonamide

TOF MS ES+ (M+H ): 946.27; HPLC retention time = 3.36 minutes.

Example 58

(R)-N-(7-(l-((4'-chlorobivhenyl-2-yl)methyl)piperidin-4-yl)- 5,^

tetmhvdwpyridoi3 -d]pyrimidin-4'yl)-4-(4-morpholino-l-(phenylthio)butan-2^ ylam ino)-3 -nitrobenzene sulfonamide

TOF MS ES+ (M+H^): 883.32 HPLC retention time = 4.76 minutes.

Example 59

N-(7-(l-((2-(4-chlomphenyl)AA-dimethylcyclohex-l-enyl)methy 7,8- ietrahydropyridof3,4-dlpyrimidin-4-vi)-4-(l'(phenylthio)pent an-2 amino)-3-

(triiluoromethylsulfonyDbenzenes lfonamide

TOF MS ES+ (M+H + ): 931.31 ; HPLC retention time = 5.33 minutes.

Example 60

(R)-N-(7-(4-(( 4 f -chlorobiphenyl-2-yl)methyl) -4-methoxycvclohexyl) -5,6, 7,8- tetra tdropryrido{3A-dlpyrim\din-4-yl)-4-(4-(dim

ylamin -3-(trifluoromethylsulfonyl)be enesulfonamide

Ή NMR (400 MHz, CHLOROFORM-i/) 6 ppm 8.22 (dd, J=17.0, 2.0 Hz, 1 H), 7.98 - 8.12 (m, 1 H), 7.74 (dd, J=10.0, 2.5 Hz, 1 H), 7.53 - 7.68 (m, 1 H), 7.38 (d, J-7.5 Hz, 1 H,) 7.1 1 - 7.36 (m, 10 H), 7.09 (t, J=7.5 Hz, 1 H), 6.43 (dd, J=17.0, 10.0 Hz, 1 H), 3.73 - 3.86 (m, 1 H), 3.53 (br. s., 1 H), 3.33 (s, 1 H), 3.12 (s, 1 H), 2.88 - 3.09 (m, 4 H), 2.71 - 2.86 (m, 2 H), 2.67 (br. s., 1 H), 2.40 (br. s., 2 H), 2.10 - 2.36 (m, 7 H), 1.92 - 2.08 (m, 1 H), 1.66 - 1 .80 (m, 1 H), 1.37 - 1.59 (m, 4 H), 1 .33 (t, J-7.5 Hz, 2 H), 1.14 - 1.29 (m, 3 H), 0.90 (d, J=3.5 Hz, 2 H); TOF MS ES+ (M+H + ): 957.29; HPLC retention times = cis/trans mixture: 3.96 and 4.02 minutes.

Example 61

(R)-N-(7-{ 4- (2-bromobenzyl ) -4-methoxycvclohexyl) -5, 6, 7, 8-tetrahydrovyridof3, 4- dlpyrimidm-4-yl)-4-(4-(dimethylamino)-l-(phenyhhio)buta^^

(trifluoromethylsulfonyl)benzenesulfonamide

Ή NMR (400 MHz, CHLOROFORM -d) δ ppm 8.27 - 8.38 (m, 1 H), 8.07 - 8.20 (m, I H), 7.79 - 7.93 (m, 1 H), 7.61 - 7.79 (m, 1 H,) 7.49 - 7.60 (m, 1 H), 7. 18 - 7.48 (m, 6 H), 7.10-7.05 (m, 1 H), 6.51 (d, J=10.0 Hz, 1 H), 3.82 - 4.02 (m, 1 H), 3.59 - 3.72 (m, 2 H), 3.26 - 3.41 (m, 2 H), 2.93 - 3.15 (m, 3 H), 2.79 (d, J= 13.5 Hz, 2 H), 2.58 (d, J=5.5 Hz, 3 H), 2.26 (br. s., 5 H), 2.01 - 2.16 (m, 1 H), 1 .17 - 1.98 (m, 14 H); TOF MS ES+ (M+H + ): 925.21 ; HPLC retention time = 3.67 minutes.

Example 62

N-C7- (4-((4'-chlorob iphenyl-2-yl)me thylene) cvclohexyl)-5, 6, 7.8-tetrahydropyrido [3.4- d]pyrimidm-4-yl)-4-((R)-4-(dimethylamino)-l-(pfa

ftrifluoromethylsulfonvDbe zenesulfonamide

1H NMR (400 MHz, CHLOROFORM-i/) δ ppm 8.26 (s, 1 H), 8.10 (s, 1 H), 7.81 (dd, J=9.0, 2.01 Hz, 1 H), 7.57 - 7.75 (m, 1 H), 7.13 - 7.44 (m, 12 H), 6.48 (d, J=9.0 Hz, 1 H), 6.03 (s, 1 H), 3.77 - 3.95 (m, 1 H), 3.61 (s, 2 H), 2.90 - 3.15 (m, 2 H), 2.43 - 2.80 (m, 6 H), 1.85 - 2.41 (m, 10 H), 1.76 (t, J=1 1.0 Hz, 4 H), 1.26 - 1.48 (m, 2 H), 0.90 - 1.10 (m, 1 H); TOF MS ES+ (M+H + ): 925.26; HPLC retentiont time = 6.71 minutes.

Example 63

N-(7-(4-benzylidemcvclohexyl)-5.6 .8-tetrahvdropyrido[3

(dimethylamino)-l-(phenylthio)butan-2-ylamino)-3 trifluoromethylsulfonyl)

benzenesulfonamide

1H NMR (400 MHz, CHLOROFORM-^ δ ppm 8.26 (s, 1 H), 8.10 (s, 1 H), 7.74 - 7.90 (m, 1 H), 7.04 - 7.46 (m, 9 H), 6.45 (d, J-9.0 Hz, 1 H), 6.24 (s, 1 H), 3.81 (d, J=3.51 Hz, 1 H), 3.63 (s, 2 H), 2.86 - 3.15 (m, 3 H), 2.78 (br. s., 3 H), 2.56 (br. s., 2 H), 2.36 - 2.50 (m, 2 H), 2.09 - 2.33 (m, 8 H), 1.84 - 2.08 (m, 5 H), 1.66 - 1.82 (m, 1 H), 1.30 - 1.61 (m, 2 H); TOF MS ES+ (M+H + ): 815.27; HPLC retention time = 3.64 minutes.

Example 64

(R)'N-(7-(4-((4'-chlorobipheml-2-yl)methyl)-4-hydroxycycl ohexyl)-5.6.7, 8- tetrahvdropyndof3 -dlwrimidin-4-yl)-4-(4-(dimethylamino)-l-(pfa

ylamino -3-(trifliwwmethylsulfonyl)bemenesulfonamide

TOF MS ES+ (M+H . 943.27; HPLC retention times = cis/trans mixture: 3.70 and 3.81 minutes.

General Reductive Amination Procedure 3.

