Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PHENYL LINKED QUINOLINYL MODULATORS OF ROR-GAMMA-T
Document Type and Number:
WIPO Patent Application WO/2015/057203
Kind Code:
A1
Abstract:
The present invention comprises compounds of Formula I. Formula I wherein: R1, R2, R3, R4, R5, R6, R7, R8, and R9 are defined in the specification. The invention also comprises a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is rheumatoid arthritis or psoriasis. The invention also comprises a method of modulating RORγt activity in a mammal by administration of a therapeutically effective amount of at least one compound of claim 1.

Inventors:
BARBAY KENT (US)
EDWARDS JAMES P (US)
KREUTTER KEVIN D (US)
KUMMER DAVID A (US)
MAHAROOF UMAR (US)
NISHIMURA RACHEL (US)
URBANSKI MAUD (US)
VENKATESAN HARIHARAN (US)
WANG AIHUA (US)
WOLIN RONALD L (US)
WOODS CRAIG R (US)
FOURIE ANNE (US)
XUE XIAOHUA (US)
CUMMINGS MAXWELL D (US)
LEONARD KRISTI A (US)
Application Number:
PCT/US2013/065040
Publication Date:
April 23, 2015
Filing Date:
October 15, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JANSSEN PHARMACEUTICA NV (BE)
International Classes:
C07D401/14; A61K31/4709; A61P29/00; C07D401/06; C07D409/14; C07D413/06; C07D417/06; C07D417/14
Domestic Patent References:
WO2012064744A22012-05-18
Other References:
THEODORE M. KAMENECKA ET AL: "Synthetic modulators of the retinoic acid receptor-related orphan receptors", MEDCHEMCOMM, vol. 4, no. 5, 1 January 2013 (2013-01-01), pages 764, XP055069085, ISSN: 2040-2503, DOI: 10.1039/c3md00005b
Attorney, Agent or Firm:
JOHNSON, Philip, S. et al. (One Johnson & Johnson PlazaNew Brunswick, New Jersey, US)
Download PDF:
Claims:
What is claimed is:

1. The compounds of Formula I:

wherein:

I is azetidinyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, thiazoiyl, pyridyl, pyridyl N- oxide, pyrazinyl, pyrimidinyi, pyridazyi, piperidinyl, tetrahydropyranyl, phenyl, oxazoiyl, isoxazolyl, thiophenyl, benzoxazolyl, or quinolinyl; wherein said piperidinyl, pyridyl, pyridyl N- oxide, imidazolyl, phenyl, thiophenyl, benzoxazolyl, and pwazoiyl are optionally substituted with SG2CHk C(0)CH3, C(0)NH2, CH3, CH2CH3, CF3, CL F, -CN, OCH ,. N(CH )2, - ίί Ή .μ.Χ Ή :. SCH3, OH, CO7H, C02C(CH3)3, or OCH2OCH3; and optionally substituted with up to two additional substituents independently selected from the group consisting of CI, OCH3, and CH3; and wherein said triazolyl, oxazoiyl, isoxazolyl, and thiazoiyl are optionally substituted with one or two CH3 groups; and wherein said azetidinyl is optionally substituted with C02C(CH3)3, ( "(())% H... CH3, S02CH3, or C(0)CH3;

Rz is 1 -methyl- 1 , 2, 3-triazo yl, pyridyl, pyridyl-N-oxide, 1 -methyl pyrazol-4-yl, pyrimidin-5-yl, pyridazyi, pyrazin-2-yl, oxazoiyl, isoxazolyl, N-acetyl-azetidin-3-yl, N- rnethy]sulfonyl-azetidin-3-yi N-Boc-azetidin-3-yl, N-methyl-azetidin-3-yl, iV-acetamidyl- azetidin-3-yl, N-acetyi piperidinyl, 1 -H-piperidinyl, N-Boc-piperidinyi, N-C(1-2)alk.yl-piperidinyl, thiazol-5-yl, 1 -methyl imidazol-2-yi l-(3-rnethoxypropyl)-irmdazol-5-yl, or l-C^alkyl imidaz.ol-5-yl; wherein said 1 -C(1..2)a3kyl imidazol-5-yl is optionally substituted with up to two additional CH3 groups, or one substituent selected from the group consisting of SCH3, and CI; and said pyridyl, and pyridyl-N-oxide are optionally substituted with up to two substituents independently selected from the group consisting of C(0)NH2, -CN, OC'I h. CF , CI, and CH3; and said thiazol-5-yl, oxazoiyl, and isoxazolyl are optionally substituted with up to two C!¾ groups; and said 1 -methyl pyrazol-4-yl is optionally substituted with up to two additional CH3 groups; R3 is H, OH, OC! I .-. NHCH3, N(CH3)2, or M¾:

R4 is H, or F:

R5 is H, CI, -CN, CFj, SCH3, OC(!-3)alkyl, OH, C(]-4)alkyl,

N(C(!-2)alkyl)2, NH-cyclopropyl, OCHF'2, 4-hydroxy-piperidinyl, azetidin-l-yl, or fur-2-yl;

R ' is 2-chloro-thiophen-5-yl, 1 -methyl-pyrazol-4-yl, phenyl, pyrimidinyl, or pyridvl, wherein said phenyl, pyrimidinyl, and pyridyl are optionally substituted with S02CH ,

HSO2CH3, CFj, F, CI, -CN, OCH3, or OCF3;

R7 is H, CI, -CN, C(i.4}alkyl, OCH2CF3, OCFI2CH2OCFi3, CF3, SCH3, S02CH3, OCFIF2, NA]A2, C(0)NHCH3, N(CH3)CH2CH2NAiA2, OC'1 1 ·( i 1/ΧΛ \Λ\ OCH2CH2NH2, 0( . ^!kyl. OCH2-(l-m.ethyl.)-imidazol-2-yl, imidazol-2-yl, fur-2-yl, pyrazol-4-yi, pyrid- -yl, or pyrimidm-

5-yl; thiophen-3-yl, l-methyl-indazol-5-yl, l-methyl-inda.zol-6-yl, phenyl, or

wherein said imidazolyl or pyrazolyl can be optionally substituted with a CH3 group ;

A1 is H or C,;j_4)alkyl;

A2 is C i-4)alkyl, cyclopropyi, C<-1-4)alkylOH, C(0)C(i_2!aikyL or OCH ; or AJ and A2 may be taken together with their attached nitrogen to form a ring selected from the isting of:

/ \ / \

-|-N O -|-N N-Rb

\— / , and — / ;

Rb is C!¾, or phenyl;

R8 is H, CH3, OCH3, or F;

R is H, or F;

and pharmaceutically acceptable salts thereof;

provided that (4-chloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)(pyridin-3- yi)methanamine, (4-chlorophenyl)(2,4-dichloro-3-(2-chlorophenyr)quinolin-6-yr)(l -methyl- 1H- imidazol-2-yl)methanol, (4-chloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)(pyridin- 4-yl)methanamine, (4-chlorophenyl)(3-(2,6-dichlorophenyl)quinolin-6-yl)( 1 -methyl- 1H- imidazol-5-yl)niethanoi, (4-chloro-3-phenylquinolin-6-yl)(2-(dimethylamino)p xidin-4-yl)(l- methyl-lH-imidazol-2-yl)methanol, 4-(2-((4-chloro-6-((4-chlorophenyi)(hydroxy)(l -methyl- 1H- imidazol-5-yl)methyl)-3-phenyiquinolin-2-yi)oxy)ethyl)thiomo holine 1 , 1 -dioxide, 1 ~(2-((4- chloro-6-((4-chiorophenyl)(hydroxy)(l -methyl- lH-imidazol-5-yl)methyl)-3-phenyiquinolin-2- yl)oxy)ethyl)pyrrolidin-2-oiie, (2-chloro-4-(dimethyia.mino)-3-phenylquinolin-6-yl)(pyridin-2- yl)(pyridin-4-yl)methanol, (4-chloro-3-phenylquinolin-6-yl)(2-f3.uoropyridin-4-yl)(l -methyl- 1 H- imidazol-2-yl)methanol, (4-cliloro-2-(l-metliyl-lH-pyrazol-4-yl)-3-phenylquinolin-6-yl)(4- chlorophenyl)(pyridin-3-yl)methanol, (2,4-diehloro-3-phenylquinolin-6-yl)di(pyridin-2- yl)methanol, 6-((3-chlorophenyl)(hydroxy)(2-(trifluoromethyl)pyridin-4-yl)m.ethyl)-3- phenylquinoline-2-carbonitrile, (2,4-dichloro-8-methyl-3-phenylquinolin-6-yl)(l -methyl- 1H- imidazol-4-yl)(6-methylpyridin-3-yl)methanol, (4-chlorophenyl)(2,4-dichloro-3-(2- chlorophenyl)quinolin-6-yl)(l -methyl- lH-imidazol-5-yl)methanol, (2,4-dic-hloro-3- phenylquinolin-6-yl)(phenyl)(pyridin-2-yl)m.ethanol, (2,4~dichloro-3-phenylquinolin-6- yl)(oxazol-2-y!)(phenyl)methanol, the second eluting enantiomer of (4-methoxy-3-phenyl-2- (irifiuoromethyl)quinolin-6-yl)(l -methyl- lH-imidazol-5-yl)^widin-2-yl)mem and the second eluting enantiomer of (4-chloro-2-methoxy-3-phenylquinolin-6-yl)(l -methyl- 1H- imidazol-5-yl)pyrimidin-2-ylmethanol (when purified on a chiralcei OD column) are excluded from the claim,

2. The compounds of claim 1 , wherein:

R1 is imidazolyl, thiazolyl, pyridyl, pyrimidinyl, or phenyl; wherein said yridyl, and said phenyl are optionally substituted with -CN, CF3, F, or CI; and wherein said imidazolyl, and thiazolyl are optionally substituted with one or two Cl¾ groups;

R is 1 -methyl- l ,2,3-triazoi-5-yl, Λ''-acetyl pi eridin-4-yl, N-Boc-piperidin-4-yl, Λ-methyl piperidin-4-yl, 1 -H-piperidin-4-yl, oxazol-2-yl, 2,4-dimethyl thiazol-5-yl, 1 -methyl imidazol-2-yl, 1 -methyl-imidazol-5-yl, or pyridyl; wherein said pyridyl is optionally substituted with CF3;

R3 is Η, ΟΗ;

R4 is Η;

R5 is Η, Ci, -CN, Ca-4)alkyl, OC(f _2)alkyl, SC! I .-. \{(Ί Ι .· ) -, or N(CH3)OCH3; R is 2-chloro-thiophen-5-yl, 1 -methyl-pyrazol-4-yl, phenyl, pyrimidinyl, or pyridyl, wherein said phenyl, pyrimidinyl, and pyridyl are optionally substituted with SG2CH3,

NHSO2CH3, CF3, CL -CN, OCH3, or GCF3;

R7 is CI, -CN, CF3, SC! I .-. OCH2CF , NA]A2, N(CH3)CH2CH2NA3A2, OC(i_3)alkyl, OCH2CH2OCH3, OCH2CH2NA!A2, or GCH2CH2NH2;

A1 is H, or CH3;

A2 is OCH3, C¾, CH2CH2OH, C(0)Cfi.2)alkyl, or CH2CH2OCH3; or A! and A2 may be taken together with their attached nitrogen to form a ring selected from the group consisting of:

* ].

R8 is H, or CH ;

R9 is H;

and pharmaceutically acceptable salts thereof:

provided that (4-chlorophenyl)(2,4-dichloro-3-(2-chlorophenyi)quinolin-6-yi)(l -methyl- 1H- imidazol-5-yl)niethanoi, 6-((3-chlorophenyl)(¾ydroxy)(2-(trifluoromethyl)pyridin-4-yl)methyl)- 3-phenylquinoiine-2-carbonitrile, (2,4-dichloro-3-phenyiquinolin-6-yl)di(pyridin-2-yl)methanol, (2,4-dichloro-3-pheny].quinolin-6-yi)(phenyl)(pyridm-2-yi)methanol, (2-chloro-4- (dimethylamino)-3-phenylquino].in-6-yl)(pyridin-2-y].)(pyridin-4-yl)methanol, (4- chlorophenyl)(2,4-dichioro~3~(2-chlorophenyl)quinoiin-6-yl)il -methyl- H-imidazol-2- yl)methanoi, (2,4-dichloro-3-phenylquinolin-6-yl)(oxazol-2-y].)(phenyl)methanol, the second eluting enantiomer of (4-methoxy-3-phenyl-2-(trifiuoromethyl)qu.moiin-6-yl)( -methyl- 1H- imidazol-5-yl)(pyridin~2-yl)methanol, and the second eluting enantiomer of (4-chloro-2- methoxy-3-phenylquinolin-6-yl)(l -methyl- l H-imidazol-5-yl)pyrimidin-2-ylmethanol (when purified on a chiralcel OD column) are excluded from the claim.

3. A compound of claim 1 selected from the group consisting of:

174

and pharmaceutically acceptable salts thereof.

4. A pharmaceutical composition, comprising a compound of claim 1 and a pharmaceutically acceptable carrier.

5. A pharmaceutical composition made by mixing a compound of claim 1 and a pharmaceutically acceptable carrier.

6. A process for making a pharmaceutical composition comprising mixing a compound of claim 5 and a pharmaceutically acceptable carrier.

7. A method for treating or ameliorating a RORvt mediated inflammatory syndrome, disorder or disease comprising administering to a subject in need thereof an effective amount of a compound of claim 5 .

8. The method of claim 8, wherein the disease is selected from the group consisting of: inflammatory bowel diseases, rheumatoid arthritis, psoriasis, chronic obstructive pulmonary disorder, psoriatic arthritis, ankylosing spondylitis, neutrophilic asthma, steroid resistant asthma, multiple sclerosis, and systemic lupus erythematosus,

9. The method of claim 7, wherein the disease is psoriasis.

10. The method of claim 7, wherein the disease is rheumatoid arthritis.

1 1. The method of claim. 8, wherein the inflammatory bowel disease is ulcerative colitis.

12. The method of claim. 8, wherein the inflammatory bowel disease is Crohn's disease.

53. The method of claim 7, wherein the disease is multiple sclerosis.

54. The method of claim 7, wherein the disease is neutrophilic asthma.

55. The method of claim 7, wherein the disease is steroid resistant asthma.

56. The method of claim 7, wherein the disease is psoriatic arthritis.

17. The method of claim 7, wherein the disease is ankylosing spondylitis.

18. The method of claim 7, wherein the disease is systemic lupus erythematosus.

19. The method of claim 7, wherein the disease is chronic obstructive pulmonary disorder.

20. A method of treating or ameliorating a syndrome, disorder or disease, in a subject in need thereof comprising administering to the subject an effective amount of a compound of claim 1 or composition or medicament thereof in a combination therapy with one or more anti- inflammatory agents, or immunosuppressive agents, wherein said syndrome, disorder or disease is selected from the group consisting of: rheumatoid arthritis, and psoriasis.

Description:
The invention is directed to substituted quinoline compounds, which are modulators of the nuclear receptor RORyt, pharmaceutical compositions, and methods for use thereof. More particularly, the RORyt modulators are useful for preventing, treating or ameliorating an RORyt mediated inflammatory syndrome, disorder or disease.

BACKGROUND OF THE INVENTION

Retinoic acid-related nuclear receptor gamma t (RORyt) is a nuclear receptor, exclusively expressed in cells of the immune system, and a key transcription factor driving Thl 7 cell differentiation. Thl 7 cells are a subset of CD4 " T cells, expressing CCR6 on their surface to mediate their migration to sites of inflammation, and dependent, on IL-23 stimulation, through the IL-23 receptor, for their maintenance and expansion. Thl7 cells produce several proinflammatory cytokines including IL-17A, IL-17F, IL-21 , and IL-22 (Korn, T., E. Bettelli, et al. (2009). "IL-17 and Thl 7 Cells." Annu Rev Immunol 27: 485-517.), which stimulate tissue cells to produce a panel of inflammatory chemokines, c tokines and metalloproteases, and promote recruitment of granulocytes ( olls, J. K. and A. Linden (2004). "Interleukin-17 family members and inflammation." Immunity 21 (4): 467-76; Stamp, L. K., M. J. lames, et al. (2004). "Interleukin-17: the missing link between T-cell accumulation and effector cell actions in rheumatoid arthritis" Immunol Cell Biol 82(1): 1 -9), Thl 7 cells have been shown to be the major pathogenic population in several models of autoimmune inflammation, including collagen- induced arthritis (CIA) and experimental autoimmune encephalomyelitis (EAE) (Dong, C. (2006). "Diversification of T-helper-cell lineages: finding the family root of IL-17-producing cells." Nat Rev Immunol 6(4): 329-33; McKenzie, B. S., R. A. astelein, et al. (2006). "Understanding the IL-23-IL-17 immune pathway," Trends Immunol 27(1): 17-23.). RORyt- deficient mice are healthy and reproduce normally, but have shown impaired Thl 7 cell differentiation in vitro, a significantly reduced ThI 7 cell population in vivo, and decreased susceptibility to EAE (Ivanov, II, B. S. McKenzie, et al. (2006), "The orphan nuclear receptor RORgamma t directs the differentiation program of proinflammatory TL-17+ T helper cells." Cell 126(6): 1 121 -33.), Mice deficient for IL-23, a cytokine required for ThI7 cell survival, fail to produce Thl7 cells and are resistant to EAE, CLA, and inflammatory bowel disease (IBD) (Cua, D. J., j. Sherlock, et al. (2003), "Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain." Nature 421(6924): 744-8.; Langrish, C. L., Y. Chen, et al. (2005). "IL-23 drives a pathogenic T cell population that induces autoimmune inflammation." J Exp Med 201(2): 233-40; Yen, D„ J. Cheung, et al. (2006). "IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6." J Clin Invest 116(5): 1310-6.). Consistent with these findings, an anti-IL23 -specific monoclonal antibody blocks development of psoriasis-like inflammation in a murine disease model (Tonel, G., C. Conrad, et, al. "Cutting edge: A. critical functional role for IL-23 in psoriasis." J Immunol 185(10): 5688- 91).

In humans, a number of observations support the role of the IL-23/Thl 7 pathway in the pathogenesis of inflammatory diseases. IL-17, the key cytokine produced by Thl7 cells, is expressed at elevated levels in a variety of allergic and autoimmune diseases (Barczyk, A., W. Pierzchala, et al. (2003). "Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine." Respir Med 97(6): 726-33.; Fujino, S., A. Andoh, et al. (2003). "Increased expression of interleukin 57 in inflammatory bowel disease." Gut 52(1): 65-70.; Lock, C, G. Hermans, et al. (2002). "Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis." Nat Med 8(5): 500-8.; Krueger, J. G., S. Fretzin, et al. "IL-17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis." J Allergy Clin Immunol 130(1 ): 145- 154 e9.). Furthermore, human genetic studies have shown association of polymorphisms in the genes for Thl7 cell-surface receptors, IL-23R and CCR6, with susceptibility to IBD, multiple sclerosis (MS), rheumatoid arthritis (RA) and psoriasis (Gazouli, M., I. Pachoula, et al. "N OD2/C ARD 15 , ATG16L1 and IL23R gene polymorphisms and childhood-onset of Crohn's disease." World J Gastroenterol 16(14): 1753-8., Nunez, C, B. Dema, et al. (2008). "IL23R: a susceptibility locus for celiac disease and multiple sclerosis?" Genes Immun 9(4): 289-93.; Bowes, J. and A. Barton "The genetics of psoriatic arthritis: lessons from genome-wide association studies." Discov Med 10(52): 177-83; ochi, Y., Y. Okada, et al. "A regulatory variant in CCR6 is associated with rheumatoid arthritis susceptibility." Nat Genet 42(6): 515-9.).

? Ustekinumab (Stelara®), an anti-p40 monoclonal antibody blocking both IL-12 and IL-23, is approved for the treatment of adult patients (18 years or older), with moderate to severe plaque psoriasis, who are candidates for phototherapy or systemic therapy. Currently, monoclonal antibodies specifically targeting only IL-23, to more selectively inhibit the Thl7 subset, are also in clinical development for psoriasis (Garber K. (201 1). "Psoriasis: from bed to bench and back" Nat Biotech 29, 563-566), further implicating the important role of the IL-23- and RORyt-driven ThI 7 pathway in this disease. Results from recent phase IS clinical studies strongly support this hypothesis, as anti-IL-17 receptor and anti-IL-17 therapeutic antibodies both demonstrated high levels of efficacy in patients with chronic psoriasis (Papp, . A., "Brodalumab, an anti- interleukin-17-receptor antibody for psoriasis." N Engl j Med 2012 366(13): 1 181 -9.; Leonard!, C, R. Matheson, et al. " Anti-interleukin- 17 monoclonal antibody ixekizumab in chronic plaque psoriasis." N Engl J Med 366(13): 1 190-9.). Anti-IL- 17 antibodies have also demonstrated clinically relevant responses in early trials in RA and uveitis (Hueber, W., Patei, D.D., Dryja, T., Wright, A.M., Koroleva, I., Bruin, G., Antoni, C, Draelos, Z., Gold, M.H., Durez, P., Tak, P.P., Gomez-Reino, J.J., Foster, C.S., Kim, R.Y., Samson, CM,, Falk, N.S., Chu, D.S., Callanan, D., Nguyen, Q.D., Rose, ,, Haider, A., Di Padova, F. (2010) Effects of AIN457, a fully human antibody to interieukin- 17 A, on psoriasis, rheumatoid arthritis, and uveitis, Sci Transl Med 2, 5272,).

Ail the above evidence supports inhibition of the Thl7 pathway by modulating RORyt activity as an effective strategy for the treatment of immune-mediated inflammatory diseases.

SUMMARY OF THE INVENTION

The present invention comprises compounds of Formula Ϊ.

I

wherein:

R s is azetidinyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, thiazoiyl, pyridyl, pyridyl N- oxide, pyrazinyl, pyrimidinyi, pyridazyl, piperidinyl, tetrahydropyranyl, phenyl, oxazoiyl, isoxazolyl, tliiophenyl, benzoxazolvL or qiiinolinyl; wherein said piperidinyl, pyridyl, pyridyl Λ- oxide, imidazolyl, phenyl, thiophenyl, benzoxazolyl, and pyrazolyl are optionally substituted with SO2CH3, C(0)CH 3 , C(Q)NH 2 , CH 3 , CH 2 CH 3 , CF 3 , CI, F, -CN, OCH 3 , N(CH 3 ) 2 , - (CI 1 . · ) )( SCH 3 , OH, CO,! I. ( Ό . ·( ' (Π Ι φ. or OCi X/l h: and optionally substituted with up to two additional substituents independently selected from the group consisting of CI, OCH 3 , and Q¾; and wherem said triazolyl, oxazolyl, isoxazolyl, and thiazolyl are optionally substituted with one or two CH 3 groups; and wherein said azetidinyl is optionally substituted with C ,αθΠ . C(0)NH 2 , CH 3 , SO2CFI3, or C(0)CH 3 ;

R 2 is 1 -methyl- 1 ,2,3-triazolyl, pyridyl, pyridyl-iV-oxide, 1 -methyl pyrazol-4-yl, pyrimidin-5-yl, pyridazyl, pyraziii-2-yL oxazolyl, isoxazolyl, N-aeetyl-azetidin-3-yl, Λ- methylsulfonyl-azetidin-3-yl, N-Boc-azetidin-3-yl, N-methyl-azetidin-3-yl, N-acetamidyi- azetidin-3-yl, N-acetyl piperidinyl, i -H-piperidinyl, N-Boc-piperidinyl, N- j -2) alkyl-piperidmyl (including N-rnethy!. piperidin-4-yl), thiazol-5-yl, 1 -methyl imidazol-2-yl, 1 -(3-methoxypropyl)- iniidazol-5-yl, or 1-C(i. 2 )aikyl imidazol-5-yl; wherein said 1 -C ( i -2) alkyl imidazoI-5-yl is optionally substituted with up to two additional CH 3 groups, or one substituent, selected from the group consisting of SCH 3 , and CI; and said pyridyl, and pyridyl-N-oxide are optionally substituted with up to two substituents independently selected from the group consisting of

C(0)NH 2 , -CN, OCFI 3 , CF 3 , CI, and CH 3 ; and said thiazol-5-yl, oxazolyl, and isoxazolyl are optionally substituted with up to two CFI 3 groups; and said 1 -methyl pyrazol-4-yl is optionally substituted with up to two additional CH 3 groups;

R 3 is H, OH, OCH3, NHCH3, N(CH 3 )2, o NH 2 ;

R 4 is I, or F;

R 5 is H, CI, -CN, CF 3 , SCH 3 , 0( ; : ^aikyi. OH, C ( i. 4 )alkyl, N(CFI 3 )OCH 3 , NH(C {3 . 2 )alkyl). N(C (1-2) alkyl) 2 , NH-cyc!opropyl, OCHF 2 , 4-hydroxy-piperidinyl, azetidin-l -yl, or fur-2-yl;

R 6 is 2-chloro-thiophen-5-yl, l -methyl-pyrazol-4-yl, phenyl, pyrimidinyl, or pyridyl, wherein said phenyl, pyrimidinyl, and pyridyl are optionally substituted with S0 2 CH 3 ,

NHSO 2 CH 3 , CF 3 , F, CI, -CN, OCH 3 , or OCF 3 ;

R 7 is H, CI, -CN, C a _ 4) alkyl, OCH 2 CF , OCH 2 CH 2 OCH 3 , CF 3 , SCH 3 , SO2CH3, OCHF 2 , NA j A 2 , C(0)NHCH 3 , Νί( Ί Ι : )( Ί 1.-( Ί Ι -N AJ A ' . OCl I ( Ί I.-N AJ A '. OCH 2 CH 2 NH 2 , OC (1 . 3) alkyl, OCH 2 -( 1 -methyl)-imidazol-2-yl, imidazol-2-yi, fur-2-yl, pyrazol-4-yi, pyrid-3-yl, or pyrimidin- 5-yl; thiophen-3-yl, l -methyl-indazol-5-yl, l-methyl-indazol-6-yl, phenyl, or ^i^ ^^^ wherein said imidazolyl or pyrazolyl can be optionally substituted with a C¾ group ;

A 1 is H or C (1-4) alkyl (including CH 3 );

A 2 is C i-4)alkyl (including CH ), cyclopropyl, C ( i-4)alkylOC(i-4 ) alkyl, C ( i-4)alkylOH, C(0)C ( j-2 ) alkyl, or OCH 3 ; or A 3 and A 2 may be taken together with their attached nitrogen to form a ring selected from the group consisting of:

- -N O -}-N N~R b

\— - , and \___/ ;

R a is 1 1 . I ' . OCH 3 , or OH;

b is CH 3 , or phenyl;

R 8 is H, CH 3 , OCH 3 , or F;

R 9 is H, or F;

and pharmaceutically acceptable salts thereof;

provided that (4-chloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)(pyridin-3- yl)meth.anamine, (4-cWorophenyl)(2,4-dichloro-3-(2-chlorophenyl.)quinolin-6-y l)(l -rn.ethyl- lH- imidazo!-2~yl)methanol, (4-chloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)(pyridin- 4-yl)methanamine, (4-chlorophenyl)(3-(2,6-dichlorophenyl)quinolin-6-yl)( I -methyl- 1 H- imidazol-5-yl)methanol, (4-chloro-3-phenylquinolin-6-yl)(2-(dimetliylarnino)pyridin- 4-y3.)(l - m.ethyl-lH-im.idazol-2-yl)methanol, 4-(2-((4-chloro-6-((4-chlorophenyl)(hydroxy)(l -methyl- 1 H- imidazol-5-yl)methyl)-3-phenylquinolin-2-yl)oxy)ethyl)thiorn orpholine 1 , 5 -dioxide, I -(2-((4- chk)ro-6-((4-chlorophenyl)(hydroxy)(5 -methyl- 5 H-imidazol-5-yl)methyl)-3-phenylquinolin-2- yi)oxy)ethyl)pyrrolidin-2-one, (2-chloro-4-(dimethyiamino)-3-phenylquinoiin-6-yl)(pyridin-2 - yl)(pyridin-4-yl)methano3, (4-chloro-3-phenylquino3in-6-yl)(2-f3.uoropyridin-4-yl)(l -methyl- 1 H- imidazol-2-yl)methanol, (4-chloro-2-(l -methyl- lH-pyrazoi-4-yl)-3-phenyiquinoiin-6-yi)(4- chlorophenyi)(pyridin-3-yl)methanoi, (2,4-clichloro-3-phenylc|uinolin-6-y3)cli(pyridin-2- yijmethanol, 6-((3-chlorophenyl)(hydroxy)(2-(tritIuoroniethyl)pyridin-4-y i)methyl)-3- phenylquinoline-2-carbonitrile, (2,4-dichloro-8-methyl-3-phenyiquino3in-6-yi)(l -methyl- 1H- imidazol-4-yl)(6-methylpyridin-3-yl)metha.nol, (4-chiorophenyl)(2,4-dichloro-3-(2- chlorophenyi)quinolin-6-yl)(l -methyi-lH~imidazol-5-yl)methanol, (2,4-dichioro-3- phenylquinolin-6-yl)(phenyl)(pyridin-2-yi)methanol, (2,4-dichloro-3-phenylquinolin-6- yl)(oxazol-2-yl)(phenyl)methanol, the second eluting enantionier of (4-methoxy-3-phenyl-2- (trifluoromethyl)quinolin-6-yl)(i -methyl- lH-imidazol-5-yl)(pyridin-2-yl)methanol, and the second eluting enantiomer of (4-chloro-2-methoxy-3-phenylquinolin-6-yl)(l -methyl- 1H- imidazol-5-yl)pyrimidin~2~ylmethano] (when purified on a chiralcel OD column) are excluded from the embodiment.

DETAILED DESCRIPTION OF THE INVENTION

The present, invention comprises compounds of Formula Ϊ,

Formula I

wherein:

R* is azetidinyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, thiazoiyL pyridyl, pyridyl N- oxide, pyrazinyl, pyrimidinyi, pyridazyi, piperidinyL tetrahydropyranyl, phenyl, oxazoiyl, isoxazolyl, tliiophenyl, benzoxazolvL or quinolinyl; wherein said piperidinyL pyridyl, pyridyl N- oxide, imidazolyl, phenyl, thiophenyl, benzoxazolyl, and pyrazolyl are optionally substituted with SO2CH3, C(0)CH 3 , C(Q)NH 2 , CH 3 , CH 2 CH 3 , CF 3 , CI, F, -CN, OCH 3 , N(CH 3 ) 2 , - (CI 1 . · ) )( SCH 3 , OH, C0 2 H, ( '( ) . ■( (CI I : ) :, or OCH 2 OC¾; and optionally substituted with up to two additional substituents independently selected from the group consisting of CI, OCH 3 , and CFI 3 ; and wherein said triazolyl, oxazoiyl, isoxazolyl, and thiazolyl are optionally substituted with one or two CFI 3 groups; and wherein said azetidinyl is optionally substituted with C .:θα ! . C(0)NFI 2 , CH 3 , S0 2 CFI 3 , or C(0)CH 3 ;

R 2 is l -methyl-i ,2,3-triazolyl, pyridyl, pyridyl-N-oxide, 1 -methyl pyrazol-4-yl, pyrimidin-5-yL pyridazyi, pyrazin-2-yl, oxazoiyl, isoxazolyl, iV-acetyI-azetidin-3-yI, iV- methyisuIfonyl-azetidin-3-yl, N-Boc-azetidin-3-yl, N-methyl-azetidin-3-yl, N-acetamidyl- azetidin-3-yl, N-acetyl piperidinyL 1 -H-piperidinyl, N-Boc-piperidinyl, A / -C (1 . 2) alk>4-piperidinyl (including Λ-methyl piperidm-4-yl), thiazo!-5-yl, 1 -methyl imidazol-2-yl, 1 -(3-methoxypropyl)- imidazol-5-yl, or imidazol-5-yl; wherein said 1-C ( i_2 ) alkyl imidazol-5-yl is optionally substituted with up to two additional CH 3 groups, or one substituent selected from the group consisting of 8CH 3 , and CI; and said pyridyl, and pyridyi-N-oxide are optionally substituted with up to two substituents independently selected from the group consisting of C(0)NH 2 , -CN, OCH3, CF3, CI, and CH 3 ; and said thiazol-5-yl, oxazolyl, and isoxazolyl are optionally substituted with up to two CH 3 groups; and said 1 -methyl pyrazol-4-yl is optionally substituted with up to two additional CH 3 groups;

R 3 is i f . OH, OCH 3 , HC I 3 , N(CS ! : ) :. or NH 2 ;

R 4 is H, or F;

R 5 is FI, CI, -CN, ( · : . SCH 3 , OC ( i- 3) alkyl, OFF C (1 . 4} alkyl, N(CH 3 )OCH 3 ,

N(C( j-2)alkyl)2, NH-cyclopropyl, OCHF 2 , 4-hydroxy-piperidinyl, azetidin-l-yl, or fur-2-yl;

R 6 is 2-chloro-thiophen-5-yl, l -rnethyl-pyrazol-4-yl, phenyl, pyrimidinyl, or pyridyl, wherein said phenyl, pyrimidinyl, and pyridyl are optionally substituted with S0 2 CH 3 ,

NFIS0 2 ai 3 , CF 3 , F, CL -CN, OCH 3 , or OCF 3 ;

R 7 is H, CI, -CN, Chalky! OCH 2 CF 3 , OC I FO FOCI F. CF 3 , SCFF, S0 2 CH 3 , ()CHF 2 , NA ! A 2 , C(0)NHCH 3 , N(CH 3 )C!:i2CH 2 NA 1 A 2 , ()( Ί l . -C ' 1 1 ·\.\ : .·\ OCH 2 CFi 2 NFI 2 , ( )C,. : ^ai kyi. OCH 2 -(l -methyl)-im.idazol-2-yl, imidazo!-2-yl, fur-2-yl, pyrazo!-4-yl, pyrid-3-yl, or pyrimidin-

5-yl; thiophen-3-yl, l -methyl-indazol-5-yl, 1 -methyl-indazol-6-yl, phenyl, or ^ wherein said imidazolyl or pyrazolyi can be optionally substituted with a CFI 3 group ;

A 1 is H or C ( i.4 ) alkyl (including CFI 3 );

A 2 is C( ) alkyi (including CFF), cyclopropyl, C i. 4) alkylOC(i 4)alkyl, C i. 4) alkylOH,

1 2

C(0)C(i-2)alkyl, or OCH 3 ; or A and A may be taken together with their attached nitrogen to

R is CH 3 , or phenyl; R 8 is H, CH 3 , OCH 3 , or F:

R 9 is H, or F;

and pharmaceutically acceptable salts thereof:

provided that (4-chloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)(pyridin-3- yi)memanamine, (4-chlorophenyi)(2,4-dichloro-3-(2-chlorophenyl)quinolin-6-y l)(l -methyl- 1H- imidazol-2-yl)methanol, (4-chloro-3-phenylquinoIin-6-yl)(l-methyl-lH-imidazol-5-yl)( pyridin- 4-yl)methanamine, (4-chlorophenyl)(3-(2,6-dichlorophenyl)quinolin-6-yl)( 1 -methyl- 1H- imidazol-5-yl)methanol, (4-chloro-3-phenylquinolin-6-yl)(2-(dimethylamino)pyridin-4- yl)(l ~ methyl- 1 H-imidazol-2-yl)methanoI, 4-(2-((4-chloro-6-((4-chlorophenyl)(hydroxy)(l-m.ethyl-lH- imidazol-5-yl)m.ethyl)-3-phenylqumolin-2-yl)oxy)ethyl)thiomo rpholine 1 , 1 -dioxide, 1 -(2-((4- chloro-6-((4-chlorophenyi)(hydroxy)(l -methyl- lH-imidazol-5-yl)methyl)-3-phenylqu.inolin-2- yl)oxy)ethyl)pyrrolidin-2~one, (2-chloro-4-(dimethylamino)-3-phenylquinolin-6-yl)(pyridin-2 - yl)(pyridin-4-yl)methanol, (4-chloro-3-phenylqumolin-6-yl)(2-fluorop>Tidin-4-yl)(l-m .ethyl-lH- imidazol-2-yl)m.ethanol, (4-chloro-2-(l -methyl- l H-pyrazo l-4-yl)-3-phenylquinolin-6-yl)(4- chlorophenyl)(pyridin-3-yl)m.ethanol, (2,4-dichloro-3-phenylquinolin-6-yl)di(pyridin-2- yl)methanol, 6-((3-chlorophenyl)(hydroxy)(2-(trifluoromethyl)pyridin-4-yl )methyl)-3- phenylquinoline-2-carbonitrile, (2,4-dichloro-8-m.ethyl-3-phenylquinolin-6-yl)(l -methyl- 1H- imidazol-4-yl)(6-m.ethylpyridin-3-yl)methanol, (4-chlorophenyl)(2,4-dichloro-3-(2- chlorophenyl)quinolin-6-yl)(l -methyl- lH-imidazol-5-yl)methanol, (2,4-dichloro-3- phenylquinolin-6-yl)(phenyl)(pyridin-2-yl)methanol, (2,4-dichloro-3-phenylquinolin-6- yl)(oxazol-2-yl)(phenyl)methanol, the second eluting enantiomer of (4-methoxy-3-phenyl-2- (trifiuoromethyl)quinolin-6-yl)(l -methyl- lH-imidazol-5-yl)(pyridin-2-yl)methanol, and the second eluting enantiomer of (4-chloro-2-methoxy-3-phenylquinolin-6-yl)(l -methyl- 1H- imidazol-5-yl)pyrimidin-2-ylmefhanol (when purified on a chiraicei OD column) are excluded from the embodiment.

In another embodiment of the invention:

R* is imidazoivl, thiazolyi, pyridyl, pyrimidinyl, or phenyl; wherein said pviidyl, and said phenyl are optionally substituted with -CN, CF 3 , F, or CI; and wherein said imidazoivl, and thiazolyi are optionally substituted with one or two CH 3 groups; R 2 is 1 -methyl- l ,2,3-triazol-5-yl, N-acetyl piperidin-4-yl, N-Boc-piperidin-4-yl, N-methyl piperidin-4-yll -H-piperidin-4-yl, oxazol-2-yl, 2,4-dimethyl thiazol-5-yi, 1 -methyl imidazol-2-yl, l-methyl-imidazol-5-yl, or pyridyl: wherein said pvridyl is optionally substituted with CF 3 ;

R 3 is Η, ΟΗ :

R 4 is Η;

R 5 is Η, CI, -CN, C( J-4) alkyl, OC (f -2) alkyl, SCH 3 , N(CH 3 ) 2 , or N(CH 3 )OCH 3 ;

R 6 is 2-chloro-thiophen-5-yl, 1 -methyI-pyrazol-4-yl, phenyl, pyrimidinyl, or pyridyl, wherein said phenyl, pyrimidinyl, and pyridyl are optionally substituted with SO 2 CH 3 ,

NHSO 2 CH 3 , CF 3 , (Ί . -CN, OCF 3 , or OCFI 3 ;

R 7 is CI, -CN, CF 3 , SCH 3 , OCFI 2 CF 3 , NA ! A 2 , Ν((Ή : )( Ή H ' Λ

OCH2CFI2OCFI3, ( ) (·Η .( ·| Ι/ΝΛ ; Λ ; . or OCH 2 CH 2 NH 2 ;

A 1 is H, or CH 3 ;

A 2 is OCH3, CH 3 , CH2CH2OH, C(0)Qi. 2) alkyl, or CH 2 CH 2 OCH 3 ; or A 1 and A 2 may be taken together with their attached nitrogen to form a ring selected from the group consisting of:

R 8 is H, or C! :

R 9 is H;

and pharmaceutically acceptable salts thereof;

provided that (4-chlorophenyl)(2,4-dichloro-3-(2-chlorophenyl)quinolm~6~yl )(l -methyl- 1 H- imidazol-5-yl)methanol, 6-((3-ch rophenyl)(hydroxy)(2-(trifluoroinethy] pyridin-4-yl)methyl)- 3-phenylquinoline~2-carbonitrile, (2,4-dichk>ro~3-phenylquinolin~6~yl)di(pyridin~2~yi)metha nol, (2,4-dichloro-3-phenylquinolin-6-yl){phenyl)(pyridin-2-yl)me thanol, (2-chloro-4- (diniethylamino)-3-phenyk}uinolin-6-yl)(pyridin-2-y])(pyridi n^ (4- chlorophenyl)(2,4-(Hchloro-3-(2-chlorophenyl)quinoHn-6-y3)(l -methyl- lH-irr)idazol-2- yl)methanol, (2,4-dichloro-3-phenylquinolin-6-yl)(oxazol-2-yl)(phenyl)met hanol, the second eluting enantiomer of (4-memoxy-3-phenyl-2-(trifiuoromethyl)quinolin-6-yl)(l -methyl- 1Η- imidazol-5-yl)(pyridin-2-yl)methanol, and the second eluting enantiomer of (4-chloro-2- methoxy-3-phenylquinolin-6-yl)(l -methyl- l H-imidazol-5-yl)pyrimidin-2-ylmethanol (when purified on a chiralcel OD column) are excluded from the embodiment.

