Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DIHYDROPYRAZOLE GPR40 MODULATORS
Document Type and Number:
WIPO Patent Application WO/2014/078610
Kind Code:
A1
Abstract:
The present invention provides compounds of Formula (I): or a stereoisomer, or a pharmaceutically acceptable salt thereof, wherein all of the variables are as defined herein. These compounds are GPR40 G protein-coupled receptor modulators which may be used as medicaments.

Inventors:
ELLSWORTH BRUCE A (US)
SHI JUN (US)
EWING WILLIAM R (US)
JURICA ELIZABETH A (US)
HERNANDEZ ANDRES S (US)
WU XIMAO (US)
Application Number:
PCT/US2013/070215
Publication Date:
May 22, 2014
Filing Date:
November 15, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BRISTOL MYERS SQUIBB CO (US)
International Classes:
C07D401/12; A61K31/4155; A61P3/10; C07D401/14
Domestic Patent References:
WO2012068529A22012-05-24
WO2009115515A12009-09-24
WO2005012249A22005-02-10
Foreign References:
US20090286758A12009-11-19
EP1743894A12007-01-17
EP1637522A12006-03-22
US20130143843A12013-06-06
US5614492A1997-03-25
Other References:
SHIMPUKADE, B. ET AL., J. MED. CHEM., vol. 55, no. 9, 2012, pages 4511 - 4515
BARLIND, J.G. ET AL., BIOORG. MED. CHEM. LETT., vol. 23, no. 9, 2013, pages 2721 - 2726
"Hawley's Condensed Chemical Dictionary", 1997, JOHN WILEY & SONS, INC.
GREENE, T.W. ET AL.: "Protecting Groups in Organic Synthesis", 2007, WILEY
"The Peptides: Analysis, Synthesis, Biology", vol. 3, 1981, ACADEMIC PRESS
REMINGTON: "The Science and Practice of Pharmacy", 2012, PHARMACEUTICAL PRESS
"Design ofProdrugs", 1985, ELSEVIER
"Methods in Enzymology", vol. 112, 1985, ACADEMIC PRESS, pages: 309 - 396
BUNDGAARD, H. ET AL.: "A Textbook of Drug Design and Development", 1991, HARWOOD ACADEMIC PUBLISHERS, article "Design and Application of Prodrugs", pages: 113 - 191
BUNDGAARD, H., ADV. DRUG DELIV. REV., vol. 8, 1992, pages 1 - 38
BUNDGAARD, H. ET AL., J. PHARM. SCI., vol. 77, 1988, pages 285
KAKEYA, N. ET AL., CHEM. PHARM. BULL., vol. 32, 1984, pages 692
"Prodrugs and Targeted Delivery (Methods and Principles in Medicinal Chemistry", vol. 47, 2011, WILEY-VCH
"Medicinal Chemistry: Principles and Practice", 2006, THE ROYAL SOCIETY OF CHEMISTRY
TESTA, B. ET AL.: "Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology", 2003, VCHA AND WILEY-VCH
"The Practice of Medicinal Chemistry", 2008, ACADEMIC PRESS
SMITH, D.K.: "A FORTRAN Program for Calculating X-ray Powder Diffraction Patterns", April 1963, LAWRENCE RADIATION LABORATORY
"Comprehensive Organic Synthesis: Selectivity, Strategy & Efficiency in Modern Organic Chemistry", 1991, PERGAMON PRESS
SMITH, M.B. ET AL.: "March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2007, WILEY & SONS
"Comprehensive Organic Functional Groups Transformations 11", 2004, ELSEVIER SCIENCE INC.
LAROCK, R.C.: "Comprehensive Organic Transformations", 1999, VCH PUBLISHERS, INC.
"Comprehensive Heterocyclic Chemistry", 1996, PERGAMON PRESS INC.
SIBI, M.P. ET AL., ORGANIC LETTERS, vol. 11, no. 23, 2009, pages 5366
SIBI, M.P. ET AL., J. AM. CHEM. SOC., vol. 127, no. 23, 2005, pages 8276
MANYEM, S. ET AL., J. COMB. CHEM., vol. 9, 2007, pages 20
GARANTI, L. ET AL., TETRAHEDRON: ASYMMETRY, vol. 13, 2002, pages 1285
MOLTENI, G., TETRAHEDRON: ASYMMETRY, vol. 15, 2004, pages 1077
BENASSUTI, L.D. ET AL., TETRAHEDRON, vol. 60, 2004, pages 4627
SHIMIZU, T. ET AL., BULL. CHEM. SOC. JPN., vol. 57, 1984, pages 787
TORTOLANI, D.R. ET AL., ORGANIC LETTERS, vol. 1, no. 8, 1999, pages 1261 - 1263
ITOH ET AL., NATURE, vol. 422, 2003, pages 173 - 176
TAN ET AL., DIABETES, vol. 57, 2008, pages 2211 - 2219
EDFALK ET AL., DIABETES, vol. 57, 2008, pages 2280 - 2287
LUO ET AL., PLOSONE, vol. 7, 2012, pages 1 - 12
YAMASHIMA, T., PROGRESS IN NEUROBIOLOGY, vol. 84, 2008, pages 105 - 115
SIMPKINS, L. ET AL., BIOORGANIC MEDICINAL CHEMISTRY LETTERS, vol. 17, no. 23, 2007, pages 6476 - 6480
FORD ET AL., J. AM. MED. ASSOC., vol. 287, 2002, pages 356 - 359
ARBEENY ET AL., CURR. MED. CHEM. - IMM., ENDOC. & METAB. AGENTS, vol. 1, 2001, pages 1 - 24
ALLEN, L.V., JR. ET AL.: "Remington: The Science and Practice ofPharmacy", vol. 2, 2012, PHARMACEUTICAL PRESS
FYFE, M.C.T. ET AL., DRUGS OF THE FUTURE, vol. 34, no. 8, 2009, pages 641 - 653
MOHLER, M.L., MEDICINAL RESEARCH REVIEWS, vol. 29, no. L, 2009, pages 125 - 195
MIZUNO, C.S. ET AL., CURRENT MEDICINAL CHEMISTRY, vol. 15, 2008, pages 61 - 74
MELNIKOVA, I. ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 5, 2006, pages 369 - 370
JONES, D., NATURE REVIEWS: DRUG DISCOVERY, vol. 8, 2009, pages 833 - 834
OBICI, S., ENDOCRINOLOGY, vol. 150, no. 6, 2009, pages 2512 - 2517
ELANGBAM, C.S., VET. PATHOL., vol. 46, no. 1, 2009, pages 10 - 24
STOUT ET AL.: "X-ray Structure Determination: A Practical Guide", 1968, MACMILLIAN
Attorney, Agent or Firm:
SUN, Jing G. et al. (P.O. Box 4000Princeton, New Jersey, US)
Download PDF:
Claims:
What is claimed is:

1. A compound of Formula I):

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

R1 is independently phenyl substituted with 0-3 R6 or pyridinyl substituted with

0-3 R6;

R2 is, at each occurrence, independently halogen or alkyl;

R3 is independently selected from: CF3, 4-halo-Ph, 4-CN-Ph,

4-C02(C1_2 alkyl)-Ph, 2-halo-4-CN-Ph, and pyrimidin-2-yl;

R4 is independently alkyl or cyclopropylmethyl;

R5 is, at each occurrence, independently halogen; and

R6, at each occurrence, is independently selected from: OH, halogen, CN, alkyl, and alkoxy.

2. A compound according to claim 1, wherein the compound is of Formula (II):

or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof. 3. A compound according to claim 1 or claim 2, wherein: R1 is independently phenyl substituted with 0-3 R6 or pyridinyl substituted with

0-2 R6;

R2 is independently halogen or C 1.4 alkyl;

R3 is independently selected from: CF3, 4-halo-Ph, 4-CN-Ph,

4-C02(C1_2 alkyl)-Ph, 2-halo-4-CN-Ph, and pyrimidin-2-yl;

R4 is independently alkyl or cyclopropylmethyl; and

R6, at each occurrence, is independently selected from: halogen, CN, alkyl, and Ci_4 alkoxy. 4. A compound according to any one of claims 1 to 3, wherein the compound is of Formula (III):

or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof. 5. A compound according to any one of claims 1 to 4, wherein:

R2 is independently halogen or alkyl;

R4 is independently alkyl; and

R6, at each occurrence, is independently selected from: halogen, CN, alkyl, and C _4 alkoxy.

6. A compound according to any one of claims 1 to 5, wherein:

R2 is independently alkyl;

and

R6, at each occurrence, is independently selected from: halogen, alkyl, and Ci_4 alkoxy. A compound according to any one of claims 1 to 6, wherein: R2 is methyl;

R4 is methyl; and

R6, at each occurrence, is independently selected from: CI and methoxy. A compound according to claim 1, wherein the compound is selected from:

- 170-

or a stereoisomer, or a pharmaceutically acceptable salt thereof.

- 174-

or a stereoisomer, or a pharmaceutically acceptable salt thereof.

11. A pharmaceutical composition, comprising: a pharmaceutically acceptable and a compound of any one of claims 1 to 10, or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof.

12. The pharmaceutical composition according to claim 1 1, further comprising one or more other additional therapeutic agents: anti-diabetic agents, anti-hyperglycemic agents, anti-hyperinsulinemic agents, anti-retinopathic agents, anti-neuropathic agents, anti-nephropathic agents, anti-atherosclerotic agents, anti-ischemic agents,

anti-hypertensive agents, anti-obesity agents, anti-dyslipidemic agents,

anti-hyperlipidemic agents, anti-hypertriglyceridemic agents, anti-hypercholesterolemic agents, anti-restenotic agents, anti-pancreatic agents, lipid lowering agents, anorectic agents, memory enhancing agents, anti-dementia agents, cognition promoting agents, appetite suppressants, treatments for heart failure, treatments for peripheral arterial disease and anti-inflammatory agents.

13. The pharmaceutical composition according to claim 1 1, further comprising a dipeptidyl peptidase-IV inhibitor and/or a sodium-glucose transporter-2 inhibitor. 14. A compound of any one of claims 1 to 10 for use in preventing, modulating or treating diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, retinopathy, neuropathy, nephropathy, diabetic kidney disease, acute kidney injury, cardiorenal syndrome, acute coronary syndrome, delayed wound healing, atherosclerosis and its sequelae, abnormal heart function, congestive heart failure, myocardial ischemia, stroke, Metabolic Syndrome, hypertension, obesity, fatty liver disease, dislipidemia, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low high-density lipoprotein, high low-density lipoprotein, non- cardiac ischemia, pancreatitis, lipid disorders, neurodegenerative disease, cognitive impairment, dementia, liver diseases and liver cirrhosis.

15. A compound for use according to claim 14, wherein the compound of any one of claims 1 to 10 is used simultaneously, separately or sequentially with an additional therapeutic agent.

Description:
DIHYDROPYRAZOLE GPR40 MODULATORS

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority under 35 U.S. C. § 119(e) to U.S. provisional patent Application No. 61/727,262, filed November 16, 2012, and U.S. provisional patent Application No. 61/777,294, filed March 12, 2013; the entire contents of these applications are incorporated herein by reference.

FIELD OF THE INVENTION

[0002] The present invention provides novel carboxylic acid substituted

dihydropyrazole compounds, and their analogues thereof, which are GPR40 G protein- coupled receptor modulators, compositions containing them, and methods of using them, for example, for the treatment or prophylaxis of diabetes and related conditions.

BACKGROUND OF THE INVENTION

[0003] Diabetes mellitus is a progressively debilitating disorder of epidemic proportions leading to various micro- and macrovascular complications and morbidity. The most common type of diabetes, type 2 diabetes, is characterized by increasing insulin resistance associated with inadequate insulin secretion after a period of compensatory hyperinsulinemia. Free fatty acids (FFAs) are evidenced to influence insulin secretion from β cells primarily by enhancing glucose-stimulated insulin secretion (GSIS).

G-protein coupled receptors (GPCRs) expressed in β cells are known to modulate the release of insulin in response to changes in plasma glucose levels. GPR40, also known as fatty acid receptor 1 (FFARl), is a membrane-bound FFA receptor which is preferentially expressed in the pancreatic islets and specifically in β cells and mediates medium to long chain fatty acid induced insulin secretion. GPR40 is also expressed in enteroendocrine cells wherein activation promotes the secretion of gut incretin hormones, such as GLP-1, GIP, CCK and PYY. To decrease medical burden of type 2 diabetes through enhanced glycemic control, GPR40 modulator compounds hold the promise of exerting an incretin effect to promote GSIS as well as potential combination with a broad range of antidiabetic drugs. [0004] The present invention relates to novel substituted dihydropyrazole compounds which have the ability to modulate GPR40. Such compounds are therefore potentially useful for the treatment or prophylaxis of diabetes and related conditions. SUMMARY OF THE INVENTION

[0005] The present invention provides substituted dihydropyrazole compounds, and their analogues thereof, which are useful as GPR40 modulators, including stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.

[0006] The present invention also provides processes and intermediates for making the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.

[0007] The present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.

[0008] The present invention also provides a crystalline form of one of the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.

[0009] The compounds of the invention may be used in the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40, such as diabetes and related conditions, microvascular complications associated with diabetes, the macrovascular complications associated with diabetes, cardiovascular diseases, Metabolic Syndrome and its component conditions, disorders of glucose metabolism, obesity and other maladies.

[0010] The compounds of the invention may be used in therapy.

[0011] The compounds of the invention may be used for the manufacture of a medicament for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40.

[0012] The compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more other agent(s).

[0013] Other features and advantages of the invention will be apparent from the following detailed description and claims. BRIEF DESCRIPTION OF THE DRAWINGS

[0014] The invention is illustrated by reference to the accompanying drawings described below.

[0015] FIG. 1 shows experimental (at rt) and simulated (at a temperature (T) of about 23 °C) powder X-ray diffraction (PXRD) patterns (CuKa λ= 1.5418 A) of the Form N-1 of 2-((4S,5S)-l-(4-(((3R, 4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid (Example 81, Isomer 2).

[0016] FIG. 2 shows a differential scanning calorimetry (DSC) thermogram of the Form N-1 of Example 81, Isomer 2.

[0017] FIG. 3 shows a thermogravimetric analysis (TGA) thermogram of the Form N-1 of Example 81, Isomer 2.

[0018] FIG. 4 shows moisture-sorption isotherms of the Form N-1 of Example 81, Isomer 2.

[0019] FIG. 5 shows a glucose excursion curve in rats an acute combination study of Example 81, Isomer 2 with BMS DPP4L

[0020] FIG. 6 shows plasma GLP-1 levels upon administration to rats from a combination study of Example 81 , Isomer 2 with BMS DPP4L

DETAILED DESCRIPTION OF THE INVENTION

I. COMPOUNDS OF THE INVENTION

[0021] In a first aspect, the present disclosure provides, inter alia, a compound of Formula (I):

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

R 1 is independently phenyl substituted with 0-3 R 6 or pyridinyl substituted with

0-3 R 6 ;

R 2 is, at each occurrence, independently halogen or alkyl;

R 3 is independently selected from: CF 3 , 4-halo-Ph, 4-CN-Ph,

4-C02(C!.2 alkyl)-Ph, 2-halo-4-CN-Ph, and pyrimidin-2-yl;

R 4 is independently alkyl or cyclopropylmethyl;

R 5 is, at each occurrence, independently halogen; and

R 6 , at each occurrence, is independently selected from: OH, halogen, CN, alkyl, and alkoxy.

[0022] a second aspect, the present disclosure provides a compound of Formula

(II):

or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the first aspect.

[0023] In a third aspect, the present disclosure includes a compound of Formula (I) (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the first or second aspect, wherein:

R 1 is independently phenyl substituted with 0-3 R 6 or pyridinyl substituted with

0-2 R 6 ;

R 2 is independently halogen or C 1.4 alkyl;

R 3 is independently selected from: CF 3 , 4-halo-Ph, 4-CN-Ph,

4-002(0! _2 alkyl)-Ph, 2-halo-4-CN-Ph, and pyrimidin-2-yl; R 4 is independently alkyl or cyclopropylmethyl; and

R 6 , at each occurrence, is independently selected from: halogen, CN, alkyl, and Ci_4 alkoxy.

[0024] In a fourth aspect, the present disclosure provides a compound of Formula (III):

or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the first, second and third aspects.

[0025] In a fifth aspect, the present disclosure includes a compound of Formula (I), (II) or (III), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein:

R 2 is independently halogen or alkyl;

R 4 is independently alkyl; and

R 6 , at each occurrence, is independently selected from: halogen, CN, alkyl, and Ci_4 alkoxy.

[0026] In a sixth aspect, the present disclosure includes a compound of Formula (I), (II) or (III), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein:

R 2 is independently alkyl; and

R 6 , at each occurrence, is independently selected from: halogen, alkyl, and Ci_4 alkoxy. [0027] In a seventh aspect, the present disclosure includes a compound of Formula (III), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein:

R 2 is methyl;

R 4 is methyl; and

R 6 , at each occurrence, is independently selected from: CI and methoxy.

[0028] In a seventh aspect, the present disclosure includes a compound selected from the exemplified examples or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

[0029] In another aspect, the present disclosure includes a compound selected from any subset list of compounds or a single compound from the exemplified examples within the scope of any of the above aspects.

[0030] In an eighth aspect, the present disclosure includes a compound selected from:

-7 -

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

[0031] In a ninth aspect, the present disclosure includes a compound selected from:

[0032] In a tenth aspect, the present disclosure includes a compound selected from:

[0033] In another aspect, the present disclosure provides or a stereoisomer, or a pharmaceutically acceptable salt, or a solvate thereof.

[0034] In another aspect, the present disclosure provides or a stereoisomer, or a pharmaceutically acceptable salt thereof.

[0035] In another aspect, present disclosure provides or a pharmaceutically acceptable salt thereof.

[0036] In another aspect, the present disclosure provides a crystalline form of or a pharmaceutically acceptable salt thereof. 0037] In another aspect, the present disclosure provides the N-l Form of

xample 81, Isomer 2).

[0038] In another aspect, the present disclosure provides the N- 1 Form of Example 81 , Isomer 2 which is characterized by unit cell parameters substantially equal to the following:

Cell dimensions:

a = 10.1890(3) A

b = 13.4473(6) A

c = 18.8524(7) A

a = 90°

^ = 90°

y = 90°

Space group: Ρΐγλγλγ

Molecules/asymmetric unit: 1

Density (calculated) = 1.391 g/cm 3

wherein said crystal is at a temperature of about 23 °C.

[0039] In another aspect, the present disclosure provides the N- 1 Form of Example 81 , Isomer 2 which is characterized by a powder X-ray diffraction pattern substantially in accordance with that shown in FIG. 1.

[0040] In another aspect, the present disclosure provides the N-l Form of Example 81, Isomer 2 which is characterized by a powder X-ray diffraction pattern comprising four or more 2Θ values selected from: 9.9 ± 0.1, 10.9 ± 0.1, 11.9 ± 0.1, 12.8 ± 0.1, 14.4 ± 0.1, 17.4 ± 0.1, 18.4 ± 0.1, 20.4 ± 0.1, 21.4 ± 0.1 and 22.2 ± 0.1, at about room temperature.

[0041] In another aspect, the present disclosure provides the N-l Form of Example 81, Isomer 2 which is characterized by a powder X-ray diffraction pattern comprising six or more 2Θ values selected from: 9.9 ± 0.1, 10.9 ± 0.1, 1 1.9 ± 0.1, 12.8 ± 0.1, 14.4 ± 0.1, 17.4 ± 0.1, 18.4 ± 0.1, 20.4 ± 0.1, 21.4 ± 0.1 and 22.2 ± 0.1, at about room temperature.

[0042] In another aspect, the present disclosure provides the N-l Form of Example 81 , Isomer 2 which is characterized by a powder X-ray diffraction pattern comprising the following 2Θ values 9.9 ± 0.1, 10.9 ± 0.1, 1 1.9 ± 0.1, 12.8 ± 0.1, 14.4 ± 0.1, 17.4 ± 0.1, 18.4 ± 0.1, 20.4 ± 0.1, 21.4 ± 0.1 and 22.2 ± 0.1, at about room temperature.

[0043] In another aspect, the present disclosure provides the N-l Form of Example 81 , Isomer 2 which is characterized by differential scanning calorimetry thermogram substantially in accordance with that shown in FIG. 2, having an endothermic transition above ca. 186 °C.

[0044] In another aspect, the present disclosure provides the N-l Form of Example 81 , Isomer 2 which is characterized by thermal gravimetric analysis thermogram substantially in accordance with that shown in FIG. 3.

[0045] In another aspect, the present disclosure provides the N-l Form of Example 81, Isomer 2 which is characterized by moisture-sorption isotherms substantially in accordance with that shown in FIG. 4.

[0046] In another embodiment, Rl is phenyl substituted with 0-3 R 6 .

[0047] In another embodiment, Rl is phenyl substituted with 0-2 R 6 .

[0048] In another embodiment, Rl is phenyl substituted with 0-1 R 6 .

[0049] In another embodiment, Rl is pyridinyl substituted with 0-2 R 6 .

[0050] In another embodiment, Rl is pyridinyl substituted with 0-1 R 6 .

[0051] In another embodiment, R2 is independently halogen.

[0052] In another embodiment, R2 is independently alkyl.

[0053] In another embodiment, R 3 is CF3.

[0054] In another embodiment, R 3 is independently selected from: 4-halo-Ph, 4-CN-Ph, 4-C0 2 (C 1 _2 alkyl)-Ph, 2-halo-4-CN-Ph, and pyrimidin-2-yl.

[0055] In another embodiment, R 3 is independently selected from: 4-halo-Ph, 4-CN-Ph, 4-C0 2 (C 1 _2 alkyl)-Ph, and 2-halo-4-CN-Ph. [0056] In another embodiment, R 4 is alkyl.

[0057] In another embodiment, R 4 is cyclopropylmethyl. [0058] In another embodiment, R 6 , at each occurrence, is independently selected from: halogen, alkyl, and alkoxy.

[0059] In another embodiment, R 6 , at each occurrence, is independently selected from: halogen and alkyl.

[0060] In another embodiment, R 6 , at each occurrence, is independently selected from: halogen and alkoxy.

[0061] In another embodiment, the compounds of the present invention have hGPR40 EC50 values < 10 μΜ.

[0062] In another embodiment, the compounds of the present invention have hGPR40 EC 50 values < 5 μΜ.

[0063] In another embodiment, the compounds of the present invention have hGPR40 EC50 values < 1 μΜ.

[0064] In another embodiment, the compounds of the present invention have hGPR40 EC 50 values < 0.5 μΜ.

[0065] In another embodiment, the compounds of the present invention have hGPR40 EC50 values < 0.2 μΜ.

[0066] In another embodiment, the compounds of the present invention have hGPR40 EC50 values < 0.1 μΜ. II. OTHER EMBODIMENTS OF THE INVENTION

[0067] In another embodiment, the present invention provides a composition comprising at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

[0068] In another embodiment, the present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

[0069] In another embodiment, the present invention provides a pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

[0070] In another embodiment, the present invention provides a process for making a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.

[0071] In another embodiment, the present invention provides an intermediate for making a compound of the present invention or a stereoisomer, a tautomer, a

pharmaceutically acceptable salt, or a solvate thereof.

[0072] In another embodiment, the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s). Examples of additional therapeutic agent(s), according to the present invention include, but are not limited to, anti-diabetic agents, anti-hyperglycemic agents, anti-hyperinsulinemic agents, anti-retinopathic agents, anti-neuropathic agents, anti-nephropathic agents,

anti-atherosclerotic agents, anti-ischemic agents, anti-hypertensive agents, anti-obesity agents, anti-dyslipidemic agents, anti-hyperlipidemic agents, anti-hypertriglyceridemic agents, anti-hypercholesterolemic agents, anti-restenotic agents, anti-pancreatic agents, lipid lowering agents, anorectic agents, memory enhancing agents, anti-dementia agents, cognition promoting agents, appetite suppressants, treatments for heart failure, treatments for peripheral arterial disease and anti-inflammatory agents.

[0073] In a preferred embodiment, the present invention provides pharmaceutical composition, wherein the additional therapeutic agents are, for example, a dipeptidyl peptidase-IV (DPP4) inhibitor (for example a member selected from saxagliptin, sitagliptin, vildagliptin, linagliptin, alogliptin, and "BMS DPP4i"), and/or a sodium- glucose transporter-2 (SGLT2) inhibitor (for example a member selected from dapagliflozin, canagliflozin, empagliflozin and remagliflozin).

[0074] In a preferred embodiment, the present invention provides pharmaceutical composition, wherein the additional therapeutic agents are, for example, a DPP4 inhibitor (for example a member selected from saxagliptin, sitagliptin, vildagliptin, linagliptin, alogliptin, and "BMS DPP4i").

[0075] In a preferred embodiment, the present invention provides pharmaceutical composition, wherein the additional therapeutic agents are, for example, an SGLT2 inhibitor (for example a member selected from dapagliflozin, canagliflozin, empagliflozin and remagliflozin).

[0076] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0077] Examples of diseases or disorders associated with the activity of the GPR40 that can be prevented, modulated, or treated according to the present invention include, but are not limited to, diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, retinopathy, neuropathy, nephropathy, diabetic kidney disease, acute kidney injury, cardiorenal syndrome, acute coronary syndrome, delayed wound healing, atherosclerosis and its sequelae, abnormal heart function, congestive heart failure, myocardial ischemia, stroke, Metabolic Syndrome, hypertension, obesity, fatty liver disease, dislipidemia, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low high-density lipoprotein (HDL), high low-density lipoprotein (LDL), non-cardiac ischemia, pancreatitis, lipid disorders, neurodegenerative disease, cognitive impairment, dementia, and liver diseases such as NASH (Non-Alcoholic SteatoHepatitis), NAFLD (Non-Alcoholic Fatty Liver Disease) and liver cirrhosis.

[0078] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of diabetes, hyperglycemia, gestational diabetes, obesity, dyslipidemia, hypertension and cognitive impairment, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent. [0079] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of diabetes, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0080] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of hyperglycemia, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0081] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of obesity, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0082] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of dyslipidemia, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0083] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of hypertension, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0084] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of cognitive impairment, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.

[0085] In another embodiment, the present invention provides a compound of the present invention for use in therapy.

[0086] In another embodiment, the present invention provides a compound of the present invention for use in therapy for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40.

[0087] In another embodiment, the present invention also provides the use of a compound of the present invention for the manufacture of a medicament for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40.

[0088] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40, comprising: administering to a patient in need thereof a therapeutically effective amount of a first and second therapeutic agent, wherein the first therapeutic agent is a compound of the present invention. Preferably, the second therapeutic agent, for example, a DPP4 inhibitor (for example a member selected from saxagliptin, sitagliptin, vildagliptin, linagliptin and alogliptin).

[0089] In another embodiment, the present invention provides a combined preparation of a compound of the present invention and additional therapeutic agent(s) for simultaneous, separate or sequential use in therapy.

[0090] In another embodiment, the present invention provides a combined preparation of a compound of the present invention and additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40.

[0091] Where desired, the compound of the present invention may be used in combination with one or more other types of antidiabetic agents and/or one or more other types of therapeutic agents which may be administered orally in the same dosage form, in a separate oral dosage form or by injection. The other type of antidiabetic agent that may be optionally employed in combination with the GPR40 receptor modulator of the present invention may be one, two, three or more antidiabetic agents or antihyperglycemic agents which may be administered orally in the same dosage form, in a separate oral dosage form, or by injection to produce an additional pharmacological benefit. [0092] The antidiabetic agents used in the combination with the GPR40 receptor modulator of the present invention include, but are not limited to, insulin secretagogues or insulin sensitizers, other GPR40 receptor modulators, or other antidiabetic agents. These agents include, but are not limited to, DPP4 inhibitors (for example, sitagliptin, saxagliptin, alogliptin, linagliptin and vildagliptin), biguanides (for example, metformin and phenformin), sulfonyl ureas (for example, glyburide, glimepiride and glipizide), glucosidase inhibitors (for example, acarbose, miglitol), PPARy agonists such as thiazolidinediones (for example, rosiglitazone and pioglitazone), PPAR α/γ dual agonists (for example, muraglitazar, tesaglitazar and aleglitazar), glucokinase activators, GPR1 19 receptor modulators (for example, MBX-2952, PSN821, and APD597), GPR120 receptor modulators (for example, as described in Shimpukade, B. et al, J. Med. Chem.,

55(9):451 1-4515 (2012)), SGLT2 inhibitors (for example, dapagliflozin, canagliflozin, empagliflozin and remagliflozin), MGAT inhibitors (for example, as described in Barlind, J.G. et al, Bioorg. Med. Chem. Lett., 23(9):2721-2726 (2013); or US 2013/0143843 Al), amylin analogs such as pramlintide, and/or insulin.

[0093] The GPR40 receptor modulator of the present invention may also be optionally employed in combination with agents for treating complication of diabetes. These agents include PKC inhibitors and/or AGE inhibitors.

[0094] The GPR40 receptor modulator of the present invention may also be optionally employed in combination with one or more hypophagic and/or weight-loss agents such as diethylpropion, phendimetrazine, phentermine, orlistat, sibutramine, lorcaserin, pramlintide, topiramate, MCHR1 receptor antagonists, oxyntomodulin, naltrexone, Amylin peptide, NPY Y5 receptor modulators, NPY Y2 receptor modulators, NPY Y4 receptor modulators, cetilistat, 5HT2c receptor modulators, and the like. The GPR40 receptor modulator of the present invention may also be employed in combination with an agonist of the glucagon-like peptide- 1 receptor (GLP-1 R), such as exenatide, liraglutide, GLP-1 (1-36) amide, GLP-l(7-36) amide, GLP-l(7-37), which may be administered via injection, intranasal, or by transdermal or buccal devices.

[0095] The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. This invention encompasses all combinations of preferred aspects of the invention noted herein. It is understood that any and all embodiments of the present invention may be taken in conjunction with any other embodiment or embodiments to describe additional embodiments. It is also understood that each individual element of the embodiments is its own independent embodiment. Furthermore, any element of an embodiment is meant to be combined with any and all other elements from any embodiment to describe an additional embodiment.

III. CHEMISTRY

[0096] Throughout the specification and the appended claims, a given chemical formula or name shall encompass all stereo and optical isomers and racemates thereof where such isomers exist. Unless otherwise indicated, all chiral (enantiomeric and diastereomeric) and racemic forms are within the scope of the invention. Many geometric isomers of C=C double bonds, C=N double bonds, ring systems, and the like can also be present in the compounds, and all such stable isomers are contemplated in the present invention. Cis- and trans- (or E- and Z-) geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. The present compounds can be isolated in optically active or racemic forms. Optically active forms may be prepared by resolution of stereoisomeric forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by chromatography or fractional crystallization. Depending on the process conditions the end products of the present invention are obtained either in free (neutral) or salt form. Both the free form and the salts of these end products are within the scope of the invention. If so desired, one form of a compound may be converted into another form. A free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present invention may be separated into the individual isomers. Compounds of the present invention, free form and salts thereof, may exist in multiple tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention. [0097] As used herein, the term "alkyl" or "alkylene" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, "C^ to alkyl" or "C^ alkyl" denotes alkyl having 1 to 6 carbon atoms. Alkyl group can be unsubstituted or substituted with at least one hydrogen being replaced by another chemical group. Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, ?-butyl), and pentyl (e.g., n-pentyl, isopentyl, neopentyl). When "Co alkyl" or "CQ alkylene" is used, it is intended to denote a direct bond.

[0098] "Alkenyl" or "alkenylene" is intended to include hydrocarbon chains of either straight or branched configuration having the specified number of carbon atoms and one or more, preferably one to two, carbon-carbon double bonds that may occur in any stable point along the chain. For example, "C2 to alkenyl" or "C2_6 alkenyl" (or alkenylene), is intended to include C2, C3, C 4 , C 5 , and alkenyl groups. Examples of alkenyl include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3, pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl,

2-methyl-2-propenyl, and 4-methyl-3 -pentenyl.

[0099] The term "alkoxy" or "alkyloxy" refers to an -O-alkyl group. For example, "Ci to alkoxy" or "C^ alkoxy" (or alkyloxy), is intended to include C2, C3, C 4 , C 5 , and alkoxy groups. Example alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), and ?-butoxy. Similarly,

"alkylthio" or "thioalkoxy" represents an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example methyl-S- and ethyl-S-.

[00100] "Halo" or "halogen" includes fluoro, chloro, bromo, and iodo. "Haloalkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogens. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoropropyl, and heptachloropropyl. Examples of haloalkyl also include

"fluoroalkyl" that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with one or more fluorine atoms.

[00101] "Haloalkoxy" or "haloalkyloxy" represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. For example, "C^ haloalkoxy", is intended to include C2, C3, C4, C5, and haloalkoxy groups. Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluorothoxy. Similarly, "haloalkylthio" or

"thiohaloalkoxy" represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example trifluoromethyl-S-, and pentafluoroethyl-S-.

[00102] The term "cycloalkyl" refers to cyclized alkyl groups, including mono-, bi- or poly-cyclic ring systems. For example, "C3 to cycloalkyl" or "C3.6 cycloalkyl" is intended to include C3, C 4 , C 5 , and cycloalkyl groups. Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbornyl. Branched cycloalkyl groups such as 1-methylcyclopropyl and

2-methylcyclopropyl are included in the definition of "cycloalkyl". The term

"cycloalkenyl" refers to cyclized alkenyl groups. C 4 _6 cycloalkenyl is intended to include C 4 , C 5 , and cycloalkenyl groups. Example cycloalkenyl groups include, but are not limited to, cyclobutenyl, cyclopentenyl, and cyclohexenyl.

[00103] As used herein, "carbocycle", "carbocyclyl", or "carbocyclic residue" is intended to mean any stable 3-, 4-, 5-, 6-, 7-, or 8-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12-, or 13-membered bicyclic or tricyclic ring, any of which may be saturated, partially unsaturated, unsaturated or aromatic. Examples of such carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane (decalin), [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, anthracenyl, and tetrahydronaphthyl (tetralin). As shown above, bridged rings are also included in the definition of carbocycle (e.g., [2.2.2]bicyclooctane). Preferred carbocycles, unless otherwise specified, are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, indanyl, and tetrahydronaphthyl. When the term "carbocycle" is used, it is intended to include "aryl". A bridged ring occurs when one or more, preferably one to three, carbon atoms link two non-adjacent carbon atoms. Preferred bridges are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

[00104] As used herein, the term "bicyclic carbocycle" or "bicyclic carbocyclic group" is intended to mean a stable 9- or 10-membered carbocyclic ring system that contains two fused rings and consists of carbon atoms. Of the two fused rings, one ring is a benzo ring fused to a second ring; and the second ring is a 5- or 6-membered carbon ring which is saturated, partially unsaturated, or unsaturated. The bicyclic carbocyclic group may be attached to its pendant group at any carbon atom which results in a stable structure. The bicyclic carbocyclic group described herein may be substituted on any carbon if the resulting compound is stable. Examples of a bicyclic carbocyclic group are, but not limited to, naphthyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, and indanyl.

[00105] "Aryl" groups refer to monocyclic or bicyclic aromatic hydrocarbons, including, for example, phenyl, and naphthyl. Aryl moieties are well known and described, for example, in Hawley's Condensed Chemical Dictionary, 13th Edition, Lewis, R.J., ed., John Wiley & Sons, Inc., New York (1997). "C 6 . 10 aryl" refers to phenyl and naphthyl.

[00106] The term "benzyl", as used herein, refers to a methyl group on which one of the hydrogen atoms is replaced by a phenyl group.

[00107] As used herein, the term "heterocycle", "heterocyclyl", or "heterocyclic group" is intended to mean a stable 3-, 4-, 5-, 6-, or 7-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered polycyclic heterocyclic ring that is saturated, partially unsaturated, or fully unsaturated, and that contains carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O and S; and including any polycyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., N→0 and S(0) p , wherein p is 0, 1 or 2). The nitrogen atom may be substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. A nitrogen in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1. When the term "heterocycle" is used, it is intended to include heteroaryl.

