Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ISOXAZOLE ANALOGS AS FXR AGONISTS AND METHODS OF USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2019/118571
Kind Code:
A1
Abstract:
The present invention provides compounds of Formula (I), and pharmaceutically acceptable salts thereof, as well as pharmaceutical compositions comprising these compounds and methods of using these compounds to prevent or treat FXR-mediated or TGR5-mediated diseases or conditions.

Inventors:
OR YAT (US)
MA JUN (US)
WANG GUOQIANG (US)
XING XUECHAO (US)
SHEN RUICHAO (US)
WANG BIN (US)
Application Number:
PCT/US2018/065166
Publication Date:
June 20, 2019
Filing Date:
December 12, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ENANTA PHARM INC (US)
International Classes:
A61K31/423; A61K31/5377; A61P15/00; A61P29/00
Domestic Patent References:
WO2017201150A12017-11-23
Foreign References:
US9751874B22017-09-05
Other References:
DATABASE PUBCHEM COMPOUND 25 January 2017 (2017-01-25), ANONYMOUS: "2-(2-Bromo-5-methoxyphenyl)-4-hydroxybicyclo[1.1.0]butane-1-carboxylic acid", XP055618415, retrieved from NCBI Database accession no. CID123486225
Attorney, Agent or Firm:
ELMORE, Carolyn, S. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED:

1 A compound represented by Formula (I),

or a pharmaceutically acceptable salt thereof, wherein:

R1 is hydrogen, halogen, cyano, optionally substituted -C1-C6 alkyl, optionally substituted - C2-C6 alkenyl, optionally substituted -C2-C6 alkynyl, optionally substituted -C3-C6 cycloalkyl or optionally substituted 3- to 6- membered heterocycloalkyl;

R2 is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted -C3- C 12 cycloalkyl or optionally substituted 3- to 12- membered heterocycloalkyl;

R3a and R3b are each independently selected from the group consisting of hydrogen, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C1-C6 alkoxy and optionally substituted -C3-C6 cycloalkyl; alternatively, R3a and R3b are taken together with the carbon atom to which they are attached to form optionally substituted -C3-C6 cycloalkyl, optionally substituted 3- to 6- membered heterocycloalkyl, or optionally substituted -C3-C6

cycloalkenyl;

L1, L2, L3 are each independently C or N;

e, f, m, and n are each independently 0, 1, 2, 3, or 4;

A1 is O, NR3c, S, S(O) or S(0)2;

A2 is absent, O, NR3c, S, S(O) or S(0)2;

A1 is attached to -CR3aR3b-, and A2 is attached to ® ;

Each X1, X2, X3 and X4 is independently selected from the group consisting of: O, C(O), S, S(O), S(0)2, NR3c, and CR3dR3e; wherein R3d and R3e are each independently selected from hydrogen, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, and optionally substituted -O-C1-C6 alkyl; alternatively, R3d and R3e are taken together with the carbon atom to which they are atached to form a optionally substituted -C3- C6 cycloalkyl or optionally substituted 3- to 6- membered heterocycloalkyl; R3c is hydrogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, formyl, or acetyl;

Y1 is absent when L1 is N, and hydrogen, hydroxyl, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted -O-C1-C6 alkyl when L1 is C; Y3 is absent when L3 is N, and hydrogen, hydroxyl, halogen, optionally substituted - C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted -O-C1-C6 alkyl when L3 is C; Y4 is absent when L4 is N, and hydrogen, hydroxyl, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted - O-Ci-Ce alkyl when L4 is C; Y2 is hydrogen, hydroxyl, halogen, optionally substituted -Ci- C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted -O-Ci-Ce alkyl;

provided that ® is not

selected from hydrogen, halogen, optionally substituted -Ci-C3 alkyl, optionally substituted - C3-Ce cycloalkyl, optionally substituted -0-Ci-C3 alkyl, alternatively two R3q groups are linked together to form a -C3-C6 cycloalkyl, aryl, heteroaryl or 3- to 6- membered heterocycloalkyl;

® is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted 3- 12 membered heterocycloalkyl;

Z is selected from the group consisting of:

1) Absent;

2) Optionally substituted -C1-C6 alkyl;

3) Optionally substituted -C2-C6 alkenyl;

4) Optionally substituted -C2-C6 alkynyl;

5) Optionally substituted -C3-C8 cycloalkyl;

6) Optionally substituted 3- to 8- membered heterocycloalkyl;

7) Optionally substituted -C3-C8 cycloalkenyl;

8) Optionally substituted aryl; and

9) Optionally substituted heteroaryl; R4 is hydroxy, protected hydroxy, -0-(hydroxy prodrug group), tetrazolyl, cyano, -CO2R5, -

Y is absent or optionally substituted -C1-C6 alkyl;

R4a and R4b are independently selected from the groups consisting of:

1) Hydrogen;

2) Optionally substituted -Ci-Ce alkyl;

3) Optionally substituted -C2-C8 alkenyl;

4) Optionally substituted -C2-C8 alkynyl; and

5) Optionally substituted -C3-C8 cycloalkyl;

R4c is hydrogen or optionally substituted -C1-C6 alkyl;

R5 is selected from the groups consisting of:

1) Hydrogen;

2)

3) Optionally substituted -Ci-Ce alkyl;

4) Optionally substituted -C2-C8 alkenyl;

5) Optionally substituted -C2-C8 alkynyl; and

6) Optionally substituted -C3-C8 cycloalkyl;

R7 is selected from the groups consisting of:

1) Optionally substituted -Ci-Ce alkyl;

2) Optionally substituted -C2-C8 alkenyl;

3) Optionally substituted -C2-C8 alkynyl;

4) Optionally substituted -C3-C8 cycloalkyl;

5) Optionally substituted -C3-C8 cycloalkenyl;

6) Optionally substituted 3- to 8-membered heterocycloalkyl;

7) Optionally substituted 3- to 8- membered heterocycloalkenyl;

8) Optionally substituted aryl; 9) Optionally substituted -Ci-Ce arylalkyl;

10) Optionally substituted heteroaryl;

11) Optionally substituted -Ci-Ce heteroarylalkyl; and

12)NR9R10; wherein R9 and R10 are each independently selected from hydrogen, optionally substituted -Ci-Cs alkyl, optionally substituted -C2-C8 alkenyl, optionally substituted -C2-C8 alkynyl, optionally substituted -C3-C8 cycloalkyl, optionally substituted aryl, optionally substituted alkylaryl, optionally substituted 3- to 8- membered heterocycloalkyl, optionally substituted heteroaryl, optionally substituted alkylheteroaryl; alternatively, R9 and R10 are taken together with the nitrogen atom to which they are attached to form a heterocyclic ring.

The compound of claim 1, wherein ® is selected from the groups set forth below:

wherein each of said groups is optionally further substituted.

3. The compound of claim 1, wherein is selected from the groups set forth below:

wherein, one of the indicated valences is the point of attachment to A and the other is the point of attachment to the Z; R3f is selected from a group consisting of halogen, -CN, -NO2, - NH2, optionally substituted C1-C6 alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted C3-C6 cycloalkyl, optionally substituted C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; R3d is selected from a group consisting of hydrogen, halogen, -CN, -NO2, -NH2, optionally substituted C1-C6 alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted C3-C6 cycloalkyl, optionally substituted C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; m’ 0, 1 or 2; and n’ is 0, 1, 2 or 3.

4. The compound of claim 1, represented by Formula (Ha), (lib), or a pharmaceutically acceptable salt thereof,

wherein R11 at each occurrence is independently selected from the group consisting of halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C1-C6 alkoxy, optionally substituted -C3-C6 cycloalkyl, optionally substituted -C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; nl is 0, 1, 2, 3, 4, or 5; n2 is 0, 1, or 3; and R1, R3a, R3b, ® , ® , Z and R4 are as defined in claim 1.

5. The compound of claim 1, represented by Formula (Vila), (Vllb), (Vile), (Vlld), or a pharmaceutically acceptable salt thereof,

wherein R11 at each occurrence is independently selected from the group consisting of halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C1-C6 alkoxy, optionally substituted -C3-C6 cycloalkyl, optionally substituted -C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; nl is 0, 1, 2, 3, 4, or 5; n2 is 0, 1, or 3;

R5, R7, ®, and ® are as defined in claim 1.

6. The compound of claim 1, represented by Formula (VIIa-1), (VIIa-2), (VIIa-3), (VIIa-4), (VIIa-5), (VIIa-6), (VIIb-1), (VIIb-2), (VIIb-3), (VIIb-4), (VIIb-5), (VIIb-6), or a pharmaceutically acceptable salt thereof,

wherein R11 at each occurrence is independently selected from the group consisting of halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C1-C6 alkoxy, optionally substituted -C3-C6 cycloalkyl, optionally substituted -C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; nl is 0, 1, 2, 3, 4, or 5; n2 is 0, 1, or 3; and R5, R7, ® , and ® are as defined in claim 1.

7. The compound of claim 1, represented by Formula (VIII), or a pharmaceutically acceptable salt thereof,

wherein ® , ® , Z and R4 are as defined in claim 1.

8. The compound of claim 7, selected from compounds of Formula (VIII) wherein ® , ® , and Z-R4 are delineated for each compound in Table 1, or a pharmaceutically acceptable salt thereof:

Table 1

9. The compound of claim 1, represented by Formula (IX), or a pharmaceutically acceptable salt thereof,

wherein ® . ®. and R7 are as defined in claim 1.

10. The compound of claim 9, selected from compounds of Formula (IX) wherein ® ® , and R7 are delineated for each compound in Table 2, or a pharmaceutically acceptable salt thereof:

Table 2

11. The compound of claim 1 , represented by Formula (X), or a pharmaceutically acceptable salt thereof,

wherein ® , ® , Z and R4 are as defined in claim 1.

12. The compound of claim 11, selected from compounds according to Formula (X), wherein ® . ®. and Z-R4 are delineated for each compound in Table 3, or a pharmaceutically acceptable salt thereof:

Table 3

13. The compound of claim 1, represented by Formula (XI), or a pharmaceutically acceptable salt thereof,

wherein ® . ®. and R7 are as defined in claim 1.

14. The compound of claim 13, selected from compounds according to Formula (XI), wherein ® . ®. and R7 are delineated for each compound in Table 4, or a pharmaceutically acceptable salt thereof:

Table 4

15. The compound of claim 1, selected from the compounds set forth below or a pharmaceutically acceptable salt thereof:

o

o

16. A method for preventing or treating an FXR-mediated disease or condition in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound according to any one of claims 1-15.

17. The method according to claim 16, wherein the FXR-mediated disease or condition is selected from the group consisting of chronic liver disease, gastrointestinal disease, renal disease, cardiovascular disease, fibrotic diseases, and metabolic disease.

18. The method according to claim 17, wherein the fibrotic diseases is selected from primary biliary cirrhosis, primary sclerosing cholangitis, pulmonary fibrosis, renal fibrosis, and liver fibrosis. 19. The method according to claim 17, wherein the chronic liver disease is selected from the group consisting of primary biliary cirrhosis (PBC), cerebrotendinous xanthomatosis (CTX), primary sclerosing cholangitis (PSC), drug induced cholestasis, intrahepatic cholestasis of pregnancy, parenteral nutrition associated cholestasis (PNAC), bacterial overgrowth or sepsis associated cholestasis, autoimmune hepatitis, chronic viral hepatitis, alcoholic liver disease, nonalcoholic fatty liver disease (NAFLD), nonalcoholic

steatohepatitis (NASH), liver transplant associated graft versus host disease, living donor transplant liver regeneration, congenital hepatic fibrosis, choledocholithiasis, granulomatous liver disease, intra- or extrahepatic malignancy, Sjogren's syndrome, Sarcoidosis, Wilson's disease, Gaucher's disease, hemochromatosis, and alpha 1 -antitrypsin deficiency. 20. The method according to claim 17, wherein the renal disease is selected from the group consisting of diabetic nephropathy, focal segmental glomerulosclerosis (FSGS), hypertensive nephrosclerosis, chronic glomerulonephritis, chronic transplant glomerulopathy, chronic interstitial nephritis, and polycystic kidney disease. 21. The method according to claim 17, wherein the cardiovascular disease is selected from the group consisting of atherosclerosis, arteriosclerosis, dyslipidemia,

hypercholesterolemia, and hypertriglyceridemia.

22. The method according to claim 17, wherein the metabolic disease is selected from the group consisting of insulin resistance, Type I and Type II diabetes, and obesity.

23. A pharmaceutical composition comprising a compound of any one of claims 1-15 and a pharmaceutically acceptable carrier. 24. Use of a compound of any one of claims 1-15 in the preparation of a medicament for the prevention or treatment of an FXR-mediated disease or condition.

Description:
ISOXAZOLE ANALOGS AS FXR AGONISTS AND METHODS OF USE THEREOF

RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 62/597,730, filed on December 12, 2017. The entire teachings of the above application are incorporated herein by reference.

TECHNICAL FIELD

The present invention relates generally to compounds and pharmaceutical

compositions useful as FXR modulators. Specifically, the present invention relates to isoxazole derivatives useful as agonists for FXR, and methods for their preparation and use.

BACKGROUND OF THE INVENTION

Famesoid X Receptor (FXR) is an orphan nuclear receptor initially identified from a rat liver cDNA library (BM. Forman, et al, Cell, 1995, 81(5), 687-693) that is most closely related to the insect ecdysone receptor. FXR is a member of the nuclear receptor family of ligand-activated transcription factors that includes receptors for the steroid, retinoid, and thyroid hormones (DJ. Mangelsdorf, et al, Cell , 1995, 83(6), 841-850). The relevant physiological ligands of FXR are bile acids (D. Parks et al., Science, 1999, 284(5418), 1362- 1365). The most potent one is chenodeoxy cholic acid (CDCA), which regulates the expression of several genes that participate in bile acid homeostasis. Famesol and derivatives, together called famesoids, are originally described to activate the rat orthologue at high concentration but they do not activate the human or mouse receptor. FXR is expressed in the liver, throughout the entire gastrointestinal tract including the esophagus, stomach, duodenum, small intestine, colon, ovary, adrenal gland and kidney. Beyond controlling intracellular gene expression, FXR seems to be also involved in paracrine and endocrine signaling by upregulating the expression of the cytokine Fibroblast Growth Factor (J. Holt et al., Genes Dev., 2003, 17(13), 1581-1591; T. Inagaki et al, CellMetab., 2005, 2(4), 217- 225).

Small molecule compounds which act as FXR modulators have been disclosed in the following publications: WO 2000/037077, WO 2002/072598, WO 2003/015771, WO 2003/099821, WO 2004/00752, WO 2004/048349, WO 2005/009387, WO 2005/082925, US

2005/0054634, WO 2007/052843, WO 2007/070796, WO 2007/076260, WO 2007/092751, WO 2007/095174, WO 2007/140174, WO 2007/140183, US 2007/0142340, WO 2008/000643, WO 2008/002573, WO 2008/025539, WO 2008/025540, WO 2008/051942, WO 2008/073825, WO 2008/157270, US 2008/0299118, US 2008/0300235, WO

2009/005998, WO 2009/012125, WO 2009/027264, WO 2009/062874, WO 2009/127321, WO 2009/149795, US 2009/0131409, US 2009/0137554, US 2009/0163474, US

2009/0163552, US 2009/0215748, WO 2010/043513, WO 2011/020615, WO 2011/117163,

WO 2012/087519, WO 2012/087520, WO 2012/087521, WO 2013/007387, WO

2013/037482, WO 2013/166176, WO 2013/192097, WO 2014/184271, US 2014/0186438, US 2014/0187633, WO 2015/017813, WO 2015/069666, WO 2016/073767,

WO 2016/116054, WO 2016/103037, WO 2016/096116, WO 2016/096115,

WO 2016/097933, WO 2016/081918, WO 2016/127924, WO 2016/130809, WO

2016/145295, WO 2016/173524, CN 106632294, CN 106588804, US 2017/0196893, WO 2017/062763, WO 2017/053826, CN 106518708, CN 106518946, CN 106478759, CN 106478447, CN 106478453, WO 2017/027396, WO 2017/049172, WO 2017/049173, WO 2017/049176, WO 2017/049177, WO 2017/118294, WO 2017/128896, WO 2017/129125, WO 2017/133521, WO 2017/147174, WO 2017/156024 Al. Further small molecule FXR modulators have been recently reviewed (R. C. Buijsman, et al., Curr. Med. Chem. 2005, 12(9), 1017-1075; Crawley, M. L. Expert Opin. Ther. Patents 2010, 20(8), 1047-1057; V. Sepe, et al, Expert Opin. Ther. Patents 2015, 25(8), 885-896).

There is a need for the development of FXR modulators for the treatment and prevention of disease. The present invention has identified compounds which modulate FXR as well as methods of using these compounds to treat disease.

SUMMARY OF THE INVENTION

In one aspect, the invention provides compounds represented by Formula I and pharmaceutically acceptable salts thereof:

wherein:

R 1 is hydrogen, halogen, cyano, optionally substituted -C1-C6 alkyl, optionally substituted - C2-C6 alkenyl, optionally substituted -C2-C6 alkynyl, optionally substituted -C3-C6 cycloalkyl or optionally substituted 3- to 6- membered heterocycloalkyl. Preferably, R 1 is isopropyl, tert-butyl, or cyclopropyl.

R 2 is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted -C3- Ci 2 cycloalkyl or optionally substituted 3- to 12- membered heterocycloalkyl;

R 3a , R 3b are each independently selected from groups consisting of hydrogen, halogen, optionally substituted -C1-C6 alkyl, , optionally substituted -C1-C6 alkoxy and optionally substituted -C3-C6 cycloalkyl. Preferably, R 3a and R 3b are both hydrogen. Alternatively, R 3a and R 3b are taken together with the carbon atom to which they are attached to form a cyclic moiety selected from optionally substituted -C3-C6 cycloalkyl, optionally substituted 3- to 6- membered heterocycloalkyl, and optionally substituted -C3-C6 cycloalkenyl.

® is

L 1 , L 2 , L 3 are each independently C or N;

e, f, m, and n are each independently 0, 1, 2, 3, or 4;

A 1 is O, NR 3c , S, S(O) or S(0) 2 ;

A 2 is absent, O, NR 3c , S, S(O) or S(0) 2 ;

A 1 is attached to -CR 3a R 3b -, and A 2 is attached t

each X 1 , X 2 , X 3 and X 4 is independently selected from group consisting of: O, C(O), S, S(O), S(0) 2 , NR 3c , and CR 3d R 3e ; wherein R 3d and R 3e are each independently selected from hydrogen, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, and optionally substituted -O-C1-C6 alkyl; alternatively, R 3d and R 3e are taken together with the carbon atom to which they are attached to form a optionally substituted -C3- C6 cycloalkyl or optionally substituted 3- to 6- membered heterocycloalkyl.

R 3c is hydrogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, formyl, or acetyl; Y 1 is absent when L 1 is N, and hydrogen, hydroxyl, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted - O-Ci-Ce alkyl when L 1 is C; Y 3 is absent when L 3 is N, and hydrogen, hydroxyl, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted -O-Ci-Ce alkyl when L 3 is C; Y 4 is absent when L 4 is N, and hydrogen, hydroxyl, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally substituted -O-C1-C6 alkyl when L 4 is C; Y 2 is hydrogen, hydroxyl, halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C3-C6 cycloalkyl, or optionally

substituted -O-Ci-Ce alkyl; provided that ® is not ni 5 n2 ; n3 i n4 is 0, 1, 2 or 3; R.3q is selected from hydrogen, halogen, optionally substituted -C1-C3 alkyl, optionally substituted -C3-C6 cycloalkyl, optionally substituted -O-C1-C3 alkyl; alternatively two R.3q groups are linked together to form a -C3-C6 cycloalkyl, aryl, heteroaryl or 3- to 6- membered heterocycloalkyl.