Amine V (1 eq) and ketone IX (1.2 eq) were dissolved in DCE (10 mL/ mmol V), and NaHC0 3 (6 eq) was added. The reaction was stirred at ambient temperature for 3 hours, and then heated to 65°C for 3 hours. Na(AcO) 3 BH (3 eq) was then added, and the reaction was stirred at 65 °C for 16 hours. The reaction was diluted with CH 2 CI 2 , washed with water followed by brine, dried over Na 2 S0 4 , filtered and concentrated. The crude residue was purified by flash chromatography on silica gel (0-10% methanol/CH 2 Cl 2 followed by 20% 7N NH 3 in methanol/CH 2 Cl 2 ). Example 65

N-(7-(l-((4 '-Chlorobiphenyl-2-yl)methyl)-3-fluoropiperidin-4-yl)-5, 6, 7, 8- tetrah dropyridof3A^lpyrimidmA )A-((R)-4-tmr^

ylamino)-3-(trifluoromethylsulfonyl)be enesulfonamide.

Following General Reductive Amination Procedure 2, (Λ)-4-(4-Μοφ1ιο1ίηο-1- ( henylthio)butan-2-ylarnino)-N-(5,6,7,8-tetrahydropyrido[3,4- d]pyrimidin-4-yl)-3- (trifluoromethylsulfonyl)benzenesulfonamide (34 mg, 0.05 mmol) and l-((4'- chlorobiphenyl-2-yl)methyl)-3-fluoropiperidin-4-one (19 mg, 0.059 mmol) afforded the title compound (6 mg, 12% yield).

'H NMR (400 MHz, CDCl 3 ) 6 ppm 1.22 (m, 1 H), 1.48 - 1.77 (m, 3H), 1.83 - 2.15 (m, 4H), 2.18 - 2.60 (m, 8H), 2.76 - 3.16 (m, 5H), 3.21 - 3.45 (m, 2H), 3.5 1 - 3.94 (m, 6H), 4.69 - 4.97 (m, 1H), 6.52 (d, J- 9.60 Hz, 1H), 6.92 (d, J= 8.59 Hz, 1 H), 7.10 - 7.51 (m, 13H), 7.79 (dd, J = 9.09, 2.02 Hz, 1H), 8.06 (br. s., 1H), 8.22 (br. s., 1 H).

MS [M+H] + = 988.27

HPLC retention time: 3.66 minutes (Agilent 1 100 HPLC system; Inertsil ODS3 100 x 3mm C 18 column; flow rate I mL/ minute; 5-95% ACN/water with 0.1% formic acid; 7.75 minute run)

Examples 66-72 were prepared by reductive amination of ketones IX with amines V following General Procedure 3 above.

Example 66 N-(7-(l-((4 '-chlorobiphenyl-2-yl)methyl)-3-fl uoropiperidin-4-yl) -5,6, 7.8- tetrahydroTryridoF3,4-d]pyrimidin-4-yl)-4-((R)-4-(dimethylam

ylamino)-3-(trifluoromethylsulfonyl)benzenesulfonamide.

Ή NMR (400 MHz, ACETONITRILE-^) δ ppm 8.07 - 8.14 (m, 2H), 7.83 (dd,

J-9.09, 2.02 Hz, IH), 7.22 - 7.40 (m, 9H), 7.06 - 7.19 (m, 4H), 6,89 (d, J=9.09 Hz, I H), 6.80 (d, J=9.60 Hz, I H), 4.69 - 4.93 (m, I H), 3.99 (dd, J=8.59, 5.05 Hz, IH), 3.58 (s, 2H), 3.20 - 3.32 (m, 2H), 3.04 - 3.20 (m, 2H), 2.85 - 2.97 (m, I H), 2.65 - 2.85 (m, 5H), 2.44 (s, 6H), 2.38 (t, J=5.81 Hz, 2H), 1.88 - 2.16 (m, 4H), 1.74 - 1.84 (m, 1 H), 1.56 (d, J= 12.63 Hz, IH); MS [M+H] + : 946.26.

Example 67

fR)-N-(7-(l-((4''Chlorobiphenyl-2-yl)methyl)piperidinA-yl )-2^

tetrahvdropyrido[3,4-dlp imidin-4-yl)-4-(4-(dimeihylamino)-l-(pheny

Ή NMR (400 MHz, ACETONITRILE- 3) δ ppm 8.11 (d, J=2.02 Hz, IH), 8.01 - 8.06 {m, IH), 7.44 - 7.49 (m, IH), 7.39 - 7.43 (m, 3H), 7.28 - 7.37 {m, 4H), 7.16 - 7.26 (m, 4H), 7.05 (d, J=8.59 Hz, IH), 6.61 (d, J-9.09 Hz, IH), 3.84 - 3.94 (m, IH), 3.48 (s, 2H), 3.34 (s, 2H), 3.13 (d, J=6.06 Hz, 2H), 2.78 (d, J=l 1.62 Hz, 2H), 2.72 (t, J=5.81 Hz, 2H), 2.52 - 2.60 (m, IH), 2.46 - 2.52 (m, 2H), 2.32 - 2.42 (m, 2H), 2.23 (s, 6H), 2.1 1 (dd, J=5.05, 2.53 Hz, IH), 1.89 (d, J=l 0.61 Hz, IH), 1.71 - 1.80 (m, 4H), 1.41 - 1.53 (m, 2H); MS (ESI) m/e (M+H) + = 996.26.

Example 68

fR)-N-i7-fl-^4'-chlorobiphenyl-2-yl)methyl)piperidin-4-yl )-2^

tetrahvdropwido[3A-d]pyrimidin-4 )-4-(4-morpholino

ylamino)- -(trifl oromethylsulfoml)benzenesulfonamide.

Ή NMR (400 MHz, ACETONITRILE-rf,) δ ppm 8.12 (d, J=2.02 Hz, IH), 8.02 - 8.07 (m, IH), 7.50 - 7.55 (m, IH), 7.39 - 7.47 (m, 4H), 7.32 - 7.39 (m, 4H), 7.18 - 7.30 (m, 4H), 6.75 (d, J=9.09 Hz, 1 H), 6.68 (d, J=8.59 Hz, 1 H), 3.95 - 4.07 (m, 1 H), 3.58 (s, 2H), 3.18 (dd, J-6.06, 4.04 Hz, 2H), 2.83 - 2.93 (m, 4H), 2.57 (br. s., 4H), 2.24 - 2.39 (m, 8H), 2.13 - 2.21 (m, 2H), 2.01 - 2.08 (m, 1H), 1.77 - 1.88 (m, 4H), 1.54 - 1.68 (m, 4H), 1.26 - 1.33 (m, 1 H); MS (ESI) m/e (M+H) + - 1038.28.

Example 69

N-(7-(l-((4'-chlorohiphenyl-2-yl)methyl)-3-fluoropiperidin-4 -yl)-2-(trifl^

5,6, 7,8-tetrahydropyrido[3.4-d]pyrimidin-4-yl)-4-((R)-4-(dimethy lamino)-l- (phenylthio)buian-2-ylamino)-3-(irifl owmethylsulfon\l)h^

Ή NMR (400 MHz, ACETONITRILE-^) δ ppm 8.10 (d, J=2.02 Hz, 1 H), 7.44 - 7.50 (m, 3H), 7.39 - 7.44 (m, 2H), 7.30 - 7.39 (m, 4H), 7.18 - 7.28 (m, 4H), 7.08 (d, J=8.08 Hz, 1H), 6.61 (d, J=9.09 Hz, 1 H), 4.91 (d, J=50.53 Hz, 1 H), 3.83 - 3.95 (m, 1 H), 3.57 (s, 2H), 3.25 - 3.38 (m, 2H), 3.10 - 3.18 (m, 2H), 2.95 - 3.06 (m, 1 H), 2.77 - 2.91 (m, 3H), 2.27 - 2.55 (m, 8H), 2.18 - 2.23 (m, 6H), 2.08 - 2.14 (m, 1 H), 1.98 - 2.04 (m, 1 H), 1.83 - 1.91 (m, 1 H), 1.72 - 1.80 (m, 1 H), 1.69 (br. s., 1 H); MS (ESI) m/e (M+H) + = 1014.25.

