Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CYCLOPENTYL ACIDS AS LPA ANTAGONISTS
Document Type and Number:
WIPO Patent Application WO/2020/060915
Kind Code:
A1
Abstract:
The present invention provides compounds of Formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof, wherein all the variables are as defined herein. These compounds are selective LPA receptor inhibitors.

Inventors:
CHENG PETER TAI WAH (US)
KALTENBACH III ROBERT F (US)
SHI JUN (US)
SHI YAN (US)
ZHANG HAO (US)
Application Number:
PCT/US2019/051274
Publication Date:
March 26, 2020
Filing Date:
September 16, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BRISTOL MYERS SQUIBB CO (US)
International Classes:
A61K31/42; A61K31/4439; A61K31/505; A61P11/00; A61P19/10; A61P35/00; C07D261/08; C07D401/04; C07D401/14; C07D403/04; C07D417/04; C07D417/14
Domestic Patent References:
WO2017223016A12017-12-28
WO2011071570A12011-06-16
WO2002062389A12002-08-15
WO2012137982A22012-10-11
WO2008097428A22008-08-14
WO2010141761A22010-12-09
WO2017223016A12017-12-28
Foreign References:
US20110082164A12011-04-07
JP2017095366A2017-06-01
Other References:
FUJIWARA ET AL., J BIOL. CHEM., vol. 280, 2005, pages 35038 - 35050
ZHAO, Y. ET AL., BIOCHIM. BIOPHYS. ACTA (BBA)-MOL. CELL BIOL. OF LIPIDS, vol. 1831, 2013, pages 86 - 92
CHOI, J. W., ANNU. REV. PHARMACOL. TOXICOL., vol. 50, 2010, pages 157 - 186
KIHARA, Y. ET AL., BR. J. PHARMACOL., vol. 171, 2014, pages 3575 - 3594
MOOLENAAR ET AL., BIOESSAYS, vol. 26, 2004, pages 870 - 881
VAN LEEWEN ET AL., BIOCHEM. SOC. TRANS., vol. 31, 2003, pages 1209 - 1212
BRINDLEY, D., J. CELL BIOCHEM., vol. 92, 2004, pages 900 - 12
BAKER ET AL., ANAL. BIOCHEM., vol. 292, 2001, pages 287 - 295
ONORATO ET AL., J. LIPID RES., vol. 55, 2014, pages 1784 - 1796
GOETZL ET AL., SCIENTIFIC WORLD J., vol. 2, 2002, pages 324 - 338
BIOORG. MED. CHEM. LETT., vol. 23, 2013, pages 3833 - 3840
SIMON ET AL., J. BIOL. CHEM., vol. 280, 2005, pages 14656 - 14662
ROCKEY, D. C. ET AL., NEW ENGL. J. MED., vol. 372, 2015, pages 1138 - 1149
TAGER ET AL., NATURE MED., vol. 14, 2008, pages 45 - 54
NAKAGAWA ET AL., CANCER CELL, vol. 30, 2016, pages 879 - 890
"IUPAC Recommendations", PURE AND APPLIED CHEMISTRY, vol. 68, 1996, pages 2193 - 2222
WIDDER, K. ET AL.: "Methods in Enzymology", vol. 112, 1985, ACADEMIC PRESS, pages: 309 - 396
BUNDGAARD, H.: "A Textbook of Drug Design and Development", article "Design and Application of Prodrugs", pages: 113 - 191
"Comprehensive Organic Synthesis: Selectivity, Strategy & Efficiency in Modern Organic Chemistry", 1991, PERGAMON PRESS
BUNDGAARD, H., ADV. DRUG DELIV. REV., vol. 8, 1992, pages 1 - 38
BUNDGAARD, H. ET AL., J. PHARM. SCI., vol. 77, 1988, pages 285
KAKEYA, N. ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
K. YASUDA ET AL., LUNG, vol. 172, 1994, pages 91 - 102
TESTA, B. ET AL.: "Chemistry, Biochemistry and Enzymology", 2003, VCHA AND WILEY-VCH, article "Hydrolysis in Drug and Prodrug Metabolism"
MUNGER ET AL., CELL, vol. 96, 1999, pages 319 - 328
CHOI ET AL., BIOCHEMICA ET BIOPHYSICA ACTA, vol. 1781, 2008, pages 531 - 539
MILLS ET AL., NAT REV. CANCER, vol. 3, 2003, pages 582 - 591
J.P. PRADERE ET AL.: "LPAi receptor activation promotes renal interstitial fibrosis", J. AM. SOC. NEPHROL., vol. 18, 2007, pages 3110 - 3118, XP002682319, doi:10.1681/ASN.2007020196
N. WIEDMAIER ET AL., INT J MEDMICROBIOL, vol. 298, no. 3-4, 2008, pages 231 - 43
A. KANTARCI ET AL., J. PATHOL., vol. 210, 2006, pages 59 - 66
N. WATANABE ET AL.: "Plasma lysophosphatidic acid level and serum autotaxin activity are increased in liver injury in rats in relation to its severity", LIFE SCI., vol. 81, 2007, pages 1009 - 1015, XP022248228, doi:10.1016/j.lfs.2007.08.013
N.WATANABE ET AL., J. CLIN. GASTROENTEROL., vol. 41, 2007, pages 616 - 623
K. KURODA ET AL.: "Phospholipid concentration in lung lavage fluid as biomarker for pulmonary fibrosis", INHAL. TOXICOL., vol. 18, 2006, pages 389 - 393
CHEN ET AL., FEBS LETT., vol. 580, no. 19, 21 August 2006 (2006-08-21), pages 4737 - 45
TAGER ET AL., NATURE MEDICINE, vol. 14, 2008, pages 45 - 54
XU ET AL., AM. J. PATHOLOGY, vol. 174, 2009, pages 1264 - 1279
C.N. INOUE ET AL., CLIN. SCI. (COLCH., vol. 96, 1999, pages 431 - 436
J.P. PRADERE ET AL., BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1781, 2008, pages 547 - 557
J.S. KOH, J. CLIN. INVEST., vol. 102, 1998, pages 716 - 727
M. INOUE ET AL.: "Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling", NAT. MED., vol. 10, 2004, pages 712 - 718, XP002693859, doi:10.1038/nm1060
MOL CANCER THER, vol. 7, no. 10, 2008, pages 3352 - 62
GARDELL ET AL., TRENDS IN MOLECULAR MEDICINE, vol. 12, no. 2, 2006, pages 65 - 75
ISHII ET AL., ANNU. REV. BIOCHEM, vol. 73, 2004, pages 321 - 354
MILLS ET AL., NAT. REV. CANCER, vol. 3, 2003, pages 582 - 591
YAMADA ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 6595 - 6605
YAMADA, CANCER SCI., vol. 99, no. 8, 2008, pages 1603 - 1610
BOUCHARABA ET AL., J. CLIN. INVEST., vol. 114, no. 12, 2004, pages 1714 - 1725
BOUCHARABA ET AL., PROC. NATL. ACAD. SCI., vol. 103, no. 25, 2006, pages 9643 - 9648
MILLS ET AL., NAT. REV. CANCER., vol. 3, 2003, pages 582 - 591
SUTPHEN ET AL., CANCER EPIDEMIOL. BIOMARKERS PREV., vol. 13, 2004, pages 1185 - 1191
J.J.A. CONTOS ET AL., MOL. PHARMACOL., vol. 58, 2000, pages 1188 - 1196
M. AMISHIMA ET AL., AM. J. RESPIR. CRIT. CARE MED., vol. 157, 1998, pages 1907 - 1912
EDIGER ET AL., EUR RESPIR J, vol. 21, 2003, pages 759 - 769
HASHIMOTO ET AL., J PHARMACOL SCI, vol. 100, 2006, pages 82 - 87
MOOLENAAR, CURR. OPIN. CELL BIOL., vol. 7, 1995, pages 203 - 10
J. NEUROCHEM, vol. 61, 1993, pages 340
J. NEUROCHEM., vol. 70, 1998, pages 66
ISHII, I. ET AL., ANNU. REV. BIOCHEM., vol. 73, 2004, pages 321 - 354
OSBORNE, N.STAINIER, D.Y, ANNU. REV. PHYSIOL., vol. 65, 2003, pages 23 - 43
MAGUIRE, J.J. ET AL., TRENDS PHARMACOL. SCI., vol. 26, 2005, pages 448 - 454
SIESS, W., BIOCHIM. BIOPHYS. ACTA, vol. 1582, 2002, pages 204 - 215
ROTHER, E. ET AL., CIRCULATION, vol. 108, 2003, pages 741 - 747
GARDELL ET AL., TRENDS IN MOLECULAR MEDICINE, vol. 12, no. 2, February 2006 (2006-02-01)
BIOCHEM BIOPHYS RES COMMUN., vol. 363, no. 4, 2007, pages 1001 - 8
BIOCHEM BIOPHYS RES COMMUN., vol. 193, no. 2, 1993, pages 497
J. IMUUNOL., vol. 162, 1999, pages 2049
J. UROLOGY, vol. 162, 1999, pages 1779 - 1784
J. UROLOGY, vol. 163, 2000, pages 1027 - 1032
KOTANI ET AL., HUM. MOL. GENET., vol. 17, 2008, pages 1790 - 1797
ZHAO ET AL., MOL. PHARMACOL., vol. 73, no. 2, 2008, pages 587 - 600
SIMON ET AL., J.BIOL. CHEM., vol. 280, no. 15, 2005, pages 14656
GREENE ET AL.: "Protective Groups in Organic Synthesis", 2006, WILEY-INTERSCIENCE
WUTS, P.G.M.: "Greene's Protective Groups in Organic Synthesis", 2014, WILEY
KATRITZKY, A.R. ET AL.: "Comprehensive Organic Functional Group Transformations II", 2004, ELSEVIER SCIENCE INC.
LAROCK, R.C.: "Comprehensive Organic Transformations", 1999, WILEY-VCH
ISHIYAMA, T. ET AL., J. ORG. CHEM., vol. 60, 1995, pages 7508 - 7510
KUMARA SWAMY, K. C., CHEM. REV., vol. 109, 2009, pages 2551 - 2651
QIAN, Y. ET AL., J. MED. CHEM., vol. 55, 2012, pages 7920 - 7939
BOREN, B. C. ET AL., J. AM. CHEM. SOC., vol. 130, 2008, pages 8923 - 8930
RAMASAMY, S. ET AL., ORG. PROCESS RES. DEV., vol. 22, 2018, pages 880 - 887
LINDGREN, B. O., ACTA CHEM. SCAND., vol. 27, 1973, pages 888
ALMOND-THYNNE ET AL., CHEM. SCI., vol. 8, 2017, pages 40 - 62
J. ORG. CHEM., vol. 48, 1983, pages 1643 - 1654
CAN. J. CHEM., vol. 68, 1990, pages 804 - 811
J. ORG. CHEM., vol. 49, 1984, pages 2079 - 2081
Attorney, Agent or Firm:
SUN, Jing G. et al. (US)
Download PDF:
Claims:
What is claimed is:

1. A compound of Formula (I):

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

X1, X2, X3, and X4 are each independently CR6 or N; provided that no more than two of X1, X2, X3, or X4 are N;

Q1, Q2, and Q3 are independently N, O, NR5a, or CR5b, and the dashed circle denotes bonds forming an aromatic ring; provided that at least one of Q1, Q2, and Q3 is not CR5b;

L is independently a covalent bond or Ci-4 alkylene substituted with 0 to 4 R9;

W is independently O, -OCH2- or -CH2O-;

Y1 is independently O or NR7;

Y2 is independently

Y3 is independently the proviso that when

Y1 is O, then Y3 is not OR4;

Y5 independently is O or NH;

alternatively, -Y2-Y3 is R4b; R1 is independently cyano, -C(0)0Ru, -C(0)NR12aR12b,

R2 is each independently halo, cyano, hydroxyl, amino, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy, Ci-4 alkylamino, -(CH2)O-I-(C3-6 cycloalkyl), -(CH2)o-i-phneyl, or Ci-6 alkyl substituted with 0 to 3 Rc;

R3a is independently hydrogen, halo, hydroxyl, or Ci-4 alkyl;

R3b is independently hydrogen, halo, cyano, hydroxyl, amino, Ci-4 alkyl, Ci-4 haloalkyl, Ci-4 alkoxy, Ci-4 haloalkoxy or Ci-6 alkyl substituted with 0 to 2 Ra;

or alternatively, R3a and R3b together, with the carbon atom they are attached to, form a C3-4 carbocyclyl;

R4 is -Li-R4a;

Li is independently a covalent bond or Ci-4 alkylene substituted with 0 to 4 R9;

R4a is independently Ci-io alkyl, Ci-io haloalkyl, Ci-io alkenyl, C3-8 cycloalkyl,

C6-10 aryl, 3 to 8-membered heterocyclyl, 5 to 6-membered heteroaryl; wherein each of the alkyl, alkenyl, alkylene, cycloalkyl, aryl, heterocyclyl, and heteroaryl, by itself or as part of other moiety, is independently substituted with 0 to 3 R10;

R4b is independently C1-10 alkyl, C1-10 haloalkyl, C1-10 alkenyl, C3-8 cycloalkyl, C6-10 aryl, 3 to 8-membered heterocyclyl, 5 to 6-membered heteroaryl; wherein each of the alkyl, alkenyl, alkylene, cycloalkyl, aryl, heterocyclyl, and heteroaryl, by itself or as part of other moiety, is independently substituted with 0 to 3 R10a;

R5a is independently hydrogen, Ci-4 haloalkyl, -(CH2)O-I-(C3-6 cycloalkyl),

-(CH2)o-i-phneyl, or Ci-6 alkyl substituted with 0 to 3 Ra; R5b is independently hydrogen, halo, cyano, hydroxyl, amino, Ci-4 alkoxy, Ci-4 haloalkyl, Ci-4 haloalkoxy, Ci-4 alkylamino, -(CH2)O-I-(C3-6 cycloalkyl), -(CH2)o-i-phneyl, or Ci-6 alkyl substituted with 0 to 3 Rb;

R6 is each independently hydrogen, halo, cyano, hydroxyl, amino,

Ci-4 alkylamino, Ci-4 haloalkyl, Ci-4 alkoxy, Ci-4 haloalkoxy or Ci-6 alkyl substituted with 0 to 1 Rb;

R7 and R8 are each independently hydrogen, Ci-6 alkyl, Ci-4 haloalkyl, C3-6 cycloalkyl or Ci-6 alkyl substituted with 0 to 1 Rc;

R9 is each independently halo, oxo, cyano, hydroxyl, amino, Ci-4 haloalkyl, Ci-4 alkoxy, Ci-4 haloalkoxy, C3-6 cycloalkyl, or Ci-6 alkyl substituted with 0 to 3 Ra;

R10 and R10a are each independently halo, hydroxyl, amino, cyano, C2-6 alkenyl, C2-6 alkynyl, Ci-4 alkylamino, Ci-4 haloalkyl, Ci-4 alkoxy, Ci-4 haloalkoxy, phenyl, or 5 to 6 membered heteroaryl, Ci-6 alkyl substituted with 0 to 3 Rb;

R11, R12a and R12b are each independently hydrogen, Ci-6 alkyl, C3-6 cycloalkyl or benzyl;

Ra is independently halo, cyano, hydroxyl, Ci-4 alkoxy, Ci-4 haloalkyl, or Ci-4 haloalkoxy;

Rb is independently halo, cyano, hydroxyl, amino, Ci-4 alkoxy, Ci-4 haloalkyl, or Ci-4 haloalkoxy;

Re is independently Ci-4 haloalkyl, C3-6 cycloalkyl, or Ci-6 alkyl substituted with 0 to 3 Ra;

m is an integer of 0, 1, or 2; and

q is an integer of 0, 1, or 2.

2 The compound according to claim 1, wherein:

the moiety is independently

* denotes the attachment point to L; and

R5a and R5b are the same as defined in Claim 1.

3. The compound according to claim 1 or 2, wherein:

4. The compound according to any one of claims 1 to 3, wherein L is a covalent bond or C1-2 alkylene.

5. The compound according to any one of claims 1 to 4, wherein: R3a and R3b are independently hydrogen or Ci-4 alkyl;

or alternatively, R3a and R3b together, with the carbon atom they are attached to, form C3-4 cycloalkyl.

6. The compound according to any one of claims 1 to 5, wherein R1 is CO2H.

7. The compound according to any one of claims 1 to 6, wherein:

X1, X2, X3, and X4 are independently CR6; or X1, X2 and X3 are CR6 and X4 is N; or X2 and X3 are CR6 and X1 and X4 are N; or X1 and X2 are CR6 and X3 and X4 are N; and R6 is independently hydrogen, halo, hydroxyl, Ci-6 alkyl, Ci-4 haloalkyl, or Ci-4 alkoxy.

8. The compound according to any one of claims 1 to 7, wherein:

X1, X2, X3, and X4 are independently CR6; or X1, X2 and X3 are CR6 and X4 is N; or X2 and X3 are CR6 and X1 and X4 are N;

the moiety is independently

* denotes the attachment point to L;

L is independently a covalent bond or -CH2-;

W is independently O or -OCH2-;

the moiety is independently -

R2 is each independently halo, cyano, hydroxyl, Ci-4 alkoxy, Ci-4 haloalkyl or Ci-4 haloalkoxy;

R4 is independently C3-6 alkyl, -(CH2)O-I-C3-6 cycloalkyl,

-(CH(CI-2 alkyl))-C3-6 cycloalkyl, -(CH2)o-i-phenyl or -(CH(CI-2 alkyl))-phenyl, wherein each of said cycloalkyl and phenyl is independently substituted with 0 to 3 R10;

R4b is independently 5 to 6-membered heteroaryl substituted with 0 to 2 R10a;

R5a is independently C1-6 alkyl, or -(CH2)O-I-(C3-6 cycloalkyl);

R5b is independently hydrogen, halo, cyano, hydroxyl, Ci-4 alkyl, Ci-4 alkoxy, Ci-4 haloalkyl, C 1-4 haloalkoxy, or -(CH2)O-I-(C3-6 cycloalkyl);

R6 is each independently hydrogen, halo, hydroxyl, C1-6 alkyl, Ci-4 haloalkyl, or Ci-4 alkoxy;

R7 and R8 are each independently hydrogen or C1-2 alkyl;

R10 is each independently halo, cyano, hydroxyl, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, or C1-6 haloalkoxy;

R10a is each independently halo, cyano, hydroxyl, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, -(CH2)o-3-C3-6 cycloalkyl, phenyl or 5 to 6 membered heteroaryl; m is 0 or 1; and

q is 0 or 1.

9. The compound according to any one of claims 1 to 8, wherein:

* denotes the attachment point to L;

L is independently a covalent bond or -CH2-;

W is independently O or -OCH2-;

the moiety is independently -

R3a and R3b are each independently hydrogen or Ci-4 alkyl;

or alternatively, R3a and R3b together, with the carbon atom they are attached to, form C3-4 cycloalkyl;

R4 is independently C3-6 alkyl, -(CH2)O-I-C3-6 cycloalkyl, -(CH2)-phenyl, or -(CH(CH3))-phenyl; wherein wherein each of said alkyl, cycloalkyl and phenyl is independently substituted with 0 to 2 R10;

R b is independently

R6 is independently hydrogen, halo or Ci-4 alkyl;

R7 and R8 are each independently hydrogen or C1-2 alkyl;

R10 is each independently halo or Ci-4 alkyl; and

R10a is independently Ci-4 alkyl, -(CH2)I-3-C3-6 cycloalkyl, phenyl or pyridyl.

10. A compound of Formula (II):

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

X4 is CH or N;

the moiety is independently

* denotes the attachment point to L;

L is independently a covalent bond or -CH2-;

W is independently O or -OCH2-;

R3a and R3b are each independently hydrogen or methyl;

or alternatively, R3a and R3b together, with the carbon atom they are attached to, form cyclopropyl;

R4 is independently C3-6 alkyl, -(CH2)O-I-C3-6 cycloalkyl, -(CH2)-phenyl, or -(CH(CH3))-phenyl; R4b is independently

R6 is independently hydrogen, halo or Ci-4 alkyl;

R7 and R8 are each independently hydrogen or methyl;

R10a is independently Ci-4 alkyl, phenyl or pyridyl; and

m is 0 or 1.

11. The compound according to claim 1, which is selected from any one of Examples 1 to 89, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or solvate thereof.

12. A pharmaceutical composition comprising one or more compounds according to any one of claims 1 to 1 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof; and a pharmaceutically acceptable carrier or diluent.

13. Use of a compound or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1 to 11, or a pharmaceutical composition according to claim 12, for therapy

14. Use of a compound or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1 to 11, or a pharmaceutical composition according to claim 12, for treating a disease, disorder, or condition associated with dysregulation of lysophosphatidic acid receptor 1 (LPA1).

15. The use according to claim 14, wherein the disease, disorder, or condition is related to pathological fibrosis, transplant rejection, cancer, osteoporosis, or inflammatory disorders.

16. The use according to claim 15, wherein the pathological fibrosis is pulmonary, liver, renal, cardiac, dernal, ocular, or pancreatic fibrosis.

17. The use according to claim 14, wherein the disease, disorder, or condition is idiopathic pulmonary fibrosis (IPF), non-alcoholic steatohepatitis (NASH), non alcoholic fatty liver disease (NAFLD), chronic kidney disease, diabetic kidney disease, and systemic sclerosis.

18. The use according to claim 15, wherein the cancer is of the bladder, blood, bone, brain, breast, central nervous system, cervix, colon, endometrium, esophagus, gall bladder, genitalia, genitourinary tract, head, kidney, larynx, liver, lung, muscle tissue, neck, oral or nasal mucosa, ovary, pancreas, prostate, skin, spleen, small intestine, large intestine, stomach, testicle, or thyroid.

19. Use of a compound or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1 to 11, or a pharmaceutical composition according to claim 12, for treating fibrosis in a mammal comprising administering a therapeutically effective amount of the compound or the pharmaceutical composition, to the mammal in need thereof.

20. The use according to claim 19, wherein the fibrosis is idiopathic pulmonary

fibrosis (IPF), nonalcoholic steatohepatitis (NASH), chronic kidney disease, diabetic kidney disease, and systemic sclerosis.

21. Use of a compound or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof according to any one of claims 1 to 11, or a pharmaceutical composition according to claim 12, for treating lung fibrosis (idiopathic pulmonary fibrosis), asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis (non alcoholic steatohepatitis), skin fibrosis, fibrosis of the gut, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud’s phenomenon, or neuropathic pain in a mammal ccomprising administering a therapeutically effective amount of the compound or the pharmaceutical composition, to the mammal in need thereof.

Description:
CYCLOPENTYL ACIDS AS LPA ANTAGONISTS

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the priority benefit of U.S. Provisional Application No. 62/732,590, filed September 18, 2018; the entire content of which is herein incorporated by reference.

FIELD OF THE INVENTION

The present invention relates to novel substituted cyclopentyl acid compounds, compositions containing them, and methods of using them, for example, for the treatment of disorders associated with one or more of the lysophosphatidic acid (LPA) receptors.

BACKGROUND OF THE INVENTION

Lysophospholipids are membrane-derived bioactive lipid mediators, of which one of the most medically important is lysophosphatidic acid (LPA). LPA is not a single molecular entity but a collection of endogenous structural variants with fatty acids of varied lengths and degrees of saturation (Fujiwara et al., JBiol. Chem ., 2005, 280 , 35038- 35050). The structural backbone of the LPAs is derived from glycerol -based

phospholipids such as phosphatidylcholine (PC) or phosphatidic acid (PA).

The LPAs are bioactive lipids (signaling lipids) that regulate various cellular signaling pathways by binding to the same class of 7-transmembrane domain G protein- coupled (GPCR) receptors (Chun, T, Hla, T., Spiegel, S., Moolenaar, W., Editors, Lysophospholipid Receptors: Signaling and Biochemistry, 2013, Wiley; ISBN: 978-0- 470-56905-4 & Zhao, Y. et al, Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Of Lipids, 2013, 1831, 86-92). The currently known LPA receptors are designated as LPAi, LPA2, LPA3, LPA 4 , LPAs and LPA 6 (Choi, J. W., Annu. Rev. Pharmacol. Toxicol., 2010, 50, 157-186; Kihara, Y., et al, Br. J. Pharmacol., 2014, 171, 3575-3594).

The LPAs have long been known as precursors of phospholipid biosynthesis in both eukaryotic and prokaryotic cells, but the LPAs have emerged only recently as signaling molecules that are rapidly produced and released by activated cells, notably platelets, to influence target cells by acting on specific cell-surface receptors (see, e.g, Moolenaar et al., BioEssays, 2004, 26, 870-881, and van Leewen et al., Biochem. Soc. Trans., 2003, 37, 1209-1212). Besides being synthesized and processed to more complex phospholipids in the endoplasmic reticulum, LPAs can be generated through the hydrolysis of pre-existing phospholipids following cell activation; for example, the sn-2 position is commonly missing a fatty acid residue due to deacylation, leaving only the sn- 1 hydroxyl esterified to a fatty acid. Moreover, a key enzyme in the production of LPA, autotaxin (lysoPLD/NPP2), may be the product of an oncogene, as many tumor types up- regulate autotaxin (Brindley, D., J. Cell Biochem. 2004, 92, 900-12). The concentrations of LPAs in human plasma & serum as well as human bronchoalveolar lavage fluid (BALF) have been reported, including determinations made using sensitive and specific LC/MS & LC/MS/MS procedures (Baker et al. Anal. Biochem., 2001, 292, 287-295; Onorato et al., J. Lipid Res., 2014, 55, 1784-1796).

LPA influences a wide range of biological responses, ranging from induction of cell proliferation, stimulation of cell migration and neurite retraction, gap junction closure, and even slime mold chemotaxis (Goetzl, et al., Scientific World J., 2002, 2, 324- 338; Chun, J., Hla, T., Spiegel, S., Moolenaar, W., Editors, Lysophospholipid Receptors: Signaling and Biochemistry, 2013, Wiley; ISBN: 978-0-470-56905-4). The body of knowledge about the biology of LPA continues to grow as more and more cellular systems are tested for LPA responsiveness. For instance, it is now known that, in addition to stimulating cell growth and proliferation, LPAs promote cellular tension and cell- surface fibronectin binding, which are important events in wound repair and regeneration (Moolenaar et al., BioEssays, 2004, 26, 870-881). Recently, anti-apoptotic activity has also been ascribed to LPA, and it has recently been reported that PPARy is a

receptor/target for LPA (Simon et al., J. Biol. Chem., 2005, 280, 14656-14662).

Fibrosis is the result of an uncontrolled tissue healing process leading to excessive accumulation and insufficient resorption of extracellular matrix (ECM) which ultimately results in end-organ failure (Rockey, D. C., et al., New Engl. J. Med., 2015, 372, 1138- 1149). Recently it was reported that the LPA1 receptor was over-expressed in idiopathic pulmonary fibrosis (IPF) patients. LPAi receptor knockout mice were also protected from bleomycin-induced lung fibrosis (Tager et al., Nature Med., 2008, 14, 45-54). LPA pathway inhibitors (e.g. an LPAi antagonist) were recently shown to be chemopreventive anti-fibrotic agents in the treatment of hepatocellular carcinoma in a rat model (Nakagawa et al., Cancer Cell, 2016, 30, 879-890). Thus, antagonizing the LPAi receptor may be useful for the treatment of fibrosis such as pulmonary fibrosis, hepatic fibrosis, renal fibrosis, arterial fibrosis and systemic sclerosis, and thus the diseases that result from fibrosis (pulmonary fibrosis-idiopathic Pulmonary Fibrosis [IPF], hepatic fibrosis-Non-alcoholic Steatohepatitis [NASH], renal fibrosis-diabetic nephropathy, systemic sclerosis-scleroderma, etc.).

SUMMARY OF THE INVENTION

The present invention provides novel cyclopentyl acid compounds including stereoisomers, tautomers, pharmaceutically acceptable salts or solvates thereof, which are useful as antagonists against one or more of the lysophosphatidic acid (LPA) receptors, especially the LPA1 receptor.

The present invention also provides processes and intermediates for making the compounds of the present invention.

The present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts or solvates thereof.

The compounds of the invention may be used in the treatment of conditions in which LPA plays a role.

The compounds of the present invention may be used in therapy.

The compounds of the present invention may be used for the manufacture of a medicament for the treatment of a condition in which inhibition of the physiological activity of LPA is useful, such as diseases in which an LPA receptor participates, is involved in the etiology or pathology of the disease, or is otherwise associated with at least one symptom of the disease.

In another aspect, the present invention is directed to a method of treating fibrosis of organs (liver, kidney, lung, heart and the like as well as skin), liver diseases (acute hepatitis, chronic hepatitis, liver fibrosis, liver cirrhosis, portal hypertension, regenerative failure, non-alcoholic steatohepatitis (NASH), liver hypofunction, hepatic blood flow disorder, and the like), cell proliferative disease [cancer (solid tumor, solid tumor metastasis, vascular fibroma, myeloma, multiple myeloma, Kaposi's sarcoma, leukemia, chronic lymphocytic leukemia (CLL) and the like) and invasive metastasis of cancer cell, and the like], inflammatory disease (psoriasis, nephropathy, pneumonia and the like), gastrointestinal tract disease (irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), abnormal pancreatic secretion, and the like), renal disease, urinary tract- associated disease (benign prostatic hyperplasia or symptoms associated with neuropathic bladder disease, spinal cord tumor, hernia of intervertebral disk, spinal canal stenosis, symptoms derived from diabetes, lower urinary tract disease (obstruction of lower urinary tract, and the like), inflammatory disease of lower urinary tract, dysuria, frequent urination, and the like), pancreas disease, abnormal angiogenesis-associated disease (arterial obstruction and the like), scleroderma, brain-associated disease (cerebral infarction, cerebral hemorrhage, and the like), neuropathic pain, peripheral neuropathy, and the like, ocular disease (age-related macular degeneration (AMD), diabetic retinopathy, proliferative vitreoretinopathy (PVR), cicatricial pemphigoid, glaucoma filtration surgery scarring, and the like).

In another aspect, the present invention is directed to a method of treating diseases, disorders, or conditions in which activation of at least one LPA receptor by LPA contributes to the symptomology or progression of the disease, disorder or condition. These diseases, disorders, or conditions may arise from one or more of a genetic, iatrogenic, immunological, infectious, metabolic, oncological, toxic, surgical, and/or traumatic etiology.

In another aspect, the present invention is directed to a method of treating renal fibrosis, pulmonary fibrosis, hepatic fibrosis, arterial fibrosis and systemic sclerosis comprising administering to a patient in need of such treatment a compound of the present invention as described above.

In one aspect, the present invention provides methods, compounds,

pharmaceutical compositions, and medicaments described herein that comprise antagonists of LPA receptors, especially antagonists of LPA1.

The compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more, preferably one to two other agent(s).

These and other features of the invention will be set forth in expanded form as the disclosure continues. DETAILED DESCRIPTION OF THE INVENTION

I. COMPOUNDS OF THE INVENTION

In a lst aspect, the present invention provides, inter alia , a compound of Formula

(I):

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

X 1 , X 2 , X 3 , and X 4 are each independently CR 6 or N; provided that no more than two of X 1 , X 2 , X 3 , or X 4 are N;

Q 1 , Q 2 , and Q 3 are independently N, O, NR 5a , or CR 5b , and the dashed circle denotes bonds forming an aromatic ring; provided that at least one of Q 1 , Q 2 , and Q 3 is not CR 5b ;

L is independently a covalent bond or Ci-4 alkylene substituted with 0 to 4 R 9 ;

W is independently O, -OCH2- or -CH2O-;

Y 1 is independently O or NR 7 ;

Y 2 is independently

Y 3 is independently the proviso that when Y 1 is O, then Y 3 is not OR 4 ; Y 5 independently is O or NH;

alternatively, -Y 2 -Y 3 is R 4b ;

R 1 is independently cyano, -C(0)0R u , -C(0)NR 12a R 12b ,

R 2 is each independently halo, cyano, hydroxyl, amino, Ci-4 alkoxy, Ci-4 haloalkyl,

Ci-4 haloalkoxy, Ci-4 alkylamino, -(CH2)O-I-(C3-6 cycloalkyl), -(CH2)o-i-phneyl, or Ci-6 alkyl substituted with 0 to 3 R c ;

R 3a is independently hydrogen, halo, hydroxyl, or Ci-4 alkyl;

R 3b is independently hydrogen, halo, cyano, hydroxyl, amino, Ci-4 alkyl, Ci-4 haloalkyl, Ci-4 alkoxy, Ci-4 haloalkoxy or Ci-6 alkyl substituted with 0 to 2 R a ;

or alternatively, R 3a and R 3b together, with the carbon atom they are attached to, form a C3-4 carbocyclyl;

R 4 is -Li-R 4a ;

Li is independently a covalent bond or Ci-4 alkylene substituted with 0 to 4 R 9 ; R 4a is independently C1-10 alkyl, C1-10 haloalkyl, C1-10 alkenyl, C3-8 cycloalkyl,

C6-10 aryl, 3 to 8-membered heterocyclyl, 5 to 6-membered heteroaryl; wherein each of the alkyl, alkenyl, alkylene, cycloalkyl, aryl, heterocyclyl, and heteroaryl, by itself or as part of other moiety, is independently substituted with 0 to 3 R 10 ;

R 4b is independently C1-10 alkyl, C1-10 haloalkyl, C1-10 alkenyl, C3-8 cycloalkyl, C6-10 aryl, 3 to 8-membered heterocyclyl, 5 to 6-membered heteroaryl; wherein each of the alkyl, alkenyl, alkylene, cycloalkyl, aryl, heterocyclyl, and heteroaryl, by itself or as part of other moiety, is independently substituted with 0 to 3 R 10a ;

R 5a is independently hydrogen, C 1-4 haloalkyl, -(CH2)O-I-(C3-6 cycloalkyl),

-(CH2)o-i-phneyl, or Ci-6 alkyl substituted with 0 to 3 R a ; R 5b is independently hydrogen, halo, cyano, hydroxyl, amino, Ci- 4 alkoxy, Ci-4 haloalkyl, Ci- 4 haloalkoxy, Ci- 4 alkylamino, -(CH2)O-I-(C3-6 cycloalkyl), -(CH2)o-i-phneyl, or Ci-6 alkyl substituted with 0 to 3 R b ;

R 6 is each independently hydrogen, halo, cyano, hydroxyl, amino,

Ci-4 alkylamino, Ci- 4 haloalkyl, Ci- 4 alkoxy, Ci- 4 haloalkoxy or Ci-6 alkyl substituted with 0 to 1 R b ;

R 7 and R 8 are each independently hydrogen, Ci-6 alkyl, Ci- 4 haloalkyl, C3-6 cycloalkyl or Ci-6 alkyl substituted with 0 to 1 R c ;

R 9 is each independently halo, oxo, cyano, hydroxyl, amino, Ci- 4 haloalkyl, Ci- 4 alkoxy, Ci- 4 haloalkoxy, C3-6 cycloalkyl, or Ci-6 alkyl substituted with 0 to 3 R a ;

R 10 and R 10a are each independently halo, hydroxyl, amino, cyano, C2-6 alkenyl, C2-6 alkynyl, Ci- 4 alkylamino, Ci- 4 haloalkyl, Ci- 4 alkoxy, Ci- 4 haloalkoxy, phenyl, or 5 to 6 membered heteroaryl, Ci-6 alkyl substituted with 0 to 3 R b ;

R 11 , R 12a and R 12b are each independently hydrogen, Ci-6 alkyl, C3-6 cycloalkyl or benzyl;

R a is independently halo, cyano, hydroxyl, Ci- 4 alkoxy, Ci- 4 haloalkyl, or Ci- 4 haloalkoxy;

R b is independently halo, cyano, hydroxyl, amino, Ci- 4 alkoxy, Ci- 4 haloalkyl, or Ci-4 haloalkoxy;

R e is independently Ci- 4 haloalkyl, C3-6 cycloalkyl, or Ci-6 alkyl substituted with 0 to 3 R a ;

m is an integer of 0, 1, or 2; and

q is an integer of 0, 1, or 2.

In one embodiment of Formula (I), within the scope of the lst aspect, wherein X 1 , X 2 , X 3 , and X 4 are CR 6 ; where R 6 is hydrogen halo or Ci- 4 alkyl, e.g., methyl.

In another aspect, within the scope of any of the lst aspect, wherein X 1 , X 2 , X 3 , and X 4 are independently CH or CR 6a ; or one of X 1 , X 2 , X 3 , and X 4 is N, and the remaining ones are CH or CR 6a ; or two of X 1 , X 2 , X 3 , and X 4 are N, and the remaining ones are CH or CR 6a ; and R 6a is independently halo, hydroxyl, Ci-6 alkyl, C i- 4 haloalkyl, or C i-4 alkoxy. In a 2nd aspect, within the scope of the lst aspect, wherein:

the moiety is independently

* denotes the attachment point to L; and

R 5a and R 5b are the same as defined in Claim 1.

In a 3rd aspect, within the scope of the lst or 2nd aspect, wherein: i A ^ y ^ v 3

the moiety is independently -OR 4b , -NR 7 R 4b

In a 4th aspect, within the scope of any of the lst to 3rd aspects, wherein L is a covalent bond or C1-2 alkylene.

In one embodiment of Formula (la) or (lb), within the scope of any of the lst to 3rd aspects, wherein L is a covalent bond or -CH2-.

In a 5th aspect, within the scope of any of the 1 st to 4th aspects, wherein R 3a and R 3b are independently hydrogen or C1-4 alkyl;

or alternatively, R 3a and R 3b together, with the carbon atom they are attached to, form C3-4 cycloalkyl.

In a 6th aspect, within the scope of any of the lst to 5th aspects, wherein R 1 is

CO2H.

In a 7th aspect, within the scope of any of the lst to 6th aspects, wherein:

X 1 , X 2 , X 3 , and X 4 are independently CR 6 ; or X 1 , X 2 and X 3 are CR 6 and X 4 is N; or X 2 and X 3 are CR 6 and X 1 and X 4 are N; or X 1 and X 2 are CR 6 and X 3 and X 4 are N; and R 6 is independently hydrogen, halo, hydroxyl, C 1-6 alkyl, C1-4 haloalkyl, or C 1-4 alkoxy.

In an 8th aspect, within the scope of any of the lst to 7th aspects, wherein:

X 1 , X 2 , X 3 , and X 4 are independently CR 6 ; or X 1 , X 2 and X 3 are CR 6 and X 4 is N; or X 2 and X 3 are CR 6 and X 1 and X 4 are N;

the moiety is independently

* denotes the attachment point to L;

L is independently a covalent bond or -CH2-; W is independently O or -OCH2-;

R 2 is each independently halo, cyano, hydroxyl, C 1-4 alkoxy, C1-4 haloalkyl or

Ci-4 haloalkoxy;

R 4 is independently C3-6 alkyl, -(CH2)O-I-C3- 6 cycloalkyl,

-(CH(CI-2 alkyl))-C 3 - 6 cycloalkyl, -(CH2)o-i-phenyl or -(CH(CI-2 alkyl))-phenyl, wherein each of said cycloalkyl and phenyl is independently substituted with 0 to 3 R 10 ;

R 4b is independently 5 to 6-membered heteroaryl substituted with 0 to 2 R 10a ;

R 5a is independently C1-6 alkyl, or -(CH2)O-I-(C 3 - 6 cycloalkyl);

R 5b is independently hydrogen, halo, cyano, hydroxyl, Ci-4 alkyl, Ci-4 alkoxy,

Ci-4 haloalkyl, C 1-4 haloalkoxy, or -(CH2)O-I-(C3-6 cycloalkyl);

R 6 is each independently hydrogen, halo, hydroxyl, C 1-6 alkyl, Ci-4 haloalkyl, or C1-4 alkoxy;

R 7 and R 8 are each independently hydrogen or C1-2 alkyl;

R 10 is each independently halo, cyano, hydroxyl, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, or C1-6 haloalkoxy;

R 10a is each independently halo, cyano, hydroxyl, C1-6 alkyl, C1-6 haloalkyl, C1-6 alkoxy, C1-6 haloalkoxy, -(CH2)o- 3 -C 3 - 6 cycloalkyl, phenyl or 5 to 6 membered heteroaryl; m is 0 or 1; and

q is 0 or 1.

In a 9th aspect, within the scope of any one of the lst to 8th aspects, wherein:

* denotes the attachment point to L;

L is independently a covalent bond or -CH2-;

W is independently O or -OCH2-;

the moiety is independently -

R 3a and R 3b are each independently hydrogen or Ci-4 alkyl;

or alternatively, R 3a and R 3b together, with the carbon atom they are attached to, form C3-4 cycloalkyl;

R 4 is independently C3-6 alkyl, -(CH2)O-I-C 3 - 6 cycloalkyl, -(CH2)-phenyl, or -(CH(CH 3 ))-phenyl; wherein wherein each of said alkyl, cycloalkyl and phenyl is independently substituted with 0 to 2 R 10 ;

R b is independently

R 6 is independently hydrogen, halo or Ci-4 alkyl;

R 7 and R 8 are each independently hydrogen or C1-2 alkyl;

R 10 is each independently halo or Ci-4 alkyl; and

R 10a is independently Ci-4 alkyl, -(CH2)I- 3 -C 3 - 6 cycloalkyl, phenyl or pyridyl. In another aspect, within the scope of any one of the 1 st to 8th aspects, wherein the compound is of Formula (la):

or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof.

In a lOth aspect, the present invention provides a compound of Formula (II):

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

X 4 is CH or N; the moiety is independently

* denotes the attachment point to L;

L is independently a covalent bond or -CH2-;

W is independently O or -OCH2-; the A v ,- Y Y 3 moiety is independently -NR 7 R 4b

R 3a and R 3b are each independently hydrogen or methyl;

or alternatively, R 3a and R 3b together, with the carbon atom they are attached to, form cyclopropyl;

R 4 is independently C3-6 alkyl, -(CH2)O-I-C 3 - 6 cycloalkyl, -(CH2)-phenyl, or -(CH(CH 3 ))-phenyl;

R 4b is independently

R 6 is independently hydrogen, halo or C1-4 alkyl;

R 7 and R 8 are each independently hydrogen or methyl;

R 10a is independently C1-4 alkyl, phenyl or pyridyl; and

m is 0 or 1.

In another aspect, within the scope of the lOth aspect, wherein the compound is of Formula (Ha):

or a stereoisomer, tautomer, or pharmaceutically acceptable salt or solvate thereof.

In one embodiment of the present invention, the compound is selected from any one of the Examples as described in the specification, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or solvate thereof.

In one embodiment, the compounds of the present invention have hLPAl ECso values < 5000 nM, using the LPA1 functional antagonist assay; in another embodiment, the compounds of the present invention have hLPAl ECso values < 1000 nM; in another embodiment, the compounds of the present invention have hLPAl ECso values < 500 nM; in another embodiment, the compounds of the present invention have hLPAl ECso values < 200 nM; in another embodiment, the compounds of the present invention have hLPAl EC50 values < 100 nM; in another embodiment, the compounds of the present invention have hLPAl EC50 values < 50 nM.

II. OTHER EMBODIMENTS OF THE INVENTION

In some embodiments, the compound of the present invention, or a

pharmaceutically acceptable salt thereof, is an antagonist of at least one LPA receptor. In some embodiments, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is an antagonist of LPAi. In some embodiments, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is an antagonist of LPA2. In some embodiments, the compound of the present invention, or a

pharmaceutically acceptable salt thereof, is an antagonist of LPA3. In some embodiments, presented herein are compounds selected from active metabolites, tautomers, pharmaceutically acceptable salts or solvates of a compound of the present invention.