Another embodiment of the invention is a compound selected from the group consisting of:

and pharmaceutically acceptable salts thereof.

Another embodiment of the invention comprises a compound of Formula I and a

pharmaceutically acceptable carrier.

The present invention also provides a method for treating or ameliorating an RORyt mediated inflammatory syndrome, disorder or disease comprising administering to a subject in need thereof an effective amount of a. compound of Formula Ϊ or a form, composition or medicament thereof.

The present invention provides a method of preventing, treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is selected from the group consisting of: ophthalmic disorders, uveitis, atherosclerosis, rheumatoid arthritis, psoriasis, psoriatic arthritis, atopic dermatitis, multiple sclerosis, Crohn's Disease, ulcerative colitis, ankylosing spondylitis, nephritis, organ allograft rejection, fibroid lung, systic fibrosis, renal insufficiency, diabetes and diabetic complications, diabetic nephropathy, diabetic retinopathy, diabetic retinitis, diabetic microangiopathy, tuberculosis, chronic obstructive pulmonary disease, sarcoidosis, invasive staphylococcia, inflammation after cataract surgery, allergic rhinitis, allergic conjunctivitis, chronic urticaria, systemic lupus erythematosus, asthma, allergic asthma, steroid resistant asthma, neutrophilic asthma., periodontal diseases, periodonitis, gingivitis, gum disease, diastolic cardiomyopathies, cardiac infarction, myocarditis, chronic heart failure, angiostenosis, restenosis, reperfusion disorders, glomerulonephritis, solid tumors and cancers, chronic lymphocytic leukemia, chronic myelocytic leukemia, multiple myeloma, malignant myeloma, Hodgkin's disease, and carcinomas of the bladder, breast, cervix, colon, lung, prostate, or stomach comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form., composition or medicament thereof.

The present, invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is selected from the group consisting of: rheumatoid arthritis, psoriasis, chronic obstructive pulmonary disorder, psoriatic arthritis, ankylosing spondylitis, Crohn ' s disease, and ulcerative colitis.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is selected from the group consisting of: rheumatoid arthritis, psoriasis, chronic obstructive pulmonary disorder, psoriatic arthritis, ankylosing spondylitis, Crohn ' s disease, and ulcerative colitis comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is selected from the group consisting of: rheumatoid arthritis, psoriasis, chronic obstructive pulmonary disorder, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, neutrophilic asthma, steroid resistant asthma, multiple sclerosis, systemic lupus erythematosus, and ulcerative colitis comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof. The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is selected from the group consisting of: rheumatoid arthritis, and psoriasis comprising administering to a. subject in need thereof an effective amount of a. compound of Formula Ϊ or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, in a subject in need thereof comprising administering to the subject an effective amount of the compound of Formula I or composition or medicament thereof in a combination therapy with one or more anti-inflammatory agents, or immunosuppressive agents, wherein said syndrome, disorder or disease is selected from the group consisting of: rheumatoid arthritis, and psoriasis.

The present, invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is rheumatoid arthritis, comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherem said syndrome, disorder or disease is psoriasis comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is chronic obstructive pulmonary disorder comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is psoriatic arthritis comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof. The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is ankylosing spondylitis comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

The present, invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is Crohn's disease comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is ulcerative colitis comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is neutrophilic asthma comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is steroid resistant asthma comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is multiple sclerosis comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof. The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is systemic lupus erythematosus comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

The invention also relates to methods of modulating RORyt activity in a mammal by administration of an effective amount of at least one compound of Formula i.

The present invention provides a method of treating or ameliorating a syndrome, disorder or disease, wherein said syndrome, disorder or disease is selected from the group consisting of: inflammatory bowel diseases, rheumatoid arthritis, psoriasis, chronic obstructive pulmonary disorder, psoriatic arthritis, ankylosing spondylitis, neutrophilic asthma, steroid resistant asthma, multiple sclerosis, and systemic lupus erythematosus comprising administering to a. subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

The present invention provides a method of treating or ameliorating an inflammatory bowel disease, wherein said inflammatory bowel disease is Crohn's disease comprising administering to a subject in need thereof an effective amount of a compound of Formula I or a form, composition or medicament thereof.

The present, invention provides a method of treating or ameliorating an inflammatory bowel diseases, wherein said inflammatory bowel disease is ulcerative colitis comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

DEFINITIONS

The term "administering" with respect to the methods of the invention, means a method for therapeutically or pro hy tactically preventing, treating or ameliorating a syndrome, disorder or disease as described herein by using a compound of Formula I or a form, composition or medicament thereof. Such methods include administering an effective amount of said compound, compound form, composition or medicament at different times during the course of a therapy or concurrently in a combination form. The methods of the invention are to be understood as embracing all known therapeutic treatment regimens.

The term "subject" refers to a patient, which may be an animal, typically a mammal, typically a human, which has been the object of treatment, observation or experiment and is at risk of (or susceptible to) developing a syndrome, disorder or disease that is associated with abberant RORyt expression or RORyt overexpression, or a patient with an inflammatory condition that accompanies syndromes, disorders or diseases associated with abberant RORyt expression or RORyt overexpression.

The term "effective amount" means that, amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human, that is being sought by a researcher, veterinarian, medical doctor, or other clinician, which includes preventing, treating or ameliorating the symptoms of a syndrome, disorder or disease being treated.

As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combinations of the specified ingredients in the specified amounts.

The term "alkyl" refers to both linear and branched chain radicals of up to 52 carbon atoms, preferably up to 6 carbon atoms, unless otherwise indicated, and includes, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentvl, hexyl, isohexyl, heptyl, octyl, 2,2,4-trimethyipentyl, nonyl, decyl, undecyi and dodecyl. Any alkyl group may be optionally substituted with one OCH 3 , one OH, or up to two fluorine atoms.

The term "G a . / ,/' (where a and b are integers referring to a designated number of carbon atoms) refers to an alkyl, alkenyi, alkynyl, alkoxy or cycloalkyl radical or to the alkyl portion of a radical in which alkyl appears as the prefix root containing from a to & carbon atoms inclusive. For example, C ( j-4) denotes a radical containing 1, 2, 3 or 4 carbon atoms.

The term "cycloalkyl" refers to a. saturated or partially unsaturated monocyclic or bicyclic hydrocarbon ring radical derived by the removal of one hydrogen atom from a single ring carbon atom. Typical cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, eyclohexyl, eyelohexenyl, cycloheptyl and cyclooctyl. Additional examples include (¾_ 6 ) Cycloalkyl, C(5..g ) Cycloalkyl, decahydronaphthalenyl, and 2,3 ,4,5,6, 7-hexahydro- 1 H-indenyl. Any cycloalkyl group may be optionally substituted with one OCH 3 , one OH, or up to two fluorine atoms.

As used herein, the term "thiophenyl" is intended to describe the radical formed by removing a hydrogen atom from the molecule with the structure: . PHARMACEUTICALLY ACCEPTABLE SALTS

Pharmaceutically acceptable acidic/anionic salts include, and are not limited to acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, citrate, dihydrochloride, edetate, edisylate, estolate, esyiate, fumarate, glyceptate, gluconate, glutamate, glycoliylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, maiate, maieate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate and triethiodide. Organic or inorganic acids also include, and are not limited to, hydriodic, perchloric, sulfuric, phosphoric, propionic, glycolic, methanesulfonic, hydroxyethanesulfonic, oxalic, 2- naphthalenesulfonic, p-toluenesulfonic, cyclohexanesulfamic, saccharinic or trifSuoroacetic acid.

Pharmaceutically acceptable basic/cationic salts include, and are not, limited to aluminum, 2- amino-2-hydroxymethyl-propane- 1 ,3-diol (also known as tris(hydroxymethyl)aminomet,hane, tromethane or "TRIS"), ammonia, benzathine, /-butylamine, calcium, calcium gluconate, calcium hydroxide, chloroprocaine, choline, choline bicarbo ate, choline chloride, cyelohexylamine, diethanolamine, ethylenediamine, lithium, LiOMe, L-iysine, magnesium, meglumine, N¾, NH 4 OH, iV-methyl-D-glucamine, piperidine, potassium, potassium-r-butoxide, potassium hydroxide (aqueous), procaine, quinine, sodium, sodium carbonate, sodium-2-ethylhexanoate, sodium hydroxide, triethanolamine or zinc,

METHODS OF USE

The present invention is directed to a method for preventing, treating or ameliorating a RORyt mediated inflammatory syndrome, disorder or disease comprising administering to a subject in need thereof an effective amount of a compound of Formula Ϊ or a form, composition or medicament thereof.

Since RORyt is an N-terminal isoform of RORy, it is recognized that compounds of the present invention which are modulators of RORyt are likely to be modulators of RORy as well. Therefore the mechanistic description "RORyt modulators" is intended to encompass RORy modulators as w r ell.

When employed as RORyt modulators, the compounds of the invention may be administered in an effective amount within the dosage range of about 0.5 mg to about 10 g, preferably between about 0.5 mg to about 5 g, in single or divided daily doses. The dosage administered will be affected by factors such as the route of administration, the health, weight and age of the recipient, the frequency of the treatment and the presence of concurrent and unrelated treatments.

It is also apparent to one skilled in the art that the therapeutically effective dose for compounds of the present invention or a pharmaceutical composition thereof wil l vary according to the desired effect. Therefore, optimal dosages to be administered may be readily determined by one skilled in the art and will vary with the particular compound used, the mode of administration, the strength of the preparation, and the advancement of the disease condition. In addition, factors associated with the particular subject being treated, including subject age, weight, diet and time of administration, will result in the need to adjust the dose to an appropriate therapeutic level. The above dosages are thus exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.

The compounds of Formula Ϊ may be formulated into pharmaceutical compositions comprising any known pharmaceutically acceptable carriers. Exemplary carriers include, but are not limited to, any suitable solvents, dispersion media, coatings, antibacterial and antifungal agents and isotonic agents. Exemplary excipients that may also be components of the formulation include fillers, binders, disintegrating agents and lubricants.

The pharmaceutically-acceptable salts of the compounds of Formula I include the conventional non-toxic salts or the quaternary ammonium salts which are formed from inorganic or organic acids or bases. Examples of such acid addition salts include acetate, adipate, benzoate, benzenesulfonate, citrate, camphorate, dodecylsulfate, hydrochloride, hydrobromide, lactate, maleate, methanesulfonate, nitrate, oxalate, pivalate, propionate, succinate, sulfate and tartrate. Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexyl amino salts and salts with amino acids such as arginine. Also, the basic nitrogen- containing groups may be quaternized with, for example, alkyl halides.

The pharmaceutical compositions of the invention may be administered by any means that accomplish their intended purpose. Examples include administration by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal or ocular routes. Alternatively or concurrently, administration may be by the oral route. Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts, acidic solutions, alkaline solutions, dextrose-water solutions, isotonic carbohydrate solutions and cyclodextrin inclusion complexes.

The present invention also encompasses a method of making a pharmaceutical composition comprising mixing a pharmaceutically acceptable carrier with any of the compounds of the present invention. Additionally, the present invention includes pharmaceutical compositions made by mixing a pharmaceutically acceptable carrier with any of the compounds of the present invention.

POLYMORPHS AND SOLVATES

Furthermore, the compounds of the present invention may have one or more polymorph or amorphous crystalline forms and as such are intended to be included in the scope of the invention. In addition, the compounds may form solvates, for example with water (i.e., hydrates) or common organic solvents. As used herein, the term "solvate" means a physical association of the compounds of the present invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. The term, "solvate" is intended to encompass both solution-phase and isolatable solvates. Non-limiting examples of suitable sol vates include ethanolates, methanolates, and the like.

It is intended that the present invention include within its scope polymorphs and solvates of the compounds of the present invention. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the means for treating, ameliorating or preventing a syndrome, disorder or disease described herein with the compounds of the present invention or a polymorph or solvate thereof, which would obviously be included within the scope of the invention albeit not specifically disclosed.

In another embodiment, the invention relates to a compound as described in Formula I for use as a medicament.

In another embodiment, the invention relates to the use of a compound as described in Formula I for the preparation of a medicament for the treatment of a disease associated with an elevated or aberrant RORyt activity.

The present invention includes within its scope prodrugs of the compounds of this invention. In genera], such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", Ed. H. Bundgaard, Elsevier, 1985.

Furthermore, it is intended that within the scope of the present invention, any element, in particular when mentioned in relation to a. compound of Formula L shall comprise all isotopes and isotopic mixtures of said element, either naturally occurring or synthetically produced, either with natural abundance or in an isotopically enriched form. For example, a. reference to hydrogen includes within its scope H, Ή (D), and Ή (T). Similarly, references to carbon and oxygen include within their scope respectively C, iJ C and l4 C and io O and l8 0. The isotopes may be radioactive or non-radioactive. Radiolabelled compounds of Formula I may comprise a radioactive isotope selected from the group of 3 H, l i C, 18 F, 122 J, l23 I, li5 I, 13 Ί, /5 Br, 76 Br, ' 'Br and 8i Br. Preferably, the radioactive isotope is selected from the group of J H, l l C and 18 F.

Some compounds of the present invention may exist as atropisomers. Atropisomers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers. It is to be understood that, all such conformers and mixtures thereof are encompassed within the scope of the present invention.

Where the compounds according to this invention have at least one stereocenter, they may accordingly exist as enantiomers or diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.

Where the processes for the preparation of the compounds according to the invention give rise to mixture of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography. The compounds may be prepared in racemic form., or individual enantiomers may be prepared either by enantiospeeific synthesis or by resolution. The compounds may, for example, be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toluoyl-D-tartaric acid and/or (+)-di-p-toiuoyl-L~tartaric acid followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column.

During any of the processes for preparation of the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protecti ve Groups in Orga ic Chemistr , ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene & P.G.M. Wuts, Protectiv e Groups i Organic Synthesis , John Wiley & Sons, 1991 , The protecting groups may be removed at a convenient subsequent stage using methods known from the art.

ABBREVIATIONS

Herein and throughout the application, the following abbreviations may be used.

A angstrom

Ac acetyl

Ac 2 0 acetic anhydride

Boc tert-butyloxy carbonyl

BHT butylated hydroxytoiuene

br urodu.

Bu butyl

«-BuLi H- butyl lithium

r-BuOH tert-butanol

d doublet

dba dibenzylideneacetone DCE dichloroethane

DCM dichioromethane

Dess-Martin periodinane 1 ,1 ,1 -tris(acetyloxy)- 1 , 1 -dihydro- 1 ,2-benziodoxol-3-( lH)-one

DIEA A',iV-diisopropylethylamine

DMA dimethylacetamide

DME dimethoxyethane

DMF N,N-dimethylformara.ide

DMSO dimethyl sulfoxide

dppf (d ipheny Iph o sphi no) ferrocene

Eaton's Reagent, 7.7 wt¾ phosphorus pentoxide solution in methanesulfonic acid

ED CI A r -(3-dimethylaminopropyl)-iV'-ethylcarbodiiniide hydrochloride

ESI electrospray ionization

Et ethyl

Et 2 0 diethyl ether

EtOAc ethyl acetate

EtOH ethyl alcohol

PCX" flash column chromatography

HATU 0-(7-azabenzotriazoI-l -y )-N,N,N,A T ' -tetramethyluronium hexafluor opho sphate

HPLC high pressure liquid chromatography

Hunig's base N, N-diisopropylethylamine

Hz hertz

/ " -PrOH isopropyl alcohol

KHMDS potassium bis(trimethylsilyl)amide

LCMS liquid chromatography-mass spectrometry

IDA lithium diisopropyiamide

m multiplet

M molar (moles/liter)

mCPBA 3-chl.oroperbenzoic acid

Me methyl

Meldrum's acid 2,2-dimethyl- 1 ,3-dioxane~4,6-dione MeOH methanol

MeONa sodium methoxide

MHz megahertz

min minutes

mL milliliters

MS mass spectrometry

MTBE methyl tertiary butyl ether

m/z mass to charge ratio

nm nanometers

NaO Pr sodium isopropoxide

NBS N-bromosuccinimide

NMP 1 -methyl-2-pyrrolidinone

NMR nuclear magnetic resonance

Ph phenyl

PPA polyphosphoric acid

ppm parts per million

Pr propyl

q quartet

RP-HPLC reverse phase high pressure liquid chromatography s singlet

t triplet

TBAF tetrabutyiammonium fluoride

TEA triethylamine

TEMPO (2,2,6,6-fetramethylpiperidin- 1 -yi)oxidanyl

TEA trifiuoroacefie acid

THE tetrahydrofuran

TLC thin layer chromatography

IJV ultra-violet

X-Phos 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl

GENERAL SCHEMES: Compounds of Formula Ϊ in the present invention can be synthesized in accordance with the general synthetic methods known to those who are skilled in the art. The following reaction schemes are only meant to represent examples of the invention and are in no way meant to be a limit of the invention.

VI! IV {R 5 , R 7 = CS) Scheme 1 describes the preparation of 6-bronio or 6-iodo-quinoiines of Formula IV by various methods (path 1 to 4), In path 1 , cyclization of 4-haloanilines II with commercially available 2- substituted malonic acids ΙΪΪ can be done in refiuxing phosphorus oxychloride to provide 6- haloquinolines IV, wherein R and R are CI, Nucleophilic displacement of 2-chloro substitution with sodium methoxide in hot MeOH or toluene (Alan Osborne et.al. J. Chem. Soc. Perkin Trans. 1 (1993) 181 - 184 and J. Chem. Research (S), 2002, 4) gives 6-halo-2-m.ethoxyquinolines IV. Path 2 illustrates the cyclization of amides VI, derived from acylation of 4-haloanilines II with substituted acid chlorides V (acid chlorides V are either commercially available or prepared from, the corresponding carboxylic acid precursors by procedures known to those skilled in the art), in the presence of DMF in hot phosphorus oxychloride to generate 6-haloquinoIines IV, wherein R "' is H and R' is CI. In path 3, methyl 2-ami obenzoates VII can undergo acylation with acid chlorides V to form an amide intermediate, which can be further treated with a base, such as sodium ethoxide or lithium. bis(trimethylsilyl)amide, to afford 6-halo-4-hydroxyquinolin-2(li?)- ones VOL Conversion of hydroxyquinolin-2(lH)-ones VIII to 2,4-dichloroqumolines IV can be carried out in refiuxing phosphorus oxychloride. Path 4 describes the condensation of anilines II and aldehydes IX in ethanol to form compound X which can be further cyclized in polyphosphoric acid at high temperatures to give quinolinones XL Conversion to the 4- chloroquino lines IV wherein R' is Η can be accomplished in phosphorus oxychloride as previously described.

IV (R 5 , R 7 = CF 3 ) CF 3 )

Scheme 2 illustrates the synthesis leading to 6-bromo or 6-iodoquinolines of Formula IV wherein R 5 is CI and R ' ' is CF 3 (path 1), and 6-iodoquinolines of Formula IV where R J and R' are CF 3 (path 2). In path 1, cyclization of 2-aniinobenzoic acids XII with 1 , 1 , 1 -trifluoropropan- 2-ones XIII in Eaton's reagent at elevated temperatures yields 4-hydroxy-2- mfluoromethylquinoiines XIV, which upon heating in phosphorus oxychloride at temperatures between 100 - 120 °C gives 6-bromo or 6-iodoquinolines IV, wherein R 3 is CI and R' is CF 3 . 6- Iodo-2,4-bis(trifluoromethyl)quinoiines IV can be formed by the reaction sequence illustrated in path 2, Treatment of l-bromo-4-fTuorobenzenes XV with lithium diisopropylamide at - 78 °C followed by addition of ethyl trifluoroacetate provides 2-fiuorophenyl-2,2,2-trifiuoroethanones XVI. Anilines XVII can be prepared by displacing 2-fluoro in XVI with sodium azide followed by reduction with tin (II) chloride dihydrate. Cyclization of XVII with 1 ,1,1 -trifluoropropan-2- ones XIH in the presence of tributylamine in a polar solvent, such as DMF or DMSO, at high temperatures can provide 6~bronio-2,4-bis(trifluoromethyl)quinolines IV. The 6-iodo-2,4- bis(trifluoromethyl)quinolines IV can then be subsequently obtained by conversion of 6- bromoqiiinoline IV, where R 3 and R' are CF 3 , with Nal, Cul, and N,N " '-dimethylethylenediamine in i-BuOH at high temperatures under microwave conditions.

-Pr gC! or EtMgCi

}

1-BuLi

(Ph 3 P) 2 PdCI;

PATH 6 ¾ 1 -B(OH) 2 + R 2 - R R 2

XXXVII Scheme 3 illustrates synthetic routes (path 1 to 6) to ketones of Formula ΧΧΪ wherein R ! and R 2 are as described in the detailed description of the invention. In path 1 , Weinreb amide XX can be prepared from acids ΧΪΧ by reacting with N, 0-dimethylhydroxylamine hydrochloride and 1 ,1-carbonyldiimidazole or with N.O-dimethylhydroxylamnie hydrochloride in the presence of a base such as triethylamine or Hunig's base and a coupling reagent such as EDCI. The amides XX can be further treated with Grignard reagents such as R 2 MgX (X is Br or CI) that can be obtained commercially or preformed by treatment of R 2 Z with organometallic reagents such as - PrMgCI or EtMgCl in THF. Alternatively, Weinreb amides XX can be obtained from, acyl chlorides ΧΧΠ and ,0-dimethylhydroxylamine hydrochloride by using triethylamine or pyridine as a base. 1 -Methyl- IH-imidazole can be treated with one equivalent of «-BuLi and one equivalent of chloro riethylsilane at - 78 °C followed by an additional equivalent of n-BuLi, to which the Weinreb amides XX can be added to yield ketones XXI wherein R" is imidazolyl (path 2).

In path 3, halogen and metal exchange of bromides or iodides XXTV with ?-PrMgCl.LiCl or n- BuLi, followed by addition of aldehydes ΧΧΙΙΪ affords alcohols XXV. Oxidation of XXV with Dess-Martin periodinane or Μη0 2 can provide ketones XXI. In path 4, ketones XXI, where R" is triazolyl, can be prepared by treatment of 1 -methyl- IH-l , 2, 3-triazoie with w-BuLi followed by reaction with aldehydes XXIII to yield alcohols XXV, which could undergo oxidation with Dess-Martin periodinane or Mn0 2 . Path 5 exemplifies the preparation of symmetrical ketones XXI, wherein R * and R 2 are the same. As illustrated, an aryl or heteroaryl group containing an acidic proton XXXIX (Y = R 1 or R" can be deprotonated in the presence of a strong base such as /z-butyl ithium once solubilized in a preferred solvent such as tetrahydrofuran at temperatures between 0 and -78 °C then added in excess to ethyl methoxy(methyl)carbamate to provide ketones XXI wherein R and R " are the same. Aryl or heteroaryl bromide or iodide XL can also be lithiated through a lithium/halogen exchange with «-butyllithium before adding in excess to ethyl methoxy(methyl)carbamate as previously described to provide symmetrical ketones XXI. Path 6, which employs palladium catalyzed cross-coupling of arylboronic acids XXXVII with acid chlorides XXXVIII using K 3 PO 4 as a base and (Pl^ ^PdCl ? , as a catalyst in a high boiling non-polar solvent such as toluene, can also be used to generate ketones XXI. Scheme 4

(R 5 = CF 3J R 7 = CGNH e)

Synthesis leading to intermediate ketones XXVH may also be achieved via chemical routes shown in Scheme 4. In path 1, treatment of 6-bromo or 6-iodoquinolines IV with «-BuLi at, - 78 °C followed by addition of aldehydes ΧΧΙΙΪ provides secondary alcohol quinolines XXVI, which can be oxidized to ketones XXVII with Dess-Martin periodinane or Mn0 2 . Alternatively, ketones XXVII may also be prepared by treatment of 6-haloquinolines IV with «-BuLi at -78 °C followed by quenching with DMF affording carboxaldehydes XXVIII. Ketones XXVII can be obtained in a two-step process by addition of aldehyde XXVIII to a reaction mixture of aryl iodides or bromides XXIX and / ' -PrMgCl-LiCl followed by oxidation with MnO? (path 2). As illustrated in path 3, a one -pot reaction of aldehydes XXX and Grignard reagents such as R - MgCl XVIII followed by treatment with -PrMgCl and addition of 2,2,2-trifluoro-N-methoxy-N- methylacetamide yields hydroxyl compounds XXXI. The hydroxyl group can be oxidized using bleach and TEMPO. Fluoro displacement can then be achieved with ammonia in hot DMSO to provide anilines XXXII. In the presence of benzenesulfonic acid, condensation of anilines XXXII and 2-(methylimino)butanamides XXXIII in hot DMSO furnishes ketoquinolines XXVII wherein R 5 is CF 3 and R 7 is CONHMe.

Scheme 5 illustrates synthetic routes leading to compounds of Formula I (path I to 3). As illustrated in path 5 , a mixture of the 6-bromo or 6-iodoquinolines IV in an appropriate solvent such as TFIF can be either prernixed with the ketones XXI at -78 °C followed by addition of n- BuLi or can be pretreated with «-BuLi at -78 °C prior to the addition of the ketones XXI to afford the tertiary alcohols of Formula I, wherein R 3 is OH. In path 2, 6-iodoquinolines IV can be treated with /-PrMgCl followed by addition of ketone XXI to yield compounds of Formula I wherein R is OH. As shown in path 3, halogen-metal exchange of aryl halides (iodide or bromide) XXIV with an organometallic reagent, such as «-BuLi, /-PrMgCLLiCl, or EtMgCl, at an appropriate temperature, such as -78 °C or 0 °C, followed by reaction with ketones XXVII may afford tertiary alcohol quinolines of Formula. Ϊ. Compounds of Formula Ϊ wherein R is N- Boc piperdinyl can be deprotected under acidic conditions using standard procedures known in the art then further functionalize on nitrogen by treatment, with an anhydride or acylating agent such as acetylchloride to provide compounds of Formula I wherein R z is N-acetyl piperidin-4-yl.

I (R 5 , R 7 = CI) f (R 5 is substituted amine and amines)

hydroxy! reagents

I RS Q _ R 7 subs ituted hydroxy!

Scheme 6 illustrates methods used to synthesize compounds of Formula I wherein either the chlorine at R ? or at both R 3 and R' positions are replaced with nitrogen, oxygen, sulfur or aikyl groups. In path 1, 4 and 5, nucleophilic displacement of 2,4-dichloroquinolines I (R 3 and R ? are CI) with NaO(alkyl), NaS(alkyl), such as NaOMe, NaSMe, NaOEt, or NaO'Pr, in an appropriate solvent, such as MeOH, EtOH, -PrOH or DMF at elevated temperatures or with substituted hydroxy reagents such as 2-methoxyethanoi, 2,2,2-trifruoroetha.nol or amine substituted hydroxyl reagents in the presence of a base like sodium hydride in a. non-polar solvent such as toluene provides compounds of Formula Ϊ wherein R " is CI and R' is Q(alkyl), S(alkyl), 0(( ' l ! . ■) h. OCH 2 CF 3 , or 0(CH 2 ) 2 NA A " wherein A and A" are defined in the detailed description of the invention and compounds of Formula Ϊ wherein R ~ and R' are O(alkyl) or S(alkyl). Likewise, nucleophilic displacement of 2,4-dichloroquinolines I (R s and R' are CI) with primary or secondary alkyl amines, heterocycle amines, or N, 0-dimethylhydroxylamine in polar solvents such as MeOFI, EtOH, or Et 2 NCHO, or DMF provides quinolines of Formula ί (path 2) wherein R 5 is NFI(alkyl), N(alkyl) 2 , N(CH 3 )OCH 3 , or CI, and R 7 is NFI(alkyl), N(alkyl) 2 , (CH 3 )OCH 3 , NA*A 2 HC 2 -3)aikyl A ! A 2 or N(CH 3 )C( 2 4 ) alkylNA i A 2 , wherein A 1 and A 2 are as defined above. Replacement of chlorine at positions 2 and 4 of quinolines I (R 5 and R' are CI) with alkyl groups could be carried out using Zn(alkyl) 2 in the presence of K. 2 C0 3 and a palladium catalyst, such as PdCl 2 (dppf), to afford 2-alkyl and 2,4-dialkylquinolines I (path 3).

Synthetic routes to compounds of Formula I, wherein R 5 is CI or CN, and R ? is CN or arvi, are illustrated in Scheme 7, In path 1 , cyanation of the 2,4-dichloroquinolines Ϊ with Zn(CN) 2 in the presence of Zn, a palladium catalyst, such as Pd 2 (dba)3, and a. ligand, such as dppf or X-phos, at high temperatures can provide 2-CN and 2,4-diCN quinolines I. The 2,4-dichloroquinolines I can also undergo a Suzuki reactions with ArB(OH) 2 or ArB(OR) 2 and a palladium catalyst, such as PdCl 2 (dppf), yielding compounds of Formula Ϊ wherein R' is phenyl, substituted phenyl and five or six-membered heteroarvls such as furan, pyridine, pyridazine, pyrazine, pyriniidine, pyrrol, pyrazole or imidazole (path 2).

S he i;¾ 8

As illustrated in Scheme 8, compounds of Formula Ϊ wherein R 5 is a chlorine can be further substituted by treatment with alkylhoronie acids or esters under Suzuki reaction conditions (path 1), with sodium alkoxides (path 2), or with zinc cyanide (path 3) using conditions previously described to pro vide compounds of Formula Ϊ wherein R 5 is alkyl, O(alkyl) or CN and R ' is as described above.

In Scheme 9, tertiary alcohols Ϊ can he treated with base, such as NaH, and alkylated with Mel in DMF to provide compounds of Formula I wherein R 3 is OMe.

Synthetic routes to compounds of Formula Ϊ, wherein R " is ¼, are illustrated in Scheme 10. Ketimines XXXV may be prepared by Ti(OEt) 4 mediated condensation of ketones XXI with 2- methyipropane-2-sulfinamide in refluxing THF. Addition of «-BuLi to the reaction mixture of ketimines XXXV and 6-bromo or 6-iodoquinolines IV at - 78 °C followed by cleavage of tert- butanesulftnyl group with HC1 in eOH liberates amines I. Alternatively, compounds of Formula I, wherein R 3 is OH, can be treated with sodium hydride followed by addition of acetic anhydride or acetyl chloride and stirred at room temperature over a 24 to 72 hour period to provide the intermediate acetate wherein R J is OAc, The acetate can then be combined with a solution of ammonia in methanol and heated at temperatures between 60 and 85 °C to provide compounds of Formula Ϊ, wherein R is NH 2 .

Scheme 11

As shown in Scheme 1 1 , the quinolines of Formula Ϊ wherein R' is CN can be hydrolyzed as described in US20080188521 by treatment with sodium carbonate and hydrogen peroxide to provide compounds of Formula I wherein R ? is CONS¾ (path 1) or can be treated with a strong acid like HQ to convert CN to a carboxylic acid XXXIV (path 2), Once formed the acid can be further coupled to substituted amines using appropriated coupling reagents such as EDO or HATU in the presence of a. base such as triethylamine or Hunig's base to provide compounds of Formula Ϊ wherein R ? is CONA'A 2 .

Scheme 12

I (R 5 = CI, R 7 = CH 2 OC (2-3) aIk l A A 2 )

Synthesis of compounds of Formula I, wherein R is an aminoalkylaminomethylene or an aminoalkoxymethylene can be prepared from 2-methylquinolines as shown in Scheme 12. Bromination of 2-methylquinolines of Formula I can be accomplished with N-bromosuccinimide in acetic acid at elevated temperatures as described in WO2010151740, to provide the methylbromide intermediate XXXVL Nuc!eophilie displacement of the bromide under basic conditions using procedures know n in the art could afford compounds of Formula Ϊ wherein R 7 is -CH 2 NHC ( 2-3 ) alkylNA ! A 2 or (path 1) or CH 2 OC( 2- 3 ) alkylNA ] A 2 (path 2) and A' and A are defined above.

Compounds of Formula I wherein R 1 , R~ or R 6 are pyridy! can be treated with m- chloroperbenzoic acid in a chlorinated solvent at ambient to 40 °C to form the pyridyl-N-oxides of Formula L

As shown in Scheme 13, compounds of the Formula Ϊ wherein R " is H can be prepared by treating compounds of Formula I wherein R J is OH with a hydride source such as triethylsiiane and an acid such as trifluoroacetic acid in a. solvent such as dichloromethane at room temperature or with heating (WO2009091735).

Scheme 14 outlines alternative synthetic methods to compounds of Formula i. Acylation of methyl 2-aminobenzoates VII (path 1) or 2-trifluoroketoanilines XVII (path 2) with benzyloxyacetyl chloride in the presence of a base such as triethylamine in a solvent such as dichloromethane can afford amides XLi and XLV respectively. Amides XLi and XLV can undergo an intramolecular cyclization reaction with a base such as potassium bis(triniethylsilyl)aniide in a solvent such as tetrahydrofuran to provide the intermediate 6- haloquinolin-2(lH)-ones which can be converted with phosphorus oxychloride, as previously described, to compounds of Formula IV wherein R and R' is CI (resulting from path 1 ) and R " is CF 3 and R' is CI (resulting from path 2). The coupling of 6-haloquinolines IV and ketones of Formula XXI to introduce R 1 and R followed by displacement of the 2 or 4-chloro using procedures previously described provides quinolines of Formula XLII wherein R 1 , R , R 5 and R ' are defined in the detailed description of the invention. Palladium-catalyzed hydrogenation of compounds of Formula XLII that are substituted with a benzyloxy at C-3 can provide intermediate quinolin-3-ols XLIM. The quinolin-3-ols XLIII can be converted into the corresponding inflates XLIV with trifSuoromethanesulfonic acid in the presence of a base, such as pyridine, in a solvent such as dichloromethane. The inflates XLIV can be converted into compounds of Formula I, wherein R 6 is aryl or heteroaryl as defined above, by a palladium- catalyzed cross coupling with organoboron reagents of the formula R 6 B(OR) 2 in the presence of a base, such as potassium carbonate, in a solvent mixture such as 1 ,4-dioxane/water.

ili (R 6 = OBn)

(Z = I or Br)

CI and R 6 = OBn)

(CF 3 S0 2 ) 2 0 pyridine, DCM

1 (R 6 = aryS or he aryi) XLIV EXAMPLES

Compounds of the present invention can be prepared by methods known to those who are skilled in the art. The following examples are only meant to represent examples of the invention and are in no way meant to be a limit of the invention. intermediate 1: step a

feri- Butyl 4-(hydro3i-y(l-methyl-lH-imidazol-5-yl)methyl)piperidine-l-c arbox ate

A. solution of 5-bromo-l -methyl- IH-imidazole (25.0 g, 155 mmol; dried over 3.4 molecular sieves, then filtered) in DCM (310 mL) was stirred in an ice bath while / ' PrMgCl (72 mL, 2.01 M solution in THF, 145 mmol) was added rapidly dropwise under argon via pressure-equalizing addition funnel . Residual /PrMgCl was rinsed down with 50 ml. THF, and the ice bath was removed and the reaction stirred for 25 minutes. A solution of eri-butyl 4-formylpiperidine-l - carboxylate (27.6 g, 130 mmol ) in THF (65 mL) was added dropwise over ~5 minutes via pressure-equalizing addition funnel at, room temperature. After stirring 1 hour at room temperature, the yellow mixture was quenched with 5 M aqueous NH C1 (250 mL) in one portion. The organic layer was dried (Na 2 S0 4 ), filtered, and concentrated to provide the crude title compound as a clear light amber oil.

A homogeneous solution of tert-butyl 4-(hydroxy(l -methyl- 1 H-imidazo l-5-yl)methyl)piperidine- 1-carboxylate (32,2 g, 109 mmol, Intermediate 1 : step a) in 1 ,4-dioxane (436 mL) was treated with n0 2 (47,6 g, 547 mmol) and stirred at 100 °C open to the air overnight (17 hours). Since the reaction was only ~5()% complete by NMR, the reaction was cooled to room temperature and additional Mn0 2 was added (48.0 g, 552 mmol) and the reaction stirred open to the air at 100 °C for 6,5 hours, then at room temperature for 18 days, then filtered through a pad of Celite^ and the black filter cake washed with EtOAc. The crude filtrate was treated with a third portion of Mn0 2 (28.5 g, 327 mmol) and stirred at room temperature overnight. The reaction was then filtered as above and concentrated to provide the crude title compound as a clear dark yellow oil. This oil was purified by FCC with an EtOAc to 50% acetone/EtOAc gradient to provide the title compound as a clear dark yellow oil.

Intermediate 1 : step c l-(4-(l-Methy1-lHf-imidazoIe-5-carbony])piperidiii-l-yi)etha none

A homogeneous yellow solution of tert-butyl 4-(l -methyl- lH-imidazo le-5-carbonyl)piperidine- 1-carboxylate (10.1 g, 34.4 mmol, Intermediate 1 : step b) in DCM (1 72 mL) was treated with TFA (26.4 mL, 344 mmol) and stirred at room temperature for 2.5 hours. The reaction was concentrated from toluene (2 x 100 mL), and the resulting clear light amber residue was taken up in DCM (344 mL) and TEA (23.9 mL, 172 mmol). Acetic anhydride (3.91 mL, 41.3 mmol) was added dropwise and the reaction stirred at room temperature for 1 hour. The reaction was concentrated under high vacuum and the residue was purified by FCC using 95:5 DCM/ΜεΟΗ with 2% TEA as eluent. The combined fractions were concentrated, dissolved in DCM (200 mL), and washed with water (2 x 200 mL) to remove TEA, The organic layer was dried (Na 2 S0 4 ), filtered, and concentrated. The residue was triturated with MTBE (75 mL) at reflux for 15 minutes and then allowed to cool to room temperature. The mixture was filtered and the off-white filter cake was washed with MTBE (2 x 3 mL) to provide, after air drying at 100 °C, the title compound as an off-white fine powder.

Intermediate 2: step a

0-(TrifiMorometiiyI)¾ie0tiiioy! chloride

To a 1 L 3-neck flask equipped with an overhead stirrer, Claisen adaptor, nitrogen bubbler, 60 mL addition funnel, and thermocouple was added 6-itrifluorometh i)nicotimc acid (45 g, 235.5 mmol), dichloromethane (540 mL) and DMF (0.910 mL, 1 1.77 mmol) via syringe. To this solution was added oxaiyl chloride (24.51 mL, 282.56 mmol) and the reaction was allowed to stir at ambient temperature overnight. The reaction was then filtered and the clear filtrate was concentrated in vacuo to afford the title compound as a brownish semisolid.