® is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted 3- 12 membered heterocycloalkyl. Preferably, the substituents are selected from group consisting of OH, halogen, CN, optionally substituted -O-C1-C6 alkyl, optionally substituted -C1-C6 -alkyl, optionally substituted 3- to 6- membered-heterocycloalkyl and optionally substituted -C3-C6-cycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;

Z is selected from the group consisting of:

1) Absent;

2) Optionally substituted -C1-C6 alkyl;

3) Optionally substituted -C2-C6 alkenyl;

4) Optionally substituted -C2-C6 alkynyl;

5) Optionally substituted -C3-C8 cycloalkyl;

6) Optionally substituted 3- to 8- membered heterocycloalkyl;

7) Optionally substituted -C3-C8 cycloalkenyl;

8) Optionally substituted aryl; and

9) Optionally substituted heteroaryl; and

R 4 is hydroxy, protected hydroxy, -0-(hydroxy prodrug group), tetrazolyl, cyano, -CO2R 5 , -

wherein

Y is absent or optionally substituted -C1-C6 alkyl; R 4a and R 4b are independently selected from the groups consisting of:

1) Hydrogen;

2) Optionally substituted -Ci-Ce alkyl;

3) Optionally substituted -C2-C8 alkenyl;

4) Optionally substituted -C2-C8 alkynyl; and

5) Optionally substituted -C3-C8 cycloalkyl;

R 4c is hydrogen or optionally substituted -C1-C6 alkyl; preferably R 4c is hydrogen or -CH3; R 5 is selected from the groups consisting of:

1) Hydrogen;

3) Optionally substituted -Ci-Ce alkyl;

4) Optionally substituted -C2-C8 alkenyl;

5) Optionally substituted -C2-C8 alkynyl; and

6) Optionally substituted -C3-C8 cycloalkyl;

R 7 is selected from the groups consisting of:

1) Optionally substituted -Ci-Ce alkyl;

2) Optionally substituted -C2-C8 alkenyl;

3) Optionally substituted -C2-C8 alkynyl;

4) Optionally substituted -C3-C8 cycloalkyl;

5) Optionally substituted -C3-C8 cycloalkenyl;

6) Optionally substituted 3- to 8-membered heterocycloalkyl;

7) Optionally substituted 3- to 8- membered heterocycloalkenyl;

8) Optionally substituted aryl;

9) Optionally substituted -Ci-Ce arylalkyl;

10) Optionally substituted heteroaryl;

11) Optionally substituted -Ci-Ce heteroarylalkyl; and

12)NR 9 R 10 ; wherein R 9 and R 10 are each independently selected from hydrogen, optionally substituted -Ci-Ce alkyl, optionally substituted -C2-C8 alkenyl, optionally substituted -C2-C8 alkynyl, optionally substituted -C3-C8 cycloalkyl, optionally substituted aryl, optionally substituted alkylaryl, optionally substituted 3- to 8- membered heterocycloalkyl, optionally substituted heteroaryl, optionally substituted alkylheteroaryl; alternatively, R 9 and R 10 are taken together with the nitrogen atom to which they are attached to form a heterocyclic ring.

In certain embodiments, the hydroxy prodrug group is phosphate or sulfamate. In certain embodiments, the hydroxy prodrug group is an acyl group derived from an amino acid, preferably an a-amino acid.

In another embodiment, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt form, stereoisomer, solvate, hydrate or combination thereof, in combination with a

pharmaceutically acceptable carrier or excipient.

In another embodiment, the present invention provides a method for the prevention or treatment of an FXR mediated disease or condition. The method comprises administering a therapeutically effective amount of a compound of Formula (I). The present invention also provides the use of a compound of Formula (I) for the preparation of a medicament for the prevention or treatment of an FXR mediated disease or condition.

DETAILED DESCRIPTION OF THE INVENTION

In one embodiment, the present invention provides a compound of Formula (I) as described above, or a pharmaceutically acceptable salt thereof.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 1 is optionally substituted isopropyl, cyclopropyl, or /e/V-butyl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 2 is optionally substituted cyclohexyl, cyclopentyl, or cyclopropyl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 2 is cyclohexyl or cyclopentyl, each of which is optionally substituted with up to 3 groups which are independently selected from halogen, optionally substituted -C1-C6 alkyl, optionally substituted -Ci-C6 alkoxy, optionally substituted -C3-C6 cycloalkyl, optionally substituted, -C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 2 is cyclopropyl which is optionally substituted with up to 2 groups which are independently selected from of halogen, optionally substituted -C1-C6 alkyl, optionally substituted -Ci-C6 alkoxy, optionally substituted -C3-C6 cycloalkyl, optionally substituted -C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 2 is optionally substituted phenyl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 2 is optionally substituted heteroaryl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 2 is selected from the groups set forth below:

wherein each of the above groups is optionally further substituted. The preferred substituents are halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted C3-C6 cycloalkyl, optionally substituted C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein at least one of R 3a and R 3b is hydrogen or halogen. In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein both R 3a and R 3b are independently hydrogen or halogen.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein ® is

where R 3c is previously defined; preferably, R 3c is hydrogen, methyl, isopropyl or formyl; Y'.Y 2 . Y 3 , and Y 4 are as previously defined; preferably, Y'.Y 2 . Y 3 and Y 4 are each independently hydrogen, methyl, hydroxyl, or halogen.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein ® is optionally substituted fused aryl, optionally substituted fused heteroaryl or optionally substituted fused 3-12 membered heterocycloalkyl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein ® is optionally substituted phenyl, pyridyl, pyrimidinyl, pyrazolyl, thienyl, thiazolyl, triazolyl, isothiazolyl, pyrrolyl, pyrazolyl, oxazolyl, oxadiazolyl, imidazolyl, furanyl, indolyl, benzothienyl, naphthyl, quinolyl, naphthyridyl, quinoxalinyl, pyridopyrazolyl, pyridooxazolyl, pyridothiazolyl, isoquinolyl, pyridofuranyl, indazolyl, benzisoxazolyl, benzofuranyl, benzotriazolyl, or benzothiazolyl. Preferred substituents include halogen, -CN, -NO2, -NH2, optionally substituted C1-C6 alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted C3-C6 cycloalkyl, optionally substituted C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein ® is selected from the groups set forth below:

wherein one of the indicated valences is the point of attachment to A and the other is the point of attachment to Z; R 3f is selected from a group consisting of halogen, -CN, -NC , - NH2, optionally substituted C1-C6 alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted C3-C6 cycloalkyl, optionally substituted C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; R 3d is selected from a group consisting of hydrogen, halogen, -CN, -NO2, -NH2, optionally substituted C1-C6 alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted C3-C6 cycloalkyl, optionally substituted C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; m’ is 0, 1 or 2; and n’ is 0, 1, 2 or 3; preferably, m’ and n’ are each independently 0 or 1.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is absent. In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is optionally substituted -CFk; preferably, Z is -CFh. -CHF or -CF2. In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is optionally substituted -CH2CH2-. In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is optionally substituted cyclopropyl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is optionally substituted'' '^·, wherein, one of the indicated valences is the point of attachment to B and the other is the point of attachment to R 4 .

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is optionally substituted aryl;

preferably Z is optionally substituted phenyl. In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein Z is optionally substituted heteroaryl; preferably Z is optionally substituted pyridyl.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 4 is -CO2R 5 , and R 5 is previously

defined. Preferably R 5 is hydrogen, methyl, ethyl, t-butyl, In certain embodiments, the present invention relates to compounds of Formula (I),

o and pharmaceutically acceptable salts thereof, wherein R 4 is

In certain embodiments, the present invention relates to compounds of Formula (I),

and pharmaceutically acceptable salts thereof, wherein R 4 is H R7 . and R 7 is previously defined.

In certain embodiments, the present invention relates to compounds of Formula (I), and pharmaceutically acceptable salts thereof, wherein R 1 is optionally substituted

cyclopropyl; R 2 is selected from ; R 3a is

hydrogen; R 3b is hydrogen; 'o'is optionally substituted

is optionally substituted and selected from:

hydrogen, methyl, ethyl, t-butyl,

In another embodiment, the compound of Formula (I) is represented by Formula (Ila), Formula (lib), or a pharmaceutically acceptable salt thereof:

wherein R 1 , R 3a , R 3b , ®, ®, Z and R 4 are as previously defined; R 11 at each occurrence is independently selected from the group consisting of halogen, optionally substituted -C1-C6 alkyl, optionally substituted -C1-C6 alkoxy, optionally substituted -C3-C6 cycloalkyl, optionally substituted -C3-C6 cycloalkenyl, optionally substituted aryl, and optionally substituted heteroaryl; nl is 0, 1, 2, 3, 4, or 5; and n2 is 0, 1, 2 or 3.

In another embodiment, the compound of Formula (I) is represented by Formula (Ila- 1), (IIa-2), (IIa-3), (IIb-1), (IIb-2), or (IIb-3), or a pharmaceutically acceptable salt thereof:

wherein R 3a , R 3b , ® , ® , Z, R 4 , R 11 , nl and n2 are as previously defined.

In another embodiment, the compound of Formula (I) is represented by Formula (Ilia) or (Illb), or a pharmaceutically acceptable salt thereof:

wherein ®, ®, Z, R 1 , R 2 , R 3a , R 3b , R 5 , and R 7 are as previously defined. In another embodiment, the compound of Formula (I) is represented by Formula (IVa) or (IVb), or a pharmaceutically acceptable salt thereof:

wherein ®, ®, Z, R 2 , R 5 , and R 7 are as previously defined.

In another embodiment, the compound of Formula (I) is represented by Formula (Va), (Vb), (Vc), or (Vd), or a pharmaceutically acceptable salt thereof:

wherein ® , ® , Z, R 5 , R 7 , R 11 , nl and n2 are as previously defined.

In another embodiment, the compound of Formula (I) is represented by Formula (Via), or (VIb), or a pharmaceutically acceptable salt thereof:

wherein ®, ®, R 2 , R 5 , and R 7 are as previously defined.

In another embodiment, the compound of Formula (I) is represented by Formula (Vila), (Vllb), (Vile), or (Vlld), or a pharmaceutically acceptable salt thereof:

, R 5 , R 7 , R 11 , nl and n2 are as previously defined.

In another embodiment, the compound of Formula (I) is represented by Formula (VIIa-1), (VIIa-2), (VIIa-3), (VIIa-4), (VIIa-5), or (VIIa-6), or a pharmaceutically acceptable salt thereof:

wherein ® , R 5 , R 11 , Y 1 and nl are as previously defined.

In another embodiment, the compound of Formula (I) is represented by

Formula(VIIb-l), (VIIb-2), (VIIb-3), (VIIb-4), (VIIb-5), or (VIIb-6), or a pharmaceutically acceptable salt thereof:

wherein ® , R 7 , R 11 , Y 1 and nl are as previously defined.

In another embodiment, the compound of Formula (I) is represented by Formula (VIII) or a pharmaceutically acceptable salt thereof:

wherein ®, ®, Z and R 4 are as previously defined.

Representative compounds of the invention include, but are not limited to, the following compounds in Table 1 according to Formula (VIII), and pharmaceutically acceptable salts thereof, wherein, ®, ® , and Z-R 4 are delineated for each example in Table

1

Table 1

In another embodiment, the compound of Formula (I) is represented by Formula (IX), or a pharmaceutically acceptable salt thereof:

wherein ®, ®, and R 7 are as previously defined.

Representative compounds of the invention include, but are not limited to, the following compounds in Table 2 according to Formula (IX), and pharmaceutically acceptable salts thereof, wherein ®, ®, and R 7 are delineated for each example in Table 2. Table 2

In another embodiment, the compound of Formula (I) is represented by Formula (X) or a pharmaceutically acceptable salt thereof:

wherein ®, ®. Z. and R 4 are as previously defined.

Representative compounds of the invention include, but are not limited to, the following compounds in Table 3 according to Formula (X), and pharmaceutically acceptable salts thereof, wherein ®, ®, and Z-R 4 are delineated for each example in Table 3. Table 3

In another embodiment, the compound of Formula (I) is represented by Formula (XI) or a pharmaceutically acceptable salt thereof:

wherein ®. ®. and R 7 are as previously defined.

Representative compounds of the invention include, but are not limited to, the following compounds in Table 4 according to Formula (XI), and pharmaceutically acceptable salts thereof, wherein, ® , ®, and R 7 are delineated for each example in Table 4.

Table 4

It will be appreciated that the description of the present invention herein should be construed in congruity with the laws and principles of chemical bonding. In some instances, it may be necessary to remove a hydrogen atom in order to accommodate a substituent at any given location.

It will be yet appreciated that the compounds of the present invention may contain one or more asymmetric carbon atoms and may exist in racemic, diastereoisomeric, and optically active forms. It will still be appreciated that certain compounds of the present invention may exist in different tautomeric forms. All tautomers are contemplated to be within the scope of the present invention.

In certain embodiments, the present invention provides a method for the prevention or treatment of an FXR mediated disease or condition. The method comprises administering a therapeutically effective amount of a compound of Formula (I). The present invention also provides the use of a compound of Formula (I) for the preparation of a medicament for the prevention or treatment of an FXR mediated disease or condition.

In certain embodiments, the FXR-mediated disease or condition is cardiovascular disease, atherosclerosis, arteriosclerosis, hypercholesterolemia, or hyperlipidemia chronic liver disease, gastrointestinal disease, fibrotic diseases such as primary biliary cirrhosis, primary sclerosing cholangitis, pulmonary fibrosis, renal fibrosis, liver fibrosis, renal disease, metabolic disease, inflammatory demyelinating disease, cancer (i.e., colorectal cancer), or neurological indications such as stroke.

In certain embodiments, the chronic liver disease is primary biliary cirrhosis (PBC), cerebrotendinous xanthomatosis (CTX), primary sclerosing cholangitis (PSC), drug induced cholestasis, intrahepatic cholestasis of pregnancy, parenteral nutrition associated cholestasis (PNAC), bacterial overgrowth or sepsis associated cholestasis, autoimmune hepatitis, chronic viral hepatitis, alcoholic liver disease, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), liver transplant associated graft versus host disease, living donor transplant liver regeneration, congenital hepatic fibrosis, choledocholithiasis, granulomatous liver disease, intra- or extrahepatic malignancy, Sjogren’s syndrome, Sarcoidosis, Wilson’s disease, Gaucher’s disease, hemochromatosis, or alpha 1 -antitrypsin deficiency. In certain embodiments, the gastrointestinal disease is inflammatory bowel disease (IBD) (including Crohn's disease and ulcerative colitis), irritable bowel syndrome (IBS), bacterial overgrowth, malabsorption, post-radiation colitis, or microscopic colitis.

In certain embodiments, the renal disease is diabetic nephropathy, focal segmental glomerulosclerosis (FSGS), hypertensive nephrosclerosis, chronic glomerulonephritis, chronic transplant glomerulopathy, chronic interstitial nephritis, or polycystic kidney disease.

In certain embodiments, the cardiovascular disease is atherosclerosis, arteriosclerosis, dyslipidemia, hypercholesterolemia, or hypertriglyceridemia.

In certain embodiments, the metabolic disease is insulin resistance, Type I and Type II diabetes, or obesity.

In one aspect, the compound is a selective FXR agonist over TGR5 activator.

Yet a further aspect of the present invention is a process of making any of the compounds delineated herein employing any of the synthetic means delineated herein.

It should be understood that the compounds encompassed by the present invention are those that are suitably stable for use as pharmaceutical agent.

DEFINITIONS

Listed below are definitions of various terms used to describe this invention. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.

The term "alkyl”, as used herein, refers to a saturated, monovalent straight- or branched-chain hydrocarbon group. Preferred alkyl radicals include C1-C6 alkyl and C i-Cx alkyl radicals. Examples of C1-C6 alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, «-butyl, tert- butyl, neopentyl, n-hexyl groups; and examples of C i-Cx alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, «-butyl, tert- butyl, neopentyl, n-hexyl, heptyl, and octyl groups.

The term“alkenyl”, as used herein, denote a monovalent group derived from a hydrocarbon moiety by the removal of a single hydrogen atom wherein the hydrocarbon moiety has at least one carbon-carbon double bond. Preferred alkenyl groups include C2-C6 alkenyl and C2-C8 alkenyl groups. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl, heptenyl, octenyl and the like.

The term“alkynyl”, as used herein, denotes a monovalent group derived from a hydrocarbon moiety by the removal of a single hydrogen atom wherein the hydrocarbon moiety has at least one carbon-carbon triple bond. Preferred alkynyl groups include C2-C6 alkynyl and C2-C8 alkynyl groups. Representative alkynyl groups include, but are not limited to, for example, ethynyl, l-propynyl, l-butynyl, heptynyl, octynyl and the like.

The term“cycloalkyl”, as used herein, refers to a monocyclic or polycyclic saturated carbocyclic ring or a bi- or tri-cyclic group fused, bridged or spiro system, and the carbon atoms may be optionally oxo-substituted or optionally substituted with exocyclic olefmic double bond. Preferred cycloalkyl groups include C3-C8 cycloalkyl and C3-C 12 cycloalkyl groups. Examples of C3-C8-cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of C3-C12- cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl,bicyclo[2.2. l]heptyl, bicyclo[2.2.2]octyl,spiro[2.5]octyl, 3- methylenebicyclo[3.2. l]octyl, spiro[4.4]nonanyl, bicycle[3. l.0]hexanyl, spiro[2.3]hexanyl, bicycle[3. l. l]heptanyl, spiro[2.5]octanyl, bicycle[4. l.0]heptanyl, bicycle[3. l.0]hexan-6-yl, spiro[2.3]hexan-5-yl, bicycle[3. l.l]heptan-3-yl, spiro[2.5]octan-4-yl, and

bicycle[4. l.0]heptan-3-yl and the like.

The term“cycloalkenyl”, as used herein, refers to monocyclic or polycyclic carbocyclic ring or a bi- or tri-cyclic group fused, bridged or spiro system having at least one carbon-carbon double bond and the carbon atoms may be optionally oxo-substituted or optionally substituted with exocyclic olefmic double bond. Preferred cycloalkenyl groups include C3-C8 cycloalkenyl and C3-C 12 cycloalkenyl groups. Examples of C3-C8-cycloalkenyl include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like; and examples of C3-Ci2-cycloalkenyl include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, bicyclo[2.2. l]hept-2-enyl, bicyclo[3.l.0]hex-2-enyl, spiro[2.5]oct-4-enyl, spiro[4.4]non-l-enyl, bicyclo[4.2. l]non-3-en-9-yl, and the like.

The terms“heterocyclic” or“heterocycloalkyl” can be used interchangeably and referred to a non-aromatic ring or a bi- or tri-cyclic group fused, bridged or spiro system, where (i) each ring system contains at least one heteroatom independently selected from oxygen, sulfur and nitrogen, (ii) each ring system can be saturated or unsaturated (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quatemized, (v) any of the above rings may be fused to an aromatic ring, and (vi) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted or optionally substituted with exocyclic olefmic double bond.. Representative

heterocycloalkyl groups include, but are not limited to, [l,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, tetrahydrofuryl, 2-azabicyclo[2.2. l]-heptyl, 8-azabicyclo[3.2.l]octyl, 5- azaspiro[2.5]octyl, l-oxa-7-azaspiro[4.4]nonanyl, 7-oxooxepan-4-yl, and tetrahydrofuryl. Such heterocyclic groups may be further substituted. Heteroaryl or heterocyclic groups can be C-attached or N-attached (where possible).

The term "aryl," as used herein, refers to a mono-, bi-, or polycyclic carbocyclic ring system comprising at least one aromatic ring, including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, and indenyl. A polycyclic aryl is a polycyclic ring system that comprises at least one aromatic ring. Polycyclic aryls can comprise fused rings, covalently attached rings or a combination thereof.

The term“arylalkyl,” as used herein, refers to a functional group wherein an alkylene chain is attached to an aryl group, e.g., -CH2CH2-phenyl. The term“substituted arylalkyl” means an arylalkyl functional group in which the aryl group is substituted. Examples include, but are not limited to, benzyl, phenethyl and the like.

The term "heteroaryl," as used herein, refers to a mono-, bi-, or tri-cyclic aromatic radical or ring having from five to ten ring atoms of which at least one ring atom is selected from S, O and N; wherein any N or S contained within the ring may be optionally oxidized. Preferred heteroaryl groups are monocyclic or bicyclic. Heteroaryl groups include, but are not limited to, pyridinyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, triazolyl, isothiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, benzothienyl, quinoxalinyl, indolyl, indazolyl, benzisoxazolyl, benzofuranyl, benzotriazolyl,

benzothiazolyl, and the like.

The term“heteroarylalkyl,” as used herein, refers to an alkylene chain is attached to a heteroaryl group. The tern“substituted heteroarylalkyl” means a heteroarylalkyl functional group in which the heteroaryl group is substituted. Examples include, but are not limited to, pyridinylmethyl, pyrimidinylethyl and the like.

As used herein, the term“alkoxy” employed alone or in combination with other terms means, unless otherwise stated, an alkyl group having the designated number of carbon atoms connected to the rest of the molecule via an oxygen atom, such as, for example, methoxy, ethoxy, l-propoxy, 2-propoxy (isopropoxy) and the higher homologs and isomers. Preferred alkoxy are (C1-C3) alkoxy.