Example 70

(R) -N- (7~(ί-((2-( 4-chlorophenyl) -5, 5-dimeihylcyclohex- 1 -enyl) methyl)piperidin-4-yl)-2- (trifluoromethyl)-5, 6, 7, 8-tetrahydropyrido[3, 4-d]pyrimidin-4-yl)-4-(4-(dimethylammo)- l-{phenylthio)butan-2-ylamino)-3-(trifluoromethylsulfo

1H NMR (400 MHz, AC ETONITRILE-i/ 3 ) δ ppm 8.13 (d, J=2.53 Hz, 1 H), 8.02 - 8.06 (m, IH), 7.31 - 7.37 (m, 4H), 7.17 - 7.26 (m, 3H), 7.08 (d, J=8.59 Hz, 2H), 7.01 (d, J=8.59 Hz, I H), 6.62 (d, J=9.09 Hz, IH), 3.83 - 3.94 (m, IH), 3.47 (s, 2H), 3.03 - 3.16 (m, 6H), 2.57 - 2.68 (m, 3H), 2.41 - 2.50 (m, 4H), 2.30 (s, 6H), 2.23 - 2.28 (m, 2H), 2.08 - 2.18 (m, IH), 1.99 (br. s., 2H), 1.65 - 1.87 (m, 6H), 1.44 (t, J=6.57 Hz, 2H), 1.25 - 1.34 (m, 2H), 0.95 (s, 6H); MS (ESI) m/e (M+H) + = 1028.33.

Example 71

(R)-N-(7-(l-((4'-chlorobipheml-2-vttmetM)piperidin-4^^

tetrahvdropyrido[3 -d1pwimidin-4-yl)-4-(4-(dimetlwlamino)-l-(p

ylamino)-3-nitrobenzenesulfonamide.

Ή NMR (400 MHz, ACETONITRI LE-i 3 ) δ ppm 8.61 (d, J=2.0 Hz, I H), 8.31 - 8.34 (d, J=10.0 Hz, I H), 7.88 - 7.91 (d, J=1 1.6 Hz, I H), 7.15 - 7.57 (m, 12H), 6.71 (d, J=9.5 Hz, IH), 3.99 - 4.04 (m, IH), 3.48 (s, 2H), 3.34 (s, 2H), 3.17 3.23 (m, 2H), 2.56- 2.82 (m, 7H), 2.49 - 2.53 (m, 2H), 2.36 (s, 6H), 2.07-2.27 (m, I H), 1.71 - 1.76 (m, 5H), 1.46 - 1.52 (m, 2H); MS (ESI) m/e (M+H) + = 909.30.

Example 72

(R)-N-(6-(l-(( 4 '-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-6, 7-dihydro-5H-pyrrolo[3, 4- d]pyrimidin-4-yl)-4-(4-morpholino-l-(phenylthio)butan-2-ylam ino)-3^

( trifl uoromethylsulfonyl) benzenesulfonamide.

! H NMR (400 MHz, MeOD) δ ppm 8.35 (d, J=2.02 Hz, I H), 8.24 (s, I H), 7.96 (dd, J=9.22, 2.15 Hz, I H), 7.56 - 7.62 (m, I H), 7.42 - 7.49 (m, 4H), 7.33 - 7.37 (m, 4H), 7.28 - 7.32 (m, I H), 7.16 - 7.27 (m, 3H), 6.83 (d, J=8.84 Hz, 1 H), 6.74 (d, J=9.35 Hz, I H), 3.90 (br. s., 6H), 3.57 - 3.65 (m, 4H), 3.43 - 3.50 (m, I H), 3.09 - 3.26 (m, 3H), 2.98 - 3.08 (m, 2H), 2.67 - 2.76 (m, 1 H), 2.26 - 2.48 (m, 7H), 1.94 - 2.13 (m, 3H), 1.55 - 1.78 (m, 3H), 1.27 - 1.33 (m, I H); MS (ESI) m/e (M+H) + = 956.27.

General Reductive Amination Procedu

A solution of amine V (1 eq), ketone IX (1 eq), acetic acid (1 eq), sodium triacetoxyborodeuturide (1.5 eq) and molecular sieves (~400 mg/ mmol V) in DCE (12.4 mI7 mmol V) was stirred at 45°C for 12 hours. The mixture was cooled to room temperature, filtered through Celite, and concentrated. The crude residue was purified by flash chromatography on silica gel (0-100% methanol in CH 2 C1 2 ).

Examples 73-91 were prepared using the general reductive amination procedure 4 described above.

Example 73

(R)-N-(7-(l-((4'-chloro-4-fluorobiphenyl-2-yl)methyl)-4-f a

tetrahvdropyridof3, 4-dlpyrimidin-4-yl) -4- ( 4- (dimethylamino)- 1 -(phenylthio) butan-2- ylamino)- -(triftuoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H + ): 947.27; HPLC retention time = 3.45 minutes.

Example 74

N-(7-(l-((S)-l-(4'-chlorobiphenyl-2-yl)ethyl)-4-deuteropiw

( trifl uoromethyl) -5, 6, 7.8-tetrahvdropyridof 3, 4-dlwrimidin-4-yl) -4-((R)-l-(2- fluorophenylthio)-4-(4-methylpiperazin-l-yl)butan-2-ylc iino)-3~

( trifluoromethylsulfonyl) benzene sulfonamide

TOF MS ES+ (M+H + ): 1048.32; HPLC retention time = 3.89 minutes.

Example 75

(R)-N-(7-(l-((2-(4-chloropheml)-4, 4-dimethylcyclohex-l-enyl)methyl)-4- deuteropiperidin-4-yl)-2-(dimethylamino)-5.6.7.8-tetrahvdrop yridoi3, 4-d]pyrimidin-4- yl)-4-(l- (2-fluoroyhenylthio)-4-( 4-me thylmperazin-l-yl) butan-2-ylamino)-3 - (trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H + ): 1077.42; HPLC retentiont time = 4.37 minutes.

Example 76

(R)-N-(7-(4-((4'-chlorobiph yl-2 )methyl)-4-methoxy-l-dueterocvclohexyl)-5, 6, 7, 8- tetrahydropyrido [ 3, 4-dJpyrimidin- 4-yl)-4- 2-fluorophenylthio)-4- ( 4-methylpiperazin- l~yl)butan-2-ylamino)-3-(trifJuoromethylsulfonyl)benzenesulf onam

TOF MS ES+ (M+H 4 ): 1029.32; HPLC retention time = 4.92 minutes.

Example 77

4-((R)-l -Benzyloxymethyl-3-dimethylam ino-yroDylamino) -N-{7-[4-de utero-l-(4 '-chloro- 4-fluoro-b iphenyl-2-ylmethyl)-piperidin-4-yl 7-5.6. 7, 8-tetrahydro-pyrido[3.4-dJpyri idin- 4-yl}-3-trifluoromethanesulfonyl-bertzertesulfortamide

TOF MS ES+ (M+H 4 ): 945.31 HPLC retention time = 3.47 minutes.