Intermediate 2: step b

A-Methoxy-A-meihyI-6-(triflHoromethyl)ok^ tinamide

To a 1 L 3-neck flask equipped with an overhead stirrer, Claisen adaptor, nitrogen bubbler, 125 mL addition funnel, and thermocouple was added 6-(triiiuoromethyl)nicotinoyl chloride (49.3 g, 235.2 mmol, Intermediate 2: step a), dichloromethane (493 mL), and N,0- dimethylhydroxylamine hydrochloride (25.63 g, 258.8 mmol). After the mixture was cooled to 7 °C, diisopropylethylaniine (90.26 mL, 517.6 mmol) was added such that the addition temperature did not exceed 16 °C. After the additio , the reaction was allowed to warm to room temperature. The reaction was then transferred to a separatory funnel and the organic layer was washed with saturated aqueous NaHCC (2 x 100 mL) followed by water (100 mL) and then dried over sodium sulfate, and filtered. The solvent was then removed in vacuo to afford the title compound as a brownish oil.

inter mediate 2: step e

(i- ethyl-l// midazol-5-yl)(6-(trifluoromethyI)pyridin-3-yi)methan

To a 3 L 4-neck flask equipped with an overhead stirrer, nitrogen bubbler, and thermocouple was added 5-bromo- 1 -methyl- 1 H-imidazole (47.96 g, 297.9 mmol), followed by TI1F (537 mL). To this room temperature solution was added isopropylmagnesium chloride/lithium chloride complex [1 .3 in THF] (246.8 mL, 320.8 mmol) (addition temperature maintained between 16.6 and 25 °C) to afford a milky suspension and the reaction was stirred for 60 minutes and then cooled to 5.3 °C in an ice bath. To this mixture was added a solution of N-methoxy-N-m.ethyl-6- (irifiuoromethyl)nicotinamide (53.66 g, 229.14 mmol, Intermediate 2: step b) in THF (268.3 mL) (addition temperature between 5.3 and 5.6 °C) to afford an orange mixture. After addition, the reaction was warmed to room temperature over 2 hours. After stirring at room temperature for 18 hours, THF (200 mL) was added and the reaction was stirred for 2 hours. The reaction was then cooled to 4 °C with an ice bath and carefully quenched with 2 N aqueous HCl to pH = 7, quenching temperature reached 12 °C. The mixture was diluted with ethyl acetate (500 mL), phases split and the organic layer was washed with brine (2 x 200 mL), dried over sodium sulfate, filtered, and the solvent was removed. Hot ether was added and suspension was filtered to provide the title compound as a solid.

Intermediate 3 -Brosiio-2,4-ciichIoro-8-iMethyl-3-p!ieiiylqiiiiioliHe

A mixture of 2-phenylmalonic acid (7.62 g, 42.3 mmol) and POCI 3 (32.8 mL, 352 mmol) was stirred at reflux (130 °C) for 10 minutes, and the resulting homogeneous yellow solution was cooled in an ice bath. 4-Bromo~2-methylaniline (6.56 g, 35.2 mmol) was added in one portion and the mixture was refluxed for 2 hours. The dark solution was allowed to cool to room temperature and was diluted with DCM (70 mL) and ice (100 raL), and stirred at roora temperature for -5-10 minutes at which point exothermic PO(¾ hydrolysis commenced (ice bath cooling), and was then stirred at room temperature for another 30 minutes. The light yellow aqueous layer was extracted with DCM (I x 30 mL), and the combined dark homogeneous organic layers were dried (Na 2 S0 4 ), filtered, and concentrated with silica gel. The silica- adsorbed residue was dry load flash chromatographed with a 20% DCM/heptane to 100% DCM gradient to provide the title compound as an off-white solid.

Intermediate 4 l~(4~Benzoylpiperidm-l-yl)ethaiione

A. mixture of phenyl(piperidin-4-yl)methanone hydrochloride (743 mg, 3.29 mmol) in dichloro methane (13.2 ml.) and triethylamine (1.10 mL, 7.90 mmol) was treated with Ac 2 0 (0.373 mL, 3.95 mmol) dropwise over 1 minute in an ice bath under argon, and the resulting translucent mixture was immediately removed from the ice bath and stirred at room temperature overnight. The reaction was then extracted with 1 M aqueous HC1 (1 x 8 mL) and 1 M aqueous NaOH (1 x 8 mL), and the organic layer was dried (Na 2 S0 4 ), filtered, and concentrated to provide the title compound as a translucent beige oil that crystallized upon standing. 2-(3-(Trifluorometfayl)pheiiyl)acetyl chloride

To a solution of 2-(3-(trifluoromethyl)phenyl)acetic acid (4.78 g, 23.4 mmol) in DCM (46 niL) was added oxalyl chloride (12.9 mL, 25.8 mmol). One drop of DMF was then added and the mixture was stirred for 1.5 hours at room temperature. The mixture was concentrated to afford the title compound.

Intermediate 5: step b

Methyl 5-bromo-2-(2-(3-(trifl¾ioromethyl)phenyI)acetamido)ben oate

To a solution of 2-(3-(trifj.uorom.ethyl)phenyl)acety3. chloride (23.4 mmol assuming quantitative yield in previous step, Intermediate 5: step a) in DCM (50 mL) in an ice bath was added methyl 2-amino-5-bromobenzoate (4.90 g, 21 .3 mmol) followed by triethylamine (6.51 mL, 46.8 mmol). The mixture was stirred at room temperature overnight. The reaction mixture was washed with saturated aqueous NH 4 C1 then water. The organic phase was dried (Na 2 S0 4 ), filtered, and concentrated and the residue was purified by flash column chromatography (50-70% DCM- heptanes). Some of the title compound was obtained in pure form from the column, along with mixed fractions containing the title compound and the starting aniline. The mixed fractions were left to stand in DCM:heptanes ( 1 : 1) overnight, forming crystalline needles of the title compound, which were collected by vacuum filtration and combined with clean fractions from the column. intermediate 5 S i ' tep c

6-Bromo-4-hydroxy-3-(3-(trifluoroniethyl)phenyl)quinolin- 2(lH)-one

To a solution of methyl 5-bromo-2-(2-(3-(trifluoromethyl)phenyl)acetamido)benzoate (3.86 g, 9.28 mniol, Intermediate 5: step b) in THF (100 mL) at -78 °C was added KHMDS (0.5 M in toluene, 55.6 mL, 27,8 mmol) over 8 minutes. The resulting light yellow solution was stirred at - 78 °C for 5 minutes, then was transferred to an ice bath and stirred for 1 hour 15 minutes. The mixture was diluted with water and extracted once with EtOAc. The aqueous phase was acidified to pH 2 by addition of 1 N aqueous HC1 solution. A white precipitate formed upon acidification and was collected by vacuum filtration and air dried to afford the title compound as a white powder.

Intermediate 5: step d

6-Bromo-2,4-dichIoro-3-(3-(trifluorometh i)phenyl)quinoline

To a suspension of 6-brom.o-4-hydroxy-3-(3-(trifiuoromethyl)phenyl)quinolin-2(l H)-one (3.17 g, 8.25 mmol, Intermediate 5: step c) in acetonitrile (35 mL) was added POCI 3 (2.31 mL, 24.8 mmol). The resulting white suspension was heated at reflux under a drying tube for 2.5 hours. The mixture was allowed to cool to room temperature and stand for 2 hours. Precipitated solid was collected by vacuum filtration to afford the title compound as a white solid. intermediate 5 S i ' tep e

6-Bro8BO-4-chloro-2-met!ioxy-3-(3-(triflMoromet!iyI)phes yl)qMi8iolliie

To a suspension of 6-bromo-2,4-dichloro-3-(3-(trifluoromethyl)phenyl)quinoline (1.48 g, 3.52 mmol, Intermediate 5: step d) in toluene (15 ml,) in a sealed tube was added sodium methoxide (1.90 g, 35.2 mmol) in one portion. The resulting white suspension was heated in a 100 °C oil bath for 24 hours. An aqueous NaHCX¾ solution (10 wt. %, 27 mL) was added and the mixture was stirred for a few minutes, and the phases were separated. The aqueous phase was extracted twice with EtOAc. The organic extracts were washed with saturated aqueous NaCi. The organic phase was dried (Na 2 S0 4 ), filtered, and concentrated. The crude product was purified by flash column chromatography (silica gel, 15-30% DCM-heptanes) to afford the title compound as a white powder,

intermediate 6: step a (l~Meih l-lH-l,2 -triazol^

A solution of 1 -methyl- lH- l ,2,3-triazole (1 .47 g, 17.7 mmol, PCT Int. Appl. 2008098104) in 20 mL THF was cooled to -40 °C in a dry-ice/acetonitrile bath. «-Butyllithium (1.6 M in hexane, 10.2 mL, 16.3 mmol) was added dropwise via syringe and the mixture was stirred at -40 °C for 30 minutes. A solution of 1 -methyl- 1 H-imidazole-5-carbaldehyde (1.50 g, 13.6 mmol) in 10 mL THF was then added and the mixture was stirred for 5 minutes, then was transferred to an ice/water bath. After 1 hour, the mixture was quenched by addition of saturated aqueous NPLCl, diluted with water, and extracted twice with EtOAc. The aqueous phase, which contained the title compound, was then concentrated. The residue w r as purified by flash column chromatography (silica gel, gradient 3-10% MeOH-DCM) to afford the title compound as a light yellow foam.

Inter mediate 6: step b

(l-Methy!~lH~l,2 ~triazol~5~yl)(l-met!iyl-lH-imidazol~S~yl)metii¾iiO!ie

A mixture of (l-methyl-lH-l,2,3-triazoi-5-yl)(l-methyl-lH-imidazoi-5-yl)m ethanol (1.90 g, 9.83 mmol, Intermediate 6: step a) and manganese dioxide (5,04 g, 49.3 mmol) in 1 ,4-dioxane (100 mL) was stirred in a 100 °C oil bath under argon for 2 hours. The mixture was allowed to cool to room temperature, then was filtered through a pad of Celite*, rinsing with DCM. The filtrate was concentrated, yielding the title compound as a brown powder.

Intermediate 7: step a

Methyl 5~hromo~2~(2-pheny ] acetamido)benzoate

To a mixture of methyl 2-amino-5-bromobenzoate (9.00 g, 39.1 mmol) and Et N (7,6 mL, 54.8 mmol) in CH 2 CI 2 (90 mL) at 4 °C was added 2-phenylacetyl chloride (7.26 g, 46.9 mmol) dropwise. After completion of the addition, the cooling bath was removed and the mixture was stirred for 27 hours. TLC showed that some of the starting material methyl 2-amino-5- bromobenzoate still remained. More 2-phenyiacetyl chloride (1.88 g, 12.2 mmol) and Et 3 N (2.2 mL, 15,9 mmol) were added, and the mixture was stirred overnight. 2 C0 3 (aqueous) was added, the organic layer was separated, and the aqueous layer was extracted with CH 2 C1 2 . The combined organic layers were washed with water, dried (Na 2 S0 4 ), filtered, and concentrated in vacuo. CH 3 CN (100 niL) was added, and the precipitated solid was filtered, washed with Et 2 0, and dried to afford the title compound. The filtrate was concentrated in vacuo, and the solid was filtered, washed with Et 2 0, and dried to provide additional title compound.

Inter mediate 7: step b

6-Bromo-4-hydroxy-3-phenylquinoIin-2(lH)~oiie

To a solution of methyl 5-bromo-2-(2-phenylacetamido)benzoate (7.71 g, 22.1 mmol, Intermediate 7: step a) in THF (50 niL) at -78 °C was added 1.0 M lithium bis(trirneth lsilyl)arnide in hexane (48.7 raL, 48.7 mmol) slowly, and the color changed from colorless to clear red. The mixture, at -78 °C, was warmed to room temperature over 4 hours, during which time the color changed to cloudy yellow. The reaction was quenched with water, and acidified with 37% HC1 until pH ~ 5. The precipitated solid was filtered, washed with water and Et 2 0, and air dried to provide the title compound. Another crop precipitated from the filtrate after standing overnight. The solid was collected by filtration, washing with water and Et 2 0, and air drying to afford additional title compound. loter mediate 7: step c

~Br mo~2,4-dicMoro~3~p!u 5 s¾ylqsiMolme

A solution of 6-bromo-4-hydroxy-3-phenylquinolin-2(lH)-one (8.50 g, 26.9 mmol, Intermediate 7: step b) in phosphoryl trichloride (51 ml,, 547 mmol) was heated at 107 °C for 3.5 hours, and then cooled to room temperature. After evaporation of POCI3 in vacuo, concentrated ΝΗ 4 ΟΗ (aqueous) was added dropwise at 4 °C until H 9. The precipitated solid was filtered, washed with water, and dried at 50 °C under vacuum overnight to provide the title compound.

Pyridine (27.6 mL, 343 mmol) was added to N.O-dirneth lhydroxylamine hydrochloride (56.7 g, 172 mmol) in DCM (400 mL). 4-Chloro benzoyl chloride (20 mL, 156 mmol) was then added and the mixture was stirred at room temperature for 3 clays. Solids were removed by vacuum filtration, washing with DCM. The filtrate was washed with 1 N aqueous HC1 followed by water. The organic phase was dried (Na 2 S() 4 ), filtered, and concentrated, affording the crude title compound as a colorless liquid which was used without purification.

Intermediate 8: step b

-Chlorophenyi)(i-methyHH-imidazoi-5-yI)methanone

Ethyl magnesium bromide (3,0 M in diethyl ether, 21 .5 mL, 64.4 mmol) was added via syringe over a few minutes to a clear colorless solution of 5-brom.o-l -methyl-lH-imidazole (10.4 g, 64.4 mmol) in TFIF (100 mL) under a nitrogen atmosphere in an ice bath, A white precipitate formed during the addition. The mixture was removed from the ice bath and was stirred for 20 minutes, then was again cooled in an ice bath before addition of 4-chloro-N-methoxy-N-methylbenzamide (10.7 g, 53.6 mmol, intermediate 8: step a). The resulting white suspension was stirred overnight at room temperature. The reaction was quenched by addition of saturated aqueous ΝΗ 4 Ο and diluted with water. The mixture was partially concentrated to remove THF and was diluted with DCM. The mixture was acidified to pH 1 with 1 N aqueous HQ, then neutralized with saturated aqueous NaHCC . The phases were separated and the aqueous phase was further extracted with DCM. The organic extracts were washed with water, then were dried ( a 2 S0 4 ), filtered, and concentrated, affording a white solid. The crude product was triturated with a mixture of EtQAc: heptanes (1 : 1 , 150 mL). The precipitated solid was collected by vacuum filtration, washing with heptanes, to afford the title compound. ί «ter mediate 9 step a

Methyl 5-bromo-2-(2-(pyridin-2- )acetamido)beiizoate

Into a 250-mL round-bottom flask was placed a solution of methyl 2-amino-5-bromobenzoate (5 g, 21.73 mmol), 2-(pyridin-2-yl)acetic acid hydrochloride (4.5 g, 25.92 mmol), HATU (10 g, 26.30 mmol) and D1EA (8.5 g, 65.77 mmol) in /V,N-dimethylformamide (100 mL). The resulting solution was stirred overnight at 20 °C. The reaction was then quenched by the addition of 100 mL of water. The resulting solution was extracted with 3x100 mL of dichloromethane and the organic layers combined and dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/mefhanol (1 :50) to afford the title compound as a red solid.

intermediate 9: step b

6-Bromo-3-(pyridin-2- )quinoline-2,4(lH, 3H)-dione

Into a 250-mL round-bottom flask was placed a solution of methyl 5-bromo-2-[2-(pyridin-2- yi)aceta.mido]benzoate (3 g, 7.73 mmol. Intermediate 9: step a) and MeONa (11.7 mL, 31 mmol, 2.64 M in MeOH) in tetrahydrofuran (30 mL). The resulting solution was stirred overnight at 20 °C. Then, the solvents were removed by a rotary evaporator and 50 mL water was added to the residue. The pH of the resulting mixture was adjusted to 7 with 1 M aqueous HCl. The solid was collected by filtration and dried under vacuum to afford the title compound as a white solid which was used in the next step without further purification. Intermediate 9: step c

6-Bromo-2,4-dichloro-3-(pyridHi-2-yl)qMi8iolliie

Into a 100-mL round-bottom flask was placed a solution of 6-bromo-3-(pyridin-2-yl)quinoline- 2,4(1H, 3H)-dione (2.54 g, 7.21 mmol, Intermediate 9: step b) in POCI 3 (50 mL). The resulting solution was stirred for 3 hours at 120 °C. The reaction was then quenched by the addition of 50 mL of water. The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate, filtered and concentrated to afford the title compound as a white solid. intermediate 10: step a

Methyl 5-bronio-2-(2-(pyridin-3-yI)acetamido)benzoate

Into a 250-mL round-bottom flask was placed a solution of methyl 2-amino-5-bromobenzoate (5 g, 21.73 mmol, 100%), 2-(pyridin-3-yl)acetic acid hydrochloride (4.5 g, 25.92 mmol), HATU (10 g, 26.30 mmol 100%) and DIEA (8.5 g, 65.77 mmol, 100%) in N,N-dimethylformamide (100 mL). The resulting solution was stirred overnight at 20 °C. The reaction was then quenched by the addition of 100 mL of water. The resulting solution was extracted with 3x100 mL of dichioro methane and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :2-1 : 1) to provide the title compound as a yellow solid. intermediate 10 S i ' tep b

6-Bromo-3-(pyridin-3-yl)qumoline-2,4(l J, 3//)-dione

Into a 100-mL round-bottom flask, was placed a solution of methyl 5-bromo-2-[2-(pyridin-3- yl)acetamido]benzoate (3 g, 7.73 mmol, Intermediate 10: step a) and MeONa (1 1.7 mL, 31 mmol, 2.64 M in MeOH) in tetrahydrofuran (30 mL). The resulting solution was stirred overnight at 20 °C. The solids were collected by filtration and washed with water (3 x 5 mL). The solids were dried under vacuum to provide the title compound as a white solid,

intermediate 10: step e

6-Bro8BO-2,4-dic!iIoro-3-(pyridiii-3-yl)qss!8iolHie

Into a 100-mL round-bottom flask, was placed a solution of 6-bromo-3-(pyridin-3-yI)quinoline- 2,4(1H, 3H)-dione (700 mg, 1 .99 mmol, Intermediate 10: step b) in phosphorus trichloride (20 mL). The resulting solution was stirred for 3 hours at, 120 °C. The resulting mixture was concentrated under vacuum. The reaction was then quenched by the addition of 20 ml. of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate, filtered and concentrated to afford the crude title compound as a white solid.

Intermediate 11: step a

Methyl 2-(2-(benzyloxy)acetamido)-5-bromobeiizoate

To a solution of methyl-2-amino-5-bromobenzoate (15 g, 62.6 mmol) in DCM (241 niL) at 0 °C was added benzyloxyacetyl chloride (12.5 mL, 75.1 nimol) followed by Et N (20 niL, 144 mmol) dropwise. The resulting white suspension was stirred at room temperature for 3 hours. The mixture was then washed with saturated aqueous NH 4 C1 (200 mL) followed by water (200 mL). The organics were dried (Na 2 S0 4 ), filtered and concentrated to dryness. The crude solid was triturated with MeOH (90 mL) and dried under vacuum to afford the title compound as a white solid.

Intermediate 11: step b

3-(Benzyloxy)-6-bromo-4-hydroxyq«inoIm-2(l /)-one

To a solution of methyl 2-(2-(benzyloxy)acetamido)-5-bromobenzoate (15 g, 39.7 mmol, Intermediate 1 1 : step a) in THF (198 mL) was added KHMDS (1 M in THF, 119 mL, 1 19 mmol). The resulting solution was stirred at room temperature for 40 minutes and then additional KHMDS (19.8 mL, 19.8 mmol) was added and stirring continued at, room temperature for 2 hours. The mixture was quenched with water (225 mL) and the layers were separated. The aqueous layer was acidified with 1 N aqueous HCl to pH 2-3. Some of the title compound precipitated out of solution and was collected by filtration. The aqueous was then extracted with EtOAc (3 x 200 mL). The organics were combined with the solid collected previously and sonicated. The solution was dried (Na 2 S0 4 ), filtered and concentrated to dryness to provide the title compound as a yellow solid. Intermediate 11: step c

3-(Benzyloxy)-6=bromo-2,4-dieMoFoqMmolme

To a suspension of 3-(benzyloxy)-6-bromo-4-liydroxyqumo1in-2(lH}-one (12.4 g, 35.8 mniol, Intermediate 1 1 : step b) in acetonitrile (119 ml.) was added POCI 3 (10 m.L, 107.5 mmol) followed by 2,6-lutidine (6.26 niL, 53.7 mmol) dropwise. The suspension was heated to 100 °C for 4 hours, then the reaction was allowed to cool to room temperature. The solids were filtered, rinsed with MeOH and dried under vacuum to afford the title compound as a tan solid.

intermediate 11: step d

(3-(Benzyloxy)-2,4-dichIoroquinoUn-6-yl)(l-methyl-lH-imid azol-5-yI)(6- (trifluoromethyl)pyridin-3-yl)methanol

A solution of 3-(benzyloxy)-6-bromo-2,4-dichloroquinoline (2.57 g, 6.71 mmol. Intermediate 11 : step c) in THF (100 mL) was cooled to -78 °C, during which it became a white suspension. Then, wBuLi (1.6 M in hexanes, 5.87 mL, 9.39 mmol) was added dropwise and the resulting dark red solution was stirred for 10 minutes at -78 °C. To this mixture was added a solution of (1- methyl-lH-imidazoi-5-yl)(6-(trifluoromethyi)pyridin-3-yl)met hanone (2.23 g, 8.72 mmol, Intermediate 2: step c) in THF (30 mL) over 4 minutes and the resulting mixture stirred at -78 °C for 2 minutes. The dry-ice/acetone bath was then replaced with an ice bath and the mixture was stirred for an additional 45 minutes. The reaction was then quenched with water and extracted with EtOAc. The organics were combined, dried (MgS0 4 ), filtered and concentrated to dryness to afford the crude product which was purified by FCC (4% MeOH/DCM) to provide the title compound.

Intermediate 11 : step e

(3-(Benzyloxy)-4-chloro-2-niethoxyquinoIin-6-yl)(l-methyl -l//-imidazoI-5-yl)(6

(trifluoromethyI)pyridin-3-yI)methanol

To a mixture of (3-{¾enzyloxy)-2,4-dicMoroquinoHn-6-yl)(l -methyl-l H-imidazol-5-yl)(6- (trit]iioromethyl)pyridin-3-yl)rnethano! (2.46 g, 4.4 mmol. Intermediate 1 1 : step d) in methanol (24.6 raL) was added NaOMe (0.5 M in MeOH, 8.8 mL, 4,4 mmol) and the resulting suspension heated to 65 °C for 8 hours. The mixture was then cooled to room temperature and concentrated to dryness. Water was added and the mixture acidified with 2 N aqueous HQ to ~pH 2. The aqueous was then extracted with EtOAc. The organics were combined and washed with water, saturated aqueous NaHCOi and brine. The organics were then dried ( gS0 4 ), filtered and concentrated to dryness to afford the title compound which was used without further purification.

Intermediate 11: step f

4-ChlorQ-6-(hydroxy(l-methyl-lH midazol-5-ylX

2~methoxyquinoIin~3~oI

To a solution of (3-(benzyloxy)-4-chloro-2-methoxyquinoiin-6-yl)(l-methyl-lH- imidazol-5- yl)(6-(trifluoromethyl)pyridin-3-yl)methanol (2.56 g, 4.61 mmol, Intermediate 1 1 : step e) in MeOH (97 mL) was added 10% Pd/C (246 mg, 0.23 mmol). The reaction vessel was evacuated and then placed under an atmosphere of hydrogen for 1.5 hours. The mixture w r as then flushed with N? and filtered through a pad of Celite ® . The filtrate was concentrated to dryness, then DCM was added and the solution concentrated to dryness. The resulting solid was dried in the oven. The solid was then purified by FCC (15% MeOH DCM) to provide the title compound.

intermediate 11: step g

4-CMoro-6-(iiydroxy(l-m ethyl- IH-im

2-methoxyquinoIin-3-yl trifissoromethaiiesiilfossate

To a suspension of 4-chloro-6-(hydroxy(l-methyl-lH-im^

3-yl)methyl)-2-methoxyquinolin-3-ol (750 mg, 1 .61 mmol, intermediate 1 1 : step f) in CH 2 C1 2 (15 mL) was added pyridine (390 xL, 4.84 mmol) and the reaction became a solution. The solution was cooled to 0 °C and trifluoromethanesulfonic anhydride (683 mg, 2.42 mmol) was added dropwise. The reaction was stirred at 0 °C for 1 hour, then the ice bath was removed and stirring continued for an additional hour. Trifluoromethanesulfonic anhydride (683 mg, 2.42 mmol) was then added and the mixture stirred at room temperature for 1 hour. The solution was poured into a mixture of 1 N aqueous HC1 (20 mL) and ice and then the aqueous was extracted with CH 2 CI 2 . The organics were washed with water followed by saturated aqueous NaHC0 3 and brine. The aqueous layers were combined and back-extracted with EtOAc. The EtOAc layers were combined and washed with water followed by saturated aqueous NaHC0 3 and brine. The CH 2 C1 2 and EtOAc layers were combined, dried (MgS0 4 ), filtered and concentrated to dryness. The crude material was purified by FCC (0-5% MeOH/CH 2 Cl 2 ) to provide the title compound. Intermediate 12: step a

l-Methyl-lH midazoI-5-yl)(pyridin-2-yl)methaiiol

A solution of isopropylmagnesium chloride/lithium chloride complex (1.3 M in THF, 19.5 mL, 25.35 mmol) was added dropwise by syringe to a solution of 5-bromo-l -methyl- lH-imidazole (4.12 g, 25.58 mmol) in dry THF (130 mL) at 0 °C. After 15 minutes, the Grignard solution was added via cannuiation to a solution of picolinaldehyde (2.0 mL, 20.93 mmol) in dry THF (55 mL) at 0 °C. The reaction mixture was stirred for 5 minutes at 0 °C, then warmed to room temperature for 1 hour. The reaction mixture was then cooled in an ice bath and quenched with saturated aqueous ammonium chloride. The mixture was partitioned between brine and ethyl acetate. The separated aqueous phase was further extracted with ethyl acetate. The organic phase was dried (Na 2 S0 4 ), filtered, and concentrated. The crude product was purified by flash column chromatography (silica gel, 0-5% MeOH-DCM) to provide the title compound as a white solid.

Intermediate 12: step b

-Methyl-lH~imidazol~5~yl)(pyridin-2-yl)methaiioiie

A heterogenous mixture of (l-methyl-lH-imidazol-5-yl)(pyridin-2-yl)methanol (1.41 g, 7.45 mmol, Intermediate 12: step a) and manganese dioxide (3.24 g, 37.27 mmol) in 1,4-dioxane (52 mL) was stirred at 100 °C for 2 hours. The reaction mixture was then cooled to room temperature, filtered through Celite ® , washed with DCM, and concentrated to provide the title compound as an off- white solid. intermediate 13: step a Diethyl 2-(4-(trifluoromethoxy)phenyl)malonate

Cul (0.26 g, 1.378 mmol), 2-picolinic acid (0.24 g, 1.969 mmol) and cesium carbonate (19.24 g, 59.061 mmol) were combined in a reaction vessel and the flask was evacuated and re-filled with argon (3 times). 1,4-Dioxane was then added followed by diethylmaionate (6 mL, 39.374 mmol) and l-iodo-4-(trifluoromethoxy)benzene (3 mL, 19.687 mmol). The resulting yellow suspension was stirred at room temperature for 48 hours and quenched with saturated NH 4 CI. The mixture was extracted with EtOAc (2x). The combined organic extracts were dried over Na 2 SQ 4 , filtered, and concentrated to provide the title compound.

Intermediate 13: step b

2-(4-(Trifluoromethoxy)phenyI)maloiiic acid

A mixture of diethyl 2-(4-(trifluoromethoxy)phenyl)malonate (5.6 g, 17.486 mmol, Intermediate 13: step a) and an aqueous 3 M NaOH solution was stirred in a 100 C C oil bath for 1 hour, cooled to room temperature, poured into ice water and acidified with 6 N aqueous HCL The aqueous mixture was extracted with EtOAc. The EtOAc extract was dried over Na 2 S0 4 , filtered, and evaporated in vacuo to provide the title compound.

Intermediate 13: step c

6-Bromo-2,4-dichloro-8-methyl-3-(4~(trifluoromethoxy)pheiiyl )quiiioliiie

A mixture of 2-(4-(trifluoromethoxy)phenyl)malonic acid (3.1 g, 1 1 .74 mmol, Intermediate 13: step b), 4-bromo-2-methylani!ine (2.1 8 g, 1 1 .74 mmol) and P0C1 3 (10 mL) was heated at 105 °C for 3 hours, cooled to room temperature, concentrated under reduced pressure then slowly poured into ice water. A NH 4 OH solution was added to a basic pH (pH 8 - 9). The precipitates were collected by filtration, rinsed with H 2 0 and dried under high vacuum. The resulting tan solids were dissolved in DCM and chromatographed (heptane/DCM) to provide the title compound.

Intermediate 13: step d

6-Bromo-4-chIoro-2-niethoxy-8-methyi-3-(4-(trifliioromethoxy )phenyl)quinoIine

A mixture of 6-bromo-2,4-dichloro-8-methyl-3-(4-(trifiuorometh.oxy)phenyl )quinoline (1.97 g, 4.37 mmol, Intermediate 13: step c) and sodium methoxide (1.1 8 g, 21.84 mmol) in toluene (20 mL) was heated in a sealed tube at 1 10 °C for 24 hours, cooled to room temperature, diluted with DCM, stirred at room temperature for 30 minutes and filtered through Celit,e s rinsing several times with DCM. The solvents were removed under reduced pressure and the off-white solid product precipitated from MeOH, filtered and dried to provide the title compound.

Intermediate 14: step a ferf-Buty] 4-(hydroxy(6-(trifluoromethyl)pyridin-3-yl)methyl)piperidine -l-carboxy]ate

A solution of isopropylniagnesium chloride (2.0 M in THF, 40.3 mL, 80,6 mmol) was added dropwise by syringe to a solution of 5-hromo-2-(t,rifluoromethyl)pyridine (19.5 g, 86.3 mmol) in dry THF (12 ml.) at 2 °C. After 30 minutes, tert-butyl 4-formylpiperidine- 1 -carboxylate (12.3 g, 57.3 mmol) was added to the Grignard solution at 2 °C as a solid. The reaction mixture was warmed to 10 °C over 1.5 hours after which it, was quenched with saturated aqueous ammonium chloride solution. The mixture was partitioned between water and ethyl acetate. The separated aqueous phase was further extracted with ethyl acetate and washed with saturated aqueous NaCl solution. The organic phase was dried (MgS0 4 ), filtered, and concentrated and used crude in the next, step.

Intermediate 14: step b ferf-Buty] 4-(6-(trifluoromethyl)nicotinoyl)piperidine-l-carbox.y]ate

Dess-Martin periodinane reagent (30.0 g, 70.8 mmol) was added to a solution of tert-butyl 4- {¾ydroxy(6-(trifluoromethyl)pyri(Hn-3-yl)methyl)piperidine- l -carboxylate (Intermediate 14: step a, 17.8 g, 49.5 mmol) in DCM at room temperature and the mixture was stirred for 2 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous solution of NaHCC . The organic phase was dried (MgSO^, filtered, and concentrated. The crude product was purified by flash column chromatography (silica gel, 0-60% EtOAc-hexanes) to provide the title compound that was 90% pure by NMR and was carried on to the next step.

Intermediate 14: step c Piperidi5i-4-yl(6-(irifliiorosBetln )pyridiii-3-yl)sBet!i¾5iose

TFA (34.4 mL, 449,3 mmol) was added to a solution of ie/ -butyi 4-(6- (trifluoromethyl)nicotinoyl)piperidine- l-carboxylate (Intermediate 14: step b, 16.1 g, 44.9 mmol) in DCM (450 mL) and the resulting solution was stirred at room temperature for 3 hours. The mixture was concentrated to remove most of the TFA on the rotary evaporator and a mixture of EtOAc/hexanes was added. The white solid that precipitated was filtered and dried and used in the next, step.

intermediate 14: step d l-(4-(6-(lYifluoromethyl)nicotinoyl)piperidiii-l- )ethaiione

TEA (32.1 mL, 230,9 mmol) was added to a solution of piperidin-4-yl(6- (trifiuoromethyi)pyridin~3-yl)methanone (Intermediate 14: step c, 14.3 g, 38.5 mmol) in DCM (427 ml.) followed by acetic anhydride (5.28 mL, 55.8 mmol). The mixture was stirred for 2 hours and transferred to a separatory funnel and washed with 100 mL of aqueous 2 M NaFi 2 PQ 4 solution. The organic layer was dried (MgS0 4 ), filtered and concentrated. The crude product was purified by flash column chromatography (silica gel , 0-3% MeOH-DCM) to provide the title compound.

Inter mediate 15: step ¾

lY-Metlioxy-A-metiiy!pyrimidisie-l-eiirboxafiiide

Sodium pyrimidine-2-carboxylate (4.00 g, 27.4 mmol), imidazole hydrochloride (3.15 g, 30.1 mmo!), and 5 -carbonyldiimidazole (5.26 g, 31 .5 mmol) were slurried in acetonitrile (30 mL) at room temperature under an N2 atmosphere. The mixture was then warmed to 52 °C over 30 minutes. Evolution of carbon dioxide was seen when reaction mixture reached approximately 50 °C. The mixture was then stirred at 52 °C for approximately 2 hours. The reaction was then cooled to room temperature, then Λ' ' ,Ο-dimethylhydroxylamine hydrochloride (3.54 g, 35.6 mmol) was then added slowly, portion wise over approximately 15 minutes and a mild exotherm was seen after each addition. The contents were stirred at room temperature overnight. To the reaction mixture was then added deionized water (25 mL) and dichioromethane (25 mL). 6 M Aqueous hydrochloric acid was added dropwise to acidify the aqueous layer to approximately pH 1. The organic phase was then separated and the aqueous phase was extracted twice with dichioromethane. The combined organics were washed with 2 M aqueous hydrochloric acid, separated, then the acidic layer was extracted twice with dichioromethane. The combined organic phases were washed with a saturated, aqueous NaHCO ? solution, then dried over MgS0 4 , filtered and the solvent was removed by distillation under reduced pressure to provide the title compound.

Intermediate 15: step b

(l-MetIiyl-lH4midazoI-S-yl)(pyrimidm-2-yl)sBetiiaiioHe

5-Bromo-l -methyl- IH-imidazole (6.66 g, 41.4 mmol) was added to a round bottom flask followed by tetrahydrofuran (150 mL) under an N? atmosphere. The contents were cooled to 0 °C in an ice water bath. EtMgBr (3.0 M solution in THF, 13.3 mL, 39.8 mmol) was added slowly via syringe over approximately 5 minutes, then the ice bath was removed and contents allowed to warm and stirred at room temperature for approximately 30 minutes. The vessel was then re-cooled to 0 C C and a solution of iV"niethoxy~A ,; -methylpyriniidine-2~carboxa.mide (3,09 g, 15.9 mmol, Intermediate 15: step a) in THF (20 mL) was cannulated into the reaction vessel. The contents were allowed to stir at 0 °C, then slowly warmed to room temperature, then heated to 40 °C in an oil bath and heated with stirring at that temperature for approximately 36 hours. The contents were then cooled to 0 °C, quenched with a saturated aqueous NH 4 C1 solution, diluted with ethyl acetate and transferred to a separatory funnel. The aqueous layer was separated, extracted twice with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered, then distilled under reduced pressure to afford an amber oil. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / (10% of a 2 M N3¾ MeOH in DCM)) to provide the title compound. intermediate 16: step a

~Dimethylthiazol-5-yl)(l-methyl-l^

1 -Methyl- lH-l ,2,3-triazole was prepared according to the literature reference WO2008/98104. To a 2 L flask containing 1 -methyl- l H-l ,2,3-triazole (9 g, 108.3 mmol) was added THF (1500 mL) and the solution was cooled to -40 °C. To this colorless homogeneous solution was added «-butyllithium (2.5 M in hexanes, 45 mL, 1 12.5 mmol) dropwise which immediately afforded a dark brown viscous mixture. The mixture was kept between -10 to -20 °C for 60 minutes, then a THF solution of 2,4-dimethylthiazole-5-carbaldehyde (17.2 g, 121 ,8 mmol in 200 mL THF) was introduced via cannula. Once the aldehyde was added the reaction was allowed to warm to room temperature. After 3 hours, the reaction was quenched by pouring it into a saturated solution of aqueous NH 4 C1. The aqueous portion was extracted with EtOAc in portions, 7 x 400 mL, The combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated to afford a brown oil. Chromatography on silica gel (10%) acetone-DCM increasing to 50% acetone and increasing to 10% MeOH-DCM) provided the title compound as an amber solid.

Intermediate 16: step b

(2,4-Dimethylthiazol-5~yl)(l-methyl-lH-l,2,3~triazol~5~yl)me thaiioiie

To a 500 ml, flask containing (2,4-dimethyltliiazol-5-yl)(l -methyl- 1H- 1 ,2,3-triazol-5- yl)methanol (10.5 g, 46.8 mmol, Intermediate 16: step a) was added 1 ,4-dioxane (400 mL) and the contents were warmed to form a homogeneous solution. Activated Mn0 2 (18 g, 207 mmol) was added and the dark brownish mixture was heated to reflux in an aluminum, heating mantle under an atmosphere of N 2 . After 1.5 hours, the contents were filtered while still hot through Celite ¾ and rinsed with warm THF. The resulting light orange solution was concentrated and passed through a silica gel column (25% acetone-DCM) to provide the title compound as a light orange solid.

Intermediate 17: step a

-Methoxy-N,2,4-trimethylthiazo1e-5-carboxamide

To a flask containing 2,4-dimethylthiazole~5~carboxylic acid (2.5 g, 15.9 mmol) was added DCM (75 mL) to give a suspension . DMF (3 mL) was added which resulted in a homogeneous solution. Then, carbonyldiimidazole (2.84 g, 17.5 mmol) was introduced and the mixture was stirred at, room temperature for 2 hours. N,0-dim.ethymydroxylamine HQ (1 .9 g, 19.9 mmol) was then added and the mixture was stirred at room temperature for 18 hours, at which time the reaction mixture was diluted with water and 5 N aqueous NaOH and the aqueous portion was extracted with DCM (4 x 50 mL). The combined organics were washed with brine, dried over a 2 S0 4 , filtered and concentrated. Chromatography on silica gel (1 0% EtOAc-DCM increasing to 40% EtOAc) provided the title compound as a colorless oil.

Intermediate 17: step b

(2,4-Dimethylthiazol-5-yi)(i -methyi-l/ -imidazol-5-yI)methanone

To a flask containing 5-bromo-l -methyl- lH-imidazole (390 mg, 2.42 mmol) was added THF (8 mL) and the solution was cooled to 0 °C. To this solution was added isopropylmagnesium chloride-LiCl complex (1.3 M in THF, 2.5 mL, 3.25 mmol) which resulted in a white suspension. The reaction mixture was stirred in a 0 °C bath for 30 minutes, then a THF (2 mL) solution of A'-methoxy-A^^-trimethylthiazole-S-carboxamide (550 mg, 2.75 mmol, Intermediate 17: step a) was introduced. The reaction mixture was stirred at 50 °C for 18 hours, cooled to room temperature and quenched with aqueous NH 4 C1 solution. The aqueous portion was extracted with DCM (4 x 50 mL) and the combined organics were washed with brine, dried over Na 2 S0 4 , filtered and concentrated. Chromatography on silica gel (25% EtOAc-DCM increasing to 5% MeOH-DCM) afforded the title compound as an amber solid.