[00108] Examples of heterocycles include, but are not limited to, acridinyl, azetidinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl,

benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl,

2H,6H-l,5,2-dithiazinyl, dihydrofuro[2,3-Z?]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, imidazolopyridinyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isothiazolopyridinyl, isoxazolyl, isoxazolopyridinyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl,

1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolopyridinyl, oxazolidinylperimidinyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolopyridinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2-pyrrolidonyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrazolyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-l,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thiazolopyridinyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, and xanthenyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.

[00109] Examples of 5- to 10-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, triazolyl, benzimidazolyl, lH-indazolyl, benzofuranyl, benzothiofuranyl, benztetrazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, oxindolyl, benzoxazolinyl, benzthiazolyl, benzisothiazolyl, isatinoyl, isoquinolinyl, octahydroisoquinolinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, isoxazolopyridinyl, quinazolinyl, quinolinyl, isothiazolopyridinyl, thiazolopyridinyl, oxazolopyridinyl, imidazolopyridinyl, and pyrazolopyridinyl.

[00110] Examples of 5- to 6-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.

[00111] As used herein, the term "bicyclic heterocycle" or "bicyclic heterocyclic group" is intended to mean a stable 9- or 10-membered heterocyclic ring system which contains two fused rings and consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S. Of the two fused rings, one ring is a 5- or 6-membered monocyclic aromatic ring comprising a 5-membered heteroaryl ring, a 6-membered heteroaryl ring or a benzo ring, each fused to a second ring. The second ring is a 5- or 6-membered monocyclic ring which is saturated, partially unsaturated, or unsaturated, and comprises a 5-membered heterocycle, a 6-membered heterocycle or a carbocycle (provided the first ring is not benzo when the second ring is a carbocycle).

[00112] The bicyclic heterocyclic group may be attached to its pendant group at any heteroatom or carbon atom which results in a stable structure. The bicyclic heterocyclic group described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1.

[00113] Examples of a bicyclic heterocyclic group are, but not limited to, quinolinyl, isoquinolinyl, phthalazinyl, quinazolinyl, indolyl, isoindolyl, indolinyl, lH-indazolyl, benzimidazolyl, 1,2,3,4-tetrahydroquinolinyl, 1,2,3,4-tetrahydroisoquinolinyl,

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

1 ,2,3 ,4-tetrahydro-quinoxalinyl, and 1 ,2,3 ,4-tetrahydro-quinazolinyl.

[00114] As used herein, the term "aromatic heterocyclic group" or "heteroaryl" is intended to mean stable monocyclic and polycyclic aromatic hydrocarbons that include at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrroyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1 ,2,4-thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl, benzodioxolanyl, and benzodioxane. Heteroaryl groups are substituted or unsubstituted. The nitrogen atom is substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., N→0 and S(0) p , wherein p is 0, 1 or 2).

[00115] Examples of 5- to 6-membered heteroaryls include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, imidazolyl, imidazolidinyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, oxazolidinyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl.

[00116] As used herein, the term "aromatic heterocyclic group" or "heteroaryl" is intended to mean stable monocyclic and polycyclic aromatic hydrocarbons that include at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrroyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1 ,2,4-thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl, benzodioxolanyl, and benzodioxane. Heteroaryl groups are substituted or unsubstituted. The nitrogen atom is substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., N→0 and S(0) p , wherein p is 0, 1 or 2).

[00117] Examples of 5- to 6-membered heteroaryls include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, imidazolyl, imidazolidinyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, oxazolidinyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl.

[00118] Bridged rings are also included in the definition of heterocycle. A bridged ring occurs when one or more, preferably one to three, atoms (i.e., C, O, N, or S) link two non-adjacent carbon or nitrogen atoms. Examples of bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and a carbon-nitrogen group. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

[00119] The term "counter ion" is used to represent a negatively charged species such as chloride, bromide, hydroxide, acetate, and sulfate or a positively charged species such as sodium (Na+), potassium (K+), calcium (Ca 2+ ), ammonium (R n NH m + where n=0-4 and m=0-4) and the like.

[00120] When a dotted ring is used within a ring structure, this indicates that the ring structure may be saturated, partially saturated or unsaturated.

[00121] As used herein, the term "amine protecting group" means any group known in the art of organic synthesis for the protection of amine groups which is stable to an ester reducing agent, a disubstituted hydrazine, R4-M and R7-M, a nucleophile, a hydrazine reducing agent, an activator, a strong base, a hindered amine base and a cyclizing agent. Such amine protecting groups fitting these criteria include those listed in Greene, T.W. et al., Protecting Groups in Organic Synthesis, 4th Edition, Wiley (2007) and The Peptides: Analysis, Synthesis, Biology, Vol. 3, Academic Press, New York (1981), the disclosure of which is hereby incorporated by reference. Examples of amine protecting groups include, but are not limited to, the following: (1) acyl types such as formyl, trifluoroacetyl, phthalyl, and p-toluenesulfonyl; (2) aromatic carbamate types such as benzyloxycarbonyl (Cbz) and substituted benzyloxycarbonyls, l-(p-biphenyl)-l-methylethoxycarbonyl, and 9-fluorenylmethyloxycarbonyl (Fmoc); (3) aliphatic carbamate types such as

tert-butyloxycarbonyl (Boc), ethoxycarbonyl, diisopropylmethoxycarbonyl, and allyloxycarbonyl; (4) cyclic alkyl carbamate types such as cyclopentyloxycarbonyl and adamantyloxycarbonyl; (5) alkyl types such as triphenylmethyl and benzyl; (6) trialkylsilane such as trimethylsilane; (7) thiol containing types such as

phenylthiocarbonyl and dithiasuccinoyl; and (8) alkyl types such as triphenylmethyl, methyl, and benzyl; and substituted alkyl types such as 2,2,2-trichloroethyl,

2-phenylethyl, and ?-butyl; and trialkylsilane types such as trimethylsilane.

[00122] As referred to herein, the term "substituted" means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound. Ring double bonds, as used herein, are double bonds that are formed between two adjacent ring atoms (e.g., C=C, C=N, or N=N).

[00123] In cases wherein there are nitrogen atoms (e.g., amines) on compounds of the present invention, these may be converted to N-oxides by treatment with an oxidizing agent (e.g., mCPBA and/or hydrogen peroxides) to afford other compounds of this invention. Thus, shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxide (N→0) derivative.

[00124] When any variable occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-3 R, then said group may optionally be substituted with up to three R groups, and at each occurrence R is selected independently from the definition of R.

[00125] When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom in which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such substituent.

[00126] Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.

[00127] The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, and/or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[00128] As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic groups such as amines; and alkali or organic salts of acidic groups such as carboxylic acids. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic,

methanesulfonic, ethane disulfonic, oxalic, and isethionic, and the like.

[00129] The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington: The Science and Practice of Pharmacy, 22nd Edition, Allen, L.V., Jr., ed., Pharmaceutical Press, London, UK (2012), the disclosure of which is hereby incorporated by reference.

[00130] In addition, compounds of formula I may have prodrug forms. Any compound that will be converted in vivo to provide the bioactive agent (i.e., a compound of formula I) is a prodrug within the scope and spirit of the invention. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:

a) Bundgaard, FL, ed., Design of Prodrugs, Elsevier (1985), and Widder, K. et al, eds., Methods in Enzymology, 1 12:309-396, Academic Press (1985);

b) Bundgaard, FL, Chapter 5, "Design and Application of Prodrugs", A Textbook of Drug Design and Development, pp. 1 13-191, Krosgaard-Larsen, P. et al, eds., Harwood Academic Publishers (1991);

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

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

e) Kakeya, N. et al., Chem. Pharm. Bull, 32:692 (1984); and f) Rautio, J., ed., Prodrugs and Targeted Delivery (Methods and Principles in Medicinal Chemistry), Vol. 47, Wiley-VCH (201 1).

[00131] Compounds containing a carboxy group can form physiologically

hydrolyzable esters that serve as prodrugs by being hydrolyzed in the body to yield formula I compounds per se. Such prodrugs are preferably administered orally since hydrolysis in many instances occurs principally under the influence of the digestive enzymes. Parenteral administration may be used where the ester per se is active, or in those instances where hydrolysis occurs in the blood. Examples of physiologically hydrolyzable esters of compounds of formula I include C^alkyl, C^alkylbenzyl,

4-methoxybenzyl, indanyl, phthalyl, methoxymethyl, C^ alkanoyloxy-C^alkyl (e.g., acetoxymethyl, pivaloyloxymethyl or propionyloxymethyl),

C^alkoxycarbonyloxy-C^alkyl (e.g., methoxycarbonyl-oxymethyl or

ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl,

(5-methyl-2-oxo-l,3-dioxolen-4-yl)-methyl), and other well known physiologically hydrolyzable esters used, for example, in the penicillin and cephalosporin arts. Such esters may be prepared by conventional techniques known in the art.

[00132] Preparation of prodrugs is well known in the art and described in, for example, King, F.D., ed., Medicinal Chemistry: Principles and Practice, The Royal Society of Chemistry, Cambridge, UK (2nd Edition, reproduced, 2006); Testa, B. et al, Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology, VCHA and Wiley-VCH, Zurich, Switzerland (2003); Wermuth, C.G., ed., The Practice of Medicinal Chemistry, Third Edition, Academic Press, San Diego, CA (2008).

[00133] The present invention also includes isotopically-labeled compounds of the invention, wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H (also represented as 'D' for deuterium) and 3 H, carbon such as ^C, 13 C, and 14 C, nitrogen, such as 13 N and 15 N, oxygen, such as 15 0, 17 0, and 18 0. Certain isotopically-labeled compounds of the invention, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, 3 H, and carbon-14, 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increase in vivo half-life or reduced dosage

requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as ^C, 15 0, and 13 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.

Isotopically-labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.

[00134] The term "solvate" means a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. The solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement. The solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules. "Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.

[00135] The names used herein to characterize a specific form, e.g., "N-l" etc., are not to be limited so as to exclude any other substance possessing similar or identical physical and chemical characteristics, but rather such names are used as mere identifiers that are to be interpreted in accordance with the characterization information presented herein.

[00136] All numbers expressing quantities of ingredients, weight percentages, temperatures, and so forth that are preceded by the word "about" are to be understood as only approximations so that slight variations above and below the stated number may be used to achieve substantially the same results as the stated number. Accordingly, unless indicated to the contrary, numerical parameters preceded by the word "about" are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.

[00137] All measurements are subject to experimental error and are within the contemplation of the invention.

[00138] When the invention is described or characterized by any of the disclosed figures or tables, it is understood that all variations within limitations and/or error margins of the experiments and technology are contemplated.

[00139] As used herein, "polymorphs" refer to crystalline forms having the same chemical structure/composition but different spatial arrangements of the molecules and/or ions forming the crystals.

[00140] As used herein, "amorphous" refers to a solid form of a molecule and/or ions that is not crystalline. An amorphous solid does not display a definitive X-ray diffraction pattern with sharp maxima.

[00141] Samples of the crystalline forms may be provided with substantially pure phase homogeneity, indicating the presence of a dominant amount of a single crystalline form and optionally minor amounts of one or more other crystalline forms. The presence of more than one crystalline form in a sample may be determined by techniques such as powder X-ray diffraction (PXRD) or solid state nuclear magnetic resonance spectroscopy (SSNMR). For example, the presence of extra peaks in the comparison of an

experimentally measured PXRD pattern with a simulated PXRD pattern may indicate more than one crystalline form in the sample. The simulated PXRD may be calculated from single crystal X-ray data. See Smith, D.K., "A FORTRAN Program for Calculating X-ray Powder Diffraction Patterns", Lawrence Radiation Laboratory, Livermore, California, UCRL-7196 (April 1963).

[00142] Preferably, the crystalline form has substantially pure phase homogeneity as indicated by less than 10%, preferably less than 5%, and more preferably less than 2% of the total peak area in the experimentally measured PXRD pattern arising from the extra peaks that are absent from the simulated PXRD pattern. Most preferred is a crystalline form having substantially pure phase homogeneity with less than 1% of the total peak area in the experimentally measured PXRD pattern arising from the extra peaks that are absent from the simulated PXRD pattern.

[00143] As used herein, "substantially pure", when used in reference to a crystalline form, means a compound having a purity greater than 90 weight %, including greater than 90, 91, 92, 93, 94, 95, 96, 97, 98, and 99 weight %, and also including equal to about 100 weight % of Compound la, based on the weight of the compound. The remaining material comprises other form(s) of the compound, and/or reaction impurities and/or processing impurities arising from its preparation. For example, a crystalline form of Compound I may be deemed substantially pure in that it has a purity greater than 90 weight %, as measured by means that are at this time known and generally accepted in the art, where the remaining less than 10 weight % of material comprises other form(s) of Compound la and/or reaction impurities and/or processing impurities.

[00144] The presence of reaction impurities and/or processing impurities may be determined by analytical techniques known in the art, such as, for example,

chromatography, nuclear magnetic resonance spectroscopy, mass spectrometry, and/or infrared spectroscopy.

[00145] As used herein, the parameter "molecules/asymmetric unit" refers to the number of molecules of the crystalline compound in the asymmetric unit.

[00146] As used herein, the unit cell parameter "molecules/unit cell" refers to the number of molecules of the crystalline compound in the unit cell.

[00147] Abbreviations as used herein, are defined as follows: " 1 x" for once, "2 x" for twice, "3 x" for thrice, "°C" for degrees Celsius, "eq" for equivalent or equivalents, "g" for gram or grams, "mg" for milligram or milligrams, "L" for liter or liters, "mL" for milliliter or milliliters, "μί" for microliter or microliters, "N" for normal, "M" for molar, "mmol" for millimole or millimoles, "min" for minute or min, "h" for hour or h, "rt" for room temperature, "RT" for retention time, "atm" for atmosphere, "psi" for pounds per square inch, "cone." for concentrate, "aq" for "aqueous", "sat" or "sat'd" for saturated, "MW" for molecular weight, "mp" for melting point, "MS" or "Mass Spec" for mass spectrometry, "ESI" for electrospray ionization mass spectroscopy, "HR" for high resolution, "HRMS" for high resolution mass spectrometry, "LCMS" for liquid chromatography mass spectrometry, "HPLC" for high pressure liquid chromatography, "RP HPLC" for reverse phase HPLC, "TLC" or "tic" for thin layer chromatography, "NMR" for nuclear magnetic resonance spectroscopy, "nOe" for nuclear Overhauser effect spectroscopy, "^R" for proton, "δ" for delta, "s" for singlet, "d" for doublet, "t" for triplet, "q" for quartet, "m" for multiplet, "br" for broad, "Hz" for hertz, and "α", "β", "R", "S", "E", and "Z" are stereochemical designations familiar to one skilled in the art.

Me methyl

Et ethyl

Pr propyl

i ' -Pr isopropyl

Bu butyl

i-Bu isobutyl

t-Bu tert-butyl

Ph phenyl

Bn benzyl

Hex hexanes

MeOH methanol

EtOH ethanol

i-PrOH or IPA isopropanol

AcOH or HOAc acetic acid

Ag 2 C0 3 silver carbonate

AgOAc silver acetate

ADDP 1 , 1 '-(azodicarbonyl)dipiperidine

DEAD diethyl azodicarboxylate

DBAD di-tert-butyl azodicarboxylate

PPh 3 triphenylphosphine

PBu 3 tributylphosphine

CDCI3 deutero-chloroform

CHCI3 chloroform

cDNA complimentary DNA

DMF dimethyl formamide

DMSO dimethyl sulfoxide EDTA ethylenediaminetetraacetic acid

EtOAc ethyl acetate

Et 2 0 diethyl ether

AICI3 aluminum chloride

Boc tert-butyloxycarbonyl

DCM dichloromethane

CH 2 C1 2 dichloromethane

CH 3 CN or ACN acetonitrile

Cs 2 C0 3 cesium carbonate

HC1 hydrochloric acid

H 2 S0 4 sulfuric acid

K 2 C0 3 potassium carbonate

KCN potassium cyanide

mCPBA or m-CPBA meto-chloroperbenzoic acid

Pd/C palladium on carbon

PhS0 2 Cl benzenesulfonyl chloride

j ' -Pr 2 NEt diisopropylethylamine

Pd(dppf)Cl 2 [l,l'-bis(diphenylphosphino)ferrocene]dichloropalladium

Pd 2 (dba) 3 tris(dibenzylideneacetone)dipalladium

Sphos 2-dicyclohexylphosphino-2',6'-dimethoxybiphenyl

Sphos pre.cat. chloro(2-dicyclohexylphosphino-2',6'-dimethoxy- 1,1'- biphenyl)[2-(2-aminoethylphenyl)]palladium(II) - methyl-?-butyl ether adduct

LiHMDS lithium bis(trimethylsilyl)amide

9-BBN 9-borabicyclo[3.3.1 Jnonane

BBr 3 boron tribromide

H 2 0 2 hydrogen peroxide

DIEA or Hiinig's base N,N-Diisopropylethylamine

PS polystyrene

Si0 2 silica oxide SnCl 2 tin(II) chloride

SELECTFLUOR® l-Chloromethyl-4-fluoro-l,4-diazoniabicyclo[2.2.2]octane bis(tetrafluoroborate)

L-selectride lithium tri-sec-butyl(hydrido)borate

TBAF tetra-w-butylammonium fluoride

TEA triethylamine

TFA trifluoroacetic acid

TFAA trifluoroacetic anhydride

THF tetrahydrofuran

TMSC1 chlorotrimethy Is ilane

TBSOTf tert-butyldimethylsilyl trifluoromethanesulfonate

TBSC1 tert-butyldimethylsilyl chloride

TMSCHN 2 trimethylsilyldiazomethane

KOAc potassium acetate

MgS0 4 magnesium sulfate

MsCl methanesulfonyl chloride

MsOH or MSA methylsulfonic acid

NaCl sodium chloride

NaH sodium hydride

NaHC0 3 sodium bicarbonate

NaOH sodium hydroxide

Na 2 S0 3 sodium sulfite

Na 2 S 2 0 3 sodium thiosulphate

Na 2 S0 4 sodium sulfate

NH 3 ammonia

NH4CI ammonium chloride

NH4OH ammonium hydroxide

LG leaving group

PG protecting group [00148] The compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis. The compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below. The reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention.

[00149] The novel compounds of this invention may be prepared using the reactions and techniques described in this section. Also, in the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, are chosen to be the conditions standard for that reaction, which should be readily recognized by one skilled in the art. Restrictions to the substituents that are compatible with the reaction conditions will be readily apparent to one skilled in the art and alternate methods must then be used.

SYNTHESIS

[00150] The compounds of Formula (I) may be prepared by the exemplary processes described in the following schemes and working examples, as well as relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter and in the working examples.

Protection and de-protection in the processes below may be carried out by procedures generally known in the art (see, for example, Greene, T.W. et al, Protecting Groups in Organic Synthesis, 4th Edition, Wiley (2007)). General methods of organic synthesis and functional group transformations are found in: Trost, B.M. et al., eds., Comprehensive Organic Synthesis: Selectivity, Strategy & Efficiency in Modern Organic Chemistry, Pergamon Press, New York, NY (1991); Smith, M.B. et al, March 's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure. 6th Edition, Wiley & Sons, New York, NY (2007); Katritzky, A.R. et al., eds., Comprehensive Organic Functional Groups Transformations II, 2nd Edition, Elsevier Science Inc., Tarrytown, NY (2004); Larock, R.C., Comprehensive Organic Transformations, VCH Publishers, Inc., New York, NY (1999), and references therein.

[00151] Methods for synthesis of a large variety of substituted dihydropyrazole compounds useful as starting materials for the preparation of compounds of the present invention are well known in the art. For examples of methods useful for the preparation of dihydropyrazole materials see the following references and citations therein: Katritzky et al, eds., Comprehensive Heterocyclic Chemistry, Pergamon Press Inc., New York (1996); Sibi, M.P. et al, Organic Letters, 1 1(23):5366 (2009); Sibi, M.P. et al, J. Am. Chem. Soc, 127(23):8276 (2005); Manyem, S. et al, J. Comb. Chem., 9:20 (2007);

Garanti, L. et al., Tetrahedron: Asymmetry, 13 : 1285 (2002); Molteni, G., Tetrahedron: Asymmetry, 15: 1077 (2004); Benassuti, L.D. et al, Tetrahedron, 60:4627 (2004);

Shimizu, T. et al, Bull. Chem. Soc. Jpn., 57:787 (1984).

[00152] Compounds of Formula (I) can be prepared as shown in Scheme 1.

Conversion of hydrazine A, which contains a LG such as = F, CI, Br and the like, to hydrazide B with trifluoroacetic anhydride followed by treatment with a phenylsulfonyl chloride forms hydrazonoyl chloride C. Hydrazonoyl chloride C can undergo a [3+2] cycloaddition with an α,β-unsaturated carbonyl compound where Y is a chiral auxiliary, such as (4S)-phenyloxazolidinone and the like, or alkoxy group to give dihydropyrazole D as depicted in Scheme 1. Reduction of the carbonyl group in D, via a reducing agent, e.g., NaBH 4 or LiBH 4 , leads to hydroxyl E. Activation of the hydroxyl group of E, via methanesulfonyl chloride, for example, and displacement with a cyanide reagent, e.g., sodium cyanide, potassium cyanide or trimethylsilylcyanide, leads to nitrile F. Nitrile F can be converted to methyl ester G by acidic methano lysis. Intermediate G can be converted to boronate H by metal catalyzed borylation, e.g., catalyzed by Pd(dppf)Cl2. Cleavage of the boronate group in H, via oxidation with, for example, H 2 02, leads to phenol M. Displacement of the hydroxyl group in L (synthesis shown vide infra) with phenol M, via e.g., PBu 3 and ADDP, or PPh 3 and DEAD, or PPh 3 and DBAD, generates intermediate N. Intermediate N can be converted to compounds of Formula (I) by hydrolysis, via a hydroxide reagent, e.g., LiOH or NaOH. Intermediate L can be synthesized from amine J. Conversion of the amine J to intermediate K via metal catalyzed amination or S ^r reaction, e.g., Pd (0), Cu(I) or heat with base. Activation of

X group in K, via toluenesulfonyl chloride, for example, leads to intermediate L.

Scheme 1

X

PG = protecting group LG = leaving group

[00153] Alternatively, compounds of Formula (I) may be synthesized starting with amine O; treatment of O with Mel affords ammonium salt P. Conversion of P to Q with R^NH^ and base (as described, for example, in Tortolani, D.R. et al, Organic Letters, 1(8): 1261-1263 (1999)) followed by treatment with reductant, e.g., L-selectride, forms alcohol S as shown in Scheme 2. Transformation of the hydroxyl group in S to leaving group gives intermediate L, via methanesulfonyl chloride, for example. Intermediate L could be converted to compounds of Formula (I) according to the synthetic steps described in Scheme 1.

Scheme 2

[00154] Alternatively, compounds of Formula (I) can be synthesized via reaction of intermediate C with a substituted acrylate AA in the presence of a Ag salt to provide dihydropyrazole AB as depicted in Scheme 4. The methyl ester AB can be hydrolyzed, via LiOH, for example, to afford carboxylic acid AC. Carboxylic acid AC can be converted to ester AD, via Arndt-Eistert homologation. Intermediate AD could be converted to compounds of Formula (I) by according to the synthetic steps described in Scheme 1.

Scheme 4

[00155] The compound of the instant invention herein described may have asymmetric centers. For example, the chiral carbon atoms in Formula (I) or Formula (II), as indicated below, exist in either as S or R configuration.

[00156] Thus, the stereoisomeric configurations of each compound of Formula (I) or Formula (II) are considered part of the invention. In structures where the stereochemistry of an intermediate or final compound is not indicated, it has not been determined. rv. BIOLOGY

[00157] Diabetes mellitus is a serious disease afflicting over 100 million people worldwide. It is diagnosed as a group of disorders characterized by abnormal glucose homeostasis resulting in elevated blood glucose. Diabetes is a syndrome with interrelated metabolic, vascular, and neuropathic components. The metabolic abnormality is generally characterized by hyperglycemia and alterations in carbohydrate, fat and protein metabolism caused by absent or reduced insulin secretion and/or ineffective insulin secretion. The vascular syndrome consists of abnormalities in the blood vessels leading to cardiovascular, retinal and renal complications. Abnormalities in the peripheral and autonomic nervous systems are also part of diabetic syndrome. Strikingly, diabetes is the fourth leading cause of global death by disease, the largest cause of kidney failure in developed countries, the leading cause of vision loss in industrialized countries and has the greatest prevalence increase in developing countries.

[00158] Type 2 diabetes, which accounts for 90% of diabetes cases, is characterized by increasing insulin resistance associated with inadequate insulin secretion after a period of compensatory hyperinsulinemia. The reasons for β cell secondary failure are not completely understood. Acquired pancreatic islet damage or exhaustion and/or genetic factors causing susceptibility to islet secretory insufficiency have been hypothesized.

[00159] Free fatty acids (FFAs) are evidenced to influence insulin secretion from β cells primarily by enhancing glucose-stimulated insulin secretion (GSIS). Although glucose is recognized as the major stimulator of insulin secretion from β cells, other stimuli, such as amino acids, hormones, and FFAs, also regulate insulin secretion. Thus, under normal settings, insulin secretion from β cells in response to food intake is evoked by the collective stimuli of nutrients, such as glucose, amino acids, and FFAs, and hormones like the incretin glucagon-like peptide 1 (GLP-1). Fatty acids are also known to stimulate the secretion of several gut satiety hormones, including cholocystokinine (CCK), GLP-1, and peptide YY (PYY).

[00160] G-protein coupled receptors (GPCRs) expressed in β cells are known to modulate the release of insulin in response to changes in plasma glucose levels. GPR40, also known as fatty acid receptor 1 (FFAR1), is a membrane-bound FFA receptor which is preferentially expressed in the pancreatic islets and specifically in β cells. GPR40 (e.g., human GPR40, RefSeq mRNA ID NM 005303; e.g., mouse GPR40 RefSeq mRNA ID NM_194057) is a GPCR located at chromosome 19ql3.12. GPR40 is activated by medium to long chain fatty acids and thereby triggering a signaling cascade that results in increased levels of [Ca 2+ ] j in β cells and subsequent stimulation of insulin secretion (Itoh et al, Nature, 422: 173-176 (2003)). Selective small molecule agonists of GPR40 have been shown to promote GSIS and reduce blood glucose in mice (Tan et al, Diabetes, 57:2211-2219 (2008)). Briefly, when activators of GPR40 are administered to either normal mice or mice that are prone to diabetes due to genetic mutation, prior to a glucose tolerance test, improvements in glucose tolerance are observed. A short-lived increase in plasma insulin levels are also observed in these treated mice. It has also been shown that GPR40 agonists restore GSIS in pancreatic β-cells from the neonatal STZ rats suggesting that GPR40 agonists will be efficacious in diabetics with compromised β-cell function and mass. Fatty acids are known to stimulate the secretion of several gut satiety hormones, including cholocystokinine (CCK), GLP-1, and peptide YY (PYY), and GPR40 has been shown to colocalize with cells that secrete such hormones (Edfalk et al., Diabetes, 57:2280-2287 (2008); Luo et al. PLOSone, 7: 1-12 (2012)). Fatty acids are also known to play a role in neuronal development and function, and GPR40 has been reported as a potential modulator of the fatty acid effects on neurons (Yamashima, T., Progress in Neurobiology, 84: 105-115 (2008)).

[00161] Given the increase in the worldwide patient population afflicted by type 2 diabetes, there is a need for novel therapies which are effective with minimal adverse events. To decrease medical burden of type 2 diabetes through enhanced glycemic control, GPR40 modulator compounds of the present invention are being investigated here for their incretin effect to promote GSIS as well as the potential combination with a broad range of anti-diabetic drugs.

[00162] The term "modulator" refers to a chemical compound with capacity to either enhance (e.g., "agonist" activity) or partially enhance (e.g., "partial agonist" activity) or inhibit (e.g., "antagonist" activity or" inverse agonist" activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, receptor internalization, and/or may be manifest only in particular cell types.

[00163] It is also desirable and preferable to find compounds with advantageous and improved characteristics compared with known anti-diabetic agents, in one or more of the following categories that are given as examples, and are not intended to be limiting: (a) pharmacokinetic properties, including oral bioavailability, half life, and clearance; (b) pharmaceutical properties; (c) dosage requirements; (d) factors that decrease blood drug concentration peak-to-trough characteristics; (e) factors that increase the concentration of active drug at the receptor; (f) factors that decrease the liability for clinical drug-drug interactions; (g) factors that decrease the potential for adverse side-effects, including selectivity versus other biological targets; and (h) improved therapeutic index with less propensity for hypoglycemia.

[00164] As used herein, the term "patient" encompasses all mammalian species.

[00165] As used herein, the term "subject" refers to any human or non-human organism that could potentially benefit from treatment with a GPR40 modulator.

Exemplary subjects include human beings of any age with risk factors for metabolic disease. Common risk factors include, but are not limited to, age, sex, weight, family history, or signs of insulin resistance such as acanthosis nigricans, hypertension, dislipidemia, or polycystic ovary syndrome (PCOS).

[00166] As used herein, "treating" or "treatment" cover the treatment of a disease-state in a mammal, particularly in a human, and include: (a) inhibiting the disease-state, i.e., arresting it development; and/or (b) relieving the disease-state, i.e., causing regression of the disease state.

[00167] As used herein, "prophylaxis" or "prevention" cover the preventive treatment of a subclinical disease-state in a mammal, particularly in a human, aimed at reducing the probability of the occurrence of a clinical disease-state. Patients are selected for preventative therapy based on factors that are known to increase risk of suffering a clinical disease state compared to the general population. "Prophylaxis" therapies can be divided into (a) primary prevention and (b) secondary prevention. Primary prevention is defined as treatment in a subject that has not yet presented with a clinical disease state, whereas secondary prevention is defined as preventing a second occurrence of the same or similar clinical disease state.

[00168] As used herein, "risk reduction" covers therapies that lower the incidence of development of a clinical disease state. As such, primary and secondary prevention therapies are examples of risk reduction.

[00169] "Therapeutically effective amount" is intended to include an amount of a compound of the present invention that is effective when administered alone or in combination to modulate GPR40 and/or to prevent or treat the disorders listed herein. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the preventive or therapeutic effect, whether administered in combination, serially, or simultaneously.

In Vitro GPR40 Assays

FDSS-based Intracellular Calcium Assay

[00170] Cell lines expressing GPR40 are generated using the pDEST 3xFLAG® gene expression system and are cultured in culture medium comprising the following components: F 12 (Gibco #11765), 10% lipid deprived fetal bovine serum, 250 μg/mL zeocin and 500 μg/mL G418. To conduct the fluorescent imaging plate reader

(FLIPR)-based calcium flux assay to measure intracellular Ca 2+ response, cells expressing GPR40 are plated on 384 well plates (BD BIOCOAT® #356697) at a density of 20,000 cells/20 μΐ, medium per well in phenol red and serum-free DMEM (Gibco #21063-029) and incubated overnight. Using BD kit #s 80500-310 or -301, the cells are incubated with 20 μΐ, per well of Hank's buffered salt solution with 1.7 mM probenecid and Fluo-3 at 37 °C for 30 min. Compounds are dissolved in DMSO and diluted to desired concentrations with assay buffer and added to the cells as 3x solution (20 μΐ, per well). Run fluorescence/luminescence reader FDSS (Hamamatsu) to read intracellular Ca 2+ response.

[00171] The exemplified Examples disclosed below were tested in the Human GRP40 In Vitro assay described above and found having hGRP40 modulating activity, reported as hGPR40 EC 50 .

GPR40 IP-One HTRF Assays in HEK293/GPR40 Inducible Cell Lines

[00172] Human, mouse and rat GPR40-mediated intracellular IP-One HTRF assays were established using human embryonic kidney HEK293 cells stably transfected with a tetracycline-inducible human, mouse or rat GPR40 receptor. Cells were routinely cultured in growth medium containing DMEM (Gibco Cat. #12430-047), 10% qualified FBS (Sigma, Cat. #F2442), 200 μ^πιΐ hygromycin (Invitrogen, Cat. #16087-010) and 1.5 μg/ml blasticidin (Invitrogen, Cat. #R210-01). About 12-15 million cells were passed into a T175 tissue culture flask (BD FALCON® 3531 12) with growth medium and incubated for 16-18 hours (overnight) at 370C with 5% CO2. The next day, assay medium was exchanged with growth medium containing 1000 ng/mL of tetracycline (Fluka Analytical, Cat. #87128) to induce GPR40 expression for 18-24 hours at 370C incubator with 5% C0 2 . After induction, the cells were washed with PBS (Gibco, Cat. #14190-036) and detached with Cell Stripper (CELLGRO®, Cat. #25-056-CL). 10-20 mL growth medium were added to the flask and cells were collected in 50 mL tubes (FALCON®, Cat.

#352098) and spun at 1000 RPM for 5 minutes. Culture medium was aspirated and the cells were resuspended in 10 mL of IX IP-One Stimulation Buffer from the Cisbio IP- One kit (Cisbio, Cat. #62IPAPEJ). The cells were diluted to 1.4 xl06 cells/mL in

Stimulation Buffer.

[00173] Test compounds were 3-fold, 1 1 -point serially diluted in DMSO in a REMP assay plate (Matrix Cat. #4307) by BIOCEL® (Agilent). The compounds were transferred into an Echo plate (LABCYTE, Cat. #LP-0200) and 20 nL of diluted compounds were transferred to an assay plate (proxi-plate from Perkin Elmer, Cat. #6008289) by Echo acoustic nano dispenser (LABCYTE, model ECHO550). 14 μΐ ^ of the diluted cells were then added to the assay plate by Thermo (SN 836 330) CombiDrop and incubated at room temperature for 45 minutes. Then 3 μϊ ^ of IP 1 coupled to dye D2 from the Cisbio IP-One kit were added to the assay plate followed by 3 μϊ ^ of Lumi4TM-Tb cryptate K from the kit. The plate was further incubated at room for 1 hour before reading on the Envision (Perkin Elmer Model2101) with an HTRF protocol. Activation data for the test compound over a range of concentrations was plotted as percentage activation of the test compound (100% = maximum response). After correcting for background [(sample read-mean of low control)/ (mean of high control - mean of low control)] (low control is DMSO without any compound), EC 5 Q values were determined. The EC 5 Q is defined as the concentration of test compound which produces 50% of the maximal response and was quantified using the 4 parameter logistic equation to fit the data. The maximal Y value observed (% Ymax) was calculated relative to a BMS standard reference compound at a final concentration of 0.625 μΜ.

[00174] Some of the exemplified Examples disclosed below were tested in the Human GRP40 In Vitro assay described above and found having hGRP40 modulating activity reported as hGPR40 IP1 EC 50 . In Vivo GPR40 Assays

Acute Oral Glucose Tolerance Test

[00175] Ten week old C57BL6 mice were housed individually and fasted for 5 hours on the day of study. Tail vein sampling was performed from nicked tails to obtain plasma samples. Baseline plasma samples were taken at t = 0. Mice were orally treated with vehicle or compounds co-administered with glucose (2g/kg). Sampling thereafter from tails of treated mice at 20, 40, 60, 120 and 180 min provided data used for generating glucose excursion curves from which 0-180 min blood glucose excursion profiles were generated. The area under the curve (AUC) allowed for assessment of glucose lowering by compound treatments. Blood samples were collected in EDTA- treated tubes

(MICROVETTE® CB300, Sarstedt, Numbrecht, Germany), stored on ice and spun @ 6000 rpm for 10 minutes. Plasma glucose was analyzed on the same day using an AU680 Clinical Chemistry Analyzer (Beckman Coulter, Brea, CA). Statistical analysis is a one way ANOVA with Dunnett's post hoc test or two-way student's t test where appropriate. P values less than 0.05 were considered statistically significant. Glucose reduction is reported as a % change in AUC (0-180 min) from the vehicle treatment group. For example, "Acute oral glucose tolerance: -50% @ 0.3 mg/kg" represents the results of a study as described above, whereupon administration of 0.3 mg/kg of the specified example results in a 50% reduction in glucose AUC (0-180 min) relative to vehicle treated animals.