In another embodiment, the present invention provides a composition comprising at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

In another embodiment, the present invention provides a pharmaceutical composition, comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

In another embodiment, the present invention provides a process for making a compound of the present invention.

In another embodiment, the present invention provides an intermediate for making a compound of the present invention.

In another embodiment, the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s).

In another embodiment, the present invention provides a method for the treatment of a condition associated with LPA receptor mediated fibrosis, comprising administering to a patient in need of such treatment a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof. As used herein, the term "patient" encompasses all mammalian species.

In another embodiment, the present invention provides a method of treating a disease, disorder, or condition associated with dysregulation of lysophosphatidic acid receptor 1 (LPA1) in a patient in need thereof, comprising administering a therapeutically effective amount of a compound of the present invention, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or solvate thereof, to the patient. In one

embodiment of the method, the disease, disorder, or condition is related to pathological fibrosis, transplant rejection, cancer, osteoporosis, or inflammatory disorders. In one embodiment of the method, the pathological fibrosis is pulmonary, liver, renal, cardiac, dernal, ocular, or pancreatic fibrosis. In one embodiment of the method, the disease, disorder, or condition is idiopathic pulmonary fibrosis (IPF), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), chronic kidney disease, diabetic kidney disease, and systemic sclerosis. In one embodiment of the method, the cancer is of the bladder, blood, bone, brain, breast, central nervous system, cervix, colon, endometrium, esophagus, gall bladder, genitalia, genitourinary tract, head, kidney, larynx, liver, lung, muscle tissue, neck, oral or nasal mucosa, ovary, pancreas, prostate, skin, spleen, small intestine, large intestine, stomach, testicle, or thyroid.

In another embodiment, the present invention provides a method of treating fibrosis in a mammal comprising administering a therapeutically effective amount of a compound of the present invention, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or solvate thereof, to the mammal in need thereof. In one embodiment of the method, the fibrosis is idiopathic pulmonary fibrosis (IPF), nonalcoholic

steatohepatitis (NASH), chronic kidney disease, diabetic kidney disease, and systemic sclerosis.

In another embodiment, the present invention provides a method of treating lung fibrosis (idiopathic pulmonary fibrosis), asthma, chronic obstructive pulmonary disease (COPD), renal fibrosis, acute kidney injury, chronic kidney disease, liver fibrosis (non alcoholic steatohepatitis), skin fibrosis, fibrosis of the gut, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, glioblastoma, bone cancer, colon cancer, bowel cancer, head and neck cancer, melanoma, multiple myeloma, chronic lymphocytic leukemia, cancer pain, tumor metastasis, transplant organ rejection, scleroderma, ocular fibrosis, age related macular degeneration (AMD), diabetic retinopathy, collagen vascular disease, atherosclerosis, Raynaud’s phenomenon, or neuropathic pain in a mammal ccomprising administering a therapeutically effective amount of a compound of the present invention, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt or solvate thereof, to the mammal in need thereof.

As used herein, "treating" or "treatment" cover the treatment of a disease-state in a mammal, particularly in a human, and include: (a) inhibiting the disease-state, i.e., arresting it development; and/or (b) relieving the disease-state, i.e., causing regression of the disease state. As used herein, "treating" or "treatment" also include the protective treatment of a disease state to reduce and/or minimize the risk and/or reduction in the risk of recurrence of a disease state by administering to a patient a therapeutically effective amount of at least one of the compounds of the present invention or a or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof. Patients may be selected for such protective therapy based on factors that are known to increase risk of suffering a clinical disease state compared to the general population. For protective treatment, conditions of the clinical disease state may or may not be presented yet. The protective treatment can be divided into (a) primary prophylaxis and (b) secondary prophylaxis. Primary prophylaxis is defined as treatment to reduce or minimize the risk of a disease state in a patient that has not yet presented with a clinical disease state, whereas secondary prophylaxis is defined as minimizing or reducing the risk of a recurrence or second occurrence of the same or similar clinical disease state.

The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. This invention encompasses all

combinations of preferred aspects of the invention noted herein. It is understood that any and all embodiments of the present invention may be taken in conjunction with any other embodiment or embodiments to describe additional embodiments. It is also to be understood that each individual element of the embodiments is its own independent embodiment. Furthermore, any element of an embodiment is meant to be combined with any and all other elements from any embodiment to describe an additional embodiment.

III. CHEMISTRY

Throughout the specification and the appended claims, a given chemical formula or name shall encompass all stereo and optical isomers and racemates thereof where such isomers exist. Unless otherwise indicated, all chiral (enantiomeric and diastereomeric) and racemic forms are within the scope of the invention. Many geometric isomers of C=C double bonds, C=N double bonds, ring systems, and the like can also be present in the compounds, and all such stable isomers are contemplated in the present invention. C/s and trans- (or E- and Z-) geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.

The present compounds can be isolated in optically active or racemic forms. Optically active forms may be prepared by resolution of racemic forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by chromatography or fractional crystallization. Depending on the process conditions the end products of the present invention are obtained either in free (neutral) or salt form. Both the free form and the salts of these end products are within the scope of the invention. If so desired, one form of a compound may be converted into another form. A free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present invention may be separated into the individual isomers. Compounds of the present invention, free form and salts thereof, may exist in multiple tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention.

The term "stereoisomer" refers to isomers of identical constitution that differ in the arrangement of their atoms in space. Enantiomers and diastereomers are examples of stereoisomers. The term "enantiomer" refers to one of a pair of molecular species that are mirror images of each other and are not superimposable. The term "diastereomer" refers to stereoisomers that are not mirror images. The term "racemate" or "racemic mixture" refers to a composition composed of equimolar quantities of two enantiomeric species, wherein the composition is devoid of optical activity.

The symbols "R" and "S" represent the configuration of substituents around a chiral carbon atom(s). The isomeric descriptors "R" and "S" are used as described herein for indicating atom configuration(s) relative to a core molecule and are intended to be used as defined in the literature (IUPAC Recommendations 1996, Pure and Applied Chemistry , 68:2193-2222 (1996)).

The term "chiral" refers to the structural characteristic of a molecule that makes it impossible to superimpose it on its mirror image. The term "homochiral" refers to a state of enantiomeric purity. The term "optical activity" refers to the degree to which a homochiral molecule or nonracemic mixture of chiral molecules rotates a plane of polarized light.

As used herein, the term "alkyl" or "alkylene" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. While“alkyl” denotes a monovalent saturated aliphatic radical (such as ethyl),“alkylene” denotes a bivalent saturated aliphatic radical (such as ethylene). For example, "Ci to Cio alkyl" or "Ci-io alkyl" is intended to include Ci, C2,

C3, C 4 , C5, Ce, Ci, Cs, C9, and Cio alkyl groups. "Ci to Cio alkylene" or "C1-10 alkylene", is intended to include Ci, C2, C3, C 4 , C5, C6, C7, Cs, C9, and Cio alkylene groups.

Additionally, for example, "Ci to C6 alkyl" or "C1-6 alkyl" denotes alkyl having 1 to 6 carbon atoms; and "Ci to C6 alkylene" or " C1-6 alkylene" denotes alkylene having 1 to 6 carbon atoms; and "Ci to C 4 alkyl" or "C1-4 alkyl" denotes alkyl having 1 to 4 carbon atoms; and "Ci to C 4 alkylene" or "Ci- 4 alkylene" denotes alkylene having 1 to 4 carbon atoms. Alkyl group can be unsubstituted or substituted with at least one hydrogen being replaced by another chemical group. Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl ( e.g ., n-propyl and isopropyl), butyl (e.g. , n-butyl, isobutyl, /-butyl), and pentyl (e.g., n-pentyl, isopentyl, neopentyl). When "Co alkyl" or "Co alkylene" is used, it is intended to denote a direct bond. Furthermore, the term “alkyl”, by itself or as part of another group, such as alkylamino, haloalkyl, hydroxyalkyl, aminoalkyl, alkoxy, alkoxyalkyl, haloalkoxyalkyl, and haloalkoxy, can be an alkyl having 1 to 4 carbon atoms, or 1 to 6 carbon atoms, or 1 to 10 carbon atoms.

“Heteroalkyl” refers to an alkyl group where one or more carbon atoms have been replaced with a heteroatom, such as, O, N, or S. For example, if the carbon atom of the alkyl group which is attached to the parent molecule is replaced with a heteroatom (e.g, O, N, or S) the resulting heteroalkyl groups are, respectively, an alkoxy group (e.g, -OCH3, etc.), an alkylamino (e.g, -NHCH3, -N(CH3)2, etc.), or a thioalkyl group (e.g, -SCH3). If a non terminal carbon atom of the alkyl group which is not attached to the parent molecule is replaced with a heteroatom (e.g, O, N, or S) and the resulting heteroalkyl groups are, respectively, an alkyl ether (e.g, -CH2CH2-O-CH3, etc.), an alkylaminoalkyl

(e.g, -CH2NHCH3, -CH 2 N(CH3)2, etc.), or a thioalkyl ether (e.g.,-CH2-S-CH3). If a terminal carbon atom of the alkyl group is replaced with a heteroatom (e.g, O, N, or S), the resulting heteroalkyl groups are, respectively, a hydroxyalkyl group (e.g, -CH2CH2-OH), an aminoalkyl group (e.g, -CH2NH2), or an alkyl thiol group (e.g, -CH2CH2-SH). A heteroalkyl group can have, for example, 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. A C1-C6 heteroalkyl group means a heteroalkyl group having 1 to 6 carbon atoms. "Alkenyl" or "alkenylene" is intended to include hydrocarbon chains of either straight or branched configuration having the specified number of carbon atoms and one or more, preferably one to two, carbon-carbon double bonds that may occur in any stable point along the chain. For example, "C2 to C6 alkenyl" or "C2-6 alkenyl" (or alkenylene), is intended to include C2, C3, C 4 , C5, and C6 alkenyl groups. Examples of alkenyl include, but are not limited to, ethenyl, l-propenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 2-methyl-2- propenyl, and 4-methyl-3-pentenyl.

"Alkynyl" or "alkynylene" is intended to include hydrocarbon chains of either straight or branched configuration having one or more, preferably one to three, carbon- carbon triple bonds that may occur in any stable point along the chain. For example, "C2 to C6 alkynyl" or "C2-6 alkynyl" (or alkynylene), is intended to include C2, C3, C 4 , C5, and C6 alkynyl groups; such as ethynyl, propynyl, butynyl, pentynyl, and hexynyl.

As used herein,“arylalkyl” (a.k.a. aralkyl),“heteroarylalkyl”“carbocyclylalkyl” or“heterocyclylalkyl” refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is replaced with an aryl, heteroaryl, carbocyclyl, or heterocyclyl radical, respectively. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-l-yl, naphthylmethyl, 2- naphthylethan-l-yl, naphthobenzyl, 2-naphthophenylethan-l-yl and the like. The arylalkyl, heteroarylalkyl, carbocyclylalkyl, or heterocyclylalkyl group can comprise 4 to 20 carbon atoms and 0 to 5 heteroatoms, e.g ., the alkyl moiety may contain 1 to 6 carbon atoms.

The term "benzyl", as used herein, refers to a methyl group on which one of the hydrogen atoms is replaced by a phenyl group, wherein said phenyl group may optionally be substituted with 1 to 5 groups, preferably 1 to 3 groups, OH, OCH3, Cl, F, Br, I, CN, NO2, NH2, N(CH 2 )H, N(CH 3 ) 2 , CF3, OCF3, C(=0)CH 3 , SCH3, S(=0)CH 3 , S(=0) 2 CH 3 , CH3, CH2CH3, CO2H, and CO2CH3. “Benzyl” can also be represented by formula“Bn”.

The term "alkoxy" or "alkyloxy" refers to an -O-alkyl group. "Ci to C6 alkoxy" or "C1-6 alkoxy" (or alkyloxy), is intended to include Ci, C2, C3, C 4 , C5, and C6 alkoxy groups. Example alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (e.g, n-propoxy and isopropoxy), and /-butoxy. Similarly, "alkylthio" or "thioalkoxy" represents an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example, methyl-S- and ethyl-S-.

The term“alkanoyl” or“alkylcarbonyl” as used herein alone or as part of another group refers to alkyl linked to a carbonyl group. For example, alkylcarbonyl may be represented by alkyl-C(O)-. "Ci to C6 alkylcarbonyl" (or alkylcarbonyl), is intended to include Ci, C2, C3, C 4 , C5, and C6 alkyl-C(O)- groups.

The term“alkylsulfonyl” or“sulfonamide” as used herein alone or as part of another group refers to alkyl or amino linked to a sulfonyl group. For example, alkylsulfonyl may be represented by -S(0)2R’, while sulfonamide may be represented by -S(0)2NR c R d . R’ is Ci to C6 alkyl; and R c and R d are the same as defined below for “amino”.

The term“carbamate” as used herein alone or as part of another group refers to oxygen linked to an amido group. For example, carbamate may be represented by N(R c R d )-C(0)-0-, and R c and R d are the same as defined below for“amino”.

The term“amido” as used herein alone or as part of another group refers to amino linked to a carbonyl group. For example, amido may be represented by N(R c R d )-C(0)-, and R c and R d are the same as defined below for“amino”.

The term“amino” is defined as -NR cl R c2 , wherein R cl and R c2 are independently

H or

Ci- 6 alkyl; or alternatively, R cl and R c2 , taken together with the atoms to which they are attached, form a 3- to 8-membered heterocyclic ring which is optionally substituted with one or more group selected from halo, cyano, hydroxyl, amino, oxo, C1- 6 alkyl, alkoxy, and aminoalkyl. When R cl or R c2 (or both of them) is C1- 6 alkyl, the amino group can also be referred to as alkylamino. Examples of alkylamino group include, without limitation, methylamino, ethylamino, propylamino, isopropylamino and the like. In one embodiment, amino is -NFh.

The term“aminoalkyl” refers to an alkyl group on which one of the hydrogen atoms is replaced by an amino group. For example, aminoalkyl may be represented by N(R cl R c2 )-alkylene-. "Ci to C6 1 ' or“C1- 6 aminoalkyl" (or aminoalkyl), is intended to include Ci, C2, C 3 , C 4 , C5, and C6 aminoalkyl groups.

The term“halogen” or“halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine, with chlorine or fluorine being preferred. "Haloalkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with one or more halogens. "Ci to C6 haloalkyl" or "Ci- 6 haloalkyl" (or haloalkyl), is intended to include Ci, C2, C3, C 4 , C5, and C6 haloalkyl groups. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl,

trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl,

heptafluoropropyl, and heptachloropropyl. Examples of haloalkyl also include

"fluoroalkyl" that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more fluorine atoms. The term“polyhaloalkyl” as used herein refers to an “alkyl” group as defined above which includes from 2 to 9, preferably from 2 to 5, halo substituents, such as F or Cl, preferably F, such as polyfluoroalkyl, for example, CF3CH2, CF3 or CF3CF2CH2.

"Haloalkoxy" or "haloalkyloxy" represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. For example, " Ci to C6 haloalkoxy" or "C1-6 haloalkoxy", is intended to include Ci, C2, C3, C 4 , C5, and C6 haloalkoxy groups. Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluorothoxy. Similarly,

"haloalkylthio" or "thiohaloalkoxy" represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example trifluoromethyl-S-, and pentafluoroethyl-S-. The term“polyhaloalkyloxy” as used herein refers to an“alkoxy” or“alkyloxy” group as defined above which includes from 2 to 9, preferably from 2 to 5, halo substituents, such as F or Cl, preferably F, such as polyfluoroalkoxy, for example, CF3CH2O, CF3O or CF3CF2CH2O.

"Hydroxy alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more hydroxyl (OH). "Ci to G hydroxy alkyl" (or hydroxy alkyl), is intended to include Ci, C2, C3, C 4 , C5, and Ce hydroxyalkyl groups.

The term "cycloalkyl" refers to cyclized alkyl groups, including mono-, bi- or poly-cyclic ring systems. "C3 to Cs cycloalkyl" or "C3-8 cycloalkyl" is intended to include C3, C 4 , C5, C6, C7, and Cs cycloalkyl groups, including monocyclic, bicyclic, and polycyclic rings. Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbomyl. Branched cycloalkyl groups such as

I-methylcyclopropyl and 2-methylcyclopropyl and spiro and bridged cycloalkyl groups are included in the definition of "cycloalkyl".

The term "cycloheteroalkyl" refers to cyclized heteroalkyl groups, including mono-, bi- or poly-cyclic ring systems. " C3 to C7 cycloheteroalkyl" or "C3-7

cycloheteroalkyl" is intended to include C3, C 4 , C5, C 6 , and C7 cycloheteroalkyl groups. Example cycloheteroalkyl groups include, but are not limited to, oxetanyl,

tetrahydrofuranyl, tetrahydropyranyl, azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, and piperazinyl. Branched cycloheteroalkyl groups, such as piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyridinylmethyl, pyridizylmethyl,

pyrimidylmethyl, and pyrazinylmethyl, are included in the definition of

"cycloheteroalkyl " .

As used herein, "carbocycle", "carbocyclyl" or "carbocyclic residue" is intended to mean any stable 3-, 4-, 5-, 6-, 7-, or 8-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-,

I I-, 12-, or l3-membered bicyclic or tricyclic hydrocarbon ring, any of which may be saturated, partially unsaturated, unsaturated or aromatic. Examples of such carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane (decalin), [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, anthracenyl, and tetrahydronaphthyl (tetralin). As shown above, bridged rings are also included in the definition of carbocycle ( e.g ., [2.2.2]bicyclooctane). Preferred carbocycles, unless otherwise specified, are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, and indanyl. When the term "carbocyclyl" is used, it is intended to include "aryl". A bridged ring occurs when one or more carbon atoms link two non- adjacent carbon atoms. Preferred bridges are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

Furthermore, the term“carbocyclyl”, including“cycloalkyl” and“cycloalkenyl”, as employed herein alone or as part of another group includes saturated or partially unsaturated (containing 1 or 2 double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 10 carbons or 3 to 6 carbons, forming the ring and which may be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl,

any of which groups may be optionally substituted with 1 to 4 substituents such as halogen, alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, nitro, cyano, thiol and/or alkylthio and/or any of the alkyl substituents.

As used herein, the term "bicyclic carbocyclyl" or "bicyclic carbocyclic group" is intended to mean a stable 9- or lO-membered carbocyclic ring system that contains two fused rings and consists of carbon atoms. Of the two fused rings, one ring is a benzo ring fused to a second ring; and the second ring is a 5- or 6-membered carbon ring which is saturated, partially unsaturated, or unsaturated. The bicyclic carbocyclic group may be attached to its pendant group at any carbon atom which results in a stable structure. The bicyclic carbocyclic group described herein may be substituted on any carbon if the resulting compound is stable. Examples of a bicyclic carbocyclic group are, but not limited to, naphthyl, l,2-dihydronaphthyl, l,2,3,4-tetrahydronaphthyl, and indanyl.

As used herein, the term "aryl", as employed herein alone or as part of another group, refers to monocyclic or polycyclic (including bicyclic and tricyclic) aromatic hydrocarbons, including, for example, phenyl, naphthyl, anthracenyl, and phenanthranyl. Aryl moieties are well known and described, for example, in Lewis, R.J., ed., Hawley's Condensed Chemical Dictionary , 13th Edition, John Wiley & Sons, Inc., New York (1997). In one embodiment, the term“aryl” denotes monocyclic and bicyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1 -naphthyl and 2-naphthyl). For example, "C6 or Cio aryl" or "C6-10 aryl" refers to phenyl and naphthyl. ETnless otherwise specified, "aryl", "C6 or Cio aryl", "C6-10 aryl", or "aromatic residue" may be unsubstituted or substituted with 1 to 5 groups, preferably 1 to 3 groups, selected from OH, OCH 3 , Cl, F, Br, I, CN, NO2, NH 2 , N(CH 2 )H, N(CH 3 ) 2 , CF 3 , OCF 3 , C(=0)CH 3 , SCH 3 , S(=0)CH 3 , S(=0) 2 CH 3 , CH 3 , CH 2 CH 3 , C0 2 H, and C0 2 CH 3.

The term "benzyl", as used herein, refers to a methyl group on which one of the hydrogen atoms is replaced by a phenyl group, wherein said phenyl group may optionally be substituted with 1 to 5 groups, preferably 1 to 3 groups, OH, OCH 3 , Cl, F, Br, I, CN, N0 2 , NH 2 , N(CH 3 )H, N(CH 3 ) 2 , CF 3 , OCF 3 , C(=0)CH 3 , SCH 3 , S(=0)CH 3 , S(=0) 2 CH 3 , CH 3 , CH 2 CH 3 , C0 2 H, and C0 2 CH 3.

As used herein, the term "heterocycle", "heterocyclyl", or "heterocyclic group" is intended to mean a stable 3-, 4-, 5-, 6-, or 7-membered monocyclic or 5-, 6-, 7-, 8-, 9-,

10-, 11-, 12-, 13-, or l4-membered polycyclic (including bicyclic and tricyclic) heterocyclic ring that is saturated, or partially unsaturated, and that contains carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from N, O and S; and including any polycyclic group in which any of the above-defined heterocyclic rings is fused to a carbocyclic or an aryl (e.g., benzene) ring. That is, the term "heterocycle",

"heterocyclyl", or "heterocyclic group" includes non-aromatic ring systems, such as heterocycloalkyl and heterocycloalkenyl. The nitrogen and sulfur heteroatoms may optionally be oxidized {i.e., N 0 and S(0)p, wherein p is 0, 1 or 2). The nitrogen atom may be substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. A nitrogen in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1. Examples of hetercyclyl include, without limitation, azetidinyl, piperazinyl, piperidinyl, piperidonyl, piperonyl, pyranyl, morpholinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, morpholinyl, di hy drofuro[2, 3 -/ijtetrahy drofuran

As used herein, the term "bicyclic heterocycle" or "bicyclic heterocyclic group" is intended to mean a stable 9- or lO-membered heterocyclic ring system which contains two fused rings and consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from N, O and S. Of the two fused rings, one ring is a 5- or 6-membered monocyclic aromatic ring comprising a 5-membered heteroaryl ring, a 6-membered heteroaryl ring or a benzo ring, each fused to a second ring. The second ring is a 5- or 6-membered monocyclic ring which is saturated, partially unsaturated, or unsaturated, and comprises a 5-membered heterocycle, a 6-membered heterocycle or a carbocycle (provided the first ring is not benzo when the second ring is a carbocycle).

The bicyclic heterocyclic group may be attached to its pendant group at any heteroatom or carbon atom which results in a stable structure. The bicyclic heterocyclic group described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1. Examples of a bicyclic heterocyclic group are, but not limited to,

1.2.3.4-tetrahydroquinolinyl, l,2,3,4-tetrahydroisoquinolinyl,

5,6,7,8-tetrahydro-quinolinyl, 2,3-dihydro-benzofuranyl, chromanyl,

1.2.3.4-tetrahydro-quinoxalinyl, and l,2,3,4-tetrahydro-quinazolinyl.

Bridged rings are also included in the definition of heterocycle. A bridged ring occurs when one or more atoms (i.e., C, O, N, or S) link two non-adjacent carbon or nitrogen atoms. Examples of bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and a carbon-nitrogen group. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

As used herein, the term "heteroaryl" is intended to mean stable monocyclic and polycyclic (including bicyclic and tricyclic) aromatic hydrocarbons that include at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrroyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, l,2,4-thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl, benzodioxolanyl, and benzodioxane. Heteroaryl groups are substituted or unsubstituted. The nitrogen atom is substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The nitrogen and sulfur heteroatoms may optionally be oxidized (, i.e ., N 0 and S(0)p, wherein p is 0, 1 or 2).

Examples of heteroaryl also include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl,

benzisothiazolyl, benzimidazolinyl, carbazolyl, 4a//-carbazolyf carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2HbH- \ ,5,2-dithiazinyl, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, liT-indazolyl, imidazolopyridinyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isothiazolopyridinyl, isoxazolyl, isoxazolopyridinyl, methyl enedioxy phenyl, naphthyridinyl,

octahydroisoquinolinyl, oxadiazolyl, l,2,3-oxadiazolyl, l,2,4-oxadiazolyl,

l,2,5-oxadiazolyl, l,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolopyridinyl, oxazolidinylperimidinyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathianyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolopyridinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2-pyrrolidonyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl,

4//-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrazolyl, tetrahydrofuranyl,

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

Examples of 5- to lO-membered heteroaryl include, but are not limited to, pyridinyl, furanyl, thienyl, pyrazolyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, oxazolidinyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, triazolyl, benzimidazolyl, 1 //-indazolyf benzofuranyl, benzothiofuranyl, benztetrazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, oxindolyl, benzoxazolinyl, benzthiazolyl, benzisothiazolyl, isatinoyl, isoquinolinyl, octahydroisoquinolinyl, isoxazolopyridinyl, quinazolinyl, quinolinyl, isothiazolopyridinyl, thiazolopyridinyl, oxazolopyridinyl, imidazolopyridinyl, and pyrazolopyridinyl. Examples of 5- to

6-membered heteroaryl include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, oxazolidinyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl. In some embodiments, the heteroaryl are selected from benzthiazolyl, imidazolpyridinyl, pyrrol opyridinyl, quinolinyl, and indolyl.

Unless otherwise indicated, "carbocyclyl" or "heterocyclyl" includes one to three additional rings fused to the carbocyclic ring or the heterocyclic ring (such as aryl, cycloalkyl, heteroaryl or cycloheteroalkyl rings), for example,

and may be optionally substituted through available carbon or nitrogen atoms (as applicable) with 1, 2, or 3 groups selected from hydrogen, halo, haloalkyl, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl-alkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxy carbonyl, arylcarbonyl, arylalkenyl,

aminocarbonylaryl, arylthio, arylsulfmyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxy, nitro, cyano, thiol, alkylthio, arylthio, heteroarylthio, arylthioalkyl, alkoxyarylthio, alkylcarbonyl, arylcarbonyl,

alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aminocarbonyl,

alkylcarbonyloxy, arylcarbonyloxy, alkylcarbonylamino, arylcarbonylamino, arylsulfmyl, arylsulfmylalkyl, arylsulfonylamino and arylsulfonaminocarbonyl and/or any of the alkyl substituents set out herein.

When any of the terms alkyl, alkenyl, alkynyl, cycloalkyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl are used as part of another group, the number of carbon atoms and ring members are the same as those defined in the terms by themselves. For example, alkoxy, haloalkoxy, alkylamino, haloalkyl, hydroxyalkyl, aminoalkyl, haloalkoxy, alkoxyalkoxy, haloalkylamino, alkoxyalkylamino, haloalkoxyalkylamino, alkylthio, and the like each independently contains the number of carbon atoms which are the same as defined for the term“alkyl”, such as 1 to 4 carbon atoms, 1 to 6 carbon atoms, 1 to 10 carbon atoms, etc. Similarly, cycloalkoxy, heterocyclyloxy,

cycloalkylamino, heterocyclylamino, aralkylamino, arylamino, aryloxy, aralkyloxy, heteroaryloxy, heteroarylalkyloxy, and the like each indepdently contains ring members which are the same as defined for the terms“cycloalkyl”,“heterocyclyl”,“aryl”, and “heteroaryl”, such as 3 to 6-membered, 4 to 7-membered, 6 to lO-membered, 5 to 10- membered, 5 or 6-membered, etc.

In accordance with a convention used in the art, a bond pointing to a bold line, such as as used in structural formulas herein, depicts the bond that is the point of attachment of the moiety or substituent to the core or backbone structure.

In accordance with a convention used in the art, a wavy or squiggly bond in a

Z'

structural formula, such as , is used to depict a stereogenic center of the carbon atom to which X’, Y’, and Z’ are attached and is intended to represent both enantiomers in a single figure. That is, a structural formula with such as wavy bond denotes each of the enantiomers individually, such as as well as a racemic mixture thereof. When a wavy or squiggly bond is attached to a double bond (such as C=C or C=N) moiety, it include cis- or trans- (or E- and Z-) geometric isomers or a mixture thereof.

It is understood herein that if a carbocyclic or heterocyclic moiety may be bonded or otherwise attached to a designated substrate through differing ring atoms without denoting a specific point of attachment, then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom. For example, the term “pyridyl” means 2-, 3- or 4-pyridyl, the term“thienyl” means 2- or 3 -thienyl, and so forth.

When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom in which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such substituent. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. One skilled in the art will recognize that substituents and other moieties of the compounds of the present invention should be selected in order to provide a compound which is sufficiently stable to provide a pharmaceutically useful compound which can be formulated into an acceptably stable pharmaceutical composition. Compounds of the present invention which have such stability are contemplated as falling within the scope of the present invention.

The term "counter ion" is used to represent a negatively charged species such as chloride, bromide, hydroxide, acetate, and sulfate. The term“metal ion” refers to alkali metal ions such as sodium, potassium or lithium and alkaline earth metal ions such as magnesium and calcium, as well as zinc and aluminum.

As referred to herein, the term "substituted" means that at least one hydrogen atom (attached to carbon atom or heteroatom) is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound. When a substituent is oxo (i.e., =0), then 2 hydrogens on the atom are replaced. Oxo substituents are not present on aromatic moieties. When a ring system e.g ., carbocyclic or heterocyclic) is said to be substituted with a carbonyl group or a double bond, it is intended that the carbonyl group or double bond be part (; i.e ., within) of the ring. Ring double bonds, as used herein, are double bonds that are formed between two adjacent ring atoms (e.g., C=C, C=N, or N=N). The term“substituted” in reference to alkyl, cycloalkyl, heteroalkyl, cycloheteroalkyl, alkylene, aryl, arylalkyl, heteroaryl, heteroarylalkyl, carbocyclyl, and heterocyclyl, means alkyl, cycloalkyl, heteroalkyl, cycloheteroalkyl, alkylene, aryl, arylalkyl, heteroaryl, heteroarylalkyl, carbocyclyl, and heterocyclyl, respectively, in which one or more hydrogen atoms, which are attached to either carbon or heteroatom, are each independently replaced with one or more non hydrogen substituent(s).

In cases wherein there are nitrogen atoms (e.g, amines) on compounds of the present invention, these may be converted to N-oxides by treatment with an oxidizing agent (e.g, mCPBA and/or hydrogen peroxides) to afford other compounds of this invention. Thus, shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxide (N— >0) derivative.

When any variable occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0, 1, 2, or 3 R groups, then said group be unsubstituted when it is substituted with 0 R group, or be substituted with up to three R groups, and at each occurrence R is selected independently from the definition of R.

Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.

As used herein, the term“tautomer” refers to each of two or more isomers of a compound that exist together in equilibrium, and are readily interchanged by migration of an atom or group within the molecule For example, one skilled in the art would readily understand that a 1,2, 3 -triazole exists in two tautomeric forms as defined above:

r^N

NH

i '

1 H-1 ,2,3-triazole 2H-1 ,2,3-triazole

Thus, this disclosure is intended to cover all possible tautomers even when a structure depicts only one of them.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, and/or other problem or complication, commensurate with a reasonable benefit/risk ratio.

The compounds of the present invention can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable salts are preferred. As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences , 18th Edition, Mack Publishing Company, Easton, PA (1990), the disclosure of which is hereby incorporated by reference.

If the compounds of the present invention have, for example, at least one basic center, they can form acid addition salts. These are formed, for example, with strong inorganic acids, such as mineral acids, for example sulfuric acid, phosphoric acid or a hydrohalic acid, with organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms, for example acetic acid, which are unsubstituted or substituted, for example, by halogen as chloroacetic acid, such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (Ci-C 4 ) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example by halogen, for example methyl- or p-toluene- sulfonic acid. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds of the present invention having at least one acid group (for example COOH) can also form salts with bases. Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as morpholine, thiomorpholine, piperidine, pyrrolidine, a mono, di or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl-propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine. Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of the present invention, or their pharmaceutically acceptable salts, are also included.

Preferred salts of the compounds of the present invention which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate, nitrate or acetate.

Preferred salts of the compounds of the present invention which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines. In addition, compounds of the present invention may have prodrug forms. Any compound that will be converted in vivo to provide the bioactive agent (i.e., a compound of formula I) is a prodrug within the scope and spirit of the invention. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:

a) Bundgaard, H., ed., Design of Prodrugs, Elsevier (1985), and Widder, K. et al., eds., Methods in Enzymology, 112:309-396, Academic Press (1985);

b) Bundgaard, H., Chapter 5, "Design and Application of Prodrugs", A Textbook of Drug Design and Development , pp. 113-191, Krosgaard-Larsen, P. et al., eds., Harwood Academic Publishers (1991);

c) Bundgaard, H., Adv. Drug Deliv. Rev., 8: 1-38 (1992);

d) Bundgaard, H. et al., J Pharm. Sci., 77:285 (1988); and

e) Kakeya, N. et al., Chem. Pharm. Bull., 32:692 (1984).

The compounds of the present invention contain a carboxy group which can form physiologically hydrolyzable esters that serve as prodrugs, i.e.,“prodrug esters”, by being hydrolyzed in the body to yield the compounds of the present invention per se. Examples of physiologically hydrolyzable esters of compounds of the present invention include C | to Cg alkyl, C ] to Cg alkylbenzyl, 4-methoxybenzyl, indanyl, phthalyl, methoxymethyl, C j _6 alkanoyloxy-C ] _g alkyl (e.g, acetoxymethyl, pivaloyloxymethyl or

propionyloxymethyl), C | to Cg alkoxycarbonyloxy-C j to Cg alkyl (e.g,

methoxycarbonyl-oxymethyl or ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl, (5-methyl-2-oxo-l,3-dioxolen-4-yl)-methyl), and other well known physiologically hydrolyzable esters used, for example, in the penicillin and cephalosporin arts. Such esters may be prepared by conventional techniques known in the art. The“prodrug esters” can be formed by reacting the carboxylic acid moiety of the compounds of the present invention with either alkyl or aryl alcohol, halide, or sulfonate employing procedures known to those skilled in the art. Such esters may be prepared by conventional techniques known in the art.

Preparation of prodrugs is well known in the art and described in, for example, King, F.D., ed., Medicinal Chemistry: Principles and Practice , The Royal Society of Chemistry, Cambridge, UK (1994); Testa, B. et al., Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology, VCHA and Wiley-VCH, Zurich, Switzerland (2003); Wermuth, C.G., ed., The Practice of Medicinal Chemistry , Academic Press, San Diego, CA (1999).

The present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include deuterium and tritium. Deuterium has one proton and one neutron in its nucleus and that has twice the mass of ordinary hydrogen. Deuterium can be represented by symbols such as " 2 H" or "D". The term "deuterated" herein, by itself or used to modify a compound or group, refers to replacement of one or more hydrogen atom(s), which is attached to carbon(s), with a deuterium atom. Isotopes of carbon include 13 C and 14 C.

Isotopically-labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed. Such compounds have a variety of potential uses, e.g ., as standards and reagents in determining the ability of a potential

pharmaceutical compound to bind to target proteins or receptors, or for imaging compounds of this invention bound to biological receptors in vivo or in vitro.

"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent. It is preferred that compounds of the present invention do not contain a N-halo, S(0)2H, or S(0)H group.

The term "solvate" means a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes 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 solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement. The solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules. "Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art. ABBREVIATIONS

Abbreviations as used herein, are defined as follows: " 1 x" for once, "2 x" for twice, "3 x" for thrice, " °C" for degrees Celsius, "eq" for equivalent or equivalents, "g" for gram or grams, "mg" for milligram or milligrams, "L" for liter or liters, "mL" for milliliter or milliliters, "pL" for microliter or microliters, "N" for normal, "M" for molar, "mmol" for millimole or millimoles, "min" for minute or minutes, "h" for hour or hours, "rt" for room temperature, "RT" for retention time,“RBF” for round bottom flask, "atm" for atmosphere, "psi" for pounds per square inch, "cone." for concentrate,“RCM” for ring-closing metathesis, "sat" or "sat'd " for saturated,“SFC” for supercritical fluid chromatography "MW" for molecular weight, "mp" for melting point, "ee" for enantiomeric excess, "MS" or "Mass Spec" for mass spectrometry, "ESI" for electrospray ionization mass spectroscopy, "HR" for high resolution, "HRMS" for high resolution mass spectrometry, "LCMS" for liquid chromatography mass spectrometry, "HPLC" for high pressure liquid chromatography, "RP HPLC" for reverse phase HPLC, "TLC" or "tlc" for thin layer chromatography, "NMR" for nuclear magnetic resonance

spectroscopy, "nOe" for nuclear Overhauser effect spectroscopy, M l H" for proton, "5" for delta, "s" for singlet, "d" for doublet, "t" for triplet, "q" for quartet, "m" for multiplet, "br" for broad, "Hz" for hertz, and "a", "b",“g”, "R", "S", "E", and "Z" are stereochemical designations familiar to one skilled in the art.

Me methyl

Et ethyl

Pr propyl

/-Pr isopropyl

Bu butyl

/-Bu isobutyl

/-Bu er -butyl

Ph phenyl

Bn benzyl

Boc or BOC tert- butyl oxy carb ony 1

B0C2O di-/er/-butyl dicarbonate

AcOH or HO Ac acetic acid

AlCb aluminum trichloride AIBN Azobis-isobutyronitrile

BBn boron tribromide

BCb boron trichloride

BEMP 2-fer/-butylimino-2-diethylamino-l,3-dimethylperhydro-l,3,2- diazaphosphorine

BOP reagent benzotriazol- 1 -yloxytris(dimethylamino)phosphonium

hexafluorophosphate

Burgess reagent l-methoxy-N-triethylammoniosulfonyl-methanimidate CBz carbobenzyloxy

DCM or CH2CI2 dichloromethane

CEECN or ACN acetonitrile

CDCh deutero-chloroform

CHCh chloroform

mCPBA or m-CPBA weto-chloroperbenzoic acid

CS2CO3 cesium carbonate

CU(OAC) 2 copper (II) acetate

Cy2NMe N-cy cl ohexy 1 -N -methyl cy cl ohexanamine

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

DCE 1,2 dichloroethane

DEA Diethylamine

DEAD Diethyl azodicarboxylate

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

DIAD Diisopropyl azodicarboxylate

DIBALH Diisobutyl aluminum hydride

DIC or DIPCDI diisopropylcarbodiimide

DIEA, DIPEA or diisopropylethylamine

Hunig's base

DMAP 4-dimethylaminopyridine

DME 1 ,2-dimethoxy ethane

DMF dimethyl formamide

DMSO dimethyl sulfoxide

cDNA complementary DNA Dppp (R)-(+)- 1 ,2-bis(diphenylphosphino)propane

DuPhos (+)-l,2-bis((2S,5S)-2,5-diethylphospholano)benzene

EDC A-(3-dimthylaminopropyl)-A r -ethyl carbodiimide

EDCI A-(3-dimthylaminopropyl)-A r -ethyl carbodiimide hydrochloride

EDTA ethylenediaminetetraacetic acid

(AX)-EtDuPhosRh(I) (+)-l,2-bis((2S,5S)-2,5-diethylphospholano)benzene(l,5- cyclooctadiene)rhodium(I) trifluoromethanesulfonate

Et 3 N or TEA triethylamine

EtOAc ethyl acetate

Et 2 0 diethyl ether

EtOH ethanol

GMF glass microfiber filter

Grubbs II (l,3-bis(2,4,6-trimethylphenyl)-2-imidazolidinylidene)dichlo ro

(phenylmethylene)(triycyclohexylphosphine)ruthenium

HC1 hydrochloric acid

HATU 0-(7-azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium

hexafluorophosphate

HEPES 4-(2-hydroxyethyl)piperaxine- 1 -ethanesulfonic acid

Hex hexane

HOBt or HOBT 1 -hydroxybenzotri azole

H2O2 hydrogen peroxide

IBX 2-iodoxybenzoic acid

H2SO4 sulfuric acid

Jones reagent Cr0 3 in aqueous H2SO4, 2 M solution

K2CO3 potassium carbonate

K2HPO4 potassium phosphate dibasic (potassium hydrogen phosphate)

KOAc potassium acetate

K3PO4 potassium phosphate tribasic

LAH lithium aluminum hydride

LG leaving group

Li OH lithium hydroxide

MeOH methanol MgSCri magnesium sulfate

MsOH or MSA methyl sulfonic acid/methanesulfonic acid

NaCl sodium chloride

NaH sodium hydride

NaHCCb sodium bicarbonate

Na 2 C0 3 sodium carbonate

NaOH sodium hydroxide

Na 2 S0 3 sodium sulfite

Na 2 S0 4 sodium sulfate

NBS N-bromosuccinimide

NCS N-chlorosuccinimide

NH 3 ammonia

NH4C1 ammonium chloride

NH40H ammonium hydroxide

NH4 + HC0 2 - ammonium formate

NMM N-methylmorpholine

OTf triflate or trifluoromethanesulfonate

Pd 2 (dba) 3 tris(dibenzylideneacetone)dipalladium(0)

Pd(OAc) 2 palladium(II) acetate

Pd/C palladium on carbon

Pd(dppf)Cl 2 [1,1 -bis(diphenylphosphino)-fenOcene]dichloropalladium(II)

Ph 3 PCl 2 triphenylphosphine dichloride

PG protecting group

POCh phosphorus oxychloride

PPTS pyridinium p-toluenesulfonate

i-PrOH or IP A isopropanol

PS Polystyrene

RT or rt room temperature

SEM-C1 2-(trimethysilyl)ethoxymethyl chloride

Si0 2 silica oxide

SnCh tin(II) chloride

TBAF tra-«-butyl ammonium fluoride TBAI tetra-//-butyl ammonium iodide

TFA trifluoroacetic acid

THF tetrahydrofuran

THP tetrahydropyran

TMSCHN2 Trimethylsilyldiazomethane

TMSCH2N3 Trimethylsilylmethyl azide

T3P propane phosphonic acid anhydride

TRIS tris (hydroxymethyl) aminomethane

pTsOH p-toluenesulfonic acid

IV. BIOLOGY

Lysophospholipids are membrane-derived bioactive lipid mediators.

Lysophospholipids include, but are not limited to, lysophosphatidic acid (l-acyl-2- hydroxy -s«-glycero-3 -phosphate; LPA), sphingosine 1 -phosphate (S1P),

lysophosphatidylcholine (LPC), and sphingosylphosphorylcholine (SPC).

Lysophospholipids affect fundamental cellular functions that include cellular

proliferation, differentiation, survival, migration, adhesion, invasion, and morphogenesis. These functions influence many biological processes that include neurogenesis, angiogenesis, wound healing, immunity, and carcinogenesis.

LPA acts through sets of specific G protein-coupled receptors (GPCRs) in an autocrine and paracrine fashion. LPA binding to its cognate GPCRs (LPAi, LPA2, LPA3, LPA 4 , LPA5, LPA 6 ) activates intracellular signaling pathways to produce a variety of biological responses.