Intermediate 18: step a

(2,4-Dichloro-3^heQylquinolin-6-yl)(l,2-dimethyl-lH midazol-5-yI)methanol

To a flask containing 6-bromo-2,4-dichioro-3-phenylquinoline (4 g, 1 1.33 mmol. Intermediate 7: step c) was added THF (200 mL) to give a. homogeneous clear solution. The solution was cooled in a dry-ice bath and «-BuLi (2,5 M in hexanes, 4,25 mL, 10.63 mmol) was added which resulted in an immediate reddish- brownish mixture. After 2 minutes, a THF solution of 1,2-dimethyl-lH- imidazole-5-carbaklehyde (1 .75 g, 14.1 mmol in 10 mL THF) was added and the reaction mixture became a light yellow color. The -78 °C bath was replaced with a 0 °C ice-bath and after 40 minutes, the reaction mixture was quenched with aqueous NH 4 C1 solution and the aqueous portion was extracted with EtOAc (4 x 100 mL). The combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated. The resulting solid was triturated with Et 2 0 to provide the title compound as an off white powder. (2,4-Dichloro-3-phenyiqiiinolin-6-yI)(l,2-dimethyi-l/ -imidazol-5-yI)meth

To a flask containing (2,4-dichloro-3-phenylquinolin-6-yl)(l ,2-dimethyl-lH-imidazol-5- yl)methanol (520 mg, 1.31 mmol, intermediate 18: step a) was added 1,4-dioxane (12 mL) followed by manganese dioxide (475 mg, 5.46 mmol). The mixture was heated to reflux for 3.25 hours and then the contents were filtered through a Celite ® pad, while still warm, and the Celite* pad was rinsed with warm THF. The effluent was concentrated under reduced pressure to provide the title compound as an off white foam.

Intermediate 19: step a

-Bromo-3-phen -2-(trifluoromethyI)q«iiioliii-4-o]

A mixture of 2-a.mino-5-bromobenzoic acid (3.01 g, 13.9 mmol), l ,l,l-trifmoro-3-phenyipropan- 2-one (3.11 g, 16,5 mmol), and Eaton's reagent (9,3 mL) in a sealed tube was heated at 100 °C for 4 hours. The reaction mixture was then allowed to cool to room temperature, water was added slowly, and the mixture was stirred vigorously for about 15 minutes. The precipitated solid was collected by filtration, washed with water, and dried to provide the title compound. intermediate 19: step h

6-Bro8BO-4-chloro-3-piieiiyl- A solution of 6-bromo-3-phenyl-2-(trifluoromethyl)quinolin-4-ol (8.29 g, 22.5 mmol, Intermediate 19: step a) in phosphoryl trichloride (25 mL, 269 mmol) was heated at 1 10 °C for 2 hours, and concentrated in vacuo. Dichloromethane and ice-water were added, and the mixture was basified at 4 °C with concentrated NH 4 OH until pH ~ 10. The organic layer was separated and the aqueous layer was extracted with CH 2 CI 2 . The combined organic phases were dried (Na 2 S0 4 ), concentrated, and purified by flash column chromatography (120 g silica gel column, 2 - 9% EtOAc in heptanes) to afford the title compound as a light yellow solid.

Intermediate 20: step a

-Iodo-3-phenyl-2-(trifl¾ioromethyI)q«moIin-4-o1

A mixture of 2-amino-5-iodobenzoic acid (5.20 g, 19.8 mmol), 1 ,1,1 -triflixoro-3-phenylpropan-2- one (3.95 g, 21.0 mmol), and Eaton's reagent (12 mL) in a sealed tube was heated at 100 °C for 2 hours. More l ,l ,l-trifiuoro-3-phenylpropan-2-one (1 .60 g, 8,50 mmol) was added and the mixture was heated for another 2 hours. The reaction was then cooled to room temperature, ice water was added, and the mixture was stirred vigorously for about 20 minutes. 50% Aqueous NaOH and concentrated NH 4 OH solutions were added until pH 9. The resulting dark brown gummy material that formed was diluted with DCM which resulted in a solid precipitate that was collected by filtration. The solids were washed with water and Et 2 0 and air dried to provide the title compound.

Intermediate 20: step h

-Chioro-6-iodo-3-phenyl-2-(trifluoromethyI)quinoHne

A solution of 6-iodo-3-phenyl-2-(trifluoromethyl)quinolin-4-oi (1.54 g, 3.71 mmol, Intermediate 20: step a) in phosphoryl trichloride (5 mL, 53.8 mmol) was heated at 1 10 °C for 1 hour 45 minutes, and then cooled to room temperature. Ice-water was added, and the mixture was basified at 4 °C with 50% aqueous NaOH and concentrated NH 4 OH until pH 9. The precipitated solid was filtered, washed with water and Et 2 0, and dried to provide the title compound. The filtrate was separated and the aqueous layer was extracted with CH 2 C1 2 . The combined organic phases were dried (Na 2 8C>4), concentrated, and purified by flash column chromatography (80 g silica gel column, 0 - 5% EtOAc in heptanes), affording a mixture of the title compound and des- iodo by-product, 4-chioro-3-phenyl-2-(trifluoromefhyi)quinoline, in about 8: 1 ratio as a thick oil, which solidified over night. inter mediate 21

feri- Butyl 4-nicotinoy piperidine-l-carhoxylate

A mixture of piperidin-4-yl(pyridin-3-yl)methanone hydrochloride (397 mg, 1.75 mmol), άι-tert- butyl dicarbonate (710 mg, 3.25 mmol), N,N-dimethylpyridin-4-amine (28 mg, 0.23 mmol) and Et 3 N (1.2 mL, 8.6 mmol) in THF (15 mL) and CH 2 C1 2 (5 mL) was stirred for 3 days and then concentrated. The residue was purified by flash column chromatography (40 g silica gel column, 50-70% EtOAc in heptane) to provide the title compound as a clear oil.

Intermediate 22: step a

(3-Chlorophenyl)(6-(trifluoromethyl)pyridin-3-yl)methan.oiie

To a solution of A r -methoxy-N-rnethyl-6-(trifluoromethyl)nicotinamide (1 .23 g, 5.25 mmol, Intermediate 2: step b) in THF (12 mL) at 4 °C was added 0.5 M (3-chlorophenyI)magnesium bromide in THF ( 12.7 mL, 6.35 mmol). The mixture was stirred at 4 °C to room temperature overnight, and quenched with NH 4 C1 (aqueous). The organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The combined organic phases were dried (Na?SG4), filtered, concentrated, and purified by flash column chromatography (40 g silica gel column, 0- 70% EtOAc in heptanes) to provide the title compound as an oil, which solidified upon standing. intermediate 22: step b

(3-Chlorophenyl)(2,4-dichloro-3-phenylquinolin~6-yl)(6-(trif luoromethyl)pyridm

yl)methaiiol

To a solution of 6-bromo-2,4-dichloro-3-phenylquinoline (422 nig, 1.20 mmol. Intermediate 7: step c) and (3-chlorophenyl)(6-(trifluoromelhyl)pyridin-3-y3.)rnethanone (340 mg, 1 .19 mmol. Intermediate 22: step a) in THF (8 mL) at -78 °C was added 1.6 M «-BuLi in hexane (1.14 mL, 1.82 mmol). The mixture was stirred at, -78 °C to 10 °C for 2 hours and then quenched with NH 4 CI (aqueous). The organic layer was separated, and the aqueous layer was extracted with CH2CI2. The combined organic phases were dried (Na 2 S0 4 ), filtered, concentrated, and purified by flash column chromatography (24 g silica gel column, 30 - 40% EtOAc in heptane) to provide the title compound as a white solid.

Intermediate 23: step a

To a suspension of 2-(trifluoromethyl)isonicotinic acid (1.03 g, 5.39 mmol), N,0- dimethyihydroxylamine hydrochloride (0.800 g, 8,20 mmol) and A /t (ethylca.rbonimidoyl)-A r ,iV- dimethyl ,3-propanediamine hydrochloride (EDO, 1.35 g, 7.04 mmol) in CH 2 CI 2 (30 mL) wa.s added Et 3 N (1.90 mL, 13,7 mmol), and the mixture immediately turned clear. After stirring at room temperature overnight, NH 4 CI (aqueous) was added. The mixture was stirred vigorously for a while, and white solid was then filtered off. The organic layer was separated, and the aqueous layer was extracted with CH 2 CI?. The combined organic phases were washed with brine, and the aqueous layer was back extracted with CH 2 CI 2 . The organic phases were combined, dried (Na. 2 S0 4 ), filtered, concentrated, and purified by flash column chromatography (40 g silica gel column, 40-70% EtOAc in heptanes) to afford the title compound as a clear oil, inter mediate 23: step h

(3-C sloroplies¾yl)(2-(trii½orometliyl)pyr!d! .-4-yl)metliasso8ie

The title compound was prepared using N~methoxy-A-methyl-2-(trifluoromethyi)isonicotiiiamide (Intermediate 23: step a) in place of jV-methoxy-A'-methyl~6-(trifluoromethyl)nicotinamide (Intermediate 2: step b) according to the procedure described in Intermediate 22: step a. intermediate 24

~ 8!orop!sessyi)(2,4-dkMoro~3~p!se&y¾

The title compound was prepared using (3-chlorophenyi)(pyridin-3-yl)methanone in place of (3- chlorophenyl)(6-(trifiuoromethyl)pyridin~3~yl)methanone (Intermediate 22: step a) according to the procedure described in Intermediate 22: step b.

A suspension of 4-picoliiiic acid (3.00 g, 24.4 mmol) and 1 , 1 -carbonyldiimidazole (4.74 g, 29.2 mmol) in CH 2 C1 2 (35 mL) was stirred for ~· 40 minutes, at which time it became a. clear solution. After the addition of N, O-dimethylhydroxylamine hydrochloride (2.85 g, 29.2 mmol), the mixture was stirred at room temperature for 22 hours. Water was added, the organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The combined organic layers were washed with water once, and the aqueous layer was back extracted with CH 2 CI 2 . The organic phases were combined, dried (Na 2 S0 4 ), filtered, concentrated, and purified by flash column chromatography (80 g silica gel column, 100% EtOAc) to provide the title compound as a clear oil. intermediate 25: step b

(l-Methyl-lH~imidazol~5~yl)(pyridiii-4-yl)methaiioiie

To a heat- un dried flask containing 1 -methyl- IH-imidazole (2.2 mL, 27.7 mmol) and THF (13 mL) at -78 °C was added 1.6 M n-BuLi in hexane ( 18.5 mL, 29.6 mmol). After stirring at -78 °C for 40 minutes, neat chlorotriethylsilane (4.9 mL, 29.2 mmol) was introduced slowly. The mixture was stirred at -78 °C for 1 hour. 1.6 M w-BuLi in hexane (18 mL, 28.8 mmol) was added, and stirring became very difficult. The cooling bath was removed, and stirring was continued for a while before the temperature reached around 10 °C, The mixture was re-cooled to -78 °C, a solution of N-methoxy-N-methylisonicotinamide (3.82 g, 23.0 mmol, Intermediate 25: step a) in THF (28 mL) was added via cannula, and stirring stopped. The cooling bath was removed, and the stirring was continued for 40 minutes before room temperature was reached. The reaction was quenched with a few drops of MeOH. Brine was added, the organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The combined organic phases were dried (Na 2 S0 4 ), filtered, concentrated, and purified by flash column chromatography (silica gel, 50- 100% EtOAc in heptane, then 5-10% MeOH in CH 2 C1 2 ) to afford the title compound as an off-white solid. intermediate 25: step c (2,4-Dichloro-3-phenyIq«inolin-6-yl)(l-methyl-lH midazol-5-yl)(pyridiii-4- yl)methanol « TFA

To a solution of 6-bromo-2,4-dichloro-3-phenylquinoline (1 ,46 g, 4.14 mmol, Intermediate 7: step c) and (l -methyl-l H-imidazol-5-yl)(pyridin-4-yl)methanone (778 nig, 4.16 mmol, Intermediate 25: step b) in THF (65 mL) at -78 °C was added 1.6 M «-BuLi in hexane (3.90 mL, 6.24 mmol). After stirring at -78 °C to room temperature overnight, the mixture was quenched with NH 4 CI (aqueous). The organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The combined organic phases were dried (Na 2 S0 4 ), filtered, concentrated, and purified by flash column chromatography (80 g silica gel column, 100% EtOAc, then 5-10% MeOH in CH 2 C1 2 ) and then reverse phase HPLC (water/acetonitrile/0.1 % TFA) to afford the title compound.

Intermediate 26

(4~Buty!~2~c oro~3~pIie!iyIqisiiiolis^

y1)methanol » TFA

The title compound was isolated as a by-product from the reaction that, formed (2,4-dichloro~3~ phenylquinolin-6-yl)( 1 -methyl- lH-imidazol-5-yl)(pyridin-4-yl)methanol (Intermediate 25 : step e).

Intermediate 27

(2,4-Dichloro-3-phenyIq«inolin-6-yl)(pyridin-2-yl)(pyridin- 4- )methanol » TFA

The title compound was prepared using pyridin-2-yl(pyridin-4-yl)methanone in place of (1- methyl-lH-imidazoi-5-yl)(pyridin-4-yl)methanone (Intermediate 25: step b) accordi g to the procedure described in Intermediate 25: step c.

Intermediate 28: step a

2-FIuoro-/V-methoxy-N-methylisonicotinamide

The title compound was prepared using 2~fiuoroisonieotinic acid in place of 4-picolinic acid according to the procedure described in Intermediate 25: step a.

Inter mediate 28: step h

-Fli5oropyridi5i-4-yI)(l-!BethyI-lH-imidazoI-2-yl)metiiaiioi ie

The title compound was prepared using 2-fluoro-A -methoxy-N-met¾ylisonicotinamide (Intermediate 28: step a) in place of /V-methoxy-N-methylisonicotinamide (Intermediate 25: step a) according to the procedure described in Intermediate 25: step b.

Intermediate 29

tert-Butyl 4-((2,4-diehlQro-3-phenylquinolm-6-ylX

1-carboxylate

To a solution of 6-bromo-2,4-dichloro-3-phenylquinoline (174 mg, 0.490 mmol, Intermediate 7: step c) and iert-butyl 4-nicotinoylpiperidine-l -carboxylate (143 mg, 0.490 mmol, Intermediate 25) in THF (5 mL) at -78 °C was added 1 .6 M «-BuLi in hexane (0.47 mL, 0.75 mmol). The mixture was stirred at -78 °C to 0 °C for 3 hours and quenched with NH 4 CI (aqueous). The organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The combined organic phases were dried (Na 2 S0 4 ), filtered, concentrated, and purified by flash column chromatography (40 g silica gel column, 50 - 70% EtOAc in heptane) to provide the title compound as a solid.

Intermediate 30

~CMorop!iesiyl)(2,4-dimetIiyIt azol~5~yl)fiiet!iaiiosie

To a flask containing N-methoxy-A ,2,4-trimethylthia.zoie-5-carboxamide (510 mg, 2.55 mmol, Intermediate 17: step a) was added THF (18 mL) and the solution was cooled to 0 °C. To this solution was added (4-ehlorophenyl)magnesiuni chloride (1 M in diethylether, 3.3 mL, 3.3 mmol). A homogeneous yellow mixture resulted and was allowed to warm to room temperature over 15 minutes. After 2 hours, the reaction mixture was quenched with aqueous NH 4 CI solution and the aqueous portion was extracted with EtOAc (4 x 50 mL). The combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated to dryness. Chromatography on silica gel (100% DCM increasing to 10% EtOAc-DCM) provided the title compound as a white solid.

Intermediate 31: step a N-(4-Bromophenyl)-2-phenyIacetamide

Into a 250-mL round-bottom flask were placed a solution of 4-bromoaniline (17.1 g, 99.42 mmol) and triethylamine (30 g, 297 mmol) in dichloromethane (100 mL). Then, 2-phenylacetyl chloride (18.6 g, 120 mmol) was added dropwise with stirring, and the resulting solution was stirred at room temperature for 6 hours. The solids were filtered out, and the filtrate was concentrated under vacuum. The crude product was purified by re-crystallization from ethyl acetate to provide the title compound as a white solid.

Intermediate 31: step b

-Bromo=2-cMoro-3-phenyIqiiinoIine

Into a 50-mL round-bottom flask were placed Λ Γ ,Λ' -dime thy] formamide (2.15 g, 29,45 mmol) and phosphoryl trichloride (20.3 g, 132.7 mmol) and the mixture was cooled to 0°C. Then, N-(4- bromophenyI)-2-phenylacetamide (5.7 g, 17.87 mmol, Intermediate 23: step a) was added in portions. The resulting solution was heated at 80 °C for 5 hours, then concentrated under vacuum, followed by dilution with 50 mL of H 2 0, and then extraction with 3 x 50 mL of ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by FCC (1 : 10 EtOAc petroleum ether) to afford the title compound as a yellow solid.

Inter mediate 32: step a

Methyl 5-bromo-2-(2-(2-chIorophenyl)acetamido) benzoate

The title compound was prepared using 2-chlorophenylacetyI chloride in place of 2-phenylacetyI chloride using the procedure described for Intermediate 7: step a, -Bromo~3-(2-chlorophenyl)-4~hydroxyquiiiolin-2(lH)-oiie

The title compound was prepared using methyl 5-bromo-2-(2-(2-chlorophenyl)a.cetamido) benzoate (Intermediate 32: step a) in place of 5-bromo-2-(2-phenylacetamido) be zoate using the procedure described for Intermediate 7: step b.

Intermediate 32: step c

~Brofiio~2,4-dicMoro~3~(2~c!i!oropiie!iyI)qisi!iolisie

The title compound was prepared using 6-bromo-3-(2-chlorophenyl)-4-hydroxyquinolin-2(lH)- one (Intermediate 32: step b) in place of 6-bromo-4-hydroxy-3-phenylqxiinolin-2(lH)-one using the procedure described for Intermediate 7: step c.

Example la l-(4-((4-Chloro-2-methoxy-3-(3-(trifluoromethyl)p

y -l /-imidazol-5-yl)methyl)piperidin-l-yr)ethanone O

«-Butyllithium (1.6 M in liexane, 0,585 mL, 0.937 mmoi) was added over 1 minute to a solution of 6-bromo-4-chloro-2-methoxy-3-(3-(trifluoromethyi)phenyl)quin oline (409.8 mg, 0.984 mmol, Intermediate 5: step e) in THF (3 mL) at -78 °C. After 1 minute, a solution of l-(4-(l -methyl- l.H-imidazole-5-carbonyl)piperidin-l -yl)ethanone (231.4 mg, 0.984 mmol, Intermediate 5 : step c) in THF (5 mL) was added via cannula. The mixture was stirred at -78 °C for 10 minutes, then was transferred to an ice bath and stirred for 45 minutes. The reactior! mixture was quenched by addition of saturated aqueous NFL ¾ C1, diluted with water, and extracted with EtOAc (3x), The organic phase was dried (Na 2 SC)4), filtered, and concentrated to dryness. The residue was purified by flash column chromatography (silica gel, 0-8% MeOH-DCM) to afford the target compound as a white solid. MS m/e 573.3 I M 1 1 j . l-(4-((4-Chloro-2-m.ethoxy~3-(3-(trifluoromethyl)phenyl)qu

imidazol-5-yl)methyl)piperidin-l -yl)ethanone was purified by chiral HPLC (Chiralcel OD-H, EtOH) to give 2 enantiomers. Example lb: (first enantiomer to elute off ' chiral column) Ή NMR (400 MHz, CDCI 3 , 1.2: 1 mixture of retainers, minor rotamer peaks marked *) δ 8.21 (s, 1 H), 8.16* (br. s., 1H), 7.85* (d, J = 2.53 Hz, 1H), 7.83 (d, J = 2.53 Hz, 1H), 7.72* (s, 1H), 7.70 (s, 1H), 7.59 - 7.66 (m, 4H), 7.54 - 7.58 (m, 2H), 7.41 - 7.51 (m, 2H), 7.34 (s, 2H), 7.23 (s, 2H), 4.78 (d, J = 12.63 Hz, 1H), 4.61 * (d, J = 12.63 Hz, 1 FI), 4.01 (s, 6H), 3.95 (d, J = 13.14 FIz, 1 F1), 3.76* (d, . 1 1 .12 Hz, H I ). 3.28 (s. M l ). 3.27* (s, 3H), 3.1 1 - 3.24* 0». i l l ). 2.94 - 3.04 (m, 1F1), 2.56 - 2.70 (m, 3H), 2.40 - 2.56 (m, 3H), 2.25 - 2.38 (m, 2H), 2.05 (s, 3H)*, 2.03 (s, 3F1), 1.13 - 1.44 (m, 6H); MS m/e 573.2 [M+H] "h and Example c: (second enantiomer to elute off chiral column) 1H NMR (400 MHz, CDCI 3 , 1.2: 1 mixture of ro tamers, minor rotamer peaks marked *) δ 8.21 (s, IH), 8.16* (br. s., 1 1 1 ). 7.84* (d, J ------ 3.03 Hz, I I I ). 7.82 (d, J ------ 2.53 Hz, lH), 7.71* (s, 1H), 7.69 (s, 1H), 7.58 - 7.65 (m, 4H), 7.52 - 7.58 (m, 2H), 7.40 - 7.51 (m, 2H), 7.29 - 7.35 (m, 2F1), 7.22 (s, 2H), 4.77 (d, J = 13.1 Hz, 1F1), 4.60 (d, J = 13.6 Hz, I FF), 4.00 (s, 6H), 3.93 (d, J = 12.(53 Hz, 1H), 3.74* (d, J = 13.14 Hz, 1H), 3.27 (s, 3H), 3.26* (s, 31 1 ). 3.1 3.22 (m, 1H), 2.92 - 3.05 (m, 1H), 2.80 (s, 1H), 2.57 - 2.74 (m, 2H), 2.39 - 2.55 (m, 3H), 2.2 2.39 (m, 2H), 2.04* (s, 3H), 2.02 (s, 3H), 1.12 - 1.46 (m, 6H); MS m/e 573.2 [M+H] + .

Example 2a: 6-((l -Acetylpiperidin-4-yl)(hydroxy)(l -methyl- 1 //-iniidazol-5-yl)methyi) meihoxy-3-(3-(trif!iioros«eihyi)plieiiyl)qiiis¾olisse-4-ca rbossiiriIe

A round bottom flask was charged with l -(4-((4-chloro-2-mefhoxy-3-(3- (trifluoromefhyi)phenyl)quinolin-6-yl)(hydroxy)( -methyl- lH-imidazo l-5-yl)methyl)piperidin-l- yi)ethanone (158 mg, 0.276 mmol, Example l a), Zn(CN) 2 (58.3 mg, 0.496 mmol), Pd 2 (dba)3 (37.9 mg, 0.041 mmol), zinc nanopowder (5.4 mg, 0.083 mmol), and dicyclohexyl(2',4 , ,6'- triisopropyl-[ 1 , 1 '-biphenyl]-2-yl)phosphine (X-Phos, 27, 1 mg, 0.055 mmol). The flask was evacuated and re-filled with argon (three cycles). Diiiiethylacetamide (1.4 niL, sparged with argon for 30 minutes) was then added and the mixture was heated at 120 °C for 4 hours. The mixture was cooled to room temperature and was filtered through Celite 1 *, washing with EtOAc. The filtrate was washed sequentially with 2 M aqueous ΝΗ 4 ΟΗ, water, and saturated aqueous NaCl. The organic phase was dried (Na 2 S0 4 ), filtered, and concentrated to dryness. The residue was purified by flash column chromatography (silica gel, 1 -8% MeOH-DCM) to afford the title compound. MS m/e 564.3 [M+H] + .

6-((l -Acety3piperidin-4-yl)(hydroxy)(l -m£thy3-EH-im azol-5-yl)methyl)-2-methoxy-3-(3- (trifluoromethyi)phenyl)c|uinoline-4-carbonitriie was purified by chiral HPLC (Chiraicel OD-H, EtOH) to give 2 enantiomers. Example 2b: (first enantiomer to eiute off chiral column) Ή NMR (400 MHz, CDCI 3 , 1.1 : 1 mixture of rotamers, minor rotamer peaks marked *) δ 8.28 (s, 1H),

8.24* (s, 1H), 7.84 - 7.91 (m, 2H), 7.71 - 7.84 (m, 6H), 7.63 - 7.71 (m, 2H), 7.43 - 7.50 (m, 2H), 7.29 - 7.33 (m, 2H), 7.21 (s, 21 !). 4.76 <d. ./ 14.65 Hz, 1H), 4.59* (d, ./ 11.12 Hz, IH), 4.06* (s, 6H), 3.94 (d, J= 12.63 Hz, IH), 3.68 - 3.80 (m, IH), 3.29 (s, 3H), 3.27* (s, 3H), 3.22 (s, IH), 3.13 - 3.22* (m, IH), 3.10* (s, IH), 2.93 - 3.04 (m, IH), 2.58 - 2.71 (m, IH), 2.23 - 2.57 (m, 5H), 2.04 (s, 3H), 2.03 (s, 3H), 1.12 - 1.47 (m, 6H); MS m/e 564.2 [M+H] + and Example 2c: (second enaiitiomer to elute off cliiral column) Ή NMR (400 MHz, CDCI 3 , 1.1 : 1 mixture of rotamers, minor rotamer peaks marked *) δ 8.28 (s, H), 8.24* (s, IH), 7.88-7.90* (m, I H), 7.85- 7.87 (m, IH), 7.71 - 7.83 (m, 6H), 7.64 - 7.71 (m, 2H), 7.42 - 7.50 (m, 2H), 7.31-7.34 (m, 2H), 7.22 (s, 2H), 4.77 (d, J ------ 13.14 Hz, IH), 4.60* (d, ./ 14.15 Hz, IH), 4.07 (s, 6H), 3.94* (d, J =

14.15 Hz, IH), 3.68 - 3.80 (m, IH), 3.28 (s, 3H), 3.27* (s, 3H), 3.13 - 3.23 (m, I H), 2.96 - 3.05* (m, IH), 2.95 (s, I H), 2.84* (s, IH), 2.58 - 2.70 (m, IH), 2.22 - 2.57 (m, 5H), 2.05* (s, 3H), 2.03 (s, 3H), 1.17 - 1.45 (m, 6H); MS m/e 564.2 [M+Hf.

Example 3a: (4-ChIoro-2-methoxy-3-(3-(trifluoroniethyl)phenyl)quinolin-6 -yl)(l-niethyl- i/ -l,2 -triazol-5-yl)(l-methyl-l//-imidazol-5-yl)methanoI

«-Butyllitbium ( 1.6 M in hexane, 0.547 mL, 0.876 mmol) was added over 1 minute to a solution of 6-bromo-4-chloro-2-rnethoxy-3-(3-(trifluoromethyl)phenyl)qui noline (383 mg, 0.919 mmol. Intermediate 5: step e) in THF (3 mL) at -78 °C. After 5 minutes, a solution of (1 -methyl- 1H- l,2,3 riazol-5-yl)(l~methyl~lH-imidazol-5-yl)methanone (176 mg, 0.919 mmol, Intermediate 6, step b) in THF (15 mL) was added via cannula. The mixture was stirred at -78 °C for 5 minutes, then was transferred to an ice bath and stirred for 30 minutes. The reaction mixture was quenched by addition of saturated aqueous NH 4 CI, diluted with water, and extracted with EtOAc (3x), The organic phase was washed twice with water. The organic phase was dried (Na 2 S0 4 ), filtered, and concentrated. The residue was purified by flash column chromatography (silica gel, 1-7% MeOH-DCM) to afford the target compound as a white solid. MS m/e 529.2 [M+H] + . (4-Chloro-2-methoxy-3-(3-(trifluoromethyi)phenyl)quinolin-6- yl)(l -methyl- lH-l ,2,3-tri yl)(l -methyl-lH-imidazol-5-yi)methanol was purified by chiral SFC (Chiralpack IC, 75% C0 2 , 25% (0,2% isopropylamine-2-propanol)) to give 2 enantiomers. The enantioniers were then further purified individually on plug silica gel columns (0-5% MeOH-DCM). Example 3b: (first enantiomer to elute off chiral column) ! H NMR (400 MHz, CDCI 3 ) δ 8.30 (br. s., 1H), 7.82 - 8.09 (br. s., 1 1 1 !. 7.78 (d, ,/ 9.1 Hz, 1 H), 7.70 - 7.75 (m, 1H), 7.61 - 7.68 (m, 21 1 ;·. 7.36 (br. s., 1 1 1 ;·. 7.26 - 7.32 (m, 1H), 7.13 (br. s., 1 1 1 ;·. 6.19 (br. s., i l l ). 3.98 (s, 3H), 3.91 (s, 3H), 3.52 is. 3H); MS m/e 529.2 | M 1 1 1 and Example 3c: (second enantiomer to elute off chiral column) Ή NMR (400 MHz, CDCI 3 ) δ 8.31 (br. s„ ! f ! ). 7.79 - 8.05 (br. s., IH), 7.78 (d, J = 8.6 Hz, 1 H), 7.71 - 7.75 (m, I H), 7.61 - 7.69 (m, 2H), 7.36 (br. s., 1H), 7.26 - 7.32 (m, 1 1 1 . 7.12 (br. s., IH), 6.19 (br. s., I H), 3.98 (s, 3H), 3.91 (s, 3H), 3.52 (s, 3H); MS m/e 529.2 | M 1 I | .

Example 4a: 6-(Hydroxy(l-methyl-1 /-l ,2,3-triazol-5-yI)(l-methyI-l// midazol-5- l)methyl)-2-methoxy-3-(3-(trifluoromethyI)phenyl)quiiioline- 4-carbonitrile

A round bottom flask was charged with (4-chloro-2-methoxy-3-(3- (trifluoromethyi)phenyl)quinolin-6-yl)(i -methyl- lH-1 ,2,3-triazol-5-yl)(l -methyl- lH-imidazol-5- yi)methaiiol (96.6 mg, 0.183 mmol, Example 3a), Zn(CN) 2 (38.6 mg, 0.329 mmol), Pd 2 (dba) 3 (25.1 mg, 0.027 mmol), zinc nanopowder (3.6 mg, 0,055 mmol), and dicyclohexyl(2 , ,4',6'- triisopropyl-[ l ,r-bipheiiyl]-2-yl)phosphine (X-Phos, 18,0 mg, 0.037 mmol). The flask was evacuated and re-filled with argon (three cycles). Dimethyiacetamide (1.8 mL, sparged with argon for 30 minutes) was then added and the mixture was heated at 120 °C for 9 hours. The mixture was cooled to room temperature and was filtered through Celite*, washing with EtOAc. The filtrate was washed sequentially with 2 M aqueous NH 4 OH, water, and saturated aqueous NaCl. The organic phase was dried (Na S0 4 ), filtered, and concentrated. To improve conversion, the crude product was resubjected to the reaction conditions as described above for an additional 4.5 hours, then was worked up as described above. The residue was partially purified by flash column chromatography (silica gel, 2-8% MeOH-DCM) then was further purified by RP-HPLC (10-90% CH 3 CN-H 2 O, 0.1 % TFA). HPLC fractions were basified (saturated aqueous NaHC0 3 ), partially concentrated, and extracted with DCM (3x). The organic extract was dried over a 2 S0 , filtered, and concentrated to afford the title compound. MS m/e 519.9 [M+H] ÷ .

6-(Hydroxy(l -methyl- 1H- 1 ,2,3-triazol-5-yl)(l -methyl- lH-imidazol-5-yi)methyl)-2-methoxy-3- (3-(trifluoromethyl)phenyl)quinoline-4-carbonitrile was purified by ehiral SFC (Chiralpack IC, 90% CO?, 10% (0.2% isopropylamine-EtOH)) to give 2 enantiomers. The enantiomers were then further purified on plug silica gel columns (0-5% MeOH-DCM). Example 4b: (first enantiomer to elute off ehiral column) H i N R (400 MHz, CDC1 3 ) δ 8.27 - 8.31 (m, 1 1 1 ). 7.88 (d. J ------ 8.6

Hz, 1H), 7.84 (s, 1 H), 7.77 - 7.82 (m, 1H), 7.73 - 7.77 (m, 1H), 7.66 - 7.73 (m, 1H), 7.35 (dd, ,/ = 8.6, 1.5 Hz, 1 H), 7.16 (s, IH), 6.33 (br. s., IH), 6.25 (br. s., IH), 4.07 (s, 3H), 3.93 (s, 3H), 3.53 (s, 3H); MS m/e 520.2 [M+H] ÷ and Example 4c: (second enantiomer to elute off ehiral column) Ή NMR (400 MHz, CDCI 3 ) δ 8.29 (d, J = 2.0 Hz, IH), 7.90 (d, J = 8.6 Hz, IH), 7.84 (s, IH), 7.77 - 7.81 (m, I H), 7.73 - 7.77 (m, 1 1 1 ). 7.66 - 7.73 (m, IH), 7.37 (dd, ./ 9.1 , 2.0 Hz, IH), 7.33 (s, IH), 7.59 (s, I H), 6.30 (br. s., I H), 5.54 (br. s., I H), 4.08 (s, 3H), 3.95 (s, 3H), 3.55 (s, 3H); MS m/e 520.0 j Y! l i | .

Example 5: (4-Ch]orophenyI)(2,4-dichIoro-3-(pyridm-2-yl)q«moIin-6-y1)( l-meth^ ^ imidazo1-5-y])methanoI » TFA

Into a 500-rnL round-bottom flask was placed a solution of 6-bromo-2,4-dichloro-3-(pyridin-2- y quinoline (380 mg, 5.07 mmol, intermediate 9: step c) in tetrahydrofuran (10 mL). This was followed by the addition of «-BuLi (0.43 mL, 5 .28 mmol, 3.0 M in hexanes) dropwise with stirring at -78 °C. The resulting solution was stirred for 30 minutes at -78 °C. To this was added a solution of 5-[(4-chlorophenyl)carbonylJ-l -methyl-l.H-imidazole (599 mg, 0.90 mmol, Intermediate 8: step b) in tetrahydrofuran (10 mL) dropwise with stirring at -78 °C. The resulting solution was allowed to react, with stirring, for an additional 2 hours at room temperature. The reaction was then quenched by the addition of 10 mL of saturated aqueous NH 4 C1. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC [(1 #waters-2767- 1 ): Column, Sunfire prep C18, 5 μηι, 19 x 100 mm; mobile phase, 0.05% TFA in water and MeOH (45% MeOH up to 65% in 10 minutes); Detector, IJV 254 nm] to afford the title compound trifluoroacetic acid salt as a white solid. MS (ES, m/z) 495 [M+H] + ; ! H NMR (300 MHz, CDCI3 + D 2 0) δ 8.86 - 8.69 (m, 1 H), 8.50 (br. s., 1 H), 8.40 (br. s., 0.5H), 8.25 (br. s., 0.51 1 . 8.07 (d, J = 8.6 Hz, IH), 7.97 (t, J ------- 7.5 Hz, 1H), 7.85 - 7.69 (m, 1H), 7.60 - 7.43 (m, 211),

7.43 - 7.30 (m, 4H), 6.70 (br. s., 0..5H), 6.6.5 (br. s., 0.5H), 3.59 (br. s., 3H).

Example 6: (4-ChlorophenyI)(2,4-dichIoro-3-(pyridin-3-y1)qumoIm-6-yl)(l -methyl-l imidazol-5~yl)methaiiol » TFA

Into a 100-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 6-bromo-2,4-dichloro-3-(pyridin-3-yl)quinoline (160 mg, 0.45 mmol, Intermediate 10: step c) in tetrahydrofuran (10 mL). This was followed by the addition of t-BuLi (0.85 mL, 1.35 mmol, 1 .6 M in pentane) dropwise with stirring at -78 °C. The resulting solution was stirred for 30 minutes at -78 °C. To this was added a solution of 5-[(4- c lorophenyl)carbonyl]-l-methyl-lH-imidazole (121 mg, 0.55 mmol, Intermediate 8, step b) in tetrahydrofuran (10 mL) dropwise with stirring at -78 °C. The resulting solution was stirred for 30 minutes at -78 °C, The resulting solution was allowed to react, with stirring, for an additional 30 minutes at -40 °C, The resulting solution was allowed to react, with stirring overnight at room temperature. The reaction was then quenched by the addition of 50 mL of saturated aqueous NH 4 CI. The resulting solution was extracted with 3x 10 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC [(IntelFlash-5 ): Column, silica gel; mobile phase, 0.05% TFA in water and CFI. 3 CN (22% CH 3 CN up to 44% within 10 minutes; Detector, IJV 254 nm] to afford the title compound trifluoroacetic acid salt as an off-white solid. MS (ES, m/z) 495 [M+Hf and 517 [M+Na] + ; ; H NMR (400 MHz, ( 1) . · ( )! ) ) δ 9.03 (s. 1H), 8.78- 8.62 (ra, 2H), 8.39-8.36 (m, IH), 8.15-8.07 (m, 2H), 7.97-7.94 (m, 1 H), 7.80-7.76 (m, IH), 7.54- 7.45 (m, 4H), 7.00 (br. s., I H), 3.74 (d, ./ 1 1.2 Hz, 3H).

Example 7s: {4-C loro-2-methoxy-3-[4-(methy1sulfoiiyI)phenyI]q«iiioMn-6-yI}( l-methyI- lH-imidazol-5-yI)[6-(trifl«oromethyl)pyridin-3-yI]methan.o]

A mixture of 4-chloro-6-(hydroxy(l -methyl- lH-imidazol-5-yl)(6-(trifluoromethyl)pyridin-3- yl)methyl)-2-methoxyquinolin-3-yl trifktoromeihanesulfonate (70 mg, 0.12 mmol, Intermediate 5 5 : step g), (4-(methylsulfonyl)phenyl)boronic acid (35 mg, 0.18 mmol), PdCl 2 (dppf) (9 mg, 0.012 mmol) and K. 2 C0 3 (16 mg, 0.12 mmol) was sparged with nitrogen three times. To this mixture was added 1,4-dioxane (2 mL) and water (0.3 mL) and the suspension purged with nitrogen. The resulting solution was heated to 65 °C for 15 hours. The reaction was allowed to cool to room temperature and concentrated to dryness. The residue was dissolved in DMSC), filtered and purified by reverse-phase HPLC (acetonitrile / ' water + TFA). The acidic fractions were neutralized by diluting with EtOAc and washing with saturated aqueous NaHC0 followed by water. The organics were dried (MgS0 4 ), filtered and concentrated to dryness to provide the title compound. Ή NMR (400 MHz, CD 3 OD) δ 8.79 - 8.76 (m, IH), 8.24 - 8.20 (m, IH), 8.08 - 8.05 (m, 2H), 8.05 - 8.01 (m, I H), 7.96 - 7.93 (m, IH ), 7.84 (d, ./ 8.3 Hz, I H), 7.76 - 7.72 (m, 2H), 7.65 - 7.62 (m, 2H), 6.35 (s, I H), 4.00 (s, 3H), 3.49 (s, 3H), 3.20 (s, 3H ); MS (ESI): mass caicd. for C28H22CIF3 4O4S, 602.1 ; m/z found, 603.2 [M+Hf.

{4-Chloro-2-methoxy-3- 4-(methylsulfonyl)phenyl]quinolin-6-yl} ( 1 -methyl- lH-imidazol-5- yi)[6-(trifluoroniethyl)pyridiii-3-yl]metlianol was purified by HPLC (KiOmasil CI 8 100 A, 5 μΜ column, Mobile phase: 35-100% acetonitrile/water + 0.25% ammonium bicarbonate) followed by FCC (0-5% MeOH/DCM) to give 2 enantiomers. The first eluting enantiomer was Example 7b: ¾ H NMR (400 MHz, CD 3 OD) δ 8,81 - 8.77 ( m. i l l ). 8.23 (d, J = 2.0 Hz, i l l). 8.09 - 8.06 (m, 2H), 8.06 - 8.02 (m, 1H), 7.97 - 7.93 (m, 1H), 7.85 (d, J = 8.2 Hz, IH), 7.78 - 7.72 (m, 2H), 7.66 - 7.62 (m, 2H), 6.37 (s, i l l). 4.01 (s, 3H), 3.50 (s, 3H), 3.21 (s, 3H); MS (ESI): mass calcd. for C 28 H 22 CIF 3 N 4 O 4 S, 602.1; m/z found, 603.2 [M+H] ' and the second eluting enantiomer was Example 7c: ! H NMR (400 MHz, CD 3 OD) δ 8.80 - 8.77 (m, IH), 8.22 (d, J= 2.1 Hz, 1H), 8.09 - 8.05 (m, 2H), 8.05 - 8.01 (m, IH), 7.96 - 7.92 (m, 1 FI), 7.85 (d, J = 8.2 FIz, 1H), 7.77 - 7.71 (m, 2FI), 7.66 - 7.61 (m, 2H), 6.36 (s, I Fl), 4.00 (s, 3H), 3.49 (s, 3F1), 3.21 (s, 3H); MS (ESI): mass calcd. for C 2S H 22 CIF 3 N 4 O 4 S, 602.1 ; m/z found, 603.2 [M+H] + .