Acute Oral Glucose Tolerance Test in Rats

[00176] Male SPRAGUE DAWLEY® rats (CRL, Wilmington MA) were used. Rats were delivered to the vivarium and acclimated for 1 week. Rats were fasted from 5 PM on the night before study. Overnight fasted rats were 180-200 grams at time of study. Tail vein sampling was performed to obtain baseline plasma samples. Rats were randomized to treatment groups based on fasting plasma glucose readings determined by Accu-Chek glucometer (Roche, Indianapolis, IN). Rats were dosed at 4mL/Kg body weight with 40% PEG400 (Sigma, St. Louis, MO) 10% CREMOPHOR® (Sigma, St. Louis, MO) and 50% distilled water with or without compounds. For rats that received BMS DPP4i combined with GPR40 agonist, administration was performed by co-dosing compounds. Plasma samples were collected one hour after compound dosing to determine baseline changes in glucose and active GLP- 1 levels in the presence and absence of BMS DPP4L Sampling thereafter from tail veins provided time point data to calculate AUCQ.^O' glucose as a marker of two hour glucose lowering efficacy. Blood samples were collected in EDTA- treated tubes (MICRO VETTE® CB300, Sarstedt, Numbrecht, Germany), stored on ice and spun @ 6000 rpm for 10 minutes. Plasma glucose was analyzed on the same day using an AU680 Clinical Chemistry Analyzer (Beckman Coulter, Brea, CA). Statistical analysis is a one way ANOVA with Dunnett's post hoc test or two-way student's t test where appropriate. P values less than 0.05 were considered statistically significant. Glucose reduction is reported as a % change in AUC (0-120 min) from the vehicle treatment group. Fasting hormone responses are the difference from basal levels 1 hour post dose. Active GLP-1 levels (GLP-1 (7-36) amide and GLP-1 (7-37)) were measured by ELISA (Millipore, Billerica, MA).

BMS DPP4i - Reference Compound

[00177] BMS DPP4i is disclosed in Simpkins, L. et al. Bioorganic Medicinal Chemistry Letters, 17(23):6476-6480 (2007) (compound 48) and in WO 2005/012249 (Example 3). BMS DPP4i has the following formula:

BMS DPP4i

[00178] BMS DPP4i was administered to rats alone, and in combination with Example 81, Isomer 2 of the present invention, at 10 mg/kg, as depicted in FIG. 5 and FIG. 6. As depicted in FIG. 5, the combination of BMS DPP4i with Example 81, Isomer 2 demonstrated greater reductions in plasma glucose during an oral glucose tolerance test than either Example 81, Isomer 2 or BMS DPP4i alone. As depicted in FIG. 6, the combination of BMS DPP4i with Example 81, Isomer 2 shows greater increases in active GLP-1 during an oral glucose tolerance test than either Example 81, Isomer 2 or BMS DPP4i alone. [00179] The compounds of the present invention possess activity as modulators of GPR40, and, therefore, may be used in the treatment of diseases associated with GPR40 activity. Via modulation of GPR40, the compounds of the present invention may preferably be employed to modulate the production/secretion of insulin and/or gut hormones, such as GLP-1, GIP, PYY, CCK and amylin.

[00180] Accordingly, the compounds of the present invention can be administered to mammals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to, treating, preventing, or slowing the progression of diabetes and related conditions, microvascular complications associated with diabetes, macrovascular complications associated with diabetes, cardiovascular diseases, Metabolic Syndrome and its component conditions, inflammatory diseases and other maladies. Consequently, it is believed that the compounds of the present invention may be used in preventing, inhibiting, or treating diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, retinopathy, neuropathy, nephropathy, diabetic kidney disease, acute kidney injury, cardiorenal syndrome, acute coronary syndrome, delayed wound healing, atherosclerosis and its sequelae (acute coronary syndrome, myocardial infarction, angina pectoris, peripheral vascular disease, intermittent claudication, myocardial ischemia, stroke, heart failure), Metabolic

Syndrome, hypertension, obesity, fatty liver disease, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL, high LDL, vascular restenosis, peripheral arterial disease, lipid disorders, liver diseases such as NASH (Non-Alcoholic SteatoHepatitis), NAFLD (Non-Alcoholic Fatty Liver Disease) and liver cirrhosis, neurodegenerative disease, cognitive impairment, dementia, and treatment of side-effects related to diabetes, lipodystrophy and osteoporosis from corticosteroid treatment.

[00181] Metabolic Syndrome or "Syndrome X" is described in Ford et al, J. Am. Med. Assoc., 287:356-359 (2002) and Arbeeny et al, Curr. Med. Chem. - Imm., Endoc. & Metab. Agents, 1 : 1-24 (2001).

[00182] GPR40 is expressed in neuronal cells, and is associated with development and maintenance of neuronal health in brain, as described in Yamashima, T., Progress in Neurobiology, 84: 105-1 15 (2008). V. PHARMACEUTICAL COMPOSITIONS, FORMULATIONS AND

COMBINATIONS

[00183] The compounds of this invention can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.

[00184] The term "pharmaceutical composition" means a composition comprising a compound of the invention in combination with at least one additional pharmaceutically acceptable carrier. A "pharmaceutically acceptable carrier" refers to media generally accepted in the art for the delivery of biologically active agents to animals, in particular, mammals, including, i.e., adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.

[00185] Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to which the agent-containing composition is to be administered; the intended route of administration of the composition; and the therapeutic indication being targeted.

Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, L.V., Jr. et al, Remington: The Science and Practice of Pharmacy (2 Volumes), 22nd Edition, Pharmaceutical Press (2012).

[00186] The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.

[00187] By way of general guidance, the daily oral dosage of each active ingredient, when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day. Intravenously, the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.

Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.

[00188] The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g., oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.

[00189] Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 2000 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.1 -95% by weight based on the total weight of the composition.

[00190] A typical capsule for oral administration contains at least one of the compounds of the present invention (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a No. 1 gelatin capsule. [00191] A typical injectable preparation is produced by aseptically placing at least one of the compounds of the present invention (250 mg) into a vial, aseptically freeze-drying and sealing. For use, the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation.

[00192] The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of the present invention, alone or in combination with a pharmaceutical carrier. Optionally, compounds of the present invention can be used alone, in

combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g., an antidiabetic agent or other pharmaceutically active material.

[00193] The compounds of the present invention may be employed in combination with other GPR40 modulators or one or more other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-diabetic agents, anti-hyperglycemic agents, anti-hyperinsulinemic agents, anti-retinopathic agents, anti-neuropathic agents, anti-nephropathic agents, anti-atherosclerotic agents, anti-ischemic agents, anti-hypertensive agents, anti-obesity agents, anti-dyslipidemic agents, anti-hyperlipidemic agents, anti-hypertriglyceridemic agents,

anti-hypercholesterolemic agents, anti-restenotic agents, anti-pancreatic agents, lipid lowering agents, anorectic agents, memory enhancing agents, anti-dementia agents, or cognition promoting agents, appetite suppressants, treatments for heart failure, treatments for peripheral arterial disease and anti-inflammatory agents.

[00194] Where desired, the compound of the present invention may be used in combination with one or more other types of antidiabetic agents and/or one or more other types of therapeutic agents which may be administered orally in the same dosage form, in a separate oral dosage form or by injection. The other type of antidiabetic agent that may be optionally employed in combination with the GPR40 receptor modulator of the present invention may be one, two, three or more antidiabetic agents or antihyperglycemic agents which may be administered orally in the same dosage form, in a separate oral dosage form, or by injection to produce an additional pharmacological benefit.

[00195] The antidiabetic agents used in the combination with the compound of the present invention include, but are not limited to, insulin secretagogues or insulin sensitizers, other GPR40 receptor modulators, or other antidiabetic agents. These agents include, but are not limited to, dipeptidyl peptidase IV inhibitors (DPP4i; for example, sitagliptin, saxagliptin, alogliptin, vildagliptin), biguanides (for example, metformin, phenformin), sulfonyl ureas (for example, gliburide, glimepiride, glipizide), glucosidase inhibitors (for example, acarbose, miglitol), PPARy agonists such as thiazolidinediones (for example, rosiglitazone, pioglitazone), PPAR α/γ dual agonists (for example, muraglitazar, tesaglitazar, aleglitazar), glucokinase activators (as described in Fyfe, M.C.T. et al, Drugs of the Future, 34(8):641-653 (2009) and incorporated herein by reference), other GPR40 receptor modulators (e.g., TAK-875), GPR1 19 receptor modulators (for example, MBX-2952, PSN821, APD597), GPR120 receptor modulators (for example, as described in Shimpukade, B. et al, J. Med. Chem., 55(9):4511-4515 (2012)), sodium-glucose transporter-2 (SGLT2) inhibitors (for example dapagliflozin, canagliflozin, empagliflozin, remagliflozin), l lb-HSD-1 inhibitors (for example MK-0736, BI35585, BMS-823778, and LY2523199), MGAT inhibitors (for example, as described in Barlind, J.G. et al, Bioorg. Med. Chem. Lett, 23(9):2721-2726 (2013); or US 2013/0143843 Al), amylin analogs such as pramlintide, and/or insulin. Reviews of current and emerging therapies for the treatment of diabetes can be found in: Mohler, M.L. et al, Medicinal Research Reviews, 29(1): 125-195 (2009), and Mizuno, C.S. et al, Current Medicinal Chemistry, 15:61-74 (2008).

[00196] The GPR40 receptor modulator of formula I may also be optionally employed in combination with agents for treating complication of diabetes. These agents include PKC inhibitors and/or AGE inhibitors.

[00197] The GPR40 receptor modulator of formula I way also be optionally employed in combination with one or more hypophagic agents such as diethylpropion, phendimetrazine, phentermine, orlistat, sibutramine, lorcaserin, pramlintide, topiramate, MCHR1 receptor antagonists, oxyntomodulin, naltrexone, Amylin peptide, NPY Y5 receptor modulators, NPY Y2 receptor modulators, NPY Y4 receptor modulators, cetilistat, 5HT2c receptor modulators, and the like. The compound of structure I may also be employed in combination with an agonist of the glucagon-like peptide- 1 receptor (GLP-1 R), such as exenatide, liraglutide, GPR- 1(1-36) amide, GLP-l(7-36) amide, GLP-l(7-37) (as disclosed in U.S. Patent No. 5,614,492 to Habener, the disclosure of which is incorporated herein by reference), which may be administered via injection, intranasal, or by transdermal or buccal devices. Reviews of current and emerging therapies for the treatment of obesity can be found in: Melnikova, I. et al, Nature Reviews Drug Discovery, 5:369-370 (2006); Jones, D., Nature Reviews: Drug Discovery, 8:833-834 (2009); Obici, S., Endocrinology, 150(6):2512-2517 (2009); and Elangbam, C.S., Vet. Pathol, 46(1): 10-24 (2009).

[00198] The above other therapeutic agents, when employed in combination with the compounds of the present invention may be used, for example, in those amounts indicated in the Physicians' Desk Reference, as in the patents set out above, or as otherwise determined by one of ordinary skill in the art.

[00199] Particularly when provided as a single dosage unit, the potential exists for a chemical interaction between the combined active ingredients. For this reason, when the compound of the present invention and a second therapeutic agent are combined in a single dosage unit they are formulated such that although the active ingredients are combined in a single dosage unit, the physical contact between the active ingredients is minimized (that is, reduced). For example, one active ingredient may be enteric coated. By enteric coating one of the active ingredients, it is possible not only to minimize the contact between the combined active ingredients, but also, it is possible to control the release of one of these components in the gastrointestinal tract such that one of these components is not released in the stomach but rather is released in the intestines. One of the active ingredients may also be coated with a material that affects a sustained-release throughout the gastrointestinal tract and also serves to minimize physical contact between the combined active ingredients. Furthermore, the sustained-released component can be additionally enteric coated such that the release of this component occurs only in the intestine. Still another approach would involve the formulation of a combination product in which the one component is coated with a sustained and/or enteric release polymer, and the other component is also coated with a polymer such as a low viscosity grade of hydroxypropyl methylcellulose (HPMC) or other appropriate materials as known in the art, in order to further separate the active components. The polymer coating serves to form an additional barrier to interaction with the other component.

[00200] These as well as other ways of minimizing contact between the components of combination products of the present invention, whether administered in a single dosage form or administered in separate forms but at the same time by the same manner, will be readily apparent to those skilled in the art, once armed with the present disclosure.

[00201] The compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents. By "administered in combination" or "combination therapy" it is meant that the compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated. When administered in combination, each component may be administered at the same time or sequentially in any order at different points in time. Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.

[00202] The compounds of the present invention are also useful as standard or reference compounds, for example as a quality standard or control, in tests or assays involving the GPR40 receptor. Such compounds may be provided in a commercial kit, for example, for use in pharmaceutical research involving GPR40 or anti-diabetic activity. For example, a compound of the present invention could be used as a reference in an assay to compare its known activity to a compound with an unknown activity. This would ensure the experimentor that the assay was being performed properly and provide a basis for comparison, especially if the test compound was a derivative of the reference compound. When developing new assays or protocols, compounds according to the present invention could be used to test their effectiveness.

[00203] The compounds of the present invention may also be used in diagnostic assays involving GPR40.

[00204] The present invention also encompasses an article of manufacture. As used herein, article of manufacture is intended to include, but not be limited to, kits and packages. The article of manufacture of the present invention, comprises: (a) a first container; (b) a pharmaceutical composition located within the first container, wherein the composition, comprises: a first therapeutic agent, comprising: a compound of the present invention or a pharmaceutically acceptable salt form thereof; and, (c) a package insert stating that the pharmaceutical composition can be used for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40 (as defined previously). In another embodiment, the package insert states that the pharmaceutical composition can be used in combination (as defined previously) with a second therapeutic agent for the treatment and/or prophylaxis of multiple diseases or disorders associated with GPR40. The article of manufacture can further comprise: (d) a second container, wherein components (a) and (b) are located within the second container and component (c) is located within or outside of the second container. Located within the first and second containers means that the respective container holds the item within its boundaries.

[00205] The first container is a receptacle used to hold a pharmaceutical composition. This container can be for manufacturing, storing, shipping, and/or individual/bulk selling. First container is intended to cover a bottle, jar, vial, flask, syringe, tube (e.g., for a cream preparation), or any other container used to manufacture, hold, store, or distribute a pharmaceutical product.

[00206] The second container is one used to hold the first container and, optionally, the package insert. Examples of the second container include, but are not limited to, boxes (e.g., cardboard or plastic), crates, cartons, bags (e.g., paper or plastic bags), pouches, and sacks. The package insert can be physically attached to the outside of the first container via tape, glue, staple, or another method of attachment, or it can rest inside the second container without any physical means of attachment to the first container. Alternatively, the package insert is located on the outside of the second container. When located on the outside of the second container, it is preferable that the package insert is physically attached via tape, glue, staple, or another method of attachment. Alternatively, it can be adjacent to or touching the outside of the second container without being physically attached.

[00207] The package insert is a label, tag, marker, etc. that recites information relating to the pharmaceutical composition located within the first container. The information recited will usually be determined by the regulatory agency governing the area in which the article of manufacture is to be sold (e.g., the United States Food and Drug

Administration). Preferably, the package insert specifically recites the indications for which the pharmaceutical composition has been approved. The package insert may be made of any material on which a person can read information contained therein or thereon. Preferably, the package insert is a printable material (e.g., paper, plastic, cardboard, foil, adhesive-backed paper or plastic, etc.) on which the desired information has been formed (e.g., printed or applied). [00208] Other features of the invention will become apparent in the course of the following descriptions of exemplary embodiments that are given for illustration of the invention and are not intended to be limiting thereof. VI. EXAMPLES

[00209] The following Examples are offered as illustrative, as a partial scope and particular embodiments of the invention and are not meant to be limiting of the scope of the invention. Abbreviations and chemical symbols have their usual and customary meanings unless otherwise indicated. Unless otherwise indicated, the compounds described herein have been prepared, isolated and characterized using the schemes and other methods disclosed herein or may be prepared using the same.

HPLC/MS AND PREPARATORY/ANALYTICAL HPLC METHODS EMPLOYED IN CHARACTERIZATION OR PURIFICATION OF EXAMPLES

[00210] Analytical HPLC/MS (unless otherwise noted) was performed on Shimadzu SCL-IOA liquid chromatographs and Waters MICROMASS® ZQ Mass Spectrometers (Desolvation Gas: Nitrogen; Desolvation Temp. 250 °C; Ion Source Temp: 120 °C; Positive Electrospray conditions) using the following method:

Linear Gradient of 0% to 100% solvent B over 2 min, with 1 minute hold at 100% B;

UV visualization at 220 nm;

Column: PHENOMENEX® Luna CI 8 (2) 30mm x 4.60mm; 5m particle (Heated to Temp. 40 °C);

Flow rate: 5 mL/min;

Solvent A: 10% ACN, 90% water, 0.1% TFA; or, 10% MeOH, 90% water, 0.1%

TFA; and

Solvent B: 90% ACN, 10% water, 0.1% TFA; or, 90% MeOH, 10% water, 0.1%

TFA. [00211] Preparatory HPLC (unless otherwise noted) was performed on a Shimadzu SCL-IOA liquid chromatograph with a linear gradient of 20-100% Solvent B over 10 or 30 min, with either a 2 or 5 min (respectively) hold at 100% Solvent B; UV visualization at 220 nm;

Column: PHENOMENEX® Luna Axia 5μ CI 8 30x100 mm;

Flow rate: 20 mL/min;

Solvent A: 10% ACN, 90% water, 0.1% trifluoroacetic Acid; and

Solvent B: 90% ACN, 10% water, 0.1% trifluoroacetic Acid.

[00212] Preparative LC/MS (unless otherwise noted) with the following conditions Gradient: 25-100% B over 25 minutes, then a 5-minute hold at 100% B;

Column: Waters XBridge CI 8, 19 x 250 mm, 5-μιη particles;

Guard Column: Waters XBridge CI 8, 19 x 10 mm, 5-μιη particles;

Mobile Phase A: 5:95 acetonitrile:water with 10-mM ammonium acetate;

Mobile Phase B: 95:5 acetonitrile:water with 10-mM ammonium acetate;

Flow rate: 20 mL/min.

[00213] Analytical HPLC (unless otherwise noted) was performed to determine compound purity on a Shimadzu SIL-10A using the following method (Unless otherwis ' stated, retention times listed in Examples refer the retention times of Column 1):

Orthogonal Method:

Linear Gradient of 10% to 100% solvent B over 15 min;

UV visualization at 220 nm and 254 nm;

Column 1 : SunFire C18 3.5 μιη, 4.6x150mm;

Column 2: Xbridge Phenyl 3.5μιη, 4.6x150 mm;

Flow rate: 1 mL/min (for both columns);

Solvent A: 5% MeCN- 95% H 2 0- 0.05% TFA; and

Solvent B: 95% MeCN -5% H 2 O-0.05% TFA.

or

Zorbax method:

Linear Gradient of stated starting percentage to 100% solvent B over 8 min;

UV visualization at 220 nm;

Column: ZORBAX® SB C18 3.5 μιη, 4.6x75mm;

Flow rate: 2.5 mL/min;

Solvent A: 10% MeOH-90% H 2 O-0.2% H 3 P0 4 ; and Solvent B: 90% MeOH-10% H 2 O-00.2% H3PO4.

or

Analytical LC/MS Method:

Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B;

UV visualization at 220 nm;

Column: Waters Acquity UPLC BEH CI 8, 2.1 x 50 mm, 1.7-μιη particles;

Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; or 5:95 acetonitrile:water with 0.05% TFA;

Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; or 95:5 acetonitrile:water with 0.05% TFA;

Temperature: 50 °C;

Flow: 1.11 mL/min.

[00214] Preparatory chiral SFC chromatography (unless otherwise noted) was performed on a Berger Multigram II SFC chromatograph using the following method:

UV visualization at 220 nm;

Column: CHIRALPAK® AD-H SFC, 250 x 21 mm ID, 5 μιη;

Flow rate: 60.0 mL/min, 150 bar backpressure; and

Mobile Phase: 60/40, C0 2 /MeOH.

[00215] Analytical chiral SFC chromatography (unless otherwise noted) was performed on an Aurora Analytical SFC chromatography using the following method:

UV visualization at 220 nm;

Column: CHIRALPAK® AD-H, 250 x 4.6 mm ID, 5 μιη;

Flow rate: 3 mL/min, 150 bar backpressure; and

Mobile Phase: 60/40,CO 2 /MeOH.

NMR EMPLOYED ΓΝ CHARACTERIZATION OF EXAMPLES

[00216] in NMR spectra (unless otherwise noted) were obtained with JEOL® or Bruker FOURIER® transform spectrometers operating at 400 MHz or 500 MHz. l R-nOe experiments were performed in some cases for regiochemistry elucidation with a 400 MHz Bruker FOURIER® Transform spectrometer.

[00217] Spectral data are reported as chemical shift (multiplicity, number of hydrogens, coupling constants in Hz) and are reported in ppm (δ units) relative to either an internal standard (tetramethyl silane = 0 ppm) for !H NMR spectra, or are referenced to the residual solvent peak (2.49 ppm for CD 3 SOCD 2 H, 3.30 ppm for CD 2 HOD, 1.94 for CHD 2 CN, 7.26 ppm for CHC1 3 , 5.32 ppm for CDHC1 2 ).

Example 1

2-((4S,5S)-l-(4-((l-(2-Fluoro-5-methoxyphenyl)piperidin-4 -yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00218] 1A. l-(2-Fluoro-5-methoxyphenyl)piperidin-4-ol: A mixture of 2-bromo-l- fluoro-4-methoxybenzene (650 mg, 3.2 mmol), piperidin-4-ol (800 mg, 7.9 mmol) and 2- dicyclohexylphosphino-2',6'-dimethoxy-l,l '-biphenyl (52 mg, 0.13 mmol) in THF (3.2 mL) was purged with argon. Tris(dibenzylideneacetone)dipalladium (0) (58 mg, 0.063 mmol) was added and followed by lithium bis(trimethylsilyl)amide (1 N in THF, 15.2 mL, 15.2 mmol). The mixture was purged with argon at rt for several min and then heated at 70 °C for 2.5 h. The reaction mixture was diluted with aqueous NaHCC^ and was extracted with EtOAc. The organic extract was washed successively with H 2 0 and brine, and the organic layer was dried (MgSC^) and concentrated. Purification of the residue via silica gel chromatography gave the desired product as an oil (330 mg, 46% yield). LC- MS Anal. Calc'd for C 12 H 16 FN0 2 : 225.26, found [M+H] 226.2. NMR (400 MHz, CDC1 3 ) δ ppm 6.92 (1 H, dd, J=12.09, 8.79 Hz), 6.51 (1 H, dd, J=7.15, 3.30 Hz), 6.40 (1

H, dt, J=8.79, 3.02 Hz), 3.85 (1 H, td, J=8.52, 4.40 Hz), 3.76 (3 H, s), 3.27 - 3.46 (2 H, m), 2.74 - 2.91 (2 H, m), 1.95 - 2.14 (2 H, m), 1.75 (2 H, dtd, J=12.85, 9.10, 9.10, 3.57 Hz), 1.45 (1 H, d, J=4.40 Hz). [00219] IB. l-(2-Fluoro-5-methoxyphenyl)piperidin-4-yl 4-methylbenzenesulfonate: To l-(2-fluoro-5-methoxyphenyl)piperidin-4-ol (1.0 g, 4.4 mmol) and 4-methylbenzene- 1-sulfonyl chloride (1.7 g, 8.9 mmol) in CH 2 Ci2 (10 mL) at rt, pyridine (3.5 g, 44 mmol) was added dropwise. The reaction was stirred at rt for 16 h. The reaction mixture was diluted with EtOAc, and the organic layer was washed successively with Η 2 0 and brine. The resulting organic layer was dried (MgS0 4 ) and concentrated. Purification of the residue via silica gel chromatography gave the desired product (light yellow solid, 1.5 g, 87% yield). LC-MS Anal. Calc'd for C 19 H 22 FN0 4 S: 379.45, found [M+H] 380.1. l H NMR (400 MHz, CDC1 3 ) δ 7.91 - 7.76 (m, J=8.2 Hz, 2H), 7.40 - 7.30 (m, J=8.2 Hz, 2H), 6.91 (dd, J=12.1, 8.8 Hz, 1H), 6.52 - 6.33 (m, 2H), 4.79 - 4.58 (m, 1H), 3.75 (s, 3H), 3.22 (ddd, J=1 1.8, 7.4, 3.8 Hz, 2H), 2.91 (ddd, J=1 1.8, 7.4, 3.8 Hz, 2H), 2.46 (s, 3H), 2.05 - 1.84 (m, 4H).

[00220] 1C. N'-(4-Bromophenyl)-2,2,2-trifluoroacetohydrazide. To a stirred suspension of (4-bromophenyl)hydrazine hydrochloride (100 g, 447 mmol) in DCM (1500 mL) at rt was added dropwise a solution of TFAA (68.4 mL, 492 mmol) in DCM (150 mL). The reaction mixture was stirred at rt for 2 h and TFAA (45 mL, 324 mmol) was added slowly to the reaction mixture. After 5 min, no more solids remained. The reaction mixture was concentrated to -half volume, and the mixture was diluted with hexane (~1L), resulting in an off- white crystalline precipitate. The mixture was filtered and the solids were rinsed with hexanes. The resulting solid was dried under high-vac for 12 h at 50 °C to give '-(4-bromophenyl)-2,2,2-trifluoroacetohydrazide (103 g, 364 mmol, 81% yield) as an off-white crystalline compound. The mother liquor was concentrated and redissolved in DCM and 3 g of N'-(4-bromophenyl)-2,2,2- trifluoroacetohydrazide was added. The mixture was stirred for 20 min and hexane was added to give a suspension that was filtered and dried to give an additional 6.3 g (+4.96% yield) of N'-(4-bromophenyl)-2,2,2-trifluoroacetohydrazide as an off-white crystalline compound. LCMS: RT = 2.52 min (86.6%), m/z calc'd 282, 284, obs 305, 307 [M+Na]. !H NMR (400 MHz, CDC1 3 ) δ 8.10 (br. s., 1H), 7.46 - 7.33 (m, 2H), 6.79 - 6.67 (m, 2H), 6.06 (br. s., 1H).

[00221] ID. (Z)-N'-(4-Bromophenyl)-2,2,2-trifluoroacetohydrazonoyl chloride. To a solution of '-(4-bromophenyl)-2,2,2-trifluoroacetohydrazide (1 11 g, 393 mmol) in EtOAc (1500 mL) at rt was added dropwise benzenesulfonyl chloride (53.8 mL, 413 mmol). The mixture was cooled to 0 °C and Hunig's Base (72.8 mL, 413 mmol) was added dropwise. The resulting mixture was stirred at rt overnight. The resulting mixture was diluted with EtOAc and the organic layer was washed with water (3 x 2L). The combined organic extracts were dried over sodium sulfate, filtered and concentrated to give (Z)-N'-(4-bromophenyl)-2,2,2-trifluoroacetohydrazonoyl chloride (1 19 g, 393 mmol, 100% yield) as a yellow liquid, which was used for next step without further purification. Analytical HPLC: RT = 3.93 min (83.1%). !H MR (400 MHz, CDC1 3 ) δ 8.00 (br. s., 1H), 7.51 - 7.36 (m, 2H), 7.11 - 6.94 (m, 2H).

[00222] IE. (S,E)-3-But-2-enoyl-4-phenyloxazolidin-2-one. To a solution of (S)-(+)-4- phenyl-2-oxazolidinone (84.4 g, 517 mmol), lithium chloride (22.81 g, 538 mmol) and triethylamine (79 mL, 564 mmol) in THF (1200 mL) in a 5L 3 -neck flask at -5 °C was added crotonic anhydride (76 mL, 512 mmol) over 5 min. The reaction temperature increased slightly during the addition. The mixture was allowed to warm to rt, resulting in a white milky solution. The reaction mixture was stirred at rt overnight. The reaction was quenched via addition of 0.2M aqueous HC1 (600 mL) to give an aqueous layer of pH~7. The two layers were separated and the aqueous layer was extracted with EtOAc (400 mL). The combined organic layers were washed with brine, dried over a 2 S04 and concentrated to give an light brown oil (200 mL). To the oil was added MeOH (200 mL). Crystals formed slowly at the beginning and continued to form over 15 min. The product was collected by filtration and the solids were rinsed with MeOH (200 mL) to give white solid. More product was collected from the MeOH filtrate, and the materials were combined and dried to give 101.6 g of (S,E)-3-But-2-enoyl-4-phenyloxazolidin-2-one as white crystals.

[00223] IF. (S)-3-((4S,5R)-l-(4-Bromophenyl)-4-methyl-3-(trifluoromethyl )-4,5- dihydro-lH-pyrazole-5-carbonyl)-4-phenyloxazolidin-2-one. To A solution of (Z)-N'-(4- bromophenyl)-2,2,2-trifluoroacetohydrazonoyl chloride (94.3 g, 313 mmol) and (S,E)-3- but-2-enoyl-4-phenyloxazolidin-2-one (65.1 g, 282 mmol) in 1,4-dioxane (782 mL) under 2 atm was added silver carbonate (103 g, 375 mmol) and the mixture was stirred at 50 °C overnight. The reaction mixture was filtered through CELITE®, and the filter cake was washed with EtOAc. The filtrate was concentrated to give an oil, which was purified via silica gel chromatography to give a yellow oil. The residue was purified via crystallization from hexanes-ether (7;3) to give (S)-3-((4S,5R)-l-(4-bromophenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazole-5-carbony l)-4-phenyloxazolidin-2- one (72 g, 145 mmol, 46.4% yield) as a white crystalline compound. l R NMR (400 MHz, CDC1 3 ) δ 7.47 - 7.31 (m, 5H), 7.29 - 7.20 (m, 3H), 6.93 - 6.81 (m, 2H), 5.81 (d, J=2.2 Hz, 1H), 5.39 (dd, J=8.8, 4.2 Hz, 1H), 4.82 (t, J=9.0 Hz, 1H), 4.43 (dd, J=9.2, 4.2 Hz, 1H), 3.16 (dd, J=7.0, 1.5 Hz, 1H), 1.50 (d, J=7.0 Hz, 3H).

[00224] 1G. ((4S,5R)-l-(4-Bromophenyl)-4-methyl-3-(trifluoromethyl)-4,5- dihydro- lH-pyrazol-5-yl)methanol. To a solution of (S)-3-((4S,5R)-l-(4-bromophenyl)-4-methyl- 3-(trifluoromethyl)-4,5-dihydro-lH-pyrazole-5-carbonyl)-4-ph enyloxazolidin-2-one (93.8 g, 189 mmol) in THF (1420 mL) was added a solution of NaBH 4 (7.15 g, 189 mmol) in water (56.8 mL) in two portions. The first portion was slowly added over 5 min and the internal temperature raised from 20.5 °C to 28.9 °C, and the second portion was added more rapidly. The resulting mixture was stirred at rt for 4 h. The reaction mixture was cooled to 2 °C, and 10% KHS0 4 (180 mL) was added while maintaining internal temperature < 10 °C. The resulting mixture was stirred for 30 min and the mixture was evaporated. The residue was diluted with EtOAc (900 mL) and water (900 mL). The layers were extracted and the organic layer was washed with brine. The organic layer was dried over a2S0 4 and the solution was filtered and evaporated to give 102 g of a white semi-solid that was purified via silica gel chromatography to give 85 g of ((4S,5R)- l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-l H-pyrazol-5-yl)methanol as a white solid. LCMS: RT = 3.64 min (93.6%), m/z calc'd 336, 338, obs 337, 339

[M+H]. !H NMR (400 MHz, CDC1 3 ) δ 7.46 - 7.35 (m, 2H), 7.10 - 6.95 (m, 2H), 4.07 (q,

J=4.6 Hz, 1H), 3.90 - 3.69 (m, 2H), 3.55 - 3.39 (m, 1H), 1.63 - 1.49 (m, 1H), 1.36 (dd, J=7.2, 0.6 Hz, 3H).

[00225] 1H. ((4S,5R)-l-(4-Bromophenyl)-4-methyl-3-(trifluoromethyl)-4,5- dihydro- lH-pyrazol-5-yl)methyl methanesulfonate. To a cooled (0-5 °C) solution of ((4S,5R)-1- (4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH- pyrazol-5-yl)methanol (85 g, 252 mmol) in THF (750 mL) was added triethylamine (70.3 mL, 504 mmol) and methanesulfonyl chloride (22.59 mL, 290 mmol). A white precipitate was observed. The mixture was stirred at rt for 30 min, and the reaction was judged complete as monitored by LCMS. The mixture was diluted with EtOAc (250 mL) and sat'd aqueous NaHCC^. The layers were extracted, and the aqueous layer was back extracted with EtOAc. The combined organic layers were washed successively with water and brine. The organic layer was dried over MgS0 4 , and the solution was filtered and evaporated to give

((4S,5R)-l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4 ,5-dihydro-lH-pyrazol-5- yl)methyl methanesulfonate (103 g, 248 mmol, 98% yield). LCMS: r.t. 3.60 min (94.7%), m/z calc'd 414, 416, obs 415, 417 [M+H].

[00226] II. 2-((4S,5S)-l-(4-Bromophenyl)-4-methyl-3-(trifluoromethyl)-4, 5-dihydro- lH-pyrazol-5-yl)acetonitrile. A suspension of KCN (9.89 g, 147 mmol) in DMSO (700 mL) was heated to 60 °C for 1 h and cooled to 40 °C. To this mixture was added a solution of ((4S,5R)-l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4,5- dihydro-lH- pyrazol-5-yl)methyl methanesulfonate (51 g, 123 mmol) in 50 mL of DMSO. The mixture was heated at 40 °C for 9 h and the reaction mixture was cooled in an ice bath and stirred overnight. The reaction mixture was diluted with ice (450 g), EtOAc (500 mL), and sat'd aqueous NaHCC^ solution (250 mL). The mixture was stirred for 15 min and the layers were separated. The aqueous layer was extracted with EtOAc (200 mL) and the combined organic layers were washed with brine. The organic layer was dried over MgS0 4 , filtered and evaporated to give 2-((4S,5S)-l-(4-bromophenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (39 g, 1 13 mmol, 92% yield) as a slight reddish oil. LCMS: RT = 3.55 min (88.3%), m/z calc'd 345, 347, obs 368, 380 [M+Na]. !H NMR (400 MHz, CDC1 3 ) δ 7.53 - 7.39 (m, 2H), 7.07 - 6.92 (m, 2H), 4.36 -

4.24 (m, 1H), 3.56 - 3.31 (m, 1H), 2.78 (dd, J=16.9, 3.5 Hz, 1H), 2.50 (dd, J=16.9, 9.0 Hz, 1H), 1.48 - 1.36 (m, 3H).