Lysophospholipids, such as LPA, are quantitatively minor lipid species compared to their major phospholipid counterparts ( e.g ., phosphatidylcholine,

phosphatidylethanolamine, and sphingomyelin). LPA has a role as a biological effector molecule, and has a diverse range of physiological actions such as, but not limited to, effects on blood pressure, platelet activation, and smooth muscle contraction, and a variety of cellular effects, which include cell growth, cell rounding, neurite retraction, and actin stress fiber formation and cell migration. The effects of LPA are predominantly receptor mediated. Activation of the LPA receptors (LPAi, LPA2, LPA3, LPA 4 , LPAs, LPA 6 ) with LPA mediates a range of downstream signaling cascades. These include, but are not limited to, mitogen-activated protein kinase (MAPK) activation, adenylyl cyclase (AC) inhibition/activation, phospholipase C (PLC) activation/Ca 2+ mobilization, arachidonic acid release, Akt/PKB activation, and the activation of small GTPases, Rho, ROCK, Rac, and Ras. Other pathways that are affected by LPA receptor activation include, but are not limited to, cyclic adenosine monophosphate (cAMP), cell division cycle 42/GTP -binding protein (Cdc42) , proto-oncogene serine/threonine-protein kinase Raf (c-RAF), proto- oncogene tyrosine-protein kinase Src (c-src), extracellular signal-regulated kinase (ERK), focal adhesion kinase (FAK), guanine nucleotide exchange factor (GEF), glycogen synthase kinase 3b (GSK3b), c-jun amino-terminal kinase (JNK), MEK, myosin light chain II (MLC II), nuclear factor kB (NF-kB), N-methyl-D-aspartate (NMDA) receptor activation, phosphatidylinositol 3 -kinase (PI3K), protein kinase A (PKA), protein kinase C (PKC), ras-related C3 botulinum toxin substrate 1 (RAC1). The actual pathway and realized end point are dependent on a range of variables that include receptor usage, cell type, expression level of a receptor or signaling protein, and LPA concentration. Nearly all mammalian cells, tissues and organs co-express several LPA-receptor subtypes, which indicates that LPA receptors signal in a cooperative manner. LPAi, LPA2, and LPA3 share high amino acid sequence similarity.

LPA is produced from activated platelets, activated adipocytes, neuronal cells, and other cell types. Serum LPA is produced by multiple enzymatic pathways that involve monoacylglycerol kinase, phospholipase Ai, secretory phospholipase A2, and

lysophospholipase D (lysoPLD), including autotaxin. Several enzymes are involved in LPA degradation: lysophospholipase, lipid phosphate phosphatase, and LPA acyl transferase such as endophilin. LPA concentrations in human serum are estimated to be 1-5 mM. Serum LPA is bound to albumin, low-density lipoproteins, or other proteins, which possibly protect LPA from rapid degradation. LPA molecular species with different acyl chain lengths and saturation are naturally occurring, including l-palmitoyl (16:0), l-palmitoleoyl (16: 1), l-stearoyl (18:0), l-oleoyl (18: 1), l-linoleoyl (18:2), and 1- arachidonyl (20:4) LPA. Quantitatively minor alkyl LPA has biological activities similar to acyl LPA, and different LPA species activate LPA receptor subtypes with varied efficacies. LPA RECEPTORS

LPAi (previously called VZG-l/EDG-2/mrecl.3) couples with three types of G proteins, Gi/ 0 , Gq, and G12/13. Through activation of these G proteins, LPA induces a range of cellular responses through LPAi including but not limited to: cell proliferation, serum- response element (SRE) activation, mitogen-activated protein kinase (MAPK) activation, adenylyl cyclase (AC) inhibition, phospholipase C (PLC) activation, Ca 2+ mobilization, Akt activation, and Rho activation.

Wide expression of LPAi is observed in adult mice, with clear presence in testis, brain, heart, lung, small intestine, stomach, spleen, thymus, and skeletal muscle.

Similarly, human tissues also express LPAi; it is present in brain, heart, lung, placenta, colon, small intestine, prostate, testis, ovary, pancreas, spleen, kidney, skeletal muscle, and thymus.

LPA2 (EDG-4) also couples with three types of G proteins, Gi/ 0 , G q , and G12/13, to mediate LPA-induced cellular signaling. Expression of LPA2 is observed in the testis, kidney, lung, thymus, spleen, and stomach of adult mice and in the human testis, pancreas, prostate, thymus, spleen, and peripheral blood leukocytes. Expression of LPA2 is upregulated in various cancer cell lines, and several human LPA2 transcriptional variants with mutations in the 3 '-untranslated region have been observed. Targeted deletion of LPA2 in mice has not shown any obvious phenotypic abnormalities, but has demonstrated a significant loss of normal LPA signaling ( e.g ., PLC activation, Ca 2+ mobilization, and stress fiber formation) in primary cultures of mouse embryonic fibroblasts (MEFs). Creation of Ipa l(-/-) I pal (-/-) double-null mice has revealed that many LPA-induced responses, which include cell proliferation, AC inhibition, PLC activation, Ca 2+ mobilization, JNK and Akt activation, and stress fiber formation, are absent or severely reduced in double-null MEFs. All these responses, except for AC inhibition (AC inhibition is nearly abolished in LPAi (-/-) MEFs), are only partially affected in either LPAi (-/-) or LPA2 (-/-) MEFs. LPA2 contributes to normal LPA- mediated signaling responses in at least some cell types (Choi et al, Biochemica et Biophysica Acta 2008, 1781, p531-539).

LPA3 (EDG-7) is distinct from LPAi and LPA2 in its ability to couple with Gi/ 0 and G q but not G12/1 3 and is much less responsive to LPA species with saturated acyl chains. LPA 3 can mediate pleiotropic LPA-induced signaling that includes PLC activation, Ca 2+ mobilization, AC inhibition/activation, and MAPK activation.

Overexpression of LPA 3 in neuroblastoma cells leads to neurite elongation, whereas that of LPAi or LPA 2 results in neurite retraction and cell rounding when stimulated with LPA. Expression of LPA 3 is observed in adult mouse testis, kidney, lung, small intestine, heart, thymus, and brain. In humans, it is found in the heart, pancreas, prostate, testis, lung, ovary, and brain (frontal cortex, hippocampus, and amygdala).

LPA 4 (p2y9/GPR23) is of divergent sequence compared to LPAi, LPA2, and LPA 3 with closer similarity to the platelet-activating factor (PAF) receptor. LPA 4 mediates LPA induced Ca 2+ mobilization and cAMP accumulation, and functional coupling to the G protein Gs for AC activation, as well as coupling to other G proteins. The LPA 4 gene is expressed in the ovary, pancreas, thymus, kidney and skeletal muscle.

LPA5 (GPR92) is a member of the purinocluster of GPCRs and is structurally most closely related to LPA 4 . LPAs is expressed in human heart, placenta, spleen, brain, lung and gut. LPAs also shows very high expression in the CD8+ lymphocyte

compartment of the gastrointestinal tract.

LPA 6 (p2y5) is a member of the purinocluster of GPCRs and is structurally most closely related to LPA 4. LPA 6 is an LPA receptor coupled to the Gl2/l3-Rho signaling pathways and is expressed in the inner root sheaths of human hair follicles.

Illustrative Biological Activity

Wound Healing

Normal wound healing occurs by a highly coordinated sequence of events in which cellular, soluble factors and matrix components act in concert to repair the injury. The healing response can be described as taking place in four broad, overlapping phases— hemostasis, inflammation, proliferation, and remodeling. Many growth factors and cytokines are released into a wound site to initiate and perpetuate wound healing processes.

When wounded, damaged blood vessels activate platelets. The activated platelets play pivotal roles in subsequent repair processes by releasing bioactive mediators to induce cell proliferation, cell migration, blood coagulation, and angiogenesis. LPA is one such mediator that is released from activated platelets; this induces platelet aggregation along with mitogenic/migration effects on the surrounding cells, such as endothelial cells, smooth muscle cells, fibroblasts, and keratinocytes.

Topical application of LPA to cutaneous wounds in mice promotes repair processes (wound closure and increased neoepithelial thickness) by increasing cell proliferation/ migration without affecting secondary inflammation.

Activation of dermal fibroblasts by growth factors and cytokines leads to their subsequent migration from the edges of the wound into the provisional matrix formed by the fibrin clot whereupon the fibroblasts proliferate and start to restore the dermis by secreting and organizing the characteristic dermal extracellular matrix (ECM). The increasing number of fibroblasts within the wound and continuous precipitation of ECM enhances matrix rigidity by applying small tractional forces to the newly formed granulation tissue. The increase in mechanical stress, in conjunction with transforming growth factor b (TGFP), induces a-smooth muscle actin (a-SMA) expression and the subsequent transformation of fibroblasts into myofibroblasts. Myofibroblasts facilitate granulation tissue remodeling via myofibroblast contraction and through the production of ECM components.

LPA regulates many important functions of fibroblasts in wound healing, including proliferation, migration, differentiation and contraction. Fibroblast proliferation is required in wound healing in order to fill an open wound. In contrast, fibrosis is characterized by intense proliferation and accumulation of myofibroblasts that actively synthesize ECM and proinflammatory cytokines. LPA can either increase or suppress the proliferation of cell types important in wound healing, such as epithelial and endothelial cells (EC), macrophages, keratinocytes, and fibroblasts. A role for LPAi in LPA-induced proliferation was provided by the observation that LPA-stimulated proliferation of fibroblasts isolated from LPAi receptor null mice was attenuated (Mills el al , Nat Rev. Cancer 2003; 3: 582-591). LPA induces cytoskeletal changes that are integral to fibroblast adhesion, migration, differentiation and contraction.

Fibrosis

Tissue injury initiates a complex series of host wound-healing responses; if successful, these responses restore normal tissue structure and function. If not, these responses can lead to tissue fibrosis and loss of function. For the majority of organs and tissues the development of fibrosis involves a multitude of events and factors. Molecules involved in the development of fibrosis include proteins or peptides (profibrotic cytokines, chemokines, metalloproteinases etc.) and phospholipids. Phospholipids involved in the development of fibrosis include platelet activating factor (PAF), phosphatidyl choline, sphingosine-l phosphate (S1P) and lysophosphatidic acid (LPA).

A number of muscular dystrophies are characterized by a progressive weakness and wasting of musculature, and by extensive fibrosis. It has been shown that LPA treatment of cultured myoblasts induced significant expression of connective tissue growth factor (CTGF). CTGF subsequently induces collagen, fibronectin and integrin expression and induces dedifferentiation of these myoblasts. Treatment of a variety of cell types with LPA induces reproducible and high level induction of CTGF (J.P. Pradere, et al., LPAi receptor activation promotes renal interstitial fibrosis, J. Am. Soc. Nephrol. 18 (2007) 3110-3118; N. Wiedmaier, et al., Int J Med Microbiol; 298(3-4):23 l-43, 2008). CTGF is a profibrotic cytokine, signaling down-stream and in parallel with TGFp.

CTGF expression by gingival epithelial cells, which are involved in the development of gingival fibromatosis, was found to be exacerbated by LPA treatment (A. Kantarci, et al., J. Pathol. 210 (2006) 59-66).

LPA is associated with the progression of liver fibrosis. In vitro , LPA induces stellate cell and hepatocyte proliferation. These activated cells are the main cell type responsible for the accumulation of ECM in the liver. Furthermore, LPA plasma levels rise during CCL-induced liver fibrosis in rodents, or in hepatitis C virus-induced liver fibrosis in humans (N. Watanabe, et al ., Plasma lysophosphatidic acid level and serum autotaxin activity are increased in liver injury in rats in relation to its severity, Life Sci. 81 (2007) 1009-1015; N.Watanabe, et al., J. Clin. Gastroenterol. 41 (2007) 616-623).

An increase of phospholipid concentrations in the bronchoalveolar lavage fluid in rabbits and rodents injected with bleomycin has been reported (K. Kuroda, et al.,

Phospholipid concentration in lung lavage fluid as biomarker for pulmonary fibrosis, Inhal. Toxicol. 18 (2006) 389-393; K. Yasuda, et al, Lung 172 (1994) 91-102).

LPA is associated with heart disease and mycocardial remodeling. Serum LPA levels are increased after myocardial infarction in patients and LPA stimulates rat cardiac fibroblast proliferation and collagen production ( Chen et al. FEBSLett. 2006 Aug

2l;580(l9):4737-45). Pulmonary Fibrosis

In the lung, aberrant wound healing responses to injury contribute to the pathogenesis of fibrotic lung diseases. Fibrotic lung diseases, such as idiopathic pulmonary fibrosis (IPF), are associated with high morbidity and mortality.

LPA is an important mediator of fibroblast recruitment in pulmonary fibrosis.

LPA and LPAi play key pathogenic roles in pulmonary fibrosis. Fibroblast

chemoattractant activity plays an important role in the lungs in patients with pulmonary fibrosis. Profibrotic effects of LPAi-receptor stimulation is explained by LPAi-receptor- mediated vascular leakage and increased fibroblast recruitment, both profibrotic events. The LPA-LPAi pathway has a role in mediating fibroblast migration and vascular leakage in IPF. The end result is the aberrant healing process that characterizes this fibrotic condition.

The LPAi receptor is the LPA receptor most highly expressed on fibroblasts obtained from patients with IPF. Furthermore, BAL obtained from IPF patients induced chemotaxis of human foetal lung fibroblasts that was blocked by the dual LPAi- LPA 3 receptor antagonist Ki 16425. In an experimental bleomycin-induced lung injury mouse model, it was shown that LPA levels were high in bronchoalveolar lavage samples compared with unexposed controls. LPAi knockout mice are protected from fibrosis after bleomycin challenge with reduced fibroblast accumulation and vascular leakage. In human subjects with IPF, high LPA levels were observed in bronchoalveolar lavage samples compared with healthy controls. Increased fibroblast chemotactic activity in these samples was inhibited by the Ki 16425 indicating that fibroblast migration is mediated by the LPA-LPA receptor(s) pathway (Tager et al. Nature Medicine , 2008, 14, 45-54).

The LPA-LPAi pathway is crucial in fibroblast recruitment and vascular leakage in pulmonary fibrosis.

Activation of latent TGF-b by the anb6 integrin plays a critical role in the development of lung injury and fibrosis { Munger et al. Cell, vol. 96, 319-328, 1999). LPA induces o^6-mediated TGF-b activation on human lung epithelial cells {Xu et al. Am. J. Pathology , 2009, 174 , 1264-1279). The LPA-induced OA^6-mediated TGF-b activation is mediated by the LPA2 receptor. Expression of the LPA2 receptor is increased in epithelial cells and mesenchymal cells in areas of lung fibrosis from IPF patients compared to normal human lung tissue. The LPA-LPA2 pathway contributes to the activation of the TGF-b pathway in pulmonary fibrosis. In some embodiments, compounds that inhibit LPA2 show efficacy in the treatment of lung fibrosis. In some embodiments, compounds that inhibit both LPA1 and LPA2 show improved efficacy in the treatment of lung fibrosis compared to compounds which inhibit only LPA1 or LPA2.

Renal Fibrosis

LPA and LPAi are involved in the etiology of kidney fibrosis. LPA has effects on both proliferation and contraction of glomerular mesangial cells and thus has been implicated in proliferative glomerulonephritis (C.N. Inoue, et al ., Clin. Sci. (Colch.) 1999, 96, 431-436). In an animal model of renal fibrosis [unilateral ureteral obstruction (UUO)], it was found that renal LPA receptors are expressed under basal conditions with an expression order of LPA2>LPA3=LPAI»LPA 4. This model mimics in an accelerated manner the development of renal fibrosis including renal inflammation, fibroblast activation and accumulation of extracellular matrix in the tubulointerstitium. UUO significantly induced LPAi-receptor expression. This was paralleled by renal LPA production (3.3 fold increase) in conditioned media from kidney explants. Contra-lateral kidneys exhibited no significant changes in LPA release and LPA-receptors expression. This shows that a prerequisite for an action of LPA in fibrosis is met: production of a ligand (LPA) and induction of one of its receptors (the LPAi receptor) (J.P. Pradere et al, Biochimica et Biophysica Acta, 2008, 1781, 582-587).

In mice where the LPAi receptor was knocked out (LPAi (-/-), the development of renal fibrosis was significantly attenuated. UUO mice treated with the LPA receptor antagonist Ki 16425 closely resembled the profile of LPAi (-/-) mice.

LPA can participate in intraperitonial accumulation of monocyte/macrophages and LPA can induce expression of the profibrotic cytokine CTGF in primary cultures of human fibroblasts (J.S. Koh ,et al, ./. Clin. Invest., 1998, 102, 716-727).

LPA treatment of a mouse epithelial renal cell line, MCT, induced a rapid increase in the expression of the profibrotic cytokine CTGF. CTGF plays a crucial role in UUO- induced tubulointerstitial fibrosis (TIF), and is involved in the profibrotic activity of TGFp This induction was almost completely suppressed by co-treatment with the LPA- receptor antagonist Ki 16425. In one aspect, the profibrotic activity of LPA in kidney results from a direct action of LPA on kidney cells involving induction of CTGF.

Hepatic fibrosis

LPA is implicated in liver disease and fibrosis. Plasma LPA levels and serum autotaxin (enzyme responsible for LPA production) are elevated in hepatitis patients and animal models of liver injury in correlation with increased fibrosis. LPA also regulates liver cell function. LPAi and LPA 2 receptors are expressed by mouse hepatic stellate cells and LPA stimulates migration of hepatic myofibroblasts.

Ocular Fibrosis

LPA is in involved in wound healing in the eye. LPAi and LPA3 receptors are detectable in the normal rabbit corneal epithelial cells, keratocytes and endothelial cells and LPAi and LPA3 expression are increased in corneal epithelial cells following injury.

LPA and its homologues are present in the aqueous humor and the lacrimal gland fluid of the rabbit eye and these levels are increased in a rabbit corneal injury model.

LPA induces actin stress fiber formation in rabbit corneal endothelial and epithelial cells and promotes contraction corneal fibroblasts. LPA also stimulates proliferation of human retinal pigmented epithelial cells

Cardiac fibrosis

LPA is implicated in myocardial infarction and cardiac fibrosis. Serum LPA levels are increased in patients following mycocardial infarction (MI) and LPA stimulates proliferation and collagen production (fibrosis) by rat cardiac fibroblasts. Both LPA1 and LPA3 receptors are highly expressed in human heart tissue.

Treatment of Fibrosis

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used to treat or prevent fibrosis in a mammal. In one aspect, a compound of Formulas (I), or a pharmaceutically acceptable salt thereof, is used to treat fibrosis of an organ or tissue in a mammal. In one aspect is a method for preventing a fibrosis condition in a mammal, the method comprising administering to the mammal at risk of developing one or more fibrosis conditions a therapeutically effective amount of a compound of Formulas (I), or a pharmaceutically acceptable salt thereof. In one aspect, the mammal has been exposed to one or more environmental conditions that are known to increase the risk of fibrosis of an organ or tissue. In one aspect, the mammal has been exposed to one or more environmental conditions that are known to increase the risk of lung, liver or kidney fibrosis. In one aspect, the mammal has a genetic predisposition of developing fibrosis of an organ or tissue. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is administered to a mammal to prevent or minimize scarring following injury. In one aspect, injury includes surgery.

The terms“fibrosis” or“fibrosing disorder,” as used herein, refers to conditions that are associated with the abnormal accumulation of cells and/or fibronectin and/or collagen and/or increased fibroblast recruitment and include but are not limited to fibrosis of individual organs or tissues such as the heart, kidney, liver, joints, lung, pleural tissue, peritoneal tissue, skin, cornea, retina, musculoskeletal and digestive tract.

Exemplary diseases, disorders, or conditions that involve fibrosis include, but are not limited to: Lung diseases associated with fibrosis, e.g ., idiopathic pulmonary fibrosis, pulmonary fibrosis secondary to systemic inflammatory disease such as rheumatoid arthritis, scleroderma, lupus, cryptogenic fibrosing alveolitis, radiation induced fibrosis, chronic obstructive pulmonary disease (COPD), scleroderma, chronic asthma, silicosis, asbestos induced pulmonary or pleural fibrosis, acute lung injury and acute respiratory distress (including bacterial pneumonia induced, trauma induced, viral pneumonia induced, ventilator induced, non-pulmonary sepsis induced, and aspiration induced); Chronic nephropathies associated with injury/fibrosis (kidney fibrosis), e.g. ,

glomerulonephritis secondary to systemic inflammatory diseases such as lupus and scleroderma, diabetes, glomerular nephritis, focal segmental glomerular sclerosis, IgA nephropathy, hypertension, allograft and Alport; Gut fibrosis, e.g. , scleroderma, and radiation induced gut fibrosis; Liver fibrosis, e.g. , cirrhosis, alcohol induced liver fibrosis, nonalcoholic steatohepatitis (NASH), biliary duct injury, primary biliary cirrhosis, infection or viral induced liver fibrosis (e.g, chronic HCV infection), and autoimmune hepatitis; Head and neck fibrosis, e.g, radiation induced; Corneal scarring, e.g, LASIK (laser-assisted in situ keratomileusis), corneal transplant, and trabeculectomy;

Hypertrophic scarring and keloids, e.g, burn induced or surgical; and other fibrotic diseases, e.g, sarcoidosis, scleroderma, spinal cord injury/fibrosis, myelofibrosis, vascular restenosis, atherosclerosis, arteriosclerosis, Wegener's granulomatosis, mixed connective tissue disease, and Peyronie's disease.

In one aspect, a mammal suffering from one of the following non-limiting exemplary diseases, disorders, or conditions will benefit from therapy with a compound of the present invention, or a pharmaceutically acceptable salt thereof: atherosclerosis, thrombosis, heart disease, vasculitis, formation of scar tissue, restenosis, phlebitis, COPD (chronic obstructive pulmonary disease), pulmonary hypertension, pulmonary fibrosis, pulmonary inflammation, bowel adhesions, bladder fibrosis and cystitis, fibrosis of the nasal passages, sinusitis, inflammation mediated by neutrophils, and fibrosis mediated by fibroblasts.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is administered to a mammal with fibrosis of an organ or tissue or with a predisposition of developing fibrosis of an organ or tissue with one or more other agents that are used to treat fibrosis. In one aspect, the one or more agents include corticosteroids. In one aspect, the one or more agents include immunosuppressants. In one aspect, the one or more agents include B-cell antagonists. In one aspect, the one or more agents include uteroglobin.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used to treat a dermatological disorders in a mammal. The term “dermatological disorder,” as used herein refers to a skin disorder. Such dermatological disorders include, but are not limited to, proliferative or inflammatory disorders of the skin such as, atopic dermatitis, bullous disorders, collagenoses, psoriasis, scleroderma, psoriatic lesions, dermatitis, contact dermatitis, eczema, urticaria, rosacea, wound healing, scarring, hypertrophic scarring, keloids, Kawasaki Disease, rosacea, Sjogren-Larsso Syndrome, urticaria. In one aspect, a compound of the present invention, or a

pharmaceutically acceptable salt thereof, is used to treat systemic sclerosis.

Pain

Since LPA is released following tissue injury, LPAi plays an important role in the initiation of neuropathic pain. LPAi, unlike LPA2 or LPA3, is expressed in both dorsal root ganglion (DRG) and dorsal root neurons. Using the antisense oligodeoxynucleotide (AS-ODN) for LPAi and LPAi-null mice, it was found that LPA-induced mechanical allodynia and hyperalgesia is mediated in an LPAi-dependent manner. LPAi and downstream Rho-ROCK activation play a role in the initiation of neuropathic pain signaling. Pretreatment with Clostridium botulinum C3 exoenzyme (BoTXC3, Rho inhibitor) or Y-27632 (ROCK inhibitor) completely abolished the allodynia and hyperalgesia in nerve-injured mice. LPA also induced demyelination of the dorsal root, which was prevented by BoTXC3. The dorsal root demyelination by injury was not observed in LPAi-null mice or AS-ODN injected wild-type mice. LPA signaling appears to induce important neuropathic pain markers such as protein kinase C g (PKCy) and a voltage-gated calcium channel a2d1 subunit (Caa25l) in an LPAi and Rho-dependent manner (M. Inoue, et al ., Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling, Nat. Med. 10 (2004) 712-718).

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of pain in a mammal. In one aspect, the pain is acute pain or chronic pain. In another aspect, the pain is neuropathic pain.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of fibromylagia. In one aspect, fibromyalgia stems from the formation of fibrous scar tissue in contractile (voluntary) muscles. Fibrosis binds the tissue and inhibits blood flow, resulting in pain.

Cancer

Lysophospholipid receptor signaling plays a role in the etiology of cancer.

Lysophosphatidic acid (LPA) and its G protein-coupled receptors (GPCRs) LPAi, LPA2, and/or LPA3 play a role in the development of several types of cancers. The initiation, progression and metastasis of cancer involve several concurrent and sequential processes including cell proliferation and growth, survival and anti-apoptosis, migration of cells, penetration of foreign cells into defined cellular layers and/or organs, and promotion of angiogenesis. The control of each of these processes by LPA signaling in physiological and pathophysiological conditions underscores the potential therapeutic usefulness of modulating LPA signaling pathways for the treatment of cancer, especially at the level of the LPA receptors or ATX/lysoPLD. Autotaxin (ATX) is a prometastatic enzyme initially isolated from the conditioned medium of human melanoma cells that stimulates a myriad of biological activities, including angiogenesis and the promotion of cell growth, migration, survival, and differentiation through the production of LPA ( Mol Cancer Ther 2008;7(l 0):3352-62).

LPA signals through its own GPCRs leading to activation of multiple downstream effector pathways. Such downstream effector pathways play a role in cancer. LPA and its GPCRs are linked to cancer through major oncogenic signaling pathways.

LPA contributes to tumorigenesis by increasing motility and invasiveness of cells. LPA has been implicated in the initiation or progression of ovarian cancer. LPA is present at significant concentrations (2-80 mM) in the ascitic fluid of ovarian cancer patients. Ovarian cancer cells constitutively produce increased amounts of LPA as compared to normal ovarian surface epithelial cells, the precursor of ovarian epithelial cancer.

Elevated LPA levels are also detected in plasma from patients with early-stage ovarian cancers compared with controls. LPA receptors (LPA2 and LPA3) are also overexpressed in ovarian cancer cells as compared to normal ovarian surface epithelial cells. LPA stimulates Cox -2 expression through transcriptional activation and post-transcriptional enhancement of Cox-2 mRNA in ovarian cancer cells. Prostaglandins produced by Cox-2 have been implicated in a number of human cancers and pharmacological inhibition of Cox-2 activity reduces colon cancer development and decreases the size and number of adenomas in patients with familial adenomatous polyposis. LPA has also been implicated in the initiation or progression of prostate cancer, breast cancer, melanoma, head and neck cancer, bowel cancer (colorectal cancer), thyroid cancer and other cancers (Gardell et al , Trends in Molecular Medicine, vol. 12, no. 2, p 65-75, 2006; Ishii et al , Anna. Rev.

Biochem , 73, 321-354, 2004; Mills et al., Nat. Rev. Cancer, 3, 582-591, 2003; Murph et al., Biochimica et Biophysica Acta, 1781, 547-557, 2008).

The cellular responses to LPA are mediated through the lysophosphatidic acid receptors. For example, LPA receptors mediate both migration of and invasion by pancreatic cancer cell lines: an antagonist of LPAi and LPA 3 (KΪ16425) and LPAi- specific siRNA effectively blocked in vitro migration in response to LPA and peritoneal fluid (ascites) from pancreatic cancer patients; in addition, Ki 16425 blocked the LPA- induced and ascites-induced invasion activity of a highly peritoneal metastatic pancreatic cancer cell line (Yamada et al, J. Biol. Chem., 279, 6595-6605, 2004). Colorectal carcinoma cell lines show significant expression of LPAi mRNA and respond to LPA by cell migration and production of angiogenic factors. Overexpression of LPA receptors has a role in the pathogenesis of thyroid cancer. LPA 3 was originally cloned from prostate cancer cells, concordant with the ability of LPA to induce autocrine proliferation of prostate cancer cells.

LPA has stimulatory roles in cancer progression in many types of cancer. LPA is produced from and induces proliferation of prostate cancer cell lines. LPA induces human colon carcinoma DLD1 cell proliferation, migration, adhesion, and secretion of angiogenic factors through LPAi signaling. In other human colon carcinoma cells lines (HT29 and WiDR), LPA enhances cell proliferation and secretion of angiogenic factors. In other colon cancer cell lines, LPA2 and LPA3 receptor activation results in

proliferation of the cells. The genetic or pharmacological manipulation of LPA metabolism, specific blockade of receptor signaling, and/or inhibition of downstream signal transduction pathways, represent approaches for cancer therapies.

It has been reported that LPA and other phospholipids stimulate expression of interleukin-8 (IL-8) in ovarian cancer cell lines. In some embodiments, high

concentrations of IL-8 in ovarian cancer correlate with poor initial response to chemotherapy and with poor prognosis, respectively. In animal models, expression of IL- 8 and other growth factors such as vascular endothelial growth factor (VEGF) is associated with increased tumorigenicity, ascites formation, angiogenesis, and invasiveness of ovarian cancer cells. In some aspects, IL-8 is an important modulator of cancer progression, drug resistance, and prognosis in ovarian cancer. In some embodiments, a compound of the present invention inhibits or reduces IL-8 expression in ovarian cancer cell lines.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of cancer. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of malignant and benign proliferative disease. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used to prevent or reduce proliferation of tumor cells, invasion and metastasis of carcinomas, pleural mesothelioma (Yamada, Cancer Sci., 2008, 99(8), 1603-1610) or peritoneal

mesothelioma, cancer pain, bone metastases (Boucharaba et al , J. Clin. Invest ., 2004, 114(12), 1714-1725; Boucharaba et al, Proc. Natl. acad. Sci., 2006, 103(25) 9643-9648). In one aspect is a method of treating cancer in a mammal, the method comprising administering to the mammal a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a second therapeutic agent, wherein the second therapeutic agent is an anti -cancer agent.

The term“cancer,” as used herein refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread). The types of cancer include, but is not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma or basal cell cancer) or hematological tumors (such as the leukemias) at any stage of the disease with or without metastases.

Additional non-limiting examples of cancers include, acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, anal cancer, appendix cancer, astrocytomas, atypical teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer (osteosarcoma and malignant fibrous histiocytoma), brain stem glioma, brain tumors, brain and spinal cord tumors, breast cancer, bronchial tumors, Burkitt lymphoma, cervical cancer, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T- Cell lymphoma, embryonal tumors, endometrial cancer, ependymoblastoma,

ependymoma, esophageal cancer, ewing sarcoma family of tumors, eye cancer, retinoblastoma, gallbladder cancer, gastric (stomach) cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), gastrointestinal stromal cell tumor, germ cell tumor, glioma, hairy cell leukemia, head and neck cancer, hepatocellular (liver) cancer, Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors (endocrine pancreas), Kaposi sarcoma, kidney cancer, Langerhans cell

histiocytosis, laryngeal cancer, leukemia, Acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, hairy cell leukemia, liver cancer, non-small cell lung cancer, small cell lung cancer, Burkitt lymphoma, cutaneous T-cell lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, lymphoma, Waldenstrom macroglobulinemia, medulloblastoma, medulloepithelioma, melanoma, mesothelioma, mouth cancer, chronic myelogenous leukemia, myeloid leukemia, multiple myeloma, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma, malignant fibrous histiocytoma of bone, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer,

papillomatosis, parathyroid cancer, penile cancer, pharyngeal cancer, pineal parenchymal tumors of intermediate differentiation, pineoblastoma and supratentorial primitive neuroectodermal tumors, pituitary tumor, plasma cell neoplasm/multiple myeloma, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell (kidney) cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma, Ewing sarcoma family of tumors, sarcoma, kaposi, Sezary syndrome, skin cancer, small cell Lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, stomach (gastric) cancer, supratentorial primitive

neuroectodermal tumors, T-cell lymphoma, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer, urethral cancer, uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, Wilms tumor.

The increased concentrations of LPA and vesicles in ascites from ovarian cancer patients and breast cancer effussions indicate that it could be an early diagnostic marker, a prognostic indicator or an indicator of response to therapy (Mills et al , Nat. Rev. Cancer ., 3, 582-591, 2003; Sutphen et al ., Cancer Epidemiol. Biomarkers Prev. 13, 1185-1191, 2004). LPA concentrations are consistently higher in ascites samples than in matched plasma samples.

Respiratory and Allergic Disorders

In one aspect, LPA is a contributor to the pathogenesis of respiratory diseases. In one aspect the respiratory disease is asthma. Proinflammatory effects of LPA include degranulation of mast cells, contraction of smooth-muscle cells and release of cytokines from dendritic cells. Airway smooth muscle cells, epithelial cells and lung fibroblasts all show responses to LPA. LPA induces the secretion of IL-8 from human bronchial epithelial cells. IL-8 is found in increased concentrations in BAL fluids from patients with asthma, chronic obstructive lung disease, pulmonary sarcoidosis and acute respiratory distress syndrome and 11-8 has been shown to exacerbate airway inflammation and airway remodeling of asthmatics. LPA1, LPA2 and LPA3 receptors have all been shown to contribute to the LPA-induced IL-8 production. Studies cloning multiple GPCRs that are activated by LPA allowed the demonstration of the presence of mRNA for the LPAi, LPA 2 and LPA 3 in the lung (J.J.A. Contos, et al., Mol. Pharmacol. 58, 1188-1196, 2000).

The release of LPA from platelets activated at a site of injury and its ability to promote fibroblast proliferation and contraction are features of LPA as a mediator of wound repair. In the context of airway disease, asthma is an inflammatory disease where inappropriate airway“repair” processes lead to structural“remodeling” of the airway.

In asthma, the cells of the airway are subject to ongoing injury due to a variety of insults, including allergens, pollutants, other inhaled environmental agents, bacteria and viruses, leading to the chronic inflammation that characterizes asthma.

In one aspect, in the asthmatic individual, the release of normal repair mediators, including LPA, is exaggerated or the actions of the repair mediators are inappropriately prolonged leading to inappropriate airway remodeling. Major structural features of the remodeled airway observed in asthma include a thickened lamina reticularis (the basement membrane-like structure just beneath the airway epithelial cells), increased numbers and activation of myofibroblasts, thickening of the smooth muscle layer, increased numbers of mucus glands and mucus secretions, and alterations in the connective tissue and capillary bed throughout the airway wall. In one aspect, LPA contributes to these structural changes in the airway. In one aspect, LPA is involved in acute airway hyperresponsiveness in asthma. The lumen of the remodeled asthmatic airway is narrower due to the thickening of the airway wall, thus decreasing airflow. In one aspect, LPA contributes to the long-term structural remodeling and the acute hyperresponsiveness of the asthmatic airway. In one aspect, LPA contributes to the hyper responsiveness that is a primary feature of acute exacerbations of asthma.

In addition to the cellular responses mediated by LPA, several of the LPA signaling pathway components leading to these responses are relevant to asthma. EGF receptor upregulation is induced by LPA and is also seen in asthmatic airways (M.

Amishima, et al., Am. J. Respir. Crit. Care Med. 157, 1907- 1912, 1998). Chronic inflammation is a contributor to asthma, and several of the transcription factors that are activated by LPA are known to be involved in inflammation (Ediger et al., Eur Respir J 21 :759-769, 2003). In one aspect, the fibroblast proliferation and contraction and extracellular matrix secretion stimulated by LPA contributes to the fibroproliferative features of other airway diseases, such as the peribronchiolar fibrosis present in chronic bronchitis, emphysema, and interstitial lung disease. Emphysema is also associated with a mild fibrosis of the alveolar wall, a feature which is believed to represent an attempt to repair alveolar damage. In another aspect, LPA plays a role in the fibrotic interstitial lung diseases and obliterative bronchiolitis, where both collagen and myofibroblasts are increased. In another aspect, LPA is involved in several of the various syndromes that constitute chronic obstructive pulmonary disease.

Administration of LPA in vivo induces airway hyper-responsiveness, itch-scratch responses, infiltration and activation of eosinophils and neutrophils, vascular remodeling, and nociceptive flexor responses. LPA also induces histamine release from mouse and rat mast cells. In an acute allergic reaction, histamine induces various responses, such as contraction of smooth muscle, plasma exudation, and mucus production. Plasma exudation is important in the airway, because the leakage and subsequent airway-wall edema contribute to the development of airway hyperresponsiveness. Plasma exudation progresses to conjunctival swelling in ocular allergic disorder and nasal blockage in allergic rhinitis (Hashimoto et al ., J Pharmacol rici 100, 82 - 87, 2006). In one aspect, plasma exudation induced by LPA is mediated by histamine release from mast cells via one or more LPA receptors. In one aspect, the LPA receptor(s) include LPAi and/or LPA 3. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of various allergic disorders in a mammal. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of respiratory diseases, disorders or conditions in a mammal. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of asthma in a mammal. In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used in the treatment of chronic asthma in a mammal.

The term“respiratory disease,” as used herein, refers to diseases affecting the organs that are involved in breathing, such as the nose, throat, larynx, eustachian tubes, trachea, bronchi, lungs, related muscles ( e.g ., diaphram and intercostal s), and nerves. Respiratory diseases include, but are not limited to, asthma, adult respiratory distress syndrome and allergic (extrinsic) asthma, non-allergic (intrinsic) asthma, acute severe asthma, chronic asthma, clinical asthma, nocturnal asthma, allergen-induced asthma, aspirin-sensitive asthma, exercise-induced asthma, isocapnic hyperventilation, child-onset asthma, adult-onset asthma, cough-variant asthma, occupational asthma, steroid-resistant asthma, seasonal asthma, seasonal allergic rhinitis, perennial allergic rhinitis, chronic obstructive pulmonary disease, including chronic bronchitis or emphysema, pulmonary hypertension, interstitial lung fibrosis and/or airway inflammation and cystic fibrosis, and hypoxia.

The term“asthma” as used herein refers to any disorder of the lungs characterized by variations in pulmonary gas flow associated with airway constriction of whatever cause (intrinsic, extrinsic, or both; allergic or non-allergic). The term asthma may be used with one or more adjectives to indicate cause.

In one aspect, presented herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the treatment or prevention of chronic obstructive pulmonary disease in a mammal comprising administering to the mammal at least once an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof. In addition, chronic obstructive pulmonary disease includes, but is not limited to, chronic bronchitis or emphysema, pulmonary hypertension, interstitial lung fibrosis and/or airway inflammation, and cystic fibrosis.

Nervous System

The nervous system is a major locus for LPAi expression; there it is spatially and temporally regulated throughout brain development. Oligodendrocytes, the myelinating cells in the central nervous system (CNS), express LPAi in mammals. In addition, Schwann cells, the myelinating cells of the peripheral nervous system, also express LPAi, which is involved in regulating Schwann cell survival and morphology. These

observations identify important functions for receptor-mediated LPA signaling in neurogenesis, cell survival, and myelination.

Exposure of peripheral nervous system cell lines to LPA produces a rapid retraction of their processes resulting in cell rounding, which was, in part, mediated by polymerization of the actin cytoskeleton. In one aspect, LPA causes neuronal

degeneration under pathological conditions when the blood-brain barrier is damaged and serum components leak into the brain (Moolenaar, Curr. Opin. Cell Biol. 7:203-10,

1995). Immortalized CNS neuroblast cell lines from the cerebral cortex also display retraction responses to LPA exposure through Rho activation and actomyosin

interactions. In one aspect, LPA is associated with post-ischemic neural damage (J. Neurochem. 61, 340, 1993; J. Neurochem ., 70:66, 1998).

In one aspect, provided is a compound of the present invention, or a

pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a nervous system disorder in a mammal. The term“nervous system disorder,” as used herein, refers to conditions that alter the structure or function of the brain, spinal cord or peripheral nervous system, including but not limited to Alzheimer’s Disease, cerebral edema, cerebral ischemia, stroke, multiple sclerosis, neuropathies, Parkinson’s Disease, those found after blunt or surgical trauma (including post-surgical cognitive dysfunction and spinal cord or brain stem injury), as well as the neurological aspects of disorders such as degenerative disk disease and sciatica.

In one aspect, provided is a compound of the present invention, or a

pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a CNS disorder in a mammal. CNS disorders include, but are not limited to, multiple sclerosis, Parkinson’s disease, Alzheimer’s disease, stroke, cerebral ischemia, retinal ischemia, post-surgical cognitive dysfunction, migraine, peripheral neuropathy/neuropathic pain, spinal cord injury, cerebral edema and head injury.

Cardiovascular Disorders

Cardiovascular phenotypes observed after targeted deletion of lysophospholipid receptors reveal important roles for lysophospholipid signaling in the development and maturation of blood vessels, formation of atherosclerotic plaques and maintenance of heart rate (Ishii, I. et al. Annu. Rev. Biochem. 73, 321-354, 2004). Angiogenesis, the formation of new capillary networks from pre-existing vasculature, is normally invoked in wound healing, tissue growth and myocardial angiogenesis after ischemic injury. Peptide growth factors ( e.g . vascular endothelial growth factor (VEGF)) and

lysophospholipids control coordinated proliferation, migration, adhesion, differentiation and assembly of vascular endothelial cells (VECs) and surrounding vascular smooth- muscle cells (VSMCs). In one aspect, dysregulation of the processes mediating angiogenesis leads to atherosclerosis, hypertension, tumor growth, rheumatoid arthritis and diabetic retinopathy (Osborne, N. and Stainier, D.Y. Annu. Rev. Physiol. 65, 23-43, 2003).

Downstream signaling pathways evoked by lysophospholipid receptors include Rac-dependent lamellipodia formation ( e.g . LPAi) and Rho-dependent stress-fiber formation (e.g. LPAi), which is important in cell migration and adhesion. Dysfunction of the vascular endothelium can shift the balance from vasodilatation to vasoconstriction and lead to hypertension and vascular remodeling, which are risk factors for atherosclerosis (Maguire, J.J. et ah, Trends Pharmacol. Sci. 26, 448-454, 2005).

LPA contributes to both the early phase (barrier dysfunction and monocyte adhesion of the endothelium) and the late phase (platelet activation and intra-arterial thrombus formation) of atherosclerosis, in addition to its overall progression. In the early phase, LPA from numerous sources accumulates in lesions and activates its cognate GPCRs (LPAi and LPA 3 ) expressed on platelets (Siess, W. Biochim. Biophys. Acta 1582, 204-215, 2002; Rother, E. et al. Circulation 108, 741-747, 2003). This triggers platelet shape change and aggregation, leading to intra-arterial thrombus formation and, potentially, myocardial infarction and stroke. In support of its atherogenic activity, LPA can also be a mitogen and motogen to VSMCs and an activator of endothelial cells and macrophages. In one aspect, mammals with cardiovascular disease benefit from LPA receptor antagonists that prevent thrombus and neointima plaque formation.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used to treat or prevent cardiovascular disease in mammal.