Example 8: [4-Chloro-2-methoxy-3-(l-methyl-lH-py

imidazol-5-yl)[6-(trifluoromethyl)pyridin-3-yl]methanol » TFA

The title compound was prepared using 1 -methyl-4-(4,4,5,5-tetramethyI- 1 ,3,2-dioxaborolan-2- yl)-l H-pyrazole in place of (4-(methyl.sulfonyl)phenyl)boronic acid using the procedure described for Example 7a. J H NMR (400 MHz, CD3OD) δ 9.03 (s, I Fl), 8.82 (d, J = 2.3 Hz, 1H), 8.25 (d, J = 2.1 Hz, i f !}. 8.10 - 8.06 (m, i f !}. 8.03 (s, I Fl), 7.93 (d. ./ 8.7 FIz, 1FI), 7.90 (d, ./ 8.3 FIz, 1H), 7.85 (s, III), 7.66 (dd, J = 8.7, 2.1 FIz, HI), 7.08 - 7.06 (m, IH), 4.09 (s, 3H), 3.98 (s, 3H), 3.71 (s, 3H); MS (EST): mass calcd. for C25H20CIF3 6O2, 528.1; m/z found, 529.2 [M+H] + .

Example 9a: 5-(4-Ch1oro-6-{hydrox '(l -methyl- lH-imidazol-5-yI)[6-

(tri.fl«oromethy1)pyridin-3-yI]methy1}-2-methoxyqumolin- 3-yl)pyrim

carbonitrile » TFA

The title compound was prepared using 5-(4,4,5,5-tetramethyl- l ,3,2-dioxaborolan-2- yi)pyrimidine-2-carbonitrile in place of (4-(methylsulfonyi)phenyl)boronic acid using the procedure described for Example 7a. S H MR (400 MHz, CD 3 OD) δ 9.06 - 9.02 (m, 3H), 8.84 - 8.79 (m, 1 1 1 ). 8.33 - 8.29 (m, i l l ). 8.12 - 8.08 (m, 1 H), 8.02 (d, J ------ 8.9 Hz, 1 1 1 ). 7.90 id. ./ 8.2

Hz, U S ). 7.82 - 7.77 (m, ! f ! ). 7.10 (s, 1H), 4.06 (s, 3H), 3.71 (s, 3H); MS (ESI): mass calcd. for m z found, 552.2 [M+Hf .

5-(4-Chloro-6- {hydroxy(l -methyl- lH~imidazol-5-yl)[6-(trifluoromethyl)pyridin~3-yl]methyl} -2- methoxyquinolin~3-yl)pyrimidine-2-carhoni†,rile was purified by HPLC (CHIRACEL OD 20 μΜ Daicel column, Mobile phase: 100% EtOH) followed by FCC (0-10% MeOH/DCM) to give 2 enantiomers. The first eluting enantiomer was Example 9b: 3 H NMR (400 MHz, CD 3 OD) δ 9.05 (s, 2H), 8.77 (m, IH), 8.25 (d, ,/ 2.1 Hz, IH), 8.04 (dd, ,/ 8.2, 2.2 Hz, 1H), 7.98 - 7.94 (m, IH), 7.85 (d, J = 8.2 Hz, IH), 7.79 (dd, J = 8.9, 2.1 Hz, I H), 7.76 - 7.72 (m, IH), 6.36 (s, IH), 4.05 (s, 3H), 3.48 (s, 3H); MS (ESI): mass calcd. for Ο,.Μ ,-αΐ- ' Λ ' . .. 551.1 ; m/z found, 552.2 [M+H and the second eluting enantiomer was Example 9c: J H NMR (400 MHz, CD 3 OD) δ 9.05 (s, 2H), 8.80 - 8.76 (m, 1 1 1 ). 8.26 (d, J ------ 2.1 Hz, IH), 8.06 - 8.02 (m, IH), 7.97 id. ./ 8.8

Hz, IH), 7.85 (d, J = 8.3 Hz, IH), 7.79 (dd, J = 8.9, 2.1 Hz, IH), 7.75 (s, IH), 6.36 (s, IH), 4.06 (s, 3H), 3.49 (s, 3H); MS (ESI): mass calcd. for CzeHnClF^Cb, 551.1 ; m/z found, 552.2

Example 10: (4-Chloro-2-methoxy-3-pyrimidin-5-ylqui^

yl) [6-(trifluoromethyl)pyridm-3-yl] methanol'TFA

The title compound was prepared using pyrimidin-5-ylboronic acid in place of (4- (methylsulfonyl)phenyl)boronic acid using the procedure described for Example 7a. Ή NMR (400 MHz, CD 3 OD) δ 9.23 (s, lH), 9.04 (s, IH), 8.88 (s. 2H), 8.84 - 8.81 (m, 1H), 8.32 - 8.28 (m, 1 H), 8.12 - 8.08 (m, 1 H), 8.03 - 8.00 (m, 1 H), 7.92 - 7.88 (m, 1 H), 7.80 - 7.76 (m, 1 H), 7.1 1 - 7.09 (m, 1H), 4.05 (s, 3H), 3.72 (s, 3H); MS (ESI): mass calcd. for

526.1 ; m/z found, 527.1 [M+H]".

Example 11 : {4-Chloro-2-methoxy-3-[3-(methylsuIfonyI)pheByl] quinolin-6-yI} (1-methyl- ί H-imidazoJ-5-yI) [6-(trifluoromethyl)pyridin-3-y 11 methanol'TFA

The title compound was prepared using (3-(methylsulfonyl)phenyl)boronic acid in place of (4- (methylsulfonyl)phenyl)boronic acid using the procedure described for Example 7a. Ή NMR (400 MHz, CD 3 OD) 5 9.02 (s, 1H), 8.84 - 8.81 (m, I I I ). 8.30 - 8.27 (m, ! f ! ). 8.12 - 8.07 (m, 1H), 8.06 - 8.03 (m, 1H), 8.00 (d, J = 8.8 Hz, 1H), 7.97 - 7.95 (m, IH), 7.90 (d, J = 8.2 Hz, 1H), 7.79 - 7.71 (m, 3H), 7.10 - 7.07 (m, IH), 4.02 (s, 3H), 3.72 (s, 3H), 3.18 (s, 3H); MS (ESI): mass calcd. for C,..H . . . <(1 l- ,() ;S. 602.1 ; m/z found, 603.2 [M+H]". Example 12 : N- [3-(4-C hIoro-6- {hydrox (l-methyl-lH-imidazol-S-yl) 16-

(trifluorometh i)pyridin-3-yIImethyI}-2-methoxyquinoliii-3- yI)phenyl]methanesulfonainide » TFA

The title compound was prepared using (3-(methylsulfonamido)phenyl)boronic acid in place of (4-(methylsuifonyl)phenyl)boronic acid using the procedure described for Example 7a. Ή NMR (400 MHz, CD 3 OD) δ 9.03 (s, I f !). 8.85 -- 8.81 (m, 1H), 8.27 - 8.24 (m, 1H), 8.11 - 8.06 (m, lH), 7.99 (d, J= 8.7 Hz, 1H), 7.90 (d, J = 8.3 Hz, 1H), 7.75 - 7.70 (m, 1H), 7.48 - 7.42 (m, lH), 7.31 - 7.27 (m, 1H), 7.26 - 7.23 (m, lH), 7.14 - 7.10 (m, 1H), 7.09 - 7.07 (m, 1H), 4.01 (s, 3H), 3.72 (s, 3H), 2.99 (s, 3H); MS (ESI): mass calcd. for C 28 H 23 CIF 3 N 5 O 4 S, 617.1; m'z found, 618.0 [M+H] + .

Example 13: {4-Chloro-2-methoxy-3-[5~(methylsulfonyl)pyridiii-3-yl]quiii olin-6-yl}(l- methyl-lH-imidazol-5-yl)[6-(trifluoromethyl)pyridiii~3-yl]me thaiiol » TFA

The title compound was prepared using (5-(mefhylsiilfonyl)pyridin~3-yl)boronic acid in place of (4-(methylsulfonyl)phenyl)boronic acid using the procedure described for Example 7a. f H NMR (400 MHz, CD 3 OD) δ 9.19 - 9.15 (m, i l l). 9.04 (s, ! H). 8.91 - 8.87 (m, 1 H), 8.82 (d, ./ 2.2 Hz, 1H), 8.46 - 8.43 (m, 1H), 8.31 (d, J= 2.1 Hz, 1 H), 8.13 - 8.08 (m, 1H), 8.02 (d, J= 8.8 Hz, 1 H), 7.90 (d, J = 8.3 Hz, H I ). 7.80 - 7.76 (m, I I I ). 7.12 - 7.09 (m, ! f ! ). 4.05 (s, 3H), 3.72 (s, 31 ! ). 3.28 (s, 3H); MS (ESI): mass calcd. for C27H21CIF3N5O4S, 603.1 : m/z found, 604.0 [M+Hf ,

Example 14a: [4-Chloro-3-(5-chlorothiopheB-2-yl)-2-methoxyq«iiioliii-6-y l](l-methyl-lH r - imidazol-5-yl)[6-(trifluoromethyl)pyridin-3-yl]methaiioI » TFA

A mixture of 4-chloro-6-(hydroxy(l -methyl- lH-imidazol-5-yl)(6-(trifluoro methyl)pyridm-3- yl)methyl)-2-methoxyquinolm-3-yi trifiuoromethanesulfonate (40 nig, 0.067 mmol, Intermediate 1 1 : step g), (5-chlorothiophen-2-yl)boronic acid (.16 mg, 0.1 mmol), PdCl 2 (dppf) (5 mg, 0.007 mmol) and K 2 CO 3 (9 mg, 0.067 mmol) was sparged with nitrogen three times. To this mixture was added 1 ,4-dioxane (1.2 niL) and water (0.17 mL) and the suspension purged with nitrogen. The resulting solution was heated to 65 °C for 15 hours. The mixture was then heated to 75 °C for 6 hours after which it was allowed to cool to room temperature. The solution was diluted with EtOAc, washed with water, dried (MgS0 ), filtered and concentrated to dryness. The crude material was purified by reverse-phase HPLC (acetonitrile / water + TFA) to provide the title compound. Ί ί NMR (400 MHz, CD 3 OD) δ 9.03 (s, I H ). 8.84 - 8.81 (m, I I I ). 8.26 (d, ./ 2,2 Hz, 1H), 8.1 1 - 8.06 (m, 1H), 7.96 (d, ./ 8.7 Hz, 1 H), 7.90 (d, J ------ 8.2 Hz, 1 H), 7.74 - 7.71 (m,

1 H), 7.09 - 7.04 (m, 3H), 4.06 (s, 3H), 3.73 (s, 3H); MS (ESI): mass calcd. for C25H17CI2F3N4O2S, 564.0; m/z found, 565.0 | M I I I .

[4-Chloro-3-(5-chlorothiophen-2-yl)-2-mefhoxyqumolin-6-yl](l -methyl- l H-imidazol-5-yl)[6- (trifluoromethyl)pyridin-3-yl]methanol was purified by HPLC (Kromasil CI 8 100 A, 5 μΜ column. Mobile phase: 35-100% acetonitrile/water + 0.25% ammonium bicarbonate) to give 2 enantiomers. The first eluting enantiomer was Example 14b: 3 H NMR. (400 MHz, CD 3 OD) δ 8.79 - 8.77 (m, I I I ). 8.21 (d, J ------ 2.0 Hz, I I I ). 8.04 - 8.00 On. IH), 7.90 (d, J ------ 8.8 Hz, I I I ). 7.84

(d, ,1 = 8.2 Hz, IH), 7.74 (s, I H), 7.72 (dd, J = 8.8, 2.1 Hz, IH), 7.08 - 7.06 (m, IH), 7.05 - 7.02 (m, I H), 6.36 - 6.34 (m, I H), 4.05 (s, 3H), 3.48 (s, 3H); MS (ESI): mass calcd. for C 25 H] 7 C1 2 F 3 4 0 2 S, 564,0; m/z found, 565.0 [M+H] and the second eluting enantiomer was Example 14c: Ή NMR (400 MHz, CD :( ) !)} δ 8.80 - 8.76 (m, 1H), 8.21 (d . J = 2.1 Hz, i l l ). 8.05 - 8.00 (m, 1H), 7.91 (d, ./ 8.8 Hz, 1H), 7.84 (d. ./ 8.2 Hz, 1H), 7.75 (s, 1H), 7.74 - 7.70 (m, 1H), 7.08 - 7.06 (m, 1H), 7.06 - 7.03 (m, 1H), 6.37 - 6.34 (m, 1H), 4.05 (s, 3H), 3.48 (s, 3H); MS (ESI): mass calcd. for C >,H r CI . -f ; :N ;O . .S. 564.0: in / found, 565.0 [M+H] + .

Example 15: [4~CMoro~2~methoxy-3-(2~methoxypyrim

lH midazol-5-yl)[6-(trifliioromethyl)pyridiii-3-yl]methaiiol » TFA

The title compound was prepared using (2-methoxypyrimidin-5-yl)boromc acid in place of (5- chlorothiophen-2-yl)boronic acid using the procedure described for Example 54a. ! H NMR (400 MHz, CD 3 OD) δ 9.05 - 9.02 (m, ! ! ! ). 8.84 - 8.80 (m, 1 1 1 ). 8.63 (s, 2H), 8.28 (d, J ------- 2.2 Hz, I I I .

8.12 - 8.08 (m, 1H), 8.00 (d, J -- 8.7 Hz, I I I . 7.90 (d, ./ 8.3 Hz, 1 H), 7.78 - 7.73 (m, 1H), 7.1 1 - 7.08 (m, 1H), 4.09 (s, 3H), 4.05 (s, 3H), 3.72 (s, 3H); MS (ESI): mass calcd. for C26H20CIF3 6O3, 556.1 ; m/z found, 557.1 [M+H] + .

Example 16: 4-(4-Chloro-6-{hydroxy(l-niethyl-l//-imidazol-5-yI)[6- (trifluoromethyl)pyridin-3-yIlmethyl}-2-methoxyquinoHn-3-yi) benzOnitrile » TFA

The title compound was prepared using (4-cyanophenyl)boronic acid in place of (5- chlorothiophen-2-yl)boronic acid using the procedure described for Example 14a, l H NMR (400 MHz, CD 3 OD) δ 9.05 - 9.03 (m, lH), 8.84 - 8.80 (m, lH), 8.27 (d, J = 2.3 Hz, 1H), 8.1 1 - 8.07 (m, H i ).. 8.00 (d, J = 8.7 Hz, 1H), 7.90 (d, J = 8.3 Hz, 1H), 7.87 - 7.83 (m, 2H), 7.74 (dd, J = 8.8, 2.2 Hz, 1H), 7.57 - 7.53 (m, 2H), 7.10 - 7.07 (m, 1H), 4.00 (s, 3H), 3.72 (s, 3H): MS (ESI): mass calcd. for C 2 ^. X9 CXE^ 5 0 2 , 549.1 ; m/z found, 550.2 [M+H] + .

Example 17: N~[4-(4-Chloro-6-{hydroxy(l-methyl-lH~iniidazol-5-yl)[6-

(trifluoromethyl)pyridin-3-yl]methyl}-2-methoxyquiiiolin- 3- yl)phenyl]methanesulfonaniide » TFA

The title compound was prepared using (4-(meth.ylsulfonamido)phenyl)boronic acid in place of (5~chlorothiophen-2~yl)boronic acid using the procedure described for Example 14a. Ή NMR (400 MHz, CD 3 OD) δ 9.05 - 9.03 (m, 1H), 8.84 - 8.81 (m, 1H), 8.25 (d, ./ 2.1 Hz, 1H), 8.1 1 - 8.06 (m, 1H), 7.97 (d, J = 8.6 Hz, 1H), 7.90 (d, J= 8.3 Hz, 1H), 7.73 - 7.69 (m, 1 H), 7.37 - 7.30 (m, 4H), 7.09 - 7.06 (m, 1H), 4.00 (s, 3H), 3.72 (s, 3H), 3.04 (s, 3H); MS (ESI): mass calcd. for C 28 H 23 CIF 3 N 5 O 4 S, 617.1 ; m/z found, 618.2 [M+H]".

Example 18a: 6~(Hydroxy(l-methyl-lH midazol-5-yl)(pyridm~2~yl)methyl)~3- p eny]quinoline~2,4~dicarbonitrile

(2,4-Dichloro-3-phenylquinolin-6-yl)(l -methyl- lH-imida.zol-5-yl)(pyridin-2-yl)methanol (227.8 mg, 0.494 mmol, Example 64), zinc cyanide (73.8 nig, 0.628 mmol), zinc dust (8.8 mg, 0.135 nimol), dppf (27.7 mg, 0.05 mmol), and Pu . '(dba) ; (22.6 mg, 0.0247 mmol) were charged to an oven-dried microwave vial. The vial was evacuated and back-filled with nitrogen. Dimethylacetamide (3 niL) was added to the mixture via. syringe. Nitrogen was bubbled through the reaction mixture for 5 minutes and the mixture was stirred and heated at 120 °C for 1 hour, followed by 100 °C for 3 hours under a positive pressure of nitrogen. The mixture was allowed to cool to ambient temperature, filtered through Celite ¾ , and rinsed with ethyl acetate. The filtrate was hasified with 5 M aqueous ammonium hydroxide, the layers were separated and the aqueous layer was extracted with excess ethyl acetate. The combined organic layers were dried (Na 2 S0 4 ), filtered, and concentrated to dryness. Due to an incomplete reaction as shown by LCMS (starting material and mono-cynated product present), the crude material was subjected to additional reaction conditions. In an oven-dried microwave vial, the dry crude material was mixed with zinc cyanide (80.1 mg, 0.682 mmol), zinc dust (8.7 mg, 0.133 mmol), X-Phos (32.7 mg, 0.0665 mmol), and Pd2(dba)3 (62.2 mg, 0,0679 mmol). The vial was evacuated and back-filled with nitrogen. Dimethylacetamide (3.4 mL) was added to the mixture via syringe. Nitrogen was bubbled through the reaction mixture for 5 minutes and the mixture was stirred and heated at 120 °C for 5 .25 hours under a positive pressure of nitrogen. The mixture was allowed to cool to ambient temperature, filtered through Celite ® , and rinsed with ethyl acetate. The filtrate was basified with 5 M aqueous ammonium hydroxide, the layers were separated and the aqueous layer was extracted with excess ethyl acetate. The combined organic layers were dried (Na 2 S0 4 ), filtered, and concentrated to dryness. The crude product was purified by flash column chromatography (silica gel, 0-5% MeOH-DCM) followed by reverse phase chromatography (acetonitrile/H 2 0 + 0.05% TFA). Product fractions were basified with saturated aqueous sodium bicarbonate and extracted with DCM, before being dried (Na 2 S0 4 ), filtered, and concentrated to provide the title compound. f H NMR (500 MHz, CDCI 3 ) 0 8.66 (d, J = 4.5 Hz, 1 1 1 ;·. 8.41 (d, J 1 .8 Hz, 1 H), 8.27 (d, J = 8.9 Hz, 1H), 8.06 (del, J = 8.9, 1.9 Hz, 1 1 1 ;·. 7.78 (td, J = 7.7, 1.7 Hz, 1 H), 7.64 - 7.58 (m, 51 1 ). 7.48 (s, 1 1 1 ). 7.38 - 7.35 (m, 1 1 1 ). 7.30 (d, J = 7.9 Hz, I I I ). 6.84 (s, 1 1 1 ;·. 6.40 (s, 1 1 1 ;·. 3.41 (s, 3H); MS m/e 443.2 | M S f j .

6-(Hydroxy(l -methyl- lH-imidazol-5-yl)(pyridin-2-yl)methyl)-3-phenylquinoline-2,4 - dicarbonitrile was purified by chirai SFC (ChiralPak AD, 100 % ethanol) to provide two pure enantiomers. The first eluting enantiomer was Example 18b: H NMR (500 MHz, CDCi 3 ) δ 8.66 (d, J = 4.3 Hz, 1H), 8.41 (d, J = 1.7 Hz, IH), 8.28 (d, J = 8.9 Hz, I f ! ). 8.06 (dd, J = 8.9, 1.9 Hz, 1H), 7.78 (td, J = 7.7, 1.7 Hz, IH), 7.66 - 7.57 (m, 5H), 7.51 (s, IH), 7.37 (ddd, J = 7.5, 4.9, 0.8 Hz, IH), 7.30 (d, J = 7.9 Hz, IH), 6.82 (s, IH), 6.41 (s, IH), 3.42 (s, 3H); MS m/e 443.2 [M+H] + . The second eluting enantiomer was Example 18c: ! H NMR (500 MHz, CDC1 3 ) δ 8.66 (d, J = 4.8 Hz, IH), 8.41 (d, J = 1.8 Hz, IH), 8.28 (d, J = 8.9 Hz, I H), 8.06 (dd, J = 8.9, 1.9 Hz, IH), 7.78 (td, J = 7.7, 1.6 Hz, IH), 7.66 - 7.58 (m, 5H), 7.48 (s, IH), 7.37 (dd, J = 7.1 , 5.2 Hz, IH), 7.29 (d, J = 7.9 Hz, IH), 6.79 (s, IH), 6.40 (s, IH), 3.41 (s, 3H); MS m/e 443.2 [M+Hf .

Example 19a: 6-(Hydroxy(l-methyI-lH midazoI-5-yl)(6- l)met!iy!)-2-met][ioxy~S~methy!~3~(4~^

To a round bottom flask containing (4-chloro-2-memoxy-8-methyl-3-(4- (trifluoromefhoxy)phenyi)quinolin-6-yi)( l -methyl- lH-imidazol-5-yl)(6-(trifluoromethyl)pyridin- 3-yl)methanol (0.19 g, 0.305 mmoi, Example 63) was added Zn(CN) 2 (64.47 mg, 0.596 mmol), Pd 2 (dba) (41.9 mg, 0.046 mmol), zinc nanopowder (5.9 mg, 0.092 mmol), and dicyclohexyl(2 , ,4',{v-misopropyl-[l, -biphenyl]-2-yl)phosphine (X-Phos, 29,9 mg, 0,061 mmol). The flask was evacuated and re-filled with argon (3x), Dmiethylacetamide (1.5 mL, degassed) was then added and the mixture was heated at 120 °C for 4 hours. The mixture was cooled to room temperature, diluted with EtOAc and filtered through Ceiite ® rinsing with EtOAc. The filtrate was diluted with saturated aqueous NH 4 CI and H 2 0 and layers were separated. The aqueous layer was again extracted with EtOAc. The combined EtOAc extract was washed with brine, dried over Na 2 S0 4 , filtered and evaporated in vacuo to provide an oil. LC/MS showed product plus a significant amount of starting material. The mixture was re-subjected to the conditions above and heated in a 120 °C oil bath for an additional 4 hours. After work-up (same as above) the crude product was purified by chromatography (10% MeOH in DCM on a gradient) to provide the title compound.

6-(Hydroxy(l -methyl- lH-imidazol-5-yl)(6-(trifluoromethyl)pyridin-3-yl)methyl)-2- methoxy-8- methyl-3-(4-(trifiuoromethoxy)phenyi)quinoline-4-carbonitril e was purified by chiral HPLC (Chiralpak (IC); solvent A = CO ? .; solvent B = methanol + 0,2% IP A; flow rate 50 mL/min: wavelength 254 nM; temperature 40 degrees) to give 2 enantiomers. The first eluting enantiomer was Example 19b: J H NMR (400 MHz, CDC1 3 ) δ 8.74 - 8.87 (m, 1 H), 8.06 - 8.16 (m, 1H), 7.91 - 8.02 (m, ! l l !. 7.65 - 7.74 (m, 1H), 7.54 - 7.65 (m, 2H), 7.42 - 7.52 (m, 2H), 7.32 - 7.41 (m, 2H), 6.44 - 6.60 (m, 1 1 1 ;·. 4.09 (s, 3H), 3.28 - 3.51 (s, 3H), 2.67 is. 3H); MS (ESI) 614 (M+H) + . The second eluting enantiomer was Example 19c: Ή NMR (400 MHz, CDCI 3 ) δ 8.75 - 8.86 (m, 1 H), 8.05 - 8.16 (m, 1 H), 7.89 - 8.02 (m, IH), 7.65 - 7.77 (m, 1 H), 7.54 - 7.63 (m, 2H), 7.42 - 7.50 (rn, 2H), 7.32 - 7.42 (m, 2H), 6.40 - 6.60 (m, 1H), 4.08 (s, 3H), 3.35 - 3.56 (broad s, 3H), 2.68 (s, 3H); MS (ESI) 614 { i l s .

Example 2Θ: i-(4-((2,4-Dichloro-8-niethyl-3-phenylquinolin-6- yl)(hydroxy)(phenyl)methyI)piperidm-l-yI)ethanone

A solution of n-butyllithium. (1 .6 M in hexanes, 0.5 mL, 0.8 rnmol) was added dropwise by syringe to a solution of 6-bromo-2,4-dichloro-8-methyl-3-phenylquinoline (0.32 g, 0.88 mmol. Intermediate 3) in dry deoxygenated THF (10 mL) at -78 °C. After 2 minutes, a solution of l-(4- benzoylpiperidin-l-yl)ethanone (0.203 g, 0.88 mmol, Intermediate 4) in dry THF (4 mL) was added dropwise by syringe. An additional 2 mL of THF was used to complete the quantitative addition. After 10 minutes, the flask was removed from the dry-ice bath and placed into an ice- water bath. After 1 hour, the reaction was quenched with saturated aqueous ammonium chloride solution and the mixture was partitioned between water and EtOAc. The layers were separated and the aqueous phase was further extracted with EtOAc and washed with saturated aqueous NaCl solution. The organic phase was dried (MgSQ 4 ), filtered, and concentrated to dryness. The crude product was purified by flash column chromatography (silica gel, 0-100% EtOAc-DCM) to provide the title compound. MS m/e 519.1 (M+H) + .

Example 21a: 6-((l-AcetyIpiperidin-4-yl)(hydroxy)(phenyI)methyl)-8-meth I-3- phenylqumol e-2,4-dicarbonitrile

A microwave vial was charged with l-(4-((2,4-dichloro-8-methyl-3-phenylquinolin-6- yl)(hydroxy)(phenyl)methyl)piperidin-l-yl)ethanone (240 mg, 0.46 mmol, Example 20), Zn(C ) 2 (55.7 mg, 0.480 mmol), Pd 2 (dba) 3 (43.3 mg, 0.047 mmol), zinc dust (6.05 mg, 0.0926 mmol), and dicyclohexyl(2',4',6'-triisopropyl- l ,l'-biphenyl]-2-yl)phosphine (X-Phos, 22.1 mg, 0.046 mmol). Dimethyiacetamide (2.4 mL) was then added and the mixture was sparged with nitrogen for 10 minutes and placed in a pre-heated aluminum block at 120 °C for 2 hours. The mixture was cooled to room temperature and was filtered through Celite*, and washed with EtOAc. The filtrate was washed sequentially with 2 M aqueous NH 4 OH and aqueous NaCl solution. The organic phase was dried (MgS0 4 ), filtered, and concentrated. The residue was purified by flash column chromatography (silica gel, 0 to 100% EtOAc-DCM). MS (ESI) 501 .3 ( M H) .

6-((l -Acetylpiperidin-4-yl)(hydroxy)(phenyl)methy3)-8-m.ethyl-3-p henylquinoline-2,4- dicarbonitrile was purified by chiral HPLC (Chiralcel OD, 80% heptane/20% ethanol) to give 2 enantiomers. The 2 enantiomers were further purified by reverse-phase HPLC (5-85% CH 3 CN- H 2 0, 0.05% TFA). The product was converted to the free base (neutralized with saturated aqueous NaHCC and extracted with EtOAc) and the organic fractions were concentrated to afford the title compound. The first eluting enantiomer was Example 21b: Ή NMR (600 MHz,

CDCI 3 , mixture of rotamers) δ ppm 8.35-8.32 (m, 1H), 7.84-7.81 (m, IH), 7.66-7.59 (m, 5H), 7.58-7.55 (m, 2H), 7.41-7.37 (m, 2H), 7.31-7.24 (m, 1H), 4.75 (d, J = 13.4 Hz, 0.5H), 4.69 (d, J = 13.5 Hz, 0.5H), 3.89 (d, J = 13.7 Hz, 0.5H), 3.82 (d, J = 13.7 Hz, 0 51 1). 3.1(S (t J = 12.2 Hz, 0.5H), 3.12-3.03 (m, 0 51 ) . 2.89-2.83 (m, 1H), 2.82 (s, 1.5H), 2.80 (s, 1.5H), 2.67-2.53 (m, lH), 2.38 (s, 0.5H), 2.36 (s, 0.5H), 2.07 (s, 1.5H), 2.06 (s, 1.5H), 1.46-1.21 (m, 4H); MS m/e 501.2 [M+H] + and the second eluting enantiomer was Example 21c: 3 H NMR (600 MHz, CDCI 3 , mixture of rotamers) 5 ppm 8.35-8.32 (m, 1 H), 7.84-7.81 (m, 1H), 7.65-7.59 (m, 5H), 7.58-7.55 (m, 2H), 7.41-7.37 (m, 2H), 7.31-7.24 (m, 1H), 4.75 (d, J = 13.5 Hz, 0.5H), 4.69 (d, J = 13.5 Hz, 0.5H), 3.89 (d, J = 13.7 Hz, 0.5H), 3.82 (d, J = 13.6 Hz, 0.5H), 3.18-3.14 (m, 0.5H), 3.1 1-3.07 (m, 0.5H), 2.86-2.83 (m, 1 H), 2.82 (s, 1 .51 1 ). 2.80 (s, 1.5H), 2.68-2.54 (m, 1 H), 2.37 (s, 0.5H), 2.35 (s, 0.5H), 2.07 (s, 1 .5H), 2.06 (s, 1.5H), 1.47-1.21 (m, 4H); MS m/e 501.2

Example 22: (4-ChlorophenyI)(2,4-dichIoro-8-methyl-3-phenylquinoUn-6-yl) (l-methyl-l/ - iniidazol-5-yl)metha»ol

The title compound was prepared analogously to the method described in Example 20 using (4- chlorophenyl)(l -methyl- lH-imidazol-5-yl)methanone (Intermediate 8: step b) in place of l -(4- benzoylpiperidin-l-yl)ethanone (Intermediate 4). MS m/e 508.1 (Μ+Η) Γ .

Example 23a: 6-((4-Chlorophenyl)(hydroxy)(l-methyl-l//-imidazol-5-yl)niet hyl)-8-niethyl- 3-phenylquinoline-2,4-dicarbonitrile

The title compound was prepared analogously to the method described in Example 25 a using (4- chk>rophenyl)(2,4-dichloro-8-m.ethyl-3-phenylquinolin-6-y l)(i-m

yl)methanol (Example 22) in place of 1 -(4-((2,4-dichloro-8-methyi-3-phenylquinolin-6- yi)(hydroxy)(phenyi)methyl)piperidin-l-yl)ethanone (Example 20). MS m/e 490.2 (M+H) "h .

6~((4-Chiorophenyl)(hydroxy)( 1 -methyl- 1 H-iniida.zol-5-yl)methyl)-8-methyl-3-phenylqirinolir!e~ 2,4-dicarbonitrile was purified by chiral HPLC (Chiralcel OD, 50% heptane/20% ethanol) to give 2 enantiomers. The 2 enantiomers were further purified by reverse-phase HPLC (5-85% CH 3 CN-H 2 O, 0.05% TFA). The product was converted to the free base (neutralized with saturated aqueous NaHC0 3 and extracted with EtOAc) and the organic fractions were concentrated to afford the title compound. The first eliiting enantiomer was Example 23b: ! H NMR (600 MHz, CDCI 3 ) δ ppm 8.21 (s, i l l ). 7.62-7.49 (m, 61 1 ). 7.24 (s, 5H), 6.29 (s, 1 H), 3.32 (s, 3H), 2.70 (s, 3H); MS m/e 490.2 [M+H] ' and the second eluting enantiomer was Example 23c: 5 H NMR (600 MHz, CDCi 3 ) δ ppm 8.29 (d, J = 1.5 Hz, i l l ). 7.67-7.56 (m, 61 1 ). 7.35-7.28 (m, 4H), 7.14 (s, 1H), 6.27 (s, 1H), 3.35 (s, 3H), 2.75 (s, 3H); MS m/e 490.2 [M+Hf .

Example 24: (2,4-Dichloro-8-methyI-3-pheny]qumolm^

(tri.fl«oromethy1)pyridin-3-yI)methanol

The title compound was prepared analogously to the method described in Example 20 using (1- methyl-lH-imidazol-5-yl)(6-(trifluoromethyl)pyridin-3-yi)met hanone (Intermediate 2: step c) in place of l-(4-benzoylpiperidin-l-yl)etha.none (Intermediate 4). MS m/e 543.1 (Μ+Η) .

Example 25a: 6-(Hydroxy(l-methyl-lH midazol-5-yI)(6-(trifluoromethyl)pyridin-3- yl)methyl)-8-methyl-3-phenyIqiiinolme-2,4-dicarbonitriIe

The title compound was prepared analogously to the method described in Example 21a using (2,4-dichloro-8-methyl-3-phenylqumolin-6-yl)(l -methyl- lH-imidazol-5-yl)(6- (trifluoroniethyl)pyridin-3-yI)methanol (Example 24) in place of l ~(4-((2,4-dichloro~8-methyl~3- phenylquinolin-6-yl)(hydTox.y)(phenyl)methyl)piperidin-l -yl)ethanone (Example 20), MS m/e 525,2 ( VI · Η ) .

6-(Hydroxy(l -methyl- ]J/-imidazol-5-yl)(6-(†Tifluoromethyl)p Tidm-3-y3)m

phenyIquinoline-2,4-dicarbonitrile was purified by chiral HPLC (Chiralcel OD, 80% heptane/20% ethanol) to give 2 enantiomers. The first eluting enantiomer was Example 25b: Ή

NMR (600 MHz, CDC ) δ ppm 8.81 (d, J = 2.1 Hz, 1H), 8.33 (d, J = 1.9 Hz, 1H), 7,98 (dd, J = 8.3, 2,2 Hz, I H), 7.71 (d, . = 8.2 Hz, i f ! }. 7.66-7.59 (m, 6H), 7.37 (s, I H ). 6.45 (s, IH), 5 ,29 (s, 1H), 3.41 ($, 3H), 2,80 (s, 3H); MS m/e 525.2 (M+H) '1' and the second eluting enantiomer was Example 25c: ; H NMR (600 MHz, CDC1 3 ) δ ppm 8.80 (d, ./ = 2.1 Hz, I H), 8.33 (dd, ./ - 2.2, 0.7 Hz, I I I ). 7.97 (dd, J - 8.3, 2.2 Hz, III), 7.70 (dd, J - 8.2, 0.8 Hz, I H), 7.67-7.58 (m, 6H), 7.31 (s, IH), 6.38 (d, J = 1 .2 Hz, IH), 5.94 (s, I H), 3.39 (s, 3H), 2.79 (s, 3H); MS m/e 525.2 (M+H) .

The title compound was prepared analogously to the method described in Example 20 using 1 - (4-(6-(trifluoromethyl)nicotinoyl)piperidin- 1 -yl)ethanone (Intermediate 14: step d) in place of 1 - (4~benzoylpiperidin-l-yl)etlianone (Intermediate 4). MS m/e 588.2 (M+H) "1" .

Example 27a: 6-((l-AcetyIpiperidin-4-yl)(hydroxy)(6-(trifluoromethyl)pyri din-3- yl)methyl)-8-methyl-3-phenylquiiioline-2,4-dicarboaitrile

The title compound was prepared analogously to the method described in Example 21a using 1 - (4-((2,4-clichloro-8-methyl-3-phenyiqui

yl)methyl)piperidin~l-yI)ethanone (Example 26). MS m e 570.3 (M+H) + . 6-((l-Acetylpiperidin-4-yl)(hydroxy)(6-(triiIuoromethyi)pyri din-3-yl)m

phenylquinoline-2,4-dicarbonitrile was purified by chiral SFC (Chiralpak AD-H, 5 μηι, 250 x 20 mm, mobile phase: 75% C0 2 , 25% mixture of methanol-isopropanol 50/50 v/v). The first eluting enantiomer was Example 27b: Ή NMR (600 MHz, CDCI 3 , mixture of rotamers) δ ppm 8.98- 8.94 (m, 1H), 8.41-8.32 (m, 1H), 8.16-8.13 (m, 1H), 7.87 (s, 0.5H), 7.79 (s, 0.5H), 7.71-7.66 (m, IH), 7.65-7.56 (m, 5H), 4.74-4.68 (m, IH), 3.96-3.64 (m, 2H), 3.21-3.08 (m, IH), 2.93- 2.88 (m, IH), 2.84 (s, 1.5H), 2.83 (s, 1.5H), 2.65-2.60 (m, IH), 2.03 (s, 1.5H), 2.02 (s, 1.5H), 1.58-1.34 (m, 4H); MS m e 570.3 (M+H) " and the second eluting enantiomer was Example 27c: Ή NMR (600 MHz, CDCk mixture of rotamers) δ ppm 9.01-8.93 (m, IH), 8.39-8.36 (m, IH), 8.16-8.13 (m, IH), 7.87 (s, 0.5H), 7.79 (s, 0.5H), 7.69-7.67 (m, IH), 7.66-7.58 (m, 5H), 4.74- 4.68 (m, I H), 3.97-3.64 (m, 2H), 3.22-3.06 (m, IH), 2.93-2.88 (m, IH), 2.84 (s, 1 .5H), 2.83 (s, 1.5H), 2.70-2.53 (m, IH), 2.03 (s, 1.5H), 2.02 (s, 1.5H) 1.58-1.34 (m, 4H); MS m/e 570.3

Example 28 : 6-((4-CyaHophenyl)(faydroxy)(l -methyI-lH-imidazol-5-yI)methyI)-3-pheiiyl-2- (trifluoromethyl)quinoHne-4-carbonitrile*TFA

The title compound was isolated from the reaction that formed Example 3.1 a. H NMR (400 MHz, CD3OD) δ 9.04 (s, IH), 8.40 (d, ,/ 9.09 Hz, IH), 8.34 (d, ,/ 2.02 Hz, IH), 8.06 (dd, ./ 2.27, 8.84 Hz, IH), 7.84 (d, J = 8.59 Hz, 2H), 7.70 (d, J = 8.59 Hz, 2H), 7.54 - 7.60 (m, 3H), 7.43 - 7.49 (m, 2H), 7.09 (d, J = 1.52 Hz, IH), 3.70 (s, 3H); MS m/e 510.1 [M+H] + .

Example 29: l-(4-((4-Chloro-3-phenyl~2~(trifluoromethyl)quinolin~6~yl)(h ydroxy)^yridin -yI)methy1)piperidm-l-yI)ethanone*TFA

A mixture of (4-chloro-3-phenyl-2-(trifluoromethyl)quinolin-6-yl)(piperid in-4-yl)(pyridin-3- yi)methanol (198 mg, 0.400 mmoi, Example 70), acetyl chloride (0,060 niL, 0.84 mmol), and Etj (0.16 mL, 1.15 mmol) in CHiCi? (4 mL) was stirred at room temperature for 1.5 hours and then concentrated to dryness. The residue was purified by reverse phase HPLC (water/acetonitrile/0.1 % TFA) to provide the title compound as a white solid. l H NM (400 MHz, CD 3 OD) δ 9.13 (s, ! H ). 8.82 (t, ./ 8.08 Hz. I I I ). 8.69 - 8.75 (m, 2H), 8.27 (d. ./ 9.09 Hz, l i l !. 8.18 - 8.24 (m, 1H), 7.94 - 8.02 (m, 1 H), 7.49 - 7.55 (m, 3H), 7.26 - 7.34 (m, 2H), 4.55 - 4.65 (m, 1 H), 3.91 - 4.05 (m, 1H), 3.15 - 3.27 (m, 2H), 2.67 - 2.77 (m, III), 2.08 (d, ./ 4.04 Hz, 3H), 1.52 (m, 4H); MS m/e 540.3 i .Yi - i l j .