[00227] U. Methyl 2-((4S,5S)-l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4, 5- dihydro-lH-pyrazol-5-yl)acetate: To a cooled (0 °C) solution of methanol (1L) and dichloromethane (1.5L) was added acetyl chloride (0.33L, 4.7 mol) over 45 min. The resulting mixture was stirred for lh, and 2-((4S,5S)-l-(4-bromophenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (90 g, 0.26 mol) was added, and the mixture was stirred for 2 h at rt. The reaction mixture was evaporated in vacuo, and the residue was treated with a solution of HCl/methanol/dichloromethane as generated above. The reaction mixture was stirred overnight at rt. The reaction mixture was evaporated in vacuo, and the residue was dissolved in HCl/methanol that was formed separately by mixing methanol with acetyl chloride as above (1L methanol treated with 0.33L of acetyl chloride at 0 °C for 45 min). The reaction mixture was stirred at rt overnight, and was heated at 40 °C for 1 h. The reaction mixture was diluted with 1 L of acetonitrile, and the mixture was evaporated in vacuo. The residue was dissolved in ethyl acetate, and the mixture was washed successively with 2x sat'd aqueous NaHCC^ and brine. The combined organic layers were dried over a 2 S03, the mixture was filtered, and the solution was evaporated. The residue was purified via silica chromatography to give 80.5 g of methyl 2-((4S,5S)-l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4, 5- dihydro-lH-pyrazol-5-yl)acetate as a light yellow oil. LC-MS Anal. Calc'd for

C 14 H 14 BrF3 202: 378.02, found [M+H] 379.5, 381.5. !pi MR (400 MHz, CDC1 3 ) δ

7.43 - 7.37 (m, 2 H), 7.01 - 6.94 (m, 2 H), 4.45 - 4.37 (m, 1 H), 3.72 (s, 3 H), 3.23 - 3.15 (m, 1 H), 2.79 (dd, J= 3.0, 16.3 Hz, 1 H), 2.41 (dd, J= 10.3, 16.3 Hz, 1 H), 1.34 (d, J = 7.0 Hz, 3 H). 13 C MR (300MHz, CDC1 3 ) δ 170.57, 142.50, 132.29, 129.58, 123.02,

119.45, 115.78, 1 13.60, 77.43, 65.06, 52.08, 45.52, 34.81, 17.70. 19 F-NMR (400 MHz, CDC1 3 ) 5 -63.25

[00228] IK. Methyl 2-((4S,5S)-4-methyl-l-(4-(4,4,5,5-tetramethyl-l,3,2-dioxabor olan- 2-yl)phenyl)-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl )acetate: A mixture of methyl 2-((4S,5S)-l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)-4, 5-dihydro-lH- pyrazol-5-yl)acetate (1.517 g, 4.0 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2- dioxaborolane) (1.63 g, 6.4 mmol), potassium acetate (1.26 g, 12.8 mmol) and 1,1 '-

(diphenylphosphino)ferrocene)palladium dichloride (0.16 g, 0.20 mmol) in DMF (8 mL) was purged with argon. The reaction mixture was heated at 80 °C for 2 h. The reaction mixture was diluted with EtOAc and the organic layer was washed successively with H2O and brine. The resulting organic layer was dried (MgS0 4 ) and concentrated.

Purification of the residue via silica gel chromatography gave the desired product as a gum (2.36 g). The product was used without further purification.

[00229] 1L. Methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3-(trifluoromethyl)- 4,5- dihydro-lH-pyrazol-5-yl)acetate: To a solution of methyl 2-((4S,5S)-4-methyl-l-(4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)-3-(trif luoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate in ethyl acetate (75 mL), was added hydrogen peroxide (30 wt.% in water, 15 mL) dropwise. The reaction mixture was stirred at rt. After 1 h, additional hydrogen peroxide solution (6.5 mL) and 15 mL EtOAc were added. After the mixture was stirred at rt for 7 h, the reaction mixture was cooled with an ice-water bath and the reaction was slowly quenched with aqueous sodium sulfite. The mixture was extracted with EtOAc. The organic extracts were washed with brine, dried (MgS0 4 ), filtered and concentrated. Purification via silica gel chromatography gave the desired product as an oil (1.1 g, 85% yield). LC-MS Anal. Calc'd for C 14 H 1 5F3 2 03: 316.28, found [M+H] 317.2.

[00230] 1M. Methyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5-methoxyphenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate: To 1 - (2-fluoro-5-methoxyphenyl)piperidin-4-yl 4-methylbenzenesulfonate (1.22 g, 2.96 mmol) and methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro- lH-pyrazol-5-yl)acetate (0.94 g, 3.0 mmol) in DMF (7 mL), cesium carbonate (2.22 g, 6.81 mmol) was added. The reaction mixture was heated at 60 °C for 16 h. The reaction mixture was cooled to rt and treated with methyl iodide (698 mg, 4.92 mmol) and stirred at rt for 1 h. The reaction mixture was diluted with EtOAc, washed with H2O, brine, dried (MgS0 4 ) and concentrated. Purification via silica gel chromatography gave the desired product as a gum (660 mg, 42% yield). LC-MS Anal. Calc'd for C 2 6H29F 4 3 0 4 : 523.52, found [M+H] 524.2. ¾ NMR (400 MHz, CDC1 3 ) δ 7.12 - 6.99 (m, 2H), 6.99 - 6.83 (m, 3H), 6.53 (dd, J=7.1, 3.3 Hz, 1H), 6.41 (dt, J=8.8, 3.3 Hz, 1H), 4.38 (tt, J=7.1, 3.6 Hz, 2H), 3.81 - 3.73 (m, 3H), 3.72 - 3.67 (m, 3H), 3.33 (ddd, J=11.4, 7.6, 3.6 Hz, 2H), 3.26 - 3.12 (m, 1H), 2.97 (ddd, J=1 1.7, 8.1, 3.3 Hz, 2H), 2.80 (dd, J=15.9, 3.3 Hz, 1H), 2.49 - 2.33 (m, 1H), 2.19 - 2.04 (m, 2H), 2.04 - 1.87 (m, 2H), 1.34 (d, J=6.6 Hz, 3H).

[00231] Example 1 : To a solution of methyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluo romethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate (533 mg, 1.02 mmol) in THF (20 mL) was added lithium hydroxide (1 N in water, 1.43 mL, 1.43 mmol) and the mixture was stirred at rt. After 16 h, lithium hydroxide (1 N, 2.64 mL, 2.64 mmol) was added. The reaction mixture was stirred at rt for 3 h. The reaction mixture was cooled with an ice-water bath and the solution was acidified with IN HC1. The mixture was extracted with CH2CI2. The organic extracts were dried (MgS0 4 ), filtered and concentrated, and the residue was purified via RP-Prep HPLC. The product-containing fractions were evaporated, and the residue was treated with CH 3 CN and IN HC1, and the mixture was lyophilized to give 2-((4S,5S)-l- (4-(( 1 -(2-fluoro-5-methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-meth yl-3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1 as a light yellow solid (480 mg, 84% yield). LC-MS Anal. Calc'd for C 2 5H2 7 F 4 304: 509.19, found [M+H] 510.2. !H MR (500 MHz, CD 3 CN) δ ppm 7.13 - 7.07 (m, 2H), 7.07 - 7.03 (m, 1H), 7.01 - 6.96 (m, 2H), 6.74 (dd, J=7.2, 3.0 Hz, 1H), 6.61 (dt, J=8.9, 3.3 Hz, 1H), 5.67 (br.s, 1H), 4.53 - 4.47 (m, 1H), 4.47 - 4.42 (m, 1H), 3.79 (s, 3H), 3.46 (ddd, J=l 1.6, 7.6, 3.6 Hz, 2H), 3.40 - 3.32 (m, 1H), 3.11 (ddd, J=l 1.9, 8.2, 3.3 Hz, 2H), 2.75 (dd, J=16.5, 3.0 Hz, 1H), 2.52 (dd, J=16.5, 9.6 Hz, 1H), 2.21 - 2.13 (m, 2H), 2.00 (s, 1H), 1.96 - 1.89 (m, 2H), 1.33 (d, J=7.2 Hz, 3H) Analytical HPLC (Orthogonal method): RT = 9.1 min, HI: 97%.

hGPR40 EC50 = 114 nM. hGPR40 IP1 EC 50 = 17 nM. Acute oral glucose tolerance: - 47% @ 0.3 mg/kg.

Example 8

2-((4S,5S)-l-(4-((l-(2,3-Difluoro-5-methoxyphenyl)-3-meth ylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid, HC1 (Isomer 1 and Isomer 2)

[00232] 8A. 4-((tert-Butyldimethylsilyl)oxy)-l-(2,3-difluoro-5-methoxyph enyl)-3- methylpiperidine: A mixture of l,2,3-trifluoro-5-methoxybenzene (530 mg, 3.3 mmol) and 4-((tert-butyldimethylsilyl)oxy)-3-methylpiperidine (300 mg, 1.3 mmol) in DMSO (3 mL) was heated at 140 °C for 20 h. The reaction mixture was quenched with aqueous aHC03 and extracted with EtOAc. The organic extract was washed with ¾0 and brine, dried ( a2S0 4 ) and concentrated. Purification via silica gel chromatography gave 4- ((ter?-butyldimethylsilyl)oxy)-l-(2,3-difluoro-5-methoxyphen yl)-3-methylpiperidine as a yellow oil (230 mg, 47% yield). LC-MS Anal. Calc'd for 371.21, found [M+H] 372.1. [00233] 8B. l-(2,3-Difluoro-5-methoxyphenyl)-3-methylpiperidin-4-ol: To a solution of cw-4-((?ert-butyldimethylsilyl)oxy)-l-(2,3-difluoro-5-methox yphenyl)-3- methylpiperidine (230 mg, 0.61 mmol) in THF (lmL) was added 1M

tetrabutylammonium fluoride (3 mL, 3 mmol) and stirred at rt for 8 h. The reaction mixture was quenched with aqueous aHC03 and extracted with EtOAc. The organic extract was washed with H 2 0 and brine, dried ( a 2 S04) and concentrated. Purification via silica gel chromatography gave l-(2,3-difluoro-5-methoxyphenyl)-3-methylpiperidin-

4- ol as an oil (105 mg, 65% yield). LC-MS Anal. Calc'd for C 13 H 17 F 2 N0 2 257.12, found [M+H] 258.0.

[00234] 8C. Methyl 2-((4S,5S)-l-(4-(((3R,4R)-l-(2,3-difluoro-5-methoxyphenyl)-3 - methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol-

5- yl)acetate: To triphenylphosphine (140 mg, 0.53 mmol) in DMF (4 mL)) was added (E)-diethyl diazene-l,2-dicarboxylate (0.072 mL, 0.46 mmol) and methyl 2-((4S,5S)-l- (4-hydroxyphenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-l H-pyrazol-5-yl)acetate (1J, 120 mg, 0.38 mmol) and then the mixture was stirred for 5 min followed by the addition of l-(2,3-difluoro-5-methoxyphenyl)- 3-methylpiperidin-4-ol (98 mg, 0.38 mmol) and then stirred at rt overnight. The reaction mixture was quenched with aqueous aHC03 and extracted with EtOAc. The organic extract was washed with H 2 0 and then brine, dried (Na 2 S0 4 ) and concentrated. Purification via silica gel chromatography gave methyl 2-((4S,5S)-l -(4-(((3R,4R)-l -(2,3 -difluoro-5-methoxyphenyl)-3 -methylpiperidin- 4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH -pyrazol-5-yl)acetate as an oil (40 mg, 19% yield). LC-MS Anal. Calc'd for C 27 H3 0 F 5 3 O4 555.22, found [M+H]

556.0.

[00235] Example 8 (Isomer 1) and (Isomer 2): To a stirred solution of methyl 2- ((4S,5S)-l-(4-(((3R,4R)-l-(2,3-difluoro-5-methoxyphenyl)-3-m ethylpiperidin-4- yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5 -dihydro- 1 H-pyrazol-5 -yl)acetate (55 mg, 0.099 mmol) in THF (1 mL) and H 2 0 (0.10 mL) was added 2M lithium hydroxide (0.074 mL, 0.15 mmol). The mixture was stirred at rt for 3 h and then diluted with EtOAc and acidified with 6N HC1. The organic layer was dried (Na 2 S0 4 ) and concentrated. They were separated by to provide 4 isomers. The resulting product was separated via chiral Prep. SFC to yield two diastereomers as yellow oil: 2-((4S,5S)-l-(4-((l-(2,3- difluoro-5 -methoxyphenyl)-3 -methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1 (1 1 mg, 19% yield, Isomer 1): LC-MS Anal. Calc'd for C26H28F5 3O4 541.20, found [M+H] 542.2. ¾ NMR (500MHz, CDC1 3 ) δ 7.13 - 6.99 (m, 2H), 6.96 - 6.85 (m, 2H), 6.40 - 6.15 (m, 2H), 4.45 - 4.32 (m, 1H), 3.94 - 3.81 (m, 1H), 3.74 (s, 3H), 3.48 - 3.39 (m, 2H), 3.26 - 3.18 (m, 1H), 2.85 (dd, J=16.4, 2.9 Hz, 2H), 2.65 - 2.58 (m, 1H), 2.49 - 2.39 (m, 1H), 2.20 - 2.09 (m, 2H), 1.86 - 1.75 (m, 1H), 1.35 (d, J=7.2 Hz, 3H), 1.12 (d, J=6.6 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 11.7 min, HI: 95%. hGPR40 EC50 = 796 nM. 2-((4S,5S)-l-(4-((l-(2,3-difluoro-5-methoxyphenyl)-3-methylp iperidin- 4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH -pyrazol-5-yl)acetic acid, HC1 (16 mg, 26% yield, Isomer 2): LC-MS Anal. Calc'd for C26H28F5 3O4 541.20, found [M+H] 542.2. ¾ NMR (500MHz, CDC1 3 ) δ 7.13 - 6.99 (m, 2H), 6.96 - 6.85 (m, 2H), 6.40 - 6.15 (m, 2H), 4.45 - 4.32 (m, 1H), 3.94 - 3.81 (m, 1H), 3.74 (s, 3H), 3.48 - 3.39 (m, 2H), 3.26 - 3.18 (m, 1H), 2.85 (dd, J=16.4, 2.9 Hz, 2H), 2.65 - 2.58 (m, 1H), 2.49 - 2.39 (m, 1H), 2.20 - 2.09 (m, 2H), 1.86 - 1.75 (m, 1H), 1.35 (d, J=7.2 Hz, 3H), 1.12 (d, J=6.6 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 1 1.7 min, HI: 95%. hGPR40 EC 50 = 237 nM.

Example 9

2-((4S,5S)-l-(4-((l-(2-Fluoro-5-isopropoxyphenyl)piperidi n-4-yl)oxy)phenyl)-4-methyl- -(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00236] 9A. l-(2-Fluoro-5-isopropoxyphenyl)piperidin-4-ol: To l,2-difluoro-4- isopropoxybenzene (950 mg, 5.52 mmol) and piperidin-4-ol (1.1 g, 1 1 mmol) was added DMSO (7 mL). The mixture was heated at 140 °C overnight. The reaction mixture was quenched with aqueous NaHC03 and extracted with EtOAc. The organic extract was washed with ¾0 and then brine, dried (Na2S0 4 ) and concentrated. Purification via silica gel chromatography gave l-(2-fluoro-5-isopropoxyphenyl)piperidin-4-ol as a yellow oil (666 mg, 70% yield). LC-MS Anal. Calc'd for C 14 H 20 FNO 2 253.15, found [M+H] 254.2.

[00237] 9B. Methyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5-isopropoxyphenyl)piperidin-4 - yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetate: A mixture of triphenylphosphine (123 mg, 0.47 mmol) and (E)-diethyl diazene-1,2- dicarboxylate (0.064 mL, 0.40 mmol) in DMF (0.5 mL) was stirred for 5 min. and methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5- yl)acetate (1J, 101 mg, 0.32 mmol) in DMF (0.2 mL) was added, followed by the addition of l-(2-fluoro-5-isopropoxyphenyl)piperidin-4-ol (85 mg, 0.34 mmol) in DMF (0.25 mL). The reaction mixture was stirred at rt overnight. The reaction mixture was quenched with aqueous aHC03 and extracted with EtOAc. The organic extract was washed with H2O and then brine, dried ( a2S0 4 ) and concentrated. Purification via silica gel

chromatography gave methyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5- isopropoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3-(trif luoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate as an oil (62 mg, 33% yield). LC-MS Anal. Calc'd for

C28 H 33 F 4 N 3°4 551.24, found [M+H] 552.0.

[00238] Example 9 (white solid, 13 mg) was prepared following the same procedure for Example 8. LC-MS Anal. Calc'd for C 27 H 31 F4 304 537.23, found [M+H] 537.9. l H NMR (500MHz, CDC1 3 ) δ 7.07 (d, J=9.1 Hz, 4H), 6.93 (d, J=9.1 Hz, 2H), 6.75 - 6.65 (m, 1H), 4.57 - 4.51 (m, 1H), 4.50 - 4.43 (m, 1H), 4.42 - 4.37 (m, 1H), 3.80 - 3.69 (m, 2H), 3.38 - 3.29 (m, 2H), 3.27 - 3.17 (m, 1H), 2.88 - 2.80 (m, 1H), 2.50 - 2.42 (m, 1H), 2.40 - 2.30 (m, 2H), 2.18 - 2.08 (m, 2H), 1.40 - 1.28 (m, 10H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 10.4 min, HI: 95%. hGPR40 EC 50 = 374 nM. Example 10

2-((4S,5S)-l-(4-((l-(5-Ethoxy-2,3-difluorophenyl)-3-methylpi peridin-4-yl)oxy)phenyl)- 4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)ace tic acid, HCl (Isomer 1 and Isomer 2)

[00239] 1 OA. as-4-((tert-butyldimethylsilyl)oxy)- 1 -(5-ethoxy-2,3 -difluorophenyl)-3 - methylpiperidine: A mixture of 5-ethoxy- 1,2,3 -trifluorobenzene (403 mg, 2.29 mmol) and c«-4-((?ert-butyldimethylsilyl)oxy)-3-methylpiperidine (350 mg, 1.53 mmol) in DMSO (2 mL) was heated at 140 °C for 18 h. The reaction mixture was quenched with H2O and extracted with EtOAc. The organic extract was washed with H2O and then brine, dried ( a 2 S0 4 ) and concentrated. Purification via silica gel chromatography gave cis-4-((tert- butyldimethylsilyl)oxy)-l-(5-ethoxy-2,3-difluorophenyl)-3-me thylpiperidine as a yellow oil (340 mg, 58% yield). LC-MS Anal. Calc'd for C 2 oH33F 2 0 2 Si 385.22, found [M+H] 386.0.

[00240] Example 10 (Isomer 1 and Isomer 2): Isomer 1 (white solid, 13mg) and Isomer 2 (white solid, 17 mg) were prepared following the procedure for Example 8. 2-((4S,5S)- l-(4-((l-(5-Ethoxy-2,3-difluorophenyl)-3-methylpiperidin-4-y l)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1 (Isomer 1): LC-MS Anal. Calc'd for C 27 H3 0 F 5 3O4 555.22, found [M+H] 555.9. l H NMR (500 MHz, CDC1 3 ) δ

7.04-7.09 (m, 2H), 6.85-6.91 (m, 2H), 6.22-6.33 (m, 2H), 4.31-4.37 (m, 1H), 3.94 (q, J=7.1 Hz, 2H), 3.82 (d, J=3.8 Hz, 1H), 3.38-3.47 (m, 2H), 3.22-3.31 (m, 1H), 2.89 (q, J=7.4 Hz, 3H), 2.81 (br. s., 1H), 2.72 (dd, J=15.5, 2.9 Hz, 1H), 2.59 (dd, J=12.2, 9.5 Hz, 1H), 2.29 (dd, J=15.4, 10.2 Hz, 1H), 2.08-2.18 (m, 2H), 1.75-1.85 (m, 1H), 1.39 (t, J=7.0 Hz, 3H), 1.21 (d, J=6.0 Hz, 1H), 1.11 (d, J=6.6 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 11.7 min, HI: 97%. hGPR40 EC 50 = 1203 nM. 2- ((4S,5S)-l-(4-((l -(5-ethoxy -2, 3-difluorophenyl)-3-methylpiperidin-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HC1 (Isomer 2): LC- MS Anal. Calc'd for C 27 H3 0 F 5 3 O4 555.2, found [M+H] 555.9. !H MR (500 MHz, CDCI3) δ 7.04-7.09 (m, 2H), 6.85-6.91 (m, 2H), 6.22-6.33 (m, 2H), 4.31-4.37 (m, 1H), 3.94 (q, J=7.1 Hz, 2H), 3.82 (d, J=3.8 Hz, 1H), 3.38-3.47 (m, 2H), 3.22-3.31 (m, 1H), 2.89 (q, J=7.4 Hz, 3H), 2.81 (br. s., 1H), 2.72 (dd, J=15.5, 2.9 Hz, 1H), 2.59 (dd, J=12.2, 9.5 Hz, 1H), 2.29 (dd, J=15.4, 10.2 Hz, 1H), 2.08-2.18 (m, 2H), 1.75-1.85 (m, 1H), 1.39 (t, J=7.0 Hz, 3H), 1.21 (d, J=6.0 Hz, 1H), 1.1 1 (d, J=6.6 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 1 1.7 min, HI: 97%. hGPR40 EC 50 =

315 nM. Example 12

2-((4S,5S)-l-(4-((l-(2-Fluoro-5-isopropoxyphenyl)-3-methylpi peridin-4-yl)oxy)phenyl)- -methyl-3 -(trifluoromethyl)-4,5 -dihydro- 1 H-pyrazol-5-yl)acetic acid, HC1.

[00241] 12A. 4-((tert-Butyldimethylsilyl)oxy)-l-(2-fluoro-5-isopropoxyphe nyl)-3- methylpiperidine: A mixture of 4-((tert-butyldimethylsilyl)oxy)-3 -methylpiperidine (250 mg, 1.09 mmol), l,2-difluoro-4-isopropoxybenzene (225 mg, 1.30 mmol) and sodium bicarbonate (92 mg, 1.09 mmol) in DMSO (1 mL) was heated at 140 °C for 28 h. The reaction mixture was quenched with H 2 0 and extracted with EtOAc. The organic extract was washed with H 2 0 and then brine, dried ( a 2 S04) and concentrated. Purification via silica gel chromatography gave the desired product as an oil (50 mg, 12.0% yield). LC- MS Anal. Calc'd for C 2 iH 36 FN0 2 Si 381.25, found [M+H] 382.0.

[00242] 12B. l-(2-Fluoro-5-isopropoxyphenyl)-3-methylpiperidin-4-ol: To a solution of 4-((tert-butyldimethylsilyl)oxy)-l-(2-fluoro-5-isopropoxyphe nyl)-3-methylpiperidine (80 mg, 0.21 mmol) in THF (1 mL) was added 1M tetrabutylammonium fluoride (1.0 mL, 1.04 mmol) and the mixture was stirred at rt for 28 h. The reaction mixture was quenched with aqueous NH 4 C1 and extracted with EtOAc. The organic extract was washed with H 2 0 and then brine, dried (Na 2 S0 4 ) and concentrated. Purification via silica gel chromatography gave the desired product as a yellow oil (25 mg, 45% yield). LC-MS Anal. Calc'd for C 15 H 22 FN0 2 267.16, found [M+H] 268.0. [00243] 12C. Methyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5-isopropoxyphenyl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetate: A mixture of methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetate (1J, 30 mg, 0.09 mmol), l-(2- fluoro-5-isopropoxyphenyl)-3-methylpiperidin-4-ol (25 mg, 0.09 mmol) and

triphenylphosphine (37 mg, 0.14 mmol) in THF (0.8 mL) was sonicated for 10 min. before (E)-di-tert-butyl diazene-l,2-dicarboxylate (32 mg, 0.14 mmol) was added. The mixture was stirred at rt overnight. The reaction mixture was quenched with aqueous aHC03 and extracted with EtOAc. The organic extract was washed with H2O and then brine, dried ( a2S0 4 ) and concentrated. Purification via silica gel chromatography gave the desired product as a yellow oil (26 mg, 49% yield). LC-MS Anal. Calc'd for C29H35F4N3O4 565.26, found [M+H] 566.0.

[00244] Example 12: To a suspension of methyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5- isopropoxyphenyl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methy l-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetate (26 mg, 0.05 mmol) in THF (0.25 mL) and H 2 0 (0.03 mL) was added 2M lithium hydroxide (0.04 mL, 0.07 mmol) and the mixture was stirred at rt for 3 h. The reaction was diluted with EtOAc and acidified with 6N HCl. The organic layer was collected and the aqueous layer was extracted with CH 2 Cl2 twice. The combined organic was dried ( a 2 S04) and concentrated. The resulting product was separated via chiral Prep. SFC to yield 2-((4S,5S)-l-(4-((l-(2-fluoro-5- isopropoxyphenyl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methy l-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HCl (oil, 6 mg, 21%). LC-MS Anal. Calc'd for C 28 H 33 F 4 N 3°4 : 55 1 · 2 > found [ M+H ] 5 52 · 1 · ^ NMR (500 MHz, CDCI3) § ppm 7.05 (d, J=8.8 Hz, 2H), 6.83-6.97 (m, 3H), 6.50 (dd, J=7.4, 3.0Hz, 1H), 6.30-6.44 (m, 1H), 4.32- 4.51 (m, 2H), 3.83 (d, J=3.8 Hz, 1H), 3.36-3.47 (m, 2H), 3.12-3.27 (m, 1H), 2.72-2.89 (m, 2H), 2.56 (dd, J=12.0, 9.8 Hz, 1H), 2.45 (dd, J=16.4, 10.3 Hz, 1H), 2.09-2.21 (m, 2H), 1.73-1.90 (m, 1H), 1.29-1.37 (m, 9H), 1.22-1.27 (m, 2H), 1.1 1 (d, J=6.6 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 11.2 min, HI: 99%. hGPR40 EC50 = 31 1 nM. hGPR40 IP1 EC 50 = 253 nM.

Example 16 2-((4S,5S)-l-(4-((l-(2,3-Difluoro-5-methoxyphenyl)piperidin- 4-yl)oxy)phi

methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)ac etic acid, HC1

[00245] 16A. l-(2,3-Difluoro-5-methoxyphenyl)piperidin-4-ol: To l,2,3-trifluoro-5- methoxybenzene (1 g, 6.17 mmol) was added piperidin-4-ol (1.87 g, 18.51 mmol) and DMSO (10 mL). The reaction was capped and placed in a 140 °C oil bath for 3 h. The reaction mixture was filtered and the solids rinsed into the filtrate with 2 mL of EtOAc. The filtrate was added to 50 mL of water and 50 mL of EtOAc, the mixture was well stirred, allowed to settle, and then the lower aqueous layer was removed. The EtOAc layer was washed with an additional 3 x 50 mL of water. The EtOAc layer was passed through a 30 mm id x 40 mm silica gel plug using a total of 200 mL of EtOAc. The filtrate was concentrated in vacuo to give the product as a tan solid (1.02 g, 68% yield). LC-MS Anal. Calc'd for C 12 H 15 F 2 02 243.1 1, found [M+H] 244.1. NMR (500MHz, CDC1 3 ) δ 6.33 (dd, J=5.8, 3.0 Hz, 1H), 6.27 - 6.17 (m, 1H), 3.97 - 3.80 (m, 1H), 3.75 (s, 3H), 3.53 - 3.19 (m, 2H), 3.02 - 2.78 (m, 2H), 2.04 (d, J=9.9 Hz, 2H), 1.84 - 1.69 (m, 2H), 1.61 - 1.40 (m, 1H).

[00246] 16B. Methyl 2-((4S,5S)-l-(4-((l-(2,3-difluoro-5-methoxyphenyl)piperidin- 4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate: To a solution of triphenylphosphine (107 mg, 0.407 mmol) and DMF (1 mL) was added (E)- diethyl diazene-l,2-dicarboxylate (0.055 mL, 0.349 mmol) and the mixture was stirred at rt for 5 min. This mixture was added to the thick amber oil methyl 2-((4S,5S)-l-(4- hydroxyphenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetate (U, 92 mg, 0.291 mmol) and the mixture was stirred for 5 minutes. To this amber solution was added l-(2,3-difluoro-5-methoxyphenyl)piperidin-4-ol (77 mg, 0.317 mmol) and the mixture was stirred for 5 hours. The reaction mixture was quenched with 5 mL of water and 20 mL of EtOAc. The organic layer was washed with 3 x 5 mL of additional water and then the solvent from the organic layer was removed in vacuo. The residue was purified via silica gel chromatography to give the desired product as a pale amber oil (39 mg, 24% yield). LC-MS Anal. Calc'd for C 26 H 28 F 5 304 541.20, found [M+H] 542.2.

[00247] Example 16: To methyl 2-((4S,5S)-l-(4-((l-(2,3-difluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluo romethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate (39 mg, 0.072 mmol) and THF (1 mL) added water (0.1 mL) and then lithium hydroxide (2 M aqueous) (0.090 mL, 0.18 mmol). After 3 h the reaction mixture was added to 10 mL of EtOAc and then 5 mL of hexane was added. The mixture was shaken and then allowed to settle. An emulsion had formed. 2-3 mL of the upper hexane layer were removed leaving 2-3 mL of an emulsion at the top of the mixture. 3 drops of 2 N aqueous HC1 were added causing the emulsion to break and a white cloudiness to now appear in the lower aqueous layer. The remaining hexane layer was removed. The aqueous layer was pH 2 (pH paper). The aqueous layer was extracted with 5 x 3 mL of CH 2 Cl2, the combined extracts dried with a 2 S04, filtered, and concentrated in vacuo to provide 2-((4S,5S)-l-(4-((l-(2,3-difluoro-5-methoxyphenyl)piperidin- 4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid, HC1 (tan foam, 34 mg, 82% yield). LC-MS Anal. Calc'd for C 2 5H 26 F 5 304 527.18, found [M+H] 528.2. !H MR (500MHZ, CDC1 3 ) δ 7.12 - 7.02 (m, 2H), 6.98 - 6.88 (m, 2H), 6.40 - 6.22 (m, 2H), 4.40 (dt, J=10.5, 3.3 Hz, 2H), 3.84 - 3.67 (m, 5H), 3.35 (ddd, J=1 1.6, 7.9, 3.3 Hz, 2H), 3.23 (td, J=3.6, 2.5 Hz, 1H), 3.01 (ddd, J=11.6, 7.9, 3.3 Hz, 2H), 2.86 (dd, J=16.5, 3.0 Hz, 1H), 2.46 (dd, J=16.5, 10.5 Hz, 1H), 2.19 - 2.05 (m, 2H), 2.03 - 1.80 (m, 4H), 1.36 (d, J=7.2 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT=10.8 min, HI: 99%. hGPR40 EC 50 = 273 nM. hGPR40 IP1 EC 50 = 133 nM. Example 17

2- ((4S,5S)-l-(4-((l-(5-Ethoxy-2,3-difluorophenyl)piperidin-4-y l)oxy)phenyl)-4-methyl- - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00248] 17A. 5-Ethoxy-l,2,3-trifluorobenzene: To 3,4,5-trifluorophenol (5 g, 33.8 mmol) and potassium carbonate (11.7 g, 84 mmol) in acetone (33 mL) was added bromoethane (3.8 mL, 50.6 mmol). The mixture was refluxed for 16 hours under nitrogen. The reaction was filtered and the solids were washed with 5 mL of CH2CI2 to provide a slightly cloudy filtrate which was concentrated to remove the CH2CI2 and then was filtered to provide product (4.5 g, 76% yield) as a faintly yellow liquid. l R NMR (500MHz, CDCI3) δ 6.50 (dd, J=9.5, 5.6 Hz, 2H), 3.96 (q, J=6.9 Hz, 2H), 1.41 (t, J=7.0 Hz, 3H).

[00249] Example 17 (pale tan foam, 52 mg) was prepared following the procedure for Example 16. LC-MS Anal. Calc'd for C26H28F5 3O4 541.20, found [M+H] 542.2. l R NMR (500MHz, CDC1 3 ) δ 7.14 - 7.02 (m, 2H), 7.00 - 6.88 (m, 2H), 6.40 - 6.21 (m, 2H),

4.47 - 4.33 (m, 2H), 3.96 (q, J=7.2 Hz, 2H), 3.35 (ddd, J=11.6, 7.8, 3.2 Hz, 2H), 3.28 - 3.15 (m, 1H), 3.00 (ddd, J=l 1.8, 7.9, 3.4 Hz, 2H), 2.87 (dd, J=16.5, 3.0 Hz, 1H), 2.47 (dd, J=16.5, 10.5 Hz, 1H), 2.19 - 2.05 (m, 2H), 2.02 - 1.79 (m, 2H), 1.46 - 1.30 (m, 6H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT=13.4 min, HI: 98%. hGPR40 EC = 286 nM. hGPR40 IP1 EC = 294 nM.

Example 18

2-((4S,5S)-l-(4-((l-(2,3-Difluoro-5-isobutoxyphenyl)piperidi n-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HC1

[00250] Example 18 (sticky, off-white foam, 24 mg) was prepared following the procedure for Example 16. LC-MS Anal. Calc'd for C28H32F5N3O4 569.23, found

[M+H] 570.2. !H NMR (500MHZ, CDCI3) δ 7.15 - 7.01 (m, 2H), 6.99 - 6.85 (m, 2H),

6.38 - 6.24 (m, 2H), 4.46 - 4.35 (m, 2H), 3.64 (d, J=6.6 Hz, 2H), 3.35 (ddd, J=11.5, 7.8, 3.3 Hz, 2H), 3.26 - 3.18 (m, 1H), 3.01 (ddd, J=l 1.7, 7.8, 3.6 Hz, 2H), 2.87 (dd, J=16.5, 3.0 Hz, 1H), 2.47 (dd, J=16.5, 10.5 Hz, 1H), 2.19 - 2.07 (m, 2H), 2.02 - 1.87 (m, 2H), 1.36 (d, J=7.2 Hz, 3H), 1.02 (d, J=6.6 Hz, 6H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT=8.5 min, HI: 98%. hGPR40 EC 50 = 4763 nM.

Example 21

2-((4S,5S)-l-(4-((l-(2-Cyano-5-methoxyphenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, TFA

[00251] 21A. 2-(4-Hydroxypiperidin-l-yl)-4-methoxybenzonitrile: A reaction mixture of 2-fluoro-4-methoxybenzonitrile (0.5g, 3.31 mmol) and piperidin-4-ol (1.0 g, 9.92 mmol) in Acetonitrile (3.31 mL) with K 2 C0 3 (1.37 g, 9.92 mmol) was stirred at 70 °C for 16h. Cooled to room temperature. Diluted with water and extracted with methylene chloride. The organic extract was washed with brine, dried over Mg 2 S04, and concentrated in vacuo to give 0.77g yellow oil. Purification via silica gel chromatography gave (0.77g, 3.24mmol, 98% yield) as a clear, colorless oil. LC-MS Anal. Calc'd for C 13 H 16 N 2 0 2 232.12, found [M+H] 233.0.