Example 78

N-(7-{l-[2-( 4-Chloro-vhenyl )-4, 4-dimethyl-cyclo hex-l-enylmethyl1-4-deutero-piperidin~ 4-yl) -5.6. 7.8-tetrahvdro-Dyrido[3.4-d]pyrimidin-4-yl)-4-[(R)-3 - (isopropyl-methyl- amino) - 1 -p enylsulfanylmethyl-propyl mirto 1-3-trifluoromethanesulfowl- benzenesulfonamide

H 3 C CH 3

TOF MS ES+ (M+H^): 989.38; HPLC retention time = 3.71 minutes.

Example 79

4-((R)-3-Amino-l^henylsulfanylmethyl-propylamino)-N'{7-[4-de utero-l-(4'-c}doro-4- fluoro-biphenyl-2-ylmethyl)-piperidin-4-yl]-5,6, 7,8-tetrahydro-pyridof3, 4-dJpyrimidin-4- yl}-3-trifluoromethanesulfonyl-benzenesulfonamide

TOF MS ES+ (M+H 4 : 919.24; HPLC retention time = 3.34 minutes.

Example 80

4- (2R)-4-f8-oxa-3-azabicychf3.2 Joctan-3'Vl)-J-(phenylthio)butan-2-ylamino)-N-(7- (l-((4'-chloro-4-fluorobiphenyl-2-yl)methyl)-4-deutempiperid in-4-yl)-5,6, 7,8- tetrahydroDyrido f 3 , 4-dlpyrimidin- 4-yl) -3-(trifl uoromethylsulfonyl ) benze e sulfonamide

TOF MS ES+ (M+rf): 1015.30; HPLC retention time = 3.61 minutes.

Example 81

4-f(2R)-4-(8-oxa-3 izabicvclo[3.2 1octc -3-yl)-l-(phenylthio)b tan-2-ylamm^ (l-((2-(4-chlorophenyl)AA-dimethyl ycloh x-l-enyl)methy

5, 6, 7, 8-tetrahvdropyridof3, 4-d1pwimidin-4-yl)-3-(trifluoromethylsulfonyl)- benzenesulfonamide

TOF MS ES+ (M+H 4 : 1029.37 HPLC retention time = 7.89 minutes.

Example 82

(R)-N-(7-(l-((4'-chloro-4-fluorobipheml-2-yl)methyl)-4-dwter opiperid 6.7, 8- tetrahydropyridof 3.4-dlpyrimidin-4-yl) -4-fl- fphenylthio) -4-(pyrrolidin- 1 butan-2- ylamino)-3-(trifluoromethylsulfonyl)ben∑enesulfonamide

TOF MS ES+ (M+H + ): 973.29; HPLC retention time = 3.46 minutes.

Example 83

(R)-N-(7-(l-((2-(4-chloropheml)-4 -dimethylcyclohex-l-enyl)metfTyl)-4- deuteropiperidin-4-yl) -5.6.7, 8-tetrahvdropyrido[3, 4-d]pyrimidin-4-yl)-4-( l-(2- fl uorophenylthio)-4- ( 4-methylpiperazin- 1 -yl)butan-2-ylamino)-3- (trifluoromethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H + ): 1034.38; HPLC retention time = 3.73 minutes.

Example 84

N-{7-f4-Deutero-l-(4'-chloro-4-fluoro-biphenyl~2'yl ethyl)^iperidm~4-yl J-5, 6, 7, 8- tetrahvdro-pyrido[3A-d]pyrimidin-4-vtt-4-((R)-3-dimethylamin o-l- phenylsulfanylmethyl-prooylamino)-3-trifluoromethamsulfonyl- bemenesulfonamide

TOF MS ES+ (M+H + ): 975.30; HPLC retention time - 3.51 minutes.

Example 85

(R)~N-(7-(l-((2-(4'Chlorophenyl)-4A-dimethylcvclohex-l-enyl) methyl)-4- deuteropiperidin-4-yl)-5,6 ,8-tetrah dropyrido[3 -d]pyrimidin-4-yl)-4-^

4- (pyrrol idin-l-yl) butan-2-ylamino)-3- (rifluoromethylsulfonyl) benzene sulfonamide

TOF MS ES+ (M+H + ): 987.36; HPLC retention time = 3.71 minutes.

Example 86

N-(7-{l-[(R)-l-(4'-Chloro-bmhenyl-2-yl)-ethyl 4-deuteropiperM^

tetrahvdro-pyrido[3A-d]pyrimidin-4-yl)~4-((R)-3-morpholin -4-yl-l- phenylsulfanylmethyl-propylamino)-3-trifluorometha su^

TOF MS ES+ (M+H 4 : 985.31 ; HPLC retention time = 3.45 minutes.

Example 87

(R)-N-(7-(l-({4'-chloro-4-ftuorobiphenyl-2-yl)methyl)-4~deut em 7,8- tetrahydropyrido[3, 4-dJpwim idin- 4-yl) -4-(l -(2-fluorophenylthio) -4-( 4-methylpiperazin- 1 -yl)b tan-2-ylamino) -3-( trifluoromethylsulfonyl) benzenesulfonam ide

TOF MS ES+ (M+H 4 ): 1020.31 ; HPLC retention time = 3.49 minutes.

Example 88

N-(7-{l-i(S)-l-(4'-Chloro pheml-2-yl)-ethyll-4-deuteropiperidin-4-yn-5

tetrahydro-pyrido[3, 4-dlpyrimidin-4-yl)-4-((R)-3-morphol in-4-yl-l- phenylsulfanylmethyl-propylamino)-3-trifluoromethanes lfonyl-benzenes lfon mide

TOF MS ES+ (M+H ): 985.31 ; HPLC retentiont time = 3.46 minutes.

Example 89

(Tl)-N-(7-(l-((4'-chloro-4-fluorobipheml-2-yl)metM)-4-deutew 6, 7, 8- tetrahydropyrido[3, 4-d]pyrimidm-4-yl) -4-( 4- ( 4-ethylpiperazin- 1 -yl)-l-(phenylthio) butan- 2-ylamino)-3-ftrifluowmethylsulfonyl)benzenesulfonamide

TOF MS ES+ (M+H ): 1016.33 HPLC retention time - 3.51 minutes.

Example 90

N-(7-(l-(l-(2-(4-chlorophenyl)-4,4-dimethylcvclohex-l-enyl)e thyl) 4-deuteropiperidin-4- yl)-5.6.7.8-tetrahydropyrido[3 -d]pyrimidin-4-yl)-4-((R)-4-D-morpholino-l- (phenylthio)butan-2-ylamino)-3-(trifl oromethylsulfony^

TOF MS ES+ (M+H 4 : 1017.37; HPLC retention time = 3.76 minutes

Example 91

(R) -N-(7-( l-((2-( 4-chlorophenyl)-4, 4-dimethylcvclohex~J-enyl)methyl)- 4- deuteropiperidin-4-yl)-5.6, 7, 8-tetrahydropyrido[3.4~d pyrimidin-4-yl}-4~(4-morpholino- l-(uhenylthio)butan-2-yla ino)-3-ftrifluoromethylsulfonyl)be enesulfonamide

TOF MS ES+ (M+H 4 ): 1003.35 HPLC retention time = 3.73 minutes.