Example 30a: 6-((l-Acetylpiperidiii-4-yl)(hydroxy)(pyridiii-3-yI)methyI)- 3-pheiiyl-2- (trifluoromethyi)quinoline-4-carbonitrile » TFA

The title compound was prepared using l -(4-((4-chloro-3-phenyl-2-(trifluoromethyl)quinolin-6- yl)(hydroxy)(pyridin-3-yl)rnethy])piperidin~l -yl)ethanone e TFA (Example 29) in place of (4- chloro-3-phenyl-2-(trifluoromethy})quinolin-6-yl)(4-chloroph eny

yl)rnethanol » TFA (Example 62) using the procedure described for Example 31 a, with the exception that the reaction mixture was heated at 120 °C for 10 hours. Ή NMR (400 MHz, CD 3 OD) 5 9.17 (s, 1H), 8.87 - 8.93 (m, 1H), 8.74 (d, J = 5.56 Hz, 1 H), 8.62 (dd, J = 2.02, 5.56 Hz, 1H), 8.37 (d, ,/ 9.09 Hz, 1 H), 8.25 - 8.32 (m, 1H), 8.00 - 8.07 (m, 1 H), 7.53 - 7.62 (m, 3H), 7.41 - 7.50 (m, 2H), 4.55 - 4.66 (m, 1H), 3.92 - 4.03 (m, 1H), 3.16 - 3.28 (m, 2H), 2.68 - 2.77 (m, 1H), 2.08 (d . ./ 5.05 Hz, 3H), 1.41 - 1.67 (m, 4H): MS m/e 531.3 | V! H |

6-((l -Acetylpiperidin-4-yl)(¾ydroxy)(pyridin-3-yl)methyl)-3-phen yl-2-

(trifluoromefhyi)quinoline-4-carbonitrile was neutralized by partitioning between saturated NaHCOa aqueous solution and DCM (xl) followed by EtOAc (x3). The organic layer was dried (Na?SQ4), filtered, concentrated to dryness, and purified by chiral HPLC (Chiralcel OD, CH 3 CN) to give two pure enantiomers. Example 30b: (first enantiomer to elute off chiral column) Ή NMR (400 MHz, CDCI 3 , -1 : 1 mixture of retainers) δ 8.77 - 8.94 (m, 1H), 8.54 (dd, J = 1 .52, 12.13 Hz, 1H), 8.38 - 8.50 (m, 1H), 8.29 (t, J = 9.09 Hz, 1H), 8.07 (dd, J = 2.02, 9.09 Hz, 0.5H), 7.99 (dd, J= 2.02, 9.09 Hz, 0.5H), 7.95 (d, J= 8.08 Hz, 1H), 7.47 - 7.65 (m, 3H), 7.36 - 7.42 (m, 2H), 7.19 - 7.34 (m, 1H), 5.55 - 5.84 (m, 2H), 5.08 - 5.18 (m, 1H), 4.56 - 4.77 (m, 1H), 3.71 - 3.93 (m, 1H), 3.00 - 3.21 (m, 1H), 2.78 - 2.96 (m, 1H), 2.52 - 2.66 (m, 1H), 1.37 - 1.75 (m, 3H), 1.22 - 1.35 (m, 1H); MS m/e 531.3 [M+H] + . Example 30c: (second enantiomer to elute off chiral column) ! H MR (400 MHz, ( IX " ! :. -4 : 1 mixture of rotamers) δ 8.79 - 8.93 (m, 1H), 8.54 (dd, J = 1.52, 11.62 Hz, 1H), 8.40 - 8.50 (m, IH), 8.29 (t, J = 8.34 Hz, 1H), 8.06 (dd, J = 2.02, 9.09 Hz, 0.5H), 7.99 (dd, J = 2.02, 9.09 Hz, 0.5H), 7.94 (d, J ------ 8.08 Hz, 1H), 7.51 - 7.59

(m, 3H), 7.36 - 7.44 (m, 2H), 7.25 - 7.34 (m, IH), 5.62 - 5.95 (m, 2H), 4.93 - 5.03 (m, IH), 4.59 - 4.76 (m, IH), 3.76 - 3.90 (m, IH), 3.03 - 3.20 (m, IH), 2.80 - 2.93 (m, IH), 2.52 - 2.68 (m, IH), 1.36 - 1.74 (m, 3H), 1.20 - 1.36 (m, IH); MS m/e 531.3 [M+H] + .

Example 31a: 6-((4-CMoropheKyI)(liydroxy)(l-me

-(trifliiorosBeiiiyI)qiiiiio iie-4-carboiiitriIe®TFA

A pressure tube containing (4-chloro-3-phenyl-2-(trifluoromethyl)quinolin-6-yl)(4- chlorophenyl)(l -methyl- lH-imidazo}-5-yl)rnethanol » TF A (145 mg, 0.190 mmo!, Example 62), Pda(dba)3 (18 mg, 0,020 rnmol), dicyclohexyl(2',4',6 , -triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (X-Phos, 10 mg, 0.021 rnmol), zinc cyanide (11.5 mg, 0,098 rnmol), and zinc nanopowder (2.5 mg, 0.038 rnmol) in N,N-dimethylacetamide (5 mL) was sparged with nitrogen for 8 minutes, and then heated at 120 °C for 2.5 hours. The mixture was allowed to cool to room temperature and filtered through a syringe filter. The filtrate was concentrated in vacuo, EtOAc and saturated NH 4 CI aqueous solution were added. The organic layer was separated, and the aqueous layer was extracted with EtOAc. The combined organic layers were dried (Na 2 S0 4 ), filtered, and concentrated to dryness. The residue was purified by reverse phase HPLC (water/acetonitrile/0.1 % TFA) to provide the title compound as a white solid, 'ϊί NMR (400 MHz, CD 3 OD) δ 9.02 (s, IH), 8.39 (d, ,/ 8.59 Hz, IH), 8.34 (d, ,/ 2.02 Hz, IH), 8.05 (dd, ./ 2.27, 8.84 Hz, 1H), 7.52 - 7.63 (m, 3H), 7.42 - 7.50 (m, 6H), 7.04 (s, 1H), 3.71 (s, 3H); MS m/e 519.2 ! - l i j ' .

6-((4-Chlorophenyl)(hydroxy)( 1 -methyl- lH-imidazoi-5-yl)methyl)-3-phenyl-2- (trifluoromethyi)quinoline-4-carbonitrile was neutralized by partitioning between saturated NaHC0 3 aqueous solution and DCM. The organic layer was dried (Na 2 S0 4 ), filtered, concentrated to dryness, and purified by chiral HPLC (Chiralcel OD, 90% heptanes/10% EtOH) to give two pure enantiomers. Example 31b: (first enantiomer to elute off chiral column) Ή NMR (400 MHz, CD(¾) δ 8.46 (s, i l l ). 8.23 (d, J = 9.09 Hz, 1 1 1 ). 7.78 (dd, J = 2.02, 9.09 Hz, l i l !. 7.52 - 7.62 (m, 3H), 7.36 - 7.43 (m, 2H), 7.28 - 7.36 (m, 4H), 7.18 - 7.28 (m, IH), 6.29 (s, 1 H), 3.37 (s, 3H); MS m/e 519.2 [M+H] " . Example 31c: (second enantiomer to elute off chiral column) 3 H NMR (400 MHz, CDCI3) δ 8.45 (s, 1H), 8.23 (d, J = 9.09 Hz, IH), 7.78 (d, J = 9.09 Hz, IH), 7.50 - 7.64 (m, 3H), 7.35 - 7.43 (m, 2H), 7.27 - 7.35 (m, 4H), 7.19 - 7.28 (m, I H), 6.28 (s, IH), 3.37 (s, 3H); MS m/e 519.2 j \I ! i j .

Example 32a: (4-Methoxy-3-phenyl-2-(trifluoromethyl)quinolin-6-yl)(l-meth yl-l//- methanoi » TFA

A. mixture of (4-chloro-3-phenyl-2-(trifiuoromethyl)quinolin-6-yl)(l -me† l-lH-imidazol-5- yl)(pyridin-2-yl)methanol » TFA (53 mg, 0.073 mmol, Example 61 ) and 0.5 M NaOMe in MeOH (0.80 mL, 0.40 mmol) in a sealed tube was heated at 70 °C for 2 hours. The solvent was evaporated, and water was added. The mixture was extracted with EtOAc, dried (Na?S()4), filtered, and concentrated. The residue was purified by reverse phase HPLC (water/acetonitrile/0.1 % TFA) to provide the title compound as a white solid. l H NMR (400 MHz, CD 3 OD) δ 8.96 (s, IH), 8.62 (d, J = 4.04 Hz, IH), 8.41 (d, J = 2.02 Hz, IH), 8.19 (d, J = 9.09 Hz, I H), 8.02 (dd, ./ 2.02, 9.09 Hz, IH), 7.93 (td, ./ 2.02, 8.08 Hz, IH), 7.79 (d, ,/ 8.08 Hz, IH), 7.47 - 7.52 (m, 3H), 7.36 - 7.45 (m, 3H), 7.08 (d, ./ 1.52 Hz, IH), 3.63 (s, 3H), 3.55 is. 3H); MS m/e 491.2 I V! - i l j .

I l l The racemate was neutralized by partitioning between saturated NaHC0 3 aqueous solution and DCM, The organic layer was dried (Na 2 S0 4 ), filtered, concentrated to dryness, and purified by chiral HPLC (Chiraicel OD, 90% heptanes/10% EtOH) to give two enantiomers. The first eluting enantiomer was Example 32b: J H NMR (400 MHz, CDC1 3 ) δ 8.64 (d, J = 4.55 Hz, 1H), 8.18 - 8.24 (m, 2H), 7.88 (dd, J = 2.02, 9.09 Hz, III), 7.72 (td, J = 1.52, 7.58 Hz, 1H), 7.50 (s, 1H),

7.43 - 7.49 (m, 3H), 7.30 - 7.40 (m, 3H), 7.23 (d, J = 8.08 Hz, 1 H), 6.37 (s, 1H), 3.50 (s, 3H),

3.44 (s, 3H); MS m/e 491.2 f X ! f ! j .

Example 33a: (4-Chlorophenyl)(4-methoxy-3^henyl-2-(trifluoromethyl)quinol iii-6-yl)(l- methyI-l// midazol-5-yl)methanol *TFA

A mixture of (4-chloro-3-phenyl-2-(tTifluOromethyl)quinolin-6-y3 (4-chloropheny].)(l -inethyl- lH-imidazol-5-yl)rnethanol*TFA (80 mg, 0.1 1 mmol, Example 62) and 0.5 M NaOMe in MeOH (0.50 niL, 0,25 mmol) in a sealed tube was heated at 70 °C for 7 hours. More 0,5 NaOMe in MeOH (0.35 niL, 0.18 mmol) was added and the mixture was heated at the same temperature for another hour. The solvent was evaporated, and DMSO was added. After filtering through a syringe filter, the filtrate was purified by reverse phase HPLC (water/acetonitrile/Q.1 % TFA) to provide the title compound as a white solid. 3 H NMR (400 MHz, CD 3 OD) δ 9.00 (s, 1 H), 8.25 (d, J ------ 2.02 Hz, 1H), 8.23 (d, ,/ 9.09 Hz, 1H), 7.88 (dd, ./ 2.02, 9.09 Hz, 1H), 7.47 - 7.52 (m,

4H), 7.41 - 7.47 (m, 3H), 7.36 - 7.41 (m, 2H), 6.97 (s, 1H), 3.71 (s, 3H), 3.53 (s, 3H); MS m/e 524.3 [M+H] + .

(4-Chlorophenyl)(4-methoxy-3-phenyl-2-(trifluoromethyl)quino lin-6-yl)(l -methyl- lH-i

5-yl)methanol was neutralized by partitioning between saturated NaHC0 3 aqueous solution and DCM. The organic layer was dried (Na 2 S0 4 ), filtered, concentrated to dryness, and purified by chiral HPLC (Chiralpak Q] , 100%) MeOH) to give two enantiomers. The first eluting enantiomer was Example 33b: Ή NMR (400 MHz, CDCI 3 ) δ 8.14 - 8.20 (m, 2H), 7.77 (dd, J = 2.27, 8.84 Hz, 1H), 7.44 - 7.55 (m, 3H), 7.34 - 7.40 (m, 2H), 7.29 - 7.34 (m, 4H), 7.24 - 7.29 (m, 1 H), 6.34 (br. s., 1 H), 3.48 (s, 3H), 3.36 (s, 3H); MS m/e 524.3 [M+H] " and the second eluting enantiomer was Example 33c: Ή NMR (400 MHz, CDC1 3 ) δ 8.14 - 8.20 (m, 2H), 7.77 (dd, J = 2.02, 9.09 Hz, 1 H), 7.44 - 7.50 (m, 3H), 7.36 (d, J = 4.04 Hz, 2H), 7.29 - 7.34 (m, 4H), 7.23 - 7.29 (rn, 1 H), 6.32 - 6.36 (m, 1 H), 3.48 (s, 3H), 3.37 (s, 3H); MS m/e 524.1 [M+H] + .

Example 34a: l-(4-((4-Chloro-3-phenyi-2-(trifliioromethyl)quinoIin-6-yI)( hydroxy)(l- methyI-l// midazoi-5-yI)methyl)piperidin-l-yl)ethanone

The racemic title compound was prepared using 1 -(4-( 1 -methyl- 1 H-imidazole-5- carbonyl)piperidin- l-yl)ethanone (Intermediate I : step c) in place of (4-chl.orophenyl)(l -methyl- l.H-imidazol-5-yl)methanone (Intermediate 8: step b) using the procedure described for Example 62, with the exception that the reaction was carried out at -78 °C ah the time. Ή NMR (400 MHz, CD 3 OD, -1 : 1 mixture of rotamers) δ 8.86 (s, 1H), 8.56 (d, J = 4.04 Hz, 1 H), 8.27 (d, J = 9.09 Hz, 1H), 8.09 (s, IH), 7.81 i d. ./ = 9.09 Hz, IH), 7.45 - 7.58 (m, 3H), 7.31 (d, ./ = 2.53 Hz, 2H), 4.66 (d, ,/ 13.14 Hz, 0.5H), 4.46 (d, J = 12.63 Hz, 0.5H), 4.04 (d, ./ 13.64 Hz, 0.5H), 3.84 (d, 13.64 Hz, 0.5H), 3.60 (s, 1.5H), 3.58 (s, 1.5H), 3.25 - 3.30 (overlap with solvent, m, 0.5H), 3.05 (td, J --- 2.53, 13.14 Hz, 0.5H), 2.67 - 2.86 (m, 1.5H), 2.55 (td, J = 3.03, 12,88 Hz, 0.5H), 2.09 - 2.28 (m, IH), 2.08 (s, 1.5H), 2.03 (s, 1.5H), 1.41 - 1.63 (rn, IH), 1.27 - 1.41 (m, IH), 1.05 - 1.27 (m, IH); MS m/e 543.2 [M+Hf .

The racemate was neutralized by partitioning between saturated NaHC0 3 aqueous solution and DCM. The organic layer was dried (Na 2 S0 4 ), filtered, concentrated to dryness, and purified by cliiral HPLC (Chiralcel OD, 100% EtOH) to give two pure enantiomers. Example 34b: (first enantiomer to elute off chiral column) 1H MR (400 MHz, CDCI 3 , - 1.5: 1 mixture of rotamers) δ 8.45 (d, ./ === 13.64 Hz, IH), 8.20 (dd, ./ === 4.04, 8.59 Hz, IH), 7.68 (t, ./ == 10.1 1 Hz, 1 1 1 ). 7.48 - 7.53 (m, 3H), 7.24 - 7.34 (m, 2H), 7.10 - 7.24 (m, 2H), 4.65 (d, ,/ 12.63 Hz, 0.6H), 4.56 (d, J = 13.14 Hz, 0.4H), 3.91 (d, ,/ 13.14 Hz, 0.41 1 ). 3.66 - 3.79 (m, 0.6H), 3.31 (s, 1.8H), 3.29 is. 1.2H), 3.18 (1. ./ 12.38 Hz, 0.4! i ). 2.97 (t, ./ 12.13 Hz, 0.61 1 ). 2.38 - 2.69 (m, 2H), 2.34 (d, ./ 1 3. 1 4 Hz, 0.41 ! ). 2.22 (d, ./ 12.63 Hz, 0.6H), 2.01 (s, 1.8H), 1.95 (s, 1.2H), 1.13 - 1.55 (m, 2.4H), 1,09 (d, J= 12.63 Hz, 0.6H); MS m/ ' e 543.2 jV! H| . Example 34c: (second enantiomer to elute off chiral column) 1H NMR (400 MHz, CDC1 3 , -1.5:1 mixture of rotamers) 88.46 (d, J = 12.63 Hz, 1H), 8.20 (del, ,/ 3.79, 8.84 Hz, 1H), 7.68 ίί../ 10.11 Hz, 1H), 7.46 - 7.53 (m, 3H), 7.24 - 7.35 (m, 2H), 7.10 - 7.23 (m, 2H), 4.64 (d, J= 13.14 Hz, 0.6H), 4.55 (d, J - 12.63 Hz, 0.4H), 3.90 id../ = 13.14 Hz, 0.4! I).3.72 (d, ./ = 13.14 Hz, 0.611;·.3.31 (s, 1.8H), 3.29 (s, 1.211).3.18 (1../ 12.38 Hz, 0.4H), 2.97 (t, ./ 12.38 Hz, 0.6H), 2.38 - 2.68 (m, 211;·.2.34 (d, J = 12.63 Hz, 0.4H), 2.21 (d, J= 13.14 Hz, 0.6H), 2.01 (s, 1.8H), 1.94 (s, 1.2H), 1.12 - 1.58 (m, 2.411).1.08 (d, ./ = 12.63 Hz, 0.611}: MS m c 543.2 jY! 11| .

Example 35: (4-CMoro~2~meihoxy-3-ptasylqMiraollii-^^

yl)metiiaiiol ® TFA

To a flask containing (2,4-dichloro-3-phenylqmnolin-6-yl)(2,4-dime1hylihiazol-5-yr )( 1 -methyl- 1H- 1 ,2,3-triazol-5-yl)methanol (100 mg, 0.2 mmol. Example 67) was added MeOH (6 mL) followed by solid NaOMe (50 mg, 0.88 mmol, 95% purity) at room temperature. The reaction mixture was heated to reflux for 24 hours, then allowed to cool to room temperature, filtered through Celite* and rinsed with MeOFI, The MeOH was removed under reduced pressure and the residue was diluted with water and extracted with EtOAc (3 x 40 mL), The combined organics were washed with brine, dried over MgS0 , filtered and concentrated to dryness. Purification by RP-HPLC (water / acetonitrile / 0.05% TFA) afforded the title compound. Ή NMR (500 MHz, CDCI 3 ) δ 8.23 (d, J = 2.1 Hz, 1H), 7.88 (d, J === 8.8 Hz, 1H), 7.57 - 7.39 (m, 4H), 7.38 - 7.30 (m, 2H), 7.17 (s, IH), 4.75 (s, 1H), 4.02 (s, 3H), 3.92 (s, 3H), 2.56 is, 3H), 2.12 (s, 311}: MS (ESI): mass calcd. for C25H22CIN5O2S, 491.1, m/z found 492.0 IV! -llj .

Example 36: (2-Chloro-4-methoxy-3-phenylqiiinoHn-6-yI)(2,4-dimethylthiaz oi-5-yI)(l- methyI-.l/-l,2,3-triazol-5-yl)methanol » TFA

To a flask containing (2,4-dichloro-3-phenylqxiinolin-6-yl)(2,4-dimethylthiazol-5- yl)( 1 -methyl- 1H- 1 ,2,3-triazol-5-yl)methanol (100 mg, 0.2 mmol. Example 67) was added MeOH (6 mL) followed by solid NaOMe (50 mg, 0.88 mmol, 95% purity) at room temperature. The reaction mixture was heated to reflux for 24 hours, then allowed to cool to room temperature, filtered through Celite 6 ' and rinsed with MeOFI, The MeOH was removed under reduced pressure and the residue was diluted with water and extracted with EtOAc (3 x 40 mL), The combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated to dryness. Purification by RP-HPLC (water / acetonitrile / 0.05% TFA) afforded the title compound. Ή NMR (500 MHz, CDC1 3 ) 6 8.13 (d, J = 2.1 Hz, IH), 8.04 (d, J = 8.9 Hz, 1H), 7.68 (dd, J = 8.9, 2.3 Hz, 1H), 7.54 - 7.44 (m, 3H), 7.43 - 7.39 (m, 2H), 7.23 (s, 1 H), 3.95 (br. s, 1H), 3.93 (s, 3H), 3.48 (s, 3H), 2.59 (s, 3H), 2.16 (s, 3H); MS (ESI): mass calcd. for C 25 H 22 CIN 5 O 2 S, 491.1 , m/z found 492,0 [M+H] + .

Example 37: (4-Chloro-2-methoxy-3-phenyiqiiinolin-6-yI)(2,4-dimethyithia zol-5-yI)(.l- methyI-lH-imidazoi-5-yl)methanol*TFA

To a microwave vial containing (2,4-dichloro-3-phenylquinolin-6-yl)(2,4-dimethylthiazoI-5- yl)(l -methyl-lH-imidazol-5-yl)methanol (120 mg, 0.24 mmol. Example 68) was added MeOH (4 mL) followed by solid NaOMe (60 mg, 1 .1 1 mmol, 95% purity) at room temperature. The vial was sealed and evacuated and the mixture was heated to 75 °C for 5.5 hours. The MeOH was removed under reduced pressure and the residue was diluted with water and extracted with EtOAc (4 x 25 mL). The combined organics were washed with brine, dried over MgS0 , filtered and concentrated to dryness. Purification by RP-HPLC (water / acetonitrile / 0.05% TFA) afforded the title compound. ! H NMR (500 MHz, CD 3 OD) δ 9.00 (s, 1H), 8.30 (d, J = 2.0 Hz, IH), 8.01 (d, J = 8.9 Hz, H i ).. 7.81 (dd, J 8.9, 2.1 Hz, 1H), 7.60 - 7.37 (m, 5H), 7.26 (s, IH), 3.75 (s, 3H), 3.55 (s, 3H), 2.62 (s, 3H), 2.25 (s, 3H); MS (ESI): mass calcd. for C 26 H 23 CIN 4 O 2 S, 490.1 : m/z found 491.1 [M+H] ; .

Example 38: (2-Chloro-4-methoxy-3-phenylq«inoliii-6-yl)(2,4-dimethylthi azol-5-yl)(l- methyI-lH-imidazol-5-yI)methanol » TFA

To a microwave vial containing (2,4-dichloro-3-phenylquinoiin-6-yl)(2,4-dim.ethylthiazol-5- yl)(l -methyl-lH-imidazol-5-yi)methanol (520 rag, 0.24 mmol, Example 68) was added eOH (4 mL) followed by solid NaOMe (60 nig, 1 .1 1 mmol, 95% purity) at room temperature. The vial was sealed and evacuated and the mixture was heated to 75 °C for 5.5 hours. The MeOH was removed under reduced pressure and the residue was diluted with water and extracted with EtOAc (4 x 25 mL). The combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated to dryness. Purification by RP-HPLC (water / acetonitri!e / 0.05% TFA) afforded the title compound. l H NMR (500 MHz, CD 3 OD) δ 9.00 (s, i l l ). 8.28 (d, ./ 2.1 Hz, IH), 7.98 (d, J 8.8 Hz, 1H), 7.74 (dd, ,/ 8.8, 2.2 Hz, i l l ). 7.51 - 7.38 (m, 3H), 7.35 - 7.27 (m, 2H), 7.24 (s, I H), 4.00 (s, 3H), 3.75 (s, 3H), 2.62 (s, 3H), 2.25 (s, 3H); MS (ESI): mass calcd. for C26H23CIN4O2S, 490.1 ; m/z found 491 .1 [M+H] + .

Example 39: (2,4-Dichloro-3-phenylquinolin-6-yl)(l,2-dimethyl-lH midazoI-5-yl)(l- methyI-.l/ -l,2,3-triazol-5-yl)methanol

To a flask containing 1 -methyl- lH-l ,2,3-triazole (125 mg, 1.5 mmol) was added THF (10 mL) and the colorless solution was cooled to -45 °C. Then, #-BuLi (2.5 M in hexaiies, 0,6 mL, 1.5 mmol) was added affordi g a suspension. The suspension was stirred between -40 °C and -10 °C for 30 minutes, then a THF solution of (2,4-dichloro-3-phenylquinolin-6-yl)(l,2-dimethyl-lH- imidazol-5-yl)niethanone (500 mg, 1.26 mmol, Intermediate 18: step b, in 5 mL THF) was introduced and the mixture was allowed to warm up to room temperature. After 1 hour, the reaction mixture was heated to 40 °C for 3 hours and then quenched with aqueous NH 4 CI solution. The aqueous portion was extracted with EtOAc (3 x 50 mL) and then with DCM (3 x 50 mL). The individual organic portions were washed with brine, dried over MgS0 4 , filtered, combined and concentrated to dryness. Chromatography on silica gel (5% MeOH-DCM increasing to 10% MeOH) provided material which was re -purified by preparative TLC (5% 2 M-NH 3 -MeOH-EtO Ac) to provide the title compound as light tan solid. ! H NMR (500 MHz,

CDCI 3 ) δ 8.45 - 8.36 (m, 1H), 7.94 (d, J= 8.8 Hz, IH), 7.66 - 7.46 (m, 4H), 7.46 - 7.30 (m, 2H), 7.01 (s, 1H), 5.94 (s, IH), 3.91 (s, 3H), 3.34 (s, 3H), 2.17 (s, 3H). MS (ESI): mass calcd. for C24H20CI2N6O, 478.1 , m/z found 479.1 [M+H] + .

Example 40s: (2-Ch1oro-4~methoxy-3-phenylqumolm-6-y^^

yl)(l~metliyl~lH~l,2,3~iriazo1~5~yl)met!iaEol®TFA

To a flask containing (2,4-dichloro-3-phenyIquinolin-6-yl)(l ,2-dimethyl-lH-imidazol-5-yl)(l- m.ethyl-lH-1 ,2,3-triazol-5-yl)methanol ( 1.1 g, 2,29 mmol, Example 39) was added MeOH (30 mL) followed by solid NaOMe (505 mg, 9.35 mmol, 95% purity) at room temperature. The mixture was heated to 80 °C for 5 hours, then cooled back to room temperature and the MeOH was removed under reduced pressure. The crude material was passed through a plug of silica gel (5% MeOH-DCM) to afford a light brown solid. This material was further purified by P-HPLC (water / acetonitrile / 0.05% TFA) to afford the title compound. f H NMR (500 MHz, CD3OD) δ 8.34 (d, J= 1.8 Hz, IH), 8.01 (d, J = 8.8 Hz, IH), 7.67 (dd, J= 8.8, 2.1 Hz, IH), 7.58 - 7.36 (m,

1 1 6H), 6.90 (s, H i ). 4.01 (s. 3H), 3.69 (s, 31 1 ). 3.55 (s, 3H), 2.66 (s, 31 ! ): MS (EST): mass calcd. for ( H.. ; ( 1 \,.i 474.2, m/z found 475.1 [M+H] ÷ .

The racemic mixture was separated by ehiral chromatography using [Chiralpak AD, 1000A, 20 μΜ (Diaeel), heptane:2-propanoi (80:20 with 0.2% TEA), to give two enantiomers. The first eluting enantiomer was Example 40b and the second eluting enantiomer was Example 40c.

Example 41a: (4-Chloro-2-methoxy~3~phenylquinotin-6-y

-methy1-lH-l,2,3-tria¾o]-5-yI)methano1 » TFA

To a flask containing (2,4-dichloro-3-phenylquinoiin-6-yl)(l ,2-dimethyl-lH-imidazol-5-yl)(l - methyl-l H-l ,2,3-triazol-5-yl)methanol (1 .1 g, 2.29 mmol, Example 39) was added MeOH (30 mL) followed by solid NaOMe (505 nig, 9.35 mmol, 95% purity) at room temperature. The mixture was heated to 80 C for 5 hours, then cooled back to room temperature and the MeOH was removed under reduced pressure. The crude material was passed through a plug of silica gel (5% MeGH-DCM) to afford a light brown solid. This material was further purified by RP-HPLC (water / acetoiiitrile / 0.05% TEA) to afford the title compound. h H NMR (500 MHz, CD 3 OD) δ 8.32 (d, J = 2.1 Hz, I II), 7.98 (d, J = 8.8 Hz, 1 H), 7.60 (dd, J = 8.8, 2.2 Hz, 1H), 7.50 - 7.37 (m, 41 ! }. 7.34 - 7.25 (m, 2H), 6.87 (s, 1H), 4.00 (s, 3H), 3.99 (s, 3H), 3.70 (s, 3H), 2.67 (s, 3H); MS (ESI): mass calcd. for ( ■;.:! I . > : C!NV.( ) , . 474.2, m/z found 475.1 [M+H] + .

The racemie mixture was separated by ehiral chromatography using [Chiralpak AD, 1000A, 20 μΜ (Diacel), heptane:2-propanol (75:25 with 0.2% TEA), to give two enantiomers. The first eluting enantiomer was Example 41b and the second eluting enantiomer was Example 41c.

Example 42: (2,4-Bis(methylthio)-3-phenylquinolin-6-yl)(2,4-dimethylthia zol-5-yl)(l- methyI-lH-imidazol-5-yI)methanol » TFA

To a flask containing (2,4-dichloro-3-phenylqxiinolin-6-yl)(2,4-dimethylthiazol-5- yl)( 1 -methyl- lH-imidazol-5-yl)methanol (198 rag, 0.4 mmol. Example 68) was added DMF (67.5 niL) followed by sodium methanethiolate (37 mg, 0.53 mmol ) at room temperature. After 15 minutes, the reaction mixture was concentrated and chromatographed directly on silica gel (2% MeOH- DCM increasing to 5% MeOH) which afforded a mixture of two products. This material was re- purified by RP-HPLC (water / acetonitrile / 0.05% TFA) to provide the title compound. l H NMR (500 MHz, CDC1 3 ) δ 8.72 (s, 1 H), 8.60 (s, IH), 8.25 i d. J = 8.3 Hz, 1 H), 7.77 (d, ,/ 8.3 Hz, IH), 7.59 - 7.45 (m, 3H), 7.39 - 7.28 (m, 2H), 7.20 (s, IH), 3.71 (s, 3H), 2.80 (s, 3H), 2.68 (s, 3H), 2.32 (s, 3H), 1 .95 (s, 3H); MS (ESI): mass calcd. for C27H26 4OS3, 518.1 , m/z found 519.1 [M+H] + .

Example 43: (4-ChIoro-2-(methylthio)-3-phenyiqiiinolin-6-yI)(2,4-dimethy ithiazol-5-yI)(.l- methyI-lH-imidazol-5-yI)methanol » TFA

To a flask containing (2,4-dichloro-3-phenylquinolin-6-y3)(2,4-dimethylthiazol-5-y l)(l-methyl- lH~imidazol-5-yl)methanol (1 98 mg, 0.4 mmol, Example 68) was added DMF (67.5 niL) followed by sodium methanethiolate (37 mg, 0.53 mmol) at room temperature. After 55 minutes, the reaction mixture was concentrated to dryness and chromatographed directly on silica gel (2% MeOH-DCM increasing to 5% MeOH) which afforded a mixture of two products. This material was re -purified by RP-HPLC (water / acetonitrile / 0.05% TFA) to provide the title compound.

Ή NMR (500 MHz, CDCI 3 ) 6 8.70 (s, IH), 8.63 (s, I H), 8.12 (d, ,/ 8.9 Hz, IH), 7.78 (d, ./ 8.7 Hz, I H), 7.56 - 7.43 (m, 3H), 7.34 (d, J ------ 7.1 Hz, 2H), 7.20 (s, I H), 3.71 (s, 3H), 2.78 (s, 3H), 2.34 (s, 3H), 1.94 (s, 3H); MS (ESI): mass calcd. for C ...H . s CI \ ,OS . .. 506.1 , m/z found 507.0 ! · Π | ' .

Example 44: [2-(2-Aziridin-l-ylethoxy)-4-chloro-3-pheiiyIquiiioIin-6-yl] (4-chloropheiiyl)(l- methyI-iH-imidazol-5-yl)metIiaHol

(4-Chlorophenyl)(2,4-dichlofo-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)mefhanol (100 mg, 0.202 mmol, Example 65), 2-(aziridin-l-yl)ethanol (16.2 μΐ,, 0.20 mmol), toluene (2 mL), and sodium hydride (60% dispersion in mineral oil, 20.2 mg, 0.51 mmol) were combined in a round bottom flask under an Na atmosphere. The reaction solution was heated to reflu and refluxed overnight, cooled and stirred at room temperature an additional night before workup. Reaction contents were transferred to a separatory funnel with EtOAc dilution, and extracted with saturated, aqueous NH 4 C1 then saturated, aqueous NaHC0 3 solutions. The organic phase was separated then dried over MgSO-j, filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / MeOH) to afford the title compound. MS (ESI): mass calcd. for C 30 H 26 Cl 2 N4O 2 , 544.1 ; m z found, 545.5 [M+H] + ; 1 H MR (600 MHz, CD 3 OD) δ ppm 8.05 (d, J = 2.0 Hz, 1 H), 7.74 (d, J = 8.7 Hz, 1 H), 7.61 - 7.57 (m, 2H), 7.40 - 7.35 (m, 2H), 7.35 - 7.31 (m, lH), 7.28 - 7.23 (m, 6H), 6.18 (d, J = 5.1 Hz, IH), 4.51 - 4.44 (m, 2H), 3.37 (s, 3H), 2.50 - 2,39 (m, 2H), 1 .48 - 1.37 (m, 2H), 0.97 - 0.94 (m, 2H).

Example 45: (4-C lorophenyI)[4-ch1oro-3-phenyl-2-(2,2,2-trifluoroet oxy)quinoHii-6-yi](l- methyI-lH-imidazol-5-y1)methanol*TFA

(4-Chlorophenyl)(2,4-dichloro-3-pheny[quinolin^

(100 mg, 0.202 rnniol, Example 65), 2,2,2 -trifluoroethanol (14,5 \L, 0.202 mmol), toluene (2 ml), and sodium hydride (60% dispersion in mineral oil, 20 mg, 0.51 mmol) were combined in a round bottom flask under an N 2 atmosphere. The reaction solution was heated to reflux and reflux ed overnight. The reaction solution was cooled to room temperature then transferred to a separatory tunnel with EtOAc dilution, and extracted with saturated, aqueous NH 4 CI then saturated, aqueous NaHCOi solutions. The organic phase was separated then dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / MeOH) then via reverse phase chromatography using acetonitrile with 0.05%) trifluoroacetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHC0 3 solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over Mg80 4 , filtered and concentrated under reduced pressure to provide the title compound. MS (ESS): mass caicd. for C28H20CI2F3N3O2, 557.1 ; m/z found, 558.3 [M+Hf; Ή NMR (600 MHz, CDCI3) δ ppm 8.38 (s, 1 H), 8.17 (d, J = 2.0 Hz, 1H), 7.87 (d, J ------ 8.8 Hz, 1H), 7.60 (dd, J ------ 8.8, 2.1 Hz,

1 1 1). 7.52 - 7.43 (m, 3H), 7.38 - 7.31 (m, 6H), 6.56 (s, 1 H), 4.92 (q, J ----- 8.4 Hz, 21 1). 3.62 (s.

(4-Cblorophenyl)(2,4-dichloro-3-phenylquinoti^

(100 mg, 0.202 mmol, Example 65), 2-(methylamino)etbanol (16.2 p.L, 0.202 mmol), toluene (2 ml), and sodium hydride (60% dispersion in mineral oil, 28.3 mg, 0.707 mmol) were combined in a round bottom, flask and heated at reflux for 48 hours under an N 2 atmosphere. The reaction solution was then cooled to room temperature, diluted with EtOAc, transferred to a separatory funnel, extracted with saturated, aqueous H 4 CI then saturated, aqueous NaHCi¾ solutions. The organic phase was separated then dried over MgS0 , filtered and concentrated under reduced pressure. The crude product was purified by flash colum chromatography (silica gel, 0-10% DCM / MeOH) then further purified via reverse phase chromatography using acetonitrile with 0.05% trifluoroacetic acid in water as eluent, then lyophilized to afford the title compound as a trifluoroacetate salt. MS (ES I): mass ca!cd. for C 2 9H 26 C1 2 4 0 2 , 532.1 ; m/z found, 533.2 [M+H] + ; Ί ΐ NM (600 MHz, CD 3 OD) δ ppm 8.98 (s, I I I ). 8.22 (s, I I I ). 7.83 i d. J = 8.8 Hz, I I I ). 7.79 (d, ./ 8.9 Hz, ! f ! ). 7.55 (t, J 7.4 Hz, 2H), 7.50 (t, ./ 7.4 Hz, 1H), 7.47 - 7.39 (m, 6H), 6.93 (d . J = 1.4 Hz, 1H), 3.80 - 3.73 (m, 2H), 3.69 (s, 3H), 3.61 - 3.57 (m, 2H), 2.75 (s, 3H).

Example 47a: 6-((2,4-Dimethylthiazol-5-yI)(hydroxy)(l-meth -lH-l,2,3-triazol-5- yI)metIiyl)-3-piiein qni5io iie-2,4-dicarboiiitriIe

(2,4-Dichloro-3-phenylquinolin-6-yl)(2,4-dimethylthiazol-5-y l)(l -methy

yl)methanol (100 mg, 0.2 mmol, Example 67), zinc cyanide (75 mg, 0.64 mmol), X-Phos (25 mg, 0.052 mmol), and Pd 2 (dba) 3 (60 mg, 0.066 mmol) were all added to a large microwave vial. DMA (3 niL) was added and the vial was sealed and evacuated and the mixture was heated to 120 °C in an oil bath. After 1.5 hours, the reaction mixture was filtered while still warm through Celite* and rinsed with EtOAc. The effluent was concentrated and the DMA was partially removed under high vacuum. The crude material was chromatographed on silica gel (3% MeOH- DCM increasing to 8% MeOH) which provided the title compound as a pale yellow foam. Ή MR (500 MHz, CDCJ ) δ 8.53 (s, 1H), 8.26 (d, ./ 8.9 Hz, lH), 7.79 (d, ./ 9.0 Hz, H I). 7.71 - 7.54 (m, 5H), 7.10 (s, 1H), 6.10 (s, H I). 4.00 (s, 31 1 ). 2.57 (s, 3H), 2.14 (s, 3H); MS (ESI): mass calcd. for C 26 H 19 N 7 OS, 477.1, m/z found 478.1 [M+H] + .

The racemic mixture was separated by chiral chromatography using [Chiralcel OJ, 1000A, 20 μΜ (Diacel), and 100% MeOH], to give two enantiomers. The first e!uting enantiomer was Example 47b and the second eluting enantiomer was Example 47c.