[00252] Example 21 (amber oil, 71 mg) was prepared following the procedure for Example 1. LC-MS Anal. Calc'd for C 26 H 27 F 3 N 4 04 516.20, found [M+H] 517.1. l H NMR (400MHz, CDC1 3 ) δ 10.17 (br. s., IH), 7.65 (d, J=8.2 Hz, IH), 7.18 - 6.89 (m, 5H), 6.82 (d, J=2.2 Hz, IH), 4.61 (br. s., IH), 4.40 (d, J=9.9 Hz, IH), 3.97 (t, J=9.6 Hz, 2H), 3.88 (s, 3H), 3.50 - 3.32 (m, 2H), 3.31 - 3.14 (m, J=5.8, 3.6 Hz, IH), 2.86 (dd, J=16.5, 2.7 Hz, IH), 2.59 - 2.39 (m, J=16.5, 10.4 Hz, IH), 2.38 - 2.24 (m, 2H), 2.24 - 2.09 (m, 2H), 1.35 (d, J=7.1 Hz, 3H). Analytical HPLC (Orthogonal method, 0% Solvent B start): RT = 9.3 min, HI: 98%. hGPR40 EC 50 = 680 nM. Example 22

2-(l-(4-((l-(2-Fluoro-5-methoxyphenyl)piperidin-4-yl)oxy)phe nyl)-4-methyl-3- (pyrimidin-2-yl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, TFA Isomer 1 and Isomer 2

[00253] Example 22 (red oil, 11 mg) was prepared following the procedure for Example 1 using methyl but-2-enoate instead of IE with chiral preparative SFC separation. Isomer 1 : LC-MS Anal. Calc'd for C28H30FN5O4 519.23, found [M+H] 520.2. !H NMR (500MHz, CDC1 3 ) δ 8.86 (d, J=4.7 Hz, 2H), 7.30 (d, J=4.7 Hz, 2H), 7.22 (t, J=4.8 Hz, IH), 7.00 - 6.94 (m, 3H), 6.63 (dd, J=7.3, 2.9 Hz, IH), 6.47 (dt, J=8.9, 3.1 Hz, IH), 4.60 (dt, J=1 1.0, 2.5 Hz, IH), 4.45 - 4.39 (m, IH), 3.80 (s, 3H), 3.74 (qd, J=7.1, 2.5 Hz, IH), 3.41 (ddd, J=11.6, 8.0, 3.6 Hz, 2H), 3.04 (ddd, J=11.6, 7.7, 3.3 Hz, 2H), 2.92 (dd, J=16.0, 2.8 Hz, IH), 2.51 (dd, J=16.0, 11.0 Hz, IH), 2.17 (td, J=8.5, 3.9 Hz, 2H), 2.06 - 1.97 (m, 2H), 1.43 (d, J=7.2 Hz, 3H). Analytical HPLC (Zorbax method, 0%

Solvent B start): RT = 5.1 min, HI: 98%. hGPR40 EC 50 = 2038 nM. Isomer 2: LC-MS

Awal. Calc'd for C 28 H3 0 F 5 O4 519.23, found [M+H] 520.2. Ή NMR (500MHz, CDC1 3 ) δ 8.87 (d, J=4.7 Hz, 2H), 7.30 (d, J=9.1 Hz, 2H), 7.23 (t, J=5.0 Hz, IH), 7.02 (dd, J=12.0, 8.9 Hz, IH), 6.96 (d, J=8.S Hz, 2H), 6.80 (dd, J=6.7, 2.9 Hz, IH), 6.58 (dt, J=8.8, 3.0 Hz, IH), 4.60 (d, J=11.0 Hz, IH), 4.48 (dt, J=6.1, 3.1 Hz, IH), 3.80 (s, 3H), 3.75 - 3.68 (m, IH), 3.59 - 3.51 (m, 2H), 3.21 - 3.1 1 (m, 2H), 2.92 (dd, J=16.1, 2.6 Hz, 1H), 2.51 (dd, J=l 6 0 , 11.0 Hz, IH), 2.30 - 2.21 (m, 2H), 2.12 - 2.02 (m, 2H), 1.43 (d, J=7.2 Hz, 3H). Analytical HPLC (Zorbax method, 0% Solvent B start): RT = 5.0 min, HI: 92%. hGPR40 EC50 = 5570 nM.

Example 25

2-((4S,5S)-l-(4-((l-(5-Ethoxy-2-fluorophenyl)-3-ethylpiperid in-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HC1 (Isomers 1 2)

[00254] 25A. Ethyl l-benzyl-3-ethyl-4-oxopiperidine-3-carboxylate: To a solution of ethyl l-benzyl-4-oxopiperidine-3-carboxylate (1.0 g, 3.8 mmol) in ACN (9.6 mL) was added KOtBu (1 N in THF) (5.7 mL, 5.7 mmol) and iodoethane (0.46 mL, 5.7 mmol). The reaction mixture was stirred at rt overnight. The reaction mixture was poured into sat. NH4CI and extracted with EtOAc. The organic layers was washed with brine, dried over Na 2 S0 4 and concentrated to give a colorless oil as a crude material as ethyl l-benzyl-3- ethyl-4-oxopiperidine-3-carboxylate (0.90 g, 3.11 mmol, 81% yield). LC-MS Anal.

Calc'd for C 17 H 23 N0 3 289.17, found [M+H] 290.0.

[00255] 25B. l-Benzyl-3-ethylpiperidin-4-one: To a sealed vial containing ethyl 1 - benzyl-3-ethyl-4-oxopiperidine-3-carboxylate (800 mg, 2.8 mmol) was added 6 N HC1 (8 mL, 48 mmol). The reaction mixture was heated at 100 °C for 16 h. To the reaction mixture was poured cold 5 N NaOH to adjust the pH~8 and the mixture was extracted with EtOAc. The organic layer was washed with brine, dried over Na 2 S0 4 , and concentrated. Purification via silica gel chromatography gave a colorless oil (0.3 lg, 1.427 mmol, 51.6% yield). LC-MS Anal. Calc'd for C 14 H 19 NO 217.15, found [M+H] 218.0. ¾ NMR (400MHz, CDC1 3 ) δ ppm 7.27 (s, 5H), 3.64 - 3.31 (m, 2H), 3.02 - 2.70 (m, 2H),

2.53 - 2.39 (m, 2H), 2.34 - 2.25 (m, 2H), 2.18 (dd, J=1 1.0, 9.5 Hz, 1H), 1.39 - 0.98 (m, 2H), 0.79 (t, J=7.5 Hz, 3H).

[00256] 25C. l-Benzyl-3 -ethyl- l-methyl-4-oxopiperidin-l-ium, iodide salt: To a solution of l-benzyl-3-ethylpiperidin-4-one (0.46 g, 2.10 mmol) in acetone (2.10 mL) was added methyl iodide (0.16 mL, 2.60 mmol). The reaction mixture was stirred at rt overnight. The solvents were removed and the residue was dried in vacuo to give a light yellow foam as l-benzyl-3 -ethyl- l-methyl-4-oxopiperidin-l-ium, iodide salt (0.58g, 1.60 mmol, 76% yield). LC-MS Anal. Calc'd for C 15 H 22 NO 232.17, found [M+H] 232.0.

[00257] 25D. l-(5-Ethoxy-2-fluorophenyl)-3-ethylpiperidin-4-one: To a solution of 5- ethoxy-2-fluoroaniline (100 mg, 0.64 mmol) in EtOH (1.3 mL) / water (0.6 mL) was added l-benzyl-3 -ethyl- l-methyl-4-oxopiperidin-l-ium, iodide salt (301 mg, 0.84 mmol) and K2CO3 (13 mg, 0.097 mmol). The reaction mixture was heated at 100 °C overnight. The reaction mixture was poured into water and was extracted with dichloromethane. The organic layer was washed with brine, dried over a2S0 4 , and concentrated via vacuum. Purification via silica gel chromatography gave a colorless oil l-(5-ethoxy-2- fluorophenyl)-3-ethylpiperidin-4-one (155 mg, 0.58 mmol, 91% yield). !H MR

(500MHz, CD3CI) δ ppm 6.95 (dd, J=12.1, 8.8 Hz, 1H), 6.53 (dd, J=7.4, 3.0 Hz, 1H),

6.44 (dt, J=8.8, 3.2 Hz, 1H), 4.14 - 3.88 (m, 2H), 3.74 - 3.48 (m, 2H), 3.25 - 3.11 (m, 1H), 2.92 (dd, J=11.8, 9.4 Hz, 1H), 2.69 (dddd, J=14.2, 10.0, 5.8, 1.4 Hz, 1H), 2.60 - 2.40 (m, 2H), 2.07 - 1.77 (m, 1H), 1.46 - 1.29 (m, 4H), 1.04 - 0.83 (m, 3H).

[00258] 25E. l-(5-Ethoxy-2-fluorophenyl)-c«-3-ethylpiperidin-4-ol: To a 0 °C solution of l-(5-ethoxy-2-fluorophenyl)-3-ethylpiperidin-4-one (0.51 g, 1.92 mmol) in MeOH (7.7 mL) was added NaBH 4 (0.084 g, 2.2 mmol). After 1 h, the reaction was quenched via addition of 1.5 M K.2HP0 4 and the mixture was concentrated to remove MeOH. The resulting mixture was diluted with water and extracted with

dichloromethane. The organic layer was washed with brine, dried with Na 2 S0 4 , and concentrated. The residue was dissolved in EtOAc and filtered through a plug of silica gel. Purification via silica gel chromatography gave l-(5-ethoxy-2-fluorophenyl)-cz ' s-3- ethylpiperidin-4-ol (180 mg, 0.673 mmol, 35.0% yield). l H NMR (400MHz, CD 3 C1) δ ppm 6.90 (dd, J=12.1, 8.8 Hz, 1H), 6.55 (dd, J=7.3, 3.0 Hz, 1H), 6.39 (dt, J=8.8, 3.2 Hz, 1H), 4.08 - 3.85 (m, 3H), 3.31 - 2.96 (m, 3H), 2.82 (t, J=1 1.0 Hz, 1H), 2.02 - 1.72 (m, 3H), 1.59 - 1.33 (m, 5H), 0.99 (t, J=7.5 Hz, 3H).

[00259] 25F. Methyl 2-(l-(4-((l-(5-ethoxy-2-fluorophenyl)-3-ethylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate: To a solution of methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3-(trifluoromethyl)- 4,5- dihydro-lH-pyrazol-5-yl)acetate (U, 56 mg, 0.18 mmol), PPh 3 (70 mg, 0.27 mmol), and di-tert-butylazodicarboxylate (61 mg, 0.27 mmol) in toluene (0.88 mL) was added l-(5- ethoxy-2-fluorophenyl)-c«-3-ethylpiperidin-4-ol (57 mg, 0.21 mmol). The reaction mixture was stirred at 60 °C for 2.5 h. The reaction mixture was loaded onto to a silica gel column for purification, followed by RP-Prep HPLC. The purified material was dissolved in EtOAc and was extracted with sat. NaHCC^. The organic layers were washed with brine and dried over a2S0 4 . The organic layer was concentrated to give methyl 2-(l-(4-((l-(5-ethoxy-2-fluorophenyl)-3-ethylpiperidin-4-yl) oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceta te (42 mg, 0.074 mmol, 41.9% yield) as a colorless foam. LC-MS Anal. Calc'd for C29H35F4N3O4: 565.26, found [M+H] 566.2.

[00260] Example 25 (Isomers 1 and 2): To methyl 2-((4S,5S)-l-(4-((l-(5-ethoxy-2- fluorophenyl)-3-ethylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-( trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetate (42 mg, 0.074 mmol) in THF (1.4 mL) /water (0.14 mL), lithium hydroxide (1 N in water, 0.22 mL, 0.22 mmol) was added. The reaction mixture was stirred at rt for 1 h. The reaction mixture was diluted with water and acidified with IN HC1. The mixture was extracted with CH2CI2. The organic extracts were dried (MgS0 4 ) and concentrated to afford a diastereomeric mixture. The diastereomers were separated by chiral Prep. SFC to provide 2 isomers. Example 25 (Isomer 1) was obtained as a colorless foam (17.5 mg, 39.0%). LC-MS Anal. Calc'd for C 28 H33F4 3 04: 551.24, found [M+H] 552.1. ¾ NMR (400MHz, CD 3 CN) δ ppm 7.86 - 7.72 (m, 1H), 7.25 - 7.15 (m, 1H), 7.12 - 7.02 (m, 2H), 7.01 - 6.94 (m, 2H), 6.93 - 6.84 (m, 1H), 4.48 - 4.38 (m, 1H), 4.29 - 4.19 (m, 1H), 4.04 (d, J=7.1 Hz, 2H), 3.73 - 3.51 (m, 3H), 3.38 - 3.24 (m, 2H), 3.03 - 2.91 (m, 1H), 2.71 (d, J=3.0 Hz, 1H), 2.51 (d, J=9.6 Hz, 3H), 2.32 (br. s., 3H), 1.37 (t, J=6.9 Hz, 3H), 1.33 - 1.22 (m, 3H), 0.94 (t, J=7.6 Hz, 3H). Analytical HPLC

(Orthogonal method): RT = 11.3 min, HI: 97%. hGPR40 EC 50 = 1019 nM. Example 25 (Isomer 2) was obtained as a colorless foam (17.0mg, 37.1%). LC-MS Anal. Calc'd for C 28 H 33 F 4 N 3°4 551.24, found [M+H] 552.1. ¾ NMR (400MHz, CD 3 CN) δ ppm 7.97 - 7.83 (m, 1H), 7.27 - 7.16 (m, 1H), 7.13 - 7.04 (m, 2H), 7.03 - 6.88 (m, 3H), 4.50 - 4.38 (m, 1H), 4.32 - 4.18 (m, 1H), 4.05 (d, J=7.1 Hz, 2H), 3.77 - 3.54 (m, 4H), 3.44 - 3.22 (m, 3H), 3.04 - 2.89 (m, 1H), 2.71 (d, J=3.3 Hz, 1H), 2.65 - 2.56 (m, 1H), 2.51 (d, J=9.6 Hz, 1H), 2.42 - 2.26 (m, 2H), 1.38 (t, J=6.9 Hz, 3H), 1.33 - 1.23 (m, 3H), 0.94 (t, J=7.5 Hz, 3H). Analytical HPLC (Orthogonal method): RT = 11.3 min, HI: 95%. hGPR40 EC 50 = 113 nM. hGPR40 IP1 EC 50 = 21 nM. Acute oral glucose tolerance: -38% @ 0.3 mg/kg. Example 26 2-((4S,5S)-l-(4-((l-(5-Ethoxy-2-fluorophenyl)-3-isopropylpip eridin-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HCl, (Isomers 1 and

[00261] Example 26 (Isomer 1) (beige foam, 20 mg) was prepared following the procedure for Example 25. LC-MS Anal. Calc'd for C29H35F4N3O4: 565.26, found

[M+H] 566.0. !H MR (500MHZ, CD 3 CN) δ ppm 7.85 - 7.74 (m, 1H), 7.25 - 7.15 (m, 1H), 7.1 1 - 7.04 (m, 2H), 6.99 (d, J=9.1 Hz, 2H), 6.94 - 6.85 (m, 1H), 4.47 - 4.38 (m, 2H),

4.04 (d, J=6.9 Hz, 2H), 3.65 - 3.53 (m, 3H), 3.41 - 3.28 (m, 2H), 2.76 - 2.67 (m, 1H), 2.54 - 2.44 (m, 1H), 2.37 - 2.22 (m, 4H), 1.37 (t, J=7.0 Hz, 3H), 1.30 (d, J=6.9 Hz, 3H), 1.00

(d, J=7.2 Hz, 3H), 0.90 (d, J=6.9 Hz, 3H). Analytical HPLC (Orthogonal method): RT = 11.5 min, HI: 100%. hGPR40 EC 50 = 1904 nM.

[00262] Example 26 (Isomer 2) (beige foam, 21.0 mg) was prepared following the procedure for Example 25. LC-MS Anal. Calc'd for C29H35F4N3O4: 565.24, found

[M+H] 566.0. !H MR (500MHZ, CD 3 CN) δ ppm 8.03 - 7.87 (m, 1H), 7.29 - 7.18 (m, 1H), 7.12 - 7.04 (m, 2H), 7.02 - 6.98 (m, 2H), 6.97 - 6.91 (m, 1H), 4.53 - 4.36 (m, 2H),

4.05 (q, J=6.9 Hz, 2H), 3.64 (d, J=3.3 Hz, 3H), 3.47 - 3.38 (m, 1H), 3.37 - 3.27 (m, 1H), 2.73 (dd, J=16.5, 3.0 Hz, 2H), 2.56 - 2.45 (m, 1H), 2.44 - 2.23 (m, 4H), 1.38 (t, J=7.0 Hz, 3H), 1.30 (d, J=7.2 Hz, 3H), 1.00 (d, J=7.2 Hz, 3H), 0.90 (d, J=7.2 Hz, 3H). Analytical HPLC (Orthogonal method): RT = 11.5 min, HI: 100%. hGPR40 EC 50 = 82 nM.

Example 27

2-((4S,5S)-l-(4-((l-(5-Ethoxy-2-fluorophenyl)-3-isobutylpipe ridin-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HCl (Isomers 1 and 2)

[00263] Example 27 (Isomer 1) (beige foam, 38 mg) was prepared following the procedure for Example 25. LC-MS Anal. Calc'd for C30H37F4N3O4: 579.27, found

[M+H] 580.3. !H MR (500MHZ, CD 3 CN) δ ppm 7.92 - 7.66 (m, IH), 7.30 - 7.13 (m, IH), 7.13 - 7.04 (m, 2H), 7.02 - 6.95 (m, 2H), 6.92 - 6.85 (m, IH), 4.53 - 4.38 (m, IH), 4.20 - 4.12 (m, IH), 4.04 (d, J=7.1 Hz, 2H), 3.73 - 3.46 (m, 3H), 3.40 - 3.18 (m, 2H), 2.94 (dd, J=12.5, 7.2 Hz, 3H), 2.73 (dd, J=16.4, 3.3 Hz, IH), 2.51 (d, J=9.6 Hz, IH), 2.31 (m., IH), 1.74 - 1.53 (m, 2H), 1.37 (t, J=7.1 Hz, 3H), 1.31 - 1.21 (m, 3H), 0.92 (dd, J=7.3, 6.3 Hz, 6H). Analytical HPLC (Orthogonal method): RT = 14.6 min, HI: 96%. hGPR40 EC50 = 412 nM. hGPR40 IP 1 EC 50 = 90 nM.

[00264] Example 27 (Isomer 2) (beige foam, 21 mg) was prepared following the procedure for Example 25. LC-MS Anal. Calc'd for C 30 H3 7 F 4 3O4: 579.27, found

[M+H] 580.4. !H MR (500MHZ, CD 3 CN) δ ppm 7.77 - 7.57 (m, IH), 7.23 - 7.12 (m, IH), 7.10 - 7.02 (m, 2H), 7.00 - 6.92 (m, 2H), 6.89 - 6.80 (m, IH), 4.47 - 4.35 (m, IH), 4.20 - 4.10 (m, IH), 4.04 (d, J=7.1 Hz, 2H), 3.66 - 3.54 (m, 2H), 3.53 - 3.42 (m, IH), 3.37 - 3.27 (m, IH), 3.25 - 3.13 (m, IH), 2.70 (m, IH), 2.54 - 2.43 (m, IH), 2.33 - 2.23 (m, 4H), 1.74 - 1.54 (m, 2H), 1.37 (t, J=6.9 Hz, 3H), 1.30 (d, J=7.1 Hz, 3H), 0.92 (t, J=6.3 Hz, 6H). Analytical HPLC (Orthogonal method): RT = 14.8 min, HI: 100%. hGPR40 EC50 = 34 nM. hGPR40 IP1 EC 50 = 84 nM.

Example 29

2-((4S,5S)-l-(4-(((3R,4R)-l-(5-Ethoxy-2-fluorophenyl)-3-meth ylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid, HC1 salt

[00265] 29A. 5-Ethoxy-2-fluoroaniline: To a solution of (5-ethoxy-2- fluorophenyl)boronic acid (10.1 g, 55.0 mmol) in MeOH (220 mL) was added 14.8 M aqueous NH4OH (18.6 mL, 275 mmol), and then cuprous oxide (1.57 g, 11.0 mmol). The reaction mixture was stirred under air for 7 h. The reaction mixture was concentrated. The crude material was dissolved in EtO Ac/Hex (2: 1). The material was filtered through CELITE® and concentrated. The crude material was purified by flash chromatography to provide 5-ethoxy-2-fluoroaniline (4.10 g, 26.4 mmol, 48% yield) as a brown oil. LC-MS Anal. Calc'd for C 8 H 20 FNO 155.17, found [M+H] 156.1. l H NMR (400MHz, CDC1 3 ) δ 6.86 (dd, J=10.9, 8.8 Hz, 1H), 6.32 (dd, J=7.5, 2.9 Hz, 1H), 6.20 (dt, J=8.8, 3.3 Hz, 1H), 3.94 (q, J=6.9 Hz, 2H), 3.68 (br. s., 2H), 1.37 (t, J=6.9 Hz, 3H).

[00266] 29B. 1 -Benzyl- 1, 3 -dimethyl-4-oxopiperidin- 1 -ium, iodide salt: To a solution of l-benzyl-3-methylpiperidin-4-one (14.0 g, 68.9 mmol) in acetone (68.9 mL) at rt was added Mel (8.61 mL, 138 mmol) dropwise. The reaction mixture was concentrated to obtain l-benzyl-l,3-dimethyl-4-oxopiperidin-l-ium, iodide salt (24 g, 69.5 mmol, 101% yield) as a light yellow foam. LC-MS Anal. Calc'd for C 14 H 20 NO: 218.15, found [M+H] 219.2.

[00267] 29C. l-(5-Ethoxy-2-fluorophenyl)-3-methylpiperidin-4-one: To a solution of 5-ethoxy-2-fluoroaniline (7.87 g, 50.7 mmol) in EtOH (103 mL) was added K 2 C0 3 (1.05 g, 7.61 mmol), 1 -benzyl- 1,3 -dimethyl -4-oxopiperidin-l-ium,iodide salt (26.3 g, 76.0 mmol), and water (46.6 mL). The reaction mixture was heated to 95 °C overnight. The reaction mixture was cooled to rt and diluted with EtO Ac/water. The layers were separated and the aqueous layer was extracted with EtOAc (2x). The combined organic layers were washed with brine, dried (MgS0 4 ) and concentrated. The crude product was purified by flash chromatography to provide l-(5-ethoxy-2-fluorophenyl)-3- methylpiperidin-4-one (10.12 g, 40.3 mmol, 79% yield) as a colorless oil, which solidified overnight. LC-MS Anal. Calc'd for C 14 H 18 FN0 2 251.13, found [M+H] 252.2. in NMR (400MHZ, CDCI3) δ 6.95 (dd, J=12.1, 8.8 Hz, IH), 6.52 (dd, J=7.5, 2.9 Hz,

IH), 6.44 (dt, J=8.8, 3.2 Hz, IH), 3.98 (q, J=7.3 Hz, 2H), 3.75 - 3.64 (m, 2H), 3.12 (td, J=l 1.7, 3.5 Hz, IH), 2.85 - 2.69 (m, 3H), 2.49 (dt, J=14.1, 3.3 Hz, IH), 1.40 (t, J=6.9 Hz, 3H), 1.09 (d, J=6.1 Hz, 3H).

[00268] 29D. (0 ' s)-l-(5-ethoxy-2-fluorophenyl)-3-methylpiperidin-4-ol: To a solution of l-(5-ethoxy-2-fluorophenyl)-3-methylpiperidin-4-one (4.920 g, 19.58 mmol) in THF (98 mL) at -78 °C was added a 1 M solution of L-Selectride (23.49 mL, 23.49 mmol) in THF. After 1 h, the reaction mixture was quenched with 1 M aqueous NaOH (23.49 mL, 23.49 mmol) and warmed to 0 °C. 30% aqueous H 2 (¾ (7.40 mL, 72.4 mmol) was added dropwise and the reaction mixture was warmed to rt and stirred for 1 h. The reaction mixture was diluted with EtO Ac/water and the layers were separated. The aqueous layer was extracted with EtO Ac (2x). The combined organic layers were washed with brine, dried (MgS0 4 ) and concentrated. The crude product was purified by flash

chromatography to provide (c«)-l-(5-ethoxy-2-fluorophenyl)-3-methylpiperidin-4-ol (4.453 g, 17.58 mmol, 90% yield) as a colorless oil. LC-MS Anal. Calc'd for

C 14 H 20 FNO 2 : 253.31, found [M+H] 254.0. l H NMR (500MHz, CDC1 3 ) δ 6.89 (dd, J=12.1, 8.8 Hz, IH), 6.52 (dd, J=7.3, 2.9 Hz, IH), 6.37 (dt, J=8.8, 3.2 Hz, IH), 3.97 (q, J=7.1 Hz, 2H), 3.90 (br. s., IH), 3.13 - 3.02 (m, 2H), 3.02 - 2.95 (m, IH), 2.84 (dd, J=1 1.4, 9.8 Hz, IH), 2.05 (dqt, J=10.1, 6.7, 3.6 Hz, IH), 2.00 - 1.91 (m, IH), 1.91 - 1.83 (m, IH), 1.50 (br. s., IH), 1.38 (t, J=6.9 Hz, 3H), 1.03 (d, J=6.9 Hz, 3H).

[00269] 29E. (3R,4S)-l-(5-Ethoxy-2-fluorophenyl)-3-methylpiperidin-4-ol: (C¾)-l-(5- ethoxy-2-fluorophenyl)-3-methylpiperidin-4-ol (29.15 g, 115 mmol) was purified by chiral SFC chromatography. (3R,4S)-l-(5-Ethoxy-2-fluorophenyl)-3-methylpiperidin-4- ol (13.54 g, 53.50 mmol, 47% yield) was obtained as a colorless oil after concentration. LC-MS Anal. Calc'd for C 14 H 18 FN0 2 : 251.13, found [M+H] 252.2. l R NMR (400MHz, CDCI3) δ 6.95 (dd, J=12.1, 8.8 Hz, IH), 6.52 (dd, J=7.5, 2.9 Hz, IH), 6.44 (dt, J=8.8, 3.2 Hz, IH), 3.98 (q, J=7.3 Hz, 2H), 3.75 - 3.64 (m, 2H), 3.12 (td, J=1 1.7, 3.5 Hz, IH), 2.85 - 2.69 (m, 3H), 2.49 (dt, J=14.1, 3.3 Hz, IH), 1.40 (t, J=6.9 Hz, 3H), 1.09 (d, J=6.1 Hz, 3H).

[00270] 29F. Methyl 2-((4S,5S)-l-(4-(((3R,4R)-l-(5-ethoxy-2-fluorophenyl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetate: To a solution of (3R,4S)-l-(5-ethoxy-2-fluorophenyl)-3-methylpiperidin-4- ol (2.48 g, 9.78 mmol), methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetate (1J, 3.09 g, 9.78 mmol), and BU3P (3.86 mL, 15.6 mmol) in toluene (122 mL) was added (E)-diazene-l,2-diylbis(piperidin- 1-ylmethanone) (3.95 g, 15.6 mmol). The reaction mixture was sonicated for 2 h. The reaction mixture was poured into hexanes and filtered. The crude product was purified by flash chromatography to provide methyl 2-((4S,5S)-l-(4-(((3R,4R)-l-(5-ethoxy-2- fluorophenyl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetate (2.60 g, 4.70 mmol, 48% yield) as a colorless oil. LC- MS Anal. Calc'd for C 28 H33F4 3 04: 551.57, found [M+H] 552.4. ¾ NMR (500MHz, CDCI3) δ 7.07 - 7.00 (m, 2H), 6.95 - 6.86 (m, 3H), 6.51 (dd, J=7.3, 2.9 Hz, IH), 6.39 (dt,

J=8.8, 3.0 Hz, IH), 4.39 (dt, J=10.1, 3.1 Hz, IH), 3.98 (q, J=7.0 Hz, 2H), 3.83 (td, J=9.1, 4.3 Hz, IH), 3.71 (s, 3H), 3.48 - 3.36 (m, 2H), 3.22 - 3.14 (m, IH), 2.85 - 2.74 (m, 2H), 2.57 (dd, J=12.1, 9.6 Hz, IH), 2.41 (dd, J=16.1, 10.3 Hz, IH), 2.20 - 2.08 (m, 2H), 1.88 - 1.76 (m, IH), 1.40 (t, J=7.0 Hz, 3H), 1.34 (d, J=7.2 Hz, 3H), 1.11 (d, J=6.6 Hz, 3H).

[00271] Example 29: To a solution of methyl 2-((4S,5S)-l-(4-(((3R,4R)-l-(5-ethoxy- 2-fluorophenyl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl- 3-(trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetate (2.57 g, 4.66 mmol) in THF (85 mL) and water (8.5 mL) was added a 1 M aqueous solution of LiOH (58.2 mL, 23.3 mmol) and the reaction mixture was stirred at rt until complete for 1 h. The reaction mixture was concentrated to remove the THF and hexanes were added. An emulsion formed. The layers were separated as much as possible. Brine was added to the emulsion and the layers separated completely. The combined aqueous layer and brine aqueous layer was acidified to pH 2 with 1 M aqueous HC1. The product was extracted with CH2CI2 (3x) and the combined organic layers were dried (MgSC^) and concentrated to provide 2-((4S,5S)-l-(4-

(((3R,4R)-l-(5-ethoxy-2-fluorophenyl)-3-methylpiperidin-4 -yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid (2.4720 g, 4.60 mmol, 99% yield) as a white foam. To a solution of 2-((4S,5S)-l-(4-(((3R,4R)-l-(5-ethoxy-2- fluorophenyl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetic acid (7.72 g, 14.4 mmol) in acetonitrile (50 mL) was added 3 N aqueous HC1 (9.57 mL, 28.7 mmol). The solution was concentrated. 2- ((4S,5S)-l-(4-(((3R,4R)-l-(5-ethoxy-2-fluorophenyl)-3-methyl piperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid, HC1 (8.08 g, 14.0 mmol, 97% yield) was isolated as an off-white solid. LC-MS Anal. Calc'd for C 21 li 3 l F 4 N 3 0 4 : 537.55, found [M+H] 538.3. !H MR (400MHZ, CD 3 CN) δ 7.99 (br. s., 1H), 7.24 (ddd, J=12.0, 9.3, 1.2 Hz, 1H), 7.12 - 7.04 (m, 2H), 7.02 - 6.93 (m, 3H), 4.47 - 4.38 (m, 1H), 4.18 (td, J=10.0, 4.1 Hz, 1H), 4.04 (q, J=6.9 Hz, 2H), 3.78 - 3.65 (m, 2H), 3.62 (dd, J=12.4, 4.1 Hz, 1H), 3.42 (td, J=l 1.8, 4.4 Hz, 1H), 3.37 - 3.27 (m, 1H), 2.83 (br. s., 1H), 2.73 (dd, J=16.5, 3.0 Hz, 1H), 2.54 - 2.38 (m, 2H), 2.36 - 2.28 (m, 1H), 1.37 (t, J=7.0 Hz, 3H), 1.30 (d, J=7.2 Hz, 3H), 1.1 1 (d, J=6.9 Hz, 3H). Analytical HPLC (Orthogonal method) RT=13.3 min, HI: 97%. hGPR40 EC 50 = 154 nM. hGPR40 IP1 EC50 = 1 1 nM. Acute oral glucose tolerance: -52% @ 0.3 mg/kg (average of two experiments).

Example 30

2-((4S,5S)- 1 -(4-(((3 S, 4S)- 1 -(5-Ethoxy-2-fluorophenyl)-3-methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid

[00272] Example 30 (off-white solid, 26.8 mg) was prepared following the procedure for Example 25. LC-MS Anal. Calc'd for C 21 li 3 l F 4 N 3 0 4 : 537.55, found [M+H] 538.3. !H NMR (500MHz, CD 3 CN) δ 7.09 - 7.03 (m, 2H), 6.97 - 6.90 (m, 3H), 6.53 (dd, J=7.4, 3.0 Hz, 1H), 6.43 (dt, J=8.9, 3.3 Hz, 1H), 4.46 - 4.38 (m, 1H), 3.98 (q, J=6.9 Hz, 2H), 3.92 (td, J=9.2, 4.4 Hz, 1H), 3.43 - 3.36 (m, 2H), 3.36 - 3.29 (m, 1H), 2.87 - 2.77 (m, 1H), 2.72 (dd, J=16.4, 3.2 Hz, 1H), 2.58 (dd, J=12.1, 9.9 Hz, 1H), 2.48 (dd, J=16.4, 9.5 Hz, 1H), 2.20 - 2.1 1 (m, 1H), 2.06 - 1.97 (m, 1H), 1.74 - 1.63 (m, 1H), 1.36 - 1.31 (m, 3H), 1.30 (d, J=6.9 Hz, 3H), 1.07 (d, J=6.6 Hz, 3H). Analytical HPLC (Orthogonal method): 13 3 min, HI: 97%. hGPR40 EC 50 = 1200 nM. Example 32

2-((4S,5S)-l-(4-((l-(5-Ethoxy-2-fluorophenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00273] 32A. 2-Bromo-4-ethoxy-l-fluorobenzene: A mixture of 3-bromo-4- fluorophenol (2 g, 10.47 mmol), potassium carbonate (1.74 g, 12.57 mmol) and bromoethane (4 mL, 52.4 mmol) and acetonitrile (10.5 mL) was heated at 60 °C overnight. Sat. aqueous NaHC0 3 (50 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x50 mL), and the combined organic extracts were washed with water (100 mL) and brine (100 mL), and dried ( a 2 S04), filtered, and concentrated to afford 2-bromo-4-ethoxy-l-fluorobenzene (clear oil, 1.9 g, 8.67 mmol, 83% yield). l H NMR (400MHz, CDC1 3 ) δ 7.05 (dt, J=6.1, 3.4 Hz, 1H), 7.03 - 6.98 (m, 1H), 6.82 - 6.76 (m, 1H), 3.98 (q, J=7.0 Hz, 2H), 1.40 (t, J=7.0 Hz, 3H).

[00274] 32B. l-(5-Ethoxy-2-fluorophenyl)piperidin-4-ol: A mixture of piperidin-4-ol (460 mg, 4.5 mmol), 2-bromo-4-ethoxy-l-fluorobenzene (495 mg, 2.26 mmol) and Sphos pre.cat. (15 mg, 0.023 mmol) in THF (4.5 mL) was purged with argon and lithium bis(trimethylsilyl)amide (9.0 mL, 9.04 mmol) was added. The reaction mixture was heated at 70 °C for 2.5 h. Sat. aqueous NaHC0 3 (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x10 mL), and the combined organic extracts were washed with water (20 mL) and brine (20 mL), and dried

( a2S0 4 ), filtered, and concentrated. Purification via silica gel chromatography gave 1-

(5-ethoxy-2-fluorophenyl)piperidin-4-ol (yellow oil, 254 mg, 1.051 mmol, 46.5% yield). LC-MS Anal. Calc'd for C 13 H 18 FN0 2 239.13, found [M+H] 240.2.

[00275] Example 32 was prepared following the procedure for Example 1. LC-MS

Anal. Calc'd for C 26 H 2 9F4 3 04 523.52, found [M+H] 524.1. !H MR (400MHZ,

CD 3 CN) δ ppm 8.00 (dd, J=5.9, 2.6 Hz, 1H), 7.29 (dd, J=12.2, 9.2 Hz, 1H), 7.14 - 7.08

(m, 2H), 7.06 - 6.98 (m, 3H), 4.70 (br. s., 1H), 4.51 - 4.40 (m, 1H), 4.08 (q, J=6.9 Hz, 2H), 3.91 (t, J=9.9 Hz, 2H), 3.65 - 3.50 (m, 2H), 3.44 - 3.29 (m, 1H), 2.76 (dd, J=16.4, 3.1 Hz, 1H), 2.62 (br. s., 2H), 2.56 - 2.48 (m, 1H), 2.38 - 2.23 (m, 2H), 1.40 (t, J=7.0 Hz, 3H), 1.33 (d, J=7.3 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 1 1.8 min, HI: 100%. hGPR40 EC 50 = 217 nM. [00276] Example 33 and Example 35 to Example 56 were prepared following the procedure for Example 32.

Example 33

2-((4S,5S)-l-(4-((l-(2-Chloro-5-methoxyphenyl)piperidin-4-yl )oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00277] Example 33 (1.2 mg): LC-MS Anal. Calc'd for C 2 5H 27 C1F 3 304 525.95, found [M+H] 526.1. ¾ NMR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.24 (d, J=8.9 Hz, 1H), 7.13 - 7.04 (m, 2H), 6.94 (d, J=8.9 Hz, 2H), 6.66 (d, J=3.0 Hz, 1H), 6.54 (dd, J=8.7, 2.7 Hz, 1H), 4.51 - 4.37 (m, 3H), 3.79 (s, 3H), 3.31 - 3.18 (m, 3H), 2.98 - 2.90 (m, 2H), 2.82 - 2.70 (m, 1H), 2.34 - 2.20 (m, 1H), 2.18 - 2.07 (m, 2H), 2.04 - 1.85 (m, 2H), 1.32 (d, J=7.4 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.4 min, HI: 100%. hGPR40 EC 50 = 103 nM. Example 35

2-((4S,5S)-l-(4-((l-(2,5-Dichlorophenyl)piperidin-4-yl)oxy)p henyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00278] Example 35 (2.2 mg): LC-MS Anal. Calc'd for C 2 4H 2 4Cl2F3 30 3 530.37, found [M+H] 530.0. ¾ NMR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.29 (d, J=8.4 Hz, IH), 7.14 - 7.05 (m, 3H), 7.01 - 6.92 (m, 3H), 4.48 - 4.38 (m, 3H), 3.31 - 3.20 (m, 2H), 2.95 (ddd, J=1 1.5, 8.1, 3.2 Hz, 2H), 2.77 (dd, J=15.9, 3.0 Hz, IH), 2.33 (dd, J=15.9, 10.9 Hz, IH), 2.18 - 2.07 (m, 2H), 2.03 - 1.93 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.7 min, HI: 100%.

hGPR40 EC50 = 254 nM.