Synthesis of Examples 92-93 by reductive amination of ketones IX with amines V.

Example 92

N-{6-[l-(4'-Chloro-biphenyl-2~ylmetM)^iperidin-4-yl]-5

tetraaza-azulen-3-yl}-4-((R)-3-morpholin-4-yl-l^hemlsulfa nylme

trifl oromet anesulfonyl-benzenesulfonamide.

To a vial containing 4-(( ^)-3-moφholin-4-yl-l-phenylsulfanylmethyl- propylamino)-N-(5,6,7,8-tetrahydro-4H-l,2,3a,6-tetraaza-azul en-3-yl)-3- trifluoromethanesulfonyl benzenesulfonamide (50 mg, 0.072 mmol) and l-(4'-chloro- biphenyl-2-ylmethyl) -piper id in-4-one (21.73 mg, 0.072 mmol) was added

dichloromethane (1.5 mL) and methanol (1.5 mL) under nitrogen. The reaction mixture was stirred in an ice bath for 10 minutes, and then acetic acid (2.176 mg, 0.036 mmol) was added. After stirring in the ice bath for another 30 minutes, sodium

cyanoborohydride (27.3 mg, 0.434 mmol) was added, and the reaction was heated to 45°C and stirred for 14 hours. The reaction mixture was concentrated under reduced pressure, and then diluted with dichloromethane and water. Organ ics were extracted three times with dichloromethane. The combined organic layers were washed with brine, dried over a 2 S0 , filtered and concentrated under reduced pressure. This crude material was purified via flash chromatography on silica gel (10-15% 2N NH in methanol in CH 2 C1 2 ) to afford the title compound (5.5 mg, 8% yield). Ή NM (600 MHz, DMSO-rf 6 ) δ ppm 8.03 (d, J = 2.27 Hz, 1 H), 7.88 (dd, J= 9.06, 1.89 Hz, 1 H), 7.40 - 7.50 (m, 5H), 7. 14 - 7.39 (m, 9H), 6.99 (d, J= 9.44 Hz, 1 H), 6.85 (d, J= 9.06 Hz, 1H), 4.06 (d, J= 5.67 Hz, 1H), 4.00 (t, J= 7.37 Hz, 1 H), 3.80 (d, J = 6.80 Hz, 1 H), 3.47 (d, J = 2.64 Hz, 4H), 3.24 - 3.31 (m, 4H), 2.77 - 2.83 (m, 2H), 2.64 - 2.76 (m, 5H), 2.41 - 2.47 (m, 1 H), 2.19 - 2.36 (m, 5H), 2.15 (d, J= 6.80 Hz, 2H), 1.93 (d, J = 5.67 Hz, 1 H), 1.81 (q, J= 1 1.96 Hz, 2H), 1.71 (td, J = 13.88, 5.85 Hz, 1 H), 1.50 - 1.61 (m, 2H), 1.31 - 1.42 (m, 2H).

HR-MS (m/z, MH+): 973.30

HPLC retention time: 3.50 minutes (Agilent 1 100 HPLC system; Inertsil ODS3 100 x 3mm C 18 column; flow rate of 1.0 mL/minute; gradient of 5-95%

acetonitrile/water with 0.1% FA)

Example 93

N-{6-[ l-(4'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl ]-5, 6, 7,8-tetrahydro-4H-l , 2, 3a, 6- tetraaza~azulert'3~yl}-4-tfR)-3-dimethylamim-l-phenyl-sulfy^

trifl uoromethanesulfonyl -benzenesulfonamide.

To a vial containing 4-((if)-3-Dimethylamino-l -phenylsulfanylmethyl- propylamino)-N-(5,6,7,8-tetrahydro-4H-l ,2,3a,6-tetraaza-azulen-3-yl)-3- trifluoromethanesulfonyl-benzenesulfonamide (1200 mg, 0.185 mmol) and l -(4'-chloro- biphenyl-2-ylmethyl)-piperidin-4-one (55.5 mg, 0. 185 mmol) was added methanol (6 mL) under nitrogen. The reaction mixture was stirred in an ice bath for 10 minutes, and then acetic acid (5.56 mg, 0.093 mmol) was added. After stirring in the ice bath for another 30 minutes, sodium cyanoborohydride (34.9 mg, 0.556 mmol) was added, and the reaction was stirred at ambient temperature for 14 hours. The reaction mixture was concentrated under reduced pressure and filtered. This crude material was purified via flash chromatography on silica gel (10-15% 2N NH 3 in methanol in CH 2 C1 2 ) to afford the title compound (24 mg, 20% yield).

1H NMR (400 MHz, DMSO-i/ 6 ) δ ppm 8.03 (d, J- 2.02 Hz, 1H), 7.88 (dd, J=

9.09, 2.53 Hz, 1H), 7.31 - 7.51 (m, 10H), 7.24 - 7.31 (m, 3H), 7.15 - 7.23 (m, 2H), 6.91 (d, J= 9.60 Hz, 1H), 4.01 (br, s, 1H), 3.80 (d, J- 7.58 Hz, 2H), 3.15 - 3.27 (m, 2H), 2.79 (d, J= 5.56 Hz, 2H), 2.65 - 2.75 (m, 6H), 2.31 - 2.48 (m, 3H), 2.12 - 2.22 (m, 1H), 2.08 (s, 6H), 1.65 - 1.96 (m, H), 1.49 - 1.60 (m, 2H), 1.29 - 1.43 (m, 2H).

HR-MS (m/z, MH+): meas. 931.28

HPLC retention time: 3.39 minutes (Agilent 1100 HPLC system; Inertsil ODS3 100 x 3mm C18 column; flow rate of 1.0 mL/minute; gradient of 5-95%

acetonitrile/water with 0.1% formic acid) Examples 94-96 were prepared by reductive amination of ketones VI with amines

V and removal of protecting group to afford Intermediates VII, followed by reductive amination with carbonyl compounds VIII.

Example 94

N-{7-[l-f4'-Chloro-biphenyl-2-ylmethyl)-piperidin-4-yl 7-5.6.7, 8-tetrahvdro-

Π.2.41triazolof 4, 3-alt>yrazin-3-yl }-4-( (R)-3-moryholin-4-yl - 1 -phenylsulfanylmethyl- propylamino)'3-trifluowmethanesulfonyl-benzenesulfonamide.