Example 48: [4-Chloro-2-(2-methoxyethoxy)-3-phenyIquiiioIin-6-yl](4-chlo ropheiiyl)(l- methyl-lH-imidazol-S-y methanol

(4-Chlorophenyl)(2,4-dichloro-3-phenyIquinoiin-6-yI)(l -methyl- lH-imidazol-5-yi)methanol (100 mg, 0.202 mmol, Example 65), 2-methoxyethanol (16.0 μΐ., 0.202 mmol), toluene (2 mL), and sodium hydride (60% dispersion in mineral oil, 20.2 mg, 0.505 mmol) were combined in a round bottom flask under an N 2 atmosphere. The contents were heated to reflux and refluxed overnight. The reaction solution turned from a heterogeneous white mixture to slightly yellowish with a moderate amount of precipitate. The contents were cooled to room temperature then transferred to a separately funnel with EtOAc dilution, and extracted with saturated, aqueous NH 4 Ci and saturated, aqueous NaHC0 3 solutions. The organic phase was separated then dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / (10% of a 2 M M MeOH in DCM)) then further purified via reverse phase chromatography using acetonitriie with ammonium hydroxide in water as eiuent to afford the title compound, MS (ESI): mass calcd. for ( \ ; H . .,C!A :() ; . 533.1 ; m/z found, 534.2 i M H j : J H NMR (600 MHz, C! ) .·()!. ) ) δ ppm 8.96 (d, J = 0.9 Hz, 1H), 8.19 (d, J = 1.9 Hz, 1 H), 7.91 (d, ./ 8.8 Hz, IH), 7.70 (dd, ./ 8.8, 2,2 Hz, 5 H), 7.48 - 7.44 (m, 4H), 7.43 - 7.39 (m, 3H), 7.36 - 7.33 (m, 2H), 6.90 (d, J = 1 .6 Hz, I H), 4.59 - 4.54 (m, 2H), 3.70 (s, 3H), 3.68 - 3.63 (m, 2H), 3.25 (s, 3H).

Example 49a: (4-Ch1oro-2-met ox.y-3-p eny]quinolin-6-yl)(4-chIorophenyl)(l-mei imidazo1-5-yi)met anoI » TFA

(4-Chlorophenyl)(2,4-dichlofo-3-phenylquinolin-6-yl)(l -memyl-lH-imidazol-5-yl)methan (100 rag, 0.202 mrao!, Example 65), toluene (2 ml.,), and sodium meth oxide (109 mg, 2,02 mmol) were combined in a round bottom flask equipped with a stirbar and condenser under an N 2 atmosphere. The reaction solution was heated to reflux and refluxed overnight. Analysis showed an incomplete reaction, so additional sodium methoxide (109 mg, 2,02 mmol) was added and contents were refluxed for an additional day. The reaction was cooled to room temperature and contents were transferred to a separator}' funnel with EtOAc dilution, and extracted with saturated, aqueous NH 4 C1 then saturated, aqueous NaHC(¾ solutions. The organic phase was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude material was purified via reverse phase chromatography using acetonitrile with 0.05% trifluoroacetic acid in water as eluent. The fractions from the purification containing the desired product were lyophilized to provide the title compound as a trifluoroaeetate salt. MS (EST): mass calcd. for ( ■· 1 1 > ,C! : N ; ( , 489.1 ; m/z found, 490.1 I M H j : 3 H NMR (600 MHz, CD 3 OD) δ ppm

8.97 (d, ,/ 0.9 Hz, I H), 8.18 (d, J = 1.9 Hz, I H), 7.94 (d, J = 8.9 Hz, I H), 7.71 (dd, J = 8.8, 2,2 Hz, IH), 7.48 - 7.44 (m, 4H), 7.43 - 7.40 (m, 31 1 ;·. 7.32 - 7.28 (m, 2H), 6.90 (d, ./ 1.6 Hz, I H),

3.98 (s, 3H), 3.70 (s, 3H).

(4-Chloro-2-methoxy-3-phenylquinolin-6-yl)(4-chlorophenyl)(l -methyl- 1 H-imidazol-5- yl)methanol*TFA was purified on a chiralcei AD column (8 cm) with ethanol to provide two enantiomers. The first eluting e antiomer was Example 49b: MS (ESI): mass calcd. for ( >Η > |(Ί :\ ' ,0 . .. 489, 1 ; ni/z found, 490.2 i VI H ] : 3 H NMR (600 MHz, CD 3 OD) δ ppm 8.14 (d, J = 2.0 Hz, IH), 7.94 (s, 1 H), 7.87 (d, ./ 8.8 Hz, IH), 7.69 (dd, ,/ 8.8, 2.1 Hz, IH), 7.45 (dd, ./ 1 1.4, 4.4 Hz, 2H), 7.42 - 7.34 (m, 5H), 7.33 - 7.27 (m, 2H), 6.40 (s, IH), 3.97 (s, 3H), 3.51 (s, 3H) and the second eluting enantiomer was Example 49c: MS (ESI): mass calcd. for C27H21CI2N3O2, 489.1 ; m/z found, 490.1 | M I I j : Ή NMR (600 MHz, CD 3 OD) δ ppm 8.20 - 8.14 (m, 2H), 7.88 (d, ./ 8.8 Hz, I H), 7.69 (dd, ./ 8.8, 2.1 Hz, I H), 7.47 - 7.42 (m, 2H), 7.42 - 7.35 (m, 5H), 7.31 - 7.27 (m, 2H), 6.51 (s, IH), 3.96 (s, 3H), 3.55 (s, 3H).

Example 50: {4-Chloro-2~[(2~methoxyethyI)(methy])am

ch1orophenyl)(l-methyl-lH-imidazol-5-yl)methaiioI

(4-Chloro " phenyl)(2,4-dichloro-3-pheny[quinolin-6-y[)( l -methyl- lH-imidazol-5-yl)methanol (100 nig, 0.202 mmol, Example 65), /V-(2-methoxymethyl)methylamine (900 μΕ, 10.1 mmol), and methanol (2 mL) were combined in a reaction tube, which was then sealed and heated to 100 °C for 72 hours. The contents were then cooled to room temperature, transferred to a round bottom flask and the solvent was removed via reduced pressure distillation. The crude residue was then taken up into EtOAc, transfeiTed to a separatory funnel, extracted twice with a saturated, aqueous NH 4 CI solution. The organic phase was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / (10% of a 2 M NH 3 MeOH in DCM)) then further purified via reverse phase chromatography using aeetomtriie with 0.05% trifiuoroacetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHCC solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure to afford the title compound. MS (ESf): mass calcd. for C30H28CI2N4O2, 546.2; m/z found, 547.3 [M+H] + ; Ή NMR (600 MHz, CD 3 OD) δ ppm 8.05 (d, J = 2.0 Hz, 1H), 7.97 (s, 1H), 7.75 (d, J = 8.8 Hz, 1H), 7.60 (dd, J 8.8, 2.1 Hz, 1 H), 7.45 (dd, J = 10.3, 4.6 Hz, 2H), 7.38 (ddd, ,/ 8.6, 4.5, 1.2 Hz, 1 1 1 !. 7.36 - 7.31 (m, 6H), 6.42 (s, i l l !. 3.51 (s, 31 1 ;·. 3.31 - 3.28 (m, 21 1 }. 3.24 (dd, J ------ 8.7, 3.0

Hz, 2H), 3.17 (s, 3H), 2.74 (s, 3H).

Example 51: A^~[2-({4-Chloro-6-[(4-ch1orophenyl)(hydroxy)(l-methyI-lH ' midazol-5- yl)methyl]-3-phenyIqumoIm-2-yI}oxy)ethy]]propaiiamide

(4-Cblorophenyl)(2,4-dich]Oro-3-phenylqum^

(100 mg, 0.202 mmol, Example 65), A'-(2-hydroxylethyl)propionamide (34.2 μΐ,, 0.303 mmol), toluene (2 mL), and sodium hydride (60% dispersion in mineral oil, 32.3 mg, 0.808 mmol) were combined in a round bottom flask under an 2 atmosphere. The reaction solution was heated to reflux and refluxed overnight. The reaction solution was cooled to room temperature then transferred to a separatory funnel with EtOAc dilution, and extracted with saturated, aqueous NH4CI then saturated, aqueous NaHC0 3 solutions. The organic phase was separated, then dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / MeOH) then via reverse phase chromatography using acetonitrile with 0.05% trifluoroacetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHC0 3 solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure to provide the title compound. MS (ESI): mass calcd. for C31H28CI2 4O3, 574.2; m/z found, 575.2 [M+H ; 1 H NMR (600 MHz, CD 3 OD) δ ppm 8.14 (s, 1 H), 7.83 (d, J = 8.8 Hz, 1H), 7.78 (s, 1H), 7.68 (dd, J = 8.8, 1.7 Hz, 1H), 7.47 - 7.41 (m, 2H), 7.41 - 7.37 (m, 1 1 1 ). 7.38 - 7.34 (m, A l l ). 7.34 - 7.30 (m, 2H), 6.32 (s, 1 1 1 ). 4.52 (t, ,/ 5.6 Hz, 2FI), 3.53 - 3.44 (m, 5H), 2.1 1 (q, J = 7.6 Hz, 2H), 1.04 (t, J = 7.6 Hz, 3H). Example 52: [2-(2-Aminoethoxy)-4-chIoro-3-phenylquinolin-6-ylj(4-chiorop henyI)(l' meth I-iH-imidazol-5-yl)metIiaHol

(4-ChlofO " phenyl)(2,4-dichloro-3-phenylquinolin-6-yl)( l -methyl- lH-imidazol-5-yl)methanol (100 mg, 0.202 mmoi, Example 65), N-(2-hydroxyethyl)-2,2,2-trifluoroacetamide (49 mg, 0.30 mmol), toluene (2 mL), and sodium hydride (60% dispersion in mineral oil, 32.3 mg, 0.808 mmol) were combined in a round bottom flask and heated at reflux for 48 hours under an N 2 atmosphere. Analysis after overnight reaction showed only partial conversion, so additional N- (2-hydroxyethyl)-2,2,2-trifluoroacetamide (33 mg, 0.21 mmol) and sodium hydride (60% dispersion in mineral oil, 25 mg, 1.03 mmol) were added and the vessel was reseaied and heated at reflux for an additional 48 hours. The reaction solution was then cooled to room temperature, diluted with EtOAc, transferred to a separatory funnel, and extracted with saturated, aqueous NH 4 Ci then saturated, aqueous N:il !( ' ( ) .· solutions. The organic phase was separated then dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified via reverse phase chromatography using acetonitrile with 0.05% trifluoroacetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHC0 3 solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure to afford the title compound. MS (ESI): mass calcd. for C 28 H 24 CI 2 N 4 O 2 , 518.1 ; m/z found, 519.2 [M+H] + ; i NM (600 MHz, CD 3 OD) 5 ppm 8.17 (d. J ------ 2.0 Hz, 1 1 1 ). 7.87 (d, ./ 8.8 Hz, i l l ). 7.72 (dd, J ------

8.8, 2.1 Hz, 1H), 7.69 (s, 1H), 7.50 - 7.45 (m, 2H), 7.44 - 7.40 (m, 1H), 7.39 - 7.33 (m, 6H), 6.26 (s, 1H), 4.67 - 4.60 (m, 2H), 3.47 (s, 3H), 3.20 (t, J= 5.3 Hz, 2H). Example 53: {4-Chloro-2-[(2-methoxyethyl)(methyl)amino]-3-phenylquiiioli ii-6- }(l- methyl-lH midazol-5-yl)[6-(trifluoromethyl)pyridin-3-yIImethaiiol

(2,4-Dichloro-3-pheny3quinolin-6-y3 (l -m£thy3-lH-imidazol-5-yl)[6-(trifiuoromethyl) pyridm-3- yljmethanol (200 rag, 0.378 mmol, Example 66), N-(2-methoxymethyl)methylamine (842 μΤ, 9.45 mmol), and methanol (2 mL) were combined in a. reaction tube, which was then sealed and heated to 100 °C for 48 hours. The contents were then cooled to room temperature, transferred to a round bottom flask and the solvent was removed via reduced pressure distillation . The crude residue was then taken up into EtOAc, transferred to a separatory funnel and extracted twice with a saturated, aqueous H 4 C1 solution. The organic phase was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified via reverse phase chromatography using acetonitrile with 0.05% trifluoro acetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHC0 3 solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure to afford the title compound. MS (ESI): mass calcd. for C30H27CIF3N5O2, 581.2; m/z found, 582.2 [M+H] ÷ ; ! H NMR (600 MHz, CD3OD) δ ppm 8.77 ( d. ./ 1.8 Hz, 1H), 8.08 (d, J = 2.0 Hz, 1H), 8.01 (dd. ./ 8.2, 1.9 Hz, I I I ). 7.92 (s, 1H), 7.84 (d, J = 8.2 Hz, 1H), 7.80 (d, J = 8.8 Hz, l H), 7.61 (dd, J= 8.8, 2.1 Hz, 1 H), 7.49 (t, J = 7.5 Hz, 2H), 7.42 (ddd, J = 6.7, 2.5, 1.2 Hz, 1H), 7.39 - 7.35 (m, 2H), 6.44 (s, 1 H), 3.52 (s, 3H), 3.35 - 3.32 (m, 2H), 3.30 - 3.27 (ni, 2H), 3.19 (s, 3H), 2.78 (s, 3H).

Example 54: [ -Chloro-3-phenyI-2-(2,2,2-trifluoroethoxy)quinoIiii-6-yl](l- methyl-lH- imidazoI-S-yl)[6-(trifli5orometliyl)pyridiii-3-yl| methanol

(2,4-Dichloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)f6-(trifluoromethyl) pyridin-3- yljmethanol (200 nig, 0.378 mmol. Example 66), 2,2,2 -trifluoroethanoi (14.0 μL, 0.19 mmol), toluene (2 mL), and sodium hydride (60% dispersion in mineral oil, 59 mg, 0.47 mmol) were combined in a round bottom flask under an N 2 atmosphere. The reaction solution was heated to reflux and ref!uxed overnight. Analysis showed the reaction had only progressed a moderate amount, so additional reagents were added, 2,2,2-trifluoroethanol (14.0 μΐ,, 0.19 mmol) and sodium hydride (60% dispersion in mineral oil, 6 mg, 0.19 mmol) and contents were heated at reflux for an additional 48 hours. The contents were cooled to room temperature then transferred to a separatory funnel with EtOAc dilution, and extracted with saturated, aqueous H 4 C1 then saturated, aqueous NaHCOs solutions. The organic phase was separated then dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude material was purified via reverse phase chromatography using acetonitrile with 0.05% trifiuoroacetic acid in water as eluent. The fractions from, the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHC0 3 solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS()4, filtered and concentrated under reduced pressure to afford the title compound. MS (ES ): mass calcd. for C 2 sHi 9 ClF 6 N 4 02, 592.1; m/z found, 593.2 j .Yi - i i i : j H NMR (600 MHz, CD 3 OD) δ ppm 8.80 (d, J = 2.1 Hz, 1H), 8.28 (d, J = 1.9 Hz, 1H), 8.17 (s, 1 H), 8.05 (dd, J = 8.2, 2.0 Hz, 1H), 7.97 (d, J= 8.7 Hz, 1H), 7.86 (dd, J = 8.3, 0.4 Hz, 1H), 7.75 (dd, J = 8.8, 2.2 Hz, I.H), 7.51 - 7.42 (m, 3H), 7.37 - 7.34 (m, 2H), 6.60 (s, 1H), 5.00 (q, J= 8.6 Hz, 2H), 3.56 (s, 3H).

Example 55 : (2,4-Dichloro-3-phen quinolin-6-yI)(l -methyI-lH-imidazol-5-yI)pyrimidin-2- ylmethanol

6-Bromo-2,4-dicliloro-3-plienyIquinoline (0,895 g, 2.54 mmol, Intermediate 7: step c) and (1- methyl- 1 H-imidazol-5-yl)(pyrimidin-2-yl)methanone (500 mg, 2.66 mmol, Intermediate 15: step b) were dissolved in THF (250 niL) in a dry round bottom, flask under an N2 atmosphere, then cooled to -78 °C in dry ice acetone bath. n-BuLi (2.5 M in hexanes, 0.966 ml,, 2.42 mmol) was then added dropwise via syringe over approximately 2 minutes. The reaction contents were stirred at -78 °C for approximately! ,5 hours, then the dry ice bath was removed and allowed to warm to room temperature and stir for approximately 1 hour. The reaction was then re-cooled to 0 °C and quenched with a saturated, aqueous NH 4 CI solution, then transferred to a separately funnel with EtOAc. The organic phase was separated, then the aqueous layer was back extracted with EtOAc, then the combined organic phases were dried over MgS0 , filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography (silica gel, 0-10% DCM / (10% of a 2M N I¾ MeOH in PCM }} then further purified via reverse phase chromatography using acetonitrile with 0.05% trifluoroacetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separately funnel with EtOAc and extracted with a saturated, aqueous NaHCC solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure to afford the title compound. MS (ESI): mass calcd. for C 24 H 17 C1 2 N 5 0, 461.1 ; m/z found, 462.1 j \ 1 H i : Ή NMR (600 MHz, CD 3 OD) δ ppm 8.88 (d, J = 4.9 Hz, 2H), 8.57 (d, J = 1.8 Hz, 1H), 8.08 (dd, J = 8.9, 2.0 Hz, lH), 7.99 (d, J = 8.9 Hz, 1 H), 7.80 (s, 1 1 1 }. 7.54 - 7.45 (m, 4H), 7.36 - 7.32 (m, 2H), 6.49 (d, ,/ 1.0 Hz, ! H), 3.43 (s, 3H).

Example 56: {4~Chloro-2-[methoxy(methyl)amino]-3-phenylquinoliii-6-yl}(l ~methyl~lH- imidazol-5-yI) 6-(iriflMoromei!iyl)pyridiii~3~yl]metiiaiioI

(2,4-Dichloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)[6-(trifluoromethyl)pyridin-3- yl] methanol (200 mg, 0.378 mmol, Example 66), Λ ' ,Ο-dimethylhydroxylamine hydrochloride (376 mg, 3.78 nimol), and dimethylformamicle (2 mL) were combined in a reaction tube which was then sealed and heated to 100 °C for 48 hours. Analysis shows desired product, but also the presence of starting material. Additional ΛζΟ-dimethylhydroxylamine hydrochloride (190 mg, 0.195 mmol) was added and the contents were heated for an additional 24 hours. The contents were then cooled, transferred to a separately funnel, diluted with EtOAc and extracted with a saturated, aqueous NH 4 C1 solution, followed by deionized water (4 x). The organic phase was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude material was purified via reverse phase chromatography using acetonitrile with ammonium hydroxide in water as eiuent to afford the title compound, MS (ESI): mass caicd. for C28H23CIF3N5O2, 553.1 ; m/z found, 554.2 [M+Hf; l H NM (400 MHz, CD 3 OD) δ ppm 8.79 (d, ,/ 2.1 Hz, IH), 8.19 id. ./ 1.9 Hz, 1 H), 8.02 (dd. 8.2, 1.9 Hz, 1 H), 7.96 id. ./ 8.8 Hz, IH), 7.83 (d, J ------ 8.2 Hz, IH), 7.75 - 7.68 (m, 2H), 7.48 (t, J ------ 7.4 Hz, 2H), 7.40 (ddd, ./ 7.4, 3.9, 1.3

Hz, IH), 7.32 - 7.24 (m, 2H), 6.35 (s, IH), 3.48 (s, 3H), 3.07 (s, 3H), 2.77 (s, 3H).

Example 57: {2,4-Bis[methoxy(methyJ)aminoj-3-phenyiqiiinolin-6-yI}(l-met hyl-l/i r - iniidazol-5-yi)[6-(trifluoromethyl)pyridin-3-yl] methanol

Purification of the crude product from the synthesis of {4-chloro-2-[methoxy(methyl)amino]-3- phenylquinolin-6-yl}(l -methyl- lH-imidazol-5-yl)[6-(trifluoromethyl)pyridin-3-yl]methanol (Example 56) also provided the title compound. MS (ESI): mass calcd. for C30H29F3N0Q3, 578.2; mlz found, 579.2 [M+H] + ; 3 H NMR (400 MHz, CD 3 OD) δ ppm 8.75 (d, J= 2.0 Hz, HI), 8.36 (d, J = 2.0 Hz, 1H), 8.02 (dd, J = 8.3, 1.8 Hz, 1H), 7.97 (d, J~ 8.9 Hz, 1H), 7.84 (d, J = 8.2 Hz, 1H), 7.78 - 7.70 (m, 2H), 7.44 - 7.37 (m, 2H), 7.36 - 7.29 (m, 1H), 7.24 (d, J = 7.3 Hz, 2H), 6.33 (s, 1H), 3.50 (s, 3H), 3.26 (s, 3H), 3.02 (s, 3H), 2.86 (s, 3H), 2.78 (s, 3H).

Example 58: {4-Chloro-2-[methoxy(meth i)amino|-3-phen iquinolin-6-yl}(4- chlorophenyl)(l-methyi-lir/-iniidazol-5-yi)methanoI

(4-C3iloropiienyl)(2,4-dichloiO-3-phenyiquinolin-6-yi)(l -methyl- 1H- imidazol-5-yl)methanol (250 mg, 0,505 mmol. Example 65), N,0-dimethylhydroxylamine hydrochloride (1.01 g, 10.1 mmol), and dimethyiformamide (2 mL) were combined in a reaction tube, which was then sealed and heated to 100 °C for 48 hours. The contents were then cooled, transferred to a. separately funnel, diluted with EtOAc and extracted with saturated, aqueous NH 4 CI solution, followed by deionized water (4 x). The organic phase was separated and dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude material was purified via reverse phase chromatography using acetonitrile with ammonium hydroxide in water as eluent to afford the title compound. MS (ESI): mass calcd. for C 28 H 2 CI 2 N O 2 , 518.1 ; m/z found, 519.2 [M+H] + ; Ή NMR (400 MHz, CD3OD) δ ppm 8.14 (d. ./ 1.9 Hz, 1 1 1). 7.91 id, J= 8.8 Hz, I H). 7.70 (dd, ./ = 8.8, 2.1 Hz, 1H), 7.67 (s, 1H), 7.47 (dd, J = 11.4, 4.3 Hz, 2H), 7.39 (dt, J= 4.4, 1.8 Hz, IH), 7.36 (d, J = 4.1 Hz, 4H), 7.26 (dd, J = 8.2, 1.3 Hz, 21 1). 6.28 (s, IH), 3.46 (s, 3H), 3.05 (s, 3H), 2.76

(s, 3 ,:H)

Example 59: {2,4-Bis{methoxy(methyI)amino]-3-phenylqiiiiioliii-6-yl}(4-c hlorophenyI)(l- methyl-lH-imidazol-S-y^methanol

Purification of the crude product from the synthesis of {4-chlQro-2-[metboxy(methyl)ammo]-3- phenylquinolin-6-yl}i4-chlorophenyl)(i -methyl- l H-imidazol-5-yl)methanol (Example 58) also provided the title compound. MS (ESI): mass calcd. for C 30 H 30 CIN 5 O 3 , 543.2; m/z found, 544.3 [M+H] + ; ! H NMR (400 MHz, CD 3 OD) 5 pm 8.36 (d, ./ 1 .8 Hz, i l l . 7.96 - 7.89 (m, ! ! ! ). 7.71 (dd . ./ 8.9, 2.2 Hz, 1H), 7.66 (s, 1H), 7.42 - 7.28 (m, 7H), 7.24 (d, J = 6.6 Hz, 2H), 6.28 (d, J = 1.0 Hz, 1H), 3.48 (s, 3H), 3.25 (s, 3H), 3.01 (s, 3H), 2.85 (s, 3H), 2.79 (s, 3H).

Example 60a: (4-Chloro-2-methoxy-3-phenylq«iBolin-6-yl)(l-methyl-lH r -imidazol-5-

(2,4-Dichloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)pyrimidin-2-ylmethanol (156 mg, 0.337 mmol, Example 55), toluene (2 niL), and sodium methoxide (365 nig, 6.75 mmol) were combined in a round bottom, flask equipped with a stirbar and condenser under an N2 atmosphere. The contents were heated to reflux and refiuxed overnight. The reaction was cooled and the contents were transferred to a separatory funnel with EtOAc dilution, and extracted with saturated, aqueous NH 4 CI then saturated, aqueous NaHC(¾ solutions. The organic phase was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude material was purified via reverse phase chromatography using acetonitri!e with 0.05% trifiuoroacetic acid in water as eluent. The fractions from the purification containing the desired product were transferred to a separatory funnel with EtOAc and extracted with a saturated, aqueous NaHCQ 3 solution. The aqueous layer was separated, extracted with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure to provide the title compound. MS (ESI): mass calcd. for C25H20CIN5O2, 457.1; m/z found, 458.2 [M+Hf; J H NMR (400 MHz, C ' iX ' h ) δ ppm 8,81 (d, ./ 4.9 Hz, 2H), 8.48 (d, ./ 5.8 Hz, ! ! ! }. 7.92 - 7.81 (m, 2H), 7.54 - 7.39 (m, 41 1 ). 7.36 - 7.28 (m, 3H), 6.55 (s, 1 H), 6.1 5 (s, 1 1 1 ;·. 3.98 (s, 31 1 ;·. 3.40 (s, 3H).

(4-Chloro-2-methoxy-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yi)pyrimidin-2- yimethanol was purified on a chiralcel OD column (20 μτη, Diacei) with methanol to provide two enantiomers. The first ehiting enantiomer was Example 60b: MS (ESI); mass calcd. for C25H20CIN5O2, 457.1 ; m/z found, 458.2 | M I I j : ] H NMR (600 MHz, CD 3 OD) δ ppm 8.86 (d, J = 4.9 Hz, 2H), 8.41 (d, ,/ 2 0 Hz, 1H), 7.90 ( dd. ./ 8.8, 2.0 Hz, 1H), 7.82 (d, J 8.8 Hz, 1H), 7.63 (s, lH), 7.46 - 7.41 (m, 3H), 7.41 - 7.37 (m, 1H), 7.29 (d, J = 7.0 Hz, 2H), 6.41 (s, 1H), 3.95 (s, 3H), 3.39 (s, 3H).

Example 61 : (4-Chloro-3^henyl-2-(MfluoromethyI)quto

yl)(pyridin-2-yI)methanol*TFA

The title compound was prepared using (1 -methyl- lH-imidazol-5-yl)(pyridin-2-yl)metha.none (Intermediate 12: step b) in place of rert-butyl 4-nicotinoylpiperidine- 1 -carboxylate using the procedure described for Example 69. 3 H NMR (400 MHz, CD3OD) δ 8.97 (s, 1H), 8.58 - 8.65 (m, 21 ! ). 8.28 (d, ./ 9.09 Hz, 1H), 8.1 1 (dd, J = 2.02, 9.09 Hz, 1H), 7.88 - 7.97 (m, 1H), 7.80 (d, J = 7.58 Hz, 1H), 7.48 - 7.55 (m, 3H), 7.38 - 7.46 (m, 1H), 7.26 - 7.34 (m, 2H), 7.1 1 (s, IH), 3.63 (s, 3H); MS m/e 495.3 [M+Hf.

Example 62: (4-Chloro-3-phenyI-2-(trifluoromethyl)quinoIin-6- i)(4-chloropheiiyI)(l- methyl-lH-imidazol-S-y^methanol^TFA

A mixture of 6-bromo-4-chloro-3-phenyl-2-(trif3.uorom.ethyl)quinoline (864 mg, 2.23 mmol, Intermediate 19: step b), (4-chiorophenyl)( 1 -methyl- lH-imidazol-5-yl)methanone (490 mg, 2.22 mmol, Intermediate 8: step b) and 22 m.L of THF was purged with N2 and cooled to -78 °C. To the mixture was added «-BuLi (1.6 M in hexanes, 1.8 mL, 2.9 mmol) dropwise and the color changed to orange then almost black. The reaction mixture was stirred at -78 °C to 0 °C for 70 minutes, then allowed to warm up to room temperature overnight. Saturated NH4CI (aqueous) was added and the organic layer was separated. The aqueous layer was extracted with dichloromethane. The combined organic phases were dried (Na 2 S0 4 ), filtered, and concentrated. The crude was purified by flash column chromatography (silica gel, 5-10% MeOH in DCM) followed by reverse phase HPLC (water/acetonitrile/0.1% TFA) to provide the title compound as a white solid. j H NMR (400 MHz, CD 3 OD) δ 9.00 (s, IH), 8.42 (d, ./ 2.02 Hz, 1H), 8.31 (d, ./ = 8.59 Hz, I H), 7.97 (dd, J = 2.02, 9.09 Hz, IH), 7.47 - 7.54 (m, 4H), 7.41 - 7.47 (m, 3H), 7.27 - 7.34 (m, 2H), 6.99 (d, J = 1 .52 Hz, IH), 3.71 (s, 3H); MS m/e 528.2 [M+Hf.

Example 63 : (4~Chloro-2-methoxy-8-methyl-3-(4-(trifluoro

l)(l-methyl~lH midazol-5-yl)(6~(trifluQrom

fi-Butyllithium (2.0 mL, 3.202 mmol) was added to a -78 °C mixture of 6-bromo-4-chloro-2- methoxy-8-methyl-3-(4-(trifiuoromethoxy)phenyl)quinoline (1.1 g, 2.463 mmol, Intermediate 13: step d) and (l-memyl-lH-imidazol-5-yl)(6-(trifluoromethyl)pyridin-3-yl)m ethanone (0.691 g, 2.709 mmol, Intermediate 2: step c) in dry THF (25 mL) over a 2 minute period. After complete addition stirring was continued at -78 °C for 10 minutes then the reaction was warmed up to 0 °C and stirred for 1 hour. Saturated aqueous NH 4 C1 was added and the reaction mixture slowly warmed to room temperature. Water was added and the layers were separated. The aqueous layer was extracted with EtOAc. The combined organic extracts were dried (Na 2 S04), filtered, evaporated in vacuo and chromatographed (DCM/10% MeOH in EtOAc) to provide the product. Further purification using reverse phase HPLC (acetonitrile / water + 0.1% TFA) provided the title compound. MS (ESI) 623.1 ( V! · ! !) ' .

Example 64: (2,4-Dichloro-3-phenylquinoUn-6-yl)(l~methyl~lH midazol~5-yl)(pyridin-2- yl)methaiiol

A solution of «-BuLi (2.5 M in hexanes, 0.34 mL, 0.85 rnmol) was added dropwise by syringe to a solution of 6-bromo-2,4-dichloro-3-phenylquinoline (305.4 mg, 0.865 mmol. Intermediate 7: step c) in dry THF (4.4 mL) at -78 °C. After 1.5 minutes, a solution of ( 1 -methyl- 1 H-imidazol- 5-yl)(pwidin-2-yl)methanone (0.175 g, 0,936 mmol. Intermediate 12: step b) in dry THF (1.8 mL) was added dropwise. The reaction mixture was stirred for 5 minutes at -78 °C, then the reaction flask was placed into an ice-water bath. After 10 minutes, the mixture was warmed to room temperature and the reaction was quenched with methanol and water. The mixture was partitioned between water and DCM. The separated aqueous phase was further extracted with DCM. The organic phase was dried (Na 2 S0 4 ), filtered, and concentrated. The crude product was purified by flash column chromatography (silica gel, 0-10% MeOH-DCM) to provide the title compound as a white solid. f H NMR (500 MHz, CDC1 3 ) δ 8.65 (ddd, J = 4.9, 1.6, 1.0 Hz, 1H), 8.30 (d, J = 1.7 Hz, lH), 8.06 (d, J = 8.8 Hz, 1H), 7.86 (dd, J = 8.8, 2.0 Hz, 1H), 7.73 (td, J = 7.7, 1.7 Hz, 1H), 7.55 - 7.47 (m, 4H), 7.36 - 7.29 (m, 3H), 7.23 (d, J = 8.0 Hz, 1H), 6.37 (d, J = 1.1 Hz, i l l ). 3.44 (s, 3H); MS m/e 461.1 j YM I j ' .

Example 65: (4-Chlorophenyl)(2,4-dichloro-3-phenylquinolin-6-yI)(l-methy I-7/ -imidazol- 5-yI)methanol

To (4-chlorophenyl)(l -methyl- 1 H-imidazol-5-yl)methanone (830 mg, 3.76 mmol, Intermediate 8: step b) under an atmosphere of nitrogen was added THF (30 mL) and the mixture was heated until a solution was obtained. To 6-bromo-2,4-dichloro-3-phenylquinoline (1.21 g, 3.42 mmol, Intermediate 7: step c) under an atmosphere of nitrogen was added THF (25 mL). The resulting colorless solution was cooled in a dry ice/acetone bath. «-BuLi (1 .6 M in hexane, 2.35 mL, 3.76 mmol) was added dropwise. The mixture was stirred for 5 minutes before addition of the THF solution of (4-chlorophenyl)(l ~methyl-lH-imidazol-5-yl)rnethanone via cannula. The reaction mixture was stirred in the dry ice/acetone bath for 30 minutes, then in an ice bath for 50 minutes and at room temperature for 55 minutes, then was quenched by addition of saturated, aqueous NH 4 C1 solution. The mixture was diluted with water and extracted three times with EtOAc. The organic phase was dried (NaiSCli), filtered, and concentrated and the residue was purified by flash column chromatography (silica gel, 0-4% MeOH-DCM) to afford the title compound. Ή NMR (400 MHz, CDCl ? ) δ ppm 8.32 (d, J = 1.96 Hz, 1H), 8.02 (d, J = 8.80 Hz, 1H), 7.72 (dd, J ----- 2.20, 8.80 Hz, 1 1 1 ). 7.48 - 7.56 (m, 3H), 7.30 - 7.38 (m, 7H), 6.40 (d, J = 5.22 Hz, I I I ). 3.39 (s, 3H); MS m/e 494.1 ( M I I ) ' .

Example 66: (2,4-Dich]oro-3-phenylquinoHii-6-yi)(l-met yl-lH midazo]-5-yI)[6- (trifluoromethyi)pyridm-3-yI]methanol » TFA

(l-Memyl-lH-irnidazol-5-yi)(6-(trifluoromemyl)p Tidin-3-yl)methanone (3,00 g, 8.50 mmol, Intermediate 2: step c) and 6-bromo-2,4-dichloro-3-phenylquinoline (2.32 g, 9.08 mmol, Intermediate 7: step c) were dissolved in THF (250 mL) under an N? atmosphere in a dry round bottom flask, then cooled to -78 °C in dry ice / acetone bath. «-BuLi (2.5 M in hexanes, 3.24 mL, 8.09 mmol) was then added dropwise via syringe over approximately 2 minutes. The contents were stirred at -78 °C for approximately 2.5 hours, then the dry ice bath was removed and the contents were allowed to warm to room temperature. The reaction was then cooled to 0 °C in an ice water bath and quenched with a saturated, aqueous NH 4 C1 solution, then transferred to a separately funnel with EtOAc dilution. The organic phase was separated, then the aqueous layer was back extracted twice with EtOAc, then the combined organic phases were dried over MgS0 4 , filtered and concentrated under reduced pressure. The crude product was purified by RP-HPLC (40-80% CH 3 CN-H 2 0, 0.1 % TF A) to afford the title compound as a yellow solid. MS (ESI): mass calcd. for C26H17CI2F3N4O, 528.1 ; m/z found, 529.4 [M+Hf; 1H NMR (600 MHz, CD 3 OD) δ ppm 8.80 (d, J = 2.1 Hz, IB), 8.35 (d, J = 1.8 Hz, 1H), 8.07 - 8.03 (m, 2H), 7.88 (dd, J 8.9, 2.1 Hz, 1 H), 7.85 (d, J = 8.3 Hz, 1 H), 7.76 (s, 1H), 7.54 - 7.50 (m, 3H), 7.37 - 7.33 (m, 2H), 6.39 (d . ./ 1.1 Hz, 1H), 3.48 (s, 3H).

Example 67: (2,4-DichIoro-3-phenyIquinoUn-6-yl)(2,4-dimethyIthiazOl-5-yl )(l-methyl-l/ - -triazol-5-yI)methanol

To a flask containing 6-bromo-2,4~dichloro-3-pheny!quinoline (750 mg, 2.12 mmol, Intermediate 7: step c) was added THF (30 mL) to give a homogeneous clear solution. The solution was cooled in a dry ice bath and «-BuLi (2.5 M in hexanes, 0.840 mL, 2.5 mmol) was introduced. After 2 minutes, a solution of (2,4-dimethylthiazol-5-yl }(! -methyl- 1H- 1 ,2,3-triazoi- 5-yl)methanone (600 mg, 2.7 mmol, Intermediate 16: step b) in 18 mL THF was added. The dry-ice bath was replaced with a 0 °C ice-bath and after 45 minutes the reaction mixture was quenched with NH 4 CI solution and the aqueous portion was extracted with EtOAc (4 x 50 mL). The combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated. The crude material was triturated with Et->0 to afford the title compound as a light tan solid. The mother liquors were chromatographed on silica gel (2% MeOH/DCM increasing to 5% MeOH) which provided additional title compound. Ή NMR (500 MHz, CDC1 3 ) δ 8.32 (d, J = 2.0 Hz, 1H), 8.09 (d, J = 8.9 Hz, IH), 7.73 (dd, J = 8.9, 2,2 Hz, 1H), 7.58 - 7.46 (m, 3H), 7.20-7.33 (m, 21 1 ). 7.20 (s, 1H), 4.38 (s, 1H), 3.94 (s, 3H), 2.58 (s, 3H), 2.1(S (s, 3H); MS (ESI): mass calcd. for C24H19CI2N5OS, 495.1 , m/z found 496.1 [M+H]".

Example 68: (2,4-DicMoro-3-phenyIquiBolin-6-yl)(2,4-dim

To a flask containing 6-bromo-2,4-dich[oro-3-phenylquinoline (450 mg, 1 .27 mmol, Intermediate 7: step c) was added THF (15 mL) to give a homogeneous clear solution. The solution was cooled in a dry ice bath and «-BuLi (2.5 M in hexanes, 0.45 mL, 1.13 mmol) was added. After 2 minutes, a solution of (2,4-dimethylthiazol-5-yi)(l -methyl- lH-imidazol-5- yijmethanone (350 mg, 1.58 mmol, Intermediate 17: step b) in 4 mL THF was introduced. The dry-ice bath was replaced with a 0 °C bath and after 35 minutes the reaction mixture was quenched with aqueous NH 4 CI solution and the aqueous portion was extracted with EtOAc (4 x 50 mL). The combined organies were washed with brine, dried over MgS0 4 , filtered and concentrated. Chromatography on silica gel (20% acetone-DCM increasing to 5% MeOH-DCM) afforded the title compound as a pale yellowish solid. ! H NMR (500 MHz, CDCI 3 ) δ 8.35 (d, J = 1.9 Hz, i l l ). 8.03 (d, J = 8.8 Hz, i l l ). 7.79 (dd, ./ 8.8, 2.1 Hz, IH), 7.59 - 7.41 (m, 31 1 ). 7.40 - 7.31 (m, 2H), 7.28 (d, J = 6.4 Hz, IH), 6.44 (s, IH), 5.46 (s, IH), 3.48 (s, 3H), 2.56 (s, 3H), 2.13 (s, 3H); MS (ESI): mass calcd. for C25H20CI2N4OS, 494.1 , m/z found 495.0 [M+H] + .

Example 69: fert-Butyl 4-((4-chloro-3-phenyl-2-(trifluoromethyl)quiiioliii-6- y1)(hydroxy)(pyridiii-3-yi)methyI)piperidine-l-carboxy1ate

To a solution of 4-chloro-6-iodo-3-phenyl-2-(trifluoromethyl)quinoline (Intermediate 20: step b, containing about 13% molar of 4-chloro-3-phenyl-2-( rifiuoromethyl)quinoline as impurity, 340 mg) in THF (4 mL) at -78 °C under N 2 was added /PrMgCl (2.0 M in THF, 0.40 mL, 0.8 mmol). After stirring for 8 minutes, the cooling bath was removed, and stirring was continued for 20 minutes, then terf-buty! 4-nicotinoylpiperidine-l-carboxylate (225 mg, 0.770 mmol, Intermediate 25 ) was added in neat. After stirring at room temperature overnight, the mixture became clear brown and was heated at 50 °C for 1.5 hours. The mixture was quenched with saturated NH 4 C1 aqueous solution, and extracted with EtOAc. The extracts were dried over Na 2 S0 4 , filtered, and concentrated. The crude mixture was purified by flash column chromatography (silica gel, 20- 100% EtOAc in heptanes) to provide the title compound. Ή NMR (400 MHz, CDCl ? ) δ 9.17 - 9.35 (m, 1H), 8.50 - 8.88 (m, 2H), 8.29 (d, J = 7.58 Hz, 1 1 1 ). 8.24 (d, J = 8.59 Hz, I I I ). 7.82 8.00 (m, 1H), 7.47 - 7.57 (m, 4H), 7.21 - 7.33 (m, 2H), 4.08 - 4.26 (m, 2H), 2.69 - 2.99 (m, 3H), 5.82 - 1.92 (m, IH), 1 .53 - 1.80 (m, 3H), 1 .47 (s, 4.51 1 ). 1.41 (s, 4.5H); MS m/e 598.3 [M+H] + .