The term“cardiovascular disease,” as used herein refers to diseases affecting the heart or blood vessels or both, including but not limited to: arrhythmia (atrial or ventricular or both); atherosclerosis and its sequelae; angina; cardiac rhythm disturbances; myocardial ischemia; myocardial infarction; cardiac or vascular aneurysm; vasculitis, stroke; peripheral obstructive arteriopathy of a limb, an organ, or a tissue; reperfusion injury following ischemia of the brain, heart or other organ or tissue; endotoxic, surgical, or traumatic shock; hypertension, valvular heart disease, heart failure, abnormal blood pressure; shock; vasoconstriction (including that associated with migraines); vascular abnormality, inflammation, insufficiency limited to a single organ or tissue.. In one aspect, provided herein are methods for preventing or treating

vasoconstriction, atherosclerosis and its sequelae myocardial ischemia, myocardial infarction, aortic aneurysm, vasculitis and stroke comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof, or pharmaceutical composition or medicament which includes a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, provided herein are methods for reducing cardiac reperfusion injury following myocardial ischemia and/or endotoxic shock comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, provided herein are methods for reducing the constriction of blood vessels in a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, provided herein are methods for lowering or preventing an increase in blood pressure of a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

Inflammation

LPA has been shown to regulate immunological responses by modulating activities/functions of immune cells such as T-/B-lymphocytes and macrophages. In activated T cells, LPA activates IL-2 production/cell proliferation through LPAi (Gardell et al, TRENDS in Molecular Medicine Vol.l2 No.2 February 2006). Expression of LPA- induced inflammatory response genes is mediated by LPAi and LPA 3 {Biochem Biophys Res Commun. 363(4): 1001-8, 2007). In addition, LPA modulates the chemotaxis of inflammatory cells {Biochem Biophys Res Commun., 1993, 15; 193(2), 497). The proliferation and cytokine-secreting activity in response to LPA of immune cells ( J Imuunol. 1999, 162, 2049), platelet aggregation activity in response to LPA, acceleration of migration activity in monocytes, activation of NF-kB in fibroblast, enhancement of fibronectin-binding to the cell surface, and the like are known. Thus, LPA is associated with various inflammatory/immune diseases.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used to treat or prevent inflammation in a mammal. In one aspect, antagonists of LPAi and/or LPA 3 find use in the treatment or prevention of inflammatory/immune disorders in a mammal. In one aspect, the antagonist of LPAi is a compound of the present invention, or a pharmaceutically acceptable salt thereof.

Examples of inflammatory/immune disorders include psoriasis, rheumatoid arthritis, vasculitis, inflammatory bowel disease, dermatitis, osteoarthritis, asthma, inflammatory muscle disease, allergic rhinitis, vaginitis, interstitial cystitis, scleroderma, eczema, allogeneic or xenogeneic transplantation (organ, bone marrow, stem cells and other cells and tissues) graft rejection, graft-versus-host disease, lupus erythematosus, inflammatory disease, type I diabetes, pulmonary fibrosis, dermatomyositis, Sjogren's syndrome, thyroiditis ( e.g ., Hashimoto's and autoimmune thyroiditis), myasthenia gravis, autoimmune hemolytic anemia, multiple sclerosis, cystic fibrosis, chronic relapsing hepatitis, primary biliary cirrhosis, allergic conjunctivitis and atopic dermatitis.

Other Diseases, Disorders or Conditions

In accordance with one aspect, are methods for treating, preventing, reversing, halting or slowing the progression of LPA-dependent or LPA-mediated diseases or conditions once it becomes clinically evident, or treating the symptoms associated with or related to LPA-dependent or LPA-mediated diseases or conditions, by administering to the mammal a compound of the present invention, or a pharmaceutically acceptable salt thereof. In certain embodiments, the subject already has a LPA-dependent or LPA- mediated disease or condition at the time of administration, or is at risk of developing a LPA-dependent or LPA-mediated disease or condition.

In certain aspects, the activity of LPAi in a mammal is directly or indirectly modulated by the administration of (at least once) a therapeutically effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof. Such modulation includes, but is not limited to, reducing and/or inhibiting the activity of LPAi. In additional aspects, the activity of LPA in a mammal is directly or indirectly modulated, including reducing and/or inhibiting, by the administration of (at least once) a therapeutically effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof. Such modulation includes, but is not limited to, reducing and/or inhibiting the amount and/or activity of a LPA receptor. In one aspect, the LPA receptor is LPAi.

In one aspect, LPA has a contracting action on bladder smooth muscle cell isolated from bladder, and promotes growth of prostate-derived epithelial cell (J.

Urology, 1999, 162, 1779-1784; J. Urology , 2000, 163, 1027-1032). In another aspect, LPA contracts the urinary tract and prostate in vitro and increases intraurethral pressure in vivo (WO 02/062389).

In certain aspects, are methods for preventing or treating eosinophil and/or basophil and/or dendritic cell and/or neutrophil and/or monocyte and/or T-cell recruitment comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In certain aspects, are methods for the treatment of cystitis, including,

e.g, interstitial cystitis, comprising administering at least once to the mammal a therapeutically effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In accordance with one aspect, methods described herein include the diagnosis or determination of whether or not a patient is suffering from a LPA-dependent or LPA- mediated disease or condition by administering to the subject a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof, and determining whether or not the patient responds to the treatment.

In one aspect provided herein are compounds of the present invention, pharmaceutically acceptable salts, pharmaceutically acceptable prodrugs, and

pharmaceutically acceptable solvates thereof, which are antagonists of LPAi, and are used to treat patients suffering from one or more LPA-dependent or LPA-mediated conditions or diseases, including, but not limited to, lung fibrosis, kidney fibrosis, liver fibrosis, scarring, asthma, rhinitis, chronic obstructive pulmonary disease, pulmonary hypertension, interstitial lung fibrosis, arthritis, allergy, psoriasis, inflammatory bowel disease, adult respiratory distress syndrome, myocardial infarction, aneurysm, stroke, cancer, pain, proliferative disorders and inflammatory conditions. In some embodiments, LPA-dependent conditions or diseases include those wherein an absolute or relative excess of LPA is present and/or observed.

In any of the aforementioned aspects the LPA-dependent or LPA-mediated diseases or conditions include, but are not limited to, organ fibrosis, asthma, allergic disorders, chronic obstructive pulmonary disease, pulmonary hypertension, lung or pleural fibrosis, peritoneal fibrosis, arthritis, allergy, cancer, cardiovascular disease, ult respiratory distress syndrome, myocardial infarction, aneurysm, stroke, and cancer.

In one aspect, a compound of the present invention, or a pharmaceutically acceptable salt thereof, is used to improve the corneal sensitivity decrease caused by corneal operations such as laser-assisted in situ keratomileusis (LASIK) or cataract operation, corneal sensitivity decrease caused by corneal degeneration, and dry eye symptom caused thereby.

In one aspect, presented herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the treatment or prevention of ocular inflammation and allergic conjunctivitis, vernal keratoconjunctivitis, and papillary conjunctivitis in a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, presented herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the treatment or prevention of Sjogren disease or inflammatory disease with dry eyes in a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, LPA and LPA receptors ( e.g . LPAi) are involved in the

pathogenesis of osteoarthritis (Kotani et al, Hum. Mol. Genet., 2008, 77, 1790-1797). In one aspect, presented herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the treatment or prevention of osteoarthritis in a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, LPA receptors (e.g. LPAi, LPA 3 ) contribute to the pathogenesis of rheumatoid arthritis (Zhao et al, Mol. Pharmacol ., 2008, 73(2), 587-600). In one aspect, presented herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the treatment or prevention of rheumatoid arthritis in a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, LPA receptors ( e.g . LPAi) contribute to adipogenesis. (Simon et al , J.Biol. Chem., 2005, vol. 280, no. 15, p.14656). In one aspect, presented herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the promotion of adipose tissue formation in a mammal comprising administering at least once to the mammal an effective amount of at least one compound of the present invention, or a pharmaceutically acceptable salt thereof. a. In Vitro Assays

The effectiveness of compounds of the present invention as LPA1 inhibitors can be determined in an LPA1 functional antagonist assay as follows:

Chinese hamster ovary cells overexpressing human LPA1 were plated overnight (15,000 cells/well) in poly-D4ysine coated 384-well microplates (Greiner bio-one,

Cat#781946) in DMEM/F12 medium (Gibco, Cat#! 1039). Following overnight culture, cells were loaded with calcium indicator dye (AAT Bioquest Inc, Cat# 34601) for 30 minutes at 37 °C. The cells were then equilibrated to room temperature for 30 minutes before the assay. Test compounds solubilized in DMSO were transferred to 384 well non binding surface plates (Coming, Cat# 3575) using the Labcyte Echo acoustic dispense and diluted with assay buffer [IX HBSS with calcium/magnesium (Gibco Cat# 14025- 092), 20 mM HEPES (Gibco Cat# 15630-080) and 0.1% fatty acid free BSA (Sigma Cat# A9205)] to a final concentration of 0.5% DMSO. Diluted compounds were added to the cells by FDSS6000 (Hamamatsu) at final concentrations ranging from 0.08 nM to 5 mM. and were then incubated for 20 min at room temperature at which time LPA (Avanti Polar Lipids Cat#857l30C) was added at final concentrations of 10 nM to stimulate the cells. The compound ICso value was defined as the concentration of test compound which inhibited 50% of the calcium flux induced by LPA alone. ICso values were determined by fitting data to a 4-parameter logistic equation (GraphPad Prism, San Diego CA). b. In Vivo Assays

LPA Challenge with plasma histamine evaluation.

Compound is dosed orally p.o. 2 hours to CD-l female mice prior to the LPA challenge. The mice are then dosed via tail vein (IV) with 0.15 mL of LPA in 0. l%BSA/ PBS (2 pg/pL). Exactly 2 minutes following the LPA challenge, the mice are euthanized by decapitation and the trunk blood is collected. These samples are collectively centrifuged and individual 75 pL samples are frozen at -20°C until the time of the histamine assay.

The plasma histamine analysis was run by standard EIA (Enzyme Immunoassay) methods. Plasma samples were thawed and diluted 1 :30 in 0.1% BSA in PBS. The EIA protocol for histamine analysis as outlined by the manufacturer was followed (Histamine EIA, Oxford Biomedical Research, EA#3 l).

The LPA used in the assay is formulated as follows: LPA (l-oleoyl-2-hydroxy-sn- glycero-3 -phosphate (sodium salt), 857130P, Avanti Polar Lipids) is prepared in

0. l%BSA/PBS for total concentration of 2 pg/pL. 13 mg of LPA is weighed and 6.5 mL 0. l%BSA added, vortexed and sonicated for ~l hour until a clear solution is achieved.

V. PHARMACEUTICAL COMPOSITIONS, FORMULATIONS AND

COMBINATIONS

In some embodiments, provided is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention, or a

pharmaceutically acceptable salt thereof. In some embodiments, the pharmaceutical composition also contains at least one pharmaceutically acceptable inactive ingredient.

In some embodiments, provided is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention, or a

pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable inactive ingredient. In one aspect, the pharmaceutical composition is formulated for intravenous injection, subcutaneous injection, oral administration, inhalation, nasal administration, topical administration, ophthalmic administration or otic administration.

In some embodiments, the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a suspension, a gel, a colloid, a dispersion, a suspension, a solution, an emulsion, an ointment, a lotion, an eye drop or an ear drop.

In some embodiments, the pharmaceutical composition further comprises one or more additional therapeutically active agents selected from: corticosteroids ( e.g ., dexamethasone or fluticasone), immunosuppresants (e.g., tacrolimus & pimecrolimus), analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchodilators, leukotriene receptor antagonists (e.g, montelukast or zafirlukast), leukotriene formation inhibitors, monoacylglycerol kinase inhibitors, phospholipase Ai inhibitors, phospholipase A 2 inhibitors, and lysophospholipase D (lysoPLD) inhibitors, autotaxin inhibitors, decongestants, antihistamines (e.g, loratidine), mucolytics, anticholinergics, antitussives, expectorants, anti-infectives (e.g, fusidic acid, particularly for treatment of atopic dermatitis), anti-fungals (e.g, clotriazole, particularly for atopic dermatitis), anti-IgE antibody therapies (e.g, omalizumab), b-2 adrenergic agonists (e.g, albuterol or salmeterol), other PGD2 antagonists acting at other receptors such as DP antagonists, PDE4 inhibitors (e.g, cilomilast), drugs that modulate cytokine production, e.g, TACE inhibitors, drugs that modulate activity of Th2 cytokines IL-4 & IL-5 (e.g, blocking monoclonal antibodies & soluble receptors), PPARy agonists (e.g, rosiglitazone and pioglitazone), 5 -lipoxygenase inhibitors (e.g, zileuton).

In some embodiments, the pharmaceutical composition further comprises one or more additional anti-fibrotic agents selected from pirfenidone, nintedanib, thalidomide, carlumab, FG-3019, fresolimumab, interferon alpha, lecithinized superoxide dismutase, simtuzumab, tanzisertib, tralokinumab, hu3G9, AM- 152, IFN-gamma-lb, IW-001, PRM- 151, PXS-25, pentoxifylline/N-acetyl-cysteine, pentoxifylline/vitamin E, salbutamol sulfate, [Sar9,Met(02)l l]-Substance P, pentoxifylline, mercaptamine bitartrate, obeticholic acid, aramchol, GFT-505, eicosapentaenoic acid ethyl ester, metformin, metreleptin, muromonab-CD3, oltipraz, IMM-124-E, MK-4074, PX-102, RO-5093151.

In some embodiments, provided is a method comprising administering a compound of the present invention, or a pharmaceutically acceptable salt thereof, to a human with a LPA- dependent or LPA-mediated disease or condition. In some embodiments, the human is already being administered one or more additional therapeutically active agents other than a compound of the present invention, or a pharmaceutically acceptable salt thereof. In some embodiments, the method further comprises administering one or more additional therapeutically active agents other than a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In some embodiments, the one or more additional therapeutically active agents other than a compound of the present invention, or a pharmaceutically acceptable salt thereof, are selected from: corticosteroids (e.g,. dexamethasone or fluticasone), immunosuppresants (e.g., tacrolimus & pimecrolimus), analgesics, anti-cancer agent, anti-inflammatories, chemokine receptor antagonists, bronchodilators, leukotriene receptor antagonists (e.g, montelukast or zafirlukast), leukotriene formation inhibitors, monoacylglycerol kinase inhibitors, phospholipase Ai inhibitors, phospholipase A 2 inhibitors, and lysophospholipase D (lysoPLD) inhibitors, autotaxin inhibitors, decongestants, antihistamines (e.g, loratidine), mucolytics, anticholinergics, antitussives, expectorants, anti-infectives (e.g, fusidic acid, particularly for treatment of atopic dermatitis), anti-fungals (e.g, clotriazole, particularly for atopic dermatitis), anti-IgE antibody therapies (e.g, omalizumab), b-2 adrenergic agonists (e.g, albuterol or salmeterol), other PGD2 antagonists acting at other receptors such as DP antagonists, PDE4 inhibitors (e.g, cilomilast), drugs that modulate cytokine production, e.g. TACE inhibitors, drugs that modulate activity of Th2 cytokines IL-4 & IL-5 (e.g, blocking monoclonal antibodies & soluble receptors), PPARy agonists (e.g, rosiglitazone and pioglitazone), 5 -lipoxygenase inhibitors (e.g, zileuton).

In some embodiments, the one or more additional therapeutically active agents other than a compound of the present invention, or a pharmaceutically acceptable salt thereof, are other anti-fibrotic agents selected from pirfenidone, nintedanib, thalidomide, carlumab, FG-3019, fresolimumab, interferon alpha, lecithinized superoxide dismutase, simtuzumab, tanzisertib, tralokinumab, hu3G9, AM- 152, IFN-gamma-lb, IW-001, PRM- 151, PXS-25, pentoxifylline/N-acetyl-cysteine, pentoxifylline/vitamin E, salbutamol sulfate, [Sar9,Met(02)l l]-Substance P, pentoxifylline, mercaptamine bitartrate, obeticholic acid, aramchol, GFT-505, eicosapentyl ethyl ester, metformin, metreleptin, muromonab-CD3, oltipraz, IMM-124-E, MK-4074, PX-102, RO-5093151.

In some embodiments, the one or more additional therapeutically active agents other than a compound of the present invention, or a pharmaceutically acceptable salt thereof, are selected from ACE inhibitors, ramipril, All antagonists, irbesartan, anti- arrythmics, dronedarone, PPARa activators, PPARy activators, pioglitazone, rosiglitazone, prostanoids, endothelin receptor antagonists, elastase inhibitors, calcium antagonists, beta blockers, diuretics, aldosterone receptor antagonists, eplerenone, renin inhibitors, rho kinase inhibitors, soluble guanylate cyclase (sGC) activators, sGC sensitizers, PDE inhibitors, PDE5 inhibitors, NO donors, digitalis drugs, ACE/NEP inhibitors, statins, bile acid reuptake inhibitors, PDGF antagonists, vasopressin antagonists, aquaretics, NHE1 inhibitors, Factor Xa antagonists, Factor XHIa antagonists, anticoagulants, anti-thrombotics, platelet inhibitors, profibroltics, thrombin-activatable fibrinolysis inhibitors (TAFI), PAI-l inhibitors, coumarins, heparins, thromboxane antagonists, serotonin antagonists, COX inhibitors, aspirin, therapeutic antibodies, GPIIb/IIIa antagonists, ER antagonists, SERMs, tyrosine kinase inhibitors, RAF kinase inhibitors, p38 MAPK inhibitors, pirfenidone, multi-kinase inhibitors, nintedanib, sorafenib.

In some embodiments, the one or more additional therapeutically active agents other than a compound of the present invention, or a pharmaceutically acceptable salt thereof, are selected from Gremlin- 1 mAb, PA1-1 mAb, Promedior (PRM-151;

recombinant human Pentraxin-2); FGF21, TGF antagonists, anb6 & anb pan antagonists; FAR inhibitors, TG2 inhibitors, LOXL2 inhibitors, NOX4 inhibitors, MGAT2 inhibitors, GPR120 agonists.

Pharmaceutical formulations described herein are administrable to a subject in a variety of ways by multiple administration routes, including but not limited to, oral, parenteral ( e.g ., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical or transdermal administration routes. The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.

In some embodiments, the compound of the present invention, or a

pharmaceutically acceptable salt thereof, is administered orally.

In some embodiments, the compound of the present invention, or a

pharmaceutically acceptable salt thereof, is administered topically. In such embodiments, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is formulated into a variety of topically administrable compositions, such as solutions, suspensions, lotions, gels, pastes, shampoos, scrubs, rubs, smears, medicated sticks, medicated bandages, balms, creams or ointments. Such pharmaceutical compounds can contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives. In one aspect, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is administered topically to the skin.

In another aspect, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is administered by inhalation. In one embodiment, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is administered by inhalation that directly targets the pulmonary system.

In another aspect, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is formulated for intranasal administration. Such formulations include nasal sprays, nasal mists, and the like.

In another aspect, the compound of the present invention, or a pharmaceutically acceptable salt thereof, is formulated as eye drops.

In another aspect is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a disease, disorder or conditions in which the activity of at least one LPA receptor contributes to the pathology and/or symptoms of the disease or condition. In one embodiment of this aspect, the LPA is selected from LPAi, LPA2, LPA 3 , LPA 4 , LPAs and LPA 6 . In one aspect, the LPA receptor is LPAi. In one aspect, the disease or condition is any of the diseases or conditions specified herein.

In any of the aforementioned aspects are further embodiments in which: (a) the effective amount of the compound of the present invention, or a pharmaceutically acceptable salt thereof, is systemically administered to the mammal; and/or (b) the effective amount of the compound is administered orally to the mammal; and/or (c) the effective amount of the compound is intravenously administered to the mammal; and/or (d) the effective amount of the compound is administered by inhalation; and/or (e) the effective amount of the compound is administered by nasal administration; or and/or (f) the effective amount of the compound is administered by injection to the mammal; and/or (g) the effective amount of the compound is administered topically to the mammal; and/or (h) the effective amount of the compound is administered by ophthalmic administration; and/or (i) the effective amount of the compound is administered rectally to the mammal; and/or (j) the effective amount is administered non-systemically or locally to the mammal.

In any of the aforementioned aspects are further embodiments comprising single administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered once; (ii) the compound is administered to the mammal multiple times over the span of one day; (iii) continually; or (iv) continuously.

In any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours. In further or alternative embodiments, the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed. In one embodiment, the length of the drug holiday varies from 2 days to 1 year.

Also provided is a method of inhibiting the physiological activity of LPA in a mammal comprising administering a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof to the mammal in need thereof.

In one aspect, provided is a medicament for treating a LPA-dependent or LPA- mediated disease or condition in a mammal comprising a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In some cases disclosed herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a LPA-dependent or LPA-mediated disease or condition.

In some cases disclosed herein is the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the treatment or prevention of a LPA- dependent or LPA-mediated disease or condition. In one aspect, is a method for treating or preventing a LPA-dependent or LPA- mediated disease or condition in a mammal comprising administering a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof.

In one aspect, LPA-dependent or LPA-mediated diseases or conditions include, but are not limited to, fibrosis of organs or tissues, scarring, liver diseases, dermatological conditions, cancer, cardiovascular disease, respiratory diseases or conditions,

inflammatory disease, gastrointestinal tract disease, renal disease, urinary tract-associated disease, inflammatory disease of lower urinary tract, dysuria, frequent urination, pancreas disease, arterial obstruction, cerebral infarction, cerebral hemorrhage, pain, peripheral neuropathy, and fibromyalgia.

In one aspect, the LPA-dependent or LPA-mediated disease or condition is a respiratory disease or condition. In some embodiments, the respiratory disease or condition is asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, pulmonary arterial hypertension or acute respiratory distress syndrome.

In some embodiments, the LPA-dependent or LPA-mediated disease or condition is selected from idiopathic pulmonary fibrosis; other diffuse parenchymal lung diseases of different etiologies including iatrogenic drug-induced fibrosis, occupational and/or environmental induced fibrosis, granulomatous diseases (sarcoidosis, hypersensitivity pneumonia), collagen vascular disease, alveolar proteinosis, langerhans cell

granulomatosis, lymphangioleiomyomatosis, inherited diseases (Hermansky-Pudlak Syndrome, tuberous sclerosis, neurofibromatosis, metabolic storage disorders, familial interstitial lung disease); radiation induced fibrosis; chronic obstructive pulmonary disease (COPD); scleroderma; bleomycin induced pulmonary fibrosis; chronic asthma; silicosis; asbestos induced pulmonary fibrosis; acute respiratory distress syndrome (ARDS); kidney fibrosis; tubulointerstitium fibrosis; glomerular nephritis; focal segmental glomerular sclerosis; IgA nephropathy; hypertension; Alport; gut fibrosis; liver fibrosis; cirrhosis; alcohol induced liver fibrosis; toxic/drug induced liver fibrosis;

hemochromatosis; nonalcoholic steatohepatitis (NASH); biliary duct injury; primary biliary cirrhosis; infection induced liver fibrosis; viral induced liver fibrosis; and autoimmune hepatitis; corneal scarring; hypertrophic scarring; Duputren disease, keloids, cutaneous fibrosis; cutaneous scleroderma; spinal cord injury/fibrosis; myelofibrosis; vascular restenosis; atherosclerosis; arteriosclerosis; Wegener's granulomatosis;

Peyronie's disease, chronic lymphocytic leukemia, tumor metastasis, transplant organ rejection, endometriosis, neonatal respiratory distress syndrome and neuropathic pain.

In one aspect, the LPA-dependent or LPA-mediated disease or condition is described herein.

In one aspect, provided is a method for the treatment or prevention of organ fibrosis in a mammal comprising administering a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof to a mammal in need thereof.

In one aspect, the organ fibrosis comprises lung fibrosis, renal fibrosis, or hepatic fibrosis.

In one aspect, provided is a method of improving lung function in a mammal comprising administering a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof to the mammal in need thereof. In one aspect, the mammal has been diagnosed as having lung fibrosis.

In one aspect, compounds disclosed herein are used to treat idiopathic pulmonary fibrosis (usual interstitial pneumonia) in a mammal.

In some embodiments, compounds disclosed herein are used to treat diffuse parenchymal interstitial lung diseases in mammal: iatrogenic drug induced,

occupational/environmental (Farmer lung), granulomatous diseases (sarcoidosis, hypersensitivity pneumonia), collagen vascular disease (scleroderma and others), alveolar proteinosis, langerhans cell granulonmatosis, lymphangioleiomyomatosis, Hermansky- Pudlak Syndrome, Tuberous sclerosis, neurofibromatosis, metabolic storage disorders, familial interstitial lung disease.

In some embodiments, compounds disclosed herein are used to treat post- transplant fibrosis associated with chronic rejection in a mammal: Bronchiolitis obliterans for lung transplant.

In some embodiments, compounds disclosed herein are used to treat cutaneous fibrosis in a mammal: cutaneous scleroderma, Dupuytren disease, keloids.

In one aspect, compounds disclosed herein are used to treat hepatic fibrosis with or without cirrhosis in a mammal: toxic/drug induced (hemochromatosis), alcoholic liver disease, viral hepatitis (hepatitis B virus, hepatitis C virus, HCV), nonalcoholic liver disease (NAFLD, NASH), metabolic and auto-immune disease.

In one aspect, compounds disclosed herein are used to treat renal fibrosis in a mammal: tubulointerstitium fibrosis, glomerular sclerosis.

In any of the aforementioned aspects involving the treatment of LPA dependent diseases or conditions are further embodiments comprising administering at least one additional agent in addition to the administration of a compound having the structure of the present invention, or a pharmaceutically acceptable salt thereof. In various embodiments, each agent is administered in any order, including simultaneously.

In any of the embodiments disclosed herein, the mammal is a human.

In some embodiments, compounds provided herein are administered to a human.

In some embodiments, compounds provided herein are orally administered.

In some embodiments, compounds provided herein are used as antagonists of at least one LPA receptor. In some embodiments, compounds provided herein are used for inhibiting the activity of at least one LPA receptor or for the treatment of a disease or condition that would benefit from inhibition of the activity of at least one LPA receptor.

In one aspect, the LPA receptor is LPAi.

In other embodiments, compounds provided herein are used for the formulation of a medicament for the inhibition of LPAi activity.

Articles of manufacture, which include packaging material, a compound of the present invention, or a pharmaceutically acceptable salt thereof, within the packaging material, and a label that indicates that the compound or composition, or pharmaceutically acceptable salt, tautomers, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, is used for inhibiting the activity of at least one LPA receptor, or for the treatment, prevention or amelioration of one or more symptoms of a disease or condition that would benefit from inhibition of the activity of at least one LPA receptor, are provided.

VI. GENERAL SYNTHESIS INCLUDING SCHEMES

The compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis. The compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below. The reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention.

It will also be recognized that another major consideration in the planning of any synthetic route in this field is the judicious choice of the protecting group used for protection of the reactive functional groups present in the compounds described in this invention. An authoritative account describing the many alternatives to the trained practitioner is Greene et ah, (. Protective Groups in Organic Synthesis , Fourth Edition, Wiley-Interscience (2006)).

The compounds of the present invention may be prepared by the exemplary processes described in the following schemes and working examples, as well as relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear herein after and in the working examples.

Protection and deprotection in the processes below may be carried out by procedures generally known in the art (see, for example, Wuts, P.G.M., Greene’s Protective Groups in Organic Synthesis , 5th Edition, Wiley (2014)). General methods of organic synthesis and functional group transformations are found in: Trost, B.M. et ah, Eds.,

Comprehensive Organic Synthesis: Selectivity, Strategy & Efficiency in Modern Organic Chemistry , Pergamon Press, New York, NY (1991); Smith, M.B. et ah, March's

Advanced Organic Chemistry: Reactions, Mechanisms, and Structure. 7th Edition, Wiley, New York, NY (2013); Katritzky, A.R. et ah, Eds., Comprehensive Organic Functional Group Transformations II, 2nd Edition, Elsevier Science Inc., Tarrytown, NY (2004); Larock, R.C., Comprehensive Organic Transformations , 2 nd Edition, Wiley-VCH, New York, NY (1999), and references therein. Scheme 1 describes the synthesis of O-carbamoyl isoxazole aryl(heteroaryl)oxy- cyclohexyl acids 13. A 4-halo (preferably bromo) phenyl or azine (e.g. pyridine) benzoic acid 1 is converted to the corresponding acid chloride (e.g. with SOCh or oxalyl chloride/ catalytic DMF). This acid chloride intermediate is reacted with a substituted b-enamino- ester 2 followed by condensation with hydroxylamine to furnish the corresponding 5- halo(hetero)aryl-isoxazole 4-carboxylate ester 3. Deprotection of ester 3 followed by reduction of the resulting acid (e.g. directly with diborane or by a 2-step procedure by reacting the acid with an alkyl chloroformate followed by reduction with, e.g. NaBH 4 , at low temperature) and protection of the resulting alcohol provides the 5-halo(hetero)aryl- isoxazole protected alcohol 4. Reaction of the haloaryl- or haloheteroaryl-isoxazoles 4 with bis(pinacolato)diboron in the presence of an appropriate palladium catalyst (e.g. Ishiyama, T. et al, J Org. Chem. 1995, 60, 7508-7510) provides the corresponding pinacol boronate 5, which is then oxidized with hydrogen peroxide to give the

corresponding phenol or hydroxyheteroarene 6 (e.g. Fukumoto, S. et al,

WO 2012137982). Reaction of phenol/hydroxyheteroarene 6 with a hydroxy cyclopentyl ester 7 under Mitsunobu reaction conditions (Kumara Swamy, K. C., Chem. Rev., 2009, 109 , 2551-2651) furnishes the corresponding isoxazole cycloalkyl ether ester 8.

Deprotection of the hydoxymethylisoxazole 8 provides the cyclopentyl ester isoxazole alcohol 9. Isoxazole alcohol 9 is reacted with an activating group in the presence of base (e.g. 4-nitrophenyl chloroformate) to give the 4-nitrophenyl carbonate 10, which is reacted with an appropriate amine 11 (R 3 R 4 NH) to give the isoxazole O-carbamate 12. Deprotection of cyclopentyl ester 12 then provides the isoxazole carbamate cyclopentyl acids 13.

Scheme 1

Scheme 2 describes the synthesis of N-carbamoyl isoxazole-aryloxy cyclopentyl acids 19. Deprotection of the isoxazole ester 3 provides the isoxazole acid 14. Acid 14 is subjected to Curtius rearrangement conditions (e.g. Pt PONri) to give the intermediate isocyanate, which is reacted in situ with an appropriate alcohol R 4 -OH to give the isoxazole NH-carbamate 15. Reaction of the bromoaryl- or bromoheteroaryl-isoxazoles 15 with bis(pinacolato)diboron in the presence of an appropriate palladium catalyst (e.g. Ishiyama, T. et al, J. Org. Chem. 1995, 60, 7508-7510) provides the corresponding pinacol boronate 16, which is then oxidized with hydrogen peroxide to give the corresponding phenol or hydroxyheteroarene 17 (e.g. Fukumoto, S. et al,

WO 2012137982). Reaction of phenol/hydroxyheteroarene 17 with a hydroxy

cyclopentyl ester 7 under Mitsunobu reaction conditions (Kumara Swamy, K. C., Chem. Rev., 2009, 109, 2551-2651) furnishes the corresponding isoxazole cyclopentyl ether ester 18. Deprotection of cyclopentyl ester 18 provides the isoxazole N-carbamoyl cyclopentyl acids 19.

Scheme 2

Scheme 3 describes the synthesis of N-carbamoyl isoxazole-aryloxy cyclopentyl acids 23. Isoxazole alcohol 9 is reacted with PBn (or another mild brominating system such as CBr4/Ph 3 P) to give the corresponding bromide 20. Displacement of isoxazole bromide 20 with NaN 3 (or another azide equivalent reagent) provides the corresponding isoxazole azide, which undergoes reduction (e.g. Staudinger reduction with Ph 3 P/water) to afford isoxazole amine 21. Isoxazole amine 21 is reacted with an appropriate acylating agent (e.g. a chloroformate 22 or a 4-nitrophenylcarbonate) to provide the cyclopentyl isoxazole N-H carbamate ester, which is then deprotected to give the NH-carbamoyl methyl isoxazole-aryloxy cyclopentyl acids 23.

Scheme 3

Scheme 4 describes the synthesis of carbamoyloxymethyl triazole-aryloxy cyclopentyl acids 34. A dihalo (e.g. dibromo) phenyl or azine (e.g. pyridine) 24 is coupled with an appro-priately protected (e.g. as a tetrahydropyranyl ether) propargyl alcohol 25 under Sonogashira conditions (e.g. Alper, P. et al, WO 2008097428) to give the corresponding bromo-aryl or bromo-heteroaryl protected propargyl alcohol 26.

Thermal reaction of alkyne 3 with an alkyl azide R 5 N 3 (with or without an appropriate catalyst; Qian, Y. et al, J. Med. Chem ., 2012, 55, 7920-7939 or Boren, B. C., et al., J. Am. Chem. Soc., 2008, 130, 8923-8930) provides the corresponding regioisomeric protected hydroxylmethyl-triazoles, from which the desired triazole regioisomer 27 can be isolated. Reaction of the bromoaryl- or bromoheteroaryl-triazoles 5 with bis(pinacolato)diboron in the presence of an appropriate palladium catalyst (e.g. Ishiyama, T. et al, J Org. Chem. 1995, 60, 7508-7510) provides the corresponding pinacol boronate 28, which is then oxidized with hydrogen peroxide to give the corresponding phenol or hydroxyheteroarene 29 (e.g. Fukumoto, S. et al, WO 2012137982). Reaction of phenol/hydroxyheteroarene 7 with a 3-hydroxy cyclopentyl ester 7 under Mitsunobu reaction conditions furnishes the corresponding triazole cycloalkyl ether ester 30. Deprotection of the hydoxytriazole 30 provides the triazole alcohol 31, which is then reacted with 4-nitrophenyl chloroformate in the presence of an appropriate base to give the corresponding triazole 4-nitrophenyl carbonate 32. The triazole 4-nitrophenyl carbonate 32 is then reacted with an amine 11 in the presence of an appropriate base to give the triazole carbamate 33, which then undergoes ester deprotection to give the desired carbamoyloxymethyl triazole-aryloxy cyclopentyl acids 34.

Scheme 4

For the specific example of analogs 34, where R 5 = CFb (Scheme 4A), trimethyl silyl azide (Qian, Y. et al, J. Med. Chem ., 2012, 55 , 7920-7939) is used instead of an alkyl azide for the cycloaddition to the protected hydroxyalkyl alkyne 26. This cycloaddition can occur under either thermal or, preferably, under transition-metal catalyzed conditions (e.g. Boren, B.C. et. al., J. Am. Chem. Soc., 2008, 130, 8923-8930; Ramasamy, S., et al., Org. Process Res. Dev., 2018, 22, 880-887) to provide the desired triazole regioisomer 35 as the major product. The trimethyl silyl group is subsequently removed under standard desilylation conditions ( e.g . B NF, as in Qian, Y. et al, J. Med. Chem., 2012, 55, 7920-7939).

Scheme 4A

Scheme 5 describes the synthesis of N-carbamoyl triazole-aryloxy cyclopentyl acids 39. Triazole alcohol 31 is oxidized to the triazole acid 37 (e.g. directly to the acid with pyridinium dichromate or via a 2-step procedure via the aldehyde [Swem oxidation or Dess-Martin periodinane followed by NaClCh oxidation to the acid, e.g. Lindgren, B. O., Acta Chem. Scand. 1973, 27, 888]). Curtius rearrangement of acid 37 in the presence of an alcohol R 4 -OH provides the triazole NH-carbamate 38. Deprotection of the triazole NH-carbamate ester 38 provides the cyclopentyl triazole NH-carbamoyl acids 39. Scheme 5

N 3 R OH

Scheme 6 describes the synthesis of triazole N-carbamoyl-aryloxy cyclopentyl acids 42. Triazole alcohol 31 is reacted with a brominating agent (e.g. PBn or

CBn/Pt P) to give the corresponding bromide 40. Displacement of bromide 40 with NaN3 (or other appropriate azide reagents) gives the corresponding azide, which undergoes reduction (e.g. Staudinger reduction with PtnP/FbO) to afford triazole amine 41. Amine 41 is then reacted with an appropriate chlorformate 22 (or the corresponding 4-nitrophenyl carbonate) in the presence of an appropriate base to give the corresponding NH-carbamate. Ester deprotection of this triazole N-H carbamate ester provides the desired triazole-carbamate cyclopentyl acids 42.

Scheme 6

Scheme 7 describes the synthesis of pyrazole carbamate cyclopentyl acids 52. A pyrazole 5-carboxylic acid 43 is reduced (e.g. by a 2 step, l-pot reaction via reaction with an alkyl chloroformate followed by low-temperature reduction with NaBH 4 , or directly with diborane) to the corresponding pyrazole alcohol, which is then protected to give pyrazole intermediate 44. Halogenation of 44 occurs preferentially at the 4-pyrazole position to give halopyrazole 45, which is then subjected to a Suzuki -Miy aura cross- coupling reaction with an appropriately substituted 4-hydroxy-aryl/heteroaryl boronate 46 to provide the corresponding 4-hydroxy-aryl(heteroaryl)-pyrazole 47. Reaction of phenol/hydroxyheteroarene 47 with a hydroxy cyclopentyl ester 7 under Mitsunobu reaction conditions (Kumara Swamy, K. C., Chem. Rev., 2009, 109, 2551-2651) furnishes the corresponding pyrazole cyclopentyl ester 48. Deprotection of 48 provides the pyrazole alcohol 49. Reaction of pyrazole alcohol 49 with 4-nitro-phenyl chloroformate provides pyrazole 4-nitrophenyl carbonate 50, which undergoes reaction with an appropriate amine 12 to provide the corresponding pyrazole carbamate 51. Deprotection of pyrazole carbamate cyclopentyl ester 51 provides the pyrazole carbamate cyclopentyl acids 52. Scheme 7

o

Halogenation

N-N N-N

2) Reduction R Sa

R 5a

3) Protect Alcohol

43 44 45

Scheme 8 describes the synthesis of pyrazole N-linked carbamate cyclohexyl acids 55. The cyclopentyl ether pyrazole-alcohol 49 is oxidized to the pyrazole carboxylic acid 53 (e.g. directly to the acid with pyridinium dichromate or via a 2-step procedure via the aldehyde [Swern oxidation or Dess-Martin periodinane followed by NaClCk oxidation to the acid, e.g. Lindgren, B. O., Acta Chem. Scand. 1973, 27, 888]). Curtius rearrangement of pyrazole acid 53 in the presence of an appropriate alcohol R 4 - OH provides the pyrazole NH-carbamate 54. Deprotection of the pyrazole NH- carbamate ester 54 provides the pyrazole NH-carbamate cyclopentyl acids 55.

Scheme 8

Scheme 9 describes the synthesis of N-carbamoyl pyrazole-aryloxy cyclopentyl acids 58. Pyrazole alcohol 52 is reacted with PBn (or another mild brominating system such as CBr 4 / Ph 3 P) to give the corresponding bromide 56. Displacement of pyrazole bromide 56 with NaN 3 (or other azide equivalent reagents) provides the corresponding pyrazole azide which undergoes reduction (e.g. Staudinger reduction with Ph 3 P/water) to afford pyrazole amine 57. Pyrazole amine 57 is reacted with an appropriate acylating agent (e.g. chloroformate 22 or a 4-nitrophenyl carbonate) to provide the corresponding pyrazole N-H carbamate ester, which is deprotected to give the N-carbamoyl pyrazole- aryloxy cyclopentyl acids 19. Scheme 9

Scheme 10 describes the synthesis of: 1) amino-azine isoxazole aryloxy cyclopentyl acids 61, 2) amino-azine triazole aryloxy cyclopentyl acids 63 and 3) amino- azine pyrazole aryloxy cyclopentyl acids 65, wherein one of A 1 , A 2 and A 3 is N and the other two are CR. For instance, isoxazole amine 21 is reacted with halo-azine 59 in the presence of an appropriate base or via Pd catalyzed amination to give the isoxazole amino-azine 60, which is then subjected to ester deprotection to provide the desired amino-azine isoxazole-aryloxy cyclopentyl acids 62. The corresponding triazole amino- azine acids 63 and the pyrazole amino-azine acids 65 are synthesized from the triazole amine 41 and the pyrazole amine 57 respectively using the same general route as described for the preparation of 61 from 21. Scheme 10

, R

57 64 65

Scheme 10A describes an alternative synthesis of triazole amino azine acids 63. Triazole bromide 41 is reacted with an appropriate amino-azine 66 in the presence of base (e.g. NaH, etc.,) to afford the corresponding triazole amino-azine 62, which is subjected to ester deprotection to give the desired triazole amino-azine cyclopentyl acids 63.

Similarly, the corresponding isoxazole bromide 20 and the pyrazole bromide 56 can be converted to the corresponding amino-azine isoxazole acids 61 and amino-azine pyrazole acids 65 respectively by the same two-step sequence as described for the synthesis of triazole acids 63 from triazole bromide 41. Scheme 10A

Scheme 11 describes an alternative synthesis of triazole amino-azine cyclopentyl acids 63. The bromo-triazole 36 is deprotected to give the alcohol, which is converted to the corresponding bromide 67 (using e.g. PBn or CBn/PhiP). Bromide 67 is converted to the amine 68 via a 2-step sequence as described in Scheme 6: 1) NaN3 displacement of the bromide, 2) Staudinger reduction of the azide (PI13P/H2O). Amine 68 is then subjected to a transition -metal-catalyzed cross-coupling (e.g. palladium-mediated) with an appropriate halo-azine 59 to furnish the triazole amino-azine 69. Conversion of the bromo-aryl/heteroaryl triazole 69 to the corresponding hydroxy-aryl/ heteroaryl triazole 70 is achieved via the corresponding boronate using the 2 step sequence [B(pin) 2 /Pd- catalysis followed by treatment with H2O2] described in Scheme 4. Intermediate 70 is then subjected to a Mitsunobu reaction with hydroxy-cyclopentyl ester 7 to give triazole cyclopentyl ester 62, followed by ester deprotection to provide the desired triazole amino- azine cyclopentyl acids 63.

Scheme 11

Br

X 1 ^ X 2 1 ) Deprotection 1 ) NaN 3

x [ x4 - of Alcohol 2) Reduction of

azide

T" r PG i 2) PBr 3 or

N-N Ph 3 P/CBr4

5a

36 67 68

A 2 OH

A 1 -^-R 1

X A N" a3 X 1 ^X 2 KOH, Pd ligand X 1 "^X 2

5 x!^x 4 2 c3 x 4

9 10a or

Y n 1 * I A 1 ^R 10a

Base » ' /---x N A. N ' .A 3 1 ) (Bpin) 2 , Pd catalyst ' / ^ N^ N^ or w

N-N N-N H

2) H 2 0 2

Pd, ligands 5a 5a

62

63

Scheme 12 describes the synthesis of: 1) amino-azole isoxazole cyclopentyl acids 73, 2) amino-azole triazole cyclopentyl acids 75 and 3) amino-azole pyrazole cyclopentyl acids 77, wherein A 4 , A 5 , A 6 and A 7 are N or CR. For instance, isoxazole amine 21 is reacted with halo-azole 71 in the presence of an appropriate base or under Pd-mediated cross-coupling conditions to give the isoxazole amino-azole 72, which is then subjected to ester deprotection to provide the desired amino-azole isoxazole-cyclopentyl acids 73. The corresponding triazole amino-azole acids 75 and the pyrazole amino-azole acids 77 are synthesized from the triazole amine 41 and the pyrazole amine 57 respectively using this same general route as described for the preparation of 73 from 21. Scheme 12

Scheme 12A describes an alternative synthesis of amino-azole triazole

cyclopentyl acids 75. Triazole bromide 40 is reacted with an appropriate amino-azole 78 in the presence of base (e.g. NaH, etc.,) or under Pd-mediated cross-coupling conditions to afford the corresponding triazole amino-azole 74, which is subjected to ester deprotection to give the desired triazole amino-azole cyclopentyl acids 75. Similarly, the corresponding isoxazole bromide 20 and the pyrazole bromide 56 can be converted to the corresponding amino-azine isoxazole acids 73 and amino-azine pyrazole acids 77 respectively by the same two-step sequence as described for the synthesis of triazole acids 75 from triazole bromide 40. Scheme 12 A

Scheme 13 describes an alternative synthetic route to the triazole amino-azole cyclo-pentyl acids 75. The bromo-triazole 67 is subjected to a transition-metal-catalyzed cross-coupling reaction (e.g. palladium-mediated) or a base-mediated SNAr reaction with an appropriate amino-azole 78 to furnish the triazole amino-azole 79. Conversion of the bromo-aryl/heteroaryl triazole 79 to the correspond- ing hydroxy-aryl/heteroaryl triazole 80 is achieved via the corresponding boronate using the 2 step sequence [B(pin) 2 /Pd- catalysis followed by treatment with H2O2] described in Scheme 4. Intermediate 80 is then subjected to a Mitsunobu reaction with hydroxy-cyclopentyl ester 7 to give triazole cyclopentyl ester 74, followed by ester deprotection to provide the desired triazole amino- azole cyclopentyl acids 73. Alternatively, the bromo-aryl/heteroaryl triazole 79 can also be synthesized by reaction of triazole amine 68 with an appropriate halo-azole 71 in the presence of base (e.g. NaH, etc.,) or under Pd-mediated cross-coupling conditions.