The term“substituted” refers to substitution by independent replacement of one, two, or three or more of the hydrogen atoms with substituents including, but not limited to, -F, -Cl, -Br, -I, -OH, Ci-Ci2-alkyl; C2-C 12-alkenyl, C2-C 12-alky nyl, protected hydroxy, -NO2, -N3, - CN, -NH2, protected amino, oxo, thioxo, -NH-Ci-Ci2-alkyl, -NH-C2-C8-alkenyl, -NH-C2-C8- alkynyl, -NH-C3-C 12-cycloalkyl, -NH-aryl, -NH-heteroaryl, -NH-heterocycloalkyl, - dialkylamino, -diarylamino, -diheteroarylamino, -0-Ci-Ci2-alkyl, -0-C2-C8-alkenyl, -O-C2- Cs-alkynyl, -O-C3-C 12-cycloalkyl, -O-aryl, -O-heteroaryl, -O-heterocycloalkyl, -C(0)-Ci-Ci2- alkyl, -C(0)-C 2 -C8-alkenyl, -C(0)-C 2 -C8-alkynyl, -C(0)-C3-Ci 2 -cycloalkyl, -C(0)-aryl, - C(0)-heteroaryl, -C(0)-heterocycloalkyl, -CONH2, -CONH-Ci-Ci2-alkyl, -CONH-C2-C8- alkenyl, -CONH-C2-C8-alkynyl, -CONH-C3-Ci2-cycloalkyl, -CONH-aryl, -CONH- heteroaryl, -CONH-heterocycloalkyl, -0C02-Ci-Ci2-alkyl, -0C02-C2-C8-alkenyl, -OCO2-C2- C8-alkynyl, -OC02-C3-Ci2-cycloalkyl, -0C02-aryl, -OC02-heteroaryl, -OCO2- heterocycloalkyl, -CO2-C1-C12 alkyl, -CO2-C2-C8 alkenyl, -CO2-C2-C8 alkynyl, CO2-C3-C12- cycloalkyl, -CO2- aryl, C02-heteroaryl, C02-heterocyloalkyl, -OCONH2, -OCONH-Ci-C 12- alkyl, -OCONH-C2-C8-alkenyl, -OCONH-C2-C8-alkynyl, -OCONH-C3-C 12-cycloalkyl, - OCONH-aryl, -OCONH-heteroaryl, -OCONH- heterocyclo-alkyl, -NHC(0)H, -NHC(0)-Ci- Ci2-alkyl, -NHC(0)-C 2 -C8-alkenyl, -NHC(0)-C 2 -C8-alkynyl, -NHC(0)-C3-Ci 2 -cycloalkyl, - NHC(0)-aryl, -NHC(0)-heteroaryl, -NHC(0)-heterocyclo-alkyl, -NHC02-Ci-Ci2-alkyl, - NHC02-C2-C8-alkenyl, -NHCO2- C2-C8-alkynyl, -NHCO2-C3-C 12-cycloalkyl, -NHC02-aryl, - NHC02-heteroaryl, -NHCO2- heterocycloalkyl, -NHC(0)NH2, -NHC(0)NH-Ci-Ci2-alkyl, - NHC(0)NH-C 2 -C8-alkenyl, -NHC(0)NH-C 2 -C8-alkynyl, -NHC(0)NH-C 3 -C 12-cycloalkyl, - NHC(0)NH-aryl, -NHC(0)NH-heteroaryl, -NHC(0)NH-heterocycloalkyl, NHC(S)NH 2 , - NHC(S)NH-Ci-Ci2-alkyl, -NHC(S)NH-C 2 -C8-alkenyl, -NHC(S)NH-C 2 -C8-alkynyl, - NHC(S)NH-C3-Ci2-cycloalkyl, -NHC(S)NH-aryl, -NHC(S)NH-heteroaryl, -NHC(S)NH- heterocycloalkyl, -NHC(NH)NH 2 , -NHC(NH)NH-Ci-C 12-alkyl, -NHC(NH)NH-C 2 -C8- alkenyl, -NHC(NH)NH-C 2 -C8-alkynyl, -NHC(NH)NH-C 3 -C 12-cycloalkyl, -NHC(NH)NH- aryl, -NHC(NH)NH-heteroaryl, -NHC(NH)NH-heterocycloalkyl, -NHC(NH)-Ci-Ci2-alkyl, - NHC(NH)-C 2 -C8-alkenyl, -NHC(NH)-C 2 -C8-alkynyl, -NHC(NH)-C3-Ci2-cycloalkyl, - NHC(NH)-aryl, -NHC(NH)-heteroaryl, -NHC(NH)-heterocycloalkyl, -C(NH)NH-Ci-C 12- alkyl, -C(NH)NH-C2-C8-alkenyl, -C(NH)NH-C2-C8-alkynyl, -C(NH)NH-C3-C 12-cycloalkyl, - C(NH)NH-aryl, -C(NH)NH-heteroaryl, -C(NH)NH-heterocycloalkyl, -S(0)-Ci-Ci2-alkyl, - S(0)-C 2 -C 8 -alkenyl, - S(0)-C 2 -C 8 -alkynyl, -S(0)-C 3 -Ci 2 -cycloalkyl, -S(0)-aiyl, -S(O)- heteroaryl, -S(0)-heterocycloalkyl, -S0 2 NH 2 , -S0 2 NH-Ci-Ci 2 -alkyl, -S0 2 NH-C 2 -C 8 -alkenyl, -SC NH- C 2 -C 8 -alkynyl, -S0 2 NH-C3-Ci 2 -cycloalkyl, -S0 2 NH-aryl, -S0 2 NH-heteroaryl, - SC NH- heterocycloalkyl, -NHS0 2 -Ci-Ci 2 -alkyl, -NHS0 2 -C 2 -C 8 -alkenyl, - NHS0 2 -C 2 -C 8 - alkynyl, -NHS0 2 -C3-Ci 2 -cycloalkyl, -NHS0 2 -aryl, -NHSC -heteroaryl, -NHSC - heterocycloalkyl, -CH 2 NH 2 , -CH 2 S0 2 CH3, -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, - heterocycloalkyl, -C3-Ci 2 -cycloalkyl, polyalkoxyalkyl, polyalkoxy, -methoxymethoxy, - methoxy ethoxy, -SH, -S-Ci-Ci 2 -alkyl, -S-C 2 -C 8 -alkenyl, -S-C 2 -C 8 -alkynyl, -S-C3-Ci 2 - cycloalkyl, -S-aryl, -S-heteroaryl, -S-heterocycloalkyl, or methylthio-methyl. In certain embodiments, the substituents are independently selected from halo, preferably Cl and F; Ci-C4-alkyl, preferably methyl and ethyl; halo-Ci-C4-alkyl, such as fluoromethyl, difluoromethyl, and trifluoromethyl; C2-C4-alkenyl; halo-C2-C4-alkenyl; C3-C6-cycloalkyl, such as cyclopropyl; C1-C4- alkoxy, such as methoxy and ethoxy; halo-Ci-C4-alkoxy, such as fluoromethoxy, difluoromethoxy, and trifluoromethoxy, -CN; -OH; N¾; Ci-C4-alkylamino; di(Ci-C4-alkyl)amino; and N0 2 . It is understood that the aryls, heteroaryls, alkyls, cycloalkyls and the like can be further substituted. In some cases, each substituent in a substituted moiety is additionally optionally substituted with one or more groups, each group being independently selected from C1-C6- alkyl, CF3, -F, -Cl, -Br, -I, -OH, -N0 2 , -CN, and -NH 2 .

The term“optionally substituted”, as used herein, means that the referenced group may be substituted or unsubstituted. In one embodiment, the referenced group is optionally substituted with zero substituents, i.e., the referenced group is unsubstituted. In another embodiment, the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from groups described herein.

In accordance with the invention, any of the aryls, substituted aryls, heteroaryls and substituted heteroaryls described herein, can be any aromatic group. Aromatic groups can be substituted or unsubstituted.

It is understood that any alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl moiety described herein can also be an aliphatic group, an alicyclic group or a heterocyclic group.

An“aliphatic group” is non-aromatic moiety that may contain any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contain one or more units of unsaturation, e.g., double and/or triple bonds. An aliphatic group may be straight chained, branched or cyclic and preferably contains between about 1 and about 24 carbon atoms, more typically between about 1 and about 12 carbon atoms. In addition to aliphatic hydrocarbon groups, aliphatic groups include, for example, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Such aliphatic groups may be further substituted. It is understood that aliphatic groups may be used in place of the alkyl, alkenyl, alkynyl, alkylene, alkenylene, and alkynylene groups described herein.

The term“alicyclic,” as used herein, denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[2.2. l]heptyl, and bicyclo[2.2.2]octyl. Such alicyclic groups may be further substituted.

It will be apparent that in various embodiments of the invention, the substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, arylalkyl, heteroarylalkyl, and heterocycloalkyl are intended to be monovalent or divalent. Thus, alkylene, alkenylene, and alkynylene, cycloaklylene, cycloalkenylene, cycloalkynylene, arylalkylene, heteroarylalkylene and heterocycloalkylene groups are to be included in the above definitions, and are applicable to provide the Formulas herein with proper valency.

The terms "halo" and "halogen," as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.

The term“hydrogen” includes hydrogen and deuterium. In addition, the recitation of an atom includes other isotopes of that atom so long as the resulting compound is pharmaceutically acceptable.

In certain embodiments, the compounds of each formula herein are defined to include isotopically labelled compounds. An“isotopically labelled compound” is a compound in which at least one atomic position is enriched in a specific isotope of the designated element to a level which is significantly greater than the natural abundance of that isotope. For example, one or more hydrogen atom positions in a compound can be enriched with deuterium to a level which is significantly greater than the natural abundance of deuterium, for example, enrichment to a level of at least 1%, preferably at least 20% or at least 50%. Such a deuterated compound may, for example, be metabolized more slowly than its non- deuterated analog, and therefore exhibit a longer half-life when administered to a subject. Such compounds can synthesize using methods known in the art, for example by employing deuterated starting materials. Unless stated to the contrary, isotopically labelled compounds are pharmaceutically acceptable.

The term“hydroxy activating group,” as used herein, refers to a labile chemical moiety which is known in the art to activate a hydroxyl group so that it will depart during synthetic procedures such as in a substitution or an elimination reaction. Examples of hydroxyl activating group include, but not limited to, mesylate, tosylate, triflate. p- nitrobenzoate, phosphonate and the like.

The term“activated hydroxyl,” as used herein, refers to a hydroxy group activated with a hydroxyl activating group, as defined above, including mesylate, tosylate, triflate, p- nitrobenzoate, phosphonate groups, for example.

The term“hydroxy protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect a hydroxyl group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the hydroxy protecting group as described herein may be selectively removed. Hydroxy protecting groups as known in the art are described generally in T.H. Greene and P.G. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of hydroxyl protecting groups include benzyloxy carbonyl, 4-methoxybenzyloxy carbonyl, tert-butoxy- carbonyl, isopropoxy carbonyl, diphenylmethoxy carbonyl, 2, 2, 2-trichloroethoxy carbonyl, allyloxy carbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t-butyl, 2,2,2-trichloroethyl, 2-trimethylsilyl ethyl, allyl, benzyl, triphenyl- methyl (trityl), methoxymethyl, methylthiomethyl, benzyloxymethyl, 2-(trimethylsilyl)- ethoxymethyl, methanesulfonyl, trimethylsilyl, triisopropylsilyl, and the like.

The term "protected hydroxy," as used herein, refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including benzoyl, acetyl, trimethylsilyl, triethylsilyl, methoxymethyl groups, for example.

The term“hydroxy prodrug group,” as used herein, refers to a promoiety group which is known in the art to change the physicochemical, and hence the biological properties of a parent drug in a transient manner by covering or masking the hydroxy group. After said synthetic procedure(s), the hydroxy prodrug group as described herein must be capable of reverting back to hydroxy group in vivo. Hydroxy prodrug groups as known in the art are described generally in Kenneth B. Sloan, Prodrugs Topical and Ocular Drug Delivery.

(Drugs and the Pharmaceutical Sciences; Volume 53), Marcel Dekker, Inc., New York (1992) and in“Prodrugs of Alcohols and Phenols” by S. S. Dhareshwar and V. J. Stella, in Prodrugs Challenges and Rewards Part-2, (Biotechnology: Pharmaceutical Aspects), edited by V. J. Stella, et al, Springer and AAPSPress, 2007, pp 31-99.

The term“amino protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect an amino group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the amino protecting group as described herein may be selectively removed. Amino protecting groups as known in the art are described generally in T.H. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis. 3rd edition, John Wiley & Sons, New York (1999). Examples of amino protecting groups include, but are not limited to, methoxy carbonyl, t-butoxy carbonyl, 9-fluorenyl- methoxy carbonyl, benzyloxy carbonyl, and the like.

The term“protected amino,” as used herein, refers to an amino group protected with an amino protecting group as defined above.

The term "amino acid" refers to naturally occurring and synthetic a, b, g, or d amino acids, and includes but is not limited to, amino acids found in proteins or intermediates in metabolism of amino acids or proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, citrulline, arginine and histidine. In certain embodiments, the amino acid is in the L-configuration. In certain embodiments, the amino acid is in the D-configuration. In certain embodiments, the amino acid is provided as a substituent of a compound described herein, wherein the amino acid is a residue selected from the group consisting of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, b-alanyl, b-valinyl, b-leucinyl, b-isoleuccinyl, b-prolinyl, b-phenylalaninyl, b-tryptophanyl, b-methioninyl, b-glycinyl, b- serinyl, b-threoninyl, b-cysteinyl, b-tyrosinyl, b-asparaginyl, b-glutaminyl, b-aspartoyl, b- glutaroyl, b-lysinyl, b-argininyl and b-histidinyl.

The term "amino acid derivative" refers to a group derivable from a naturally or non-naturally occurring amino acid, as described and exemplified herein. Amino acid derivatives are apparent to those of skill in the art and include, but are not limited to, ester, amino alcohol, amino aldehyde, amino lactone, and N-methyl derivatives of naturally and non-naturally occurring amino acids. In an embodiment, an amino acid derivative is provided as a substituent of a compound described herein, wherein the substituent is - NR u -G(Sc)-C(0)-Q 1 , wherein Q 1 is -SR V , -NR V R V or alkoxyl, R v is hydrogen or alkyl, Sc is a side-chain of a naturally occurring or non-naturally occurring amino acid, G is C1-C2 alkyl, and R u is hydrogen; or R u and S c are taken together with the atoms to which they are attached to form a five-membered heterocyclic ring. In an embodiment, an amino acid derivative is provided as a substituent of a compound described herein, wherein the substituent is -0-C(0)-G(S c )-NH-Q 2 , wherein Q 2 is hydrogen or alkoxyl, S c is a side- chain of a naturally occurring or non-naturally occurring amino acid and G is C1-C2 alkyl. In certain embodiments, Q 2 and Sc are taken together with the atoms to which they are attached to form a five-membered heterocyclic ring. In certain embodiments, G is an optionally substituted methylene and S c is selected from the group consisting of hydrogen, alkyl, arylalkyl, heterocycloalkyl, carboxylalkyl, heteroarylalkyl, aminoalkyl, hydroxylalkyl, aminoiminoaminoalkyl, aminocarbonylalkyl, sulfanylalkyl,

carbamoylalkyl, alkylsulfanylalkyl and hydroxylarylalkyl. In an embodiment, an amino acid derivative is provided as a substituent of a compound described herein, wherein the amino acid derivative is in the D-configuration. In an embodiment, an amino acid derivative is provided as a substituent of a compound described herein, wherein the amino acid derivative is in the L-configuration.

The term "leaving group" means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction. By way of example, representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; and acyloxy groups, such as acetoxy, trifluoroacetoxy and the like.

When the compounds described herein contain one or more asymmetric centers they give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- , or as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optical isomers may be prepared from their respective optically active precursors by the procedures described above, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques, which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et al, Enantiomers, Racemates, and Resolutions (John Wiley & Sons, 1981). When the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon- carbon double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion. The term“subject” as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be referred to herein as a patient.

As used herein, the term "pharmaceutically acceptable salt" refers to those salts of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are

commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art.

Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reaction of the free base function with a suitable organic acid. Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts e.g., salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate. /Moluenesulfonate. undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate.

As used herein, the term "pharmaceutically acceptable ester" refers to esters of the compounds formed by the process of the present invention which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.

The term“pharmaceutically acceptable prodrugs” as used herein refers to those prodrugs of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like,

commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the present invention. "Prodrug", as used herein means a compound, which is convertible in vivo by metabolic means (e.g. by hydrolysis) to afford any compound delineated by the Formulae of the instant invention. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, Vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). "Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 5: 1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer,“Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzymology,” John Wiley and Sons, Ltd. (2002).

The term "treating", as used herein, means relieving, lessening, reducing, eliminating, modulating, or ameliorating, i.e. causing regression of the disease state or condition. Treating can also include inhibiting, i.e. arresting the development, of an existing disease state or condition, and relieving or ameliorating, i.e. causing regression of an existing disease state or condition, for example when the disease state or condition may already be present.

The term "preventing", as used herein means, to completely or almost completely stop a disease state or condition, from occurring in a patient or subject, especially when the patient or subject is predisposed to such or at risk of contracting a disease state or condition.

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

"Solvates" means solvent addition forms that contain either stoichiometric or non- stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate, when the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one of the substances in which the water retains its molecular state as H2O, such combination being able to form one or more hydrate.

As used herein, the term "analog" refers to a chemical compound that is structurally similar to another but differs slightly in composition (as in the replacement of one atom by an atom of a different element or in the presence of a particular functional group, or the replacement of one functional group by another functional group). Thus, an analog is a compound that is similar to or comparable in function and appearance to the reference compound.

The term "aprotic solvent," as used herein, refers to a solvent that is relatively inert to proton activity, i.e., not acting as a proton-donor. Examples include, but are not limited to, hydrocarbons, such as hexane and toluene, for example, halogenated hydrocarbons, such as, for example, methylene chloride, ethylene chloride, chloroform, and the like, heterocyclic compounds, such as, for example, tetrahydrofuran and /V-methylpyrrolidinone. and ethers such as diethyl ether, bis-methoxymethyl ether. Such solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of aprotic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of

Purification. 4th ed., edited by John A. Riddick et al , Vol. II, in the Techniques of Chemistry Series. John Wiley & Sons, NY, 1986.

The terms "protogenic organic solvent” or“protic solvent” as used herein, refer to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like. Such solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of protogenic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification. 4th ed., edited by John A. Riddick et al, Vol. II, in the Techniques of Chemistry Series. John Wiley & Sons, NY, 1986.

Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term“stable”, as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).

The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and variation of the reaction conditions can produce the desired isoxazole products of the present invention. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein include, for example, those described in R. Larock, Comprehensive Organic Transformations. VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis. 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis. John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis. John Wiley and Sons (1995).

The compounds of this invention may be modified by appending various

functionalities via synthetic means delineated herein to enhance selective biological properties. Such modifications include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.

PHARMACEUTICAL COMPOSITIONS

The pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention Formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term "pharmaceutically acceptable carrier" means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or Formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are sugars such as lactose, glucose and sucrose; starches such as com starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; com oil and soybean oil; glycols; such a propylene glycol; esters such as ethyl oleate and ethyl laurate; agar;

buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid;

pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the Formulator. The pharmaceutical compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracistemally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, or as an oral or nasal spray.

The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the Formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the Formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrastemal, intrathecal, intralesional and intracranial injection or infusion techniques.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol,

tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or weting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.

The injectable Formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable Formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa buter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.

Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical Formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.

Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic Formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.

The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to the compounds of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.

Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art. All publications, patents, published patent applications, and other references mentioned herein are hereby incorporated by reference in their entirety.

ABBREVIATIONS

Abbreviations which have been used in the descriptions of the schemes and the examples that follow are:

ACN for acetonitrile;

AcOH for acetic acid;

BINAP for 2,2'-bis(diphenylphosphino)-l,r-binaphthyl;

BrettPhos for 2-(dicyclohexylphosphino)3,6-dimethoxy-2',4',6'-triisopropyl -l,r- biphenyl;

BOM-C1 for Benzyl chloromethyl ether

BOP-C1 for bis(2-oxo-3-oxazolidinyl)phosphinic chloride;

CDI for carbonyldiimidazole;

DavePhos for 2-dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl;

DBU for l,8-diazabicycloundec-7-ene;

DCC for A. V -di cy cl ohexy 1 carbodi i mi de DCM for dichloromethane;

DIAD for Diisopropyl azodicarboxylate;

DMA for Dimethylacetamide

DMAP for AiV-dimethylaminopyridine:

DMF for A A-di methyl formamide;

DMP for Dess-Martin periodinane;

DMSO for Dimethyl sulfoxide;

DPPA for diphenylphosphoryl azide;

DPPF for l,l'-Ferrocenediyl-bis(diphenylphosphine);

EDC or EDCI for l-(3-diethylaminopropyl)-3-ethylcarbodiimide hydrochloride; EbN for triethylamine;

EtOAc for ethyl acetate;

HATU for l-[bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyri dinium 3- oxid hexafluorophosphate;

HC1 for hydrochloric acid;

LAH for lithium aluminium hydride;

LHMDS for Lithium bis(trimethylsilyl)amide;

Mor-Dalphos for Di(l-adamantyl)-2-morpholinophenylphosphine;

MTBE for Methyl tert-butyl ether;

NCS for N-Chlorosuccinimide;

NaHMDS for Sodium bis(trimethylsilyl)amide;

PyAOP for 7-azabenzotriazol-l-yloxy)tripyrrolidinophosphonium

hexafluorophosphate;

PyBOP for benzotriazol-l-yl-oxytripyrrolidinophosphonium hexafluorophosphate; TFA for trifluoroacetic acid;

TFFH for tetramethylfluoroformamidinium hexafluorophosphate;

THF for tetrahydrofuran;

Xantphos for 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene;

XPhos for dicyclohexyl(2',4',6'-triisopropyl-[l,r-biphenyl]-2-yl)phosp hane or 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl.