Example 36

2-((4S,5S)-l-(4-((l-(2-Fluoro-5-methylphenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00279] Example 36 (5.9 mg): LC-MS Anal. Calc'd for C 2 5H 27 F 4 303 493.50, found [M+H] 494.2. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.10 - 7.06 (m, 2H), 6.96 - 6.92 (m, 2H), 6.89 (dd, J=12.4, 8.4 Hz, IH), 6.84 (dd, J=8.4, 2.0 Hz, IH), 6.78 - 6.73 (m, IH), 4.47 - 4.36 (m, 2H), 3.32 - 3.21 (m, 2H), 2.96 (ddd, J=1 1.5, 8.3, 3.5 Hz, 2H), 2.77 (dd, J=15.9, 3.0 Hz, IH), 2.36 - 2.31 (m, 2H), 2.29 (s, 3H), 2.16 - 2.08 (m, 2H), 2.00 - 1.89 (m, 2H), 1.33 (d, J=7.4 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.9 min, HI: 97%. hGPR40 EC 50 = 440 nM. hGPR40 IP1 EC 50 = 86 nM. Example 37

2-((4S,5S)- 1 -(4-(( 1 -(2-Chlorophenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00280] Example 37 (3.3 mg): LC-MS Anal. Calc'd for C 24 H 2 5C1F 3 303 495.93, found [M+H] 496.1. ¾ NMR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.35 (dd, J=7.9, 1.5 Hz, IH), 7.23 (td, J=7.7, 1.5 Hz, IH), 7.14 - 7.06 (m, 3H), 7.03 - 6.91 (m, 3H), 4.48 - 4.38 (m, 2H), 3.31 - 3.20 (m, 3H), 2.99 - 2.90 (m, 2H), 2.77 (dd, J=15.9, 3.0 Hz, IH), 2.32 (dd, J=15.6, 10.7 Hz, IH), 2.20 - 2.07 (m, 2H), 2.04 - 1.87 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.4 min, HI: 98%. hGPR40 EC 50 = 448 nM.

Example 38

2-((4S,5S)-l-(4-((l-(2,5-Difluorophenyl)piperidin-4-yl)oxy)p henyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00281] Example 38 (2.8 mg): LC-MS Anal. Calc'd for C 2 4H 2 4F5 3 03 497.46, found [M+H] 498.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.09 (d, J=8.9 Hz, 2H), 7.01 - 6.90 (m, 3H), 6.73 (ddd, J=10.3, 7.1, 3.0 Hz, IH), 6.66 - 6.57 (m, IH), 4.49 - 4.37 (m, 3H), 3.28 - 3.19 (m, 2H), 3.04 - 2.95 (m, 2H), 2.76 (dd, J=15.4, 3.0 Hz, IH), 2.29 (dd, J=15.6, 10.7 Hz, IH), 2.21 - 2.10 (m, 2H), 2.02 - 1.84 (m, 2H), 1.32 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.4 min, HI: 100%. hGPR40 EC50 = 475 nM.

Example 39

2-((4S,5S)-4-Methyl-l-(4-((l-(o-tolyl)piperidin-4-yl)oxy) phenyl)-3-(trifluoromethyl)- -dihydro-lH-pyrazol-5-yl)acetic acid

[00282] Example 39 (8.6 mg): LC-MS Anal. Calc'd for C 2 5H 28 F 3 303 475.51, found [M+H] 476.2. !H MR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.19 - 7.12 (m, 2H), 7.10 - 7.03 (m,3H), 6.98 - 6.92 (m, 3H), 4.46 - 4.35 (m, 2H), 3.31 - 3.21 (m, 2H), 3.18 - 3.10 (m, 2H), 2.85 - 2.72 (m, 2H), 2.31 (s, 3H), 2.16 - 2.09 (m, 2H), 2.00 - 1.88 (m, 2H), 1.33 (d, J=7.4 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.8 min, HI: 96%. hGPR40 EC 50 = 498 nM. Example 40

2-((4S,5 S)- 1 -(4-(( 1 -(3 -Methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3 - (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00283] Example 40 (1.2 mg): LC-MS Anal. Calc'd for C 2 5H 28 F 3 304 491.51, found [M+H] 492.2. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.16 (t, J=8.2 Hz, IH), 7.09 (d, J=8.9 Hz, 2H), 6.97 - 6.88 (m, 2H), 6.61 (dd, J=8.4, 2.0 Hz, IH), 6.53 (t, J=2.2 Hz, IH), 6.44 (dd, J=7.9, 2.0 Hz, IH), 4.48 - 4.37 (m, 2H), 3.79 (s, 3H), 3.59 - 3.46 (m, 2H), 3.25 (d, J=4.0 Hz, IH), 3.14 - 3.04 (m, 2H), 2.76 (dd, J=15.9, 3.0 Hz, IH), 2.26 (dd, J=15.4, 10.9 Hz, IH), 2.15 - 2.05 (m, 2H), 1.97 - 1.84 (m, 2H), 1.32 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.7 min, HI: 97%. hGPR40 EC 50 = 620 nM. hGPR40 IP1 EC 50 = 107 nM.

Example 41

2-((4S,5S)-l-(4-((l-(3-Fluoropyridin-2-yl)piperidin-4-yl)oxy )phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00284] Example 41 (5.6 mg): LC-MS Anal. Calc'd for C 2 3H 2 4F 4 403 480.46, found [M+H] 481.1. !H MR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.96 - 7.93 (m, IH), 7.32 (ddd, J=13.4, 7.9, 1.5 Hz, IH), 7.14 - 7.05 (m, 2H), 7.00 - 6.89 (m, 2H), 6.82 (ddd, J=7.9, 5.0, 3.0 Hz, IH), 4.53 - 4.39 (m, 3H), 3.88 - 3.72 (m, 2H), 3.32 - 3.21 (m, 2H), 2.77 (dd, J=15.9, 2.5 Hz, IH), 2.32 (dd, J=15.6, 10.7 Hz, IH), 2.10 (ddd, J=9.8, 6.8, 3.7 Hz, 2H), 1.89 (dt, J=12.9, 4.0 Hz, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.9 min, HI: 100%. hGPR40 EC 50 = 628 nM. Example 42

2-((4S,5S)-l-(4-((l-(2-Fluoro-6-methylphenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00285] Example 42 (1.2 mg): LC-MS Anal. Calc'd for C 2 5H 27 F 4 303 493.50, found

[M+H] 494.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.07 (d, J=8.4 Hz, 2H), 7.00

- 6.93 (m, 4H), 6.89 - 6.79 (m, IH), 4.46 - 4.32 (m, 2H), 3.29 - 3.03 (m, 5H), 2.78 (dd, J=16.1, 2.7 Hz, IH), 2.41 - 2.36 (m, IH), 2.34 (s, 3H), 2.08 (br. s., 2H), 1.85 (d, J=7.4 Hz, 2H), 1.34 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.6 min, HI: 100%. hGPR40 EC 50 = 796 nM.

Example 43

2-((4S,5S)-l-(4-((l-(2,5-Dimethylphenyl)piperidin-4-yl)oxy)p henyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00286] Example 43 (8.3 mg): LC-MS Anal. Calc'd for C 26 H3 0 F 3 3O3 489.54, found [M+H] 490.2. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.12 - 7.06 (m, 2H), 7.04 (d, J=7.4 Hz, IH), 6.97 - 6.93 (m, 2H), 6.87 (s, IH), 6.77 (d, J=7.4 Hz, IH), 4.46 - 4.34 (m, 2H), 3.24 (dd, J=5.4, 4.0 Hz, IH), 3.17 - 3.09 (m, 2H), 2.85 - 2.73 (m, 3H), 2.33 (dd, J=16.1, 10.7 Hz, IH), 2.29 (s, 3H), 2.26 (s, 3H), 2.12 (dd, J=11.9, 3.0 Hz, 2H), 1.96 - 1.87 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.9 min, HI: 98%. hGPR40 EC 50 = 1074 nM.

Example 44

2-((4S,5S)-l-(4-((l-(2,6-Dimethylphenyl)piperidin-4-yl)ox y)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00287] Example 44 (5.7 mg): LC-MS Anal. Calc'd for C 26 H3 0 F 3 3O3 489.54, found [M+H] 490.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.12 - 7.06 (m, 2H), 7.03 - 6.88 (m, 5H), 4.45 - 4.31 (m, 3H), 3.28 - 3.08 (m, 5H), 2.78 (dd, J=16.3, 3.0 Hz, 1H), 2.34 (s, 3H), 2.32 (s, 3H), 2.08 (dt, J=8.2, 3.8 Hz, 2H), 1.85 (dtd, J=12.7, 8.8, 4.0 Hz, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.5 min, HI: 99%. hGPR40 EC 50 = 1338 nM. Example 45

2-((4S,5S)- 1 -(4-(( 1 -(4-Methoxypyridin-2-yl)piperidin-4-yl)oxy)phenyl)-4-methyl- 3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00288] Example 45 (4.2 mg): LC-MS Anal. Calc'd for C 2 4H2 7 F 3 404 492.50, found [M+H] 493.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.91 (d, J=5.9 Hz, 1H), 7.14

- 7.04 (m, 2H), 6.97 - 6.90 (m, 2H), 6.31 (dd, J=5.9, 2.0 Hz, 1H), 6.24 (d, J=2.0 Hz, 1H), 4.48 (tt, J=7.4, 3.5 Hz, 1H), 4.42 (d, J=10.4 Hz, 1H), 3.84 (s, 4H), 3.88 - 3.81 (m, 1H), 3.47 - 3.37 (m, 2H), 3.24 (dd, J=5.4, 4.0 Hz, 1H), 2.77 (dd, J=15.9, 3.0 Hz, 1H), 2.32 (dd, J=15.6, 10.7 Hz, 1H), 2.10 - 1.98 (m, 2H), 1.93 - 1.79 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.6 min, 100%. hGPR40 EC50 = 1523 nM.

Example 46

2-((4S,5S)-l-(4-((l-(2-Methoxyphenyl)piperidin-4-yl)oxy)phen yl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00289] Example 46 (3.6 mg): LC-MS Anal. Calc'd for C 2 5H 28 F 3 304 491.51, found [M+H] 492.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.10 - 7.06 (m, 2H), 7.04 - 7.00 (m, 2H), 6.96 - 6.89 (m, 4H), 4.49 - 4.34 (m, 2H), 3.91(s, 3H), 3.32 - 3.20 (m, 2H), 2.95 - 2.88 (m, 3H), 2.77 (dd, J=15.9, 3.0 Hz, IH), 2.30 (dd, J=15.6, 10.7 Hz, IH), 2.17 - 2.08 (m, 2H), 1.97 (dt, J=12.9, 4.0 Hz, 2H), 1.32 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.6 min, HI: 99%. hGPR40 EC 50 = 1697 nM.

Example 47

2-((4S,5S)- 1 -(4-(( 1 -(2-Methoxypyridin-4-yl)piperidin-4-yl)oxy)phenyl)-4-methyl- 3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00290] Example 47 (4.8 mg): LC-MS Anal. Calc'd for C 2 4H 27 F 3 404 492.50, found [M+H] 493.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.76 (d, J=6.4 Hz, IH), 7.13 - 7.03 (m, 2H), 6.98 - 6.88 (m, 2H), 6.52 (dd, J=6.4, 2.0 Hz, IH), 6.14 (d, J=2.5 Hz, IH), 4.51 (tt, J=6.9, 3.5 Hz, IH), 4.42 (d, J=10.4 Hz, IH), 3.88 (s, 3H), 3.72 - 3.64 (m, 2H), 3.37 (td, J=8.7, 4.0 Hz, 2H), 3.25 (dd, J=5.9, 4.0 Hz, IH), 2.76 (dd, J=15.9, 2.5 Hz, IH), 2.32 (dd, J=15.9, 10.4 Hz, IH), 2.09 - 1.98 (m, 2H), 1.92 - 1.77 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.6 min, HI: 99%. hGPR40 EC 50 = 1910 nM.

Example 48

2-((4S,5 S)- 1 -(4-(( 1 -(3 -Fluorophenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3 - (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00291] Example 48 (7.4 mg): LC-MS Anal. Calc'd for C 2 4H 2 5F4 3 03 479.47, found [M+H] 480.1. !H MR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.23 - 7.16 (m, IH), 7.10 - 7.05 (m, 2H), 7.00 - 6.90 (m, 2H), 6.74 (dd, J=8.2, 2.2 Hz, IH), 6.65 (dt, J=12.6, 2.1 Hz, IH), 6.50 (td, J=8.1, 2.2 Hz, IH), 4.49 - 4.36 (m, 2H), 3.59 - 3.45 (m, 2H), 3.24 (dd, J=5.4, 4.0 Hz, IH), 3.12 (ddd, J=12.4, 8.4, 3.5 Hz, 2H), 2.77 (dd, J=15.9, 3.0 Hz, IH), 2.34 (dd, J=15.9, 10.4 Hz, IH), 2.14 - 2.02 (m, 2H), 1.95 - 1.81 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.1 min, HI: 93%. hGPR40 EC 50 = 2272 nM.

Example 49

2-((4S,5 S)- 1 -(4-(( 1 -(4-Fluorophenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3 - (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00292] Example 49 (1.8 mg): LC-MS Anal. Calc'd for C 2 4H 2 5F4 3 03 479.47, found [M+H] 480.2. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.11 - 7.04 (m, 2H), 7.01 - 6.88 (m, 6H), 4.45 - 4.34 (m, 2H), 3.44 - 3.36 (m, 2H), 3.24 (dd, J=5.4, 4.0 Hz, IH), 3.06 - 2.99 (m, 2H), 2.77 (dd, J=15.9, 2.5 Hz, IH), 2.33 (dd, J=15.9, 10.4 Hz, IH), 2.15 - 2.06 (m, 2H), 1.97 - 1.88 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.7 min, HI: 98%. hGPR40 EC 50 = 2646

Example 50

2-((4S,5S)-l-(4-((l-(3-Chlorophenyl)piperidin-4-yl)oxy)pheny l)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00293] Example 50 (7.4 mg): LC-MS Anal. Calc'd for C 2 4H 2 5C1F3 3 03 495.93, found [M+H] 496.1. ¾ NMR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.17 (t, J=8.2 Hz, 1H), 7.08 (d, J=8.9 Hz, 2H), 6.97 - 6.91 (m, 3H), 6.87 (dd, J=8.2, 2.2 Hz, 1H), 6.78 (dd, J=7.9, 1.5 Hz, 1H), 4.48 - 4.39 (m, 2H), 3.58 - 3.45 (m, 2H), 3.25 (dd, J=5.4, 4.0 Hz, 1H), 3.12 (ddd, J=12.4, 8.4, 3.5 Hz, 2H), 2.77 (dd, J=15.9, 3.0 Hz, 1H), 2.34 (dd, J=16.1, 10.7 Hz, 1H), 2.17 - 2.04 (m, 2H), 1.97 - 1.84 (m, 2H), 1.33 (d, J=7.4 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.2 min, HI: 97%. hGPR40 EC 50 = 2839 nM.

Example 51

2-((4S,5S)-l-(4-((l-(4-Methoxy-2,6-dimethylphenyl)piperidin- 4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid

[00294] Example 51 (0.9 mg): LC-MS Anal. Calc'd for C 27 H3 2 F 3 304 519.56, found [M+H] 520.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.10 - 7.04 (m, 2H), 6.99 - 6.93 (m, 2H), 6.60 - 6.48 (m, 2H), 4.46 - 4.32 (m, 2H), 3.76 - 3.72 (m, 3H), 3.28 - 3.07 (m, 5H), 2.78 (dd, J=16.3, 3.0 Hz, 1H), 2.41 - 2.36 (m, 1H), 2.36 - 2.33 (m, 2H), 2.31 (s, 3H), 2.08 (br. s., 3H), 1.87 (br. s., 2H), 1.34 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 2.3 min, HI: 97%. hGPR40 EC 50 = 3063 nM.

Example 52

2-((4S,5S)-l-(4-((l-(3-Fluoro-4-methoxyphenyl)piperidin-4 -yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00295] Example 52 (4.4 mg): LC-MS Anal. Calc'd for C 2 5H 27 F 4 304 509.50, found [M+H] 510.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.07 (d, J=8.9 Hz, 2H), 6.97 - 6.91 (m, 3H), 6.79 (dd, J=14.1, 2.7 Hz, IH), 6.71 (ddd, J=8.8, 2.8, 1.2 Hz, IH), 4.45 - 4.37 (m, 2H), 3.84 (s, 3H), 3.41 - 3.35 (m, 2H), 3.23 (d, J=3.5 Hz, IH), 3.05 - 2.97 (m, 2H), 2.77 (dd, J=16.1, 2.7 Hz, IH), 2.33 (dd, J=15.9, 10.4 Hz, IH), 2.14 - 2.05 (m, 2H), 1.97 - 1.86 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.7 min, HI: 97%. hGPR40 EC 50 = 3621 nM.

Example 54

2-((4S,5S)-l-(4-((l-(2-Fluoro-6-methoxyphenyl)piperidin-4-yl )oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00296] Example 54 (0.5 mg): LC-MS Anal. Calc'd for C 2 5H 27 F 4 304 509.50, found [M+H] 510.1. !H MR (500MHz, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.11 (d, J=8.9 Hz, 2H), 6.98 (td, J=8.3, 6.2 Hz, IH), 6.93 (d, J=8.9 Hz, 2H), 6.73 - 6.61 (m, 2H), 4.44 (d, J=10.4 Hz, IH), 4.37 - 4.28 (m, 3H), 3.86 (s, 3H), 3.31 - 3.18 (m, 2H), 3.14 - 3.08 (m, 2H), 2.74 (d, J=12.4 Hz, IH), 2.24 - 2.13 (m, IH), 2.06 (d, J=12.9 Hz, 2H), 1.92 - 1.81 (m, 2H), 1.31 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.8 min, HI: 100%. hGPR40 EC 50 = 5896 nM.

Example 56

2-((4S,5S)-l-(4-((l-(4-Chlorophenyl)piperidin-4-yl)oxy)ph enyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00297] Example 56 (9 mg). LC-MS Anal. Calc'd for C 2 4H 2 5C1F3 3 03 495.93, found [M+H] 496.1. !H MR (500MHZ, 1 : 1 MeOD:CDCl 3 ) δ ppm 7.19 (d, J=8.9 Hz, 2H), 7.07 (d, J=9.4 Hz, 2H), 6.99 - 6.89 (m, 4H), 4.42 (dt, J=7.3, 3.5 Hz, 2H), 3.50 - 3.41 (m, 2H), 3.24 (dd, J=5.4, 4.0 Hz, 1H), 3.08 (ddd, J=12.3, 8.5, 3.5 Hz, 2H), 2.77 (dd, J=16.3, 3.0 Hz, 1H), 2.34 (dd, J=15.9, 10.4 Hz, 1H), 2.17 - 2.02 (m, 2H), 1.98 - 1.78 (m, 2H), 1.33 (d, J=6.9 Hz, 3H). Analytical HPLC (Analytical LC/MS method, 0% Solvent B start): RT = 1.9 min, HI: 94%. hGPR40 EC 50 = 6268 nM.

Example 57

2-((4S,5S)-l-(4-((3-Fluoro-l-(2-fluoro-5-methoxyphenyl)piper idin-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HCl (Isomers 1, 2, 3 and 4)

[00298] 57A. Benzyl 4-((trimethylsilyl)oxy)-5,6-dihydropyridine-l(2H)-carboxylat e: To a solution of benzyl 4-oxopiperidine-l-carboxylate (1.27g, 5.44 mmol) in DMF (4 mL) was added chlorotrimethylsilane (0.83 mL, 6.53 mmol) followed by triethylamine (1.52 mL, 10.89 mmol). The resulting heterogeneous mixture was warmed to 80 °C and stirred for 16 h. The cooled mixture was diluted with hexanes (50 mL), washed with sat. aHC03 (30 mL) and brine (30 mL), then dried over MgS0 4 , filtered and concentrated to afford benzyl 4-((trimethylsilyl)oxy)-5,6-dihydropyridine-l(2H)-carboxylat e (1.5 g, 4.91 mmol, 90% yield) as a clear oil. l H NMR (400MHz, CDC1 3 ) δ 7.22 - 7.08 (m, 5H), 4.98 - 4.94 (m, 2H), 4.60 (br. s., 1H), 3.77 (q, J=2.3 Hz, 2H), 3.42 (t, J=5.6 Hz, 2H), 1.94 (br. s., 2H), 0.04 -0.04 (m, 9H).

[00299] 57B. Benzyl 3-fluoro-4-oxopiperidine-l-carboxylate: To a solution of benzyl 4-((trimethylsilyl)oxy)-5,6-dihydropyridine-l(2H)-carboxylat e (1.52 g, 4.98 mmol) in acetonitrile (31 mL) at rt was added SELECTFLUOR® (2.1 g, 6.0 mmol) portionwise over 10 min. The mixture was stirred for 2 h, then concentrated to dryness and partitioned between EtOAc and brine. The aqueous layer was extracted with EtOAc and the combined organic phases were washed with brine, dried over MgS0 4 , filtered and concentrated. Purification via silica chromatography gave benzyl 3-fluoro-4- oxopiperidine-l-carboxylate (1.2 g, 4.78 mmol, 96% yield) as a solid. !H NMR

(400MHz, CDC1 3 ) δ 7.51 - 7.30 (m, 5H), 5.27 - 5.16 (m, 2H), 4.97 - 4.70 (m, 1H), 4.47 (br. s., 1H), 4.33 - 4.18 (m, 1H), 3.59 - 3.28 (m, 2H), 2.72 - 2.41 (m, 2H).

[00300] 57C. Benzyl 3-fluoro-4-hydroxypiperidine-l-carboxylate: To a solution of benzyl 3-fluoro-4-oxopiperidine-l-carboxylate (880 mg, 3.50 mmol) in MeOH (7 mL) was added sodium borohydride (130 mg, 3.50 mmol) slowly. After 1 h, 10% KHSO4 (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x30 mL), and the combined organic extracts were dried ( a 2 S04), filtered, and concentrated. Purification via silica gel chromatography gave benzyl 3-fluoro-4- hydroxypiperidine-l-carboxylate (870 mg, 3.44 mmol, 98%> yield). l R NMR (400MHz, CDCI3) δ ppm 7.47 - 7.27 (m, 5H), 5.13 (s, 2H), 4.75 - 4.52 (m, 1H), 4.07 - 3.68 (m, 3H), 3.61 - 3.04 (m, 2H), 2.22 (d, J=5.0 Hz, 1H), 1.93 - 1.63 (m, 2H).

[00301] 57D. Benzyl 4-((?ert-butyldimethylsilyl)oxy)-3-fluoropiperidine-l- carboxylate: To a solution of benzyl 3-fluoro-4-hydroxypiperidine-l-carboxylate (410 mg, 1.62 mmol) in DCM (1.62 mL) and triethylamine (670 μΐ, 4.86 mmol) was added tert-butyldimethylsilyl trifluoromethanesulfonate (390 μΐ, 1.70 mmol) at 0 °C. After 1 h, sat. aq. aHC03 (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x30 mL), and the combined organic extracts were dried ( a2S04), filtered, and concentrated to afford benzyl 4-((tert-butyldimethylsilyl)oxy)-3- fluoropiperidine-l-carboxylate (450 mg, 1.22 mmol, 76% yield). l R NMR (400MHz, CDC1 3 ) δ ppm 7.36 - 7.20 (m, 5H), 5.05 (s, 2H), 4.43 - 4.22 (m, 1H), 4.07 - 3.96 (m, 1H), 3.88 - 3.62 (m, 2H), 3.59 - 3.24 (m, 2H), 1.75 - 1.50 (m, 2H), 0.84 (s, 9H), 0.02-0.05 (m, 6H).

[00302] 57E. 4-((ter?-Butyldimethylsilyl)oxy)-3-fluoropiperidine: A mixture of benzyl 4-((ter?-butyldimethylsilyl)oxy)-3-fluoropiperidine-l-carbox ylate (440 mg, 1.20 mmol) and Pd on activated carbon (127 mg, 0.120 mmol) in MeOH (12 mL) was purged with H 2 for 30 min and stirred under a H 2 balloon at rt for 1 h. The mixture was filtered through CELITE®, washed with EtOAc (30 mL) and MeOH (30 mL) and concentrated to afford 4-((tert-butyldimethylsilyl)oxy)-3-fluoropiperidine (270 mg, 1.16 mmol, 97% yield) as a clear oil. ¾ NMR (400MHz, CDC1 3 ) δ ppm 4.50 - 4.27 (m, 1H), 3.97 - 3.77 (m, 1H), 3.19 - 3.05 (m, 1H), 2.93 (ddd, J=13.1, 6.3, 4.1 Hz, 1H), 2.82 - 2.65 (m, 2H), 2.62 - 2.46 (m, 1H), 1.73 - 1.50 (m, 2H), 0.88 (s, 9H), 0.00 (d, J=3.8 Hz, 6H).

[00303] 57F. 4-(4-((tert-Butyldimethylsilyl)oxy)-3-fluoropiperidin-l-yl)- 5-chloro-2- methoxypyridine: A mixture of 4-((ter?-butyldimethylsilyl)oxy)-3-fluoropiperidine (194 mg, 0.83 mmol), 4-bromo-5-chloro-2-methoxypyridine (185 mg, 0.83 mmol) and Sphos pre.cat. (6 mg, 8 μιηοΐ) in THF (1.7 mL) was purged with argon and lithium

bis(trimethylsilyl)amide (1 mL, 1.0 mmol) was added. The reaction mixture was heated at 70 °C for 2 h. Sat. aqueous NaHCC^ (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x10 mL), and the combined organic extracts were washed with water (20 mL) and brine (20 mL), and dried ( a 2 S04), filtered, and concentrated. Purification via silica gel chromatography gave 4-(4-((tert- butyldimethylsilyl)oxy)-3-fluoropiperidin-l-yl)-5-chloro-2 -methoxypyridine (182 mg, 0.49 mmol, 58% yield). LC-MS Anal. Calc'd for C 17 H 28 ClFN 2 0 2 Si 374.16, found

[M+H] 374.9.

[00304] 57G. l-(5-Chloro-2-methoxypyridin-4-yl)-3-fluoropiperidin-4-ol: To a mixture of 4-(4-((ter?-butyldimethylsilyl)oxy)-3-fluoropiperidin- 1 -yl)-5-chloro-2- methoxypyridine (192 mg, 0.51 mmol) in THF (1.0 mL) was added TBAF (0.6 mL, 0.61 mmol). The reaction was stirred at 23 °C for 2 h. Sat. aqueous aHC03 (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x10 mL), and the combined organic extracts were washed with water (20 mL) and brine (20 mL), and dried ( a 2 S04), filtered, and concentrated. Purification via silica gel chromatography gave l-(5-chloro-2-methoxypyridin-4-yl)-3-fluoropiperidin-4-ol (1 10 mg, 0.42 mmol, 82% yield) as a white foam. LC-MS Anal. Calc'd for CHH24CIFN2O2 260.07, found [M+H] 261.0.

[00305] 57H. 3-Fluoro-l-(2-fluoro-5-methoxyphenyl)piperidin-4-yl 4- methylbenzenesulfonate: To a solution of 3-fluoro-l-(2-fluoro-5- methoxyphenyl)piperidin-4-ol (74 mg, 0.30 mmol), 4-methylbenzene-l-sulfonyl chloride (290 mg, 1.52 mmol) and N,N-dimethylpyridin-4-amine (4 mg, 0.03 mmol) in DCM (600 μΐ), pyridine (245 μΐ, 3.04 mmol) was added dropwise. The reaction mixture was stirred at rt for 20 h. The reaction mixture was diluted with EtOAc and the organic layer was washed successively with H 2 0 and brine. The organic layer was dried and concentrated. Purification by chromatography gave 3-fluoro-l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl 4-methylbenzenesulfonate (waxy solid, 74 mg, 0.19 mmol, 61% yield). LC-MS Anal. Calc'd for C 19 H 21 F 2 N0 4 S 397.12, found [M+H] 398.0.

[00306] 571. Methyl 2-((4S,5S)-l-(4-((3-fluoro-l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluo romethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetate: A solution of 3-fluoro-l-(2-fluoro-5-methoxyphenyl)piperidin- 4-yl 4-methylbenzenesulfonate (96 mg, 0.24 mmol), methyl 2-((4S,5S)-l-(4- hydroxyphenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetate (1J, 80 mg, 0.25 mmol) and cesium carbonate (160 mg, 0.48 mmol) in DMF (0.48 mL) was heated at 60 °C for 48 h. The reaction mixture was diluted with EtOAc (10 mL), and the organic layer was washed with water (10 mL) three times and brine (10 mL). The organic layer was dried (Na 2 S0 4 ), filtered, and concentrated. Purification by

chromatography and RP-Prep HPLC afforded a diastereomeric mixture. The

diastereomers were separated by chiral Prep. SFC to provide 4 isomers.

Isomer 1 : LC-MS Anal. Calc'd for C26H28F5N304 541.20, found [M+H] 542.2. Isomer 2: LC-MS Anal. Calc'd for C26H28F5N304 541.20, found [M+H] 542.2. Isomer 3: LC-MS Anal. Calc'd for C26H28F5N304 541.20, found [M+H] 542.2. Isomer 3: LC-MS Anal. Calc'd for C 26 H 28 F 5 N 3 04 541.20, found [M+H] 542.2. [00307] Four isomers of Example 57 were prepared following the procedure for Example 1.

[00308] Example 57 (Isomer 1, yellow solid, 0.9 mg): LC-MS Anal. Calc'd for C 25 H 26 F 5 N 3°4 527.49, found [M+H] 528.1. l H NMR (400MHz, CD 3 CN) δ ppm 7.13 - 7.09 (m, 3H), 7.08 - 7.01 (m, 2H), 6.95 (dd, J=7.2, 3.1 Hz, IH), 6.68 (dt, J=9.0, 3.3 Hz, IH), 5.04 - 4.84 (m, IH), 4.58 - 4.42 (m, 2H), 3.80 (s, 3H), 3.79 - 3.68 (m, IH), 3.53 - 3.43 (m, 2H), 3.40 - 3.25 (m, 2H), 3.13 (ddd, J=12.3, 9.3, 3.0 Hz, IH), 2.76 (dd, J=16.4, 3.1 Hz, IH), 2.53 (dd, J=16.6, 9.5 Hz, IH), 2.35 (dd, J=8.9, 4.1 Hz, IH), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 1 1.5 min, HI: 99%. hGPR40 EC 50 = 292 nM.

[00309] Example 57 (Isomer 2, yellow solid, 1.2 mg): LC-MS Anal. Calc'd for C 25 H 26 F 5 N 3°4 527.49, found [M+H] 528.1. l H NMR (400MHz, CD 3 CN) δ ppm 7.14 - 7.07 (m, 3H), 7.07 - 7.02 (m, 2H), 6.99 (dd, J=7.2, 2.9 Hz, IH), 6.69 (dt, J=9.0, 3.2 Hz, IH), 5.06 - 4.87 (m, IH), 4.58 - 4.42 (m, 2H), 3.80 (s, 3H), 3.78 - 3.70 (m, IH), 3.53 - 3.45 (m, IH), 3.42 - 3.26 (m, 2H), 3.15 (ddd, J=12.3, 9.4, 3.4 Hz, IH), 2.76 (dd, J=16.4, 3.1 Hz, 1H), 2.52 (dd, J=16.6, 9.5 Hz, IH), 2.41 - 2.31 (m, IH), 2.08 - 2.01 (m, IH), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 1 1.4 min, HI: 99%. hGPR40 EC 50 = 119 nM.

[00310] Example 57 (Isomer 3, yellow solid, 0.9 mg): LC-MS Anal. Calc'd for C 2 5H 26 F 5 N304 527.49, found [M+H] 528.1. l H NMR (400MHz, CD 3 CN) δ ppm 7.18 -

7.00 (m, 6H), 6.78 - 6.69 (m, IH), 5.1 1 - 4.91 (m, IH), 4.61 - 4.50 (m, IH), 4.50 - 4.42 (m, IH), 3.81 (s, 3H), 3.80 - 3.72 (m, IH), 3.58 - 3.48 (m, IH), 3.36 (dt, J=12.2, 7.6 Hz, 2H), 3.21 (ddd, J=12.3, 9.3, 3.3 Hz, 2H), 2.76 (dd, J=16.6, 3.3 Hz, IH), 2.53 (dd, J=16.6, 9.5 Hz, IH), 2.39 (dd, J=9.0, 4.5 Hz, IH), 2.18 - 2.04 (m, IH), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 11.6 min, HI: 100%. hGPR40 EC 50 = 716 nM.

[00311] Example 57 (Isomer 4, yellow solid, 1.2 mg): LC-MS Anal. Calc'd for C 25 H 26 F 5 N 3°4 527.49, found [M+H] 528.0. l H NMR (400MHz, CD 3 CN) δ ppm 7.26 - 7.20 (m, IH), 7.19 - 7.15 (m, IH), 7.14 - 7.09 (m, 2H), 7.08 - 7.02 (m, 2H), 6.83 - 6.75 (m, IH), 5.16 - 4.96 (m, IH), 4.62 - 4.53 (m, IH), 4.51 - 4.43 (m, IH), 3.81 (s, 3H), 3.89 - 3.74 (m, IH), 3.63 - 3.53 (m, IH), 3.48 - 3.22 (m, 4H), 2.76 (dd, J=16.4, 3.1 Hz, IH), 2.53 (dd, J=16.6, 9.5 Hz, IH), 2.42 (td, J=9.3, 4.5 Hz, IH), 2.20 - 2.07 (m, IH), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 11.4 min, HI: 97%. hGPR40 EC 50 = 66 nM. hGPR40 ΙΡ1 EC 50 = 11 nM. Example 58

2-((4S,5S)-l-(4-((l-(3-Chloropyridin-2-yl)piperidin-4-yl)oxy )phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00312] 58A. Methyl 2-((4S,5S)-l-(4-((l-(3-chloropyridin-2-yl)piperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetate: To a stirred solution of triphenylphosphine (43 mg, 0.16 mmol) and (E)-diethyl diazene-1,2- dicarboxylate (22 μΐ, 0.14 mmol) in THF (1 18 μΐ) at rt was added methyl 2-((4S,5S)-l-(4- hydroxyphenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetate (37 mg, 0.12 mmol) followed by l-(3-chloropyridin-2-yl)piperidin-4-ol (synthesized following the procedure for Example 32 (25 mg, 0.12 mmol). The reaction mixture was stirred at 50 °C for 30 min. The solvent was evaporated and the residue was purified. Purification by chromatography gave methyl 2-((4S,5S)-l-(4-((l-(3-chloropyridin-2- yl)piperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4 ,5-dihydro-lH-pyrazol-5- yl)acetate (54 mg, 0.11 mmol, 90% yield). LC-MS Anal. Calc'd for C 2 4H 26 C1F 3 403 510.17, found [M+H] 51 1.1.