Scheme 13

Scheme 14 describes an alternative synthesis of the triazole carbamate cyclopentyl acids 34. Deprotection of triazole 27 provides the triazole alcohol 81, which is treated with 4-nitro- phenyl chloroformate to provide the intermediate triazole 4-nitrophenyl carbonate, which is reacted with an appropriate amine 11 to provide the triazole carbamate 82. Reaction of the bromo-aryl/heteroaryl-triazoles 82 with

bis(pinacolato)diboron in the presence of an appropriate palladium catalyst (e.g.

Ishiyama, T. et al, J Org. Chem. 1995, 60, 7508-7510) provides the corresponding pinacol boronate, which is then oxidized with hydrogen peroxide to give the

corresponding phenol or hydroxyheteroarene 83 (e.g. Fukumoto, S. et al,

WO 2012137982). Mitsunobu reaction of hydroxyheteroarene 83 with an appropriate hydroxycyclopentyl ester 7, followed by acid deprotection provides the desired triazole carbamate cyclopentyl acids 34. Scheme 14

Similarly, the analogous synthetic sequence (starting from the bromo-isoxazole 4) provides the isoxazole carbamate cyclopentyl acids 13, via the key intermediate hydroxyaryl isoxazole carbamate 84.

Scheme 14 A

The synthesis of pyrazine triazole carbamate cyclopentyl acids 91 is shown in Scheme 15. A chloro- substituted pyrazine undergoes a base-mediated SNAr reaction with an appropriate hydroxycyclopentyl ester 86 to give the pyrazine ether 87. Bromination of pyrazine 87 ((e.g. with N-bromo-succinimide, with concomitant ester deprotection) followed by reprotection of the acid, provides bromopyrazine ester 88. Sonogashira coupling reaction (e.g. Alper, P. et al, WO 2008097428) of bromide 88 with an appropriately protected (e.g. as a tetrahydropyranyl ether) propargyl alcohol 25 provides the corresponding alkynyl-pyrazine 89. Cycloaddition of alkyne 89 with trimethyl silyl azide (Qian, Y. et al, J Med. Chem ., 2012, 55 , 7920-7939) under transition-metal catalyzed conditions (e.g. Boren, B.C. et. al., J. Am. Chem. Soc., 2008, 130, 8923-8930; Ramasamy, S., et al., Org. Process Res. Dev., 2018, 22, 880-887) furnishes the trimethyl silyl methyl triazole with the desired regiochemistry. The trimethyl silyl group is subsequently removed under standard desilylation conditions (e.g. B NF, as in Qian, Y. et al, J. Med. Chem., 2012, 55, 7920-7939) to give the protected triazole alcohol 90. The key pyrazine triazole alcohol intermediate 90 is carried forward to the synthesis of pyrazine triazole carbamate cyclopentyl acids 91 according to the procedures described in Scheme 4 (i.e. from 30 -> 34).

Scheme 15

The synthesis of pyrazine pyrazole carbamate cyclopentyl acids 95 is shown in Scheme 16. A 4-bromo-substituted pyrazole 92 is reacted with bis(pinacolato)diboron in the presence of an appropriate palladium catalyst (e.g. Ishiyama, T. et al, J Org. Chem. 1995, 60, 7508-7510) provides the corresponding pinacol boronate 93. Suzuki-Miyaura cross-coupling reaction of boronate 93 with bromopyrazine ester 88 (e.g. Almond-Thynne et al., Chem. Sci. 2017, 8, 40-62) provides the key pyrazine pyrazole cyclopentyl ester intermediate 94, which is then converted to pyrazine pyrazole carbamate cyclopentyl acids according to the general sequence described in Scheme 4 for the corresponding triazole cyclopentyl acids.

Scheme 16

Pd-catalyzed

cross-coupling

The synthesis of homologated cyclopentyl acids 97 is shown in Scheme 17.

Cyclopentyl acid 34 is reacted with oxalyl chloride, followed by treatment with either diazomethane or trimethyl silyl diazomethane to provide the a-diazoketone 96. Diazoketone 96 is then subjected to the Wolff rearrangement reaction (e.g., with aqueous silver benzoate) to provide the corresponding homologated cyclopentyl acids 97.

Scheme 17

An alternative synthetic route to the pyrazole carbamate cyclopentyl acids 52 is shown in Scheme 18. A 4-bromopyrazole 5-carboxylic acid ester 98 is subjected to a Suzuki-Miyaura cross-coupling reaction with an appropriately substituted 4-hydroxy- aryl/heteroaryl boronate 46 to provide the corresponding 4-hydroxy-aryl(heteroaryl)- pyrazole 99. Reaction of phenol/hydroxyheteroarene 99 with an orthogonally protected hydroxy cyclopentyl acid ester 7 under Mitsunobu reaction conditions (Kumara Swamy, K. C., Chem. Rev., 2009, 109, 2551-2651) furnishes the corresponding pyrazole cyclopentyl ester 100. Selective deprotection of the pyrazole ester followed by reduction of the acid product (e.g. by a 2 step, l-pot reaction via reaction with an alkyl chloro- formate followed by low-temperature reduction with NaBH 4 , or directly with diborane) to the corresponding pyrazole alcohol 49. Curtius rearrangement (e.g. with (PhO)2PON3) of an appropriate carboxylic acid R 4 C02H in the presence of pyrazole alcohol 49 provides the pyrazole NH-carbamate 101. Deprotonation of NH-carbamate 101 with a suitable base (e.g. NaH or NaN(TMS)2) followed by alkylation with an appropriate alkyl bromide or iodide (R¾r or R 8 I) followed by ester deprotection provides the desired pyrazole carbamate cyclopentyl acids 52. Scheme 18

VII. EXAMPLES

The following Examples are offered as illustrative, as a partial scope and particular embodiments of the invention and are not meant to be limiting of the scope of the invention. Abbreviations and chemical symbols have their usual and customary meanings unless otherwise indicated. ETnless otherwise indicated, the compounds described herein have been prepared, isolated and characterized using the schemes and other methods disclosed herein or may be prepared using the same.

As appropriate, reactions were conducted under an atmosphere of dry nitrogen (or argon). For anhydrous reactions, DRISOLV® solvents from EM were employed. For other reactions, reagent grade or HPLC grade solvents were utilized. Unless otherwise stated, all commercially obtained reagents were used as received.

Microwave reactions were carried out using a 400W Biotage Initiator instrument in microwave reaction vessels under microwave (2.5 GHz) irradiation.

HPLC/MS and preparatory/analytical HPLC methods employed in

characterization or purification of examples.

NMR (nuclear magnetic resonance) spectra were typically obtained on Bruker or JEOL 400 MHz and 500 MHz instruments in the indicated solvents. All chemical shifts are reported in ppm from tetramethylsilane with the solvent resonance as the internal standard. In the examples where ¾ NMR spectra were collected in d 6 -DMSO, a water- suppression sequence is often utilized. This sequence effectively suppresses the water signal and any proton peaks in the same region usually between 3.30-3.65 ppm which will affect the overall proton integration.

1 HNMR spectral data are typically reported as follows: chemical shift, multiplicity (s = singlet, br s = broad singlet, d = doublet, dd = doublet of doublets, t = triplet, q = quartet, sep = septet, m = multiplet, app = apparent), coupling constants (Hz), and integration.

The term HPLC refers to a Shimadzu high performance liquid chromatography instrument with one of following methods:

HPLC-l : Sunfire C18 column (4.6 x 150 mm) 3.5 pm, gradient from 10 to 100% B:A for 12 min, then 3 min hold at 100% B.

Mobile phase A: 0.05% TFA in water:CH 3 CN (95:5)

Mobile phase B: 0.05% TFA in CH 3 CN:water (95:5)

TFA Buffer pH = 2.5; Flow rate: 1 mL/ min; Wavelength: 254 nm, 220 nm.

HPLC-2: XBridge Phenyl (4.6 x 150 mm) 3.5 pm, gradient from 10 to 100% B:A for 12 min, then 3 min hold at 100% B.

Mobile phase A: 0.05% TFA in water:CH 3 CN (95:5)

Mobile phase B: 0.05% TFA in CH 3 CN:water (95:5)

TFA Buffer pH = 2.5; Flow rate: 1 mL/ min; Wavelength: 254 nm, 220 nm. HPLC-3: Chiralpak AD-H, 4.6 x 250 mm, 5 pm.

Mobile Phase: 30% EtOH-heptane (1 : 1) / 70% C0 2

Flow rate = 40 mL/min, 100 Bar, 35 °C; Wavelength: 220 nm

HPLC-4: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7-mih particles;

Mobile Phase A: 5:95 CH 3 CN:water with 10 mM NH 4 OAc;

Mobile Phase B: 95:5 CH3CN: water with 10 mM NEEOAc;

Temperature: 50 °C; Gradient: 0-100% B over 3 min, then a 0.75-min hold at 100% B; Flow: 1.11 mL/min; Detection: LTV at 220 nm.

HPLC-5: Waters Acquity EIPLC BEH C18, 2.1 x 50 mm, 1.7-mih particles;

Mobile Phase A: 5:95 CH3CN: water with 0.1% TFA;

Mobile Phase B: 95:5 CH 3 CN:water with 0.1% TFA;

Temperature: 50 °C; Gradient: 0-100% B over 3 min, then a 0.75-min hold at 100% B; Flow: 1.11 mL/min; Detection: LTV at 220 nm.

Intermediate 1. (±)-cis-isopropyl 2-(3-hydroxycyclopentyl)acetate

Acetyl chloride (2.13 mL, 30.0 mmol) was added portionwise to z-PrOH (25 mL) at 0°C and the mixture was warmed to RT and stirred at RT for 30 min, after which 2-oxa -bicyclo- [3.2. l]octan-3-one (prepared according to the procedure described in ./. Org. Chem., 1983, 48, 1643-1654; 2.91 g, 23.1 mmol) was added. The reaction mixture was stirred at RT overnight, then was concentrated in vacuo, diluted with toluene, and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 20-50% EtOAc in hexanes) to give the title compound (2.93 g, 68 % yield) as an oil. 1 H NMR (500 MHz, CDCh) d 5.01 (dt, =l2.6, 6.2 Hz, 1H), 4.31 (m, 1H), 2.40 (s, 1H), 2.39 (s, 1H), 2.28(m, 1H), 2.20 (m, 1H), 1.80 (m, 2H), 1.70 - 1.63 (m, 1H), 1.60 (m, 2H), 1.49 (m, 1H), 1.23 (d, J=6.3 Hz, 6H). Intermediate 2. (±)-trans-isopropyl 2-(3-hydroxycyclopentyl)acetate

Intermediate 2A. (±)-trans-3-(2-isopropoxy-2-oxoethyl)cyclopentyl 4-nitrobenzoate A0996-142

To a 0°C solution of Intermediate 1 (1.16 g, 6.23 mmol), 4-nitrobenzoic acid (1.56 g, 9.34 mmol), and Ph 3 P (2.45 g, 9.34 mmol) in THF (25 mL) was added DEAD (1.63 g, 9.34 mmol) dropwise. After stirring at 0°C for 1 h, the reaction mixture was allowed to warm to RT and stirred overnight at RT, then was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 5-30% EtOAc in hexanes) to give the title compound (822 mg, 39 % yield). ¾ NMR (500 MHz, CDCh) 5 8.28 (d, J=8.5 Hz, 2H), 8.18 (d, =8.5 Hz, 2H), 5.48 (m, 1H), 5.03 (m, 1H), 2.60 (m, 1H), 2.36 (d, J=1 A Hz, 2H), 2.28 - 2.06 (m, 3H), 1.86 (m, 1H), 1.64 (m, 1H), 1.34 (m, 1H), 1.25 (s, 3H), 1.24

(s, 3H).

Intermediate 2

To a solution of Intermediate 2A (822 mg, 2.45 mmol) in THF (15 mL) was added aq. 1M LiOH (2.94 mL, 2.94 mmol). The reaction mixture was stirred at RT overnight, then was partially concentrated in vacuo, diluted with ThO, and extracted with EtOAc (2x). The combined organic extracts were washed with ThO, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 25-50% EtOAc in hexanes) to give the title compound (275 mg, 60 % yield) as an oil. ¾ NMR (500 MHz, CDCh) 55.00 (m, 1H), 4.39 (m, 1H), 2.58 (m, 1H), 2.29 (d,

J=1 A Hz, 2H), 2.08 - 1.94 (m, 2H), 1.85 (m, 1H), 1.58 (m, 2H), 1.43 (m, 1H), 1.33 (br d, J=3.3 Hz, 1H), 1.24 (s, 3H), 1.22 (s, 3H).

Intermediate 3. (±)-cis-isopropyl 2-(3-hydroxycyclopentyl)-2-methylpropanoate and Intermediate 4. (±)-cis-isopropyl-2-(3-hydroxycyclopentyl)propanoate

3 4

These intermediates were prepared according to the procedure described in Can. J Chem., 1990, 68, 804-811, but as the respective isopropyl esters. Intermediate 4 was prepared as a ~2: 1 mixture of diastereomers at the methyl chiral center.

Intermediate 5. (±)-Cis-m ethyl l-(3-hydroxycyclopentyl)cyclopropane-l-carboxylate

Intermediate 5 A. (±)-4-oxaspiro[bicyclo[3.2.l]octane-2,l'-cyclopropan]-3-one

To a suspension of NaH (111 mg, 2.77 mmol, 60%) in DMSO (3 mL) was added portionwise trimethyl sulfoxonium iodide (731 mg, 3.32 mmol). The mixture was stirred at RT for 1 h, after which a solution of 4-methylene-2-oxabicyclo[3.2. l]octan-3-one (synthesized according to the procedure described in J. Org. Chem. 1984, 49, 2079-2081, 255 mg, 1.85 mmol) in DMSO (3 mL) was added. The reaction mixture was stirred at RT for 1 h, then was quenched cautiously with satd aq. NLLCl and extracted with EtOAc (2x). The combined organic extracts were washed with ThO (4x), brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 20-50% EtOAc in hexanes) to give the title compound (178 mg, 63 % yield) as an oil. LC-MS, [M+H] + = 153; ¾ NMR (500 MHz, CDCh) d 4.95 (m, 1H), 2.27 (br d, =l 1.8 Hz, 1H), 2.22 - 2.15 (m, 1H), 2.02 - 1.91 (m, 2H), 1.89 - 1.80 (m, 1H), 1.77 - 1.70 (m, 2H), 1.47 - 1.25 (m, 2H), 0.89 - 0.71 (m, 2H).

Intermediate 5

Acetyl chloride (0.10 mL; 1.43 mmol) was added portionwise to MeOH (5 mL) at 0°C and the mixture was allowed to warm to RT and stirred for 30 min at RT, after which Intermediate 4A (87 mg, 0.57 mmol) was added. The reaction mixture was stirred at RT overnight, then was heated at 60° for 3 h, then was cooled to RT, and concentrated in vacuo. The residue was diluted with toluene and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 15-50% EtOAc in hexanes) to give the title compound (18 mg, 17 % yield) as an oil. ¾ NMR (500 MHz, CDCh) d 4.29 (m,

1H), 3.65 (s, 3H), 2.88 (br s, 1H), 2.18 (m, 1H), 2.02 (m, 1H), 1.74 (m, 2H), 1.65 (m, 2H), 1.43 (m, 1H), 1.24 (m, 2H), 0.80 (m, 2H).

Intermediate 6. 2,5-dibromo-3-fluoro-6-methylpyridine

Intermediate 6 A. 3-fluoro-6-methylpyridin-2-amine

To a solution of 2-bromo-3-fluoro-6-methylpyridine (5.0 g, 26.3 mmol) in ethylene glycol (50 ml) and aq. 28% NH 4 OH (63 mL; 450 mmol) was added C O (0.19 g, 1.32 mmol), K2CO3 (0.73 g, 5.26 mmol), and N, N-dimethylethylenediamine (0.29 mL, 2.63 mmol). The reaction mixture was purged with N2 and was heated at 80°C overnight in a sealed tube, then was cooled to RT and extracted with CH2CI2 (3x). The combined organic extracts were dried (Na2S0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0-100% EtOAc in hexanes) to give the title compound (2.81 g, 85 % yield). ¾ NMR (500 MHz, CDCh) d 7.11 (dd, =l0.6, 8.1 Hz, 1H), 6.47 (dd, =8.0, 3.0 Hz, 1H), 4.55 (br s, 2H), 2.38 (s, 3H).

Intermediate 6B. 5-bromo-3-fluoro-6-methylpyridin-2-amine

To a solution of Intermediate 6A (3.91 g, 31.0 mmol) in CH3CN (100 mL) at 0°C and added NBS (5.52 g, 31.0 mmol) portionwise while maintaining the reaction temperature at <5°C. The reaction mixture was stirred at RT for 30 min, then was concentrated in vacuo. The residue was chromatographed (S1O2; isocratic 30% EtOAc in hexanes) to give the title compound (6.14 g, 97 % yield). ¾ NMR (500 MHz, CDCh) d 7.37 (d, J=9.6 Hz, 1H), 4.59 (br s, 2H), 2.48 (d, =l. l Hz, 3H).

Intermediate 6

To aq. 48% HBr (23.72 mL, 210 mmol, 48%) at 0°C was added Intermediate 6B (6.l4g, 29.9 mmol) slowly portionwise. Bn (3.09 mL, 59.9 mmol) was added dropwise while maintaining the reaction temperature at <5°C. The reaction mixture was stirred at 0°C for 30 min, after which a solution of NaNCh (5.17 g, 74.9 mmol) in water (10 mL) was added dropwise while maintaining the reaction temperature at <5°C. The reaction mixture was stirred for 30 min at 0°C, then was poured into ice water, basified with 50% aq. NaOH and extracted with EtOAc (2x). The combined organic extracts were washed with aq. 10% Na2S20 3 , brine, dried (Na2S0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0-25% EtOAc in hexanes) to give the title compound (3.90g, 48 % yield). ¾ NMR (500 MHz, CDCh) d 7.60 (d, J=6.6 Hz, 1H), 2.64 (d, .7=1.4 Hz, 3H).

Intermediate 7. 4-nitrophenyl ((l-propylcyclopropyl)methyl) carbonate

7 A. Tert-butyl l-propylcyclopropane-l-carboxylate

To a solution of (ΐ-R NH (4.74 mL, 33.2 mmol) in THF (40 mL) at 0°C was added n-BuLi (13.3 mL of a 2.5M solution in hexanes, 33.2 mmol) portionwise. The LDA solution was stirred for 15 min at 0°C, then was warmed to RT, and stirred for 30 min at RT, then was cooled to-78°C. Tert- butyl cyclopropanecarboxylate (3.78 g, 26.6 mmol) was added dropwise over 10 min. The reaction was stirred at -78°C for 2 h, after which l-bromopropane (4.84 mL, 53.2 mmol) was added dropwise over 20 min. The reaction was allowed to slowly warm to RT and stirred overnight at RT, then was quenched with satd aq. NH 4 Cl and extracted with EtOAc (2x). The combined organic extracts were washed with brine, dried (MgSCri), and concentrated in vacuo. The residue was distilled under reduced pressure (20 torr, BP = 95°C) to give the title compound (2.99 g, 61 % yield) as an oil. ¾ NMR (500 MHz, CDCb) d 1.48 (m, 4H), 1.45 (s, 9H), 1.12 (m, 2H), 0.92 (m, 3H), 0.61 (m, 2H).

Intermediate 7B. (l-propylcyclopropyl)methanol

To a solution of Intermediate 7A (250 mg, 1.36 mmol) in Et 2 0 (5 mL) was cautiously added LiAlH 4 (103 mg, 2.71 mmol) portionwise. The reaction mixture was stirred overnight at RT, then was sequentially treated with water (0.1 mL), 15% aq.

NaOH (0.1 mL), and water (0.3 mL). The mixture was stirred for 1 h at RT, dried (MgS0 4 ) and concentrated in vacuo. The residue was distilled under reduced pressure to give the slightly impure title compound (186 mg, 120%; contains some t-BuOH) as an oil. 1 H NMR (500 MHz, CDCb) d 3.44 (br s, 2H), 1.48 - 1.36 (m, 4H), 0.93 (t, J=7.0 Hz,

3H), 0.44 - 0.27 (m, 4H).

Intermediate 7

To a solution of Intermediate 7B (155 mg, 1.36 mmol) in CH2CI2 (10 mL) were added pyridine (0.44 mL, 5.43 mmol) and 4-nitrophenyl chloroformate (410 mg, 2.04 mmol). The reaction mixture was stirred at RT for 2 h, then was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0-25% EtOAc in hexanes) to give the title compound (226 mg, 60 % yield) as a white solid. ¾ NMR (500 MHz, CDCb) d 8.31 (d, =9.l Hz, 2H), 7.42 (d, J=9A Hz, 2H), 4.15 (s, 2H), 1.45 (m,

4H), 0.96 (t, =7.0 Hz, 3H), 0.58 (m, 2H), 0.51 (m, 2H).

The following Intermediates 8 to 11 were prepared using analogous synthetic routes to the procedures described for the synthesis of Intermediate 7, starting from either /<2/7-butyl cyclopropanecarboxylate or tert- butyl cyclobutanecarboxylate. Intermediate 8. (l-methylcyclopropyl)methyl (4-nitrophenyl) carbonate

¾ NMR (400 MHz, CDCh) d 8.28 (d, J=92 Hz, 2H), 7.40 (d, =9.2 Hz, 2H), 4.10 (s, 2H), 1.22 (s, 3H), 0.60 (m, 2H), 0.47 (m, 2H).

Intermediate 9. (l-ethylcyclopropyl)methyl (4-nitrophenyl) carbonate

¾ NMR (400 MHz, CDCh) d 8.28 (d, =9.2 Hz, 2H), 7.39 (d, =9.2 Hz, 2H), 4.14 (s, 2H), 1.48 (q, =7.3 Hz, 2H), 0.98 (t, J=1 A Hz, 3H), 0.54 (m, 4H).

Intermediate 10. (l-ethylcy cl obutyl)m ethyl (4-nitrophenyl) carbonate

¾ NMR (500 MHz, CDCh) d 8.31 (d, =9.4 Hz, 2H), 7.42 (d, =9.4 Hz, 2H), 4.27 (s, 2H), 1.99 - 1.83 (m, 6H), 1.63 (q, J=1 A Hz, 2H), 0.90 (t, J=1 A Hz, 3H).

Intermediate 11. 4-nitrophenyl ((l-propylcyclobutyl)methyl) carbonate

¾ NMR (500 MHz, CDCh) d 8.31 (d, =9.4 Hz, 2H), 7.42 (d, =9.4 Hz, 2H), 4.26 (s, 2H), 1.99 - 1.85 (m, 6H), 1.56 (m, 2H), 1.32 (m, 2H), 0.97 (t, =7.3 Hz, 3H). Intermediate 12. (±)-cis-benzyl 3-hydroxycyclopentane-l-carboxylate

Intermediate 12A. (±)-cis-3-((benzyloxy)carbonyl)cyclopentyl 4-nitrobenzoate

To a 0°C solution of trans-benzyl 3-hydroxycyclopentane-l-carboxylate (prepared according to the procedure described in Bioorg. Med. Chem. Lett., 2013, 23, 3833-3840; 285 mg, 1.29 mmol), 4-nitrobenzoic acid (324 mg, 1.94 mmol), and Ph 3 P (509 mg, 1.94 mmol) in THF (25 mL) was added DEAD (1.17 mL, 1.94 mmol, 40%) slowly portion- wise. The reaction mixture was stirred at 0°C, then was warmed to RT and stirred at RT overnight, after which it was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 5-30% EtOAc in hexanes) to give the title compound (430 mg, 90 % yield). ¾ NMR (500 MHz, CDCh) d 8.23 (d, =8.8 Hz, 2H), 8.11 (d, .7=9.1 Hz, 2H), 7.32 (m, 5H), 5.45 (m, 1H), 5.12 (d, .7=1.9 Hz, 2H), 2.99 (m, 1H), 2.04 (s, 6H).

Intermediate 12

To a RT solution of Intermediate 12A (328 mg, 0.89 mmol) in THF (10 mL) was added aq. 1M LiOH (0.89 mL, 0.89 mmol). The reaction mixture was stirred at RT overnight, then was acidified with aq. 1N HC1, diluted with H2O, and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried

(MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2;

continuous gradient from 15-50% EtOAc in hexanes) to give the title compound (55 mg, 28 % yield) as an oil. ¾ NMR (400 MHz, CDCh) d 7.35 (m, 5H), 5.14 (s, 2H), 4.34 (m, 1H), 2.93 (m, 1H), 2.13 - 1.94 (m, 4H), 1.79 (m, 2H). Intermediate 13. 6-(3-methyl-4-(((4-phenylpyrimi din-2 -yl)amino)methyl)isoxazol-5-yl) pyri din-3 -ol

Intermediate 13A. N-((5-(5-bromopyridin-2-yl)-3-methylisoxazol-4-yl)methyl)-4- phenyl- pyrimidin-2-amine

To a solution of 4-phenylpyrimidin-2-amine (116 mg, 0.678 mmol) in THF (2 mL) was added n-BuLi (0.42 mL of a 1.5 M solution in hexanes, 0.68 mmol) at -78 °C. The reaction mixture was allowed to warm slowly to RT and stirred for 5 min at RT. A solution of Example 29B (150 mg, 0.452 mmol) in THF (1 mL) was quickly added at RT, and the mixture was stirred at RT for 48 h. The reaction mixture was diluted with H2O (2 mL) and extracted with EtOAc (3 x 5 mL). The combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo. The residue was chromatographed (12 g S1O2, continuous gradient from 0 to 50 % EtOAc in hexanes over 12 min) to provide the title compound (172 mg, 0.407 mmol, 90 % yield) as a slightly colored solid. ¾ NMR (500 MHz, CDCh) d 8.86 (d, J= 2.4 Hz, 1H), 8.35 (dd, J= 11.3, 3.8 Hz, 1H), 7.99 (ddd, J = 14.5, 8.0, 4.9 Hz, 3H), 7.83 (dd, J= 20.3, 8.5 Hz, 1H), 7.48 (dd, J= 5.0, 2.0 Hz, 3H), 6.98 (d, J= 5.2 Hz, 1H), 6.44 (t, J= 6.8 Hz, 1H), 4.85 (d, J= 6.6 Hz, 2H), 2.55 (s, 3H);

[M+H]+ = 422. Intermediate 13

A mixture of Pd2(dba)3 (20 mg, 0.022 mmol), di-tert-butyl(2',4',6'-triisopropyl- [l,l'-biphenyl]-2-yl)phosphine (37 mg, 0.088 mmol), KOH (124 mg, 2.20 mmol), and Example 30A (155 mg, 0.367 mmol) in l,4-dioxane (2 mL) and EhO (2 mL) was degassed (by evacuating & backfilling with Ar 3X). The reaction mixture was stirred at 85°C for 14 h, then was cooled to RT and acidified to pH 4 with aq. 1N HC1. EtOH (2 mL) was added, and the mixture was extracted with EtOAc (8 x 5 mL). The combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo. The residual brown solid was chromatographed (12 g S1O2, continuous gradient from 0 to 100 % EtOAc in hexanes over 13 min) to provide the title compound (49 mg, 0.136 mmol, 37.1 % yield) as a red solid. ¾ NMR (500 MHz, DMSO-^e) d 8.35 (d, J= 2.9 Hz, 2H), 7.99 (s, 2H), 7.79 (d, J= 8.6 Hz, 1H), 7.49 (t, J= 7.2 Hz, 1H), 7.43 (s, 2H), 7.37 (dd, J= 8.6, 2.9 Hz,

1H), 7.18 (d, J= 5.2 Hz, 1H), 4.82 (s, 2H), 2.33 (s, 3H); [M+H]+ = 360.3.

Intermediate 14. 2-methyl-6-(3-methyl-4-(((4-(pyridin-2-yl)pyrimidin-2-yl)ami no) methyl)isoxazol-5-yl)pyridin-3-ol

The title compound was synthesized following the same sequence as for the synthesis of Example 29D from Example 29A, but using (5-(5-bromo-6-methylpyridin-2- yl)-3-methyl-isoxazol-4-yl)methanol as the starting material instead of (5-(5-bromo- pyridin-2-yl)-3-methylisoxazol-4-yl)methanol. ¾ NMR (500 MHz, DMSO-i¾) d 8.76 - 8.57 (m, 1H), 8.44 (s, 1H), 8.07 (br s, 1H), 7.81 (br s, 1H), 7.63 (d, j= 8.4 Hz, 1H), 7.50 (t, j= 5.7 Hz, 2H), 7.29 (d, j= 8.4 Hz, 1H), 4.83 (s, 2H), 2.41 (s, 3H), 2.32 (s, 3H);

[M+H]+ = 375.2.

Intermediate 15. 2-methyl-6-(l-methyl-5-(((4-(pyridin-2-yl)pyrimidin-2-yl)ami no) methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-ol

The title compound was synthesized following the same sequence as for the synthesis of Example 29D from Example 29A, but using (4-(5-bromo-6-methylpyridin-2- yl)-l-methyl-lH-l,2,3-triazol-5-yl)methanol 30B as the starting material instead of (5-(5- bromopyridin-2-yl)-3-methylisoxazol-4-yl)methanol. ¾ NMR (500 MHz, DMSO-Ts) d 8.66 (d, J= 4.8 Hz, 1H), 8.45 (s, 1H), 7.99 (br s, 1H), 7.82 (s, 1H), 7.76 (d, J= 8.4 Hz, 1H), 7.54 - 7.45 (m, 2H), 7.26 (d, J= 8.4 Hz, 1H), 5.10 (s, 2H), 4.13 (s, 3H), 2.40 (s,

3H); [M+H]+ = 375.1. Intermediate 16. 6-(l-methyl-5-(((4-(pyridin-2-yl)pyrimidin-2-yl)amino)methyl )-lH- l,2,3-triazol-4-yl)pyridin-3-ol

The title compound was synthesized following the same sequence as for the synthesis of Example 29D from Example 29A, but using (4-(5-bromopyridin-2-yl)-l- methyl-lH-l,2,3-triazol-5-yl)methanol as the starting material instead of (5-(5-bromo- pyridin-2-yl)-3-methylisoxazol-4-yl)methanol. ¾ NMK (500 MHz, DMSO-Ts) d 8.71 (d, J= 4.8 Hz, 1H), 8.66 (d, 7= 4.6 Hz, 1H), 8.45 (d, 7= 5.1 Hz, 1H), 8.12 (d, J= 8.0 Hz, 1H), 7.95 (t, J= 7.9 Hz, 1H), 7.75 (br s, 2H), 7.52 (d, J= 5.0 Hz, 1H), 7.47 (d, J= 6.6 Hz,

1H), 7.40 (t, J= 6.2 Hz, 1H), 5.19 (s, 2H), 4.16 (s, 3H); [M+H]+ = 361.1.

Intermediate 17. 6-(5-(((4-isopropylpyrimidin-2-yl)amino)methyl)-l -methyl- 1H- 1,2, 3- triazol-4-yl)-2-methylpyridin-3-ol

The title compound was synthesized following the same sequence as for the synthesis of Example 29D from Example 29A, but using (4-(5-bromo-6-methylpyridin-2- yl)-l-methyl-lH-l,2,3-triazol-5-yl)methanol 2B as the starting material instead of (5-(5- bromopyridin-2-yl)-3-methylisoxazol-4-yl)methanol. ¾ NMR (500 MHz, DMSO-Tis) d 8.19 (s, 1H), 7.75 (d, J= 8.3 Hz, 1H), 7.49 (d, J= 6.2 Hz, 1H), 7.24 (d, J= 8.3 Hz, 1H), 6.54 (d, J= 5.0 Hz, 1H), 4.92 (d, J= 5.9 Hz, 2H), 4.14 (s, 3H), 2.69 (sep, J= 7.0 Hz, 1H),

2.41 (s, 3H), 1.11 (d, 7= 6.9 Hz, 6H); [M+H]+ = 340.0.

Intermediate 18. 5-(2-cyclobutylethyl)-l,2,4-oxadiazol-3-amine

To a 0 °C solution of 3-cyclobutylpropanoic acid (0.29 g, 2.26 mmol) in DMF (4.5 mL) at 0°C were successively added NaBH 3 CN (145 mg, 2.26 mmol), DIEA (1.98 ml, 11.3 mmol) and HATH (1.03 g, 2.72 mmol). The reaction was allowed to warm to RT, stirred for 18 h at RT, then was concentrated in vacuo. The residue was suspended in EtOH (3 mL), after which NH2OH.HCI (0.24 g, 3.39 mmol) was added, followed by pyridine (0.73 mL, 9.04 mmol). The reaction was stirred at RT for 18 h, then was concentrated in vacuo. The residue was partitioned between CH2CI2 and water; the aqueous layer was extracted with CH2CI2 (2x). The combined organic layers were dried (Na2S0 4 ) and concentrated in vacuo. The crude product was chromatographed (S1O2, continuous gradient from 0-100% EtOAc in hexane) to afford the title compound (210 mg, 56 %) as a white solid. LC-MS, [M+H] + = 168; ¾ NMR (500 MHz, CDCh) d 4.34 (br s, 2H), 2.68 (t, 7=7.6 Hz, 2H), 2.34 (quin, 7=7.8 Hz, 1H), 2.10 (m, 2H), 1.8 (m, 4H), 1.66 (m, 2H).

Examples 1 and 2. Cis and trans-3-(4-(3-methyl-4-((((R)-l-phenylethoxy)carbonyl) amino)isoxazol-5-yl)phenoxy)cyclopentane-l -carboxylic acid

Example 1 Example 2

1A. (R)-l-phenylethyl (5-(4-hydroxyphenyl)-3-methylisoxazol-4-yl)carbamate

To a solution of (R)-l-phenylethyl (5-(4-bromophenyl)-3-methylisoxazol-4-yl) carbamate (4.3 g, 10.7 mmol; prepared according to W02010/141761, Example 1) and B 2 (OH)2 (1.25 g, 13.9 mmol) in THF (30 mL) and MeOH (12 mL) was added iPnNEt (4.49 mL, 25.7 mmol). The mixture was degassed with N2 and bis(di-/-Bu(4-dimethyl- aminophenyl)phosphine)Pd(II)Cl2 (0.152 g, 0.21 mmol) was added. The mixture was degassed with N2 and was heated at 50°C for 6 h, then was cooled to RT. The mixture was diluted with water, acidified with 1N aq. HC1 and extracted with EtOAc (3X). The combined organic extracts were washed with water, brine, dried (MgSCri), and concentrated in vacuo. The residue was taken up in MeOH (100 mL) and water (30 mL), after which aq. 30% H2O2 (1.07 mL, 10.5 mmol) was added. The reaction was stirred overnight, then was partially concentrated in vacuo, diluted with water and extracted with EtOAc (3X). The combined organic extracts were washed with water, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2;

continuous gradient from 25-75% EtOAc in hexanes) to give the title compound (1.62 g, 45 % yield) as a white solid. LC-MS, [M+H] + = 339; ¾ NMR (400 MHz, CD3OD) d 7.61 (br d, =8.4 Hz, 2H), 7.48 - 7.32 (m, 5H), 6.85 (br d, =8.4 Hz, 2H), 5.83 (m, 1H),

2.16 (s, 3H), 1.62 (br d, =6.4 Hz, 3H). 1B. Ethyl 3-(4-(3-methyl-4-((((R)-l-phenylethoxy)carbonyl)amino)isoxaz ol-5- yl)phenoxy)cyclopentane- 1 -carboxylate

To a 0°C solution of compound 1A (100 mg, 0.296 mmol) and ethyl 3-hydroxy- cyclopentane carboxylate (mixture of racemic cis & trans isomers; 61 mg, 0.38 mmol) in THF (3 mL) were added Ph 3 P (101 mg, 0.384 mmol) and DEAD (0.23 mL, 0.384 mmol). The reaction was allowed to warm to RT and stirred overnight at RT, then was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 15-70% EtOAc in hexanes) to give the title compound (127 mg, 90 % yield) as a mixture of the cis and trans isomers. LC-MS, [M+H] + = 479.

Examples 1 and 2

To a solution of compound 1B (l27mg, 0.265 mmol) in MeOH/THF (3 mL each) was added aq. 2M LiOH (3 mL, 6 mmol). The reaction mixture was heated at 50°C for 30 min, then was cooled to RT, acidified (1N aq. HC1) and extracted with EtOAc (2X). The combined organic extracts were washed with water, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was purified by reverse phase preparative HPLC (Phenomenex Lumina Axia; 30 X 250 mm column; continuous gradient from 50-100% MeOEl/EhO with 0.1% TFA) to separate the cis and trans cyclopentane isomers and gave the title compounds.

Example 1 (Peak 1 to elute): (racemic cis isomer) (8 mg, 7 % yield). LC-MS, [M+H] + = 451; ¾ NMR (500 MHz, DMSO-de) d 7.64 (br d, J=7.9 Hz, 2H), 7.49 - 7.20 (m, 4H), 6.98 (br d, J=1.9 Hz, 2H), 5.74 (m, 1H), 4.89 (m, 1H), 2.81 (m, 1H), 2.38 (m, 1H), 2.09 (s, 3H), 1.98 - 1.88 (m, 4H), 1.83 (m, 1H), 1.55 (m, 2H), 1.29 (m, 1H); hLPAl IC50 = 104 nM.

Example 2 (Peak 2 to elute): (racemic trans isomer) (41 mg, 34% yield). LC-MS, [M+H] + = 451; ¾ NMR (500 MHz, DMSO-de) d 7.65 (br d, J=1.9 Hz, 2H), 7.47 - 7.20 (m, 5H), 7.00 (br d, J=1.6 Hz, 2H), 5.75 (m, 1H), 4.97 (m, 1H), 2.94 (m, 1H), 2.19 - 1.96 (m, 7H), 1.79 (m, 2H), 1.65 (m, 2H), 1.28 (m, 1H); hLPAi IC50 = 31 nM.

Example 3. (±)-Trans-3-(4-(4-(((cyclopentyl(methyl)carbamoyl)oxy)methy l)-3- methylisoxazol-5-yl)phenoxy)cyclopentane-l -carboxylic acid

3 A. (5-(4-bromophenyl)-3-methylisoxazol-4-yl)methanol

To a solution of 5-(4-bromophenyl)-3-methylisoxazole-4-carboxylic acid

(synthesized according to the procedure described in US2011/82164 Al, 2.0 g, 7.09 mmol) in THF (50 mL) was added BH3 THF (28.4 mL of a 1M solution in THF, 28.4 mmol) portionwise at 0°C and the solution was allowed to warm to RT and stirred overnight at RT. The reaction mixture was carefully quenched with H2O, acidified with 1N aq. HC1 (50 mL), stirred for 1 h at RT, then was extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgSCE), and

concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 35-75% EtOAc in hexanes) to give the title compound (1.65 g, 87 % yield) as a white solid. LC-MS, [M+H] + = 268; ¾ NMR (CDCh , 400 MHz) d 7.73 - 7.64 (m, 4H), 4.66 (d, .7=5.1 Hz, 2H), 2.42 (s, 3H). 3B. 5-(4-bromophenyl)-3-methyl-4-(((tetrahydro-2H-pyran-2-yl)oxy )methyl)isoxazole

To a solution of compound 3A (626 mg, 2.33 mmol) in CH2CI2 (10 mL) was added 3,4-dihydro-2H-pyran (0.64 mL, 7.0 mmol) and PPTS (29 mg, 0.12 mmol). After stirring overnight at RT, the mixture was quenched with sat. aq. NaHCCb and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgSCri), and concentrated in vacuo. The residue was chromatographed (S1O2;

continuous gradient from 35-100% EtO Ac/Hexanes) to give the title compound (811 mg, 99 % yield) as a white solid. LC-MS, [M+H] + = 358; ¾ NMR (500 MHz, CDCh) d 7.82 - 7.55 (m, 4H), 4.69 (m, 1H), 4.65 (m, 1H), 4.46 (m, 1H), 3.87 (m, 1H), 3.54 (m, 1H),

2.37 (s, 3H), 1.86 - 1.55 (m, 6H).

3 C . 4-(3 -methyl -4-(((tetrahy dro-2H-pyran-2-yl)oxy)methyl)i soxazol-5 -yl)phenol

To a solution of KOH (2.70 g, 48.1 mmol) in H2O (50 mL) was added compound

3B (5.65g, 16.0 mmol) and dioxane (50 mL) and the solution was degassed with N2. /-BuXphos (0.545 g, 1.28 mmol) and Pd2(dba)3 (0.294 g, 0.321 mmol) were added and the suspension was degassed with N2, then was stirred at 90°C overnight. The reaction mixture was cooled to RT, acidified with 1N aq. HC1 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and

concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 25-75% EtO Ac/Hexanes) to give the title compound (3.63 g, 78 % yield) as a white solid. LC-MS, [M+H] + = 290; ¾ NMR (500 MHz, CDCh) d 7.72 (d, =8.8 Hz, 2H), 6.94 (d, =8.8 Hz, 2H), 4.70 (m, 1H), 4.66 (d, =l2.4 Hz, 1H), 4.48 (d, =l2.4 Hz, 1H), 3.89 (m, 1H), 3.53 (m, 1H), 2.36 (s, 3H), 1.88 - 1.70 (m, 2H), 1.65 - 1.57 (m, 4H).

3D. 5-(4-((tert-butyldimethylsilyl)oxy)phenyl)-3-methyl-4-(((tet rahydro-2H-pyran-2-yl) oxy)methyl)isoxazole

To a RT solution of compound 3C (2.0 g, 6.91 mmol) in DMF (50 ml) was added TBSC1 (2.08 g, 13.8 mmol) and imidazole (1.88 g, 27.7 mmol). The reaction was stirred at RT for 6 h, after which H2O (4 mL) was added. The mixture was partially concentrated in vacuo, diluted with H2O, acidified with 1N aq. HC1 and extracted with EtOAc (2x).