SYNTHETIC METHODS

The compounds and processes of the present invention will be better understood in connection with the following synthetic schemes that illustrate the methods by which the compounds of the invention may be prepared, which are intended as an illustration only and not to limit the scope of the invention. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims.

Scheme 1

wherein, R 1 , R 2 , R 3a , R 3b , ® , ® , Z and R 4 are as previously defined. Lg is a leaving group such as halides, -OMs, -OTf, -OTs, -OAr. Pg is hydrogen or a protecting group for hydroxyl or amine whenever applicable such as, but not limited to, BOM, Boc, Cbz and benzyl. The protecting groups are common practices in organic synthesis (see T.W. Greene and P.G.M Wuts,“protective Groups in Organic Chemistry”, 4 th Ed., Wiley-Interscience, 2006).

As shown in Scheme 1, the compounds of formula (I-c) can be obtained through the coupling between the compounds of formula (I-a) and compounds of formula (I-b) employing suitable base such as but not limited to sodium /er/-butoxide, potassium tot- butoxide, or cesium carbonate in the presence or absence of phase transfer reagent such as but not limited to l8-Crown-6, l5-Crown-5 or tetrabutylammonium iodide. The reaction temperature is from -20 °C to 140 °C. The protecting group in compounds of formula (I-c) can be removed whenever applicable and coupled with the compounds of fomula (I-d) to afford the compounds of formula (I). This coupling can be achieved employing suitable base such as but not limited to sodium tert-butoxide, potassium tert-butoxide, or cesium carbonate in the presence or absence of phase transfer reagent such as but not limited to 18-Crown-6, l5-Crown-5 or tetrabutylammonium iodide. Alternatively, the compounds of formula (I) could also be prepared from the deprotected form of compounds of formula (I-c) and the compounds of fomula (I-d) via Buchwald-Hartwig amination. This process employing suitable palladium catalysts such as but not limited to Pd(OAc) 2 , Pd 2 (dba)3, PdCl 2 (P(o- Tolyl)3) 2 , PdCh(DPPF) and Pd(PPh3)4 in presence or absence of a suitable ligand such as but not limited to XPhos, Xantphos, BINAP, BrettPhos, DavePhos, DPPF, P/B , P(o-tolyl)3 and Mor-Dalphos. This amination process may use a suitable base such as but not limited to K3PO4, Cs 2 C03, NaOtBu, LiHMDS and NaHMDS. This amination process is carried out in an suitable solvent such as, but not limited to, toluene, dioxane or THF and the temperature can vary from -20 °C to 120 °C. More detail about Buchwald-Hartwig amination could be found in literature. (Buchwald, S.L. et al, Topics in Curr. Chem., 2002, 219, 131; Lundgren, R. J. et al. , Aldrichimica Acta, 2012, 45, 59; Senra, J. D. et al, Current Organic Synthesis, 2011, 81, 53).

Scheme 2

wherein, R 1 , R 2 , R 3a , R 3b , Z, R 4a , R 7 , ® and ® are as previously defined.

As shown in Scheme 2, the hydrolysis of compounds of Formula (Il-a) to the acids of Formula (Il-b) can be achieved in the presence of suitable bases such as but not limited to sodium hydroxide, lithium hydroxide or potassium hydroxide. The novel isoxazole acylsulfonamide analogs of the compounds of Formula (Il-d) can be prepared from the coupling between compounds of Formula (Il-b) and sulfonamide (II-c) using suitable coupling reagents in presence of suitable bases. The coupling reagent can be selected from, but not limited to, DCC, EDCI, CDI, diisopropyl carbodiimide, BOP-C1, PyBOP, PyAOP, TFFH and HATU. Suitable bases include, but are not limited to, triethylamine,

diisopropylethylamine, DBU, X-methyl morpholine and DMAP. The coupling reaction is carried out in an aprotic solvent such as, but not limited to, DCM, DMF or THF. The reaction temperature can vary from -20 °C to 120 °C.

Scheme 3

wherein, R 1 , R 2 , R 3a , R 3b , Z, R 7 , ® and ® are as previously defined. As shown in Scheme 3, novel isoxazole sulfonyl urea analogs of the compound of formula (Ill-b) are prepared from the compounds of formula (Il-b), wherein R 1 , R 2 , R 3a , R 3b ,

R 7 ,Z, ® and® are as previously defined. Thus, the compounds of formula (Il-b) can be converted to the acyl azide compounds of formula (III-a) using suitable reagents such as, but not limited to, DPPA. The reaction solvents can be, but not limited to, THF, DCM and toluene. The reaction temperature is from -20°C to 80°C. Alternatively, the acids of formula (Il-b) could be transformed to the acyl azides of formula (III-a) via activated acid derivatives such as acyl chlorides or anhydrides in presence of azide source. The reagents for activation of acid includes, but not limited to, tetramethylfluoroformadinium hexafluorophosphate, phenyl dichlorophosphate, SOCh-DMF. triphosgene, cyanuric chloride, NCS-Ph3P and CbCCN-Ph3P The azide source includes, but not limited to, sodium azide,

tetrabutylammonium azide, trimethylsilyl azide and N,N,N’,N’-tetramethylguanidinium azide. Curtius rearrangement of the compounds of formula (III-a) at elevated temperature preferably from 50 °C to 120 °C can lead to the isocyanate intermediates, which then can react with sulfonamides comound of formula (II-c) to afford the compounds of formula (III- b).

Scheme 4

wherein, R 1 , R 2 , R 3a , R 3b , Z, R 7 , ® and ® are as previously defined.

As shown in Scheme 4, the compounds of formula (IV-d) can be prepared from the compounds of formula (Il-b), wherein R 1 , R 2 , R 3a , R 3b , R 7 , Z, ® and® are as previously defined and Lg is a leaving group such as halides, -OMs, -OTf, -OTs, -OAr. Thus, the compounds of formula (Il-b) can be converted to alcohols of formula (IV-a) using suitable reducing reagents such as, but not limited to, LAH, LiBFb, BFb. Alternatively, the alcohols of formula (IV-a) can also be synthesized via the reduction of the derivatives of acid of formula (II-V). Such derivatives include, but not limited to, acyl chloride, mixed anhydride or ester derivatives of acids (Il-b). The compounds of formula (IV-a) could be transformed to the carbamates of formula (IV-d) via coupling with sulfonamides of formula (II-C) employing CDI or phosgene as coupling reagent with or without addition of suitable bases such as, but not limited to, , triethylamine, diisopropylethylamine, DBU, N- methylmorpholine and DMAP. Alternatively, this transformation could be achieved via direct coupling of alcohols of formula (IV-a) with isocyanates of formula (IV-b) in the presence or absence of suitable bases such as, but not limited to, triethylamine, diisopropylethylamine, DBU, /V-methyl morpholine and DMAP. Moreover, the isocyanates of formula (IV-b) could be generated in situ from compounds of formula (IV-c).

Scheme 5

wherein, R 1 , R 2 , R 3a , R 3b , ® , ® , Z and R 4 are as previously defined. Lg is a leaving group such as halides, -OMs, -OTf, -OTs, -OAr. Pg is hydrogen or a protecting group for hydroxyl or amine whenever applicable such as, but not limited to, BOM, Boc, Cbz and benzyl. The protecting groups are common practices in organic synthesis (see T.W. Greene and P.G.M Wuts,“protective Groups in Organic Chemistry”, 4 th Ed., Wiley -Interscience, 2006).

As shown in Scheme 5, the compounds of formula (V-b) can be synthesized through the coupling between the compounds of formula (V-a) and compounds of formula (I-d) employing suitable base such as but not limited to sodium / -butoxide. potassium tert- butoxide, or cesium carbonate in the presence or absence of phase transfer reagent such as but not limited to l8-Crown-6, l5-Crown-5 or tetrabutylammonium iodide. The reaction temperature is from -20 °C to 140 °C. The protecting group in compounds of formula (V-b) can be removed whenever applicable and coupled with the compounds of fomula (I-a) to afford the compounds of formula (I). This coupling can be achieved employing suitable base such as but not limited to sodium tert-butoxide, potassium tert-butoxide, sodium hydride, LHMDS, NaHMDS or cesium carbonate in the presence or absence of phase transfer reagent such as but not limited to l8-Crown-6, l5-Crown-5 or tetrabutylammonium iodide.

In the reactions described, reactive functional groups such as hydroxyl, amino, imino, thio or carboxy groups, may be protected to avoid unwanted participation in the reactions. These protecting groups may be removed at suitable steps via solovolysis, reduction, photolysis. The protection and deprotection are common practices in organic synthesis (see T.W. Greene and P.G.M Wuts,“protective Groups in Organic Chemistry”, 4 th Ed., Wiley- Interscience, 2006). Preparations and Examples

The following preparations and examples are intended to further illustrate the invention only and are not intended to limit the scope of the invention in any way.

Example 1

Step la

To (3R,3aR,6S,6aR)-hexahydrofuro[3,2-b]furan-3,6-diol (isosorbide, la-2) (2 g, 13.69 mmol) in THF (30 ml) was added l8-crown-6 (3.62 g, 13.69 mmol) and potassium tert-butoxide (27.4 ml, 27.4 mmol). The resulting mixture was stirred at RT for lh, and 4-(chloromethyl)-

5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazole (la-2) (4.14 g, 13.69 mmol) was added in one portion. The mixture was stirred at RT for l6h and diluted with EtOAc, washed with NaHCCb soution, water, brine. The organic layer was dried (Na 2 S04), filtered and concentrated. The residue was purified by CombiFlash eluting with 0 to 90% Acetone/hexane to give (3S,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-ol (la-3) (802 mg). LC/MS observed [M+Na], 434.04; Ή NMR (400 MHz, Chloroform^/) d 7.38 - 7.23 (m, 3H), 4.57 - 4.42 (m, 2H), 4.34 - 4.22 (m, 2H), 4.22 - 4.15 (m, 1H), 3.86 (td, J= 6.8, 4.8 Hz, 1H), 3.82 - 3.71 (m, 2H), 3.66 (dd, J= 9.0, 6.6 Hz, 1H), 3.37 (dd, J= 9.0, 7.1 Hz, 1H), 2.21 - 2.06 (m, 1H), 1.73 (s, 1H), 1.29 - 1.14 (m, 2H), 1.12 - 0.98 (m, 2H).

(3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4- yl)methoxy)hexahydro-furo[3,2-b]furan-3-ol (la-4) (182 mg) was also isolated. LC/MS observed [M+Na], 434.04; ¾ NMR (500 MHz, Chloroform-r ) d 7.51 - 7.31 (m, 3H), 4.45 (t, J= 4.9 Hz, 1H), 4.41 - 4.29 (m, 2H), 4.24 (dd, J= 11.0, 5.1 Hz, 2H), 4.01 - 3.90 (m, 1H), 3.86 - 3.73 (m, 3H), 3.51 (dd, J= 9.4, 5.8 Hz, 1H), 2.61 (s, 1H), 2.13 (tt, J= 8.2, 5.0 Hz,

1H), 1.36 - 1.21 (m, 2H), 1.20 - 1.03 (m, 2H).

Note: The stereochemistry of compounds (la-3) and (la-4) was assigned based on NOE observed as shown.

To (3S,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (la-3) (65 mg, 0.158 mmol) and methyl 2-bromo-4-isopropoxy- benzo[d]thiazole-6-carboxylate (lb-l) (78 mg, 0.236 mmol) in DMF (2 ml) was added NaH (3.78 mg, 0.158 mmol, 60% dispersion in mineral oil) and the mixture was stirred at RT for 6 h. The mixture was diluted with EtOAc, washed with NaHCCb solution, water, brine, dried (Na2SCh). filtered and concentrated. The residue was purified by CombiFlash eluting with 0 to 45% acetone/hexane to give methyl 2-(((3S,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)hexahydrofuro[3,2-b]fur an-3-yl)oxy)-4- isopropoxybenzo[d]thiazole-6-carboxylate (lb-2) (46 mg, 0.070 mmol, 44.1 % yield).

LC/MS observed [M+H], 661.13; ¾ NMR (400 MHz, Chloroform-ri) d 7.91 (d, J= 1.5 Hz, 1H), 7.50 (d , J= 1.5 Hz, 1H), 7.35 (dq, J= 8.4, 1.4 Hz, 2H), 7.32 - 7.23 (m, 1H), 5.58 (d, J = 3.3 Hz, 1H), 4.78 (p, .7= 6.1 Hz, 1H), 4.59 (h, J= 6.9, 5.7 Hz, 2H), 4.52 (d, J= 12.4 Hz,

1H), 4.29 (d , J= 12.4 Hz, 1H), 4.16 - 3.95 (m, 2H), 3.91 (td, J= 6.6, 4.3 Hz, 1H), 3.85 (s, 5H), 3.72 (dd, J= 9.1, 6.4 Hz, 1H), 3.47 (dd, J= 9.1, 6.8 Hz, 1H), 2.27 - 2.06 (m, 1H), 1.93 (s, 3H), 1.76 (d, J= 5.3 Hz, 1H), 1.35 dd, .7= 6.1, 1.4 Hz, 7H), 1.23-1.17 (m, 2H), 1.16 - 1.00 (m, 2H). Step lc

To methyl 2-(((3S,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxyb enzo[d]thi azole-6- carboxylate (lb-2) (43 mg, 0.065 mmol) in Methanol (1 ml) and THF (1 ml) was added LiOH (0.097 ml, 0.097 mmol, 1M) and the mixture was stirred at 45 °C for l6h. The mixture was concentrated, azeotroped with ACN. To the residue was added DCM/MeOH, and the mixture was filtered. The filtrate was collected and concentrated and the residue was lyophilized to give lithium 2-(((3S,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexa-hydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxy benzo[d]thiazole-6- carboxylate (Example 1) (44.8 mg, 0.069 mmol) as a white powder. LC/MS observed

[M+H], 647.12.

Example 2

Step 2a

To (3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (la-4) (50 mg, 0.121 mmol) and methyl 2-bromo-4- isopropoxybenzo-[d]thiazole-6-carboxylate (lb-l) (60.1 mg, 0.182 mmol) in DMF (2 ml) was added NaH (4.85 mg, 0.121 mmol) and the mixture was stirred at RT for l6h. The mixture was diluted with EA, washed with NaHCCb solution, water, brine, dried (Na2S0 4 ), filtered and concentrated. The residue was purified by CombiFlash eluting with hexane to 45% acetone/hexane to give methyl 2-(((3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)-isoxazol-4-yl)methoxy)hexa-hydro-furo[3,2-b] furan-3-yl)oxy)-4- isopropoxybenzo[d]thiazole-6-carboxylate (2a) (58 mg, 0.088 mmol, 72.3 % yield). LC/MS observed [M+H], 661.13 ^ NMR (400 MHz, Chloroform-r/) d 7.82 (d, ./ = 1.5 Hz, 1H), 7.40 (d, J= 1.6 Hz, 1H), 7.32 - 7.22 (m, 2H), 7.16 (t, = 8.1 Hz, 1H), 5.47 (td, J= 6.3, 5.0 Hz, 1H), 4.77 - 4.58 (m, 2H), 4.31 - 4.09 (m, 3H), 3.94 (dd, J= 9.6, 6.3 Hz, 1H), 3.84 - 3.55 (m,

5H), 3.76 (s, 3H), 2.01 - 1.85 (m, 1H), 1.27 (dd, J= 6.1, 0.8 Hz, 6H), 1.18 - 1.04 (m, 2H), 1.04 - 0.79 (m, 2H).

Step 2b

Example 2 was synthesized by following the similar experimental procedure in step lc for Example 1. LC/MS observed [M+H], 647.11.

Example 3

Step 3 a

To 2-(((3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexa-hydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxy benzo[d]thiazole-6- carboxylic acid (Example 2) (30 mg, 0.046 mmol), cyclopropanesulfonamide (3a) (8.42 mg, 0.069 mmol) and EDC (13.32 mg, 0.069 mmol) in DCM (0.8 ml) was added DMAP (8.49 mg, 0.069 mmol) and the mixture was stirred at RT for l6h. The mixture was concentrated and the residue was purified by HPLC eluting with 0.1% formic acid in Water and ACN to give 2-(((3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-N-(cyclopropy lsulfonyl)-4- isopropoxybenzo[d]thiazole-6-carboxamide (example 3) (l5mg, 0.020 mmol, 43.1 % yield). LC/MS observed [M+H], 750.14; ¾ NMR (400 MHz, Chloroform-d) d 7.78 - 7.56 (m, 1H), 7.48 - 7.21 (m, 4H), 5.54 (q, J = 5.9 Hz, 1H), 4.76 (dq, J = 12.1, 6.1, 5.3 Hz, 2H), 4.38 - 4.17 (m, 3H), 4.01 (dd, J = 9.7, 6.3 Hz, 1H), 3.94 - 3.64 (m, 4H), 3.07 (tt, J = 8.3, 4.7 Hz, 1H), 2.03 (tt, J = 8.3, 5.0 Hz, 1H), 1.35 (ddd, J = 8.5, 6.3, 2.8 Hz, 9H), 1.26 - 0.98 (m, 6H).

Example 4

Step 4a

To (3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (la-4) (58 mg, 0.141 mmol) in DMF (2 ml) was added NaH (8.44 mg, 0.211 mmol) and the mixture was stirred at RT for 30 min then added ethyl 2- chlorobenzo[d]thiazole-6-carboxylate (4a-l) (51.0 mg, 0.211 mmol). The mixture was stirred at RT for 16 h, then quenched with NaHCCb solution, extracted with EtOAc, organic layer separated and washed with water, brine, dried (Na2SCh). filtered and concentrated. The residue was purified by CombiFlash eluting with hexane to 60% acetone/hexane to give ethyl 2-(((3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydro-furo[3,2-b]furan-3-yl)oxy)benzo[d]thiaz ole-6-carboxylate (4a-2) (65 mg). LC/MS observed [M+H], 617.10; ¾ NMR (400 MHz, Chloroform-d) d 8.30 (d, J = 1.6 Hz, 1H), 7.99 (dd, J = 8.5, 1.7 Hz, 1H), 7.61 (d, J = 8.5 Hz, 1H), 7.39 - 7.29 (m, 2H), 7.24 (t, J = 8.0 Hz, 1H), 5.48 (q, J = 5.7 Hz, 1H), 4.80 (t, J = 4.8 Hz, 1H), 4.44 - 4.16 (m, 5H), 4.10 - 3.95 (m, 2H), 3.94 - 3.81 (m, 2H), 3.81 - 3.65 (m, 2H), 2.09-1.99 (m, 1H), 1.25 - 1.13 (m,

4H), 1.13 - 1.00 (m, 2H). Step 4b

To ethyl 2-(((3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy) hexahydrofuro[3,2-b]furan-3-yl)oxy)benzo[d]thiazole-6-carbox ylate (4a-2) (65 mg, 0.105 mmol) in MeOH (1 ml) and Tetrahydrofuran (1 ml) was added LiOH (0.158 ml, 0.158 mmol, 1M), the mixture was stirred at RT for l6h. The mixture was concentrated under reduced pressure and the residue was purified by HPLC eluting with 0.1% formic acid in Water and ACN to give 2-(((3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)hexahydro-furo[3,2-b]fu ran-3- yl)oxy)benzo[d]thiazole-6-carboxylic acid (Example 4) (17 mg). LC/MS observed [M+H], 589.06; ¾ NMR (400 MHz, Chloroform-d) d 8.33 (s, 1H), 8.02 (d, J = 8.5 Hz, 1H), 7.63 (d,

J = 8.4 Hz, 1H), 7.40 - 7.20 (m, 3H), 5.49 (q, J = 5.6 Hz, 1H), 4.82 (t, J = 4.9 Hz, 1H), 4.44 - 4.18 (m, 3H), 4.01 (dd, J = 9.8, 6.0 Hz, 1H), 3.95 - 3.67 (m, 4H), 2.03 (tt, J = 8.4, 5.1 Hz,

1H), 1.32 - 1.12 (m, 2H), 1.10 - 0.81 (m, 2H). Example 5

Step 5 a

To (3R,3aR,6R,6aR)-hexahydrofuro[3,2-b]furan-3,6-diol compound 5a-l (isomannide, 5a-l) (1.008 g, 6.90 mmol) in DMSO (18 ml) was added potassium tert-butoxide (1.161 g g, 10.35 mmol) and DMA (6 ml). The resulting cloudy mixture was heated up to 60 °C, stirred for 15 min. 4-(chloromethyl)-5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazo le (la-l) (2.087 g, 6.90 mmol) was added in one portion and the mixture was stirred at 60 °C for 2 h. The mixture was diluted with EtOAc/NaHCCb solution and the organic layer was separated, washed with water, brine, dried (Na2S04), filtered and concentrated. The residue was purified by

CombiFlash purification eluting with 0% to 90% acetone/hexane to give (3R,3aR,6R,6aR)-6- ((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazol-4-yl)methoxy) hexahydrofuro[3,2-b]furan-3- ol (5a-2) (l.89g, 66.5%). LC/MS observed [M+H], 412.06; 1H NMR (400 MHz,

Chloroform-d) d 7.42 - 7.23 (m, 3H), 4.51 (d, J = 12.4 Hz, 1H), 4.42 - 4.31 (m, 2H), 4.26 (d, J = 12.5 Hz, 1H), 4.14 (s, 1H), 3.96 - 3.73 (m, 3H), 3.47 (dt, J = 9.3, 7.0 Hz, 2H), 2.64 (s, 1H), 2.17-2.08 (m, 1H), 1.29 - 1.15 (m, 2H), 1.15 - 0.99 (m, 2H).