Example 70: (4-Chloro-3~phenyl-2-(trifliioromethyI^^

3- I)methan.ol

A solution of ieri-butyl 4-((4-chloro-3-phenyl-2-(trifluoromethyl)quinolin-6- yi)(hydroxy)(pyridin-3-yl)methyl)piperidine-l-carboxylate (313 mg, 0.520 mmol, Example 69) in DCM (6 mL) was treated with 1.8 mL of TFA., stirred for 3 hours, and concentrated. The residue was partitioned between saturated NaHC0 3 aqueous solution and DCM. The organic layer was dried (Na 2 S0 4 ), filtered and concentrated to dryness to provide the title compound as a light brown solid, Ή NMR (400 MHz, CDCI 3 ) δ 8.86 (s, IH), 8.55 (s, IH), 8.44 (d, J= 4.55 Hz, I I I). 8.21 (d, ./ 9.09 Hz, I H), 7.89 - 7.96 (m, 2H), 7.46 - 7.53 (m, 41 1). 7.23 -7.32 (m, 2H), 3.23 - 3.36 (m, 2H), 2.77 - 2.91 (m, 3H), 1.66 - 1.86 (m, 4H).

Example 71: 4-Ch1oro~6~((4~chlorophenyI)(l-methy1-l/iM

dimethyl~3~phenylqumo] ~2~am e

To (4~chloro-2-(dimethyiamino)-3-phen^

5-yl)methanol (83.6 mg, 0.166 mmol, Example 90) in NMP (1.5 mL), was added copper (I) cyanide (16.4 mg, 0.1 83 rarnol). The mixture was heated by microwave irradiation for 10 minutes at 120 °C. The mixture was partitioned between MTBE and saturated aqueous Η 4 ΟΗ. The organic phase was washed with water. The organic phase was dried (Na 2 SC"4), filtered, and concentrated in vacuo, The residue was purified by RP-HPLC (10-90% CH 3 CN- H 2 0, 0.1% TFA) and fractions were converted to the corresponding tree base by extraction from saturated aqueous NaHC0 3 with DCM. Further purification by flash column chromatography (silica gel, 10-70% acetone-DCM) afforded the title compound 3 H NMR (400 MHz, CDCI 3 ) δ 7.82 (d, J = 1.96 Hz, IH), 7.76 (d, J = 8.80 Hz, IH), 7.55 (br. s., IH), 7.43 - 7.50 (m, 2H), 7.34 - 7.42 (m, 4H), 7.30 (d, J= 8.56 Hz, 2H), 7.12 (d, J= 8.56 Hz, 2H), 6.48 (br. s., IH), 5.45 (s, IH), 3.37 (s, 3H), 2.71 (s, 6H); MS ni/e 487.0 (M+Hf.

Example 72: 4-Chloro~6~((4-chlorophenyl)(l-methyl-lH midazol-5-yl)methyl)-3- pIie!iylqisiiiolisie~2~carbosiitrile e TFA

Copper (I) cyanide (44.4 mg, 0.496 mmol) was added to a colorless solution of (4- chlorophenyl)(2,4-dichioro~3~phenylquinolin~6~yl)(l -methyl- 1 H-imidazol-5-yl)methanol (123 nig, 0,248 mmol, Example 65) in NMP (1 mL). The mixture was stirred until CuCN dissolved, then was heated by microwave irradiation at 145 °C for 40 minutes. The mixture was partitioned between saturated aqueous NH 4 OH and MTBE. The aqueous phase was extracted twice with MTBE. The organic phases were combined, dried (Na 2 S0 4 ), filtered, and concentrated in vacuo. The residue was purified by RP-HPLC (10-90% (Ί hCN-i !.·( ) . 0.1% TFA) to afford the title compound. Ή NMR (400 MHz, CDC1 3 ) δ 8.68 (s, IH), 8.26 (d, J = 8.80 Hz, 1H), 8.05 (d, J ----- 1 .96 Hz, IH), 7.66 (dd, ./ 1.96, 8.80 Hz, 1H), 7.52 - 7.62 (m, 3H), 7.37 - 7.49 (m, 4H), 7.12 (d. J ------ 8.31 Hz, 21 1). 6.78 (s, 1 1 1). 5.63 (s, 1 1 1). 3.62 (s, 3H); MS in e 469.0 (M H ) . -Dichloro-3-(2-chIorophenyl)q«ino1in-6-y1)(phenyl)(jpyridin -4-yI)n^

To a solution of 6-bromo-2,4-dichloro-3-(2-chlorophenyl)quinoline (387.5 mg, 1 mmol, Intermediate 32: step c) in THF (15 mL) at -78 °C was added «BuLi (2.5 M in hexanes, 0.52 mL, 1.3 mmol) dropwise and the resulting mixture stirred at -78 °C for 15 minutes. Then, phenyl(pyridin-4-y])met,hanone (183 mg, 1 mmol) was added, and the resulting mixture stirred at -78 °C for 15 minutes. The dry ice - acetone bath was then removed and the mixture was allowed to warm to room temperature over 2 hours. The reaction was quenched by the addition of water (50 mL) and then extracted with DCM (2 x 20 mL). The organics were combined, dried (MgS0 ), filtered and concentrated to dryness. The residue was purified by FCC (1 :20 MeOH/DCM) to provide the title compound. MS (ESI): mass calcd. for C 27 H 17 CI 3 N 2 O, 490.0, m/z found 491.0 [M+Hf. j H NMR (300 MHz, CD 3 OD) δ 8.54 (dd, J ------ 4.7, 1.7 Hz, 21 1). 8.25 -

8.22 (m, IH), 8.06 - 8.02 (m, I H), 7.93 - 7.88 (m, IH), 7.64 - 7.60 (m, IH), 7.57 - 7.52 (m, IH), 7.52 - 7.50 (m, IH), 7.50 - 7.47 (m, 2H), 7.42 - 7.33 (m, 6H).

Example 74: (2,4-Dichloro-3-(2-chlorophenyI)quinolin-6-yi)(oxazol-2-yl)( phenyi)methanol

To a solution of 6-bromo-2,4-dichloro-3-(2-chlorophenyl)quinoline (387.5 mg, 1 mmol, Intermediate 32: step c) in THF (15 mL) at -78 °C was added wBuLi (2.5 M in hexanes, 0.52 mL, 1.3 mmol) dropwise and the resulting mixture stirred at -78 °C for 15 minutes. Then, phenyl(pyridin-4-yl)methanone (183 mg, 1 mmol) was added, and the resulting mixture stirred at -78 °C for 15 minutes. The dry ice - acetone bath was then removed and the mixture was allowed to warm to room temperature over 2 hours. The reaction was quenched by the addition of water (10 rnL) and then extracted with DCM (2 x 20 mL). The organics were combined, dried (MgS0 4 ), filtered and concentrated to dryness. The residue was purified by FCC (1 :5 EtO Ac/petroleum ether) to provide the title compound. MS (ESI): mass calcd. for

( .,Η ,^Ί ΓΝ . -Ο , 480.0, m/z found 481.0 [M+Hf. Ή NM (300 MHz, CD 3 OD) δ 8.34 (d, ./ 1.8 Hz, IH), 8.03-7.89 (m, 3H), 7.64-7.57 (m, 1H), 7.56-7.44 (m, 2H), 7.42-7.29 (m, 6H), 7.23 (s,

IH).

Example 75: 6~((3-Chloropheiiyl)(hydroxy)(6-(trifluoromethyl)pyridiii-3~ yl)methyl)~3- phenylqumoline~2~carfoomtrile

A pressure tube containing (3-chloiOphenyl)(2,4-dichloro-3-phenylquinolin-6-yl)(6- (trifluoromefhyl)pyridin-3-yl)methanol (94 mg, 0.17 mmol, Intermediate 22: step b), S : 'd >- dba) .- (8.0 mg, 0.0087 mmol), l, -bis(diphenylphosphino)ferrocene (dppf, 10 mg, 0.018 mmol), zinc cyanide (25 mg, 0.21 mmol), and zinc nanopowder (3.0 mg, 0.046 mmol) in N,N- dimefhylacetamide (1 mL) was sparged with nitrogen for 5 minutes, and then heated at 120 °C for 1 hour followed by 100 °C for 3 hours. The mixture was allowed to cool to room temperature and filtered through a syringe filter. The filtrate was concentrated in vacuo, then EtOAc and H 4 OH (aqueous) were added. The organic layer was separated, and the aqueous layer was extracted with EtOAc, The combined organic layers were dried (Na 2 S0 4 ), filtered, and concentrated. The residue was purified by flash column chromatography (silica, gel column, 30-50% EtOAc in heptane), and then by reverse phase HPLC (water/acetonitrile/O. l % TEA) to provide the title compound as a by-product. X H NMR (400 MHz, CDCI 3 ) δ 8.76 (d, J = 2.02 Hz, 1 1 1 ). 8.25 (s, 1 1 1 ). 8.20 (d, ./ 9.60 Hz, 1 H), 7.93 (dd, 2.02, 8.08 Hz, 1 H), 7.75 - 7.79 (m, 21 1 ;·. 7.71 (d, J = 8.08 Hz 1 H), 7.60 - 7.65 (m, 2H), 7.51 - 7.58 (m, 3H), 7.32 - 7.40 (m, 31 1 ;·. 7.15 (dt. ./ 1.52, 7.58 Hz, 1 H).

Example 76: 6-((3-Chloropheiiyi)(hydroxy)(2-(trifluoromethyI)pyridiii-4- yl)methyI)-3- heny]qumolme~2,4~dkarbonitrile

The title compound was prepared using (3-chlorophenyl)(2,4~dichloro-3-phenylquinolin~6-yl)(2~ (txifluoromethyl)pyridin-4-yl)methanol (Example 77) in place of (3-chlorophenyl)(2,4-dichloro- 3~phenylquinolin~6~yl)(6-(trifluoromethyl)pyridin-3-yl)metha nol (Intermediate 22: step b) according to the procedure described in Example 75. 5 H NMR (400 MHz, CDCI 3 ) δ 8.81 (d, J :::: 5.56 Hz, 1H), 8.46 (d, J = 1.52 Hz, 1H), 8.31 (d, J = 9.09 Hz, 1H), 7.90 (d, J = 1.52 Hz, 1H), 7.84 (dd, J = 2.02, 8.59 Hz, 1 H), 7.60 - 7.67 (m, 5H), 7.58 (dd, J = 1 .52, 5.05 Hz, 1H), 7.35 - 7.46 (m., 2H), 7.23 - 7.27 (m, 1H), 7.10 (dt, ./ 1.52, 8.08 Hz, H I ).

Example 77: (3-Chlorophenyl)(2,4-dichloro-3-pheiiylquiiiolin-6-yl)(2-

(trifluoromethyl)pyridin-4-yl)methaiiol

To a solution of 6-bromo-2,4-dichloro-3-phenylquinoline (286 mg, 0.810 mmol, Intermediate 7: step c) and (3-c3iloroplienyl)(2-(trifluorometliyl)pyridin-4-yl)methanon e (231 mg, 0.810 mmol, Intermediate 23: step b) in THF (6 mL) at -78 °C was added 1.6 M #-BuLi in hexaiie (0,76 mL,

1.22 mmol). The mixture was stirred at -78 °C to 10 °C for 2 hours and then quenched with NH 4 CI (aqueous). The organic layer was separated, and the aqueous layer was extracted with CH 2 CI 2 . The combined organic phases were dried (Na 2 S04), filtered, concentrated, and purified by flash column chromatography (40 g silica gel column, 10 - 40% EtOAc in heptane) to afford the title compound as a yellow solid. ! H NMR. (400 MHz, CDCI 3 ) δ 8.74 (d, J = 5.56 Hz, 1H),

8.23 (d, J 2.53 Hz, 1H), 8.09 (d, J = 9.09 Hz, 1H), 7.84 (d, J ------ 1.52 Hz, I H), 7.68 (dd, J ------

2.02, 8.59 Hz, IH), 7.46 - 7.56 (m, 4H), 7.30 - 7.40 (m, 4H), 7,25 - 7.28 (m, IH), 7.13 (dt, J = 1.52, 7.58 Hz, IH).

Example 78: (2,4-Dichloro-3-pheBylquinolin-6-yl)(l-methylpiperidin-4- i)(pyridin-3- yl)methanol « TFA

To a mixture of (2,4-dichloro-3-phenylquinolin-6-yl)(piperidin-4-yl)( yridin-3-yl)methanol*TFA (15 mg, 0.022 mmol, Example 91), formaldehyde (0.010 mL, 0.13 mmol, 37% in water), and eOH (1 mL) was added NaCNBEL (4.0 mg, 0.063 mmol). After stirring at room temperature overnight, the mixture was concentrated and purified by reverse phase HPLC (water / acetonitri!e / 0.1% TFA) to provide the title compound as a white solid. ] H NMR (400 MHz, CD 3 OD) 5 8.97 (s, I H), 8.56 - 8.62 (m, 2H), 8.44 (d, J = 8.31 Hz, IH), 8.03 (s, 2H), 7.70 - 7.76 (m, IH), 7.47 - 7.57 (m, 3H), 7.33 (d, J = 6.85 Hz, 2H), 3.50 - 3.60 (m, 2H), 3.03 - 3.19 (m, 3H), 2.86 (s, M l ). 1.82 ~- 1.98 (m, 2H), 1.64 ~- 1.77 (m, 2H): MS m/e 478.0 [M+H] + ,

Example 79: (3-ChlorophenyI)(2-isopropoxy-3-pheBylqmnoIin-6-yI)(pyridiii -3- yI)methanol » TFA

A. mixture of (3-chlorophenyl)(2,4-dichloro-3-phenyl.quinolin-6-y].)(pyrid in-3-yl)methanol (21 mg, 0.043 mmol, Intermediate 24) and NaQj ' Pr (1 1 mg, 0.13 mmol) in ?PrOH (0.4 mL) was heated in a sealed tube at 80 °C for 5 hours. More NaO/Pr (15 mg, 0.1 8 mmol) was added and the mixture was heated at the same temperature for 21 hours. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / acetonitrile / 0, 1% TFA) to provide the title compound as a by-product. 3 H NMR (400 MHz, CD 3 OD) δ 8.86 (s, 1H), 8.75 - 8.81 (m, 1 1 1 ). 8.52 (d, ./ = 8.31 Hz, 1 1 1 ). 8.03 (s, IH), 7.97 - 8.03 ( m. IH), 7.82 (d, 8.80 Hz, 1H), 7.65 (d, J ~ 2.20 Hz, IH), 7.56 - 7.63 (m, 3H), 7.46 - 7.48 (m, I H), 7.33 - 7.45 (m, 5H), 7.25 - 7.29 (m, IH), 5.59 - 5.66 (m, I H), 1.38 (d, ,/ 6.36 Hz, 6H).

Example 80: (4-Butyl-2-isopropoxy-3-pheBylquinolin-6-yI)(l-methyI-lH-imi dazol-5- yl)(pyridin-4-yl)methanoI » TFA

A mixture of (4-butyl-2-chloro-3-phenylquinolin-6-yl)( 1 -methyl- 1 H-imidazol-5-yl }(pyridin-4- yi)methanol » TFA (16 mg, 0.023 mmol, Intermediate 26) and NaO/Pr (19 mg, 0.23 mmol) in / " PrOH (0.4 mL) was heated in a sealed tube at 80 °C for 17 hours. More NaO/Pr (7.0 mg, 0.085 mmol) was added and the mixture was heated at the same temperature for 64 hours. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / acetonitrile / 0.1 % TFA) to provide the title compound. h H NMR (400 MHz, CD 3 OD) δ 9.06 (s, I H), 8.78 (d, ./ 6.57 Hz, 2H), 7.86 - 7.90 (m, 4H), 7.67 (dd, ./ 2,02, 9.09 Hz, 1H), 7.36 - 7.47 (m, 3H), 7.16 - 7.20 (m, 2H), 7.12 (d, ./ 1 .52 Hz, IH), 5.44 - 5.52 (m, 1 H), 3.72 (s, 3H), 2.66 - 2.74 i m. 2H), 1.34 - 1 .44 (m, 21 1 ). 1.22 (d, J = 6.06 Hz, 6H), 1.12 - 1.19 (m, 2H), 0.73 (t, J 7.33 Hz, 3H); MS m/e 507.3 [M+H] + .

Example 81 : (3-Chlorophenyl)(2,4-diethoxy-3-phenylquinolin-6-yl)(pyridin -3- yl)methanol » TFA

A. mixture of (3-cUorophenyl)(2,4-dich].oro-3-phenylquinolin-6-yl)(pyridin -3-yl)m.ethanol (27 nig, 0.055 mmol, Intermediate 24) and NaOEt (0.30 mL, 3.8 mmol, 21% w in EtOH) was heated in a sealed tube at 82 °C for 24 hours. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / acetonitrile / 0.1 % TFA) to provide the title compound. ! H NMR (400 MHz, CD 3 OD) δ 8.89 (s, 1H), 8.81 (s, 1H), 8.55 (d, J = 8.07 Hz, 1 H), 8.05 (br. s., I I I ). 7.87 (s, IH), 7.84 { ± ./ 8.80 Hz, IH), 7.61 (d, ./ 7.09 Hz, IH), 7.49 (s, IH), 7.33 - 7.45 (m, 7H), 7.29 (br. s., IH), 4.47 (q, ./ 7.09 Hz, 2H), 3.51 - 3.63 (m, 2H), 1.30 (t, J = 6.97 Hz, 3H), 0.95 (!. ./ 7.09 Hz, 3H).

Example 82 : (4-Chloro-2-(methyl(2-(met ylamino)et yl)amiiio)-3-pheiiyiqumoMn-6-yI)(l - methyI-lH-imidazoI-5-yI)(pyridiii-4-yl)methano1 » TFA

A mixture of (2 ,4~dichloro-3-pheny!quinolin-6-y!)( 1 -methyl- lH-imidazol-5-yl)(pyridm-4- yl)methanol » TFA (18 nig, 0.026 mmol, Intermediate 25: step c) and

diamine (500 mg, 5.67 mmol) was heated in a sealed tube at 80 °C for 24 hours. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / acetonitrile / 0.1%

14 ' TFA) to provide tlie title compound. ! H NMR (400 MHz, CD 3 OD) δ 9.09 (s, III).8.87 (br. s.. 211).8.25 (d, J= 2.02 Hz, III).8.09 (d, J = 6.57 Hz, 211).7.90 (d, J= 9.09 Hz, III).7.72 (dd, J = 2.02, 8.59 Hz, 1H), 7.49 - 7.59 (m, 2H), 7.43 - 7.49 (m, 1H), 7.35 - 7.43 (m, 2H), 7.21 (d, J 1.52 Hz, 1H), 3.76 - 3.88 (m, 2H), 3.70 (s, 3H), 3.22 - 3.29 (m, 2H), 2.76 (s, 3H), 2.51 (s, 3H); MS m/e 513.0 iV!-ilj .

Example 83: 2-((4-CMoro-6-(hydroxy(l-niethyl-l J

phenylquinolin-2-yl)(inethyl)ainino)etha»ol*TFA

A mixture of (2,4-dic oro-3-phenylquinolin-6-yl)( 1 -methyl- lH-imidazol-5-yl)(pyridin-4- yi)methanol » TFA (20 mg, 0.029 mmol, Intermediate 25: step c) and 2~(niethylamino)etlianol (408 mg, 5.43 mmol) was heated in a sealed tube at 80 °C for 16 hours. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / acetonitrile / 0.1% TFA) to provide the title compound. J H NMR (400 MHz, CD 3 OD) δ 9.08 (s, IH), 8.81 (br. s., 2H), 8.37 (s, 1H), 7.97 (d, J= 6.06 Hz, 2H), 7.89 (s, 2H), 7.59 - 7.64 (m, 1H), 7.52 - 7.58 (m, 2H), 7.42 - 7.47 (m, 2H), 7.21 (d,J= 1.52 Hz, 1H), 3.81 (t, J= 4,55 Hz, 2H), 3.69 (s, 3H), 3.64 (t, J = 4.55 Hz, 2H), 2.78 (s, 3H); MS m e 500.0 |M II| .

Example 84: (4-ChIoro-2-(dimethylammo)-3-phenylquinoHii-6-yi)(pyridin-2- y1)(pyri^^ yl)methan o1*TFA

A mixture of (2,4-dichloro-3-phenylquinolin-6-yr)(pyridin-2-yl)(pyridin-4 -yl)methanol*TFA (35 mg, 0.051 mmol. Intermediate 27) and dimethylamine (0.8 mL, 1.6 mmol, 2.0 M in MeOH) was heated in a sealed tube at 80 °C for 4,5 days. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / aeetonitriie / 0, 1% TFA) to provide the title compound, f H N.MR (400 MHz, CD 3 OD) δ 8.81 (d, J = 7.09 Hz, 2H), 8.57 - 8.61 (m, 1H), 8,20 - 8.26 (m, 3H), 8.06 (d. J ------ 8.80 Hz, IH), 7.92 - 7.98 (m, H i ). 7.85 - 7.92 i m. 2H), 7.50 - 7.61 (m, 3H),

7.37 - 7.47 (m, 3H), 3.00 (s, 6H).

Example 85: (2~Chloro-4-(dimethylammo)-3-phenylquinoHn-^^

yl)(pyridin-2-yl)methaiiol*TFA

A mixture of (2,4-dichloro-3-phenylquinolin-6-yl)( l -methyl- lH-imidazol-5-yl)(pyridm-2- yl)methanol (28 mg, 0.41 mrnol, Example 64) and dimethyl amine (0,8 rnL, 1.6 mrnol, 2.0 M in MeOH) was heated in a sealed tube at 80 °C for 4.5 days. After cooling to room temperature, the mixture was purified by reverse phase HPLC (water / aeetonitriie / 0.1% TFA) to afford the title compound. ! H NMR (400 MHz, CD 3 OD) δ 8.94 (s, IH), 8.59 - 8.64 (m, IH), 8.1 8 - 8.20 (m, IH), 7.87 - 7.96 (m, 3H), 7.77 (d, J = 8.07 Hz, IH), 7.41 - 7.53 (ni, 4H), 7.25 - 7,32 (m, 2H), 7.03 (d, ./ 1.71 Hz, IH), 3.64 (s, 3H), 2.69 (s, 6H); MS m/e 470.0 [M+H] + .

Example 86: (2,4-DichIoro-3-phenylquinoUn-6-yI)(2-fluoropyridin-4-yI)(l- methyI-lH-

The title compound was prepared using (2-fuoropyridin-4-yl)(l -methyl- lH-imidazol-2- yi)methanone (Intermediate 28: step b) in place of (3-chlorophenyl)(2-(trifluoromethyl)pyridin- 4-yl)methanone (intermediate 23: step b) according to the procedure described in Example 77.

Ή NMR (400 MHz, CDC1 3 ) δ 8.18 (d, J = 1.96 Hz, IH), 8.15 (d, J = 5.38 Hz, IH), 8.05 (d, J = 8.80 Hz, I H), 7.73 (dd, J --- 2,20, 8.80 Hz, I H), 7.49 - 7.53 (m, 3H), 7.27 - 7.34 (m, 2H), 7.21 (dt, ./ 1.59, 5.38 Hz, 1H), 6.99 (s, lH), 6.91 (d, ./ 1.22 Hz, 1H), 6.88 (d, ./ 1.22 Hz, IH), 3.39 (s, M l ): MS m/e 478.8 [M+H] + .

Example 87: (4-ChloropheiiyI)(2,4-dichIoro-3-pheiiyIquinoMn-6-y1)(2,4-di methyIt iazol-5-

To a flask containing 6-bromo-2,4-dichioro-3-phenylquinoline (255 mg, 0,72 mniol, Intermediate 7: step c) was added THF (8 mL) to give a homogeneous clear solution. The solution was cooled in a -78 °C bath and then «-BuLi (2.5 M in hexanes, 0.26 mL, 0.65 mmol) was added which resulted in an immediate orange homogeneous solution. After approximately 2 minutes, (4-chlorophenyl)(2,4-dimethylthiazol-5-yl)methanone (210 mg, 0.83 mmol, Intermediate 30, in 3 mL THF) was added. The reaction mixture was maintained at -75 °C for 5 minutes then replaced with a 0 C C ice-bath. After 45 minutes, the reaction mixture was quenched with aqueous NH 4 CI solution. The aqueous portion was extracted with EtOAc (3 x 50 mL) and the combined organics were washed with brine, dried over MgS0 4 , filtered and concentrated to dryness. Chromatography on silica gel (100% DCM increasing to 20% EtOAc-DCM) provided the title compound as a white amorphous solid. Ή MR (500 MHz, CD 2 CI 2 ) δ 8.33 (s, 1H), 8.01 (d, J = 8.8 Hz, IH), 7.82 (dd, J = 8.9, 2.1 Hz, IH), 7.59 - 7.46 (m, 3H), 7.46 - 7.28 (m, 6H), 2.54 (s, 3H), 2.03 (s, 3H). MS (ESI): mass calcd. for Q27H19CI3N2OS, 524.0, m/z found 525.0 [M+H] + .

(2,4-Dimethoxy-3-pheiiyIqumoMn-6-yI)(l-methy1-lH ' midazoI-5-

Purification of the crude reaction mixture from the synthesis of (4-ehloro-2-mefhoxy-3- phenylquinolin-6-yl)( 1 -methyl- lH-imidazol-5-yi)pyrimidin-2-ylmethanol (Example 60a) also afforded the title compound as a regioisomer. MS (ESI): mass calcd. for C20H23N5Q3, 453,2; m/z found, 454.3 [M+Hf. ! H MR (400 MHz, CD 3 OD) δ 8.86 (d, J = 4.9 Hz, 2H), 8.26 (d, J = 1.7 Hz, 1 H), 7.86 - 7.75 (m, 3H), 7.47 - 7.36 (m, 6H), 6.52 (s, 1H), 3.95 (s, 3H), 3.48 (s, 3H), 3.44 (s, 3H).

Example 89: (2- hloro-^-^heny1quinolm-6-yl)(4-cM

yl)methaiiol

To a 50-mL round-bottom flask containing a solution of 6-bromo-2-chloro-3-phenylquinoline (210 mg, 0.66 mmol, Intermediate 31 : step b) in ietrahydrofuran (10 mL) was added 2.5 M n- BuLi in hexanes (0.29 mL, 0.72 mmol) dropwise with stirring at -78 °C. After 45 minutes at - 78 °C, a solution of (4-chlorophenyl)(l-methyl-lH-imidazol-5-yl)methanone (132 mg, 0.60 mmol, intermediate 8: step b) in tetrahydrofuran (2 mL) was added dropwise. The resulting solution was stirred at -78 °C for an additional 2 hours, then quenched with 30 mL of aqueous ΝΗ 4 Ο and extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by FCC (20: 1 DCM/MeOH) to provide the title compound as a white solid. MS (ES): mass calcd. for C26H19CI2N3O, 459.1 ; m/z found, 460.0 [ M i l l . Ή NMR (400 MHz, CD 3 OD) δ 8.96 (d, ./ = 3.9 Hz, 1 1 1 }. 8.33 (s, 1 1 1 ). 8.05 (d, ./ = 6.0 Hz, 1 H), 7.94 (s, 1 H), 7.88 (d, ./ = 6.6 Hz, I I I ). 7.42- 7.56 (m, 9H), 6.93 (s, 1 1 1 ;·. 3.71 (s, 3H).

Example 90: (4-Chloro-2~(dimethylamino)-3-phenylquinoIiii-6-yl)(4~chloro pheiiyl)(l- methyl~lH-imidazol~5-yl)methanol

(4-Chl.orophenyl)(2,4-dichloro-3-phenylquinolin-6-yl)(l -methyl- lH-imidazol-5-yl)methanol (124 mg, 0.251 mmol, Example 65) was treated with dimethy!amine (2 M in MeOH, 2 mL, 4 mmol) and the resulting suspension was heated in a sealed tube in an 85 °C oil bath for 2 days. The mixture was concentrated in vacuo and the residue was purified by flash column chromatography (Biotage NH column, 0-1% MeOH-DCM) to provide the title compound as a cream-colored foam . 3 H NMR (400 MHz, CDCI 3 ) δ 8.10 (d, J = 2.20 Hz, 1H), 7.72 (d, J = 8.80 Hz, 1H), 7.43 - 7.52 (m, 3H), 7.34 - 7.43 (m, 3H), 7.22 - 7.34 (m, 6H), 6.33 (s, 1H), 3.36 (s, 3H), 2.72 (s, 61 1 ).

Example 91: (2,4-Dic loro-3-phenylq«moMn-6-yI)(piperidin-4-yl)(jpyridin y1)methanol » TFA

A solution of tert-butyl 4~((2,4-dichloro-3~phenylquinoHn-6-yl)(hydroxy)(pyridin-3- yl)m.ethyl)piperidine-i-carboxylate (149 mg, 0.260 mmol. Intermediate 29) and TFA (1 ml.) was stirred at room temperature for 1 hour and concentrated to provide the title compound. X NMR (400 MHz, CD 3 OD) δ 9.12 (s, 1 H), 8.69 - 8.77 (m, 2H), 8.62 (s, 1H), 8.02 - 8.1 1 (m, 2H), 7.97 (dd, J = 5.56, 8.08 Hz, IH), 7.49 - 7.58 (m, 3H), 7.30 - 7.36 (m, 2H), 3.40 - 3.50 (m, 2H), 3.17 - 3.27 (m, i l l ). 3.05 ~- 3.17 (m, 2H), 1.75 - 1.94 (m, 2H), 1.60 4.74 (m, 2H).

IN VITRO BIOLOGICAL DATA ThermoFluor® Assay

ThermoFluor® is a fluorescence based assay that estimates ligand binding affinities by measuring the effect of a ligand on protein thermal stability (Pantoliano, M. W., Petrella, E, C, Kwasnoski, J. D., Lobanov, V. S., Myslik, J., Graf, E., Carver, T,, Asel, E., Springer, B, A,, Lane, P., and Salemme, F. R. (2001) High-density miniaturized thermal shift assays as a general strategy for drug discovery, J Biomol Screen 6, 429-40, and Matulis, D., Kranz, J. ., Salemme, F, R., and Todd, M. J. (2005) Thermodynamic stability of carbonic anhydrase: measurements of binding affinity and stoichiometry using ThermoFluor. Biochemistry 44, 5258-66). This approach is applicable to a wide variety of systems, and rigorous in theoretical interpretation through quantitation of equilibrium, binding constants

In a ThermoFluor® experiment where protein stability is monitored as the temperature is steadily increased, an equilibrium binding ligand causes the midpoint of an unfolding transition (T in ) to occur at a higher temperature. The shift in the melting point described as a AT m is proportional to the concentration and affinity of the ligand. The compound potency may be compared as a rank order of either AT m values at, a single compound concentration or in terms of Kp. values, estimated from concentration response curves.

RORyt ThermoFluor® Assay Construct

For the RORyt construct used in the ThermoFluor® assay, numbering for the nucleotide sequences was based on the reference sequence for human RORyt, transcript variant 2, NCBl Accession: NM_001001523.1 (SEQ ID NO: l). Nucleotides 850-1635 (SEQ ID NO: 2) coding for the wild type human RORyt ligand binding domain (RORyt LBD) were cloned into the pHIS l vector, a modified pET E. coli expression vector (Accelagen, San Diego), containing an in-frame N-terminal His-tag and a TurboTEV protease cleavage site (ENLYFQG, SEQ ID NO: 3) upstream of the cloned insert sequence. The amino acid sequence for the RORyt construct used in the Thermofluor assay is shown as SEQ ID NO:4. ThermoFluor® experime ts were carried out using instruments owned by Janssen Research and Discovery, L.L.C. through its acquisition of 3-Dimensional Pharmaceuticals, Inc. 1 ,8-ANS (Invitrogen) was used as a fluorescent dye. Protein and compound solutions are dispensed into black 384- well polypropylene PCR microplates (Abgene) and overlayed with silicone oil (1 ,uL, Fluka, type DC 200) to prevent evaporation.

Bar-coded assay plates are robotically loaded onto a thermostatically controlled PCR -type thermal block and then heated at a typical ramp-rate of 1 °C/min for all experiments. Fluorescence was measured by continuous illumination with UV light (Hamamatsu LC6) supplied via fiber optic and filtered through a band-pass filter (380-400 nm; >6 OD cutoff). Fluorescence emission of the entire 384-well plate was detected by measuring light, intensity using a CCD camera (Sensys, Roper Scientific) filtered to detect 500 ± 25 nm, resulting in simultaneous and independent readings of all 384 wells. Images were collected at each temperature, and the sum of the pixel intensity in a given area of the assay plate was recorded versus temperature. Reference wells contained RORyt without compounds, and the assay conditions were as follows:

0,065 mg/niL RORyt

60 μΜ 1 ,8-ANS

100 mM Hepes, pH 7.0

lO mM ' NaCl

2,5 mM GSH

0.002% Tween-20

Project compounds were arranged in a pre-dosed mother plate (Greiner Bio-one) wherein compounds are serially diluted in 100% DMSO by 1 :2 from a high concentration of 10 mM over 12 columns within a. series (column 12 is a reference well containing DMSO, no compound). The compounds were robotically dispensed directly into assay plates (lx = 46 iiL) using a Hummingbird capillary liquid handling instrument (Digilab). Following compound dispense, protein and dye in buffer was added to achieve the final assay volume of 3 μΐ.., followed by 1 μΐ., of silicone oil. The binding affinity was estimated as described previously (Matulis, D., Kranz, J. K., Salemme, F. R., and Todd, M. j. (2005) Thermodynamic stability of carbonic anhydrase: measurements of binding affinity and stoichiometry using ThermoFluor®. Biochemistry 44, 5258-66) using the following thermodynamic parameters of protein unfolding:

Reference RORyt T m : 47.8 °C

ΔΗ ,„) = 1 15 kcal/mol

CELL BASED BIOLOGICAL DATA

RORyt Reporter Assay

A reporter assay was used to test functional activity of RORyt modulatory compounds on transcriptional activation driven by the RORyt LBD. Ceils used in the assay were co-transfected with two constructs. The first construct, pBIND-RORyt LBD, contained the wild type human RORyt LBD fused to the DNA binding domain of the GAL4 protein. The second construct, pGL4.31 (Promega Cat no. C935A), contained multiple GAL4 responsive DNA elements upstream of firefly luciferase. To generate a background control, cells were similarly co- transfected with two constructs, but in the first construct the AF2 amino acid motif in the RORyt LBD was changed from LYKELF (SEQ ID NO: 5) to LFKELF (SEQ ID NO:6). The AF2 mutation has been shown to prevent co-activator binding to the RORyt LBD, thus preventing transcription of firefly luciferase. The mutant construct was called pBIND -RORyt- AF2.

For the RORyt constructs used in the reporter assay, numbering for the nucleotide sequences was also based on the reference sequence for human RORyt, transcript variant 2, NCBI Accession: N V! 00 ; 001 23. 1 (SEQ ID NOT). For the wild type human RORyt LBD construct, pBIND- RORyt LBD, nucleotides 850-1635 (SEQ ID NO:2) coding for the wild type human RORyt LBD were cloned into EcoRI and Notl sites in the pBIND vector (Promega cat. No E245A). The pBIND vector contains the GAL4 DNA Binding Domain (GAL4 DBD) and the renilla luciferase gene under control of the SV40 promoter. Renilla luciferase expression serves as a control for transfection efficiency and cell viability. For the background control construct, pBIND-RORyt- AF2, the AF2 domain of RORyt LBD was mutated using the Quik Change IS Site Directed Mutagenesis System (Stratagene Cat. No. 200519). The nucleotide sequence coding for the RORyt LBD sequence with the mutated AF2 domain is shown as SEQ ID NO:7. The amino acid sequences for the wild type RORyt LBD and RORyt LBD with the mutated AF2 domain are shown as SEQ ID N():8 and SEQ ID NO:9, respectively.

The reporter assay was performed by transiently transfecting HE 293T cells with 5 μg of pBIND-RORyt LBD or BIND -RORyt LBD-AF2 and 5 p.g pGL4.31 (Promega Cat no. C935A) using Fugene 6 (Invitrogen Cat no. E2691) at a 1 :6 ratio of DNA: Fugene 6 in a T-75 flask in which cells were at least 80% confluent. Twenty four hours after bulk transfection, cells were plated into 96-w r ell plates at 50,000 cells/well in phenol-red free DMEM containing 5% Lipid Reduced FCS and Pen/Strep, Six hours after plating, cells were treated with compounds for 24 hours. Media was removed and cells were lysed with 50 μΕ lx Glo Lysis Buffer (Promega). Dual Glo Luciferase Reagent (50 .aL/well) was then added and firefly luciferase luminescence was read on an Envision after a ten minute incubation. Finally, Stop and Glo reagent, (50 pL/well) was added and renilla luciferase luminescence was read on an Envision instrument, after a ten minute incubation. To calculate the effect of compounds on RORyt activity, the ratio of firefly to renilla luciferase was determined and plotted against compound concentration. Agonist compounds increase RORyt-driven luciferase expression, and antagonist or inverse agonist compounds decrease luciferase expression.

Human Thl7 Assay

The human Thl7 assay tests the effect of RORyt modulatory compounds on IL-17 production by CD4 T cells under conditions which favor ThI7 differentiation. Total CD4 1 T cells were isolated from the peripheral blood mononuclear cells (PBMC) of healthy donors using a CD4 : T cel l isolation kit II. following the manufacturer ' s instructions (Miltenyi Biotec). Cells were resuspended in a medium of RPMS-I640 supplemented with 50% fetal bovine serum, penicillin, streptomycin, giutamate, and β-mercaptoethanol and were added to 96-weil plates at 1.5x10 s per 100 μί_ per well. 50 μΕ of conipoimd at titrated concentrations in DMSO were added into each well at final DMSO concentration at 0.2%. Cells were incubated for 1 hour, then 50 μL· of Thl 7 cell differentiation medium was added to each well. The final concentrations of antibodies and cytokines (R&D Systems) in differentiation medium were: 3xl0 6 /mL anti-CD3/CD28 beads (prepared using human T cell activation/expansion kit, Miltenyi Biotec), 10 μg/mI, anti-IL4, 10 μ^ηΛ anti-IFNy, 10 ng mL ΙΙ.1 β, 10 ng mL 11,23, 50 ng/mL IL6, 3 ng/mL TGFp and 20 U/mL 1L2. Cells were cultured at 37 °C and 5% C0 2 for 3 days. Supernatants were collected and the accumulated lL-17 in culture was measured by using MULTI-SPOT® Cytokine Plate following manufacture's instruction (Meso Scale Discovery). The plate was read using Sector Imager 6000, and IL-1 ' 7 concentration was extrapolated from the standard curve. The IC50s were determined by GraphPad.

Table 1

Table 1 , continued

Table 1 , continued

Table 1 , continued

Table 1 , continued

Table 1 , continued

All data shown in Table 1 is either the value of one data point or the average of more than one data point. In cases where more than one value is shown in a table cell, values with qualifiers such as ~, > or < shown on the right side of the table cell could not be included in the averaging calculation for the value shown on the left side of the table cell . ND - no data.

While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, it will be understood that the practice of the invention encompasses all of the usual variations, adaptations and'Or modifications as come within the scope of the following claims and their equivalents.

Ail documents cited herein are incorporated by reference.