[00313] Example 58 (yellow oil, 9.5 mg) was prepared following the procedure for Example 32. LC-MS Anal. Calc'd for C 2 3H 2 4C1F 3 403 496.15, found [M+H] 497.0. l H NMR (400MHz, CD 3 CN) δ ppm 8.26 - 8.10 (m, 2H), 7.19 (dd, J=7.8, 6.3 Hz, IH), 7.10 (d, J=9.0 Hz, 2H), 7.04 - 6.94 (m, 2H), 4.63 (dt, J=6.8, 3.4 Hz, IH), 4.51 - 4.39 (m, IH), 4.01 - 3.85 (m, 2H), 3.81 - 3.58 (m, 2H), 3.35 (dd, J=5.5, 4.5 Hz, IH), 2.76 (dd, J=16.4, 3.1 Hz, IH), 2.52 (dd, J=16.3, 9.5 Hz, IH), 2.26 - 2.1 1 (m, 2H), 2.08 - 2.-01 (m, 2H),1.32 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 12.4 min, HI: 99%. hGPR40 EC 50 = 293 nM. [00314] Example 59 to Example 64 were prepared following the procedure for Example 58. Example 59

2-((4S,5S)-l-(4-((l-(2-Chloro-5-fluorophenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00315] Example 59 (white foam, 7 mg): LC-MS Anal. Calc'd for C 2 4H 2 4C1F4 3 03 513.14, found [M+H] 514.0. l H NMR (400MHz, CD 3 CN) δ ppm 7.85 (br. s., 1H), 7.57

(dd, J=8.9, 5.6 Hz, 1H), 7.18 - 7.08 (m, 3H), 7.05 - 6.99 (m, 2H), 4.64 (br. s., 1H), 4.50 - 4.41 (m, 1H), 3.85 (br. s., 2H), 3.46 - 3.28 (m, 3H), 2.75 (dd, J=16.6, 2.5 Hz, 1H), 2.62 - 2.39 (m, 3H), 2.16 (d, J=9.3 Hz, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC

(Orthogonal method, 50% Solvent B start): RT = 10.2 min, HI: 100%. hGPR40 EC 50 = 361 nM.

Example 60

2-((4S,5S)-l-(4-((l-(2-Chloro-5-methylphenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00316] Example 60 (clear oil, 6 mg): LC-MS Anal. Calc'd for C 2 5H2 7 C1F 3 303 509.17, found [M+H] 510.0. l H NMR (400MHz, CD 3 CN) δ ppm 8.21 (br. s., 1H), 7.52 (d, J=8.3 Hz, 1H), 7.36 (d, J=8.3 Hz, 1H), 7.16 - 7.10 (m, 2H), 7.08 - 7.02 (m, 2H), 4.74 (br. s., 1H), 4.51 - 4.41 (m, 1H), 4.15 (br. s., 2H), 3.63 (br. s., 2H), 3.42 - 3.31 (m, 1H), 2.81 - 2.65 (m, 3H), 2.53 (dd, J=16.6, 9.5 Hz, 1H), 2.42 (s, 3H), 2.32 (d, J=13.3 Hz, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 11.7 min, HI: 99%. hGPR40 EC 50 = 364 nM.

Example 61

2-((4S,5S)-l-(4-((l-(3-Fluoro-6-methylpyridin-2-yl)piperi din-4-yl)oxy)phenyl)-4-methyl- -(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00317] Example 61 (yellow solid, 35 mg): LC-MS Anal. Calc'd for C 2 4H2 6 F4 4 03 494.19, found [M+H] 495.1. l H NMR (400MHz, CD 3 CN) δ ppm 7.71 (dd, J=13.1, 8.3 Hz, IH), 7.14 - 7.06 (m, 2H), 7.02 - 6.93 (m, 2H), 6.80 (dd, J=8.2, 3.4 Hz, IH), 4.67 - 4.55 (m, IH), 4.49 - 4.40 (m, IH), 4.08 - 3.94 (m, 2H), 3.84 - 3.68 (m, 2H), 3.46 - 3.29 (m, IH), 2.76 (dd, J=16.4, 3.1 Hz, IH), 2.69 (s, 3H), 2.51 (dd, J=16.3, 9.5 Hz, IH), 2.27 - 2.11 (m, 2H), 2.02 - 1.83 (m, 2H), 1.32 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 7.6 min, HI: 99%. hGPR40 EC 50 = 366 nM.

Example 62

2-((4S,5S)-l-(4-((l-(2-Chloro-4-fluorophenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00318] Example 62 (white foam, 3 mg): LC-MS Anal. Calc'd for C 2 4H 2 4C1F4 3 03 513.14, found [M+H] 514.0. l H NMR (400MHz, CD 3 CN) δ ppm 8.08 (d, J=4.5 Hz, IH), 7.42 - 7.36 (m, IH), 7.30 (dd, J=12.2, 8.9 Hz, IH), 7.16 - 7.08 (m, 2H), 7.05 - 6.98 (m, 2H), 4.68 - 4.60 (m, IH), 4.50 - 4.42 (m, IH), 3.81 - 3.69 (m, 2H), 3.46 - 3.32 (m, 3H), 2.81 - 2.67 (m, IH), 2.58 - 2.42 (m, 2H), 2.19 (br. s., IH), 1.98 - 1.92 (m, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method (column 2), 50% Solvent B start): RT = 10.0 min, HI: 97%. hGPR40 EC 50 = 382 nM.

Example 63

2-((4S,5S)-l-(4-((l-(4-Chloro-2-fluorophenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00319] Example 63 (white foam, 15 mg): LC-MS Anal. Calc'd for C24H24CIF4N3O3

513.14, found [M+H] 514.0. l H NMR (400MHz, CD 3 CN) δ ppm 8.42 (dd, J=6.7, 2.6 Hz,

1H), 7.58 - 7.48 (m, 1H), 7.38 (dd, J=12.0, 9.0 Hz, 1H), 7.16 - 7.09 (m, 2H), 7.07 - 6.99 (m, 2H), 4.75 - 4.66 (m, 1H), 4.51 - 4.36 (m, 1H), 3.97 - 3.82 (m, 2H), 3.62 - 3.48 (m, 2H), 3.44 - 3.28 (m, 1H), 2.76 (dd, J=16.4, 3.1 Hz, 1H), 2.66 - 2.47 (m, 3H), 2.36 - 2.22 (m, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method (column 2), 50% Solvent B start): RT = 6.8 min, HI: 99%. hGPR40 EC 50 = 398 nM.

Example 64

2-((4S,5S)-l-(4-((l-(5-Chloro-2-fluorophenyl)piperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00320] Example 64 (white foam, 9 mg): LC-MS Anal. Calc'd for C 2 4H 2 4C1F4N 3 03

513.14, found [M+H] 514.0. l H NMR (400MHz, CD 3 CN) δ ppm 7.98 (d, J=8.3 Hz, 1H),

7.60 (dd, J=9.0, 5.5 Hz, 1H), 7.24 - 7.17 (m, lH), 7.15 - 7.09 (m, 2H), 7.06 - 7.00 (m, 2H), 4.73 - 4.63 (m, 1H), 4.51 - 4.40 (m, 1H), 4.04 - 3.90 (m, 2H), 3.56 - 3.43 (m, 2H), 3.41 - 3.30 (m, 1H), 2.76 (dd, J=16.4, 3.1 Hz, 1H), 2.62 - 2.47 (m, 3H), 2.31 - 2.18 (m, 2H), 1.33 (d, J=7.3 Hz, 3H). Analytical HPLC (Orthogonal method, 50% Solvent B start): RT = 13.3 min, HI: 98%. hGPR40 EC 50 = 512 nM.

Example 65

2-((4S,5S)-l-(4-((l-(5-Ethyl-2-fluorophenyl)piperidin-4-y l)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00321] Example 65 (grey solid, 26 mg) was prepared following the procedure for Example 32. LC-MS Anal. Calc'd for C26H29F4 3O3 507.22, found [M+H] 508.1. l R NMR (400MHz, CD 3 CN) δ ppm 8.23 (dd, J=7.4, 1.9 Hz, 1H), 7.45 - 7.38 (m, 1H), 7.30

(dd, J=12.5, 8.5 Hz, 1H), 7.15 - 7.07 (m, 2H), 7.06 - 6.99 (m, 2H), 4.72 (br. s., 1H), 4.51 - 4.41 (m, 1H), 3.94 (br. s., 2H), 3.61 (br. s., 2H), 3.45 - 3.31 (m, 1H), 2.81 - 2.61 (m, 4H), 2.52 (dd, J=16.4, 9.7 Hz, 2H), 2.33 (br. s., 2H), 1.32 (d, J=7.3 Hz, 3H), 1.25 (t, J=7.7 Hz, 3H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 13.5 min, HI: 100%. hGPR40 EC 50 = 583 nM. hGPR40 IP1 EC 50 = 297 nM.

Example 66

2-((4S,5S)-l-(4-((l-(2-Fluoro-5-isopropylphenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00322] Example 66 (grey solid, 10 mg) was prepared following the procedure for Example 32. LC-MS Anal. Calc'd for 02^3^4^03 521.23, found [M+H] 522.2. ¾ NMR (400MHz, CD 3 CN) δ ppm 8.29 (dd, J=7.3, 1.8 Hz, 1H), 7.47 - 7.40 (m, 1H), 7.30 (dd, J=12.4, 8.7 Hz, 1H), 7.18 - 7.10 (m, 2H), 7.07 - 7.00 (m, 2H), 4.72 (br. s., 1H), 4.53 - 4.40 (m, 1H), 4.07 - 3.87 (m, 2H), 3.59 (d, J=1 1.3 Hz, 2H), 3.44 - 3.31 (m, 1H), 3.07 - 2.94 (m,lH), 2.81 - 2.58 (m, 3H), 2.52 (dd, J=16.6, 9.5 Hz, 1H), 2.33 (br. s., 2H), 1.33 (d, J=7.3 Hz, 3H), 1.27 (d, J=7.0 Hz, 6H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 13.9 min, HI: 100%. hGPR40 EC 50 = 600 nM.

Example 71

2-((4S,5S)-l-(4-((l-(3-Ethylphenyl)piperidin-4-yl)oxy)phenyl )-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00323] Example 71 (grey solid, 29 mg) was prepared following the procedure for Example 32. LC-MS Anal. Calc'd for C26H30F3 3O3 489.22, found [M+H] 490.0. Ή NMR (400MHz, CD 3 CN) δ ppm 7.98 - 7.67 (m, 2H), 7.49 (t, J=7.9 Hz, 1H), 7.40 (d, J=7.5 Hz, 1H), 7.18 - 7.04 (m, 3H), 4.74 (br. s., 1H), 4.54 - 4.42 (m, 2H), 3.80 (br. s., 2H), 3.46 (br. s., 2H), 3.38 - 3.28 (m, 2H), 2.86 - 2.66 (m, 5H), 2.53 (dd, J=16.6, 9.5 Hz, 2H), 1.33 (d, J=7.0 Hz, 3H), 1.27 (t, J=7.5 Hz, 3H). Analytical HPLC (Orthogonal method (column 2), 10% Solvent B start): RT = 10.1 min, HI: 90%. hGPR40 EC 50 =

1182 nM. hGPR40 IP1 EC 50 = 224 nM.

Example 72

2-((4S,5S)-4-Methyl-l-(4-((l-phenylpiperidin-4-yl)oxy)phe nyl)-3-(trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00324] Example 72 (slightly yellow foam, 46 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C24H26F3 3O3 461.48, found

- I l l - [M+H] 462.1. !H MR (400MHZ, CD 3 CN) δ ppm 7.95 (d, J=7.3 Hz, 2H), 7.70 - 7.46 (m, 3H), 7.22 - 6.94 (m, 4H), 4.72 (br. s., 1H), 4.53 - 4.39 (m, 1H), 3.78 (br. s., 2H), 3.49 (br. s., 1H), 3.36 (td, J=5.6, 1.3 Hz, 1H), 2.92 - 2.69 (m, 3H), 2.53 (dd, J=16.6, 9.5 Hz, 3H), 2.23 (br. s., 1H), 1.32 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 6.3 min, HI: 97%. hGPR40 EC 50 = 1932 nM.

Example 73

2-((4S,5 S)- 1 -(4-(( 1 -(3 -Isopropylphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00325] Example 73 (grey solid, 29 mg) was prepared following the procedure for Example 32. LC-MS Anal. Calc'd for C 27 H3 2 F 3 303 503.24, found [M+H] 504.1. Ή NMR (400MHz, CD 3 CN) δ ppm 7.84 (s, 1H), 7.74 (d, J=8.0 Hz, 1H), 7.54 - 7.47 (m, 1H), 7.46 - 7.40 (m, 1H), 7.17 - 7.10 (m, 2H), 7.06 (br. s., 2H), 4.72 (br. s., 1H), 4.53 - 4.38 (m, 1H), 3.77 (d, J=7.3 Hz, 2H), 3.48 (br. s., 2H), 3.36 (td, J=5.7, 1.4 Hz, 1H), 3.11 - 2.96 (m, 1H), 2.76 (dd, J=16.4, 3.1 Hz, 2H), 2.52 (dd, J=16.6, 9.5 Hz, 2H), 2.33 - 2.15 (m, 2H), 1.32 (d, J=7.0 Hz, 3H), 1.29 (d, J=7.0 Hz, 6H). Analytical HPLC (Orthogonal method, 10% Solvent B start): RT = 9.5 min, HI: 100%. hGPR40 EC 50 = 2121 nM. Example 74

2-((4S,5S)-4-Methyl-l-(4-((l-(pyridin-2-yl)piperidin-4-yl)ox y)phenyl)-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00326] Example 74 (clear oil, 2.5 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C 2 3H 2 5F 3 403 462.19, found [M+H] 462.9. l R NMR (400MHz, CD 3 CN) δ ppm 8.03 - 7.88 (m, IH), 7.20 (d, J=9.3 Hz, IH), 7.14 - 7.07 (m, 2H), 7.04 - 6.96 (m, 2H), 6.90 (t, J=6.7 Hz, IH), 4.64 (tt, J=6.7, 3.5 Hz, IH), 4.51 - 4.40 (m, IH), 4.14 - 3.98 (m, 2H), 3.81 (ddd, J=13.7, 7.3, 3.8 Hz, 2H), 3.41 - 3.29 (m, 2H), 2.76 (dd, J=16.4, 3.1 Hz, IH), 2.52 (dd, J=16.4, 9.4 Hz, IH), 2.22 - 2.04 (m, 2H), 1.94 - 1.84 (m, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 4.9 min, HI: 99%. hGPR40 EC 50 = 2606 nM.

Example 75

2-((4S,5 S)-4-Methyl- 1 -(4-(( 1 -(pyridin-3 -yl)piperidin-4-yl)oxy)phenyl)-3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid, HC1

[00327] Example 75 (clear oil, 2.2 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C23H25F3 4O3 462.19, found [M+H] 463.1. l R NMR (400MHz, CD 3 CN) δ ppm 8.21 (d, J=2.8 Hz, IH), 8.02 - 7.92 (m, 2H), 7.76 (dd,

J=9.0, 5.5 Hz, IH), 7.16 - 7.07 (m, 2H), 7.02 - 6.94 (m, 2H), 4.65 - 4.52 (m, IH), 4.51 - 4.39 (m, IH), 3.79 - 3.66 (m, 2H), 3.49 - 3.32 (m, 3H), 2.75 (dd, J=16.4, 3.1 Hz, 2H), 2.52 (dd, J=16.4, 9.7 Hz, 2H), 2.14 - 2.06 (m, IH), 1.90 - 1.80 (m, IH), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 30% Solvent B start): RT = 5.0 min, HI: 100%. hGPR40 EC = 7680 nM.

Example 76

2-((4S,5S)-l-(4-((l-(3-Chloro-6-methoxypyridin-2-yl)piperidi n-4-yl)oxy)phi

methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)ac etic acid, HC1

[00328] Example 76 (yellow solid, 12 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C 2 4H 26 C1F 3 404 526.16, found [M+H] 527.0. l H NMR (400MHz, CD 3 CN) δ ppm 7.88 (d, J=8.5 Hz, IH), 7.14 - 7.08 (m, 2H), 7.06 - 6.99

(m, 2H), 6.94 (d, J=8.8 Hz, IH), 6.28 (br. s., IH), 4.69 (dt, J=6.0, 2.9 Hz, IH), 4.50 - 4.40 (m, IH), 4.00 (s, 3H), 3.98 - 3.90 (m, 2H), 3.76 - 3.62 (m, 2H), 3.45 - 3.28 (m, IH), 2.76 (dd, J=16.6, 3.0 Hz, IH), 2.65 - 2.47 (m, 3H), 2.36 - 2.25 (m, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 40% Solvent B start): RT = 11.8 min, HI: 100%. hGPR40 EC50 = 649 nM. Example 77

2-((4S,5S)-l-(4-((l-(5-Chloro-2-methoxypyridin-4-yl)piperidi n-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HC1

[00329] Example 77 (white solid, 4 mg) was prepared following the procedure for Example 81 (Isomer 2). LC-MS Anal. Calc'd for C 2 4H 26 C1F 3 404 526.16, found [M+H]

526.8. !H MR (400MHZ, CD 3 CN) δ ppm 7.97 (s, IH), 7.11 (d, J=9.0 Hz, 2H), 7.04 -

6.96 (m, 2H), 6.49 (s, IH), 4.67 - 4.58 (m, IH), 4.51 - 4.42 (m, IH), 4.10 (s, 3H), 3.83 (ddd, J=12.7, 8.5, 3.6 Hz, 3H), 3.61 - 3.52 (m, 3H), 3.42 - 3.32 (m, 2H), 2.76 (dd, J=16.4, 3.1 Hz, IH), 2.52 (dd, J=16.3, 9.5 Hz, IH), 2.26 - 2.10 (m, 2H), 1.33 (d, J=7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 40% Solvent B start): RT = 11.3 min, HI: 100%. hGPR40 EC 5() = 110 nM. hGPR40 ΙΡ1 EC 50 = 20 nM. Acute oral glucose tolerance: - 38% @ 0.3 mg/kg.

Example 78

2-((4S,5S)-l-(4-((l-(4-Chloro-6-methoxypyridin-2-yl)piperidi n-4-yl)oxy)phi

methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)ac etic acid, HC1

[00330] Example 78 (white solid, 1 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C 2 4H 26 C1F 3 404 526.16, found [M+H] 526.8. l H NMR (400MHz, CD 3 CN) δ ppm 8.84 (d, J=2.0 Hz, IH), 8.28 (d, J=2.3 Hz, IH), 7.16 - 7.09 (m, 2H), 7.08 - 7.02 (m, 2H), 4.70 (br. s., IH), 4.46 (d, J=9.5 Hz, IH), 4.12 (s, 3H), 4.03 (d, J=l 1.8 Hz, 2H), 3.55 (br. s., 2H), 3.37 (br. s., 2H), 2.76 (dd, J=16.4, 3.1 Hz, 2H), 2.62 - 2.50 (m, 3H), 2.29 (br. s., 3H), 1.33 (d, J=6.8 Hz, 3H). Analytical HPLC

(Orthogonal method, 40% Solvent B start): RT = 13.6 min, HI: 98%. hGPR40 EC 50 =

1990 nM.

Example 79

2-((4S,5S)-l-(4-((l-(5-Fluoro-2-methoxypyridin-4-yl)piperidi n-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceti c acid, HC1

[00331] Example 79 (white foam, 5 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C 2 4H 26 F 4 404 510.19, found [M+H] 511.0. ¾ NMR (400MHz, CD 3 CN) δ ppm 7.83 (d, J=8.3 Hz, IH), 7.11 (d, J=9.3 Hz, 2H), 7.03 -

6.96 (m, 2H), 6.36 (d, J=7.3 Hz, IH), 4.68 - 4.59 (m, IH), 4.49 - 4.42 (m, IH), 4.08 (s, 3H), 3.99 - 3.90 (m, 2H), 3.68 (ddd, J=13.7, 7.7, 3.5 Hz, 2H), 3.42 - 3.32 (m, IH), 2.76 (dd, J= 16.4, 3.1 Hz, IH), 2.52 (dd, J=16.6, 9.5 Hz, IH), 2.18 - 2.08 (m, 2H), 1.94 - 1.84 (m, 2H), 1.33 (d, J=7.0 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 7.6 min, HI: 98%. hGPR40 EC 50 = 163 nM. hGPR40 IP1 EC 50 = 94 nM.

Example 80 2-((4S,5S)-l-(4-((l-(4-Fluoro-6-methoxypyridin-2-yl)piperidi n-4-yl)oxy)phi

methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)ac etic acid, HC1

[00332] Example 80 (white solid, 9 mg) was prepared following the procedure for Example 58. LC-MS Anal. Calc'd for C 2 4H 26 F 4 404 510.19, found [M+H] 511.0. ¾ NMR (400MHz, CD 3 CN) δ ppm 8.72 (dd, J=8.4, 2.9 Hz, IH), 8.24 (d, J=2.8 Hz, IH),

7.17 - 7.08 (m, 2H), 7.08 - 7.00 (m, 2H), 4.76 - 4.69 (m, IH), 4.49 - 4.43 (m, IH), 4.12 (s, 3H), 4.09 - 4.01 (m, 2H), 3.59 (d, J=11.5 Hz, 2H), 3.37 (ddd, J=7.0, 4.3, 1.5 Hz, 2H), 2.76 (dd, J= 16.4, 3.1 Hz, IH), 2.63 - 2.48 (m, 3H), 2.30 (d, J=13.1 Hz, 2H), 1.36 - 1.31 (m, 3H). HPLC (Orthogonal method, 40% Solvent B start): RT = 12.2 min, HI: 99%. hGPR40 EC50 = 2559 nM.

Example 81

2-((4S,5S)-l-(4-((l-(5-Chloro-2-methoxypyridin-4-yl)-3-methy lpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

Isomer 2

[00333] 81 A. 4-Bromo-5-chloropyridin-2-amine: To a stirred solution of 4- bromopyridin-2 -amine (30 g, 173 mmol) in DMF (350 mL) at -20 °C was added 1- chloropyrrolidine-2,5-dione (24 g, 182 mmol). The reaction mixture was allowed to stir at rt for 24 h. The reaction mixture was poured into a cold solution of 1M NaOH (300 mL) and the mixture was extracted with Et20 (2 x 400 mL). The combined extracts were washed with water (3 x 200 mL), brine (200 mL), dried over a 2 S04, filtered and concentrated. The crude material was recrystallized from DCM which afforded 4-bromo- 5-chloropyridin-2-amine as red solid (22 g, 106 mmol, 61% yield). LC-MS Anal. Calc'd for C 5 H 4 BrClN 2 205.93, found [M+H] 206.9. ¾ NMR (400 MHz, CDC1 3 ) δ 8.08 (s, 1H), 6.81 (s, 1H), 4.49 (br. s., 2H).

[00334] 8 IB. 4-Bromo-5-chloro-2-methoxypyridine: To MeOH (390 mL) was added chlorotrimethylsilane (49 mL, 386 mmol) at 0 °C, and the solution was warmed to rt and stirred for 30 min. To the resulting solution was added 4-bromo-5-chloropyridin-2-amine (20 g, 96 mmol), and the mixture was stirred for 15 min. To the reaction mixture was added sodium nitrite (2.7 g, 40 mmol) and the solution was stirred at 50 °C for 3 h. The reaction mixture was evaporated in vacuo, and the residue was diluted with EtOAc. The pH of the aqueous layer was adjusted to pH = ~12 via addition of 1 N NaOH, and the solution was extracted 3x with EtOAc. The combined organic layers were concentrated, and the residue was purified via recrystallization from MeOH and H 2 0 to give 4-bromo- 5-chloro-2-methoxypyridine as white crystalline needles (18 g, 81 mmol, 84% yield). LC- MS Anal. Calc'd for C 6 H 5 BrClNO 222.92, found [M+H] 223.9. l R NMR (400 MHz, CDCI3) δ 8.18 (s, 1H), 7.08 (s, 1H), 3.94 (s, 4H).

[00335] 81C. Rac-l-benzyl-3-czs-methylpiperidin-4-ol: To a solution of l-benzyl-3- methylpiperidin-4-one (24.8 g, 122 mmol) in THF (102 mL) at -78 °C was added dropwise 1 M solution of L-Selectride (183 mL, 183 mmol) in THF. The reaction mixture was stirred at -78 °C for 1 h 30 min. To this solution were added EtOH (22 mL), water (55 mL) and 1 M NaOH (55 mL). The reaction mixture was warmed to 0 °C and 30% aqueous H 2 0 2 (55 mL) was added dropwise. The reaction mixture was warmed to rt with stirring for 2h. The reaction mixture was diluted with EtOAc and the insoluble white solid was filtered. The filtrate was washed with sat. NaHC03, H 2 0/brine, brine, dried (MgS0 4 ) and concentrated to give the crude product as an oil. Purification via silica gel chromatography gave rac-(3R, 4S)-l-benzyl-3-czs-methylpiperidin-4-ol as a white solid (22.2 g, 88% yield). l H NMR (500 MHz, CDC1 3 ) δ 7.40 - 7.20 (m, 5H), 3.84 (s, 1H), 3.55 (s, 2H), 2.60 - 1.73 (m, 7H), 0.97 (d, 3H). [00336] 8 ID. Rac-l-benzyl-4-c«-((?ert-butyldimethylsilyl)oxy)-3-methylpi peridine: To a solution of rac-l-benzyl-3-czs-methylpiperidin-4-ol (21.9 g, 107 mmol) in CH 2

(107 mL) and triethylamine (45 mL, 320 mmol) at 0 °C was added tert-butyldimethylsilyl trifluoromethanesulfonate (29 mL, 130 mmol). The reaction mixture was stirred at 0 °C for 1 h. Sat. aqueous aHC03 (180 mL) was added slowly to the reaction mixture. The solution was concentrated, diluted with EtOAc, washed successively with H 2 0 and brine, and the resulting organic layer was dried (MgS0 4 ) and concentrated. Purification via silica gel chromatography gave rac-l-benzyl-4-c«-((?ert-butyldimethylsilyl)oxy)-3- methylpiperidine as an oil (32 g, 92% yield). LC-MS Anal. Calc'd for C 19 H 33 NOSi 319.56, found [M+H] 320.3. !H MR (400 MHz, CDC1 3 ) δ 7.33 - 7.19 (m, 5H), 3.73 (d, J=3.3 Hz, 1H), 3.45 (br. s., 2H), 2.44 (br. s., 1H), 2.34 (br. s., 2H), 2.12 (t, J=9.9 Hz, 1H), 1.84 - 1.50 (m, 3H), 0.92 - 0.84 (m, 9H), 0.82 (d, J=7.0 Hz, 3H), 0.00 (s, 6H).

[00337] 8 IE. Rac-cw-4-((tert-butyldimethylsilyl)oxy)-3-methylpiperidine: A mixture of rac- l-benzyl-4-c«-((tert-butyldimethylsilyl)oxy)-3-methylpiperi dine (16 g, 49 mmol) and 10% palladium on carbon (3.2 g) in MeOH (500 mL) was stirred at rt under hydrogen atmosphere (1 atm, balloon) for 24 h. The mixture was filtered through CELITE® and the filtrate was concentrated to give rac-4-cis-((tert- butyldimethylsilyl)oxy)-3-methylpiperidine as an oil (1 1.3 g, 100% yield). ^H MR (500 MHz, CDC1 3 ) δ 3.80 (s, 1H), 2.90 (m, 1H), 2.70 - 2.50 (m, 4H), 1.60 - 1.50 (m, 3H), 0.86 (s, 9H), 0.80 (d, 3H), 0.00(s, 6H).

[00338] 81F. Rac-4-(c«-4-((tert-butyldimethylsilyl)oxy)-3-methylpiperidi n-l-yl)-5- chloro-2-methoxypyridine: A mixture of 4-bromo-5-chloro-2-methoxypyridine (9.7 g, 44 mmol), rac-4-c«-((ter?-butyldimethylsilyl)oxy)-3-methylpiperidine (10 g, 44 mmol), and K 2 C0 3 (12 g, 87 mmol) in DMSO (15 mL) was vigorously stirred at 1 10 °C overnight. The reaction mixture was diluted with water and the mixture was extracted with EtOAc. The organic extract was washed successively with H 2 0 and brine, and the resulting organic layer was dried (MgS0 4 ) and concentrated. Purification via silica gel chromatography gave rac-4-(c«-4-((ter?-butyldimethylsilyl)oxy)-3-methylpiperidi n-l- yl)-5-chloro-2-methoxypyridine as an oil (14.3 g, 77% yield). LC-MS Anal. Calc'd for C 18 H 3 1 ClN 2 O 2 Si:370.18, found [M+H] 371.2. !H MR (400 MHz, CDC1 3 ) δ 7.88 (s, 1H), 6.21 (s, 1H), 3.83 - 3.79 (m, 4H), 3.19 (dtd, J=11.7, 3.9, 1.8 Hz, 1H), 3.07 - 2.99 (m, 2H), 2.78 (t, J=1 1.0 Hz, 1H), 1.93 - 1.81 (m, 1H), 1.81 - 1.74 (m, 1H), 1.73 - 1.63 (m, 1H), 0.89 - 0.82 (m, 12H), 0.00 (s, 6H).

[00339] 81G. Rac-l-(5-chloro-2-methoxypyridin-4-yl)-3-cw-methylpiperidin- 4-ol: To a mixture of rac-4-(c«-4-((tert-butyldimethylsilyl)oxy)-3-methylpiperidi n-l-yl)-5-chloro-

2- methoxypyridine (10 g, 27 mmol)) in THF (27 mL) was added TBAF (81 mL, 81 mmol). The reaction was stirred at 23 °C for 16 h. To the reaction mixture was slowly added sat. aq. aHC03 (100 mL) and the resulting mixture was extracted with EtOAc (2 x 100 mL). The combined organic extracts were washed successively with water (50 mL) and brine (50 mL), and the organic layer was dried ( a 2 S04), filtered, and concentrated. Purification via silica gel chromatography gave rac-l-(5-chloro-2-methoxypyridin-4-yl)-

3- cz ' s-methylpiperidin-4-ol as white foam (7g, 99% yield). LC-MS Anal. Calc'd for C 12 H 17 ClN 2 O 2 :256.10, found [M+H] 257.0. ¾ NMR (400 MHz, CDC1 3 ) δ 7.98 (s, 1H), 6.30 (s, 1H), 3.99 - 3.94 (m, 1H), 3.91 (s, 3H), 3.30 - 3.21 (m, 1H), 3.18 - 3.09 (m, 2H), 2.93 (dd, J=11.5, 10.0 Hz, 1H), 2.08 (qd, J=6.8, 3.1 Hz, 1H), 2.02 - 1.87 (m, 2H), 1.42 (d, J=3.8 Hz, 1H), 1.06 (d, J=7.0 Hz, 3H).

[00340] 81H. Methyl 2-((4S,5S)-l-(4-((l-(5-chloro-2-methoxypyridin-4-yl)-3-?rara - methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetate: To a solution of methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetate (1 J, 11 1 mg, 0.35 mmol) and rac- l-(5-chloro-2-methoxypyridin-4-yl)-3-cw-methylpiperidin-4-ol (100 mg, 0.39 mmol) in THF (130 μΐ) was added triphenylphosphine (143 mg, 0.55 mmol). The reaction vessel was then lowered into a sonication bath and was sonicated for several minutes giving a clear and highly viscous solution. While sonicating, (E)-diethyl diazene-l,2-dicarboxylate (74 μί, 0.47 mmol) was added dropwise to the reaction mixture, and sonication was continued for 120 min. To the reaction mixture was slowly added sat. aq. NaHCC^ (10 mL), and the mixture was extracted with EtOAc (2 x 10 mL). The combined organic extracts were washed successively with water (20 mL) and brine (20 mL), and the organic layer was dried (Na 2 S0 4 ), filtered, and concentrated. Purification by chromatography gave methyl 2-((4S,5S)-l-(4-((-l-(5-chloro-2-methoxypyridin-4-yl)-3-?ra s- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetate (65 mg, 0.12 mmol, 30% yield) as white foam. LC-MS Anal. Calc'd for C 26 H3 0 ClF 3 4O4: 554.19, found [M+H] 555.1. !H MR (400 MHz, CDC1 3 ) δ 8.00 (s, 1H), 7.1 1 - 7.04 (m, 2H), 6.98 - 6.89 (m, 2H), 6.29 (s, 1H), 4.41 (d, J=10.1 Hz, 1H), 3.91 (s, 3H), 3.77 - 3.71 (m, 4H), 3.54 (dd, J=12.0, 2.4 Hz, 2H), 3.28 - 3.17 (m, 1H), 2.94 - 2.64 (m, 3H), 2.43 (dd, J=16.0, 10.2 Hz, 1H), 2.20 (dd, J=8.8, 3.8 Hz, 2H), 1.92 - 1.77 (m, 1H), 1.36 (d, J=7.1 Hz, 3H), 1.19 - 1.13 (m, 3H).

[00341] Example 81 (Isomers 1 and 2): To a stirred solution of methyl 2-((4S,5S)-l-(4- ((l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl) oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetate (65 mg, 0.12 mmol) in THF (0.53 mL) and water (53 μΚ) at rt was added IN LiOH solution (350 μί, 0.35 mmol). The reaction mixture was stirred at rt for 1 h. The reaction mixture was cooled to 0 °C and a solution of 3N HC1 (0.4 mL) was added. The reaction mixture was warmed to rt. The solvent was evaporated and the residue was dissolved in CH 3 CN, filtered. Purification by RP-Prep HPLC afforded a diastereomeric mixture. The diastereomers were separated by chiral Prep. SFC. Product-containing fractions were evaporated and the residue was dissolved in DCM and was treated with 3N HC1 (3 mL). The resulting mixture was concentrated to provide Example 81, Isomer 1 and Isomer 2 as single stereoisomers. Example 81, Isomer 1 (white foam, 20 mg) LC-MS Anal. Calc'd for C 2 5H2 8 C1F 3 404

540.18, found [M+H] 541.0. l H NMR (400 MHz, CD 3 CN) δ ppm 8.02 - 7.92 (m, 1H), 7.18 - 7.05 (m, 2H), 7.03 - 6.92 (m, 2H), 6.48 (s, 1H), 4.51 - 4.40 (m, 1H), 4.25 (br.s,

1H), 4.16 (td, J=8.5, 4.0 Hz, 1H), 4.1 1 (s, 3H), 3.99 (dd, J=13.2, 3.6 Hz, 2H), 3.43 - 3.32 (m, 2H), 3.16 (dd, J=13.3, 9.5 Hz, 1H), 2.77 (dd, J=16.4, 3.1 Hz, 1H), 2.52 (dd, J=16.4, 9.7 Hz, 1H), 2.31 - 2.21 (m, 1H), 2.16 - 2.04 (m, 1H), 1.82 - 1.68 (m, 1H), 1.33 (d, J=7.0 Hz, 3H), 1.13 (d, J=6.8 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 11.9 min, HI: 97%. hGPR40 EC 50 = 319 nM. Example 81 , Isomer 2 (white foam, 20 mg)

LC-MS Anal. Calc'd for C 2 5H 28 C1F 3 4 04 540.18, found [M+H] 541.0. !H MR (400 MHz, CD 3 CN) δ ppm 8.02 - 7.92 (m, 1H), 7.18 - 7.05 (m, 2H), 7.03 - 6.92 (m, 2H), 6.50 (br.s, 1H), 6.48 (s, 1H), 4.51 - 4.40 (m, 1H), 4.16 (td, J=8.5, 4.0 Hz, 1H), 4.1 1 (s, 3H), 3.99 (dd, J=13.2, 3.6 Hz, 2H), 3.43 - 3.32 (m, 2H), 3.16 (dd, J=13.3, 9.5 Hz, 1H), 2.77 (dd, J=16.4, 3.1 Hz, 1H), 2.52 (dd, J=16.4, 9.7 Hz, 1H), 2.31 - 2.21 (m, 1H), 2.16 - 2.04 (m, 1H), 1.82 - 1.68 (m, 1H), 1.33 (d, J=7.0 Hz, 3H), 1.13 (d, J=6.8 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 1 1.9 min, HI: 97%. hGPR40 EC 50 = 71 nM.