Step 5b To methyl 4-amino-3-fluorobenzoate (5b-l) (45g, 266 mmol) and sodium thiocyanate (86 g, 1064 mmol) in acetic acid (350 ml) at 0 °C was added bromine (13.57 ml, 263 mmol) in AcOH (100 ml) via additional funnel over lh, and the mixture was warmed up to RT and stirred for 2 days. The mixture was filtered to collect the first crop of solid, washed with water and dried in the open air to give methyl 2-amino-4-fluorobenzo[d]thiazole-6- carboxylate (5b-2) (65 g) as yellow solid. ¾ NMR (400 MHz, DMSO-ri6) d 8.28 - 8.02 (m, 1H), 7.58 (dd, J= 11.5, 1.6 Hz, 1H), 3.84 (s, 3H).

Step 5 c

To copper(II) bromide (29.6 g, 133 mmol) and methyl 2-amino-4-fluorobenzo[d]thiazole-6- carboxylate (5b-2) (20 g, 88 mmol) in acetonitrile (300 ml) with water bath was slowly added tert-butyl nitrite (12.85 ml, 97 mmol) over 10 min. The resulting mixture was stirred for 2 day sand diluted with MTBE and water, stirred for lOmin, a lot of precipitate formed. The mixture was filtered through celite andorganic layer was separated, washed with water (3X), brine (2X), dried, filtered, concentrated. The residue was purified by CombiFlash eluting with 0 to 25% acetone/Hexane. The fractions containing product was combined and concentrated then trituated with hexane. The white solid was collected via filtration to give methyl 2-bromo-4-fluorobenzo[d]thiazole-6-carboxylate (5c) ( 5.43 g). Ή NMR (400 MHz, Chloroform-d) d 8.34 (dd, J = 1.4, 0.6 Hz, 1H), 7.85 (dd, J = 10.5, 1.4 Hz, 1H), 3.97 (s, 3H), 1.56 (s, 9H).

Step 5d

To (3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (5a-2) (33 mg, 0.080 mmol) and methyl 2-bromo-4- fluorobenzo[d]thiazole-6-carboxylate (5c) (27.9 mg, 0.096 mmol) in DMA (0.8 ml) and acetonitrile (0.800 ml) was added cesium carbonate (78 mg, 0.240 mmol). The mixture was heated up at 90 °C for l6h and then diluted with EtOAc, washed with water, brine, dried, filtered and concentrated. The mixture was purified by CombiFlash eluting with 0 to 50% EtO Ac/hexane to give methyl 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)hexahydrofuro[3,2-b]fur an-3-yl)oxy)-4- fluorobenzo[d]thiazole-6-carboxylate (5d) (20 mg, 40%). LC/MS observed [M+H], 621.08;

¾ NMR (400 MHz, Chloroform-d) d 8.16 (d, J = 1.5 Hz, 1H), 7.76 (dd, J = 10.9, 1.5 Hz, 1H), 7.51 - 7.32 (m, 3H), 5.62 (td, J = 6.4, 5.5 Hz, 1H), 4.94 (t, J = 5.2 Hz, 1H), 4.58 (d, J =

12.5 Hz, 1H), 4.48 (t, J = 5.0 Hz, 1H), 4.34 (d, J = 12.4 Hz, 1H), 4.13 (dd, J = 9.7, 6.3 Hz, 1H), 4.07 - 3.90 (m, 2H), 3.94 (s, 3H), 3.84 (dd, J = 8.7, 6.7 Hz, 1H), 3.57 (t, J = 8.5 Hz,

1H), 2.32 - 2.20 (m, 1H), 1.35 - 1.24 (m, 2H), 1.22 - 1.09 (m, 2H).

Step 5e

To methyl 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-fluorobenzo [d]thiazole-6-carboxylate (5d) (20 mg, 0.032 mmol) in tetrahydrofuran (1 ml) was added LiOH (1 M) (0.064 ml, 0.064 mmol) and the mixture was stirred at RT for l6h. The mixture was diluted with EtOAc, adjust pH to ~5 with 1N HC1, the organic layer was separted, washed with water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with 0% to 60% acetone/hexane to give 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)hexahydrofuro[3,2-b]fur an-3-yl)oxy)-4- fluorobenzo[d]thiazole-6-carboxylic acid (Example 5) (11.3 mg, 0.019 mmol, 57.8 % yield). LC/MS observed [M+H], 607.06; Ή NMR (400 MHz, Chloroform-d) d 8.19 (d, J = 1.4 Hz, 1H), 7.79 (dd, J = 10.7, 1.5 Hz, 1H), 7.49 - 7.39 (m, 2H), 7.39 - 7.31 (m, 1H), 5.63 (q, J =

6.2 Hz, 1H), 4.97 (t, J = 5.2 Hz, 1H), 4.58 (d, J = 12.5 Hz, 1H), 4.49 (t, J = 5.0 Hz, 1H), 4.35 (d, J = 12.5 Hz, 1H), 4.24 - 4.09 (m, 1H), 4.06 - 3.94 (m, 2H), 3.85 (dd, J = 8.7, 6.8 Hz, 1H), 3.58 (t, J = 8.5 Hz, 1H), 2.29 - 2.19 (m, 1H), 1.36 - 1.27 (m, 2H), 1.20 - 1.09 (m, 2H).

Example 6

Example 6 was synthesized synthesized by following the similar experimental procedure as for Example 5. LC/MS observed [M+H], 623.12; 'H NMR (400 MHz, Chloroform-6/) 5 8.21 (d, = 1.5 Hz, 1H), 7.80 (dd, = 10.7, 1.5 Hz, 1H), 7.63 - 7.47 (m, 2H), 7.47 - 7.36 (m, 2H),

5.65 (q, .7= 6.1 Hz, 1H), 4.97 (t, .7= 5.2 Hz, 1H), 4.66 (d, J= 11.9 Hz, 1H), 4.51 (t, .7= 5.0 Hz, 1H), 4.38 (d, J= 11.8 Hz, 1H), 4.26 - 4.07 (m, 1H), 4.07 - 3.92 (m, 2H), 3.84 (dd, J = 8.7, 6.7 Hz, 1H), 3.60 (t, J= 8.6 Hz, 1H), 2.22 (tt, J= 8.5, 5.1 Hz, 1H), 1.33 - 1.22 (m, 2H), 1.13 (dd, J= 8.2, 3.2 Hz, 2H).

Example 7

step 7 a

Method A:

To (3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (5a-2) (41 mg, 0.099 mmol) and methyl 2-bromo-4- isopropoxybenzofd] thiazole-6-carboxylate (lb-l) (49.3 mg, 0.149 mmol) in DMF (2 ml) was added sodium hydride (5.97 mg, 0.149 mmol) and the mixture was stirred at RT for l6h. The mixture was diluted with EtOAc, washed with NaHCCh solution, water, brine, dried

(Na2SCh). fitlered and concentrated. The residue was purified by CombiFlash eluting with hexane to 50% EA/hexane to give methyl 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)-isoxazol-4-yl)methoxy)hexahy-drofuro[3,2-b]f uran-3-yl)oxy)-4- isopropoxybenzo[d]thiazole-6-carboxylate (7a) (22 mg, 33 % yield). LC/MS observed

[M+H], 661.13.

Method B:

To (3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (5a-2) (313 mg, 0.759 mmol) and methyl 2-bromo-4- isopropoxybenzo [d]thiazole-6-carboxylate (lb-l) (376 mg, 1.139 mmol) in DMA (3 ml) and acetonitrile (3.00 ml) was added cesium carbonate (742 mg, 2.278 mmol). The resulting mixture was stirred at 90 °C for 20 h. The mixture was diluted with EA, washed with 1N HC1, water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with a gradient of 0% to 80% ethyl aceate/hexane to give methyl 2- (((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)is oxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxyb enzo[d]thi azole-6- carboxylate (7a ) (402 mg, 0.608 mmol, 80 % yield). LC/MS observed [M+H], 661.13; ¾ NMR (500 MHz, Chloroform-d) d 8.00 (d, J = 1.5 Hz, 1H), 7.58 (d, J = 1.5 Hz, 1H), 7.50 - 7.41 (m, 2H), 7.41 - 7.32 (m, 1H), 5.65 (td, J = 6.7, 5.4 Hz, 1H), 4.91 (t, J = 5.1 Hz, 1H), 4.84 (hept, J = 6.1 Hz, 1H), 4.60 (d, J = 12.5 Hz, 1H), 4.51 (t, J = 4.9 Hz, 1H), 4.37 (d, J =

12.5 Hz, 1H), 4.19 (dd, J = 9.6, 6.5 Hz , 1H), 3.99 (ddd, J = 8.2, 6.8, 5.0 Hz, 1H), 3.94 (s,

3H), 3.87 (dd, J = 8.7, 6.8 Hz, 2H), 3.60 (t, J = 8.5 Hz, 1H), 2.32 - 2.15 (m, 1H), 1.44 (dd, J = 6.1, 3.0 Hz, 6H), 1.39 - 1.24 (m, 2H), 1.24 - 1.09 (m, 2H). step 7b

Method A:

To methyl 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxyb enzo[d]thiazole-6- carboxylate (7a) (400mg, 0.605 mmol) in Tetrahydrofuran (8 ml) was added potassium trimethylsilanolate (172 mg, 1.209 mmol) and the mixture was heated up to 40°C for 2h. The mixture was diluted with EtO Ac/water, the organic layer was washed with water. The aq. layer was combined, acidified with 1N HC1 and then extracted with EtO Ac (3X). The organic layer was washed with brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with a gradient of 0% to 60% (10% MeOH in ethyl acetate)/hexane to give 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexa-hydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxy benzo[d]thiazole-6- carboxylic acid (Example 7) (193 mg, 0.298 mmol, 49.3 % yield). LC/MS observed [M+H], 647.11; ¾ NMR (400 MHz, Chloroform-ri) d 7.96 (d, J= 1.5 Hz, 1H), 7.51 (d, J= 1.5 Hz,

1H), 7.39 - 7.21 (m, 3H), 5.56 (td, J= 6.7, 5.4 Hz, 1H), 4.82 (t, J= 5.1 Hz, 1H), 4.74 (hept, J = 6.1 Hz, 1H), 4.49 (d, J= 12.5 Hz, 1H), 4.41 (t, .7= 4.9 Hz, 1H), 4.26 (d, J = 12.5 Hz, 1H), 4.09 (dd, J= 9.6, 6.5 Hz, 1H), 3.94 - 3.70 (m, 3H), 3.50 (t, J= 8.5 Hz, 1H), 2.17-2.08 (m,

1H), 1.35 (dd, J= 6.1, 2.0 Hz, 6H), 1.24-1.16 (m, 2H), 1.14 - 0.97 (m, 2H).

Method B:

To methyl 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxyb enzo[d]thi azole-6- carboxylate (7a) (278 mg, 0.420 mmol) in tetrahydrofuran (2 ml) was added LiOH (0.504 ml, 0.504 mmol) and the mixture was stirred at RT for 28 h. The mixture was washed with MTBE, and the aq. layer was separated, acidified and extracted with EtO Ac, the organic layer was separated and washed with water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with a gradient of 0% to 35% acetone - hexane to give 2- (((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)is oxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxyb enzo[d]thiazole-6-carboxylic acid (Example 7) (203 mg, 74.6 % yield).

Example 8

Step 8a

Method A:

To a suspension of 2-chlorobenzo[d]thiazole-6-carboxybc acid (8a-l) (10.7 g, 50.1 mmol) in DCM (180 ml) was added DMF (0.194 ml, 2.504 mmol). To the resulting suspesion was added oxalyl dichloride (4.72 ml, 55.1 mmol) dropwise at 0 °C, and the resulting mixture was stirred at RT for 20 h. Another portion of oxalyl chloride (0.94 ml) was added and the suspension became a slightly cloudy solution after 6h. The mixture was concentrated and chased with DCM to give a light yellow solid.

To this solid was added DCM (180 ml), DMAP (0.306 g, 2.504 mmol) and 1- methylcyclopropane-l -sulfonamide (8a-2) (7.11 g, 52.6 mmol) at 0 °C. Triethylamine (10.47 ml, 75 mmol) was added dropwise at 0 °C and the suspension turn into a light yellow clear solution. The resulting mixture was stirred at RT for l4h and then concentrated. The residue was diluted with EtOAc, washed with 1N HC1, water, brine, dried and concentrated to give crude product as light yellow solid. The crude product was purified by crystallization from MTBE to give 2-chloro-N-((l-methylcyclopropyl)sulfonyl)benzo[d]thiazole-6 -carboxamide (8a-3) (11.34 g, 68.4%). ¾ NMR (400 MHz, Chloroform-d) d 8.46 (s, 1H), 8.38 (d, J = 1.8 Hz, 1H), 8.06 (d, J = 8.5 Hz, 1H), 7.92 (dd, J = 8.6, 1.9 Hz, 1H), 1.96 - 1.77 (m, 2H), 1.62 (s, 3H), 1.07 - 0.96 (m, 2H).

Method B:

To 3-(((ethylimino)methylene)amino)-N,N-dimethylpropan-l -amine hydrochloride (8a- 1) (1.974 g, 10.30 mmol) and l-methylcyclopropane-l -sulfonamide (8a-2) (1.266 g, 9.36 mmol) in DCM (20 ml) was added 3-(((ethylimino)methylene)amino)-N,N-dimethylpropan-l-amine hydrochloride (1.974 g, 10.30 mmol) and DMAP (2.52 g, 20.60 mmol). The resulting mixture was stirred at RT for 16 h and then concentrated under reduced pressure. The residue was diluted with EtOAc, washed with 1N HC1, water, brine, dried, filtered and concentrated to give 2-chloro-N-((l-methylcyclopropyl)sulfonyl)benzo[d]thiazole-6 -carboxamide (8a-3) (1.085 g, 35%). Step 8b

To (3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (5a-2) (29 mg, 0.070 mmol), 2-chloro-N-((l-methylcyclopropyl)- sulfonyl) benzo[d]thiazole-6-carboxamide (8a-3) (34.9 mg, 0.106 mmol) in DMA (1 ml) was added cesium carbonate (68.8 mg, 0.211 mmol) and the mixture was run in microwave reactor at 120 °C for 30 min, and then 140 °C for lh. The mixture was filtered through celite and purified by HPLC to give 2-(((3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)hexa hydrofuro[3,2-b]furan-3-yl)oxy)-N-((l- methylcyclopropyl)sulfonyl)benzo -[d]thiazole-6-carboxamide (Example 8) (38 mg, 76%). LC/MS observed [M+H], 706.10.

Example 9

Example 9 was synthesized by following the similar experimental procedure as for Example 8. LC/MS observed [M+H], 692.09; ¾ NMR (400 MHz, Chloroform-d) d 9.20 (s, 1H), 8.08

(d, J = 1.8 Hz, 1H), 7.71 (dd, J = 8.5, 1.9 Hz, 1H), 7.53 (d, J = 8.5 Hz, 1H), 7.39 - 7.20 (m, 3H), 5.46 (q, J = 6.1 Hz, 1H), 4.87 (t, J = 5.2 Hz, 1H), 4.50 (d, J = 12.5 Hz, 1H), 4.42 (t, J = 5.0 Hz, 1H), 4.27 (d, J = 12.5 Hz, 1H), 4.04 (dd, J = 9.8, 6.2 Hz, 1H), 3.98 - 3.85 (m, 2H), 3.78 (dd, J = 8.7, 6.8 Hz, 1H), 3.51 (t, J = 8.6 Hz, 1H), 3.08 (tt, J = 8.1, 4.8 Hz, 1H), 2.13 (tt, J = 8.5, 5.1 Hz, 1H), 1.34 (dt, J = 7.5, 3.5 Hz, 2H), 1.20 (ddd, J = 6.9, 5.2, 3.8 Hz, 2H), 1.12 0.90 (m, 4H). Example 10

Example 10 was synthesized by following the similar experimental procedure in step 3a as for Example 3.

LC/MS observed [M+H], 779.17; ¾ NMR (400 MHz, Chloroform-d) d 8.97 (s, 1H), 7.90 - 7.66 (m, 1H), 7.52 - 7.33 (m, 4H), 5.60 (q, J = 5.9 Hz, 1H), 4.94 (t, J = 5.2 Hz, 1H), 4.86 (p, J = 6.0 Hz, 1H), 4.59 (d, J = 12.5 Hz, 1H), 4.51 (t, J = 5.0 Hz, 1H), 4.37 (d, J = 12.4 Hz, 1H), 4.15 (dd, J = 9.9, 6.1 Hz, 1H), 4.07 - 3.94 (m, 2H), 3.87 (dd, J = 8.7, 6.8 Hz, 1H), 3.71 - 3.48 (m, 4H), 2.32 - 2.18 (m, 2H), 2.00 - 1.89 (m, 3H), 1.43 (d, J = 6.0 Hz, 6H), 1.36-1.23 (m, 2H), 1.24 - 1.13 (m, 2H), 0.95 - 0.81 (m, 1H).

Example 11

Example 11 was synthesized by following the similar experimental procedure in step 3 a as for Example 3. LC/MS observed [M+H], 750.14; Ή NMR (400 MHz, Chloroform-d) d 7.78 (d, J = 1.6 Hz, 1H), 7.49 - 7.30 (m, 4H), 5.61 (q, J = 6.0 Hz, 1H), 4.94 (t, J = 5.2 Hz, 1H),

4.84 (p, J = 6.1 Hz, 1H), 4.59 (d, J = 12.5 Hz, 1H), 4.51 (t, J = 5.0 Hz, 1H), 4.36 (d, J = 12.5 Hz, 1H), 4.14 (dd, J = 9.8, 6.2 Hz, 1H), 4.06 - 3.93 (m, 2H), 3.87 (dd, J = 8.7, 6.8 Hz, 1H), 3.59 (t, J = 8.6 Hz, 1H), 3.15 (tt, J = 8.1, 4.8 Hz, 1H), 2.36 - 2.12 (m, 2H), 1.51 - 1.44 (m, 2H), 1.42 (d, J = 6.1 Hz, 6H), 1.36 - 1.26 (m, 2H), 1.26 - 1.04 (m, 4H). Example 12

Example 12 was synthesized by following the similar experimental procedure in step 3a as for Example 3. LC/MS observed [M+H], 764.15; Ή NMR (400 MHz, Chloroform-d) d 8.89 (s, 1H), 7.80 (d, J = 1.6 Hz, 1H), 7.51 - 7.33 (m, 4H), 5.61 (q, J = 6. l Hz, 1H), 4.94 (t, J = 5.2

Hz, 1H), 4.85 (h, J = 6.1 Hz, 1H), 4.60 (d, J = 12.5 Hz, 1H), 4.52 (t, J = 5.0 Hz, 1H), 4.37 (d,

J = 12.5 Hz, 1H), 4.15 (dd, J = 9.8, 6.2 Hz, 1H), 4.08 - 3.95 (m, 2H), 3.88 (dd, J = 8.7, 6.8 Hz, 1H), 3.60 (t, J = 8.6 Hz, 1H), 2.31 - 2.12 (m, 4H), 1.92 - 1.77 (m, 2H), 1.43 (d, J = 6.1 Hz, 6H), 1.38 - 1.26 (m, 2H), 1.26 - 1.12 (m, 2H), 1.12 - 0.94 (m, 2H).

Example 13

Example 13 was synthesized by following the similar experimental procedure in step 3a as for Example 3. LC/MS observed [M+H], 793.19; Ή NMR (400 MHz, Chloroform-d) d 8.98 (s, 1H), 7.75 (t, J = 1.7 Hz, 1H), 7.53 - 7.29 (m, 4H), 5.62 (qd, J = 6.3, 1.8 Hz, 1H), 4.91 - 4.76 (m, 2H), 4.59 (dd, J = 12.5, 1.7 Hz, 1H), 4.51 (td, J = 4.9, 1.7 Hz, 1H), 4.36 (dd, J =

12.5, 1.7 Hz, 1H), 4.15 (ddd, J = 9.7, 6.2, 1.7 Hz, 1H), 4.04-3.92 (m, 2H), 3.86 (ddd, J = 8.6, 6.7, 1.7 Hz, 1H), 3.59 (td, J = 8.5, 1.7 Hz, 1H), 3.46 (t, J = 4.9 Hz, 4H), 2.21 (m, 1H), 1.78- 1.53 (m, 5H), 1.42 (dd, J = 6. l, 1.6 Hz, 6H), 1.39 - 1.25 (m, 2H), 1.17 (dq, J = 8.5, 2.4 Hz, 2H). Example 14

Example 14 was synthesized by following the similar experimental procedures as for Example 7. LC/MS observed [M+H], 589.06; ¾ NMR (400 MHz, Chloroform-d) d 8.40 (d, J = 1.6 Hz, 1H), 8.08 (dd, J = 8.5, 1.7 Hz, 1H), 7.69 (d, J = 8.5 Hz, 1H), 7.47 - 7.31 (m, 3H),

5.56 (q, J = 6.3 Hz, 1H), 4.94 (t, J = 5.2 Hz, 1H), 4.58 (d, J = 12.5 Hz, 1H), 4.50 (t, J = 5.0 Hz, 1H), 4.35 (d, J = 12.4 Hz, 1H), 4.14 (dd, J = 9.7, 6.3 Hz, 1H), 4.06 - 3.93 (m, 2H), 3.86 (dd, J = 8.7, 6.8 Hz, 1H), 3.58 (t, J = 8.5 Hz, 1H), 2.31 - 2.09 (m, 1H), 1.33 - 1.24 (m, 3H), 1.18 - 1.06 (m, 2H).