STEP A: To a vial containing 4-((/i)-3-morpholin-4-yl-l -phenyl-suIfanylmethyl- propylamino)-N-(5,6,7,8-tetrahydro-[ l ,2,4]triazolo[4,3-a]pyrazin-3-yl)-3- trifluoromethanesulfonyl-benzenesulfonamide (50 mg, 0.074 mmol) and 4-oxo- piperidine- l -carboxylic acid tert-buty\ ester (14.74 mg, 0.074 mmol) was added methanol (2 mL) under nitrogen. The reaction mixture was stirred in an ice bath for 10 minutes, and then acetic acid (0.424μΕ, 7.40 μηιοΐ) was added. The reaction mixture was stirred for another 30 minutes. Sodium cyanoborohydride (41 .85 mg, 0.666 mmol) was then added, and the reaction was stirred at ambient temperature for 72 hours. The reaction mixture was concentrated, filtered and purified via flash chromatography on silica gel ( 10-50% 7N NH 3 in methanol in CH 2 CI 2 ) to afford 4-{3-[4-((/ -3-Morpholin-4-yl-l - phenylsulfanylmethyl-propylamino)-3-trifluoromethanesulfonyl -benzenesulfonylamino]- 5,6-dihydro-8H-[l,2,4]triazolo[4,3-a]pyrazin-7-yl}-piperidin e- l -carboxylic acid tert- butyl ester (65 mg, 100% yield). MS [m/z; (M+l) + ]: 859.7

STEP B: To a solution of 4-{3-[4-((A)-3-morpholin-4-yl-l -phenylsulfanyl- methyl-propylamino)-3-trifluoromethanesulfonyl-benzenesulfon ylamino]-5,6-dihydro- 8H-[ l ,2,4]triazolo[4,3-a]pyrazin-7-yl}-piperidine- l-carboxylic acid /e/-/-butyl ester (65 mg, 0.076 mmol) in CH 2 C1 2 (2 mL) under nitrogen was added trifluoroacetic acid (0.146 mL, 1 .892 mmol), and the reaction was stirred for 2 hours. The reaction mixture was then concentrated under reduced pressure. The residue was dissolved in methanol and converted to the free base by elution through a Si-Carbonate SPE filter to afford 4-((Λ)-3- Mo holin-4-yl-l -phenylsulfan lmeth l-prop lamino)-N-(7-piperidin-4-yl-5, 6,7,8- tetrahydro- [1,2 ,4]tr iazolo [4, 3 -a]pyrazin-3 -y I)-3 -tr ifluoromethanesulfony 1- benzenesulfonamide (37 mg, 65% yield). MS [m/z; (M+l) ): 759.7

STEP C: To a vial containing 4-((i^)-3-mo holin-4-y^-l-phenylsulfanyl-methyl- propylamino)-N-(7-piperidin-4-yl-5,6,7,8-tetrahydro-[l,2,4]t riazolo[4,3-a]pyraz ' in-3-yl)- 3-trifluoromethanesulfonyl-benzenesulfonamide (37 mg, 0.049 mmol) and 4'-chloro- biphenyl-2-carbaldehyde (21.13 mg, 0.098 mmol) was added methanol (1.5 mL) under nitrogen. The reaction mixture was stirred in an ice bath for 10 minutes and then acetic acid (2.79μ1_,, 0.049 mmol) was added. After stirring in the ice bath for an additional 30 minutes, sodium cyanoborohydride (18.38 mg, 0.293 mmol) was added. The reaction was then allowed to warm to ambient temperature and stir for 72 hours. The solvent was removed in vacuo, and the residue purified via flash chromatography on silica gel (10- 50% methanol in CH 2 C1 2 ) to afford the title compound (18 mg, 39 % yield).

1H NMR (400 MHz, DMSO-</ 6 ) ) δ ppm 1.26 - 1.38 (m, 1H), 1.44 (q, J= 9.87 Hz, 2H), 1.51 - 1.78 (m, 4H), 1.87 - 2.01 (m, 1H), 2.09 - 2.19 (m, 2H), 2.20 - 2.34 (m, 4H), 2.34 - 2.40 (m, 2H), 2.41 - 2.47 (m, 1H), 2.65 - 2.74 (m, 1H), 2.74 - 2.82 (m, 2H), 3.28 (d, J= 7.03 Hz, 1H), 3.45 - 3.53 (m, 8H), 3.58 (s, 2H), 4.05 (d, J= 5.52 Hz, 1H), 6.84 (d, J= 9.03 Hz, 1H), 6.95 (d, J= 9.54 Hz, 1H), 7.15 - 7.22 (m, 2H), 7.27 (t, J= 7.78 Hz, 2H), 7.30 - 7.38 (m, 4H), 7.39 - 7.51 (m, 6H), 7.90 (dd, J= 9.03, 1.51 Hz, 1H), 8.06 (s, 1H).

HR-MS (m/z, MH+): meas. 959.28

HPLC retention time: 3.60 minutes (Agilent 1 100 HPLC system; Inertsil ODS3 100 x 3mm CI 8 column; flow rate of 1.0 mL/minute; gradient of 5-95%

acetonitrile/water with 0.1% FA) Example 95

N-{7-[l-(4'-Chloro-biphenyl-2-ylmethyl)-azepan-4-yll-5.6.7.8 -tetrahvdro- fl Altriazolof4 -a]pyrazm-3-yl)-4-((R)-3-morphol\n-4^

propylamino)-3-trifluoromethamsulfonyl-benzenesulfonamide

STEP A: To a vial containing 4-(( i)-3-morpholin-4-yl- l-phenyl-sulfanylmethyl- propylamino)-N-(5,6,7,8-tetrahydro-[l ,2,4]triazolo[4,3-a]pyrazin-3-yl)-3- trifiuoromethanesulfonyl-benzenesulfonamide (80 mg, 0.1 18 mmol) and 4-oxo-azepane- 1 -carboxylic acid tert-buty\ ester (63.1 mg, 0.296 mmol) was added methanol (4 mL) under nitrogen. The reaction mixture was stirred in an ice bath for 10 minutes and then acetic acid (8.13μί, 0.142 mmol) was added. After stirring in the ice bath for an additional 15 minutes, sodium cyanoborohydride (59,5 mg, 0.947 mmol) was added. The reaction was then allowed to warm to ambient temperature and stir for 72 hours. The reaction mixture was then concentrated, filtered and purified via flash chromatography on silica gel (10-50% 7N NH 3 in methanol in CH 2 C1 2 ) to afford 4-{3-[4-((tf)-3-Morpholin-4- yl-l-phenylsulfanylmethyl-propylamino)-3-trifluoromethanesul fonyl- benzenesulfonylamino]-5,6-dihydro-8H-[ l ,2,4]triazolo[4,3-a]pyrazin-7-yl}-azepane-l - carboxylic acid r/-butyl ester (76 mg, 74 % yield). MS [m/z; (M+ l ) + ]: 873.8

STEP B: To a solution of 4-{3-[4-((-?)-3-morpholin-4-yl- l-phenylsulfanyl- methyl-propylamino)-3-trifluoromethanesulfonyl-benzenesulfon ylamino]-5,6-dihydro- 8H-[ l,2,4]triazolo[4,3-a]pyrazin-7-yl}-azepane-l -carboxylic acid tert-butyl ester (76 mg, 0.087 mmol) in CH 2 CI 2 (2 mL) under nitrogen was added trifluoroacetic acid (0.168 mL, 2.176 mmol), and the reaction was stirred for 2.5 hours. The reaction mixture was concentrated, water and CH 2 CI 2 were added, and the solution was basified to pH approximately 8 with saturated a 2 C0 3 . The aqueous layer was extracted three times with CH 2 C1 2 . The combined organic layers were washed with brine, dried over a 2 S0 , filtered and concentrated under reduced pressure to afford N-(7-Azepan-4-yl-5,6,7,8- tetrahydro-[l,2,4]triazolo[4,3-a]ρ razin-3- l)-4-(( f)-3-mo ho ln-4- l-l- phenylsulfanylmethyl-propylamino)-3-trifluoromethanesulfonyl -benzenesulfonamide (62 mg, 92% yield). MS [m/z; (M+l) + ]: 773.6

STEP C: To a vial containing N-(7-azepan-4-yl-5,6,7,8-tetrahydro- [l,2,4]triazolo[4,3-a]pyrazin-3-yl)-4-(( f)-3-mo holin-4-yl-l-phenylsulfanylmethyl- propylamino)-3-trifluoromethanesulfonyl-benzenesulfonamide (62 mg, 0.080 mmol) and 4'-chloro-biphenyl-2-carbaldehyde (52.1 mg, 0.241 mmol) was added methanol (2.5 mL) under nitrogen. The reaction was stirred in an ice bath for 10 minutes and then acetic acid (5.51 μί., 0.096 mmol) was added. After stirring in the ice bath for an additional 15 minutes, sodium cyanoborohydride (40.3 mg, 0.642 mmol) was added. The reaction was then allowed to warm to ambient temperature and stir for 72 hours. The reaction mixture was concentrated, filtered and purified via flash chromatography on silica gel (10-50% methanol in CH 2 C1 2 ) to afford the title compound (50 mg, 64% yield).