The combined organic extracts were washed with H2O, 10% aq. LiCl, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0-30% EtOAc/Hexanes) to give the title compound (2.55 g, 91 % yield) as a white solid. LC-MS, [M+H] + = 404; ¾ NMR (500 MHz, CDCh) d 7.75 - 7.67 (m, 2H), 6.93 (d, =8.8 Hz, 2H), 4.71 - 4.68 (m, 1H), 4.66 (d, =l2.4 Hz, 1H), 4.48 (d, =l2.4 Hz, 1H), 3.88 (m, 1H), 3.53 (m, 1H), 2.36 (s, 3H), 1.87 - 1.78 (m, 1H), 1.76 - 1.69 (m, 1H), 1.65 - 1.54 (m, 4H), 1.00 (s, 9H), 0.23 (s, 6H).

3E. (5-(4-((tert-butyldimethylsilyl)oxy)phenyl)-3-methylisoxazol -4-yl)methanol

To a RT solution of compound 3D (2.45g, 6.07 mmol) in MeOH (75 mL) was added PPTS (0.30 g, 1.21 mmol). The reaction was heated at 50°C for 2 h, then was cooled to RT and concentrated in vacuo. The mixture was diluted with TbO and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 25-50% EtOAc in hexanes) to give the title compound (1.18 g, 61 % yield) as a white solid. LC-MS, [M+H] + = 320; ¾ NMR (500 MHz, CDCh) d 7.68 (d, 7=8.8 Hz, 2H), 6.94 (d, 7=8.8 Hz, 2H), 4.63 (s, 2H), 2.37 (s, 3H), 1.00 (s, 9H), 0.24 (s, 6H).

3F. (5-(4-((tert-butyldimethylsilyl)oxy)phenyl)-3-methylisoxazol -4-yl)methyl (4-nitro- phenyl) carbonate

To a RT solution of compound 3E (l.8g, 5.63 mmol) in CH2CI2 (50 mL) were successively added pyridine (2.28 mL, 28.2 mmol) and 4-nitrophenyl chloroformate (1.82 g, 9.01 mmol) portionwise. The reaction mixture was stirred at RT for 1 h, then was concentrated in vacuo and the residue was chromatographed (S1O2; continuous gradient from 0-40% EtO Ac/Hexanes) to give the slightly impure title compound (2.95 g, 108 % yield) as a white solid. LC-MS, [M+H] + = 485. ¾ NMR (500MHz, CDCh) d 8.28 (d, .7=9.1 Hz, 2H), 7.68 (d, .7=8.8 Hz, 2H), 7.40 (d, 7=9.4 Hz, 2H), 6.98 (d, 7=8.8 Hz, 2H), 5.27 (s, 2H), 2.43 (s, 3H), 1.00 (s, 9H), 0.25 (s, 6H).

3G. (5-(4-((tert-butyldimethylsilyl)oxy)phenyl)-3-methylisoxazol -4-yl)methyl cyclo- pentyl(methyl)carbamate

To a RT solution of compound 3F (1.5 g, 3.10 mmol) in THF (24 mL) was added iPnNEt (1.62 mL, 9.29 mmol) and N-methylcyclopentanamine (0.61 g, 6.19 mmol). The reaction mixture was stirred overnight at RT, then was diluted with H2O and extracted with EtO Ac (2x). The combined organic extracts were washed with sat aq. NaHC03, H2O, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 10-40% EtO Ac/Hexanes) to give the title compound (1.08 g, 78 % yield) as a white solid. LC-MS, [M+H] + = 445; ¾ NMR (500 MHz, CDCb) d 7.67 (d, J=8.5 Hz, 2H), 6.94 (d, J=8.8 Hz, 2H), 5.08 (s, 2H), 4.70 - 4.29 (m, 1H), 2.76 (br s, 3H), 2.38 (s, 3H), 1.82 - 1.49 (m, 8H), 1.00 (s, 9H), 0.24 (s, 6H). 3H. (5-(4-hydroxyphenyl)-3-methylisoxazol-4-yl)methyl cyclopentyl(methyl)carbamate

To a solution of compound 3G (1.08 g, 2.429 mmol) in THF (24 mL) was added BU 4 NF (3.04 mL of a 1M solution in THF, 3.04 mmol). The reaction mixture was stirred at RT for 1.5 h, then was concentrated in vacuo, diluted with H2O, acidified with 1N aq. HC1 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgSCri), and concentrated in vacuo. The residue was

chromatographed (S1O2; continuous gradient from 25-60% EtO Ac/Hexanes) to give the title compound (619 mg, 77 % yield) as a white solid. LC-MS, [M+H] + = 331; ¾ NMR (500MHz, CDCb) d 7.63 (d, =8.5 Hz, 2H), 6.97 (d, =8.8 Hz, 2H), 5.12 (s, 2H), 4.79 - 4.19 (m, 1H), 2.78 (br s., 3H), 2.39 (s, 3H), 2.00 - 1.33 (m, 8H).

31. (±)-trans-benzyl 3-(4-(4-(((cyclopentyl(methyl)carbamoyl)oxy)methyl)-3-methyl - isoxazol-5-yl)phenoxy)cyclopentane-l-carboxylate

To a RT solution of compound 3H (60 mg, 0.182 mmol), Intermediate 12 (52.0 mg, 0.236 mmol), and Ph 3 P (62 mg, 0.24 mmol) in THF (3 mL) was added DEAD (0.14 mL, 0.236 mmol) dropwise. The reaction mixture was stirred overnight at RT, then was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 15-70% EtOAc in hexanes) to give the title compound (55 mg, 57 % yield) as a white solid. LC-MS, [M+H] + = 533; ¾ NMR (500 MHz, CDCb) d 7.70 (d, J=8.8 Hz, 2H), 7.35 (m, 5H), 6.94 (d, =8.8 Hz, 2H), 5.14 (s, 2H), 5.07 (s, 2H), 4.92 (m, 1H), 4.63 - 4.29 (m, 1H), 3.14 (m, 1H), 2.76 (br s, 3H), 2.38 (s, 3H), 2.21 (dd, J=8.4, 4.0 Hz, 2H), 2.14 (m, 2H), 1.99 - 1.89 (m, 2H), 1.67 (s, 8H).

Example 3

To a RT solution of compound 31 (55 mg, 0.103 mmol) in MeOH/THF (1 mL each) was added aq. 1M LiOH (1 mL, 1.0 mmol) and the reaction was stirred for lh at RT. The mixture was diluted with H2O, acidified with 1N aq. HC1 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried

(MgS0 4 ), and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC: Phenomenex Lumina Axia; 30X100 mm column; 60-100%

MeOH/H20 with 0.1% TFA; 10 min gradient to give the title compound (36 mg, 80 % yield) as a white solid. LC-MS, [M+H] + = 443; ¾ NMR (400 MHz, CD3OD) d 7.73 (d, J=9.0 Hz, 2H), 7.05 (d, J=9.0 Hz, 2H), 5.12 (s, 2H), 5.00 (m, 1H), 4.42 (m, 1H), 3.04 (m, 1H), 2.76 (s, 3H), 2.36 (s, 3H), 2.22 - 2.10 (m, 4H), 1.92 (m, 2H), 1.76 - 1.47 (m, 8H); hLPAi ICso = 171 nM.

Examples 4 and 5. (±)-Trans-2-(3-((6-(5-(((butyl(methyl)carbamoyl)oxy)methyl) -l- methyl-lH-l,2,3-triazol-4-yl)-2-methylpyridin-3-yl)oxy)cyclo pentyl)acetic acid

4A. (±)-trans-isopropyl 2-(3-((2-methyl-6-(l-methyl-5-(((tetrahydro-2H-pyran-2- yl)oxy)methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-yl)oxy)cyclop entyl)acetate

To a 0°C solution of 2-methyl-6-(l-methyl-5-(((tetrahydro-2H-pyran-2-yl)oxy) methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-ol (synthesized according to the procedure described for the synthesis of Example 1C in WO 2017/223016; 100 mg, 0.33 mmol), Intermediate 1 (92 mg, 0.49 mmol) and Ph 3 P (129 mg, 0.49 mmol) in THF (3 mL) was added DEAD (215 mg, 0.49 mmol, 40% in toluene) portionwise. The solution was allowed to warm to RT and stirred at RT overnight, then was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 35-100% EtOAc in hexanes) to give the title compound (120 mg, 77 % yield) as a white solid. LC-MS, [M+H] + = 473.

4B. (±)-trans-isopropyl 2-(3-((6-(5 -(hydroxymethyl)- 1 -methyl- 1H- 1 ,2,3-triazol-4-yl)-2- methylpyridin-3-yl)oxy)cyclopentyl)acetate

To a solution of compound 4A (1.21 g, 2.56 mmol) in MeOH (25 mL) was added p-TsOH (0.24 g, 1.28 mmol). The reaction was heated at 50°C for 3h, then was cooled to RT, partially concentrated in vacuo, and basified with satd aq. NaHCCb. The mixture was diluted with ThO and extracted with EtOAc (2x). The combined organic extracts were washed with ThO, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 35-100% EtO Ac/Hexanes) to give the title compound (650 mg, 65 % yield) as a white solid. LC-MS, [M+H] + = 389; ¾ NMR (500 MHz, CDCh) d 8.09 (d, J=8.8 Hz, 1H), 7.23 (d, J=8.5 Hz, 1H), 5.05 (m, 1H), 4.87 (m, 1H), 4.84 (s, 2H), 4.09 (s, 3H), 2.64 (m, 1H), 2.50 (s, 3H), 2.38 (d, J=1 A Hz, 2H), 2.25 - 2.09 (m, 3H), 1.91 (m, 1H), 1.61 (m, 1H), 1.37 (m, 1H), 1.26 (dd, J=6.2, 2.1 Hz, 6H).

4C. (±)-trans-isopropyl 2-(3-((2-methyl-6-(l-methyl-5-((((4-nitrophenoxy)carbonyl)ox y) methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-yl)oxy)cyclopentyl)a cetate

To a solution of compound 4B (122 mg, 0.31 mmol) in CH2CI2 (5 mL) were added pyridine (0.10 mL, 1.26 mmol), and 4-nitrophenyl chloroformate (95 mg, 0.47 mmol).

The reaction mixture was stirred at RT for 2 h, then was concentrated in vacuo. The residue was triturated with 1 : 1 EtO Ac/hexanes; a white solid was filtered off and the filtrate was concentrated in vacuo and chromatographed (S1O2; continuous gradient from 25-75% EtOAc in hexanes) to give the title compound (160 mg, 92 % yield) as a white solid. LC-MS, [M+H] + = 554.

Examples 4 and 5

To a solution of compound 4C (160 mg, 0.29 mmol) in THF (4 mL) were added iPnNEt (0.25 mL, 1.44 mmol) and N-methylbutan-l -amine (76 mg, 0.87 mmol). The reaction mixture was stirred at RT overnight, then was concentrated in vacuo. The residue was dissolved in EtOAc, washed with aq. 0.5 N aq. LiOH (2x), H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was taken up in MeOH and THF (3 mL each) and aq. 2M LiOH (3 mL, 6.0 mmol) was added. The reaction was heated at 50°C for lh, then was cooled to RT, diluted with H2O, acidified with 1N aq. HC1 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo to give the title compound (132 mg, 98 % yield) as a white solid. The racemate was separated into the individual enantiomers using preparative chiral SFC. Column: Chiralcel OJ-H, 21 x 250 mm, 5 micron Mobile Phase: 10% MeOH / 90% C02 . Flow Conditions: 45 mL/min, 120 Bar, 40°C. The first eluting enantiomer was the more active enantiomer. The absolute configuration of the two eluted enantiomers was not determined.

Example 4 (Enantiomer 1; Peak 1 to elute): (47 mg); LCMS, [M+H] + = 460; 'H NMR (500 MHz, CDCb) d 7.92 (br d, =8.5 Hz, 1H), 7.11 (br d, J=8.5 Hz, 1H), 5.75 (br d, J=5.5 Hz, 2H), 4.82 (m, 1H), 4.14 (s, 3H), 3.28 (m, 1H), 3.13 (m, 1H), 2.98 (m, 1H),

2.92 - 2.79 (m, 3H), 2.64 (br s, 1H), 2.45 (s, 3H), 2.24 - 2.08 (m, 3H), 1.93 - 1.84 (m,

1H), 1.68 - 1.47 (m, 2H), 1.42 - 1.24 (m, 4H), 1.15 (m, 1H), 0.99 - 0.74 (m, 3H); hLPAl IC50 = 15 nM.

Example 5 (Enantiomer 2; Peak 2 to elute): (53 mg); LCMS, [M+H] + = 460; ¾

NMR (500 MHz, CDCb) d 7.92 (br d, =8.3 Hz, 1H), 7.11 (br d, J=8.3 Hz, 1H), 5.76 (br d, =5.0 Hz, 2H), 4.82 (m, 1H), 4.14 (s, 3H), 3.29 (m, 1H), 3.14 (m, 1H), 2.98 (m, 1H),

2.93 - 2.79 (m, 3H), 2.64 (m, 1H), 2.45 (s, 3H), 2.24 - 2.08 (m, 3H), 1.95 - 1.85 (m, 1H), 1.65 - 1.48 (m, 2H), 1.41 - 1.28 (m, 4H), 1.16 (m, 1H), 0.96 - 0.75 (m, 3H); hLPAl IC50 = 952 nM.

Examples 6 and 7. (±)-Trans-2-((l,3)-3-((6-(5-((((cyclobutylmethyl)(methyl)ca rbamoyl) oxy)methyl)-l-methyl-lH-l,2,3-triazol-4-yl)-2-methylpyridin- 3-yl)oxy)cyclopentyl) acetic acid

6 A. 3 -(5 -bromo-6-methylpyridin-2-yl)prop-2-yn- 1 -ol

A solution of 3,6-dibromo-2-methylpyridine (3.0 g, 11.96 mmol), Et 3 N (5.00 mL, 35.9 mmol) and prop-2-yn-l-ol (1.044 mL, 17.93 mmol) in MeCN (25 mL) was degassed with N2 (sparged 3X with N2). Pd(dppf)Cl2 (0.874 g, 1.196 mmol) and Cul (0.114 g, 0.598 mmol) were added and the mixture was degassed with N2 again. The reaction mixture was heated at 50°C for 3h, then was cooled to RT. The mixture was filtered through a Celite plug, which was washed with EtOAc (3 X 50 mL). The combined filtrates were concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0% to 100% EtOAc in hexanes over 20 min) to give the title compound as a white solid (1.81 g, 67 % yield). ¾ NMR (500 MHz, CDCh) d 7.77 (d, J= 8.2 Hz,

1H), 7.14 (d, J= 8.2 Hz, 1H), 4.51 (s, 2H), 2.66 (s, 3H).

6B. (4-(5-bromo-6-methylpyridin-2-yl)- 1 -methyl- 1H- 1,2, 3-triazol-5-yl)methanol

To a solution of (pentamethylcyclopentadienyl)Ru(II)(Ph3P)2Cl (440 mg, 0.553 mmol) in l,4-dioxane (100 mL) was added compound 6A (5.00 g, 15.92 mmol). The mixture was degassed (evacuation/releasing into Ar; repeated 3X) and TMSCH2N3 (4.09 g, 28.8 mmol) was added. The mixture was degassed with Ar (evacuation/releasing into Ar; repeated 3X). The homoge-neous reaction mixture was then heated at 50°C for 20 h, then was cooled to RT and concentrated in vacuo. The residue was dissolved in THF (60 mL), after which TBAF (31.8 mL of a 1M solution in THF, 31.8 mmol) was added. The reaction mixture was stirred at RT for 30 min, then was quenched with satd aq. NaHCCb (50 mL) and extracted with EtOAc (4 x 50 mL). The combined organic extracts were washed with brine (40 mL), dried (MgSCL) and concentrated in vacuo. The crude product was chromatographed several times (330 g Gold ISCO S1O2 column; continuous gradient from 0% to 100% EtOAc in hexanes over 35 min, then hold at 100% EtOAc for 5 min) to afford the title compound (3.0 g, 10.60 mmol, 66.5 % yield) as a white solid. The regiochemistry of the triazole was confirmed by 1D-NOE NMR experiments. 'H

NMR (500 MHz, CDCh) d 8.02 (d, J= 8.2 Hz, 1H), 7.49 (d, J= 8.2 Hz, 1H), 4.82 (d, J =

6.3 Hz, 2H), 4.29 (s, 3H), 3.39 (br s, 1H), 2.77 (s, 3H).

6C. (4-(5-bromo-6-methylpyridin-2-yl)-l -methyl- 1H- 1,2, 3-triazol-5-yl)methyl (4- nitrophenyl) carbonate

To a solution of compound 6B and 4-nitrophenyl chloroformate (1645 mg, 8.16 mmol) in THF (50 mL) was added pyridine (1.32 mL, 16.32 mmol) at RT, after which a white solid was formed. The reaction mixture was stirred at RT for 16 h, then was concentrated in vacuo. The crude product was chromatographed (330 g S1O2; continuous gradient from 0 to 100% EtOAc in CH2CI2 over 15 min) to afford the title compound (2.3 g, 5.13 mmol, 94 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 8.34 - 8.27 (m, 2H), 7.98 (d, J= 8.4 Hz, 1H), 7.93 (d, J= 8.3 Hz, 1H), 7.43 - 7.36 (m, 2H), 6.05 (s, 2H),

4.25 (s, 3H), 2.70 (s, 3H); LCMS, [M+H]+ = 448.0.

6D. (4-(5-bromo-6-methylpyridin-2-yl)-l-methyl-lH-l,2,3-triazol- 5-yl)methyl

(cyclobutyl-methyl)(methyl)carbamate

To a solution of compound 6C (6.5 g, 14.50 mmol) in THF (100 mL) added 1- cyclobutyl-N-methylmethanamine (1.817 g, 17.40 mmol) and iPnNEt (10.13 mL, 58.0 mmol). The reaction mixture was stirred at RT for 16 h, then was concentrated in vacuo. The residue was chromatographed (330 g S1O2; continuous gradient from 0 to 50%

EtOAc in CH2CI2 over 20 min) to afford the title compound (6.0 g, 14.70 mmol, 101 % yield) as a white solid. ¾ NMR (400 MHz, CDCh; mixture of rotamers) d 7.96 - 7.78 (m, 2H), 5.75 (s, 1H), 5.73 (s, 1H), 4.16 (s, 1.5H), 4.13 (s, 1.5H), 3.32 (d, j= 7.4 Hz, 1H), 3.16 (d, j= 7.3 Hz, 1H), 2.88 (s, 1.5H), 2.78 (s, 1.5H), 2.68 (s, 3H), 2.64 - 2.50 (m, 0.5H), 2.39 (q, J= 7.9 Hz, 0.5H), 2.07 - 1.49 (m, 6H); LCMS, [M+H]+ = 409.0.

6E. (4-(5-hydroxy-6-methylpyridin-2-yl)-l -methyl- 1H-1, 2, 3-triazol-5-yl)methyl

(cyclobutyl-methyl)(methyl)carbamate

To a degassed (sparged with Ar 3X) solution of compound 6D (6.0 g, 14.7 mmol), bis(pinacolato)diboron (5.60 g, 22.04 mmol), and KOAc (5.77 g, 58.8 mmol) in 1,4- dioxane (100 mL) was added Pd(dppf)Cl2 (1.075 g, 1.470 mmol). The reaction was heated at 80°C for 16 h under Ar, then was cooled to RT. Water (50 mL) was added and the mixture was extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with water (50 mL), brine (50 mL), dried (MgSCri) and concentrated in vacuo.

The crude pinacol boronate was used in the next step without further purification.

To a 0°C solution of the crude pinacol boronate in EtOAc (75 mL) was added 30% aq. H2O2 (6.43 mL, 73.5 mmol) portionwise. The reaction was stirred at 0°C for 15 min, then was warmed to RT and stirred for 1 h at RT. The reaction was cooled 0°C and quenched with satd aq. Na2S203 (20 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo. The residue was heated at 60 °C in EtOAc (50 mL) and CH2CI2 (10 mL) until all solids were dissolved, then was cooled to RT. The solid was filtered off to afford the title compound as a slightly off-white solid (2.3 g). The filtrate was concentrated in vacuo, then was chromatographed (330 g ISCO Gold S1O2 column; continuous gradient from 0 to 100% EtOAc in CH2CI2 over 20 min, then hold at 100% EtOAc for 10 min) to afford an additional 2.0 g of title compound to provide a combined total of 4.3 g of title compound (12.45 mmol, 85 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 7.80 (d, J= 8.3 Hz, 1H), 7.08 (d, J= 8.3 Hz, 1H), 6.49 (s, 1H), 5.81 - 5.68 (m, 2H), 4.17 - 4.11 (m, 3H), 3.32 (d, j= 7.4 Hz, 1H), 3.18 (d, j= 7.3 Hz, 1H), 2.88 (s, 1.5H), 2.80 (s, 1.5H), 2.56 (q, J = 8.2 Hz, 0.5H), 2.50 (s, 3H), 2.48 - 2.37 (m, 0.5H), 2.07 - 1.47 (m, 6H); LCMS,

[M+H]+ = 346.3.

6F. (±)-Trans-isopropyl 2-(3-((6-(5-((((cyclobutylmethyl)(methyl)carbamoyl)oxy) methyl)-l-methyl-lH-l,2,3-triazol-4-yl)-2-methylpyridin-3-yl )oxy)cyclopentyl)acetate

To a RT solution of compound 6E (30 mg, 0.087 mmol), Intermediate 1 (29 mg, 0.156 mmol) and Ph 3 P (41.0 mg, 0.16 mmol) in THF (1 mL) was added DEAD (68 mg, 0.16 mmol, 40% in toluene) portionwise. The reaction mixture was stirred at RT for 48 h, then was concentrated in vacuo. The residue was chromatographed (SiCh; continuous gradient from 25-100% EtOAc in hexanes) to give the title compound (41 mg, 92 % yield) as a white solid. LC-MS, [M+H] + = 514.

Examples 6 and 7

To a solution of compound 6F (158 mg, 0.308 mmol) in 1 : 1 MeOH (1.5 mL each) was added aq. 2M LiOH (1.5 mL, 3.0 mmol, 2M). The reaction was heated at 50°C for lh, then was cooled to RT, diluted with H2O, acidified with 1N aq. HC1 to pH ~4 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was purified by reverse phase HPLC: Phenomenex Lumina Axia; 30X100 mm column; 60-100% CH3CN/H2O with 0.1% TFA; 10 min gradient to give the racemic title compound (115 mg, 79 % yield) as a white solid. This racemate (77 mg) was separated into the two individual enantiomers using preparative chiral SFC. Column: Chiralcel OJ-H, 30 x 250 mm, 5 Dm, Mobile Phase: 15% MeOH / 85% CO2. Flow Conditions: 85 mL/min, 120 Bar, 40°C. The absolute configuration of the eluted peaks was not determined.

Example 6: (Enantiomer 1; first eluting peak from chiral SFC; 18 mg; 23% yield); LCMS, [M+H] + = 472; ¾ NMR (500 MHz, CDCb) d 7.93 (br d, J=8.3 Hz, 1H), 7.11 (br d, =8.3 Hz, 1H), 5.79 - 5.71 (m, 2H), 4.83 (br s, 1H), 4.17 - 4.09 (m, 3H), 3.35 - 3.13 (m, 2H), 2.92 - 2.76 (m, 3H), 2.68 - 2.54 (m, 2H), 2.45 (s, 3H), 2.43 - 2.38 (m, 1H), 2.24 - 2.09 (m, 3H), 2.04 - 1.99 (m, 1H), 1.94 - 1.84 (m, 3H), 1.82 - 1.50 (m, 5H), 1.35 (br s, 1H); hLPAi IC50 = 11 nM.

Example 7: (Enantiomer 2; second eluting peak from chiral SFC; 19 mg; 25% yield); LCMS, [M+H] + = 472; ¾ NMR (500 MHz, CDCb) d 7.92 (br d, J=1 A Hz, 1H), 7.10 (br d, =8.0 Hz, 1H), 5.78 - 5.72 (m, 2H), 4.82 (br s, 1H), 4.16 - 4.11 (m, 3H), 3.34 - 3.13 (m, 2H), 2.91 - 2.76 (m, 3H), 2.69 - 2.52 (m, 2H), 2.45 (s, 3H), 2.42 - 2.38 (m, 1H), 2.22 - 2.10 (m, 3H), 2.04 - 1.97 (m, 1H), 1.94 - 1.83 (m, 3H), 1.80 - 1.51 (m, 5H), 1.34

(br s, 1H); hLPAi IC50 = 750 nM.

Example 8. (±)-Trans-2-(3-((6-(4-(((cyclopentyl(methyl)carbamoyl)oxy)m ethyl)-3- methylisoxazol-5-yl)-2-methylpyridin-3-yl)oxy)cyclopentyl)ac etic acid

8 A. /er/-Butyl 5-(5-bromo-6-methylpyridin-2-yl)-3-methylisoxazole-4-carboxy late

To a RT solution of 5-bromo-6-methylpicolinic acid (3.0 g, 13.9 mmol) in CH2CI2 (25 mL) and DMF (1 mL) under N2 was added SOCb (3.0 mL, 41.7 mmol) and the reaction mixture was stirred at 55 °C for 15 h, then was cooled to RT and concentrated in vacuo. The crude acid chloride product was dissolved in THF (10 mL) and added to a solution of /er/-butyl 3-(methylamino)but-2-enoate (4.67 g, 27.3 mmol) and pyridine (2.2 mL, 27.3 mmol) in THF (10 mL). The reaction mixture was stirred at RT for 24 h, then was concentrated in vacuo. The crude product was dissolved in EtOH (40 mL) and water (2 mL), after which NH2OH.HCI (1.98 g, 41.7 mmol) was added. The reaction mixture was stirred at 60 °C for 15 h, then was cooled to RT and concentrated in vacuo. Water (50 mL) was added and the mixture was extracted with EtOAc (2 x 50 mL). The combined organic extracts were washed with brine (50 mL), dried (Na2S0 4 ) and concentrated in vacuo. The crude product was chromatographed (24 g S1O2; 10% EtOAc in hexanes) to afford the title compound (2.55 g, 52%, for 3 steps) as an orange liquid. LC-MS, [M+H] + = 355; ¾ NMR (300 MHz, CDCh) d 8.16 (d, =8.l0 Hz, 1H), 7.67 (d, =8. l0 Hz, 1H), 2.73 (s, 3H), 2.48 (s, 3H), 1.49 (s, 9H).

8B. 5-(5-Bromo-6-methylpyridin-2-yl)-3-methylisoxazole-4-carboxy lic acid

To a stirred solution of compound 8 A (2.50 g, 7.08 mmol) in CH2CI2 (4 mL) was added TFA (3.82 mL, 49.5 mmol) and the reaction mixture was stirred at RT for 15 h, then was concentrated in vacuo to afford the title compound (1.75 g, 83%) as a yellow solid, which was used in the next reaction without further purification. LC-MS, [M+H] + = 297; ¾ NMR (400 MHz, CDCh) d 8.21 (d, J=8.80 Hz, 1H), 7.92 (d, J=8.80 Hz, 1H), 2.79 (s, 3H), 2.63 (s, 3H).

8C. (5-(5-Bromo-6-methylpyridin-2-yl)-3-methylisoxazol-4-yl)meth anol

To a 0°C solution of compound 8B (1.50 g, 5.05 mmol) in THF (80 mL) was added ethyl chloroformate (1.64 g, 12 mmol), TEA (1.41 mL, 10.1 mmol). The reaction was stirred at RT for 16 h, then was filtered through Celite and the filtrate was concentrated in vacuo. The residue was dissolved in EtOH (15 mL) and to this 0 °C solution was added NaBEL (0.573 g, 15.2 mmol). The reaction mixture was stirred at RT for 1 h, then was quenched with 0 °C aq. 1.5 N HC1 (50 mL) and extracted with CH2CI2 (2 x 50 mL). The combined organic extracts were washed with brine (50 mL), dried (Na2S0 4 ), filtered and concentrated in vacuo. The crude product was chromatographed (12 g S1O2; 20% EtOAc in n-hexanes) to afford the title compound (1.22 g, 85%) as a white solid. LC-MS, [M+H] + = 283; ¾ NMR (300 MHz, CDCh) d 8.00 (d, =8.40 Hz, 1H), 7.70 (d, =8.40 Hz, 1H), 4.62 (s, 2H), 2.75 (s, 3H), 2.36 (s, 3H).

8D. 5-(5-Bromo-6-methylpyridin-2-yl)-3-methyl-4-(((tetrahydro-2H -pyran-2yl)oxy) methyl) isoxazole

To a RT solution of compound 8C (1.20 g, 4.24 mmol) in l,4-dioxane (15 mL) was added 3,4-dihydro-2H-pyran (0.46 g, 5.51 mmol) and PPTS (0.533 g, 2.12 mmol). The reaction mixture was stirred at RT for 15 h, then was diluted with water (20 mL) and extracted with EtOAc (2 x 30 mL). The combined organic extracts were washed with brine (30 mL), dried (Na2S0 4 ), and concentrated in vacuo. The crude product was chromatographed (24 g S1O2; 20% EtOAc in n-hexanes) to afford the title compound (1.30 g, 84%) as a colorless liquid. LC-MS, [M+H] + = 367; ¾ NMR (400 MHz, CDCh) d 7.91 (d, =8.40 Hz, 1H), 7.59 (d, =8.40 Hz, 1H), 5.02 (ABq, ^=11.60 Hz, 2H), 4.75 (t, J=3.60 Hz, 1H), 4.00 - 4.10 (m, 1H), 3.80 - 3.95 (m, 1H), 2.70 (s, 3H), 2.41 (s, 3H), 1.40 - 1.90 (m, 6H).

8E. 3-Methyl-5-(6-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborol an-2-yl)pyridin-2-yl)-4 (((tetrahydro-2H-pyran-2-yl)oxy)methyl)isoxazole

To a degassed solution of compound 8D (1.30 g, 3.54 mmol), bis(pinacolato) diboron (1.80 g, 7.1 mmol) and KOAc (0.695 g, 7.08 mmol) in dioxane (40 mL) was added l,l'-bis(diphenyl- phosphino)ferrocene-Pd(II)Cl2-CH2Cl2 adduct (0.578 g, 0.708 mmol) and the reaction mixture was heated at 90 °C for 8 h, then was cooled to RT. The mixture was filtered through Celite, which was washed with EtOAc (50 mL). The combined filtrates were concentrated in vacuo to give the title compound product (1.25 g, 85%) as a colorless oil. This crude product was used in the next step without further purification. LC-MS, [M+H] + = 415.

8F. 2-Methyl-6-(3-methyl-4-(((tetrahydro-2H-pyran-2-yl)oxy)methy l)isoxazol-5- yl)pyridine-3-ol

To a mixture of compound 8E (1.25 g, 3.02 mmol) in THF (15 mL) and water (2 mL) was added NaBCh.LhO (1.21 g, 12.1 mmol) and the reaction mixture was stirred at 55 °C for 90 min, then was cooled to RT. The mixture was diluted with EtOAc (80 mL), washed with water (2 x 50 mL), dried (Na2S0 4 ) and concentrated in vacuo. The crude product was chromatographed (24 g S1O2, 25 % EtOAc in hexanes) to afford the title compound (0.78 g, 85%) as colorless oil. LC-MS, [M+H] + = 303; ¾ NMR (300 MHz, DMSO-de) d 10.44 (s, 1H), 7.59 (d, =8.40 Hz, 1H), 7.25 (d, =8.40 Hz, 1H), 5.03 (ABq, =l2.40 Hz, 2H), 4.70 (br. s., 1H), 3.75 - 3.90 (m, 1H), 3.40 - 3.50 (m, 1H), 2.39 (s, 3H), 2.29 (s, 3H), 1.35 - 1.80 (m, 6H).

8G. 5-(5-((tert-butyldimethylsilyl)oxy)-6-methylpyridin-2-yl)-3- methyl-4-(((tetrahydro- 2H-pyran-2-yl)oxy)methyl)isoxazole

To a solution of compound 8F in DMF (15 ml) was added TBSC1 (0.990 g, 6.57 mmol) and imidazole (0.895 g, 13.1 mmol). The reaction was stirred at RT for 3 days, after which H2O (4 mL) was added. The mixture was partially concentrated in vacuo, diluted with H2O, then was carefully neutralized with 1N aq. HC1 and extracted with

EtOAc (2x). The combined organic extracts were washed with H2O, 10% aq. LiCl, dried (MgSCri), and concentrated in vacuo. The residue was chromatographed (S1O2;

continuous gradient from 10-30% EtOAc in hexanes) to give the title compound (1.21 g, 88 % yield) as a white solid. LC-MS, [M+H] + = 419.

8H. (5-(5-((tert-butyldimethylsilyl)oxy)-6-methylpyridin-2-yl)-3 -methylisoxazol-4- yl)methanol

To a solution of compound 8G (1.21 g, 2.89 mmol) in MeOH (15 mL) was added PPTS (73 mg, 0.29 mmol). The reaction was heated at 50°C for 2 h, then was cooled to RT. Solid NaHC03 was added, and the mixture was concentrated in vacuo. The mixture was diluted with H2O, neutralized with 1N aq. HC1, and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 25-50% EtOAc in hexanes) to give the title compound (426 mg, 44 % yield) as a white solid. LC- MS, [M+H] + = 335. ¾ NMR (500 MHz, CDCb) d 7.74 (d, =8.3 Hz, 1H), 7.19 (d, J=8.5 Hz, 1H), 6.60 (t, =6.7 Hz, 1H), 4.60 (d, =6.9 Hz, 2H), 2.52 (s, 3H), 2.33 (s, 3H), 1.04 (s, 9H), 0.28 (s, 6H).

81. (5-(5-((tert-butyldimethylsilyl)oxy)-6-methylpyridin-2-yl)-3 -methylisoxazol-4- yl)methyl (4-nitrophenyl) carbonate

To a RT solution of compound 8H (426 mg, 1.27 mmol) in CH2CI2 (10 mL) were successively added pyridine (0.51 mL, 6.37 mmol) and 4-nitrophenyl chloroformate (513 mg, 2.55 mmol) portionwise. The reaction mixture was stirred at RT for lh, then was concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0-70% EtOAc in hexanes) to give the slightly impure title compound (666 mg,

105 % yield). LC-MS, [M+H] + = 500.

8J. (5-(5-hy droxy-6-methylpyri din-2 -yl)-3-methylisoxazol-4-yl)methylcy cl opentyl (methyl)carbamate

To a RT solution of compound 81 (666 mg, 1.333 mmol) in THF (10 mL) was added iPnNEt (1.16 ml, 6.67 mmol) and N-methylcyclopentanamine (397 mg, 4.00 mmol). The reaction mixture was stirred overnight at RT, then was concentrated in vacuo. The residue was dissolved in THF (10 mL), and 1M TBAF in THF (1.33 mL, 1.33 mmol) was added. The reaction was stirred at RT for 30 min, then was diluted with H2O, neutralized with 1N aq. HC1, and extracted with EtOAc (2x). The combined organic extracts were washed with H 2 0, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (Si0 2 ; continuous gradient from 20-75% EtOAc in hexanes) to give the title compound (330 mg, 72 % yield) as a white solid. LC-MS,

[M+H] + = 346; ¾ NMR (500 MHz, CDCh) d 7.46 (d, =8.5 Hz, 1H), 6.97 (d, =8.5 Hz, 1H), 5.59 (s, 2H), 4.49 (m, 1H), 2.77 (s, 3H), 2.46 (s, 3H), 2.39 (s, 3H), 1.77 (br s, 2H), 1.66 (m, 2H), 1.58 - 1.46 (m, 4H).

8K. (±)-Trans-isopropyl 2-(3-((6-(4-(((cyclopentyl(methyl)carbamoyl)oxy)methyl)-3- methylisoxazol-5-yl)-2-methylpyridin-3-yl)oxy)cyclopentyl)ac etate

To a RT solution of compound 8J (20 mg, 0.058 mmol), Intermediate 1 (19 mg, 0.10 mmol) and Ph 3 P (27 mg, 0.104 mmol) in THF (1 mL) was added DEAD (45 mg,

0.104 mmol, 40% in toluene). The reaction mixture was stirred at RT for 48 h, then was concentrated in vacuo. The residue was chromatographed (Si0 2 ; continuous gradient from 15-60% EtOAc in hexanes) to give the slightly impure title compound (31 mg,

104 % yield) as a white solid. LC-MS, [M+H] + = 514.

Example 8

To a solution of compound 8K (30mg, 0.058 mmol) in MeOH/THF (0.75 mL each) was added aq. 2M LiOH (0.75 mL, 1.5 mmol). The reaction was heated at 50°C for lh, then was cooled to RT, diluted with H 2 0, acidified with 1N aq. HC1 to pH ~4 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgSCri), and concentrated in vacuo. The residue was purified by preparative LC/MS using the following conditions: Column: XBridge Cl 8, 19 x 200 mm, 5 pm particles; Mobile Phase A: 5:95 MeCN:H 2 0 with 10 mM aq. NH 4 OAc; Mobile Phase B: 95:5 MeCN:H 2 0 with 10 mM aq. NH 4 OAc; Gradient: 20-60% B:A over 19 min, then a 5 min hold at 100% B; Flow: 20 mL/min. to give the title compound (16 mg, 58 % yield) as a white solid. LCMS, (M+H) + = 472; ¾ NMR (500 MHz, DMSO-de) d 7.69 (d, =8.5 Hz, 1H), 7.42 (d, =8.6 Hz, 1H), 5.31 (s, 2H), 4.93 (br s, 1H), 2.62 (br s, 3H), 2.43 - 2.37 (m, 1H), 2.35 (s, 3H), 2.28 (s, 4H), 2.20 - 2.10 (m, 1H), 1.99 - 1.91 (m, 2H), 1.73 - 1.64 (m, 1H), 1.62 - 1.30 (m, 9H), 1.29 - 1.19 (m, 1H); hLPAi ICso = 13 nM.

Example 9. (±)-Trans-2-(3-((2-methyl-6-(l-methyl-5-(((((l-propylcyclop ropyl)methoxy) carbonyl)amino)methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-yl)ox y)cyclopentyl)acetic acid

9A. (±)-trans-isopropyl 2-(3-((6-(5-(bromomethyl)-l-methyl-lH-l,2,3-triazol-4-yl)-2- methylpyridin-3-yl)oxy)cyclopentyl)acetate

To a 0°C solution of compound 4B (250 mg, 0.644 mmol) in DME (6 mL) was added PBn (0.121 mL, 1.29 mmol) and the reaction was allowed to warm to RT and stirred at RT overnight. The reaction was cooled to 0°C, neutralized with sat. aq.

NaHCCb, diluted with H2O and extracted with EtOAc (2x). The combined organic extracts were dried (MgSCri), and concentrated in vacuo to give the crude title compound (273 mg, 94 % yield) as a white solid. LC-MS, [M+H] + = 451. 9B. (±)-trans-isopropyl 2-(3-((6-(5-(aminomethyl)-l-methyl-lH-l,2,3-triazol-4-yl)-2- methylpyridin-3-yl)oxy)cyclopentyl)acetate

To a solution of compound 9A (273mg, 0.60 mmol) in DMF (3 mL) was added NaN3 (79 mg, 1.21 mmol). The reaction was stirred at 80 °C for 2 h, then was cooled to RT, diluted with H2O and extracted with EtOAc (2x). The combined organic extracts were washed with aq. 10% Li OH, dried (MgS0 4 ), and concentrated in vacuo. The residue was dissolved in THF (3 mL) and H2O (1 mL), after which Ph 3 P (175 mg, 0.665 mmol) was added. The reaction mixture was stirred at RT overnight, then was diluted with H2O and extracted with EtOAc (2x). The combined organic extracts were washed with brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0% to 15% MeOH in CH2CI2) to give the title compound (164 mg, 70 % yield). LCMS, [M + H] + = 388; ¾ NMR (500 MHz, CDCh) d 7.98 (d, =8.5 Hz, 1H), 7.15 (d, J=8.5 Hz, 1H), 5.04 (m, 1H), 4.85 (m, 1H), 4.19 (s, 2H), 4.11 (s, 3H), 2.65 (m, 1H), 2.49 (s, 3H), 2.37 (d, J=1 A Hz, 2H), 2.23 - 2.06 (m, 3H), 1.92 (m,

1H), 1.58 (m, 1H), 1.35 (m, 1H), 1.26 (dd, J=6.2, 1.8 Hz, 6H).

Example 9

To a solution of compound 9B (20 mg, 0.052 mmol) in THF (1 mL) was added iPnNEt (0.018 mL, 0.103 mmol) and Intermediate 7 (16 mg, 0.057 mmol). The reaction mixture was stirred at RT for 2 h, then was diluted with H2O and extracted with EtOAc (2x). The combined organic extracts were washed with aq. 0.5 N LiOH (2x), H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The crude carbamate ester product was dissolved in MeOH and THF (0.5 mL each) and aq. 2M LiOH (0.5 mL, 1.0 mmol) was added. The reaction was heated at 50°C for lh, then was cooled to RT, diluted with H2O, acidified with 1N aq. HC1 to pH ~6 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The crude material was purified by preparative LC/MS using the following conditions: Column: XBridge C18, 19 x 200 mm, 5 pm particles; Mobile Phase A: 5:95 MeCN:H20 with 10 mM aq. NH 4 OAc; Mobile Phase B: 95:5 MeCN:H 2 0 with 10 mM aq. NH 4 OAc; Gradient: 20-60% B:A over 20 min, then a 5 min hold at 100% B; Flow: 20 mL/min to give the title compound (11 mg, 44 % yield). LCMS, [M + H] + = 486; ¾ NMR (500 MHz, DMSO-de) d 7.82 (br d, =8.5 Hz, 1H), 7.61 (br s, 1H), 7.40 (br d, =8.6 Hz, 1H), 4.91 (br s, 1H), 4.72 (br s, 2H), 4.04 (s, 3H), 3.81 - 3.71 (m, 2H), 2.43 (m, 1H), 2.38 (s,

3H), 2.28 (m, 1H), 2.16 (m, 1H), 1.97 (m, 2H), 1.70 (m, 1H), 1.54 (m, 1H), 1.32 - 1.14 (m, 5H), 0.99 (br d, J=62 Hz, 1H), 0.78 (m, 3H), 0.38 - 0.15 (m, 4H); hLPAi ICso = 111 nM. Example 10. (±)-Trans-2-(3-((2-methyl-6-(l-methyl-5-(((methyl(l-propylc yclopropyl) carbamoyl)oxy)methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-yl)oxy )cyclopentyl)acetic acid

10A. 1 -propyl cy cl opropane-l -carboxylic acid

To a 0°C solution of cone. HC1 (7.00 mL, 85 mmol) in l,4-dioxane (20 mL) was added Intermediate 7A (500 mg, 2.71 mmol). The solution was allowed to warm to RT and stirred overnight at RT, then was diluted with H2O and extracted with EtOAc (3x). The combined organic extracts were dried (Na 2 S0 4 ), and concentrated in vacuo to give the title compound (339 mg, 97 %) as an oil. ¾ NMR (500 MHz, CDCh) d 1.52 (m, 4H), 1.29 (m, 2H), 0.92 (m, 3H), 0.78 (m, 2H).