Example 15

Step l5a

To (3R,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (la-4) (61 mg, 0.148 mmol), benzoic acid (27.1 mg, 0.222 mmol) and triphenylphosphine (58.2 mg, 0.222 mmol) in tetrahydrofuran (1.5 ml) was added DIAD (0.043 ml, 0.222 mmol) and the mixture was stirred at RT for 20 h. The mixture was concentrated and the residue was purified by CombiFlash eluting with 0 to 70%

acetone/hexane to give (3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4- yl)methoxy)hexa-hydrofuro[3,2-b]furan-3-yl benzoate (l5a) (63 mg, 0.122 mmol, 82 % yield). LC/MS observed [M+H], 516.09. Step l5b

To (3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-yl benzoate (l5a) (63 mg, 0.122 mmol) in tetrahydrofuran (1 ml) and MeOH (1 ml) was added LiOH (0.183 ml, 0.183 mmol, 1M) and the mixture was stirred at RT for 3h. The mixture was diluted with EtOAc, washed with water, brine, dried (Na 2 S04), filtered and concentrated. The resiude was purified by CombiFlash eluting with 0 to 70% acetone-hexane to give (3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol- 4-yl)methoxy)hexa-hydrofuro[3,2-b]furan-3-ol (15b) (29 mg, 0.070 mmol, 57.7 % yield). 'H NMR (400 MHz, Chloroform-ri) d 7.50 - 7.30 (m, 3H), 4.48 - 4.41 (m, 1H), 4.37 (d, J= 5.5 Hz, 3H), 4.31 - 4.21 (m, 1H), 3.94 - 3.85 (m, 1H), 3.84 - 3.74 (m, 2H), 3.68 (qd, J= 10.1,

3.0 Hz, 2H), 2.17 - 2.09 (m, 1H), 1.28 (ddt, J= 7.1, 5.3, 3.0 Hz, 2H), 1.22 - 1.03 (m, 2H).

Step l5c

To (3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-ol (l5b) (29 mg, 0.070 mmol) and methyl 2-bromo-4- isopropoxybenzofd] thiazole-6-carboxylate (lb-l) (34.8 mg, 0.106 mmol) in DMA (0.8 ml) and acetonitrile (0.800 ml) was added cesium carbonate (34.4 mg, 0.106 mmol). The resulting mixture was stirred at in microwave reactor at l20°C for 2.5 h. The mixture was diluted with EtOAc, washed with water, brine, dried (Na^SCri). filtered and concentrated. The residue was purified by CombiFlash eluting with a gradient of 0% to 45% acetone - hexane to give methyl 2-(((3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)-hexahydrofuro[3,2-b]fu ran-3-yl)oxy)-4- isopropoxybenzo[d]thiazole-6-carboxylate (l5c) (25 mg, 0.038 mmol, 53.7 % yield). LC/MS observed [M+H], 661.13.

Step l5d

To methyl 2-(((3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl) isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-yl)oxy)-4-isopropoxyb enzo[d]thi azole-6- carboxylate (l5c) (25 mg, 0.038 mmol) in tetrahydrofuran (1 ml) was added LiOH (0.057 ml, 0.057 mmol, 1M ) and the mixture was stirred at RT for 24 h. The mixture was diluted with EA/1N HC1, the organic layer was separated and washed with water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with 0 to 70% acetone/hexane to give 2-(((3S,3aR,6S,6aR)-6-((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methoxy)-hexahydrofuro[3,2-b]fu ran-3-yl)oxy)-4- isopropoxybenzo[d]thiazole-6-carboxylic acid (Example 15) (11.5 mg, 0.018 mmol, 47.0 % yield). LC/MS observed [M+H], 647.11; Ή NMR (500 MHz, Chloroform-d) d 8.00 (d, J = 1.5 Hz, 1H), 7.56 (d, J = 1.5 Hz, 1H), 7.41 - 7.30 (m, 2H), 7.26 (dd, J = 17.1, 7.6 Hz, 1H),

5.61 (d, J = 3.3 Hz, 1H), 4.80 (p, J = 6.1 Hz, 1H), 4.58 (d, J = 3.8 Hz, 1H), 4.38 (dd, J = 9.0, 3.8 Hz, 1H), 4.36 - 4.25 (m, 3H), 4.06 (d, J = 11.1 Hz, 1H), 3.95 (dd, J = 11.1, 3.6 Hz, 1H), 3.91 - 3.84 (m, 1H), 3.75 - 3.64 (m, 2H), 2.05 (tt, J = 8.3, 5.0 Hz, 1H), 1.37 (dd, J = 6.1, 2.8 Hz, 6H), 1.28 - 1.15 (m, 2H), 1.07 (ddd, J = 8.4, 3.3, 1.8 Hz, 2H).

Example 16

Example 16 was synthesized by following the similar experimental procedure in step 7b as for Example 7. LC/MS observed [M+H], 583.10; Ή NMR (400 MHz, Chloroform-d) d 8.40 (d, J = 1.9 Hz, 1H), 8.14 (dd, J = 8.8, 2.0 Hz, 1H), 8.02 (d, J = 8.8 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 7.41 - 7.21 (m, 3H), 6.97 (d, J = 8.9 Hz, 1H), 5.64 - 5.44 (m, 1H), 4.91 (t, J = 5.0 Hz, 1H), 4.60 - 4.44 (m, 2H), 4.29 (d, J = 12.5 Hz, 1H), 4.18 (dd, J = 9.1, 6.8 Hz, 1H), 3.92 (ddd, J = 8.1, 6.8, 5.0 Hz, 1H), 3.87 - 3.74 (m, 2H), 3.55 (t, J = 8.5 Hz, 1H), 2.17 (dt, J = 8.4, 5.1 Hz, 1H), 1.32 - 1.17 (m, 2H), 1.11-1.04 (m, 2H).

Example 17

Step l7a

To (3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy) hexahydrofuro[3,2-b]furan-3-ol (5a-2) (1.6 g, 3.88 mmol) in DCM (30 ml) was added DMP (2.469 g, 5.82 mmol) in portions, and the mixture was stirred at RT for l6h. To the mixture was added MTBE/water, and the mixture was filtered through celite. The organic layer was separated and washed with water, brine, dried (Na 2 S04 for l6h), filtered, concentrated to give (3aS,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazo l-4-yl)methoxy)tetrahydro- furo[3,2-b]furan-3(2H)-one (l7a) (1.63 g) as crude Product. 'H NMR (400 MHz,

Chloroform-d) d 7.51 - 7.31 (m, 4H), 4.81 (dd, J = 6.1, 5.0 Hz, 1H), 4.53 (d, J = 12.4 Hz,

1H), 4.39 (d, J = 12.4 Hz, 1H), 4.24 (d, J = 6.2 Hz, 1H), 4.09 (d, J = 17.5 Hz, 1H), 3.96 (d, J = 17.5 Hz, 1H), 3.88 (dd, J = 9.4, 5.9 Hz, 1H), 3.61 (dd, J = 9.4, 6.3 Hz, 1H), 2.17 (tt, J = 8.4, 5.1 Hz, 1H), 1.35 - 1.24 (m, 2H), 1.15 (ddd, J = 8.2, 7.0, 4.2 Hz, 2H).

Step l7b

To methyl 2-bromo-4-isopropoxybenzo[d]thiazole-6-carboxylate (lb-l) (127 mg, 0.384 mmol) in tetrahydrofuran (2 ml) at -78 °C was added isopropylmagnesium chloride (0.192 ml, 0.384 mmol) dropwise, color getting darker but still a clear solution. After 30 min, to this mixture was added (3aS,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazo l-4- yl)methoxy)tetra-hydrofuro[3,2-b]furan-3(2H)-one (l7a) (105 mg, 0.256 mmol) in THF (2 ml) and the mixture was stirred at -78 °C for lh, then warmed up to 0 °C and quenched with NaHCCb solution. The mixture was diluted with EtO Ac/water, and the organic layer was separated and washed with water, brine, dried, filtered, concentrated. The residue was purified by CombiFlash eluting with 0 to 45% EtO Ac/hexane to give methyl 2- ((3R,3aS,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)iso xazol-4-yl)methoxy)-3- hydroxyhexahydrofuro[3,2-b]furan-3-yl)-4-isopropoxybenzo[d]t hiazole-6-carboxylate(l7b) (41 mg, 24.2%). LC/MS observed [M+H], 661.13; Ή NMR (500 MHz, Chloroform-d) d 8.19 (d, J = 1.4 Hz, 1H), 7.59 (d, J = 1.5 Hz, 1H), 7.47 - 7.40 (m, 2H), 7.40 - 7.33 (m, 1H), 4.94 - 4.87 (m, 2H), 4.82 - 4.74 (m, 1H), 4.68 - 4.61 (m, 1H), 4.49 - 4.40 (m, 1H), 4.33 (d, J = 9.8 Hz, 1H), 4.17 - 4.10 (m, 1H), 3.97 - 3.88 (m, 5H), 3.81 (dd, J = 8.6, 6.7 Hz, 1H), 2.23 (tt, J = 8.4, 5.0 Hz, 1H), 1.47 (dd, J = 6.1, 1.7 Hz, 6H), 1.37 - 1.24 (m, 2H), 1.24 - 1.07 (m, 2H).

Step l7c

Example 17 was synthesized by following the similar experimental procedure in step lc for Example 1. LC/MS observed [M+H], 647.11 ; ¾ NMR (400 MHz, Chloroform-d) d 8.27 (d, J = 1.4 Hz, 1H), 7.63 (d, J = 1.4 Hz, 1H), 7.46 - 7.31 (m, 3H), 4.96 - 4.86 (m, 2H), 4.80 (t, J = 5.0 Hz, 1H), 4.65 (d, J = 12.6 Hz, 1H), 4.42 (d, J = 12.5 Hz, 1H), 4.34 (d, J = 9.8 Hz, 1H), 4.17-4.12 (m, 1H), 3.96 (ddd, J = 16.4, 7.7, 5.2 Hz, 2H), 3.82 (dd, J = 8.4, 6.4 Hz, 1H), 2.23 (tt, J = 8.3, 5.1 Hz, 1H), 1.50 - 1.45 (d, J = 6.0 Hz, 6H), 1.34 - 1.24 (m, 2H), 1.20-1.11 (m, 2H).

Example 18

Step l8a

To methyl 2-((3R,3aS,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)i soxazol-4- yl)methoxy) -3-hydroxyhexahydrofuro[3,2-b]furan-3-yl)-4-isopropoxybenzo[ d]thiazole-6- carboxylate (l7b) (32 mg, 0.048 mmol) in DCM (1 ml) at -78 °C was added deoxofluor (0.089 ml, 0.484 mmol) and the mixture was stirred at -78 °C for 2h, then slowly warmed up to at 0 °C and quenched with NaHCCb solution. The mixture was diluted with EtO Ac/water, and the organic layer was separated and washed with water, brine, and dried, concentrated. The residue was purified by CombiFlash eluting with 0 to 40% EtOAc-hexane to give methyl 2-((3S,3aS,6R,6aR) -6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazol-4-yl)metho xy)-3- fluorohexahydrofuro[3,2-b]furan-3-yl)-4-isopropoxybenzo[d]th iazole-6-carboxylate (l8a)

(24 mg, 0.036 mmol, 74.8 % yield) as yellow foam. LC/MS observed [M+H], 663.13; ¾ NMR (500 MHz, Chloroform-d) d 8.21 (d, J = 1.4 Hz, 1H), 7.60 (d, J = 1.5 Hz, 1H), 7.43 (ddd, J = 8.4, 4.1, 1.3 Hz, 2H), 7.35 (t, J = 8.1 Hz, 1H), 4.95 (hept, J = 6.1 Hz, 1H), 4.89 - 4.81 (m, 1H), 4.74 (ddd, J = 11.7, 4.7, 1.3 Hz, 1H), 4.64 (d, J = 12.2 Hz, 1H), 4.58 (dd, J = 30.8, 11.6 Hz, 1H), 4.42 (ddd, J = 20.3, 11.6, 1.4 Hz, 1H), 4.36 (d, J = 12.3 Hz, 1H), 4.01 (ddd, J = 8.1, 7.0, 5.2 Hz, 1H), 3.95 (s, 3H), 3.81 (ddd, J = 9.4, 6.9, 2.7 Hz, 1H), 3.60 (t, J = 8.6 Hz, 1H), 2.22 (tt, J = 8.4, 5.1 Hz, 1H), 1.46 (dd, J = 6. l, 1.8 Hz, 6H), 1.35-1.25 (m, 2H), 1.22 - 1.08 (m, 2H).

Step 18b

Example 18 was synthesized by following the similar experimental procedure in step lc for Example 1. LC/MS observed [M+H], 649.11; ¾ NMR (400 MHz, Chloroform-d) d 8.28 (s, 1H), 7.65 (s, 1H), 7.51 - 7.29 (m, 3H), 4.96 (hept, J = 6.2 Hz, 1H), 4.85 (t, J = 4.9 Hz, 1H),

4.75 (dd, J = 11.7, 4.7 Hz, 1H), 4.68-4.30 (m, 4H), , 4.02 (td, J = 7.4, 5.1 Hz, 1H), 3.83 (ddd, J = 9.5, 6.9, 2.6 Hz, 1H), 3.61 (t, J = 8.6 Hz, 1H), 2.29 - 2.18 (m, 1H), 1.48 (d, J = 6.0 Hz, 6H), 1.34-1.24 (m, 2H), 1.23 - 1.11 (m, 2H). Example 19

Step l9a

To a solution of (3aS,6R,6aR)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazo l-4- yl)methoxy)-tetrahydrofuro[3,2-b]furan-3(2H)-one (l7a) (90 mg, 0.219 mmol) in MeOH (1 ml) under N2 at RT was added ammonium acetate (169 mg, 2.194 mmol). The mixture was stirred under N2 for 7 hours sodium cyanoborohydride (13.79 mg, 0.219 mmol) was added into the mixture. The mixture was stirred under N2 for 36 hours and the reaction mixture was concentrated and quenched by addtion of water (2 ml) and 1M aqueous HC1 (1 ml) dropwise. The reation mixture was diluted by DCM (5 ml). The pH of reaction mixture was adjusted to 8.5 by addition of 1M aqueous KOH. The mixture was extracted by DCM (10 mlx3). The combined organic layers was dried over Na2S0 4 , filtered and concentrated to give a crude product. The crude product was purified by CombiFlash eluting with 0 to 10% MeOH/DCM to give (3R,3aR,6R,6aS)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4- yl)methoxy)hexa-hydrofuro[3,2-b]furan-3-amine ( l9a) (13 mg) as an oil. LC/MS observed [M+H], 411.07; ¾ NMR (400 MHz, Chloroform-d) d 7.45 - 7.28 (m, 3H), 4.53 (d, J = 12.4 Hz, 1H), 4.46 (t, J = 4.7 Hz, 1H), 4.28 (d, J = 12.4 Hz, 1H), 4.22 (t, J = 4.7 Hz, 1H), 3.96 - 3.86 (m, 2H), 3.78 (dd, J = 9.0, 6.5 Hz, 1H), 3.52 - 3.38 (m, 2H), 3.33 - 3.20 (m, 1H), 2.22 - 2.11 (m, 1H), 2.05 - 1.95 (m, 2H), 1.32 - 1.20 (m, 2H), 1.17 - 1.04 (m, 2H).

Step l9b

To (3R,3aR,6R,6aS)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)isox azol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-amine (l9a) (12 mg, 0.030 mmol), methyl 2-bromo-4- isopropoxybenzo[d]thiazole-6-carboxylate (lb-l) (12 mg, 0.036 mmol) and cesium carbonate (30 mg, 0.090 mmol) was added DMA (0.6 ml). The mixture was stirred at 70 °C for 24 hours. The solvent was removed. The mixture was treated with DCM, filtered, the filtrate was purified by Combiflash eluting with 0 to 100% EtO Ac/hexane to give methyl 2- (((3R,3aR,6R,6aS)-6-((5-cyclopropyl-3-(2,6-dichlorophenyl)is oxazol-4-yl)methoxy)hexa- hydrofuro[3,2-b]furan-3-yl)amino)-4-isopropoxybenzo[d]thiazo le-6-carboxylate (l9b) (3 mg) as a colorless oil. LC/MS observed [M+H], 660.15; ¾ NMR (400 MHz, Chloroform-d) d 7.91 (d, J = 1.5 Hz, 1H), 7.51 (d, J = 1.5 Hz, 1H), 7.43 - 7.29 (m, 3H), 4.79 (hept, J = 6.0 Hz, 1H), 4.62 - 4.52 (m, 3H), 4.31 (d, J = 12.3 Hz, 1H), 4.26 - 4.19 (m, 1H), 4.01 - 3.93 (m, 1H), 3.89 (s, 3H), 3.87 - 3.80 (m, 1H), 3.58 - 3.44 (m, 2H), 2.21 - 2.12 (m, 1H), 1.42 (d, J = 6.0 Hz, 6H), 1.29 - 1.20 (m, 2H), 1.16 - 1.08 (m, 2H).

Step l9c

Example 19 was synthesized by following the similar experimental procedure in step lc for Example 1. LC/MS observed [M+H], 646.13; ¾ NMR (500 MHz, Methanol-ώ) d 7.91 (d, J = 1.5 Hz, 1H), 7.58 - 7.43 (m, 4H), 4.85 - 4.80 (m, 1H), 4.67 - 4.61 (m, 3H), 4.59 (d, J =

12.0 Hz, 1H), 4.34 (d, J= 12.0 Hz, 1H), 4.15 - 4.09 (m, 1H), 4.05 - 3.99 (m, 1H), 3.82 (dd, J = 9.0, 6.5 Hz, 1H), 3.52 (dd, J= 9.0, 6.5 Hz, 1H), 3.48 - 3.39 (m, 1H), 2.39 - 2.30 (m, 1H), 1.40 (d, J= 6.0 Hz, 6H), 1.21 - 1.16 (m, 2H), 0.94 - 0.87 (m, 2H).