Ή NMR (400 MHz, DMSO-rf 6 ) δ ppm 8.06 (d, J = 2.01 Hz, 1H), 7.90 (d, J = 7.53 Hz, 1H), 7.39 - 7.52 (m, 6H), 7.31 - 7.38 (m, 4H), 7.27 (t, J = 7.78 Hz, 2H), 7.18 - 7.22 (m, 2H), 6.95 (d, J= 9.54 Hz, IH), 6.84 (d, J= 9.03 Hz, 1H), 4.05 (d, J= 5.52 Hz, 1H), 3.58 (s, 2H), 3.44 - 3.52 (m, 8H), 3.22 - 3.33 (m, 2H), 2.64 - 2.86 (m, 4H), 2.45 (d, J - 5.02 Hz, IH), 2.23 - 2.40 (m, 7H), 2.1 1 - 2.20 (m, 2H), 1.88 - 1.98 (m, IH), 1.57 - 1.77 (m, 4H), 1.44 (q, J= 9.87 Hz, 2H).

HR-MS (m/z, MH+): measured 973.29

HPLC retention time: 3.57 minutes (Agilent 1100 HPLC system; Inertsil ODS3 100 x 3mm C 18 column; flow rate of 1.0 mL/minute; gradient of 5-95%

acetonitrile/water with 0.1% formic acid) Example 96

N-{7-[l-(4'-Chloro-bipheml~2~ylmethvB-2-methyl^iperidin-4^

f 2 ltriazolof4 -alDyrazin-3-v -4-((R)-3-mo^holin-4-yl-l^hemlsulfam

propylamino)-3-trifluoromethanesulfonyl-benzene sulfonamide.

STEP A: To a vial containing 4-((Λ)-3-πιο 1ιοΗη-4-γ1-1 -ρ1ιεη Ί5υ1Γ3η Ί-πΐ€ΐ1ι>Ί- propylamino)-N-(5,6,7,8-tetrahydro-[l,2,4]triazolo[4,3-a]pyr azin-3-yl)-3- trifluoromethanesulfonyl-benzenesulfonamide (60 mg, 0.089 mmol) and 2-methyl-4-oxo- piperidine- l -carboxylic acid tert-butyl ester (47.3 mg, 0.222 mmol) was added methanol (3 mL) under nitrogen. The reaction mixture was stirred in an ice bath for 10 minutes and then acetic acid (6.10μί, 0.107 mmol) was added. After stirring in the ice bath for an additional 1 5 minutes, sodium cyanoborohydride (44.6 mg, 0.710 mmol) was then added, and the reaction was stirred at ambient temperature for 15 hours. The reaction mixture was concentrated, filtered, and purified via flash chromatography on silica gel

(10-50% 7 NH 3 in methanol in CH 2 C1 2 ) to afford 2-methyl-4-{3-[4-(( J ί^)-3-mo holin-4- yl-l-phenylsulfanylmethyl-propylamino)-3-trifluoromethanesul fonyl- benzenesulfonylamino]-5,6-dihydro-8H-[ l ,2,4]triazolo[4,3-a]pyrazin-7-yl}-piperidine-l - carboxylic acid fer/-butyl ester (42 mg, 53 % yield). MS [m/z; (M+l ) + ]: 873.8

STEP B: To a solution of 2^ε 1-4-{3-[4-((Λ)-3^οφ1ιο1ίη-4^1- 1- phenylsulfanylmethylpropyl- amino)-3-trifluoromethanesulfonyl-benzenesulfonylamino]- 5,6-dihydro-8H-[ 1 ,2,4] triazolo [4,3-a]pyrazin-7-yI}-piperidine-l -carboxylic acid tert- butyl ester (42 mg, 0.048 mmol) in CH 2 C1 2 (1 mL) under nitrogen was added

trifluoroacetic acid (0.093 mL, 1 .203 mmol) and the reaction was stirred for 2.5 hours. The reaction mixture was then concentrated and diluted with water and CH 2 C1 2 . It was then basified to pH approximately 8 with saturated Na 2 C0 3 . The aqueous layer was extracted three times with CH2CI2. The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure to afford N- [7-(2-methyl-piperidin-4-yl)-5,6,7,8-tetrahydro-[l,2,4]triaz olo[4,3-a]pyrazin-3-yl]-4-((i¾)- 3 -morphol in-4-y 1 - 1 -phen l sulfany lmethy 1-propy lamino)-3-tri fluoromethanesu lfonyl - benzenesulfonamide (22 mg, 59% yield). MS [m/z; (M+l) + ]: 773.6

STEP C: To a vial containing N-[7-(2-methyl-piperidin-4-yl)-5,6,7,8-tetrahydro- [ 1 ,2 ,4] triazo lo [4 ,3 -a]pyrazin-3 -y l]-4-((_¾)-3 -morphol i n-4-y 1- 1 - henyl sul fany 1 methyl - propylamino)-3-trifluoro methanesulfonyl-benzenesulfonamide (20 mg, 0.026 mmol), 4'- chloro-biphenyl-2-carbaldehyde (16.82 mg, 0.078 mmol) and zinc chloride (3.53 mg, 0.034 mol) was added methanol (1 mL) under nitrogen and the mixture was stirred for 10 minutes. Sodium cyanoborohydride (13.01 mg, 0.207 mmol) was then added, and the reaction was stirred at ambient temperature for 14 hours. The reaction mixture was then concentrated and partitioned between ethyl acetate and 2 M NaOH. The aqueous layer was extracted three times with EtOAc. The combined organic layers were washed with 1M NaOH, dried under Na 2 S0 4 , filtered and concentrated under reduced pressure. The crude material was purified via HPLC (0.1% TFA Modifier in water in MeCN) to afford the title compound as a TFA salt (5 mg, 18 % yield).

Ή NMR (400 MHz, DMSO-< 6 ) δ ppm 13.15 (d, J= 10.54 Hz, 1H), 8.08 - 8.18 (m, 1H), 7.90-8.01 (m, 1H), 7.66 - 7.84 (m, 1H), 7.51 - 7.61 (m, 4H), 7.34 - 7.44 (m, 3H), 7.28 - 7.34 (m, 2H), 7.22 - 7.27 (m, 2H), 7.14 - 7.21 (m, 1H), 7.02 - 7.1 1 (m, 1H), 6.84 (t, J= 8.03 Hz, 1H), 4.06 - 4.21 (m, 3H), 3.89 - 4.03 (m, 4H), 3.62 - 3.71 (m, 3H), 3.54 - 3.64 (m, 4H), 3.48 - 3.56 (m, 2H), 3.15 - 3.28 (m, 1H), 2.95 - 3.14 (m, 4H), 2.81 - 2.92 (m, 2H), 2.63 - 2.76 (m, 3H), 2.03 - 2.22 (m, 2H), 1.75 (d, J= 12.55 Hz, 1H), 1.57 (br. s., 1H), 1.31 (d, J= 8.01 Hz, 1H), 1.21-1.27 (m, 2H), 0.94 (d, J= 7.03 Hz, 1H).