10B. (±)-Trans-isopropyl 2-(3-((2-methyl-6-(l-methyl-5-((((l-propylcyclopropyl) carbamoyl)oxy)methyl)-lH-l,2,3-triazol-4-yl)pyridin-3-yl)oxy )cyclopentyl)acetate

To a solution of compound 10A (35 mg, 0.27 mmol) and Example 4B (35 mg,

0.09 mmol) in toluene (1.5 mL) were added Et 3 N (0.038 mL, 0.27 mmol) and

(PhO)2PON3 (0.039 mL, 0.18 mmol). The reaction mixture was heated at 80°C overnight, then was cooled to RT and concentrated in vacuo. The residue was

chromatographed (S1O2; continuous gradient from 40-75% EtOAc in hexanes) to give the title compound (18 mg, 39 % yield) as a white solid. LC-MS, [M+H] + = 514; ¾ NMR (500 MHz, CDCb) d 7.93 (d, J=8.5 Hz, 1H), 7.12 (d, J=8.5 Hz, 1H), 5.75 (s, 2H), 5.15 (br s, 1H), 5.04 (m, 1H), 4.83 (m, 1H), 4.12 (s, 3H), 2.63 (m, 1H), 2.47 (s, 3H), 2.36 (d, J=1 A Hz, 2H), 2.23 - 2.06 (m, 3H), 1.90 (m, 1H), 1.63 - 1.52 (m, 3H), 1.34 (m, 2H), 1.26 (dd, =6.3, 1.7 Hz, 6H), 0.93 (m, 3H), 0.77 (m, 2H), 0.66 (m, 2H).

Example 10

To a solution of compound 10B in DMF (1 mL) was added 60% NaH in oil (4 mg, 0.105 mmol). The reaction was stirred at RT for 5 min, then Mel (70 pL of a 2M solution in t-BuOMe, 0.140 mmol,) was added. The reaction mixture was stirred at RT for lh, then was quenched with H2O and extracted with EtOAc (2x). The combined organic extracts were washed with aq 10% Li Cl, dried (MgS0 4 ), and concentrated in vacuo. The crude residue was dissolved in MeOH and THF (0.75 mL each) and aq. 2M LiOH (0.75 mL, 1.5 mmol) was added. The reaction was heated at 50°C for lh, then was cooled to RT, diluted with H2O, acidified with 1N aq. HC1 to pH ~4 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The crude material was purified by preparative LC/MS using the following conditions: Column: XBridge Cl 8, 19 x 200 mm, 5-pm particles; Mobile Phase A: 5:95 MeCN:H 2 0 with 10 mM aq. MLOAc; Mobile Phase B: 95:5 MeCN:H 2 0 with 10 mM aq. NH 4 OAc; Gradient: 15-55% B:A over 20 min, then a 4 min hold at 100% B; Flow: 20 mL/min. to give the title compound (16 mg, 88 %). LCMS, (M+H) + = 486; ¾ NMR (500 MHz, DMSO-de) d 7.83 (d, =8.5 Hz, 1H), 7.40 (br d, =8.6 Hz, 1H), 5.61 (br s, 2H), 4.91 (br s, 1H), 4.14 - 4.06 (m, 3H), 2.78 - 2.69 (m, 3H), 2.43 (m, 1H), 2.35 (s, 3H), 2.30 (br d, J=1.2 Hz, 2H), 2.15 (m, 1H), 1.97 (m, 2H), 1.76 - 1.41 (m, 4H), 1.30 - 1.04 (m, 4H), 0.84 (m, 1H), 0.67 - 0.57 (m, 4H), 0.46 (m, 1H); hLPAi ICso = 372 nM.

Examples 11 and 12 were synthesized according to the methods described for the synthesis of Example 3 using Intermediate 12.

Example 13. (±)-Trans-3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoyl)o xy)methyl)-l- methyl-lH-l,2,3-triazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclo pentanecarboxylic acid

13A. (±)-Trans-tert-butyl 3-hydroxycyclopentanecarboxylate

A mixture of tert-butyl cyclopent-3-enecarboxylate (5.0 g, 29.7 mmol) and

Rh(I)(Ph3P)Cl (0.412 g, 0.446 mmol) in THF (50 mL) was cooled to -13 °C (internal temperature) and catecholborane (10.2 mL of a 50% solution in toluene; 38.6 mmol) was added portionwise over 10 min, during which a slight exotherm occurred. The reaction temperature was maintained at <-l l°C and the reaction was stirred for 1 h at -l0°C, then was slowly quenched with solid Na2HP0 4 (2.24 g, 15.8 mmol) while the temperature was kept at <0°C. Aq. 35% H2O2 (20.8 mL, 238 mmol) was added slowly dropwise to the reaction mixture while maintaining the reaction temperature at <0°C. The reaction was stirred for 1 h at 0°C, then was allowed to warm to RT and stirred at RT for 18 h. Water (20 mL) was added to the mixture, which was extracted with EtOAc (10 mL x 2). The combined organic extracts were washed with aq. 0.1N NaOH (3 X 10 mL; to remove catechol), brine, dried (MgSCri) and concentrated in vacuo. The crude oil was

chromatographed (80 g S1O2; continuous gradient from 0% to 100% EtOAc in hexane over 30 min) to give the title compound (4.50 g, 24.2 mmol, 81% yield) as a clear oil. ¾ NMR (400 MHz, CDCh) d 4.36 - 4.26 (m, 1H), 2.96 - 2.79 (m, 2H), 2.05 - 1.62 (m, 5H), 1.61 - 1.48 (m, 1H), 1.39 - 1.30 (m, 9H)

13B. (±)-Trans-tert-butyl 3-((3-methylpyrazin-2-yl)oxy)cyclopentanecarboxylate To a solution of compound 13A (100 mg, 0.537 mmol) in THF (1 mL) was added 1 M KOtBu in THF (0.644 mL, 0.644 mmol) at 0 °C under N2. After stirring for 10 min, 2-chloro-3-methylpyrazine (0.065 mL, 0.622 mmol) was added. The reaction mixture was slowly warmed to RT and stirred for 1 h at RT, then was quenched with water (2 mL) and 1 N aq. HC1 (3 mL) and extracted with EtOAc (5 mL). The organic layer was washed with brine, dried (MgSCh) and concentrated in vacuo. The crude oil was purified by preparative HPLC (Phenomenex Luna 5u C18 30 x 250 mm column; detection at 220 nm; flow rate = 30 mL/min; continuous gradient from 30% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90:10:0.1 H 2 0:MeCN:TFA and B = 90:10:0.1

MeCN:H 2 0:TFA) to give the title compound (87 mg, 0.313 mmol, 58.2 % yield) as a clear oil. ¾ NMR (400 MHz, CDCh) d 8.26 (d, J=2.9 Hz, 1H), 8.09 (d, =2.4 Hz, 1H), 5.63 - 5.53 (m, 1H), 3.06 - 2.90 (m, 1H), 2.57 (s, 3H), 2.32 - 2.05 (m, 4H), 2.02 - 1.80 (m, 2H), 1.47 (s, 9H).

13C. (±)-Trans-3-((5-bromo-3-methylpyrazin-2-yl)oxy)cyclopentane carboxylic acid

To a 0 °C solution of compound 13B (58 mg, 0.208 mmol) in DMF (1 mL) was added N-bromosuccinimide (74.2 mg, 0.417 mmol). The reaction was warmed to 90 °C, stirred for 3 days at 90 °C, then was cooled to RT and concentrated in vacuo. The residue was purified by preparative HPLC (Sunfire Cl8 30 x l00 mm column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 10% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 H 2 0:MeCN:TFA and B = 90: 10:0.1 MeCN:H 2 0:TFA) to give the title compound (3 mg, 9.96 pmol, 4.78 % yield) as a light brown oil. ¾ NMR (500 MHz, CDCh) d 8.03 (s, 1H), 5.49 (tt, =5.3, 2.6 Hz, 1H), 3.21 - 3.09 (m, 1H), 2.45 (s, 3H), 2.36 - 2.15 (m, 4H), 2.06 - 1.87 (m, 2H). 13D. (±)-Trans-ethyl 3-((5-bromo-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxylate

A mixture of compound 13C (4 mg, 0.013 mmol) and l-chloro-N,N,2- trimethylpropenylamine (3.5 pL, 0.027 mmol) in CH2CI2 (0.5 mL) was stirred at RT for 30 min, after which EtOH (0.1 mL) was added. The reaction was stirred for 30 min, then was concentrated in vacuo. The crude oil was chromatographed (4 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (4 mg, 0.012 mmol, 91 % yield) as a clear oil. ¾ NMR (500 MHz, CDCh) d 8.02 (d, J=0.8 Hz, 1H), 5.47 (tt, =5.4, 2.5 Hz, 1H), 4.18 (q, J=7.2 Hz, 2H), 3.07 (quin, J=8.0 Hz, 1H), 2.44 (s, 3H), 2.28 - 2.14 (m, 4H), 2.01 - 1.86 (m, 2H), 1.29 (t, J=7.2 Hz, 3H).

13E. ( ± )-Trans-ethyl 3 -((5 -(3 -hy droxyprop- 1 -yn- 1 -yl)-3 -methylpyrazin-2-yl)oxy)cy clo- pentanecarboxylate

To a degassed solution of compound 13D (0.12 g, 0.365 mmol), propargyl alcohol

(0.05 mL, 0.729 mmol), Et 3 N (0.10 mL, 0.729 mmol) and Cul (3.5 mg, 0.018 mmol) in DMF (5 mL) was added (Ph 3 P)4Pd(0); 20 mg, 0.018 mmol). The reaction was stirred at 50 °C for 16 h, then was cooled to RT and partitioned between EtOAc (3 mL) and water (5 mL). The aqueous phase was extracted with EtOAc (3 x 3 mL); the combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo. The crude oil was

chromatographed (12 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (0.10 g, 0.329 mmol, 90 % yield) as a yellowish oil. ¾ NMR (400 MHz, CDCh) d 8.05 (d, J=0A Hz, 1H), 5.51 (tt, =5.3, 2.6 Hz, 1H), 4.52 (s, 2H), 4.16 (q, J=1.3 Hz, 2H), 3.06 (quin, =8. l Hz, 1H), 2.42 (d, =0.4 Hz, 3H), 2.28 - 2.06 (m, 4H), 1.99 - 1.80 (m, 2H), 1.27 (t, J=1.2 Hz, 3H); [M + H] + = 305.1.

13F. (±)-Trans-ethyl 3-((5-(5-(hy droxymethyl)-l -methyl- 1H- 1,2, 3-triazol-4-yl)-3- methylpyrazin-2-yl)oxy)cyclopentanecarboxylate

To a degassed solution of compound 13E (100 mg, 0.329 mmol) in l,4-dioxane (5 mL), TMSCH2N3 (0.049 mL, 0.329 mmol), Cul (3.13 mg, 0.016 mmol), and

Ru(II)(Ph3P)2(pentamethylcyclopentadienyl)Cl (13.1 mg, 0.016 mmol) were added. The reaction mixture was stirred at 50°C for 2 h, then was cooled to RT. The cloudy reaction mixture was filtered and the filtrate was concentrated in vacuo to give a crude brown oil. The residue was dissolved in THF (5 mL); 1.0 M B NF in THF (0.657 mL, 0.657 mmol) was added, and the mixture was stirred at RT for 60 min, then was quenched with satd aq. NaHCCb (2 mL). The mixture was extracted with EtOAc (4 x 2 mL). The combined organic extracts were washed with brine (5 mL), dried (MgSCL) and concentrated in vacuo. The crude oil was chromatographed (12 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (70 mg, 0.194 mmol, 58.9 % yield) as a light yellowish oil. ¾ NMR (500M Hz, CDCh) d 8.88 (s, 1H), 6.11 (br. s., 1H), 5.61 (tt, =5.2, 2.5 Hz, 1H), 4.82 (s, 2H), 4.18 (q, J=1.2 Hz, 2H), 4.10 (s, 3H), 3.09 (quin, =8.2 Hz, 1H), 2.50 (s, 3H), 2.32 - 2.10 (m, 4H), 2.01 - 1.85 (m, 2H), 1.29 (t, J=1.2 Hz, 3H); [M + H] + = 362.0. l3G.(±)-Trans-ethyl 3-((3-methyl-5-(l-methyl-5-((((4-nitrophenoxy)carbonyl)oxy) methyl)-lH-l,2,3-triazol-4-yl)pyrazin-2-yl)oxy)cyclopentanec arboxylate

A solution of 4-nitrophenyl chloroformate (47 mg, 0.232 mmol) in CH2CI2 (0.5 mL) was added dropwise to a solution of compound 13F (70 mg, 0.194 mmol) and pyridine (0.08 mL, 0.968 mmol) in CH2CI2 (1 mL) over 1 h. The reaction was stirred at RT for 1 h, then was concentrated in vacuo. The crude oil was chromatographed (4 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (80 mg, 0.152 mmol, 78 % yield) as a light yellowish oil. . [M + H] + = 527.0

Example 13

To a solution of compound 13G (20 mg, 0.038 mmol) in CH2CI2 (1 mL) were added l-cyclobutyl-N-methylmethanamine (4.6 mg, 0.046 mmol) and iPnNEt (0.02 mL, 0.114 mmol). The reaction mixture was stirred at RT for 2 h, then was concentrated in vacuo. To the crude oily product was added 1.0 M aq. NaOH (0.370 mL, 0.370 mmol) in THF (0.5 mL)/MeOH (0.1 mL), and the reaction was stirred at RT for 3 h, then was concentrated in vacuo. The residue was purified by preparative HPLC (Sunfire C18 30 x 100 mm column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 10% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 H20:MeCN:TFA and B = 90: 10:0.1 MeCN/FLCfTFA) to give Example 1 compound (17 mg, 0.029 mmol, 79 % yield) as a clear oil. 1 H NMR (400 MHz, CDCh) d 8.70 (s, 1H),

5.73 - 5.53 (m, 3H), 4.16 (br. s., 3H), 3.32 (d, J=7.0 Hz, 1H), 3.25 - 3.05 (m, 2H), 2.93 -

2.73 (m, 3H), 2.65 - 1.43 (m, 16H); MS (ESI) m/z: 459.1 (M+H) + ; hLPAi ICso = 618 nM.

Example 14. (±)-trans -3-((5-(5-((((cyclo- pentylmethyl)(methyl)carbamoyl)oxy) methyl)-l -methyl- 1H-1, 2, 3-triazol-4-yl)-3-methylpyrazin-2-yl)oxy)

cyclopentanecarboxylic acid

Example 14 was synthesized according to the methods described and the intermediates used for the synthesis of Example 13. [M + H] + = 473.1; ¾ NMR (400 MHz, CDCb) d 8.66 (s, 1H), 5.69 - 5.56 (m, 3H), 4.18 (s, 3H), 3.28 - 3.04 (m, 3H), 2.94 - 2.80 (m, 3H), 2.46 (s, 3H), 2.32 - 2.17 (m, 4H), 2.09 - 1.89 (m, 3H), 1.76 - 1.39 (m, 6H),

1.27 - 0.95 (m, 2H); hLPAi ICso = 184 nM.

Example 15. (±)-Trans-3-((5-(5-((((benzyloxy)carbonyl)amino)methyl)-l-m ethyl-lH- l,2,3-triazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecar boxylic acid

15 A. (±)-Trans-ethyl 3-((5-(5-((((benzyloxy)carbonyl)amino)methyl)-l -methyl- 1H-1, 2,3- triazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxyla te

A solution of (±)-trans-ethyl 3-((5-(5-(hydroxymethyl)-l-methyl-lH-l,2,3- triazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxyla te (20 mg, 0.055 mmol), benzyl N-[(tert-butoxy)carbonyl]carbamate (21 mg, 0.083 mmol), n-B P (0.020 mL, 0.083 mmol), and l,l'-(azodicarbonyl)di piperidine (21 mg, 0.083 mmol) in THF (1 mL) was stirred at 50 °C for 3 h, then was cooled to RT. TFA (0.5 mL) was added, and the reaction was stirred at RT for 1 h, then was concentrated in vacuo. The crude oil was partitioned between EtOAc (2 mL) and water (2 mL). The organic layer was washed with water, dried (MgSCri), and concentrated in vacuo. The crude oil was chromatographed (4 g Si0 2 ; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (18 mg, 0.036 mmol, 65.8 % yield) as a clear oil. MS (ESI) m/z: 495.1 (M+H) + .

Example 15

A mixture of compound 15A (18 mg, 0.036 mmol) and 1.0 M aq. NaOH (0.37 mL, 0.37 mmol) in THF (0.5 mL)/MeOH (0.1 mL) was stirred at RT for 3 h, then was concentrated in vacuo. The residue was purified by preparative HPLC (Sunfire C18 30 x 100 mm column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 10% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 H 2 0:MeCN:TFA and B = 90: 10:0.1 MeCN:H 2 0:TFA) to give the title compound (9 mg, 0.015 mmol, 42.2 % yield) as a clear oil. [M - H] + = 467.1; ¾ NMR (400 MHz, CDCh) d 8.80 (s, 1H), 7.40 - 7.28 (m, 5H), 6.53 - 6.35 (m, 1H), 5.64 - 5.56 (m, 1H), 5.08 (s, 2H), 4.63 (br s, 2H), 4.22 (s, 3H), 3.21 - 3.08 (m, 1H), 2.46 (s, 3H), 2.30 - 2.17 (m, 4H), 2.06 - 1.91 (m, 2H); hLPAi ICso = 714 nM.

Example 16. (±)-Trans-3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoyl)o xy) methyl)- l-methyl-lH-pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopent anecarboxylic acid (racemate)

16A. (4-bromo- 1 -methyl- lH-pyrazol-5-yl)methanol

A mixture of 4-bromo-l-methyl-lH-pyrazole-5-carboxylic acid (4.9 g, 23.90 mmol) and BH3DTHF complex (36 ml, 35.9 mmol) in THF (50 ml) was stirred at RT for lh, then at 50 °C for 3 days and cooled to RT. The reaction was cautiously quenched with with 1N aq. HC1 (10 mL). The mixture was partitioned between CH2CI2 (20 mL) and water (20 mL); the aqueous phase was extracted with EtOAc (3 x 10 mL), and the combined organic layers were dried (MgSCri) and filtered through a pad of S1O2 to give the crude title compound (4.30 g, 22.5 mmol, 94 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 7.43 (s, 1H), 4.73 (br s, 2H), 3.98 (s, 3H), 3.53 - 3.36 (m, 1H).

16B. 4-bromo-l-methyl-5-(((tetrahydro-2H-pyran-2-yl)oxy)methyl)-l H-pyrazole

A mixture of compound 16A (4.20 g, 22.0 mmol), dihydropyran (4.0 mL, 44.0 mmol), and PPTS (0.55 g, 2.20 mmol) in CH2CI2 (20 mL) was stirred at RT for 18 h, then was concentrated in vacuo. The crude oil was chromatographed (80 g S1O2; continuous gradient from 0% to 40% EtOAc in hexane over 30 min) to give the title compound (5.90 g, 21.44 mmol, 98 % yield) as a clear oil. ¾ NMR (400 MHz, CDCh) d 7.41 (s, 1H),

4.76 - 4.54 (m, 3H), 3.93 (s, 3H), 3.88 (ddd, J=\ 1.4, 8.1, 3.2 Hz, 1H), 3.61 - 3.53 (m, 1H), 1.89 - 1.49 (m, 6H). 16C. l-Methyl-5-(((tetrahydro-2H-pyran-2-yl)oxy)methyl)-4-(4,4,5, 5-tetramethyl-l,3,2- di oxab orol an-2-y 1)- 1 H-py razol e

A mixture of compound 16B (2.00 g, 7.27 mmol), bis(pinacolato)diboron (2.77 g, 10.9 mmol), KOAc (2.85 g, 29.1 mmol) in l,4-dioxane (20 mL) was degassed with N2 for

5 min and Pd(dppf)Ch-CH2Cl2 (0.29 g, 0.363 mmol) was added. The reaction was stirred at 80 °C under N2 for 18 h, then was cooled to RT and partitioned between CH2CI2 and water (50 mL each). The mixture was stirred vigorously, after which the organic layer was dried (NaSCL), and concentrated in vacuo. The crude oil was chromatographed (80 g S1O2; continuous gradient from 0% to 100% EtOAc in hexane over 25 min) to give the title compound (0.90 g, 2.79 mmol, 38.4 % yield) as a light brownish oil. LCMS: m/z = 323.0; ¾ NMR (500 MHz, CDCh) d 7.69 (s, 1H), 4.94 - 4.76 (m, 2H), 4.69 (t, J=3.6 Hz, 1H), 3.95 - 3.86 (m, 4H), 3.58 - 3.48 (m, 1H), 1.74 - 1.46 (m, 6H), 1.30 (s, 12H). 16D. (±)-Trans-ethyl 3-((3-methyl-5-(l-methyl-5-(((tetrahydro-2H-pyran-2-yl)oxy) methyl)-lH-pyrazol-4-yl)pyrazin-2-yl)oxy)cyclopentanecarboxy late

A degassed mixture of compound 13D (0.15 g, 0.456 mmol), Example 16C compound (0.22 g, 0.683 mmol), K3PO4 (0.2 g, 0.911 mmol), and PdCl2(dppf)-CH2Cl2 adduct (0.02 g, 0.023 mmol) in l,4-dioxane (10 mL) and water (0.1 mL) was stirred at 80°C under N2 for 18 h, then was diluted with water (25 mL) and extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with water (50 mL), brine (50 mL), dried (Na2S0 4 ), and concentrated in vacuo. The crude oil was chromatographed (12 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (0.153 g, 0.344 mmol, 76 % yield) as a clear oil. LCMS: m/z = 445.0; ¾ NMR (400 MHz, CDCb) d 8.10 (s, 1H), 7.76 (s, 1H), 5.56 - 5.48 (m, 1H), 5.05 - 4.90 (m, 2H), 4.77 - 4.69 (m, 1H), 4.16 (q, J=7.2 Hz, 2H), 3.98 (s, 3H), 3.95 - 3.81 (m, 2H), 3.16 - 2.99 (m, 1H), 2.44 (d, 7=0.7 Hz, 3H), 2.23 - 2. l7 (m, 4H), 1.96 - 1.49 (m, 8H), 1.27 (t, J=7.2 Hz, 3H).

16E. (±)-Trans-ethyl 3-((5-(5-(hydroxymethyl)-l-methyl-lH-pyrazol-4-yl)-3-methyl- pyrazin-2-yl)oxy)cyclopentanecarboxylate

A mixture of compound 16D (153 mg, 0.344 mmol) and PPTS (86 mg, 0.344 mmol) in EtOH (5 mL) was stirred at 50 °C for 3 h, then was cooled to RT and concentrated in vacuo. The residue was partitioned between EtOAc and water. The organic layer was washed with water, dried (MgSCE), and concentrated in vacuo. The crude oil was chromatographed (4 g S1O2; continuous gradient from 0% to 100% EtOAc in hexane over 10 min) to give the title compound (100 mg, 0.277 mmol, 81 % yield) as a white solid. LCMS: m/z = 361.0; ¾ NMR (500 MHz, CDCb) d 8.22 (d, J=0.6 Hz, 1H), 7.75 (s, 1H), 5.99 (br. s., 1H), 5.52 (tt, 7=5.2, 2.4 Hz, 1H), 4.71 (br. s., 2H), 4.16 (q, 7=7.2 Hz, 2H), 3.94 (s, 3H), 3.07 (quin, 7=8.1 Hz, 1H), 2.46 (s, 3H), 2.24 - 2.13 (m, 4H), 1.97 - 1.86 (m, 2H), 1.27 (t, 7=7.2 Hz, 3H).

16F. (±)-Trans-ethyl 3-((3-methyl-5-(l-methyl-5-((((4 nitrophenoxy)carbonyl)oxy) methyl)-lH-pyrazol-4-yl)pyrazin-2-yl)oxy)cyclopentanecarboxy late

A solution of 4-nitrophenyl chloroformate (47.0 mg, 0.233 mmol) in CH2CI2 (3 mL) was added dropwise to a solution of compound 16E (70 mg, 0.194 mmol) and pyridine (0.079 mL, 0.971 mmol) in CH2CI2 (5 mL) over 1 h. The reaction was stirred at RT for 2 h, thenw was concentrated in vacuo. The crude product was chromatographed (4 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (90 mg, 0.171 mmol, 88 % yield) as a light yellow oil. [M + H] + = 526.0

16G. (±)-Trans-ethyl 3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoyl)oxy)methyl)- l- methyl-lH-pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentan ecarboxylate

To a solution of compound 16F (93 mg, 0.177 mmol) in THF (1 mL) was added (cyclobutylmethyl)(methyl)amine (21 mg, 0.21 mmol) and iPnNEt (0.124 mL, 0.708 mmol). The reaction mixture was stirred at RT for 2 h, then was concentrated in vacuo. The crude product was chromatographed (4 g S1O2; continuous gradient from 0% to 30% EtOAc in hexane over 10 min) to give the title compound (80 mg, 0.165 mmol, 93 % yield) as a clear oil. [M + H] + = 486.1

Example 16

A solution of compound 16G (7 mg, 0.014 mmol) and 1N aq. NaOH (0.22 mL,

0.22 mmol) in THF (0.5 mL) was stirred at RT for 18 h, then was concentrated in vacuo. The crude product was purified by preparative HPLC (Sunfire 08 30 x 100 mm- regenerated column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 30% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 H 2 0:MeCN: TFA and B = 90: 10:0.1 MeCN:H 2 0:TFA) to give the title compound (6 mg, 0.01 mmol, 71 % yield) as a clear oil. [M + H] + = 458.1; ¾ NMR (500 MHz, CDCb) d 8.15 (s, 1H), 7.87 (s, 1H), 5.60 - 5.54 (m, 1H), 5.50 (s, 2H), 4.03 (s, 3H), 3.39 - 3.11 (m,

3H), 2.96 - 2.80 (m, 3H), 2.65 - 2.40 (m, 4H), 2.34 - 2.17 (m, 4H), 2.09 - 1.69 (m, 7H), 1.65 - 1.54 (m, 1H); hLPAi ICso = 202 nM

Example 17. (±)-Trans-ethyl 3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoyl)oxy) methyl)-l -methyl- lH-pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclo-pentanecarbo xylate

(single enantiomer)

17A. (±)-Trans-Ethyl 3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoyl)oxy)methyl)- l- methyl-lH-pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentan e-l-carboxylate

One of enantiomers (30 mg, 99.5% ee) was separated from Example 16F (70 mg, 0.144 mmol) by chiral SFC (Instrument: PIC Solution SFC, Column: Chiralpak IC, 30 x 250 mm, 5 Dm; Mobile Phase: 40% MeOH/60% C0 2 , Flow Conditions: 85 mL/min, 110 Bar, 40°C, Detector Wavelength: 254 nm, Injection Details: 0.5 mL of 40 mg/mL solution in MeOH, peak 2). The absolute stereochemistry of Example 17A was not determined.

Example 17

A mixture of compound 17A (30 mg, 0.062 mmol) and 1 M aq. NaOH (0.62 mL,

0.618 mmol) in THF (1 mL) was stirred at RT for 3 h, then was concentrated in vacuo and purified by preparative HPLC (Sunfire 08 30 x 100 mm column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 30% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 EhCfMeCIOFA and B = 90: 10:0.1 MeCN:H20:TFA) to give the title compound (30 mg, 0.051 mmol, 83 % yield) as a clear oil. The absolute stereochemistry of Example 17 was not determined. [M + H] + = 458.0; 1 H NMR (500 MHz, CDCb) d 8.21 (s, 1H), 7.90 (s, 1H), 5.57 (br s, 1H), 5.48 (s, 2H), 4.05 (s, 3H), 3.38 - 3.20 (m, 2H), 3.16 (quin, J=8.0 Hz, 1H), 2.95 - 2.82 (m, 3H), 2.65 - 2.43 (m, 4H), 2.35 - 2.18 (m, 4H), 2.10 - 1.55 (m, 8H); hLPAi ICso = 416 nM.

Example 18. (±)-Trans-3-((5-(5-((((benzyloxy)carbonyl)amino)methyl)- 1 -methyl- 1H- pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxyli c acid

18A. (±)-Trans-ethyl 3-((5-(5-((((benzyloxy)carbonyl)amino)methyl)-l-methyl-lH- pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxyla te

A solution of compound 16E (33 mg, 0.092 mmol), benzyl N-[(tert- butoxy)carbonyl]carbamate (35 mg, 0.137 mmol), n-B P (0.034 mL, 0.137 mmol), and l,r(-azodicarbonyl)dipiperidine (35 mg, 0.137 mmol) in toluene (2 mL) was stirred at 50°C for 3 h, then was cooled to RT. TFA (1 mL) was added to the reaction mixture, which was stirred at RT for 1 h, then was concentrated in vacuo. The residue was partitioned between EtOAc and water. The organic layer was washed with water, dried (MgSCri) and concentrated in vacuo. The crude oil was chromatographed (4 g SiCh;

continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (40 mg, 0.081 mmol, 89 % yield) as a clear oil. ¾ NMR (500 MHz, CDCb) d 8.17 (s, 1H), 7.74 (s, 1H), 7.44 - 7.30 (m, 5H), 6.54 (br. s., 1H), 5.58 - 5.49 (m, 1H), 5.12 (s, 2H), 4.58 (d, =6. l Hz, 2H), 4.18 (q, J=7.2 Hz, 2H), 4.07 (s, 3H), 3.09 (quin, =8.0 Hz, 1H), 2.46 (s, 3H), 2.29 - 2.10 (m, 4H), 2.00 - 1.88 (m, 2H), 1.29 (t, J=7.2 Hz, 3H).

Example 18

A solution of compound 18A (6.9 mg, 0.014 mmol) and 1 N aq. NaOH (0.2 mL, 0.210 mmol) in THF (0.5 mL) was stirred at RT for 18 h, then was concentrated in vacuo. The crude product was purified by preparative HPLC (Sunfire Cl8 30 x l00 mm column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 30% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 H20:MeCN:TFA and B = 90: 10:0.1 MeCN:H 2 0:TFA) to give the title compound as the TFA salt (8 mg, 0.014 mmol, 97 % yield) as an oil. [M + H] + = 466.0; ¾ NMR (500 MHz, CDCb) d 8.22 (s, 1H), 7.91 (s, 1H), 7.40 - 7.34 (m, 5H), 5.61 - 5.53 (m, 1H), 5.13 (s, 2H), 4.61 (s, 2H),

4.15 (br. s., 3H), 3.22 - 3.12 (m, 1H), 2.50 (s, 3H), 2.35 - 2.20 (m, 4H), 2.08 - 1.92 (m, 2H); hLPAi ICso = 369 nM. Example 19. Trans-3-((5-(5-((((benzyloxy)carbonyl)amino)methyl)- 1 -methyl- 1H- pyrazol-4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxyli c acid, single enantiomer; absolute configuration not determined

19A. Trans-ethyl 3-((5-(5-((((benzyloxy)carbonyl)amino)methyl)-l-methyl-lH-py razol- 4-yl)-3-methylpyrazin-2-yl)oxy)cyclopentanecarboxylate (single enantiomer)

One of the two enantiomers were separated (30 mg, 99.5% ee) from Example 18A (80 mg, 0.165 mmol) by chiral SFC (Instrument: PIC Solution SFC, Column: Chiralpak

IC, 30 x 250 mm, 5 pm, Mobile Phase: 40%MeOH / 60% CO2, Flow Conditions: 85 mL/min, 110 Bar, 40°C, Detector Wavelength: 254 nm, Injection Details: 0.5 mL of 10 mg/mL solution in MeOH, 2 nd eluting peak under these conditions). The absolute stereochemistry of the compound was not determined.

Example 19

A mixture of compound 19A (14 mg, 0.028 mmol) and 1 M aq. NaOH (0.284 mL, 0.284 mmol) in THF (1 mL) was stirred at RT for 3 h, then was concentrated in vacuo. The residue was purified by preparative HPLC (Sunfire Cl8 30 x l00 mm column;

detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 30% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 FhCkMeCN/TFA and B = 90: 10:0.1 MeCN/EhCfTFA) to give the title compound (13 mg, 0.022 mmol, 78 % yield) as a clear oil. The absolute stereochemistry of the compound was not determined. [M + H] + = 466.0; ¾ NMR (500 MHz, CDCb) d 8.22 (s, 1H), 7.91 (s, 1H), 7.40 - 7.34 (m, 5H), 5.61 - 5.53 (m, 1H), 5.13 (s, 2H), 4.61 (s, 2H), 4.15 (br. s., 3H), 3.22 - 3.12 (m, 1H), 2.50 (s, 3H), 2.35 - 2.20 (m, 4H), 2.08 - 1.92 (m, 2H), hLPAi ICso = 202 nM.

The examples in the following table was synthesized according to the general methods described and the intermediates used for the synthesis of Example 4, 5 and 18.

Example 26. (±)-2-(Trans-3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoy l)oxy)methyl) - 1 -methyl- 1H- 1 ,2,3 -triazol-4-yl)-3 -methylpyrazin-2-yl)oxy)cyclopentyl)acetic acid and active enantiomer

26A. (±)-(4-(5-((trans-3-(2-diazoacetyl)cyclopentyl)oxy)-6-methy lpyrazin-2-yl)-l- methyl-lH-l,2,3-triazol-5-yl)methyl (cyclobutylmethyl)(methyl)carbamate

Oxalyl chloride (0.038 mL, 0.436 mmol) was added to a solution of Example 13 (100 mg, 0.218 mmol) in CH2CI2 (1 mL), along with a catalytic amount of DMF at 0 °C. The reaction was stirred at RT for 1 h, then was concentrated in vacuo. The residue was dissolved in THF/MeCN (1 :1; 1.0 mL) and TMSCHN2 (0.218 mL of a 2 M solution in hexane, 0.436 mmol) was added at 0 °C. The reaction was allowed to warm to RT and stirred at RT for 18 h, then was concentrated in vacuo. The crude oily product was chromatographed (4 g S1O2; continuous gradient from 0% to 50% EtOAc in hexane over 10 min) to give the title compound (53 mg, 0.110 mmol, 50.4 % yield) as a light yellowish oil. [M + H] + = 483.2

Example 26

A mixture of compound 26A (53 mg, 0.110 mmol) and silver benzoate (50 mg, 0.220 mmol) in l,4-dioxane (2 mLyEbO (0.2 mL) was stirred at 70 °C for 5 h, then was cooled to RT and diluted with EtOAc (5 mL). The mixture was washed with 1N aq. HC1 (5 mL), water (5 mL), and filtered. The filtrate was concentrated in vacuo. The crude oil was purified by preparative HPLC (Sunfire Cl8 30 x l00 mm column; detection at 220 nm; flow rate = 40 mL/min; continuous gradient from 30% B to 100% B over 10 min + 2 min hold time at 100% B, where A = 90: 10:0.1 H20:MeCN:TFA and B = 90: 10:0.1 MeCN:H20:TFA) to give the title compound (47 mg, 0.097 mmol, 89 % yield) as a light brownish oil. [M + H] + = 473.4; ¾ NMR (400 MHz, CDCh) d 8.65 (s, 1H), 5.64 (s, 2H), 5.54 - 5.39 (m, 1H), 4.17 (s, 3H), 3.40 - 3.10 (m, 2H), 2.94 - 2.75 (m, 3H), 2.69 - 1.47 (m, 18H), 1.42 - 1.21 (m, 1H); hLPAi ICso = 371 nM.

Examples 27 & 28. 2-(trans-3-((5-(5-((((cyclobutylmethyl)(methyl)carbamoyl)oxy ) methyl)-l-methyl-lH-l,2,3-triazol-4-yl)-3-methylpyrazin-2-yl )oxy)cyclopentyl)acetic acid, enantiomers

The enantiomers of Example 26 (41 mg, 0.087 mmol) were separated by chiral SFC (Instrument: PIC Solution SFC Prep-200, Column: Chiralpak OJ-H, 21 x 250 mm, 5 pm Mobile Phase: 10% MeOH / 90% CO2 Flow Conditions: 45 mL/min, 120 Bar, 40°C Detector Wavelength: 248 nm Injection Details: 0.5 mL of ~2l mg/mL in MeOH) to give the a faster eluting enantiomer (7.4 mg, 0.015 mmol, 17.7 % yield) and the other, slower eluting enantiomer (5.6 mg, 0.012 mmol, 13.4 % yield) as clear oils.

Example 27 (the faster eluting enantiomer; absolute stereochemistry

undetermined): [M + H] + = 473.3; ¾ NMR (400 MHz, CDCb) d 8.65 (s, 1H), 5.64 (s,

2H), 5.54 - 5.39 (m, 1H), 4.17 (s, 3H), 3.40 - 3.10 (m, 2H), 2.94 - 2.75 (m, 3H), 2.69 - 1.47 (m, 18H), 1.42 - 1.21 (m, 1H), hLPAi ICso = 260 nM.

Example 28 (the slower eluting enantiomer; absolute stereochemistry

undetermined): [M + H] + = 473.3; ¾ NMR (400 MHz, CDCb) d 8.65 (s, 1H), 5.64 (s, 2H), 5.54 - 5.39 (m, 1H), 4.17 (s, 3H), 3.40 - 3.10 (m, 2H), 2.94 - 2.75 (m, 3H), 2.69 -

1.47 (m, 18H), 1.42 - 1.21 (m, 1H); hLPAi ICso = 3750 nM.

Example 29: (±)2-(Trans-3-((6-(3-methyl-4-(((4-(pyridin-2-yl)pyrimidin- 2-yl)amino) methyl)isoxazol-5-yl)pyridin-3-yl)oxy)cyclopentyl)acetic acid

29A. (5-(5-bromopyridin-2-yl)-3-methylisoxazol-4-yl)methanol

A solution of 1M BH3.THF in THF (14.8 mL, 14.8 mmol) was added dropwise to a solution of 5-(5-bromopyridin-2-yl)-3-methylisoxazole-4-carboxylic acid (1.67 g, 5.90 mmol) (prepared according to the procedure of Nagasue, H., JP 2017095366) in THF (25 mL). The reaction was stirred at 60 °C for 2 h, then was cooled to 0°C and cautiously quenched with HO Ac (1 mL) and MeOH (10 mL) at 0°C. The mixture was allowed to warm to RT and stirred at RT for 30 min, then was concentrated in vacuo. MeOH (30 mL) was added and the mixture was stirred for 30 min at RT, then was concentrated in vacuo. The residue was diluted with satd aq. NaHC03 (15 mL) and extracted with EtOAc (5 x 20 mL). The combined organic extracts were washed with brine, dried (MgS0 4 ) and concentrated in vacuo. The crude product was chromatographed (40 g S1O2, continuous gradient from 0 to 100 % EtOAc over 15 min) to afford the title compound (1.20 g, 4.46 mmol, 76 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 8.75 (dd, J= 2.3, 0.7 Hz, 1H), 8.05 (dd, J= 8.5, 2.4 Hz, 1H), 7.90 (dd, J= 8.5, 0.8 Hz, 1H), 4.63 (s, 2H), 2.36 (s, 3H); [M+H] + = 269.

29B. 4-(bromomethyl)-5-(5-bromopyridin-2-yl)-3-methylisoxazole

PBn (0.21 mL, 2.23 mmol) was added to a solution of compound 29 A (200 mg, 0.743 mmol) in DME (10 mL) at 0 °C. The reaction mixture was allowed to warm to RT and stirred for 2 h at RT, then was cooled to 0°C and neutralized to pH 7 with sat’d aq. NaHC0 3. The mixture was partitioned between CH2CI2 (10 mL) and H2O (10 mL), and the aqueous layer was extracted with CH2CI2 (3 x 10 mL). The combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo. The residue was

chromatographed (24 g S1O2, continuous gradient from 0 to 50 % EtOAc in hexanes over 15 min) to provide the title compound (240 mg, 0.723 mmol, 97 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 8.82 (dd, J= 2.5, 0.7 Hz, 1H), 7.99 (dd, J= 8.5, 2.3 Hz, 1H), 7.83 (dd, j= 8.4, 0.8 Hz, 1H), 4.97 (s, 2H), 2.44 (s, 3H); [M+H] + = 330.9.

29C . N-((5 -(5 -bromopyri din-2 -yl)-3 -methyli soxazol-4-yl)methyl)-4-(pyridin-2-yl) pyrimidin-2-amine

To a solution of 4-(pyridin-2-yl)pyrimidin-2-amine (270 mg, 1.57 mmol) in THF (10 mL) was added n-BuLi (0.98 mL of a 1.6 M solution in hexane, 1.57 mmol) at -78 °C. The reaction mixture was allowed to warm to RT and stirred for 5 min at RT. This solution was added quickly to a solution of compound 29B (400 mg, 1.21 mmol) in THF (20 mL) at RT. The reaction mixture was stirred at RT for 24 h, then was diluted with H2O (10 mL) and extracted with EtOAc (3 x 5 mL). The combined organic extracts were dried (MgSCL) and concentrated in vacuo. The residue was chromatographed (40 g S1O2, continuous gradient from 0 to 100 % EtOAc in hexanes over 15 min) to provide the title compound (190 mg, 0.449 mmol, 37.3 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 8.69 (d, J= 2.2 Hz, 1H), 8.55 (dt, J= 4.8, 1.3 Hz, 1H), 8.29 (d, J= 5.2 Hz, 1H), 8.20 (d, J= 7.9 Hz, 1H), 7.84 (d, J= 2.4 Hz, 1H), 7.69 (d, J= 8.3 Hz, 1H), 7.66 (dd, J = 7.8, 1.8 Hz, 1H), 7.44 (d, J= 5.0 Hz, 1H), 7.23 (ddd, J= 7.5, 4.8, 1.2 Hz, 1H), 6.32 (t, J = 6.5 Hz, 1H), 4.71 (d, J= 6.4 Hz, 2H); [M+H] + = 423.1.

29D. 6-(3-methyl-4-(((4-(pyridin-2-yl)pyrimidin-2-yl)amino)methyl )isoxazol-5-yl) pyri din-3 -ol

A solution of compound 29C (180 mg, 0.425 mmol), bis(pinacolato) diboron (162 mg, 0.638 mmol), and KOAc (167 mg, 1.701 mmol) in l,4-dioxane (4 mL) was degassed by bubbling N2 through the solution. After 5 min, PdCl2(dppf) (31 mg, 0.043 mmol) was added and the mixture was further degassed by bubbling N2 through the solution for 2 min. The reaction mixture was sealed and heated at 80°C for 16 h, then was cooled to RT. Water (2 mL) was added and the mixture was extracted with EtOAc (3 x 5 mL).

The combined organic extracts were washed with brine (5 mL), dried (MgSCri) and concentrated in vacuo.

The crude product was dissolved in EtOAc (5 mL) and cooled to 0°C. Aq. 30% H2O2 (0.065 mL, 2.13 mmol) was added dropwise. The reaction was stirred at 0°C for 15 min, then was allowed to warm to RT, stirred at RT for 1 h, then was cooled to 0°C and quenched with satd aq. Na2S203 (5 mL) and extracted with EtOAc (3 x 5 mL). The combined organic extracts were washed with brine (5 mL), dried (MgS0 4 ) and concentrated in vacuo. The residue was chromatographed (12 g S1O2; continuous gradient from 0 to 100% EtOAc in CH2CI2, then hold at 100% EtOAc for 10 min) to afford the title compound (90 mg, 0.250 mmol, 58.7 % yield) as a white solid. ¾ NMR (500 MHz, DMSO-£¾) d 8.68 (d, J= 4.7 Hz, 1H), 8.44 (s, 1H), 8.35 (d, J= 2.8 Hz, 1H), 8.03 (br s, 1H), 7.93 - 7.77 (m, 2H), 7.50 (t, J= 6.0 Hz, 2H), 7.36 (dd, J= 8.6, 2.8 Hz, 1H). [M+H] + = 361.2.