Example 20

Step 20a

A mixture of (7S,8aR)-octahydropyrrolo[l,2-a]pyrazin-7-ol (20a-l) ( 213 mg, 1.5 mmol), methyl 2-bromo-4-isopropoxybenzo[d]thiazole-6-carboxylate (lb-l) (495 mg, 1.500 mmol), and cesium carbonate (977 mg, 3.00 mmol) in DMA (4 ml) was stirred overnight at RT. To the mixture was added water, and extracted with EtOAc. The combined organic layers were washed with brine and concentrated. The residue was purified by CombiFlash eluting with 0 to 65% acetone/hexane to give methyl 2-((7S,8aR)-7-hydroxyhexahydropyrrolo[l,2- a]pyrazin-2(lH)-yl)-4-isopropoxybenzo[d]thiazole-6-carboxyla te (20a-2) (329 mg, 56%) as an off-white solid. ¾ NMR (400 MHz, DMSO-rie) d 8.05 (t, J= 1.2 Hz, 1H), 7.38 (d , J= 1.6 Hz, 1H), 4.91 - 4.79 (m, 2H), 4.30 - 4.23 (m, 1H), 4.12 (d , J= 12.1 Hz, 1H), 4.00 (d , J = 12.7 Hz, 1H), 3.84 (s, 3H), 3.28 - 3.17 (m, 1H), 3.03 (d, J= 10.7 Hz, 1H), 2.88 (t, J= 11.3

Hz, 1H), 2.39 - 2.23 (m, 2H), 2.06 - 1.97 (m, 1H), 1.72 - 1.62 (m, 2H), 1.31 (d, .7= 6.0, 6H). Step 20b

To a solution of methyl 2-((7S,8aR)-7-hydroxyhexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl )-4- isopropoxybenzo[d]thiazole-6-carboxylate (20a-2) (200 mg, 0.511 mmol) in THF (4 ml) at rt was added l8-crown-6 (162 mg, 0.613 mmol), followed by potassium tert-butoxide in THF 1M (1.533 ml, 1.533 mmol). After 15 min stirring, 4-(chloromethyl)-5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazole (la-l) (201 mg, 0.664 mmol) was added. The reaction mixture was stirred at RT for 2h, quenched with water/brine, and extracted with ethyl acetate. The combined organic layers were washed with brine and concentrated. The residue was purified by CombiFlash eluting with 0 to 60% acetone/hexane to give the 2-((7S,8aR)-7-((5- cyclopropyl-3-(2,6-dichlorophenyl)isoxazol-4-yl)methoxy)hexa hydropyrrolo[l,2-a]pyrazin- 2(lH)-yl)-4-isopropoxybenzo[d]thiazole-6-carboxylic acid (Example 20) as a pale yellow foam. LC/MS observed [M+H], 643.17; ¾ NMR (500 MHz, DMSO-ri6) d 12.75 (s, 1H), 8.01 (d, J= 1.4 Hz, 1H), 7.63 (d, J= 8.0 Hz, 2H), 7.55 (dd, J= 8.8, 7.4 Hz, 1H), 7.39 (d, J =

1.5 Hz, 1H), 4.84 (hept, = 6.1 Hz, 1H), 4.25 - 4.15 (m, 2H), 4.07 (d, J = 12.1 Hz, 1H), 3.98 - 3.89 (m, 2H), 3.29 (dd, .7= 9.5, 6.6 Hz, 1H), 3.24 - 3.15 (m, 1H), 2.95 (dd, J= 10.7, 2.7 Hz, 1H), 2.85 - 2.77 (m, 1H), 2.37 - 2.28 (m, 1H), 2.15 - 2.06 (m, 1H), 2.01 - 1.91 (m, 1H), 1.77 (dd, J= 9.4, 5.0 Hz, 1H), 1.49-1.42 (m, 2H), 1.32 (t, J= 6.1 Hz, 6H), 1.20 - 1.06 (m, 4H). Example 21

Example 21 was synthesized by following the similar experimental procedure as for Example 20. LC/MS observed [M+H], 643.17; ¾ NMR (400 MHz, DMSO-r e) d 12.73 (s, 1H), 8.01 (d, = 1.5 Hz, 1H), 7.67 - 7.60 (m, 2H), 7.60 - 7.50 (m, 1H), 7.39 (d, J= 1.6 Hz, 1H), 4.84

(p, .7= 6.1 Hz, 1H), 4.26 - 4.14 (m, 2H), 4.07 (d, J= 12.1 Hz, 1H), 3.98 - 3.89 (m, 2H), 3.28 (d, J = 6.6 Hz, 1H), 3.18 (dd, J= 12.1, 3.3 Hz, 1H), 2.95 (d , J= 10.5 Hz, 1H), 2.81 (dd, J = 12.2, 10.5 Hz, 1H), 2.39 - 2.27 (m, 1H), 2.18 - 2.03 (m, 1H), 1.96 - 1.89 (m, 1H), 1.77 (dd, J = 9.4, 5.0 Hz, 1H), 1.44 (dd, .7= 9.1, 5.4 Hz, 2H), 1.32 (d, .7= 6.0 Hz, 6H), 1.23 - 1.05 (m, 4H).

Example 22

To a solution of methyl 2-((7S,8aR)-7-hydroxyhexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl )-4- isopropoxybenzo[d]thiazole-6-carboxylate (20a-2) (100 mg, 0.255 mmol) and

triphenylphosphine (167 mg, 0.639 mmol) in THF (3 ml) at 0 °C was added TFA (0.049 ml, 0.639 mmol) and DIAD (0.124 ml, 0.639 mmol). The mixture was stirred at 0 °C for 10 min and then warmed up to rt. Sodium benzoate (92 mg, 0.639 mmol) was added, and the reaction mixture was stirred at rt for 3h, quenched with water/brine, and extracted with ethyl acetate. The organic layer was washed with brine and concentrated. The residue was purified by chromatography on silica gel eluting with 0 to 50% acetone/hexane to give compound 22a as a colorless oil (60 mg).

Step 22b

A solution of compound 22a (60 mg) in MeOH (1 ml) was treated with potassium carbonate (34 mg). The reaction mixture was stirred at rt for 3h, quenched with water/brine, and extracted with ethyl acetate. The combined organic layers were washed with brine and concentrated. The residue was purified by chromatography on silica gel eluting with 0 to 60% acetone/hexane to give a methyl 2-((7R,8aR)-7-hydroxyhexahydropyrrolo[l,2-a]pyrazin- 2(lH)-yl)-4-isopropoxybenzo[d]thiazole-6-carboxylate (22b) as a white solid (31 mg).

LC/MS observed, [M+H], 392.15. ¾ NMR (400 MHz, DMSO-rie) d 8.04 (d, J= 1.4 Hz, 1H), 7.38 (d, J= 1.5 Hz, 1H), 4.90 - 4.79 (m, 2H), 4.22 (s, 1H), 4.12 (d, J= 12.3 Hz, 1H), 4.00 (d, J= 12.6 Hz, 1H), 3.84 (s, 3H), 3.59 (s, 1H), 3.03 (t, J= 11.4 Hz, 1H), 2.89 (d, J= 9.7 Hz, 1H), 2.27 (td, J = 10.7, 9.2, 6.5 Hz, 2H), 2.20 - 2.07 (m, 2H), 2.01 (d, J= 9.1 Hz, 1H), 1.31

(d, J = 6.0 Hz, 6H).

Step 22c

To a solution of methyl 2-((7R,8aR)-7-hydroxyhexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl )-4- isopropoxybenzo[d]thiazole-6-carboxylate (compound 22b) (30 mg, 0.077 mmol) in THF (1 ml) at rt was added l8-crown-6 (30.4 mg, 0.115 mmol), followed by potassium tert-butoxide in THF 1M (0.230 ml, 0.230 mmol). After 15 min stirring, 4-(chloromethyl)-5-cyclopropyl-3- (2,6-dichlorophenyl)isoxazole (la-l) (34.8 mg, 0.115 mmol) was added. The reaction mixture was stirred at rt for 3h, quenched with water/brine, and extracted with ethyl acetate. The combined organic layers were washed with brine and concentrated. The residue was purified by chromatography on silica gel eluting with 0 to 60% acetone/hexane to give 2- ((7R,8aR)-7-((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazol-4 - yl)methoxy)hexahydropyrrolo[l,2-a]pyrazin-2(lH)-yl)-4-isopro poxybenzo[d]thiazole-6- carboxylic acid (Example 22) as a pale yellow foam (17 mg). LC-MS M+H LC/MS observed [M+H], 643.17; ¾ NMR (400 MHz, DMSO-rie) d 12.80 (s, 1H), 7.99 (s, 1H), 7.70 - 7.62 (m, 2H), 7.58 (ddd , J= 9.3, 6.8, 1.1 Hz, 1H), 7.37 (t, J= 1.4 Hz, 1H), 4.83 (p, J= 6.1 Hz,

1H), 4.21 (s, 2H), 4.06 (d, ./= 12.1 Hz, 1H), 3.95 (d, = l l.9 Hz, 1H), 3.88 (s, 1H), 3.23 (d, J = 10.8 Hz, 1H), 2.97 (d, J= l l.O Hz, 1H), 2.94 - 2.81 (m, 2H), 2.34 (t, J= 6.8 Hz, 1H), 2.11 (q, .7= 9.9, 7.5 Hz, 4H), 1.97 (d, .7= 7.4 Hz, 1H), 1.31 (dd, .7= 6.1, 1.1 Hz, 6H), 1.26 - 1.11 (m, 2H), 1.15 - 1.06 (m, 2H).

Example 23

Step 23 a

To (3R,3aR,6R,6aR)-hexahydrofuro[3,2-b]furan-3,6-diol (isomannide) (5a-l) (0.918 g, 6.28 mmol) in DMSO (30 ml) was added potassium tert-butoxide (0.846 g, 7.54 mmol) and the suspension was heated at 60 °C for 30 min. To this milky mixture was added 4-

(bromomethyl)-5-cyclopropyl-3-(2-(trifluoromethoxy)phenyl )isoxazole (23a-l) (1.82 g, 5.03 mmol) in DMSO (5 ml) and the mixture was stirred at 60 °C, after 30 min, most of the suspension went into solution. The mixture was stirred for another l.5h. The mixture was diluted with MTBE, washed with water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with a 0-60% gradient of acetone/hexane to give (3R,3aR,6R,6aR)-6-((5-cyclopropyl-3-(2-(trifluoromethoxy)phe nyl)isoxazol-4- yl)methoxy)hexahydrofuro[3,2-b]furan-3-ol (23a-2) (l.28g, 3.00 mmol, 59.6 % yield) as light yellow oil. LC/MS observed [M+H], 428.12; ¾ NMR (400 MHz, Chloroform-d) d 7.67 - 7.45 (m, 2H), 7.39 (dd, J = 8.6, 5.8 Hz, 2H), 4.64 (d, J = 11.8 Hz, 1H), 4.43 (dt, J = 13.0, 5.0 Hz, 2H), 4.35 (d, J = 11.8 Hz, 1H), 4.19 (dt, J = 12.1, 5.9 Hz, 1H), 3.95 (td, J = 6.9, 4.7 Hz, 1H), 3.87 (dt, J = 10.9, 5.7 Hz, 2H), 3.65 - 3.44 (m, 2H), 2.92 (d, J = 8.4 Hz, 1H), 2.21 (tt, J = 8.8, 5.1 Hz, 1H), 1.21 (td, J = 9.3, 8.8, 5.1 Hz, 2H), 1.11 (dt, J = 8.4, 4.0 Hz, 2H). Step 23 b

Compound 23b was synthesized following a similar experimental procedure as in Step 5d. LC/MS observed [M+H], 677.20; ¾ NMR (400 MHz, Chloroform-d) d 7.98 (d, J = 1.5 Hz, 1H), 7.62 - 7.46 (m, 3H), 7.39 (tt, J = 7.6, 1.0 Hz, 2H), 5.65 (td, J = 6.6, 5.4 Hz, 1H), 4.90 (t, J = 5.1 Hz, 1H), 4.82 (p, J = 6. l Hz, 1H), 4.65 (d, J = l l.8 Hz, 1H), 4.51 (t, J = 4.9 Hz, 1H), 4.38 (d, J = 11.8 Hz, 1H), 4.17 (dd, J = 9.6, 6.5 Hz, 1H), 4.02 - 3.88 (m, 2H), 3.92 (s, 3H), 3.84 (dd, J = 8.8, 6.8 Hz, 1H), 3.60 (t, J = 8.5 Hz, 1H), 2.22 (tt, J = 8.4, 5.1 Hz, 1H), 1.42 (dd, J = 6.1, 2.5 Hz, 6H), 1.33 - 1.19 (m, 2H), 1.19 - 1.03 (m, 2H).

Example 23 was synthesized following a similar experimental procedure as in Step 5e.

LC/MS observed [M+H], 663.18; 1H NMR (400 MHz, Chloroform-d) d 8.17 - 7.92 (m, 1H), 7.59 (dd, J = 7.8, 1.7 Hz, 2H), 7.51 (td, J = 7.8, 1.8 Hz, 1H), 7.45 - 7.33 (m, 2H), 5.66 (q, J = 6.3 Hz, 1H), 4.92 (t, J = 5.1 Hz, 1H), 4.82 (hept, J = 6.2 Hz, 1H), 4.65 (d, J = 11.9 Hz, 1H), 4.52 (t, J = 4.9 Hz, 1H), 4.38 (d, J = 11.8 Hz, 1H), 4.22 - 4.12 (m, 1H), 4.04 - 3.90 (m, 2H), 3.84 (t, J = 8.0 Hz, 1H), 3.61 (t, J = 8.5 Hz, 1H), 2.22 (tt, J = 8.4, 5.1 Hz, 1H), 1.43 (dd, J = 6.1, 2.1 Hz, 6H), 1.30 - 1.19 (m, 2H), 1.19 - 1.05 (m, 2H).

Example 24

Example 24 was synthesized following a similar experimental procedure as in Step 3a.

LC/MS observed [M+H], 780.22; Ή NMR (400 MHz, Chloroform-d) d 8.79 (s, 1H), 7.76 (d, J = 1.7 Hz, 1H), 7.58 (dd, J = 7.9, 1.8 Hz, 1H), 7.52 (td, J = 7.8, 1.8 Hz, 1H), 7.46 - 7.33 (m, 3H), 5.65 (q, J = 6.1 Hz, 1H), 4.92 (t, J = 5.2 Hz, 1H), 4.65 (d, J = 11.9 Hz, 1H), 4.51 (t, J = 4.9 Hz, 1H), 4.38 (d, J = 11.8 Hz, 1H), 4.15 (dd, J = 9.7, 6.3 Hz, 1H), 3.96 (dd, J = 9.9, 6.5 Hz, 2H), 3.84 (dd, J = 8.7, 6.8 Hz, 1H), 3.60 (t, J = 8.6 Hz, 1H), 2.41 - 2.15 (m, 1H), 1.82 (q, J = 5.3 Hz, 2H), 1.59 (s, 3H), 1.42 (d, J = 6.1 Hz, 6H), 1.25 (q, J = 4.5 Hz, 3H), 1.12 (dd, J = 8.3, 3.2 Hz, 2H), 1.08 - 0.93 (m, 2H).

Example 25

Example 25 was synthesized following a similar experimental procedure as in Step 3a. LC/MS observed [M+H], LC/MS observed [M+H], 766.20.

Example 26

Step 26a

To copper (I) bromide dimethyl sulfide complex (650 mg) and tert-butyl 6-oxa-3-azabicyclo- [3.l.0]hexane-3-carboxylate (26a-l) (2.9 g, 15.66 mmol) in tetrahydrofuran (90 ml) at -30 °C was added vinylmagnesium chloride (39.1 ml, 62.6 mmol) and the mixture was stirred at -30 °C to -10 °C for 2.5 h, slowly warmed up to 0 °C and stirred for lh, then quenched with NaHCCh solution, filtered through celite. The organic layer was separtated, washed with water, brine, dried, filtered and concentrated to give racemic tert-butyl (3S,4R)-3-hydroxy-4- vinylpyrrolidine-l-carboxylate (26a-2) (3.12 g, 14.63 mmol, 93 % yield) as crude product and used without further purification. ¾ NMR (400 MHz, Chloroform-d) d 5.78 - 5.64 (m, 1H), 5.26 - 5.10 (m, 2H), 4.10 (q, J = 6.0 Hz, 1H), 3.80 - 3.57 (m, 2H), 3.35 - 3.10 (m, 1H), 2.69 (dd, J = 12.3, 6.3 Hz, 1H), 1.46 (s, 9H).

Step 26b To the solution of tert-butyl (3S,4R)-3-hydroxy-4-vinylpyrrolidine-l-carboxylate ( 26a-2) (3.12 g, 14.63 mmol), benzoic acid (2.322 g, 19.02 mmol) and triphenylphosphine (4.99 g, 19.02 mmol) in tetrahydrofuran (100 ml) was added DIAD (3.70 ml, 19.02 mmol) and the mixture was stirred at RT for lh. The mixture was concentrated and purified by CombiFlash eluting with 0 to 30% EtO Ac/hexane to give tert-butyl (3R,4R)-3-(benzoyloxy)-4- vinylpyrrobdine-l-carboxylate (26b) (1.557 g, 4.91 mmol, 33.5 % yield). ¾ NMR (400 MHz, Chloroform-d) d 8.02 (d, J = 7.4 Hz, 2H), 7.58 (t, J = 7.4 Hz, 1H), 7.45 (t, J = 7.4 Hz, 2H), 6.01 - 5.74 (m, 1H), 5.63 - 5.46 (m, 1H), 5.19 (dd, J = 19.6, 14.2 Hz, 2H), 3.87 - 3.63 (m, 2H), 3.63 - 3.30 (m, 2H), 3.05 (b, 1H), 1.45 and 1.49 (s, 9H).

Step 26c

To tert-butyl (3R,4R)-3-(benzoyloxy)-4-vinylpyrrobdine-l-carboxylate (26b) (1.45 g, 4.57 mmol) in tetrahydrofuran (1 ml) and MeOH (0.5 ml) was added NaOH (6.85 ml, 6.85 mmol) and the mixture was stirred at RT for 3h. The mixture was diluted with EtO Ac, washed with 1N NaOH, water, brine, dried, filtered and concentrated to give tert-butyl (3R,4R)-3- hydroxy-4-vinylpyrrobdine-l-carboxylate (26c) (940 mg, 4.41 mmol, 96 % yield) as crude Product. ¾ NMR (400 MHz, Chloroform-d) d 6.07 - 5.65 (m, 1H), 5.38 - 5.07 (m, 3H), 4.28 (d, J = 3.8 Hz, 1H), 3.64 - 3.24 (m, 5H), 2.99 - 2.60 (m, 1H), 1.46 (s, 9H).

Step 26d

Method A:

To tert-butyl (3R,4R)-3-hydroxy-4-vinylpyrrolidine-l-carboxylate (compound 26c) (640 mg, 3.00 mmol) and sodium bicarbonate (756 mg, 9.00 mmol) in DCM (20 ml) was added m- CPBA (1036 mg, 6.00 mmol) and the mixture was stirred at RT for l6h. The volatiles were removed under vacuo and the residue was partitioned between EtO Ac and water, organic layer was separated and washed with 1N LiOH, water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with 0 to 80% EtO Ac/hexane to afford tert- butyl (3R,4R)-3-hydroxy-4-((R)-oxiran-2-yl)pyrrolidine-l-carboxyla te (compound 26d-l) (255 mg). ¾ NMR (400 MHz, Chloroform-d) d 4.44 (q, J = 3.3 Hz, 1H), 3.70 - 3.30 (m, 4H), 3.22 (ddd, J = 6.7, 4.1, 2.8 Hz, 1H), 3.08 (d, J = 3.3 Hz, 1H), 2.98 - 2.85 (m, 1H), 2.68 (td, J = 5.1, 2.6 Hz, 1H), 2.03 - 1.90 (m, 1H), 1.46 (s, 9H).

tert-butyl (3R,4R)-3-hydroxy-4-((S)-oxiran-2-yl)pyrrolidine-l-carboxyla te (compound 26d-2) (37 mg) was also isolated . ¾ NMR (400 MHz, Chloroform-d) d 4.56 - 4.25 (m, 1H), 3.57 (dd, J = 10.6, 8.3 Hz, 1H), 3.53 - 3.43 (m, 2H), 3.20 (dt, J = 6.7, 3.3 Hz, 1H), 2.82 (tt, J = 10.0, 5.0 Hz, 2H), 2.62 - 2.53 (m, 1H), 1.46 (s, 9H).

Method B:

To tert-butyl (3R,4R)-3-hydroxy-4-vinylpyrrolidine-l-carboxylate (compound 26c) (190 mg, 0.891 mmol) and sodium bicarbonate (599 mg, 7.13 mmol) in acetone (5.00 ml) and water (5 ml) at 0 °C was added oxone (2738 mg, 4.45 mmol) in water (5 ml) (a suspension) portionwise over lh. The mixture was stirred at 0 °C for another lh and then diluted with EtO Ac/water. The organic layer was separated, washed with water and brine, then dried, filtered and concentrated. The residue was purified by CombiFlash eluting with with 0 to 70% EtO Ac/hexane to afford tert-butyl (3R,4R)-3-hydroxy-4-((R)-oxiran-2-yl)pyrrolidine-l- carboxylate (compound 26d-l) (60 mg). ¾ NMR (400 MHz, Chloroform-d) d d 4.37 (t, J = 3.7 Hz, 1H), 3.66 - 3.27 (m, 4H), 3.15 (ddd, J = 6.8, 4.0, 2.9 Hz, 1H), 2.89 - 2.78 (m, 1H), 2.61 (td, J = 4.6, 2.3 Hz, 1H), 1.91 (pd, J = 8.9, 8.1, 3.5 Hz, 1H), 1.39 (s, 9H).

To tert-butyl (3R,4R)-3-hydroxy-4-((S)-oxiran-2-yl)pyrrolidine-l-carboxyla te (compound 26d-2) (70 mg) was also isolated . ¾ NMR (400 MHz, Chloroform-d) d 4.45 (q, J = 4.1, 3.2 Hz, 1H), 3.65 - 3.55 (m, 1H), 3.54 - 3.31 (m, 3H), 3.21 (b, 1H), 2.83 (m,M), 2.57 (b, 1H), 2.01 (b, 1H), 1.47 (s, 9H).