HR-MS (m/z, MH+): 973.29

HPLC retention time: 3.59 minutes (Agilent 1100 HPLC system; lnertsil ODS3 100 x 3mm CI 8 column; flow rate of 1.0 mL/minute; gradient of 5-95%

acetonitrile/water with 0.1% formic acid) Suzuki coupling with 3-chloro-phenylboronic acid.

Example 97 Ν-{7-[1-β '-Chloro-biphenyl-2-ylmethyl) -pipehdin-4-yl 1-5,6, 7, 8-tetrahydro-pyridof 3,4- d]pyrimidin-4-yl}-4-((R)-3-dimethylamino-l-phenyls lf^

trifliioromethanesulfonyl-benzenesulfonamide.

A microwave vial equipped with a stir bar was charged with sodium carbonate

( 1 1.8 mg, 0.1 1 mmol). The vial was then placed in an oven and dried for 30 minutes. It was then removed from the oven and allowed to cool to room temperature under nitrogen. Example 7 (50 mg, 0.056 mmol), 3-chloro-phenylboronic acid (13.1 mg, 0.084 mmol) and Pd(PPh 3 ) 4 (3.2 mg, 0.0028 mmol) were added, followed by DME: EtOH: water (2 mL, 2: 1 : 1 ratio). The resulting mixture was degassed by bubbling nitrogen through the solution, and then heated conventionally to 80°C for 16 hours. The reaction was then cooled to room temperature and filtered. The filtrate was concentrated and the crude residue was purified by flash chromatography on silica gel (0-100% methanol in CH 2 CI 2 ) to afford the title compound (26 mg, 50% yield).

' H NMR (400 MHz, MeOD) δ ppm: 8.35 (S, I H), 8.12 (s, IH), 7.95 (dd, J = 2.01 ,

9.03 Hz, I H), 7.52 (s, I H), 7.46-7.50 (m, I H), 7.12-7.43 (m, 1 1H), 6.71 (d, J = 9.54 Hz, IH), 3.93 (m, IH), 3.60 (s, 2H), 3.49 (s, 2H), 3.10-3.27 (m, 2H), 2.62-2.96 (m, 8H), 2.49- 2.58 (m, 7H), 2.12 (m, I H), 1.97-2.07 (m, 2H), 1.84-1.95 (m, 3H), 1.51 -1.63 (m, 2H).

HR-MS (m/z, MH+): measured 928.55

HPLC retention time = 3.38 minutes (Agilent 1 100 HPLC system; lnertsil ODS3

1 00 x 3mm C 18 column; flow rate of 1.0 mL/minute; gradient: 5-95% acetonitrile/water with 0.1% FA over 7.75 minutes). The following compounds were prepared by Suzuki coupling of Example 7 with the requisite boronic acid following the procedure for Example 97 above.

Example 98

N-(7-(l-((2 '-chlorobiphenyl-2-yl)methyl)piperidin-4-yl)-5.6, 7, 8-tetrahvdropyridof 3.4- d ]pwimidin-4-yl)-4- ((R)-4- (dimethylamino)-! - (phenylthio)butan-2-ylamino)-3- (trifluoromethylsulfonyl)benzene sulfonamide

TOF MS ES+ (M+H " "): 928.27; HPLC retention time =3.32 minutes

Example 99

(R)-4-(4-(dimethylamino)-l-(phenylthio)butan-2-ylamino)-N-(7 -(I-((4'- (trifluoromethyl)biphenyl-2-yl)methyl)piperxdin-4-yl)-5, 6, ? 8-tetrahydropyridof3, 4- dJpyrimidin-4-yl)-3- ( trifluoromethylsulfonvDbenzenesulfonam ide

TOF MS ES+ (M+F : 962.30; HPLC Retention time = 3.69 minutes.

PHARMACOLOGICAL DATA

Biology Assay Section

Method for determining IC50s

The present method includes utility of a Surface plasmon resonance (SPR)-based biosensor (Biacore™' GE Healthcare, Uppsala, Sweden) to characterize BCL-2 inhibitors.

Biacore™ utilizes the phenomenon of surface plasmon resonance (SPR) to detect and measure binding interactions. In a typical Biacore experiment, one of the interacting molecules (ligand) is immobilized on a flexible dextran matrix while the interacting partner (analyte) is allowed to flow across that surface. A binding interaction results in an increase in mass on the sensor surface and a corresponding direct change in the refractive index of the medium in the vicinity of the sensor surface. Changes in refractive index or signal are recorded in resonance units (R.U.) Signal changes due to association and dissociation of complexes are monitored in a non-invasive manner, continuously and in real-time, the results of which are reported in the form of a sensorgram.

The SPR assay is configured to examine solution inhibition of BCL-2 binding to peptide derivatized sensor surfaces to generate 1C50 values as a measure of inhibitor potency. Solution inhibition assay format:

Biacore™ A 100 (GE Healthcare, Uppsala, Sweden) was used to conduct all experiments reported herein. Sensor surface preparation and ail interaction analyses experiments were performed at 25°C. Reagents were purchased from GE Healthcare. Running buffer containing lOm Hepes, pH7.4, 150m sodium chloride, 1.25mM Dithiothreitol, 3% Dimethyl sulfoxide and 0.05% polysorbate 20 were utilized throughout all analyses.

Biotinylated BAK, BAD and NOXA peptides were diluted to ΙΟη in running buffer and captured onto a sensor surface pre-derivatized with streptavidin (sensor chip SA) to peptide surface densities in the range 50 - 100 R.U. Peptide captured surfaces were blocked with 500μ PE0 2 -Biotin. A blank detection spot in each flowcell was similarly blocked with PE0 2 -biotin and served as a reference spot in the competition assay.

Interaction analyses were performed by first equilibrating each sample within a 6 point three fold compound dilution series in the range 16μ to 0.004n with 56nM BCL2 for one hour during instrument start-up procedures. Protein compound mixtures were then injected over each peptide surface in parallel for 60 seconds at a flow-rate of 30μ1_Ληίη. 56n BCL2 control samples were also prepared and run at regular intervals during the assay. Surface regeneration was performed at the end of each analysis cycle by two 30 second injections of lOmM Glycine, pH 2.5, 1 Sodium Chloride, 0.05% polysorbate 20. Samples and control compound samples were run in duplicate and controls are also run at regular intervals during the assay to monitor surface and assay performance.

Data analyses are carried out using Biacore™ A 100 evaluation software vl. l to validate assay quality. Binding level report points were used relative to BCL2 control samples to calculate % inhibition values for each compound protein mixture. These data are then plotted versus compound concentration and analyzed in Tibco ® Spotfire ® v2.1 via logistic regression to calculate IC50 values for each compound. Table 4 shows the IC50 value of selected compounds.

TABLE 4

tr uoromet ysu ony enzenesu onam e