Example 29

To a solution of compound 29D (8 mg, 0.022 mmol), (±)-(isopropyl 2-cis-3- hydroxycyclopentyl)acetate (8 mg, 0.044 mmol), and Ph 3 P (11 mg, 0.055 mmol) in toluene (1 mL) was added (E)-diazene-l,2-diylbis(piperidin-l-ylmethanone; 14 mg, 0.055 mmol). The reaction mixture was stirred overnight at RT, then was concentrated in vacuo. The residue was dissolved in a solution of LiOH.H20 (5 mg, 0.12 mmol) in 1 : 1 THF:water (1 mL each), and MeOH (1 mL) was added. The reaction mixture was stirred for 20 h at RT, then was acidified to pH ~ 4 with 1N aq. HC1 and extracted with EtOAc (3 x 5 mL), dried (Na2S0 4 ) and concentrated in vacuo. The crude residue was dissolved in DMF and purified by preparative LC/MS: Column: XBridge Cl 8, 19 x 200 mm, 5-pm particles; Mobile Phase A: 5:95 MeCN:H20 with 0.1% TFA; Mobile Phase B: 95:5 MeCN:H20 with 0.1% TFA; Gradient: 10-50% B over 20 min, then a 5-min hold at 100% B; Flow rate: 20 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation to provide the title compound (3.1 mg, 0.0052 mmol, 23% yield; purity by LCMS analysis = 100%). ¾ NMR (500 MHz, DMSO-i¾) d 8.67 (d, J= 4.7 Hz, 1H), 8.47 - 8.41 (m, 2H), 7.99 (br s, 1H), 7.86 (d, J= 8.8 Hz, 1H), 7.76 (br s, 1H), 7.55 (dd, j= 8.8, 3.0 Hz, 1H), 7.52 - 7.42 (m, 2H), 4.98 (s, 1H), 4.85 (s, 2H), 2.44 - 2.21 (m, 7H), 2.04 - 1.77 (m, 3H), 1.47 - 1.35 (m, 2H); [M+H] + = 487.2; hLPAl ICso = 54 nM.

Example 30. (±)-Cis-3-(((6-(5-((((cyclobutylmethyl)(methyl)carbamoyl)ox y)methyl)-l- methyl-lH-l,2,3-triazol-4-yl)-2-methylpyridin-3-yl)oxy)methy l)cyclopentane-l- carboxylic acid

30 A. 3 -(5 -bromo-6-methylpyridin-2-yl)prop-2-yn- 1 -ol

A solution of 3,6-dibromo-2-methylpyridine (3.0 g, 11.96 mmol), Et 3 N (5.00 mL, 35.9 mmol) and prop-2-yn-l-ol (1.044 mL, 17.93 mmol) in MeCN (25 mL) was degassed with N2 (sparged 3X with N2). Pd(dppf)Cl2 (0.874 g, 1.196 mmol) and Cul (0.114 g, 0.598 mmol) were added and the mixture was degassed with N2 again. The reaction mixture was heated at 50°C for 3h, then was cooled to RT. The mixture was filtered through a Celite plug, which was washed with EtOAc (3 X 50 mL). The combined filtrates were concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from 0% to 100% EtOAc in hexanes over 20 min) to give the title compound as a white solid (1.81 g, 67 % yield). ¾ NMR (500 MHz, CDCh) d 7.77 (d, J= 8.2 Hz,

1H), 7.14 (d, J= 8.2 Hz, 1H), 4.51 (s, 2H), 2.66 (s, 3H).

30B. (4-(5-bromo-6-methylpyridin-2-yl)-l -methyl- 1H- 1,2, 3-triazol-5-yl)methanol

To a solution of (pentamethylcyclopentadienyl)Ru(II)(Ph3P)2Cl (440 mg, 0.553 mmol) in l,4-dioxane (100 mL) was added compound 30A (5.00 g, 15.92 mmol). The mixture was degassed (evacuation/releasing into Ar; repeated 3X) and TMSCH2N3 (4.09 g, 28.8 mmol) was added. The mixture was degassed with Ar (evacuation/releasing into Ar; repeated 3X). The homoge-neous reaction mixture was then heated at 50°C for 20 h, then was cooled to RT and concentrated in vacuo. The residue was dissolved in THF (60 mL), after which TBAF (31.8 mL of a 1M solution in THF, 31.8 mmol) was added. The reaction mixture was stirred at RT for 30 min, then was quenched with satd aq. NaHCCb (50 mL) and extracted with EtOAc (4 x 50 mL). The combined organic extracts were washed with brine (40 mL), dried (MgSCL) and concentrated in vacuo. The crude product was chromatographed several times (330 g S1O2 column; continuous gradient from 0% to 100% EtOAc in hexanes over 35 min, then hold at 100% EtOAc for 5 min) to afford the title compound (3.0 g, 10.60 mmol, 66.5 % yield) as a white solid. The regiochemistry of the triazole was confirmed by lD-nOe NMR experiments. 'H NMR (500 MHz, CDCh) d 8.02 (d, J= 8.2 Hz, 1H), 7.49 (d, J= 8.2 Hz, 1H), 4.82 (d, J= 6.3 Hz, 2H), 4.29 (s, 3H), 3.39 (br s, 1H), 2.77 (s, 3H).

30C. (4-(5-bromo-6-methylpyridin-2-yl)- 1 -methyl- 1H- 1,2, 3-triazol-5-yl)methyl (4- nitrophenyl) carbonate

To a solution of compound 30B and 4-nitrophenyl chloroformate (1645 mg, 8.16 mmol) in THF (50 mL) was added pyridine (1.32 mL, 16.32 mmol) at RT, after which a white solid was formed. The reaction mixture was stirred at RT for 16 h, then was concentrated in vacuo. The crude product was chromatographed (330 g S1O2; continuous gradient from 0 to 100% EtOAc in CH2CI2 over 15 min) to afford the title compound (2.3 g, 5.13 mmol, 94 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 8.34 - 8.27 (m, 2H), 7.98 (d, J= 8.4 Hz, 1H), 7.93 (d, J= 8.3 Hz, 1H), 7.43 - 7.36 (m, 2H), 6.05 (s, 2H),

4.25 (s, 3H), 2.70 (s, 3H); LCMS, [M+H]+ = 448.0. 30D. (4-(5-bromo-6-methylpyridin-2-yl)-l -methyl- 1H- 1,2, 3-triazol-5-yl)methyl

(cyclobutyl-methyl)(methyl)carbamate

To a solution of compound 30C (6.5 g, 14.50 mmol) in THF (100 mL) added 1- cyclobutyl-N-methylmethanamine (1.817 g, 17.40 mmol) and iPnNEt (10.13 mL, 58.0 mmol). The reaction mixture was stirred at RT for 16 h, then was concentrated in vacuo. The residue was chromatographed (330 g SiCh; continuous gradient from 0 to 50%

EtOAc in CH2CI2 over 20 min) to afford the title compound (6.0 g, 14.70 mmol, 101 % yield) as a white solid. ¾ NMR (400 MHz, CDCh; mixture of rotamers) d 7.96 - 7.78 (m, 2H), 5.75 (s, 1H), 5.73 (s, 1H), 4.16 (s, 1.5H), 4.13 (s, 1.5H), 3.32 (d, J= 7.4 Hz, 1H), 3.16 (d, J= 7.3 Hz, 1H), 2.88 (s, 1.5H), 2.78 (s, 1.5H), 2.68 (s, 3H), 2.64 - 2.50 (m,

0.5H), 2.39 (q, J= 7.9 Hz, 0.5H), 2.07 - 1.49 (m, 6H); LCMS, [M+H]+ = 409.0.

30E. (4-(5-hydroxy-6-methylpyri din-2 -yl)-l-methyl-lH- 1,2, 3-triazol-5-yl)methyl (cyclobutyl-methyl)(methyl)carbamate

To a degassed (sparged with Ar 3X) solution of compound 30D (6.0 g, 14.7 mmol), bis(pinacolato)diboron (5.60 g, 22.04 mmol), and KOAc (5.77 g, 58.8 mmol) in l,4-dioxane (100 mL) was added Pd(dppf)Cl2 (1.075 g, 1.470 mmol). The reaction was heated at 80°C for 16 h under Ar, then was cooled to RT. Water (50 mL) was added and the mixture was extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with water (50 mL), brine (50 mL), dried (MgSCri) and concentrated in vacuo.

The crude pinacol boronate was used in the next step without further purification.

To a 0°C solution of the crude pinacol boronate in EtOAc (75 mL) was added 30% aq. H2O2 (6.43 mL, 73.5 mmol) portionwise. The reaction was stirred at 0°C for 15 min, then was warmed to RT and stirred for 1 h at RT. The reaction was cooled 0°C and quenched with satd aq. Na2S2Ch (20 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo. The residue was heated at 60 °C in EtOAc (50 mL) and CH2CI2 (10 mL) until all solids were dissolved, then was cooled to RT. The solid was filtered off to afford the title compound as a slightly off-white solid (2.3 g). The filtrate was concentrated in vacuo, then was chromatographed (330 g ISCO Gold S1O2 column; continuous gradient from 0 to 100% EtOAc in CH2CI2 over 20 min, then hold at 100% EtOAc for 10 min) to afford an additional 2.0 g of title compound to provide a combined total of 4.3 g of title compound (12.45 mmol, 85 % yield) as a white solid. ¾ NMR (500 MHz, CDCh) d 7.80 (d, J= 8.3 Hz, 1H), 7.08 (d, J= 8.3 Hz, 1H), 6.49 (s, 1H), 5.81 - 5.68 (m, 2H), 4.17 - 4.11 (m, 3H), 3.32 (d, j= 7.4 Hz, 1H), 3.18 (d, j= 7.3 Hz, 1H), 2.88 (s, 1.5H), 2.80 (s, 1.5H), 2.56 (q, J = 8.2 Hz, 0.5H), 2.50 (s, 3H), 2.48 - 2.37 (m, 0.5H), 2.07 - 1.47 (m, 6H); LCMS,

[M+H]+ = 346.3.

30F. Methyl cis-3-(((6-(5-((((cyclobutylmethyl)(methyl)carbamoyl)oxy)met hyl)-l- methyl-lH-l,2,3-triazol-4-yl)-2-methylpyridin-3-yl)oxy)methy l)cyclopentane-l- carboxylate

A mixture of compound 30E (100 mg, 0.290 mmol), cis-methyl 3- (hydroxymethyl) cyclopentanecarboxylate (82 mg, 0.521 mmol), n-B P (0.181 mL, 0.724 mmol) and (E)-diazene-l,2-diylbis(piperidin-l-ylmethanone) (183 mg, 0.724 mmol) in toluene (10 mL) was heated at 80°C for 2h, then was cooled to RT. The mixture was diluted with CH2CI2 (5 mL) and filtered through Celite ® , which was washed with additional CH2CI2 (5 mL). The combined filtrates were concentrated in vacuo , and the residue was chromatographed (S1O2; continuous gradient from 0% to 100% EtOAc in hexanes, 20 min) to give the title compound (141 mg, 0.290 mmol, 100 % yield) as an oil. ¾ NMR (400 MHz, CDCh) d 7.96 (d, J= 8.4 Hz, 1H), 7.16 (d, J= 8.6 Hz, 1H), 5.77 (br s, 2H), 4.16 (br s, 3H), 3.96 (dd, j= 6.7, 3.7 Hz, 2H), 3.71 (s, 3H), 3.34 (d, j= 7.4 Hz, 1H), 3.18 (d, J= 7.4 Hz, 1H), 2.95 - 2.77 (m, 4H), 2.64 - 2.52 (m, 1H), 2.50 (s, 3H), 2.48

- 2.36 (m, 1H), 2.31 - 1.51 (m, 13H); LCMS, [M+H]+ = 486.3.

Example 30

A mixture of compound 30F (141 mg, 0.290 mmol) and LiOH.H20 (61 mg, 1.45 mmol) in 1 : 1 THF : water (1 mL each) was stirred at RT for 4 h. The reaction was acidified to pH = 4 with 1N aq. HC1, extracted with EtOAc (3 x 5 mL). The combined organic extracts were dried (Na2S0 4 ) and concentrated in vacuo. The crude product was purified by preparative LC/MS using the following conditions: Column: XBridge Cl 8, 19 x 200 mm, 5-pm particles; Mobile Phase A: 5:95 MeCN:H20 with l0-mM aq. NH 4 OAc;

Mobile Phase B: 95:5 MeCN:H20 with l0-mM aq. NH 4 OAc; Gradient: 20-60% B over 20 min, then a 5-min hold at 100% B; Flow: 20 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation. The yield of the product was 96.0 mg, and its estimated purity by LCMS analysis was 100%. Two analytical LC/MS injections were used to determine the final purity. Injection 1 conditions:

Column: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7-mih particles; Mobile Phase A: 5:95 MeCN:H 2 0 with 10 mM MLOAc; Mobile Phase B: 95:5 MeCN:H 2 0 with 10 mM NH 4 OAc; Temperature: 50 °C; Gradient: 0-100% B over 3 min, then a 0.75-min hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm. Injection 2 conditions: Column: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7-mih particles; Mobile Phase A: 5:95 MeCN:H 2 0 with 0.1% TFA; Mobile Phase B: 95:5 MeCN:H 2 0 with 0.1% TFA; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-min hold at 100%

B; Flow: 1.0 mL/min; Detection: UV at 220 nm. LCMS, [M + H] + = 472.1 ; ¾ NMR (500 MHz, DMSO-r/e) d 7.84 (d, J= 8.5 Hz, 1H), 7.43 (d, J= 8.6 Hz, 1H), 5.62 (br s, 2H),

4.09 (br s, 3H), 3.97 (d, j= 6.6 Hz, 2H), 3.23 (br s, 1H), 3.10 (br s, 1H), 2.75 (br s, 3H), 2.71 (br s, 2H), 2.45 - 2.35 (m, 4H), 2.30 (s, 1H), 2.10 (dt, J= 12.7, 7.9 Hz, 1H), 1.98 - 1.39 (m, 10H); hLPAl ICso = 21 nM.

Examples 31 and 32

Example 31 Example 32

(enatniomer 2; arbitrary (enantiomer 2; arbitrary absolute stereochemistry) absolute stereochemistry)

The above racemic Example 30 was separated by chiral SFC using the following conditions: Column: Chiralcel OJ-H, 30 x 250mm, 5 pm; Flow Rate: 100 mL/min; Oven Temperature: 40°C; BPR Setting: 120 bar; ETV wavelength: 220 nm; Mobile Phase: 85% C02/l5% Isopropanol -0.1% DEA (isocratic); Injection: 1500 uL of 93 mg/5 mL Methanol chiral column to afford two enantiomers. The chiral purity of both compounds are determined to be >95% ee ud ^ er the following analytical conditions: Column:

Chiralcel OJ-H, 4.6 x 100 m, 5 m (analytical); Flow Rate: 2 mL/min; Oven

Temperature: 40°C; BPR setting: 1700 psi; ETV wavelength: 220 nm; Mobile Phase: 85% C0 2 /l5% iPrOH-0. l% Et 2 NH (isocratic).

Example 31 (first eluting enantiomer): ¾ NMR (500 MHz, DMSO-i¾) d 7.84 (d, J= 8.5 Hz, 1H), 7.43 (d, j= 8.6 Hz, 1H), 5.62 (br s, 2H), 4.09 (br s, 3H), 3.97 (d, j= 6.6 Hz, 2H), 3.23 (br s, 1H), 3.10 (br s, 1H), 2.75 (br s, 3H), 2.71 (br s, 2H), 2.45 - 2.35 (m, 4H), 2.30 (s, 1H), 2.10 (dt, J= 12.7, 7.9 Hz, 1H), 1.98 - 1.39 (m, 10H). LCMS, [M + H] + = 472.1; hLPAl ICso = 10.8 nM.

Example 32 (second eluting enantiomer): ¾ NMR (500 MHz, DMSO-r/r,) d 7.84 (d, ./ = 8.5 Hz, 1H), 7.43 (d, J= 8.6 Hz, 1H), 5.62 (br s, 2H), 4.09 (br s, 3H), 3.97 (d, J= 6.6 Hz, 2H), 3.23 (br s, 1H), 3.10 (br s, 1H), 2.75 (br s, 3H), 2.71 (br s, 2H), 2.45 - 2.35 (m, 4H), 2.30 (s, 1H), 2.10 (dt, J= 12.7, 7.9 Hz, 1H), 1.98 - 1.39 (m, 10H). LCMS, [M + H] + = 472.1; hLPAl ICso = 9.7 nM.

The examples in the following table were synthesized according to the methods and procedures described for the preparation of Examples 29 and 30.

Example 42. 3-((6-(3-methyl-4-(((4-phenylpyrimi din-2 -yl)amino)methyl)isoxazol-5- yl)pyri din-3 -yl)oxy)cy cl opentane-l -carboxylic acid

A mixture of compound 29D (200 mg, 0.56 mmol), ethyl 3 -hydroxy cyclopentane carboxylate (176 mg, 1.11 mmol), Et 3 N (0.16 mL, 1.11 mmol) and Ph 3 P (292 mg, 1.1 1 mmol) in THF (5 mL) was cooled 0 °C and DIAD (0.22 mL, 1.11 mmol) was added.

The reaction was stirred at RT overnight, then was concentrated in vacuo. The crude product was chromatographed (24 g SiCh; continuous gradient from 0-50% EtOAc in hexanes for 30 min, then isocratic at 50% EtOAc for 10 min) to give the ethyl ester product, which was used in the next step without further purification.

LiOEl.EhO (187 mg, 4.46 mmol) was added to a solution of the above ethyl ester in THF (2 mL), water (1 mL) and MeOH (1 mL) at RT. The reaction was stirred at RT overnight, then was concentrated in vacuo. The residue was diluted with H2O (5 mL), and the mixture was adjusted with aq. 1N HC1 to pH ~5 and extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (2 mL), dried (MgS0 4 ) and concentrated in vacuo to afford the crude product. This material was purified by preparative LC/MS with the following conditions: Column: XBridge C18, 19 x 200 mm, 5-mih particles; Mobile Phase A: 5:95 MeCN:H 2 0 with lO-mM aq. NH 4 OAc; Mobile Phase B: 95:5 MeCN:H 2 0 with lO-mM aq. NH 4 OAc; Gradient: 15-55% B over 20 min, then a 5-min hold at 100% B; Flow: 20 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation. The title compound was obtained (18.8 mg, 7 % yield) as an oil. LCMS, [M+H] + = 472.2; ¾ NMR (500 MHz, DMSO-de) d 8.51 - 8.28 (m, 2H), 8.02 - 7.90 (m, 2H), 7.86 (br d, =8.7 Hz, 1H), 7.60 - 7.34 (m, 5H), 7.16 (br d, =4.8 Hz, 1H), 5.11 - 5.04 (m, 1H), 4.90 - 4.73 (m, 2H), 3.00 -

2.89 (m, 1H), 2.32 (s, 3H), 2.21 - 1.97 (m, 4H), 1.87 - 1.75 (m, 2H); hLPAl ICso = 26 nM.

Example 42 was further separated into individual isomers by chiral preparative chromatography. Instrument: Berger MGII-SFC¾ ep-200 (HPW-L2501); Columns: Chiralpak IC and Chiralpak ID, 21 x 250 mm, 5 m; Mobile Phase: lst SFC: 45% MeOH w/ 0.1% DEA / 55% C02; Subsequent separations: IC or ID with 45% MeOH / 55% C0 2 ; Flow Conditions: 45 mL/min, 110 Bar, 40°C; Detector Wavelength: 220 nm; Injection Details: 1 mL injections of 3.34 mg / mL solution in MeOH w/ 0.1% DEA.

Example 43 (cis isomer - first eluting enantiomer): LCMS, [M+H] + = 472.0; 'H NMR (500 MHz, DMSO-de) d 8.39 - 8.36 (m, 1H), 8.32 - 8.26 (m, 1H), 7.95 - 7.77 (m, 3H), 7.51 - 7.31 (m, 5H), 7.10 (br d, =5.0 Hz, 1H), 4.96 - 4.89 (m, 1H), 4.83 - 4.71 (m, 2H), 2.75 - 2.63 (m, 1H), 2.34 - 2.18 (m, 5H), 1.96 - 1.76 (m, 4H); hLPAl ICso = 50 nM.

Example 44 (cis isomer - second eluting enantiomer): LCMS, [M+H] + = 472.0;

¾ NMR (500 MHz, DMSO-de) d 8.41 (br d, =2.l Hz, 1H), 8.35 - 8.29 (m, 1H), 7.97 - 7.87 (m, 2H), 7.85 (d, =8.7 Hz, 1H), 7.55 - 7.34 (m, 5H), 7.14 (br d, =5.0 Hz, 1H),

5.01 - 4.95 (m, 1H), 4.88 - 4.73 (m, 2H), 2.87 - 2.76 (m, 1H), 2.42 - 2.22 (m, 5H), 2.02 -

1.89 (m, 4H); hLPAl ICso = 158 nM.

Example 45 (trans isomer - first eluting enantiomer): LCMS, [M+H] + = 472.1; 'H NMR (500 MHz, DMSO-de) d 8.43 (d, =2.5 Hz, 1H), 8.35 - 8.29 (m, 1H), 7.97 - 7.88 (m, 2H), 7.85 (d, =8.8 Hz, 1H), 7.58 - 7.52 (m, 1H), 7.49 - 7.32 (m, 4H), 7.14 (d, =5.0 Hz, 1H), 5.08 - 5.02 (m, 1H), 4.89 - 4.73 (m, 2H), 2.99 - 2.89 (m, 1H), 2.31 (s, 3H), 2.20 - 1.99 (m, 4H), 1.85 - 1.74 (m, 2H); hLPAl ICso = 17 nM.

Example 46 (trans isomer - second eluting enantiomer): LCMS, [M+H] + = 471.9; ¾ NMR (500 MHz, DMSO-de) d 8.43 (d, J=23 Hz, 1H), 8.36 - 8.26 (m, 1H), 7.98 - 7.81 (m, 3H), 7.58 - 7.32 (m, 5H), 7.14 (br d, =5.0 Hz, 1H), 5.09 - 5.02 (m, 1H), 4.91 - 4.73 (m, 2H), 3.00 - 2.86 (m, 1H), 2.31 (s, 3H), 2.20 - 1.98 (m, 4H), 1.85 - 1.73 (m, 2H); hLPAl ICso = 56 nM.

The examples in the following table were synthesized according to the methods and procedures described for the synthesis of Examples 3-10 (using Intermediates 1-12).

Example 80. (±)-2-(3-((6-(5-(((5-(2-cyclobutylethyl)-l,2,4-oxadiazol-3- yl)amino)methyl)- 1 -methyl- 1H- 1 ,2,3 -triazol-4-yl)-2-methylpyri din-3 - yl)oxy)cyclopentyl)acetic acid

80A. (±)-Trans-isopropyl 2-(3-((6-(5-formyl-l -methyl- 1H-1, 2, 3-triazol -4-yl)-2- methylpyridin-3-yl)oxy)cyclopentyl)acetate

To a solution of compound 4B (300 mg, 0.77 mmol) in CH2CI2 (7.5 mL) was added NaHCCb (324 mg, 3.86 mmol) and Dess-Martin periodinane (393 mg, 0.93 mmol). The reaction was stirred at RT for 1 h, after which the white solids were filtered through Celite, which was rinsed with EtOAc. The combined filtrates were concentrated in vacuo. The residue was chromatographed (S1O2; continuous gradient from l0%-75% EtOAc/ hexanes) to give the title compound (280 mg, 94 % yield) as a white solid.

LCMS, [M+H] + = 387; ¾ NMR (500 MHz, CDCh) d 10.95 (s, 1H), 8.07 (d, =8.5 Hz, 1H), 7.14 (d, =8.5 Hz, 1H), 5.03 (m, 1H), 4.85 (m, 1H), 4.36 (s, 3H), 2.63 (m, 1H), 2.46 (s, 3H), 2.36 (d, J=1.2 Hz, 2H), 2.23 - 2.08 (m, 3H), 1.90 (m, 1H), 1.59 (m, 1H), 1.35 (m, 1H), 1.24 (dd, =6.2, 2.1 Hz, 6H).

Example 80

To a solution of compound 80A (30 mg, 0.078 mmol) and Intermediate 18 (26 mg, 0.155 mmol) in DCE (1 mL) was added HOAc (9 pL, 0.016 mmol, 10% in DCE). The reaction was stirred 30 min at RT and added NaBH(OAc)3 (49 mg, 0.23 mmol). The reaction mixture was stirred overnight, quenched with satd aq. NaHC0 3 , stirred for 15 min and extracted with EtOAc (2x). The combined organic extracts were washed with brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was taken up in MeOH and THF (0.75 mL each) and aq. 2M LiOH (0.75 mL, 1.5 mmol) was added. The reaction was heated at 50°C for lh, then was cooled to RT, diluted with H2O, neutralized with 1N aq. HC1, treated with pH 6 aq. phosphate buffer and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was purified by preparative LC/MS using the following conditions: Column: XBridge Cl 8, 200 mm x 19 mm, 5-pm particles; Mobile Phase A: 5:95 acetonitrile: water with 0.1% TFA; Mobile Phase B: 95:5 acetonitrile: water with 0.1% TFA; Gradient: a 0-min hold at 27% B, 27-67% B over 20 min, then a 4- min hold at 100% B; Flow Rate: 20 mL/min; to give the title compound (22 mg, 56 % yield). LC-MS, [M+H] + = 496; ¾ NMR (500 MHz, DMSO-de) d 7.84 (d, =8.5 Hz, 1H), 7.42 (d, =8.5 Hz, 1H), 7.21 (br t, J=5.5 Hz, 1H), 4.93 (m, 1H), 4.77 (br d, J=5.5 Hz, 2H), 4.09 (s, 3H), 2.60 (br t, =7.5 Hz, 2H), 2.43 (m, 1H), 2.38 (s, 3H), 2.30 (br d, =7.0 Hz, 2H), 2.22 (m, 1H), 2.15 (m, 1H), 2.03 - 1.92 (m, 4H), 1.82 - 1.67 (m, 5H), 1.61 - 1.50 (m,

3H), 1.26 (m, 1H); hLPAi ICso = 108 nM.

Example 81. (±)-Trans-2-(3-((6-(5-((isobutoxycarbonyl)amino)-l-methyl-l H-l,2,3- triazol-4-yl)-2-methylpyri din-3 -yl)oxy)cy cl opentyl)acetic acid

81 A. (±)-Trans-4-(5-((3-(2-isopropoxy-2-oxoethyl)cyclopentyl)oxy )-6-methylpyridin-2- yl)-l-methyl-lH-l,2,3-triazole-5-carboxylic acid

To a mixture of compound 80A (150 mg, 0.39 mmol), NaH 2 P04 (233 mg, 1.94 mmol), 2-methyl-2-butene, (1.55 mL of a 2.0M solution in THF; 3.11 mmol), H2O (0.4 mL), and t-BuOH (2 mL) at RT was added NaClCh (70 mg, 0.78 mmol). The reaction mixture was stirred at RT overnight, then was poured into water and extracted with EtOAc (2x). The combined organic extracts were dried (MgSCri) and concentrated in vacuo to give the title compound (140 mg, 90% yield). LC-MS, [M+H] + = 403; ¾ NMR (500 MHz, CDCh) d 8.35 (d, J=8.8 Hz, 1H), 7.42 (d, J=8.8 Hz, 1H), 5.02 (m, 1H), 4.91 (m, 1H), 4.45 (s, 3H), 2.61 (m, 1H), 2.55 (s, 3H), 2.37 (m, 2H), 2.29 - 2.07 (m, 3H), 1.91 (m, 1H), 1.64 (m, 1H), 1.37 (m, 1H), 1.24 (dd, J=6.3, 1.9 Hz, 6H).

81B. (±)-trans-isopropyl 2-(3-((6-(5-((isobutoxycarbonyl)amino)-l-methyl-lH-l,2,3- triazol-4-yl)-2-methylpyridin-3-yl)oxy)cyclopentyl)acetate

A mixture of compound 81 A (30 mg, 0.075 mmol), (PhO)2PON3 (96 pL, 0.447 mmol), 2-methylpropan-l-ol (11 mg, 0.149 mmol), TEA (42 pL, 0.298 mmol) in THF (0.5 mL) was stirred at 65°C for 2 h. The mixture was cooled to RT, diluted with satd aq. NaHCCb and extracted with EtOAc (2X). The combined organic extracts were washed with brine, dried (MgSCE), and concentrated in vacuo. The residue was chromatographed (S1O2, continuous gradient from 25% to 100% EtOAc/ hexanes) to afford the title compound (24 mg, 68 % yield). LC-MS, [M+H] + = 474.

Example 81

To a solution of compound 81B (24 mg, 0.051 mmol) in MeOH and THF (0.75 mL each) was added aq. 2M LiOH (0.75 mL, 1.5 mmol). The reaction was heated at 50°C for 30 min, then was cooled to RT, diluted with H2O, neutralized with 1N aq. HC1, treated with pH 6 aq. phosphate buffer and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was purified via preparative LC/MS with the following conditions: Column: XBridge C18, 200 mm x 19 mm, 5-pm particles; Mobile Phase A: 5:95 MeCN:H20 with l0-mM aq. NH 4 OAc; Mobile Phase B: 95:5 MeCN:H20 with l0-mM aq. NH 4 OAc;

Gradient: a 0-min hold at 20% B, 20-42% B over 25 min, then a 2-min hold at 100% B; Flow Rate: 20 mL/min; to give the title compound (13 mg, 57 % yield). LC-MS, [M+H] + = 432. ¾ NMR (500 MHz, DMSO-de) d 7.73 (br d, =8.5 Hz, 1H), 7.36 (br d, J=8.5 Hz, 1H), 4.90 (m, 1H), 3.86 (s, 3H), 3.50 - 3.24 (m, 1H), 2.42 (m, 1H), 2.33 (s, 3H), 2.27 (br d, =7.3 Hz, 2H), 2.13 (m, 1H), 2.00 - 1.91 (m, 2H), 1.89 (s, 1H), 1.69 (m, 1H), 1.54 (m, 1H), 1.24 (m, 1H), 0.83 (br s, 6H). hLPAi ICso = 613 nM. The examples in the following table were synthesized according to the methods and procedures described for the synthesis of Examples 80 and 81.

Example 84. (±)-Trans-3-(4-(5-(((cyclopentyl(methyl)carbamoyl)oxy)methy l)-l -methyl- lH-pyrazol-4-yl) phenoxy)cyclopentane-l -carboxylic acid

84A. tert-butyl 4-(4-hydroxyphenyl)- 1 -methyl-lH-pyrazole-5-carboxylate

To a degassed mixture of (4-hydroxyphenyl)boronic acid (58.1 mg, 0.421 mmol), tert-butyl 4-bromo-l-methyl-lH-pyrazole-5-carboxylate (100 mg, 0.383 mmol) and CS2CO3 (250 mg, 0.766 mmol) in DMF (5 mL) was added (Ph3P) 4 Pd° (44.3 mg, 0.038 mmol), and the reaction mixture was heated at 90 °C for 2 h, then was cooled to RT and concentrated in vacuo. The residue was partitioned between water and EtOAc (8 mL each); the aqueous layer was extracted with EtOAc (2 x 15 mL), dried (Na2S0 4 ) and concentrated in vacuo. The crude product was chromatographed (S1O2; continuous gradient from 15-20% EtOAc in hexanes) to give the title compound (80 mg, 0.289 mmol, 75 % yield) as a yellow solid. [M + H] + = 275.2; ¾ NMR (300 MHz, DMSO-de) d 9.43 (s, 1H), 7.51 (s, 1 H), 7.15 - 7.18 (d, =8.4 Hz, 2H), 6.75 - 6.78 (d, =8.4 Hz, 2H), 4.03 (s, 3H), 1.38 (s, 9H).

84B. Tert-butyl 4-(4-(((±)-trans-3-(ethoxycarbonyl)cyclopentyl)oxy)phenyl)- l-methyl- 1 H-py razol e- 5 -carb oxy 1 ate

To a 0°C solution of compound 84A (500 mg, 1.823 mmol) and (±)-trans-ethyl 3- hydroxy- cyclopentane carboxylate (577 mg, 3.65 mmol) in THF (25 mL) were added Ph 3 P (956 mg, 3.65 mmol) and di-tert-butyl azodi carboxyl ate (839 mg, 3.65 mmol). The reaction mixture was heated at 50°C for 16 h, then was cooled to RT and concentrated in vacuo. The residue was diluted with water (50 mL) and extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with water (50 mL), brine (50 mL), dried (Na2S0 4 ) and concentrated in vacuo. The crude material was chromatographed (24 g S1O2, isocratic 15% EtOAc in petroleum ether) to give the title compound (800 mg) as a gummy liquid. [M + H] + = 415.2.

84C. 4-(4-(((±)-trans-3-(ethoxycarbonyl)cyclopentyl)oxy)phenyl)- l-methyl-lH-pyrazole- 5-carboxylic acid

To a 0°C solution of compound 84B (0.80 g, 1.93 mmol) in DCM (5 mL) was added TFA (5.0 mL, 64.9 mmol). The reaction mixture was allowed to warm to RT and stirred at RT for 4 days, then was concentrated in vacuo. The residue was triturated with Et20 (2 X 25 mL) and dried under vacuum to give the title compound (400 mg, 59 % yield) as an off-white solid. [M + H] + = 359.2; ¾ NMR (300 MHz, DMSO-de) d 7.55 (s,

1 H), 7.18 - 7.35 (d, J=8.7 Hz, 2H), 6.88 - 6.91 (d, =8.7 Hz, 2H), 5.91 (m, 1H), 4.00- 4.15 (m, 5H), 2.95-3.03 (m, 1 H), 1.97 - 2.15 (m, 4 H), 1.75 - 1.85 (m, 2 H), 1.19 -1.20 (t, =7.2 Hz, 3H).

84D. Ethyl (±)-trans-3-(4-(5-(hydroxymethyl)-l-methyl-lH-pyrazol-4- yl)phenoxy)cyclopentane- 1 -carboxylate

To a 0° solution of compound 84C (400 mg, 1.116 mmol) in THF (10 mL) were added Et 3 N (0.17 mL, 1.23 mmol) and ethyl chloroformate (0.117 mL, 1.23 mmol). The reaction mixture was stirred at 0° for 1 h, then was diluted with EtOH (10 mL) and re- cooled to 0°C. NaBLL (84 mg, 2.23 mmol) was added, and the reaction was stirred at RT for 16 h, then was concentrated in vacuo. The residue was diluted with water (50 mL) and extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with water (50 mL), brine (50 mL), dried (Na2S0 4 ) and concentrated in vacuo. The crude material was chromatographed (12 g S1O2; continuous gradient from 15-30% EtOAc in petroleum ether) to give the title compound (50 mg, 13 % yield) as an off white solid. [M + H] + = 345.2; ¾ NMR (300 MHz, DMSO-de) d 7.51 (s, 1 H), 7.35 - 7.38 (d, J=9 Hz, 2H), 6.95 - 6.92 (d, J=9.2 Hz, 2H), 5.33 (t, =5.4 Hz, 1H), 4.88-4.95 (m, 1 H), 4.49 - 4.51 (m, 2 H), 4.00-4.15 (m, 2H), 3.86 (s, 3 H), 2.95-3.03 (m, 1 H), 1.97 - 2.15 (m, 4 H), 1.75 - 1.85 (m, 2 H), 1.19 -1.23 (t, =7.0 Hz, 3H).

84E. Ethyl (±)-trans-3-(4-(5-(((cyclopentylcarbamoyl)oxy)methyl)-l-met hyl-lH-pyrazol- 4-yl) phenoxy)cyclopentane-l-carboxylate

To a solution of compound 84D (50 mg, 0.145 mmol) and cyclopentanecarboxylic acid (33.1 mg, 0.290 mmol) in toluene (2 mL) were successively added TEA (0.040 mL, 0.290 mmol) and (PhO)2PON3 (0.038 mL, 0.174 mmol). The reaction mixture was heated at 1 l0°C for 16 h, then was cooled to RT and concentrated in vacuo. The crude product was chromatographed (4 g SiCh; isocratic at 25% EtOAc in petroleum ether) to give the title compound (40 mg, 60 % yield) as an off-white solid. [M + H] + = 456.4; ¾ NMR (300 MHz, DMSO-de) d 7.58 (s, 1 H), 7.31 - 7.34 (m, 2 H), 6.93 - 7.96 (m, 2 H), 5.08 (s, 1H), 4.92 - 4.98 (s, 1 H), 4.06 - 4.08 (q, =7.0 Hz, 2H), 3.87 (s, 3 H), 3.81 - 3.82 (m, 3 H), 2.95 - 3.05 (m, 1 H), 1.15 - 2.15 (m, 17 H).

84F. (±)-Trans-ethyl 3-(4-(5-(((cyclopentyl(methyl)carbamoyl)oxy)methyl)- 1 -methyl- 1H- pyrazol-4-yl)phenoxy)cyclopentanecarboxylate

To a 0°C solution of compound 84E (40 mg, 0.088 mmol) in DMF (2 mL), was added NaH (5.3 mg, 0.13 mmol). The reaction mixture was stirred at 0°C for 10 min, after which Mel (11 pL, 0.176 mmol) was added. The reaction mixture was allowed to warm to RT and stirred at RT for 2 h, then was quenched with ice-cold water (5 mL) and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with water (10 mL), brine (10 mL), dried (Na2S0 4 ) and concentrated in vacuo to give the title compound (7 mg, 18 % yield) as an off white solid. [M + H] + = 470.2.

Example 84

To a 0°C solution of compound 84F (40 mg, 0.09 mmol) in THF (1 mL) & MeOH (1 mL) was added a solution of LiOH.H 2 0 (10.7 mg, 0.256 mmol) in water (0.5 mL). The reaction mixture was stirred at RT for 5 h, then was diluted with water (5 mL) and washed with Et 2 0 (2 x 10 mL). The aqueous layer was acidified with 1.5N aq. HC1 and extracted with DCM (2 x 10 mL); the combined organic layers were dried (Na 2 S0 4 ) and concentrated in vacuo. The crude material was purified by reverse phase preparative HPLC (Column: KINETICS BIPHENY (250x21.2x5pm); M.Phase A: 0.1% TFA in H 2 0. M.Phase B: MeOH; Flow: 16 mL/min; Time (min)/%B: 0/90(90min)) to give the title compound (7 mg, 18 % yield) as an off-white solid. [M + H] + = 442.2; ¾ NMR (300 MHz, MeOH-d 4 ) d 7.58 (s, 1 H), 7.30 - 7.40 (m, 2 H), 6.90 - 7.00 (m, 2 H), 5.26 (s, 2 H), 4.92-4.98 (m, 1 H), 4.23 - 4.67 (m, 1 H), 3.97 (s, 3 H), 3.03-3.10 (m, 1 H), 2.79 (s, 3 H), 2.07 - 2.25 (m, 4 H), 1.85 - 2.04 (m, 2 H), 1.62-1.72 (m, 4 H), 1.48-1.60 (m, 4 H).

Example 85. (±)-Cis-2-(2-((6-(4-(((cyclopentyl(methyl)carbamoyl)oxy)met hyl)-3- methylisoxazol-5-yl)-2-methylpyridin-3-yl)oxy)cyclopentyl)ac etic acid

Example 85A. (±)-Trans-ethyl 2-(2-hydroxycyclopentyl)acetate

To a solution of ethyl 2-(2-oxocyclopentyl)acetate (1.0 g, 5.88 mmol) in EtOH (20 mL) at RT was added NaBEE (0.222 g, 5.88 mmol) and the solution was stirred at RT for lh. The reaction mixture was diluted with EhO, then was carefully acidified with 1N aq. HC1 and extracted with EtOAc (2x). The combined organic extracts were washed with EhO, brine, dried (MgSCri), and concentrated in vacuo. The residue was

chromatographed (S1O2; continuous gradient from 25-75% EtOAc in hexanes) to give the title compound (741 mg, 73 % yield) as an oil. ¾ NMR (500 MHz, CDCh) 5 4.15 (q, J=1.2 Hz, 2H), 3.87 (m, 1H), 2.87 (d, =2.8 Hz, 1H), 2.46 (dd, =l6.2, 5.8 Hz, 1H), 2.38 (dd, =l6.2, 8.8 Hz, 1H), 2.13 - 2.03 (m, 1H), 2.01 - 1.92 (m, 2H), 1.78 - 1.67 (m, 2H), 1.63 - 1.56 (m, 2H), 1.27 (t, J=12 Hz, 3H).

Example 85B. (±)-Cis-ethyl 2-(2-((6-(4-(((cyclopentyl(methyl)carbamoyl)oxy)methyl)- 3-methylisoxazol-5-yl)-2-methylpyridin-3-yl)oxy)cyclopentyl) acetate

To a RT solution of compound 8J (30 mg, 0.087 mmol), Example 85A (27 mg, 0.16 mmol) and Ph 3 P (41 mg, 0.16 mmol) in THF (1 mL) was added DEAD (68 mg, 0.16 mmol, 40% in toluene). The reaction mixture was stirred at RT for 48 h, then was concentrated in vacuo. The residue was chromatographed (Si0 2 ; continuous gradient from 15-60% EtOAc in hexanes) to give the slightly impure title compound (49 mg,

113 % yield) as a white solid. LC-MS, [M+H] + = 500.

Example 85

To a solution of Example 85B (43 mg, 0.086 mmol) in MeOH/THF (0.75 mL each) was added aq. 2M LiOH (0.75 mL, 1.5 mmol). The reaction was heated at 50°C for lh, then was cooled to RT, diluted with EhO, acidified with 1N aq. HC1 to pH ~4 and extracted with EtOAc (2x). The combined organic extracts were washed with H2O, brine, dried (MgS0 4 ), and concentrated in vacuo. The residue was purified by preparative LC/MS using the following conditions: Column: XBridge Cl 8, 19 x 200 mm, 5 pm particles; Mobile Phase A: 5:95 MeCN:H 2 0 with 10 mM aq. NH 4 OAc; Mobile Phase B: 95:5 MeCN:H 2 0 with 10 mM aq. NH 4 OAc; Gradient: 30-70% B over 19 min, then a 5 min hold at 100% B; Flow: 20 mL/min. to give the title compound (34 mg, 84 % yield. LCMS, (M+H) + = 472. ¾ NMR (500 MHz, DMSO-de) d 7.70 (d, J=8.5 Hz, 1H), 7.45 (d, =8.7 Hz, 1H), 5.35 (s, 2H), 4.86 (m, 1H), 2.65 (br s, 3H), 2.58 (m, 1H), 2.44 (m, 1H), 2.38 (s, 3H), 2.35 (m, 1H), 2.30 (s, 3H), 2.06 (m, 1H), 1.93 - 1.85 (m, 1H), 1.77 - 1.38 (m, 12H); hLPAi ICso = 29 nM.

The examples in the following table were synthesized according to the methods and procedures described for the synthesis of Example 4 and Example 1C in WO

2017/223016, except that 2,5-dibromo-6-ethyl-pyridine was used in the first step of the synthesis instead of 2,5-dibromo-6-methyl-pyridine.

The examples in the following table were synthesized according to the methods and procedures described for the synthesis of Example 85, but using triazole intermediates which were synthesized in an analogous fashion to Example 6E.