Step 26e

To tert-butyl (3R,4R)-3-hydroxy-4-(oxiran-2-yl)pyrrolidine-l-carboxylate (26d-l) (12 mg, 0.052 mmol) in DCM (1 ml) at -78 °C was added BF3 diethyl etherate (0.013 ml, 0.105 mmol) and the mixture was stirred for lh then warmed up to 0 °C and quenched with MeOH. The mixture was concentrated under vacuo. To the residue was added methyl 2-bromo-4- isopropoxybenzo-[d]thiazole-6-carboxylate (lb-l) (34.6 mg, 0.105 mmol) and cesium carbonate (51.2 mg, 0.157 mmol) and DMA (1.000 ml). The mixture was stirred at RT for 2 days. The mixture was diluted with EtOAc, washed with water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with 0 to 60% EtO Ac/hexane to give methyl 2-((3aS,6aR)-3-hydroxyhexahydro-5H-furo[2,3-c]pyrrol-5-yl)-4 - isopropoxybenzo [d]thiazole-6-carboxylate (26e) (3.5 mg). LC/MS observed [M+H], 379.11; ¾ NMR (400 MHz, Chloroform-d) d 7.97 (d, J = 1.6 Hz, 1H), 7.53 (d, J = 1.6 Hz, 1H), 4.85 (p, J = 6.1 Hz, 1H), 4.74 (ddd, J = 7.1, 5.4, 1.5 Hz, 1H), 4.52 (d, J = 7.9 Hz, 1H), 4. l8 (dd, J = 11.3, 3.5 Hz, 1H), 3.95 (dd, J = 12.0, 1.5 Hz, 1H), 3.93 - 3.78 (m, 2H), 3.90 (s, 3H), 3.66 (dd,

J = 12.0, 5.4 Hz, 1H), 3.56 (dd, J = 11.2, 8.6 Hz, 1H), 3.09 (dtd, J = 8.5, 6.9, 3.5 Hz, 1H), 1.52 - 1.34 (m, 6H).

Step 26f

To methyl 2-((3R,3aS,6aR)-3-hydroxyhexahydro-5H-furo[2,3-c]pyrrol-5-yl )-4- isopropoxybenzo [d]thiazole-6-carboxylate (26e) (5.3 mg, 0.014 mmol) and 4- (chloromethyl)-5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazole (la-l) (8.48 mg, 0.028 mmol) in DMSO (1 ml) was added potassium tert-butoxide (2.357 mg, 0.021 mmol) and the mixture was stirred at RT for l.5h. The mixture was qunched with 1N HC1 at 0 °C and then partitioned between EtOAc and water. The organic layer was separated, washed with water, brine, dried, filtered and concentrated. The residue was purified by HPLC eluting with 0.1% formic acid in acetonitrile/water to give methyl 2-((3R,3aR,6aR)-3-((5-cyclopropyl-3-(2,6- dichlorophenyl)-isoxazol-4-yl)methoxy)hexahydro-5H-furo[2,3- c]pyrrol-5-yl)-4- isopropoxybenzo[d]thiazole-6-carboxylate (26f) (2.3 mg, 25.5 % yield). LC/MS observed [M+H], 644.15; ¾ NMR (500 MHz, Chloroform-d) d 7.90 (d, J = 1.5 Hz, 1H), 7.50 - 7.40 (m, 3H), 7.30 (t, J = 8.2 Hz, 1H), 4.78 (h, J = 6.1 Hz, 1H), 4.65 - 4.49 (m, 1H), 4.24 (s, 2H), 4.15 (q, J = 6.6 Hz, 1H), 3.84 (s, 3H), 3.79 (dd, J = 9.2, 6.4 Hz, 2H), 3.62 (dd, J = 10.9, 6.1 Hz, 2H), 3.56 (dd, J = 9.2, 6.4 Hz, 1H), 3.40 (t, J = 10.3 Hz, 1H), 2.96 (dq, J = 9.2, 6.4 Hz, 1H), 2.04 (tt, J = 8.3, 5.1 Hz, 1H), 1.38 (dd, J = 6.1, 4.8 Hz, 6H), 1.28 - 1.14 (m, 2H), 1.16 -

1.00 (m, 2H)

Example 26 was synthesized following a similar experimental procedure as in Step 5e.

LC/MS observed [M+H], 630.13.

Example 27

Step 27 a

To 4-(chloromethyl)-5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazo le (la-l) (147 mg, 0.486 mmol) and racemic tert-butyl 4-aminohexahydrocyclopenta[c]pyrrole-2(lH)-carboxylate ((±)-27a-l) (100 mg, 0.442 mmol) in acetonitrile (3 ml) was added cesium carbonate (288 mg, 0.884 mmol) and the mixture was stirred at RT for l6h and at 50 °C for 6h. To the mixture was added sodium iodide (33.1 mg, 0.221 mmol) and the resulted mixture was stirred at 50 °C for l6h. The mixture was diluted with EtOAc, washed with water, brine, dried, filtered and concentrated. The residue was purified byl CombiFlash eluting with 0 to 70%

EtO Ac/hexane to give racemic tert-butyl (3aS,4R,6aR)-4-(((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methyl)amino)hexa-hydrocyclopen ta[c]pyrrole-2(lH)- carboxylate ((±)-27a) (150 mg, 0.305 mmol, 68.9 % yield). LC/MS observed [M+H], 492.17; ¾ NMR (400 MHz, Chloroform-ri) d 7.57 - 7.30 (m, 3H), 3.59-3.33 (m, 4H), 3.07 (b, 2H), 2.80 (b, 1H), 2.59 (b,lH), 2.33 - 2.06 (m, 2H), 1.97 - 1.78 (m, 2H), 1.45 (s, 9H), 1.41 - 1.21 (m, 3H), 1.21 - 1.05 (m, 2H). Step 27b

To tert-butyl (3aS,4R,6aR)-4-(((5-cyclopropyl-3-(2,6-dichlorophenyl)isoxaz ol-4- yl)methyl)amino)hexahydrocyclopenta[c]pyrrole-2(lH)-carboxyl ate ((±)-27a ) (75 mg, 0.152 mmol) in CH2CI2 (1.5 ml) was added TFA (1.5 ml), and the mixture was stirred at RT for 2h. The mixture was concentrated, and chased with DCM. To the residue was added methyl 2- bromo-4-isopropoxybenzo[d]thiazole-6-carboxylate (75 mg, 0.228 mmol), cesium carbonate (298 mg, 0.914 mmol) and DMA (3 ml). The mixture was stirred at RT for l6h and then diluted with EtO Ac/Water. The organic layer was separated, washed with water, brine, dried, filtered and concentrated. The residue was purified by CombiFlash eluting with 0 to 70%

EtO Ac/Hexane to give methyl 2-((3aS,4R,6aR)-4-(((5-cyclopropyl-3-(2,6- dichlorophenyl)isoxazol-4-yl)methyl)amino)hexahydrocyclopent a[c]pyrrol-2(lH)-yl)-4- isopropoxybenzo[d]thiazole-6-carboxylate ((±)-27b) (65 mg, 0.101 mmol, 66.5 % yield). LC/MS observed [M+H], 641.19; ¾ NMR (500 MHz, Chloroform-ri) d 7.88 (d, J= 1.4 Hz, 1H), 7.44 (d, J= 1.5 Hz, 1H), 7.41 - 7.33 (m, 2H), 7.29 (t, J= 8.1 Hz, 1H), 4.77 (hept, J =

6.2 Hz, 1H), 3.83 (s, 3H), 3.62 (dt, J= 11.0, 8.3 Hz, 2H), 3.50 (d , J= 13.7 Hz, 1H), 3.42 (d, J = 13.6 Hz, 1H), 3.34 - 3.21 (m, 2H), 2.81 (q, J= 5.5 Hz, 1H), 2.74 (qt, J= 8.5, 4.8 Hz, 1H), 2.33 (tt, J= 8.8, 4.6 Hz, 1H), 2.15 - 2.03 (m, 1H), 1.94 - 1.78 (m, 2H), 1.37 (dd, .7= 6.1, 1.5 Hz, 6H), 1.32 - 1.23 (m, 1H), 1.23 - 1.16 (m, 3H), 1.06 (tt, J= 10.5, 6.6 Hz, 2H).

Step 27 c

To methyl 2-((3aS,4R,6aR)-4-(((5-cyclopropyl-3-(2,6-dichlorophenyl)iso xazol-4- yl)methyl)amino)hexahydrocyclopenta[c]pyrrol-2(lH)-yl)-4-iso propoxybenzo[d]thiazole-6- carboxylate ((±)-27b) (65 mg, 0.101 mmol) in tetrahydrofuran (1.000 ml) and MeOH (1 ml) was added NaOH (0.152 ml, 0.152 mmol, 1N in water) and the mixture was stirred at RT for l6h. Another portion ofNaOH (0.152 ml, 0.152 mmol, 1N) was added and the mixture was stirred at 50 °C for 6h. The mixture was acidified with 1N HC1, and diluted with

EtO Ac/water, the organic layer was separated and washed with water, brine, dried, filtered and concentrated to give (±)-Example 27 (53 mg, 0.084 mmol, 83 % yield). LC/MS observed [M+H], 627.17; ¾ NMR (500 MHz, Chloroform-ri) d 9.87 (s, 1H), 7.84 (s, 1H), 7.52 - 7.24 (m, 4H), 5.00 (s, 1H), 4.68 (p, J= 6.2 Hz, 1H), 3.81 - 3.50 (m, 3H), 3.50 - 3.18 (m, 3H), 3.18 - 3.00 (m, 1H), 3.00 - 2.66 (m, 2H), 1.31 (dd, = 24.7, 6.0 Hz, 6H), 1.23 - 1.01 (m, 4H).

Example 28

To a flask containing 5-cyclopropyl-3-(2,6-dichlorophenyl)isoxazole-4-carbaldehyde (317 mg, 1.124 mmol) and tert-butyl (3aR,5r,6aS)-5-aminohexahydrocyclopenta[c]pyrrole-

2(lH)-carboxylate (242 mg, 1.067 mmol), was added 2,2,2-trifluoroethan-l-ol (1.3 ml, 18.61 mmol). The suspension was heated up to 50 °C for 30 min to form a light yellow solution. Then it was cooled to 45°C. Sodium tetrahydroborate (powder) (51 mg, 1.348 mmol) was added. Followed by adding 2,2,2-trifluoroethan-l-ol (1 ml) to wash NaBFE on the flask wall into the solution. The mixture was stirred at 45 °C for 16 h. The mixture was cooled down and concentrated under vacuum. The residue was precipitated in EtO Ac (50 ml) and water (15 ml), organic layer was separated. The aqueous layer was extracted by EtO Ac. The combined organic layers were washed with 10% aqueous potassium sodium tartrate solution and brine. The organic layer was dried and concentrated to give crude product. The crude product was purified by combiflash (12 g silica gel, 0-100% EtO Ac in hexane) to give product 28a (248 mg) as a colorless oil. LCMS: 492.19 (M+l); ¾ NMR (400 MHz, Chloroform-ri) d 7.41 - 7.36 (m, 2H), 7.34 - 7.29 (m, 1H), 3.46 (s, 2H), 3.43 - 3.33 (m, 2H), 3.18 - 3.06 (m, 2H), 3.06 - 2.95 (m, 1H), 2.52 - 2.39 (m, 2H), 2.17 - 2.07 (m, 1H), 2.03 - 1.93 (m, 2H), 1.42 (s, 9H), 1.24 - 1.17 (m, 2H), 1.10 - 1.04 (m, 2H), 1.04 - 0.95 (m, 2H).

To a solution of compound 28a (152 mg, 0.309 mmol) in DCM (1.5 ml) at 0 °C, was added TFA (0.76 ml, 9.88 mmol) dropwise. The mixture was stirred at 0 °C for 2h. The solvent was removed by rotovap. The trace amount of TFA was removed by adding DCM (3 times) to the crude mixture and then removing by rotovap three times. The crude product (175 mg, 0.309 mmol) is a pale yellow oil. The crude product (28b) was used directly in next steps without purification. LCMS: 392.14 (M+l).

To a vial containing the above crude oil (28b, 0.154 mmol), methyl 2-bromo-4- isopropoxybenzo[d]thiazole-6-carboxylate (55.9 mg, 0.169 mmol) and cesium carbonate (201 mg, 0.616 mmol), was added N,N-dimethylacetamide (1.5 ml). The mixture was stirred at 70 °C for 20h. The solvent was removed by N2 blowing. The mixture was treated with DCM. The resulting slurry was filtered. The filtrate was loaded into a silica cartridge and purified by combiflash (8 silica gel, 0-100% EtOAc in hex) to give the product (28c) (103 mg) as a white solid. LCMS: 641.17 (M+l); ¾ NMR (400 MHz, Chloroform-r ) d 7.94 (d, J= 1.2 Hz, 1H), 7.49 (d, J= 1.2 Hz, 1H), 7.39 - 7.34 (m, 2H), 7.31 - 7.25 (m, 1H), 4.82 (hept, J= 6.0 Hz,

1H), 3.88 (s, 3H), 3.73 - 3.65 (m, 2H), 3.50 - 3.39 (m, 4H), 3.16 - 3.05 (m, 1H), 2.75 - 2.64 (m, 2H), 2.14 - 2.03 (m, 3H), 1.42 (d, J= 6.0 Hz, 6H), 1.21 - 1.17 (m, 2H), 1.17 - 1.10 (m,

2H), 1.08 - 1.00 (m, 2H).

To a solution of the methyl ester above (28c; 103 mg, 0.161 mmol) in MeOH (1.6 ml) and THF (3.3 ml), was added aqueous lithium hydroxide (1M) (1.6 ml, 1.6 mmol). The resulting solution was stirred at rt for l6h. Most of solvent was removed. Water (3 ml) and EtO Ac (10 ml) were added, Added 1M HC1 was added to pH=5. Two clear layers were formed, the organic layer was separated, and the aqueous layer was extracted with EtO Ac (2x10 ml). The combined organic layers were washed with brine, dried, filtered, and concentrated to give the crude product (103 mg) as a syrup. The crude product was treated with MeOH (3 ml). The mixture was stirred at rt for 10 min to form a white slurry, The slurry was filtered to give the pure example 28 as a white solid (26 mg). LCMS: 627.16 (M+l); 'H NMR (400 MHz, DMSO-rie) d 7.99 (d, J= 1.5 Hz, 1H), 7.68 - 7.43 (m, 3H), 7.37 (d, J= 1.6 Hz, 1H), 4.86 (hept, J= 6.0 Hz, 1H), 3.68 - 3.57 (m, 2H), 3.42 - 3.29 (m, 4H), 3.10 - 2.92 (m, 1H), 2.74 - 2.62 (m, 2H), 2.39 - 2.26 (m, 1H), 2.03 - 1.92 (m, 2H), 1.31 (d, J= 6.0 Hz, 6H), 1.20 - 0.95 (m, 6H).

Example 29

To a mixture of Example 20 (65 mg, 0.101 mmol) and sodium bicarbonate (85 mg, 1.01 mmol) in THF (3 ml)/Water (1.2 ml) was added iodine (192 mg, 0.757 mmol) at rt. The reaction mixture was stirred at rt for 4h, quenched with brine/lN HC1 (pH 4), and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over Na 2 S04, and concentrated. The residue was purified by chromatography on silica gel using hexane/acetone (100/0 to 30/70, 15 min) to give Example 29 as a brown solid. LC-MS: M+H = 657.12, calcd. 657.13; ¾ NMR (400 MHz, DMSO-rie) d 12.77 (s, 1H), 8.03 (t, J= 1.3 Hz, 1H), 7.63 (ddt, = 8.0, 6.8, 1.2 Hz, 2H), 7.55 (td, .7= 8.1, 1.0 Hz, 1H), 7.40 (d , J= 1.4 Hz, 1H), 4.84 (p , J= 6.0 Hz, 1H), 4.57-4.50 (m, 2H), 4.16 (d, J= 12.4 Hz, 1H), 4.08 (d, J= 12.8

Hz, 1H), 3.95 - 3.84 (m, 2H), 3.63-3.57 (m, 1H), 3.14 - 3.03 (m, 1H), 2.97 - 2.86 (m, 2H), 2.38 (dd, J= 8.9, 4.3 Hz, 1H), 1.99 - 1.84 (m, 1H), 1.80 (ddd, J= 14.0, 7.3, 3.3 Hz, 1H), 1.32 (d, J = 6.0 Hz, 6H), 1.17 (s, 2H), 1.20 - 1.07 (m, 2H).

Example 30

Example 30 was synthesized by following the similar experimental procedure as for Example 29. Example 30: LC-MS M+H = 257.12, calcd. 257.13; ¾ NMR (400 MHz, DMSO-rie) d 12.79 (s, 1H), 8.02 (d , J= 1.5 Hz, 1H), 7.63 (ddd, J= 8.2, 6.7, 1.3 Hz, 2H), 7.62 - 7.47 (m, 1H), 7.40 (d, J= 1.6 Hz, 1H), 4.84 (p, J= 6.0 Hz, 1H), 4.56 (d, J= 12.2 Hz, 1H),

4.50 (d, J= 12.2 Hz, 1H), 4.16 (d, J= 12.5 Hz, 1H), 4.08 (d, J= 12.9 Hz, 1H), 3.89 (ddd, J = 18.9, 9.9, 3.2 Hz, 2H), 3.60 (h, J= 6.0 Hz, 1H), 3.08 (td, J= 12.6, 3.6 Hz, 1H), 2.91 (td, J = 12.5, 12.1, 4.1 Hz, 2H), 2.38 (tt, J= 8.2, 5.2 Hz, 1H), 1.95 - 1.74 (m, 2H), 1.32 (d, .7= 6.0 Hz, 6H), 1.22 - 1.04 (m, 4H). Example 31

Example 31 was synthesized by following the similar experimental procedure as for Example 29. Example 31: LC-MS M+H = 257.12, calcd. 257.13; ¾ NMR (400 MHz, DMSO-rie) d 12.79 (s, 1H), 7.99 (d, J= 11.6 Hz, 1H), 7.69 - 7.62 (m, 2H), 7.62 - 7.53 (m, 1H), 7.42 - 7.35 (m, 1H), 4.83 (t, .7= 6.2 Hz, 1H), 4.68 (d , J= 12.2 Hz, 1H), 4.53 (d, J= 12.1 Hz, 1H), 4.20 (d, J= 15.1 Hz, 1H), 4.14 (s, 1H), 4.11 - 3.98 (m, 2H), 3.87 (d, J = 12.0 Hz, 1H), 3.57-3.50 (m, 2H), 3.06-3.02 (m, 1H), 2.93 - 2.79 (m, 2H), 2.43 - 2.35 (m, 1H), 1.32 (d, = 5.9 Hz, 6H), 1.19 - 1.10 (m, 4H).

ASSAYS

Human FXR (NR1H4) Assay

Determination of a ligand mediated Ga!4 promoter driven transactivation to quantify ligand binding mediated activation of FXR. FXR Reporter Assay kit purchased from Indigo Bioscience (Catalogue number: IB00601) to determine the potency and efficacy of compound developed by Enanta that can induce FXR activation. The principle application of this reporter assay system is to quantify functional activity of human FXR. The assay utilizes non-human mammalian ceils, CHO (Chinese hamster ovary) cells engineered to express human NR1H4 protein (referred to as FXR). Reporter cells also incorporate the cDNA encoding beetle luciferase which catalyzes the substrates and yields photon emission.

Luminescence intensity of the reaction is quantified using a plate-reading luminometer, Envision. Reporter Cells include the luciferase reporter gene functionally linked to an FXR responsive promoter. Thus, quantifying changes in luciferase expression in the treated reporter cells provides a sensitive surrogate measure of the changes in FXR activity. ECso and efficacy (normalize to CDCA set as 100%) is determined by XLFit. The assay is according to the manufacturer’s instructions. In brief, the assay was performed in white, 96 well plates using final volume of lOOul containing cells with different doses of compounds. Retrieve Reporter Cells from -80°C storage. Perform a rapid thaw of the frozen cells by transferring a 10 ml volume of 37°C cell recovery medium into the tube of frozen cells. Recap the tube of Reporter Cells and immediately place it in a 37°C water bath for 5 - 10 minutes. Retrieve the tube of Reporter Cell Suspension from the water bath. Sanitize the outside surface of the tube with a 70% alcohol swab, and then transfer it into the cell culture hood. Dispense 90 pl of cell suspension into each well of the 96-well Assay Plate. Transfer the plate into 37°C incubator, allowing the cells adherent to the bottom of the well. Dilute compounds in Dilution Plate (DP), and administrate to cells at Assay Plate (AP). DMSO content of the samples was kept at 0.2%. Cells were incubated for additional 22 hours before luciferase activities were measured. Thirty minutes before intending to quantify FXR activity, remove Detection Substrate and Detection Buffer from the refrigerator and place them in a low-light area so that they may equilibrate to room temperature. Remove the plate’s lid and discard all media contents by ejecting it into an appropriate waste container. Gently tap the inverted plate onto a clean absorbent paper towel to remove residual droplets. Cells will remain tightly adhered to well bottoms. Add 100 mΐ of luciferase detection reagent to each well of the assay plate. Allow the assay plate to rest at room temperature for at least 5 minutes following the addition of LDR. Set the instrument (Envision) to perform a single 5 second“plate shake” prior to reading the first assay well. Read time may be 0.5 second (500mSec) per well. ECso and Efficacy (normalize to CDCA set as 100%) is determined by XLFit.

To assess the FXR agonistic potency of the example compounds as well as for reference compound, potency ranges were determined in the Human FXR (NR1H4) Assay as listed below in Table 9. The efficacy was normalized to CDCA set as 100%. (A = EC50 < 0.030 mM; B = 0.030 mM < EC50 < 0.2 mM; C = 0.2mM < EC50 < 1.0 mM; D= EC50 > 1.0 mM)

Table 9

While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.