[00342] 811. (3R,4S)-l-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperidin -4-ol: Rac-l-(5-chloro-2-methoxypyridin-4-yl)-3-cw-methylpiperidin- 4-ol (81G, 9.6 g, 37.5 mmol) isomers were separated by chiral Prep. SFC to provide (3R,4S)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-ol (white foam, 4 g, 15.6 mmol) as a single enantiomer. LC-MS Anal. Calc'd for C 12 H 17 ClN 2 O 2 :256.10, found [M+H] 257.0. l R NMR (400 MHz, CDC1 3 ) δ 7.98 (s, 1H), 6.30 (s, 1H), 3.99 - 3.94 (m, 1H), 3.91 (s, 3H),

3.30 - 3.21 (m, 1H), 3.18 - 3.09 (m, 2H), 2.93 (dd, J=1 1.5, 10.0 Hz, 1H), 2.08 (qd, J=6.8, 3.1 Hz, 1H), 2.02 - 1.87 (m, 2H), 1.42 (d, J=3.8 Hz, 1H), 1.06 (d, J=7.0 Hz, 3H).

[00343] 81 J. 2-((4S,5 S)- 1 -(4-(((3R,4R)- 1 -(5-Chloro-2-methoxypyridin-4-yl)-3 - methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetate: To a solution of methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetate (U, 4.6 g, 15 mmol) and (3R,4S)- l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-ol (3.4 g, 13 mmol) in toluene (166 mL) was added tributylphosphine (5.5 mL, 21 mmol). While stirring, (E)-diazene- l,2-diylbis(piperidin-l-ylmethanone) (5.4 g, 21 mmol) was added portionwise to the reaction mixture, and the resulting mixture was heated at 50 °C for 120 min. The mixture was cooled to rt and 150 mL of hexanes was added to the mixture and a white precipitate formed. The mixture was filtered, and the filtrate was concentrated and was purified via silica gel chromatography to give methyl 2-((4S,5S)-l-(4-(((3R,4R)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-me thyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetate (4.7 g, 8.5 mmol, 64% yield). LC-MS Anal. Calc'd for C26H30CIF3 4O4 554.19, found [M+H] 555.2. ¾ NMR (400 MHz, CDC1 3 ) δ ppm 8.00 (s, 1H), 7.15 - 7.04 (m, 2H), 7.01 - 6.89 (m, 2H), 6.30 (s, 1H), 4.49 - 4.36 (m, 1H), 3.91 (s, 4H), 3.73 (s, 3H), 3.62 - 3.49 (m, 2H), 3.28 - 3.15 (m, 1H), 2.95 - 2.85 (m, 1H), 2.82 (dd, J= 16.2, 3.2 Hz, 1H), 2.67 (dd, J=12.3, 9.0 Hz, 1H), 2.43 (dd, J=16.1, 10.3 Hz, 1H), 2.26 - 2.12 (m, 2H), 1.91 - 1.78 (m, 1H), 1.36 (d, J=7.0 Hz, 3H), 1.16 (d, J=6.6 Hz, 3H).

[00344] Example 81, Isomer 2 (neutral form): To a stirred solution of methyl 2- ((4S,5S)-l-(4-(((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3 -methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5 -dihydro- 1 H-pyrazol-5 -yl)acetate (5.5 g, 9.9 mmol) in THF (90 mL) and water (9 mL) at rt was added 2N LiOH solution (12 mL, 24 mmol). The reaction mixture was stirred at rt for 16 h and IN HC1 (25 mL, 25 mmol) was added at 0 °C to pH = 4-5. The solvent was evaporated and the residue was extracted 3x with EtOAc. The organic extracts were dried over a 2 S04; the solution was filtered and concentrated. The residue was recrystallized from isopropanol to give Example 81, Isomer 2 (neutral form) as white solid (4.3 g, 7.7 mmol, 78% yield). LC-MS Anal. Calc'd for C 2 5H 28 C1F 3 404 540.18, found [M+H] 541.0. l H NMR (500 MHz, DMSO-d 6 ) δ ppm 12.52 (br. s., 1H), 8.01 (s, 1H), 7.05 (d, J=9.1 Hz, 2H), 6.96 (d, J=9.1 Hz, 2H), 6.40 (s, 1H), 4.49 - 4.33 (m, 1H), 4.02 (td, J=8.8, 4.1 Hz, 1H), 3.80 (s, 3H), 3.56 - 3.39 (m, 2H), 3.37 - 3.29 (m, 1H), 2.94 - 2.85 (m, 1H), 2.72 - 2.66 (m, 1H), 2.64 (dd, J=16.1, 2.9 Hz, 1H), 2.49 - 2.41 (m, 1H), 2.22 - 2.05 (m, 1H), 2.01 - 1.86 (m, 1H), 1.68 - 1.50 (m, 1H), 1.25 (d, J=7.2 Hz, 3H), 1.03 (d, J=6.9 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 1 1.9 min, HI: 97%. hGPR40 EC 50 = 71 nM. hGPR40 IP1 EC 50 = 9 nM. Acute oral glucose tolerance: -54% @ 0.3 mg/kg.

Form N- 1 of Example 81 , Isomer 2

[00345] Crystals suitable for X-ray diffraction analysis were grown by adding 3 mg of Example 81, Isomer 2 (neutral form) to 0.7 mL of ethyl acetate. Colorless prism shaped crystals were obtained after one day of slow evaporation of solution at room temperature. A single crystal was isolated from the solution for X-ray analysis.

[00346] Single crystal data were collected on a Bruker-AXS APEX2 CCD system using Cu Ka radiation (λ = 1.5418 A). Indexing and processing of the measured intensity data were carried out with the APEX2 software package/program suite (APEX2 Data collection and processing user interface: APEX2 User Manual, vl.27; BRUKER AXS, Inc., 5465 East Cheryl Parkway Madison, WI 5371 1 USA).

[00347] When indicated, crystals were cooled in the cold stream of an Oxford cryo system (Oxford Cryosystems Cryostream cooler: Cosier, J. et al, J. Appl. Cryst., 19: 105 (1986)) during data collection.

[00348] The structures were solved by direct methods and refined on the basis of observed reflections using the crystallographic package SHELXTL (APEX2 Data collection and processing user interface: APEX2 User Manual, vl.27; BRUKER AXS, Inc., 5465 East Cheryl Parkway Madison, WI 5371 1 USA).

[00349] The derived atomic parameters (coordinates and temperature factors) were refined through full matrix least-squares. The function minimized in the refinements was ∑ W (|F 0 | - |F C |)2. R is defined as∑ ||F 0 | - |FJ/∑ |F 0 | while R w = [∑ W (|F 0 | - |F C |) 2 /∑ W

|F 0 | 2 ] 1/2 where w is an appropriate weighting function based on errors in the observed intensities. Difference maps were examined at all stages of refinement. Hydrogens were introduced in idealized positions with isotropic temperature factors, but no hydrogen parameters were varied.

Crystal Structure Data of the Form l of Example 81, Isomer 2

Unit cell dimensions:

a = 10.1890(3) A

b = 13.4473(6) A

c = 18.8524(7) A

a = 90°

^ = 90°

y = 90°

Volume = 2583.05(17) A 3

Crystal system: Orthorhombic

Space group: 2 1 2 1 2 1

Molecules/asymmetric unit: 1

Density (calculated) = 1.391 g/cm 3

Measurement of the crystalline form is at a temperature of about 23 °C.

[00350] The unit cell parameters were obtained from single crystal X-ray

crystallographic analysis according to the procedure described in Stout et al, X-ray Structure Determination: A Practical Guide, MacMillian (1968), previously herein incorporated by reference.

Table 1 : Fractional Atomic Coordinates for the Form 1 of Example 81 , Isomer 2 at rt ATOM X Y Z ATOM X Y Z

1 CI 0.8041 0.2154 0.4137 34 H17 0.1774 0.1229 -0.0341

2 C2 0.9001 0.2380 0.3555 35 H18 0.1294 0.0133 -0.0465

3 HI 0.9426 0.3023 0.3640 36 C18 0.1973 0.0361 0.0550

4 C3 0.8049 0.2453 0.2919 37 H19 0.1478 -0.0202 0.0745

5 H2 0.8371 0.2041 0.2527 38 C19 0.3353 -0.0002 0.0394

6 C4 0.7813 0.3516 0.2658 39 H20 0.3315 -0.0561 0.0078

7 H3 0.6987 0.3547 0.2403 40 H21 0.3771 -0.0197 0.0829

8 H4 0.7756 0.3962 0.3062 41 H22 0.3848 0.0523 0.0176

9 C5 0.8909 0.3843 0.2180 42 C20 -0.0942 0.0974 -0.0546

10 C6 1.0036 0.1563 0.3465 43 C21 -0.0565 0.0934 -0.1249

11 H5 1.0554 0.1516 0.3888 44 H23 0.0322 0.0935 -0.1365

12 H6 1.0592 0.1721 0.3070 45 C22 -0.1500 0.0891 -0.1782

13 H7 0.9607 0.0939 0.3379 46 C23 -0.3160 0.0884 -0.0978

14 C7 0.8340 0.2163 0.4898 47 H24 -0.4054 0.0853 -0.0879

15 C8 0.5723 0.1745 0.2834 48 C24 -0.2305 0.0944 -0.0419

16 C9 0.5695 0.1799 0.2100 49 C25 -0.1909 0.0901 -0.3024

17 H8 0.6428 0.2025 0.1855 50 H25 -0.2472 0.0330 -0.3000

18 CIO 0.4584 0.1517 0.1729 51 H26 -0.1428 0.0891 -0.3462

19 H9 0.4572 0.1558 0.1236 52 H27 -0.2430 0.1495 -0.3001

20 C11 0.3489 0.1176 0.2087 53 CLl -0.2951 0.0883 0.0430

21 C12 0.3521 0.1127 0.2816 54 Fl 0.7345 0.1997 0.5309

22 H10 0.2785 0.0900 0.3060 55 F2 0.9228 0.1516 0.5089

23 C13 0.4616 0.1408 0.3189 56 F3 0.8838 0.3020 0.5093

24 H11 0.4617 0.1373 0.3682 57 Nl 0.6849 0.2017 0.3213

25 C14 0.1930 0.1193 0.1095 58 N2 0.6882 0.1927 0.3928

26 H12 0.2501 0.1739 0.0942 59 N3 -0.0048 0.0985 0.0013

27 C15 0.0540 0.1576 0.1196 60 N4 -0.2780 0.0869 -0.1666

28 H13 0.0014 0.1067 0.1424 61 01 0.8794 0.3954 0.1554

29 H14 0.0555 0.2154 0.1503 62 02 1.0029 0.3957 0.2518

30 C16 -0.0076 0.1852 0.0491 63 H28 1.0760 0.4130 0.2247 31 H15 -0.0976 0.2065 0.0564 64 03 0.2354 0.0831 0.1775

32 H16 0.0405 0.2400 0.0278 65 04 -0.1014 0.0883 -0.2444

33 C17 0.1295 0.0681 -0.0130

[00351] Powder X-ray diffraction (PXRD) data were obtained using a Bruker C2 GADDS. The radiation was Cu Ka (40 KV, 40mA). The sample-detector distance was 15 cm. Powder samples were placed in sealed glass capillaries of 1mm or less in diameter; the capillary was rotated during data collection. Data were collected approximately for 2<2Θ<35° with a sample exposure time of at least 1000 seconds. The resulting two-dimensional diffraction arcs were integrated to create a traditional 1 - dimensional PXRD pattern with a step size of 0.05 degrees 2Θ in the approximate range of 2 to 35 degrees 2Θ.

[00352] Characteristic diffraction peak positions (degrees 2Θ±0.1) at rt are based on a high quality pattern collected with a diffractometer (CuKa) with a spinning capillary with 2Θ calibrated with a NIST other suitable standard.

[00353] Differential scanning calorimetry (DSC) experiments were performed in a TA INSTRUMENTS® model Q2000, Q1000 or 2920. The sample (about 1-10 mg) was weighed in an aluminum pan and accurately recorded to a hundredth of a milligram, and transferred to the DSC. The instrument was purged with nitrogen gas at 50mL/min. Data were collected between room temperature and 300 °C at 10 °C/min heating rate. The plot was made with the endothermic peaks pointing down.

[00354] Thermogravimetric analysis (TGA) experiments were performed in a TA INSTRUMENTS® model Q5000, Q500 or 2950. The sample (about 4-30 mg) was placed in a platinum pan previously tared. The weight of the sample was measured accurately and recorded to a thousandth of a milligram by the instrument. The furnace was purged with nitrogen gas at 100 mL/min. Data were collected between room temperature and 300 °C at 10 °C/min heating rate.

[00355] Moisture sorption isotherms were collected in a VTI SGA-100 Symmetric Vapor Analyzer using approximately 10 mg of sample. The sample was dried at 60 °C until the loss rate of 0.0005 wt.%/min was obtained for 10 minutes. The sample was tested at 25 °C and 3 or 4, 5, 15, 25, 35, 45, 50, 65, 75, 85, and 95% RH. Equilibration at each RH was reached when the rate of 0.0003 wt%/min for 35 minutes was achieved or a maximum of 600 minutes.

Example 82

2-((4S,5S)-l-(4-(((3S,4S)-l-(5-Chloro-2-ethoxypyridin-4-yl)- 3-methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid, HC1

[00356] Example 82 (white foam, 15 mg) was prepared following the procedure for Example 81. LC-MS Anal. Calc'd for C26H30CIF3N4O4 554.19, found [M+H] 555.0. l R NMR (400MHz, CD 3 CN) δ ppm 7.98 (s, 1H), 7.09 (d, J=9.0 Hz, 2H), 6.97 (d, J=9.0 Hz,

2H), 6.34 (s, 1H), 4.50 - 4.40 (m, 1H), 4.30 (q, J=7.0 Hz, 2H), 4.00 (td, J=9.0, 4.1 Hz, 1H), 3.61 - 3.49 (m, 2H), 3.41 - 3.31 (m, 1H), 2.96 - 2.86 (m, 1H), 2.79 - 2.63 (m, 2H), 2.51 (dd, J=16.6, 9.5 Hz, 1H), 2.19 - 1.93 (m, 2H), 1.79 - 1.67 (m, 1H), 1.37 - 1.31 (m, 6H), 1.12 (dd, J=6.5, 4.0 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 13.5 min, HI: 100%. hGPR40 EC 50 = 760 nM.

Example 83

2-((4S,5S)-l-(4-(((3R,4R)-l-(5-Chloro-2-ethoxypyridin-4-y l)-3-methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid,

[00357] Example 83 (white foam, 15 mg) was prepared following the procedure for Example 81. LC-MS Anal. Calc'd for C 26 H3 0 ClF 3 4O4 554.19, found [M+H] 555.0. l H NMR (400MHz, CD 3 CN) δ ppm 7.98 (s, 1H), 7.1 1 - 7.06 (m, 2H), 6.99 - 6.95 (m, 2H), 6.34 (s, 1H), 4.49 - 4.41 (m, 1H), 4.30 (q, J=7.0 Hz, 2H), 4.01 (td, J=8.9, 4.3 Hz, 1H), 3.61 - 3.49 (m, 2H), 3.40 - 3.31 (m, 1H), 2.96 - 2.87 (m, 1H), 2.79 - 2.67 (m, 2H), 2.51 (dd, J=16.6, 9.5 Hz, 1H), 2.19 - 1.93 (m, 2H), 1.78 - 1.67 (m, 1H), 1.37 - 1.30 (m, 6H), 1.12 (dd, J=6.3, 3.5 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 13.5 min, HI: 100%. hGPR40 EC 50 = 224 nM.

Example 84

2-((4S,5S)-l-(4-(((3S,4S)-l-(2-Ethoxy-5-fluoropyridin-4-yl)- 3-methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid, HCl

[00358] Example 84 (white foam, 20 mg) was prepared following the procedure for Example 81. LC-MS Anal. Calc'd for C 26 H3 0 F 4 4O4 538.22, found [M+H] 539.2. ¾ NMR (400MHz, CD 3 CN) δ ppm 7.85 - 7.69 (m, 1H), 7.17 - 7.04 (m, 2H), 7.02 - 6.89 (m,

2H), 6.29 - 6.12 (m, 1H), 4.44 (d, J=9.0 Hz, 1H), 4.26 (q, J=7.0 Hz, 2H), 4.00 (td, J=9.0, 3.9 Hz, 1H), 3.71 - 3.58 (m, 2H), 3.34 (d, J=5.5 Hz, 1H), 3.07 - 2.93 (m, 2H), 2.86 - 2.65 (m, 2H), 2.46 (dd, J=16.2, 9.7 Hz, 1H), 2.21 - 2.12 (m, 1H), 2.02 (dt, J=4.8, 2.5 Hz, 1H), 1.73 - 1.58 (m, 1H), 1.38 - 1.29 (m, 6H), 1.09 (d, J=6.5 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 7.5 min, HI: 99%. hGPR40 EC 50 = 2575 nM.

Example 85

2-((4S,5S)-l-(4-(((3R,4R)-l-(2-Ethoxy-5-fluoropyridin-4-yl)- 3-methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid,

HCl

[00359] Example 85 (white foam, 20 mg) was prepared following the procedure for Example 81. LC-MS Anal. Calc'd for C 26 H3 0 F 4 4O4 538.22, found [M+H] 539.2. ¾ NMR (400MHz, CD 3 CN) δ ppm 7.85 - 7.69 (m, 1H), 7.17 - 7.04 (m, 2H), 7.02 - 6.89 (m,

2H), 6.29 - 6.12 (m, 1H), 4.44 (d, J=9.0 Hz, 1H), 4.26 (q, J=7.0 Hz, 2H), 4.00 (td, J=9.0, 3.9 Hz, 1H), 3.71 - 3.58 (m, 2H), 3.34 (d, J=5.5 Hz, 1H), 3.07 - 2.93 (m, 2H), 2.86 - 2.65 (m, 2H), 2.46 (dd, J=16.2, 9.7 Hz, 1H), 2.21 - 2.12 (m, 1H), 2.02 (dt, J=4.8, 2.5 Hz, 1H), 1.73 - 1.58 (m, 1H), 1.38 - 1.29 (m, 6H), 1.09 (d, J=6.5 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 7.6 min, HI: 100%. hGPR40 EC 50 = 277 nM.

Example 86

2-(l-(4-((l-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperid in-4-yl)oxy)phenyl)-4- ethyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid (Isomers 1, 2 and 3)

[00360] 86A. 2,2,2-Trifluoro-N'-(4-methoxyphenyl)acetohydrazide: To a stirred suspension of (4-methoxyphenyl)hydrazine (6 g, 43.4 mmol) in THF (150 mL) at 0 °C was added TFAA (6.0 mL, 43.4 mmol) dropwise. The resulting mixture was stirred at 0 °C for 2 h. LCMS showed the completion of the reaction. The mixture was then concentrated and purified by chromatography afforded 2,2,2-trifluoro-N'-(4- methoxyphenyl)acetohydrazide (9 g, 38.4 mmol, 89% yield) as red solid. LC-MS Anal. Calc'd for C9H9F3 2O2 234.06, found [M+H] 235.0.

[00361] 86B. (Z)-2,2,2-Trifluoro-N'-(4-methoxyphenyl)acetohydrazonoyl chloride: To a stirred solution of 2,2,2-trifluoro-N'-(4-methoxyphenyl)acetohydrazide (5 g, 21.4 mmol) in EtOAc (100 mL) at 0 °C was added benzenesulfonyl chloride (3.4 mL, 25.6 mmol) followed by N-ethyl-N-isopropylpropan-2-amine (5.6 mL, 32.0 mmol) dropwise. The resulting mixture was then warmed up to rt and stirred at rt for 12 h. LCMS showed the completion of the reaction. Then, sat. aq. NaHCC^ (50 mL) was added slowly to the reaction mixture. The mixture was then extracted with EtOAc (2x50 mL), and the combined organic extracts were dried ( a 2 S04), filtered, and concentrated. Purification by chromatography gave (Z)-2,2,2-trifluoro-N'-(4-methoxyphenyl)acetohydrazonoyl chloride (3 g, 11.9 mmol, 56% yield) as red oil. LC-MS Anal. Calc'd for C 9 H 8 C1F 3 N 2 0 252.03, found [M-H] 251.0.

[00362] 86C. Methyl 4-ethyl-l -(4-methoxyphenyl)-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazole-5-carboxylate: To a mixture of (Z)-2,2,2-trifluoro-N'-(4- methoxyphenyl)acetohydrazonoyl chloride (3.4 g, 13.46 mmol) and (E)-methyl pent-2- enoate (1.5 g, 13.5 mmol) in 1,4-dioxane (27 mL) was added silver carbonate (3.7 g, 13.5 mmol). The mixture was purged with argon three times. Then, the mixture was heat to 65 °C for overnight. The mixture was filtered through CELITE® and washed with DCM. The combined filtrates were concentrated. Purification by chromatography gave methyl 4- ethyl- 1 -(4-methoxyphenyl)-3 -(trifluoromethyl)-4,5 -dihydro- 1 H-pyrazole-5-carboxylate (2.1 g, 6.4 mmol, 47% yield) as a yellow oil. LC-MS Anal. Calc'd for C 15 H 17 F 3 203 330.12, found [M+H] 331.1.

[00363] 86D. 4-Ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5-dihydro- lH- pyrazol-5-yl)methanol: Dissolved methyl 4-ethyl-l-(4-methoxyphenyl)-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazole-5-carboxylate (2.3 g, 6.96 mmol) in EtOH (23.2 mL) and THF (11.61 mL) and added NaBH 4 (0.53 g, 13.93 mmol) slowly into the solution at 23 °C. After lhour, 10% KHS0 4 (10 mL) was added slowly to the reaction mixture. The mixture was then extracted with EtOAc (2x30 mL), and the combined organic extracts were dried (Na 2 S0 4 ), filtered, and concentrated. Purification via silica gel chromatography gave 4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)methanol (1.8 g, 5.95 mmol, 86%> yield) as an oil. LC-MS Anal. Calc'd for C 14 H 17 F 3 202 302.12, found [M+H] 303.0.

[00364] 86E. 4-Ethyl- 1 -(4-methoxyphenyl)-3 -(trifluoromethyl)-4,5-dihydro- 1 H- pyrazol-5-yl)methyl methanesulfonate: To a solution of 4-ethyl-l-(4-methoxyphenyl)-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)methanol (2.2 g, 7.28 mmol) in dichloromethane (14.5 mL) added triethylamine (5.0 mL, 36.4 mmol) and

methanesulfonyl chloride (1.7 mL, 21.8 mmol) at 0 °C and stirred for 30 mins. LCMS showed that the reaction was completed. TLC (1 :2 EtOAc:Hexanes) showed starting material were completely consumed. Then, sat. NaHC0 3 (200 mL) was added slowly to the reaction mixture. The mixture was then extracted with EtOAc (2x200 mL), and the combined organic extracts were dried (Na 2 S0 4 ), filtered, and concentrated. Purification via silica gel chromatography gave (4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)methyl methanesulfonate (2.4 g, 6.31 mmol, 87% yield) as a white foam. LC-MS Anal. Calc'd for C 15 H 19 F 3 N 2 0 4 S 380.10, found [M+H] 381.0.

[00365] 86F. 2-(4-Ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5-dihyd ro-lH- pyrazol-5-yl)acetonitrile: A solution of 4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)methyl methanesulfonate (1.24 g, 3.26 mmol) in DMSO (6.5 mL))was treated with cyanopotassium (0.32 g, 4.89 mmol) and stirred at 40 °C under argon. Reaction was monitored by LC/MS. After 16 h, LCMS showed completion. After cooling to rt, it was diluted with NaHC0 3 (-200 mL) and EtOAc (200 mL). After separation of the layers, the aqueous layer was then extracted with EtOAc (3x200mL). The combined organic extracts were washed with water (3x), brine, dried ( a2S04), and concentrated. Purification via silica gel chromatography gave 2-(4-ethyl-l-(4- methoxyphenyl)-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5- yl)acetonitrile as an oil. LC-MS Anal. Calc'd for C 15 H 16 F 3 N 3 0 31 1.13, found [M+H] 312.0.

[00366] 86G. 2-(4-Ethyl-l-(4-hydroxyphenyl)-3-(trifluoromethyl)-4,5-dihyd ro-lH- pyrazol-5-yl)acetonitrile: To a stirred solution of 2-(4-ethyl-l-(4-methoxyphenyl)-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (900 mg, 2.89 mmol) in DCM (14 mL) at -78 °C was added tribromoborane (547 μΐ, 5.78 mmol). After the addition was complete, the reaction mixture was stirred at -78 °C for 15 min and then allowed to warm up to 0 °C over a 2 h period. LCMS showed no starting material. The reaction mixture was quenched at 0 °C with dry MeOH (10 mL) and then allowed to warm up to rt. The mixture was evaporated and, the residue was partitioned between 0.1M HC1 (20 mL) and CH2CI2 (40 mL). The aqueous layer was extracted with CH2CI2 (2x20 mL) and, the combined organic layers were dried ( a2S0 4 ) and concentrated. Purification via silica gel chromatography gave 2-(4-ethyl-l-(4-hydroxyphenyl)-3-(trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetonitrile (600 mg, 2.02 mmol, 70% yield) as a yellow oil. LC-MS Anal. Calc'd for C^H^^O 297.1 1, found [M+H] 298.0.

[00367] 86H. 2-(l-(4-((l-(5-Chloro-2-methoxypyridin-4-yl)-3-methylpiperid in-4- yl)oxy)phenyl)-4-ethyl-3-(trifluoromethyl)-4,5-dihydro-lH-py razol-5-yl)acetonitrile: To a solution of 2-(4-ethyl-l-(4-hydroxyphenyl)-3-(trifluoromethyl)-4,5-dihyd ro-lH- pyrazol-5-yl)acetonitrile (174 mg, 0.58 mmol) and (3R,4S)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-ol (150 mg, 0.58 mmol) in THF (195 μΐ) was added triphenylphosphine (215 mg, 0.82 mmol). The reaction vessel was then lowered into a sonication bath and sonicated for several minutes (to allow for mixing) giving a clear and highly viscous solution. While sonicating, (E)-di-tert-butyl diazene-1,2- dicarboxylate (161 mg, 0.70 mmol) was added dropwise to the reaction mixture, and the reaction mixture was sonicated for 120 min. LCMS showed the desired mass of product. Then, sat. aq. NaHCC^ (10 mL) was added slowly to the reaction mixture. The mixture was then extracted with EtOAc (2x10 mL), and the combined organic extracts were washed with water (20 mL) and brine (20 mL), and dried (Na 2 S0 4 ), filtered, and concentrated. Purification by chromatography gave 2-((4S,5S)-l-(4-((l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-et hyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (80 mg, 0.15 mmol, 26% yield) an oil. LC-MS Anal. Calc'd for C 26 H 2 9C1F3 5 02 535.20, found [M+H] 536.0.

[00368] Example 86 (Isomers 1, 2 and 3): To a solution of 3.6 M HC1 in

MeOH/MeOAc (326 μί, 1.174 mmol) was added to 2-( 1 -(4-(( 1 -(5 -chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-et hyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (37 mg, 0.069 mmol) and the reaction stirred at rt for 48 h. The reaction mixture was diluted with acetonitrile and was evaporated to remove methanol and acetonitrile. The residue was dissolved in ethyl acetate and washed with a saturated aqueous NaHC0 3 (2 x 100 mL), water, and brine. The organic layer was then dried ( a 2 S04), filtered, and concentrated. The residue was dissolved in THF (124 μΐ) and water (13 μΐ) at rt was added 2M LiOH (345 μΐ, 0.69 mmol). The reaction mixture was stirred at rt fori h and LCMS showed no starting material. Then, 3N HC1 (0.4 mL) was added at 0 °C and warmed up to rt. The solvent was evaporated and the residue was dissolved in CH 3 CN, filtered. Purification by RP-Prep HPLC afforded a diastereomeric mixture. The diastereomers were separated by chiral Prep. SFC to provide Example 86, Isomers 1, 2 and 3 as single stereoisomers, prepared following the procedure for Example 81. Example 86, Isomer 1 (off-white solid, 5 mg): LC-MS Anal. Calc'd for C 26 H3 0 ClF 3 4O4 554.19, found [M+H] 555.1. !H MR (400MHZ, CD 3 CN) δ ppm 8.00 (s, 1H), 7.13 - 7.06 (m, 2H), 7.02 - 6.94 (m, 2H), 6.38 (s, 1H), 4.60 (d, J=9.3 Hz, 1H), 4.01 (td, J=8.9, 4.3 Hz, 1H), 3.87 (s, 3H), 3.59 - 3.50 (m, 2H), 3.31 (dd, J=3.5, 2.3 Hz, 1H), 2.97 - 2.87 (m, 1H), 2.76 - 2.66 (m, 2H), 2.52 (dd, J=16.2, 9.4 Hz, 1H), 2.25 - 2.16 (m, 1H), 2.10 - 2.02 (m, 1H), 1.79 - 1.58 (m, 4H), 1.12 (d, J=6.8 Hz, 3H), 0.95 (t, J=7.5 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 12.5 min, HI: 98%. hGPR40 EC 50 = 73 nM. Example 86, Isomer 2 (off-white solid, 5 mg): LC-MS Anal.

Calc'd for C 26 H3 0 ClF 3 4 O4 554.19, found [M+H] 555.1. l H NMR (400MHz, CD 3 CN) δ ppm 7.89 (s, 1H), 7.01 - 6.94 (m, 2H), 6.91 - 6.83 (m, 2H), 6.27 (s, 1H), 4.49 (d, J=9.3 Hz, 1H), 3.89 (td, J=8.9, 4.2 Hz, 1H), 3.76 (s, 3H), 3.51 - 3.39 (m, 2H), 3.20 (dd, J=3.7, 2.1 Hz, 1H), 2.86 - 2.78 (m, 1H), 2.64 - 2.54 (m, 2H), 2.41 (dd, J=16.3, 9.5 Hz, 1H), 2.14 - 2.06 (m, 1H), 1.96 (dd, J=12.4, 5.8 Hz, 1H), 1.71 - 1.48 (m, 3H), 1.01 (d, J=6.6 Hz, 3H), 0.87 - 0.80 (m, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 12.5 min, HI: 96%. hGPR40 EC 50 = 599 nM. Example 86, Isomer 3 (off-white solid, 5 mg):. LC- MS Anal. Calc'd for C26H30CIF3 4O4 554.19, found [M+H] 555.1. !H MR (400MHZ, CD 3 CN) δ ppm 8.00 (s, IH), 7.13 - 7.06 (m, 2H), 7.01 - 6.92 (m, 2H), 6.38 (s, IH), 4.60 (d, J=9.8 Hz, IH), 4.01 (td, J=8.8, 4.1 Hz, IH), 3.87 (s, 3H), 3.60 - 3.50 (m, 2H), 3.31 (dd, J=3.5, 2.0 Hz, IH), 2.97 - 2.86 (m, IH), 2.75 - 2.65 (m, 2H), 2.52 (dd, J=16.3, 9.3 Hz, IH), 2.25 - 2.17 (m, IH), 2.07 (d, J=6.5 Hz, IH), 1.81 - 1.59 (m, 3H), 1.12 (d, J=6.5 Hz, 3H), 0.95 (t, J=7.5 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 12.5 min, HI: 95%. hGPR40 EC 50 = 1932 nM. Example 87

2-((4S,5 S)- 1 -(4-(((3R,4R)- 1 -(5 -Chloro-2-i/3 -methoxypyridin-4-yl)-3 -methylpiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid,

[00369] Example 87 (white foam, 60 mg) was prepared following the procedure for Example 81. LC-MS Anal. Calc'd for C 2 5H 2 5D3C1F3 40 4 543.19, found [M+H] 544.2.

!H NMR (400MHz, CD 3 CN) δ ppm 7.96 (s, IH), 7.1 1 (d, J=9.0 Hz, 2H), 6.99 (d, J=8.8 Hz, 2H), 6.48 (s, IH), 4.54 - 4.41 (m, IH), 4.16 (td, J=8.4, 4.0 Hz, IH), 3.98 (dd, J=12.7, 3.6 Hz, 2H), 3.43 - 3.34 (m, 2H), 3.16 (dd, J=13.2, 9.7 Hz, 2H), 2.76 (dd, J=16.3, 3.0 Hz, IH), 2.53 (dd, J=16.3, 9.5 Hz, IH), 2.33 - 2.20 (m, IH), 2.12 (br. s., IH), 1.82 - 1.71 (m, IH), 1.33 (d, J=7.0 Hz, 3H), 1.13 (d, J=6.8 Hz, 3H). HPLC (Orthogonal method, 30% Solvent B start): RT = 1 1.6 min, HI: 100%. hGPR40 EC 50 = 74 nM. hGPR40 IP1 EC 50 = 6 nM. Example 88 2-((4S,5S)- and (4R,5R)-4-Ethyl-l-(4-((l-(2-fluoro-5-methoxyphenyl)piperidin -4- yl)oxy)phenyl)-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5- yl)acetic acid (Isomers 1 and 2)

[00370] 88A. 2,2,2-Trifluoro-N'-(4-methoxyphenyl)acetohydrazide: To a stirred suspension of (4-methoxyphenyl)hydrazine (6.0 g, 42.1 mmol) in THF (150 mL) at 0 °C was added 2,2,2-trifluoroacetic anhydride (6.44 mL, 46.3 mmol) dropwise. The resulting mixture was stirred at 0 °C for 2.5 h and then an additional amount of trifluoroacetic anhydride (2.38 mL, 16.9 mmol) was added dropwise. After stirred for an additional 5 min at 0 °C, the mixture was concentrated and purified via silica gel chromatography to give the desired product (6.48 g, 65.7% yield) as an off-white solid: LC-MS [M+Na] 257.

[00371] 88B. 2,2,2-Trifluoro-N'-(4-methoxyphenyl)acetohydrazonoyl chloride: To a stirred solution of 2,2,2-trifluoro-N'-(4-methoxyphenyl)acetohydrazide (6.48 g, 22.14 mmol) in EtOAc (120 mL) was added benzenesulfonyl chloride (3.44 mL, 26.6 mmol) at rt. The solution was cooled to 0 °C, and to the solution was added N,N- diisopropylethylamine (5.80 mL, 33.2 mmol) dropwise. The resulting mixture was stirred at 0 °C for 20 min, and then warmed to rt. After stirring at rt overnight, the mixture was diluted with EtOAc and, washed with sat'd NaHCC^ and water. The organic layer was dried ( a 2 S04) and concentrated and the crude was purified via silica gel

chromatography (0 to 10% EtOAc) to provide the desired product (4.22 g, 75% yield) as a reddish liquid: LC-MS [M+H] 253, 255.

[00372] 88C. (4S,5R)- and (4R,55)-Methyl 4-ethyl-l-(4-methoxyphenyl)-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazole-5-carboxylate: A stirred mixture of 2,2,2- trifluoro-N'-(4-methoxyphenyl)acetohydrazonoyl chloride (1.20 g, 4.51 mmol), (E)- methyl pent-2-enoate (1.593 g, 13.54 mmol) and silver carbonate (2.489 g, 9.03 mmol) in dioxane (20 mL) was sonicated under vacuum and refilled with argon three times. The mixture was heated to 65 °C and stirred at this temperature overnight. Then, the mixture was filtered and the filter cake was rinsed with CH2CI2. The combined filtrates were concentrated and purified via silica gel chromatography to afford the desired product (910 mg, 61.1% yield) as a reddish oil: LC-MS [M+H] 331.

[00373] 88D. ((4S.5R) and (4R,55)-4-Ethyl- 1 -(4-methoxyphenyl)-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)methanol: To a stirred solution of (AS,5R) and (4R,5S - methyl 4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazole-5- carboxylate (910 mg, 2.76 mmol) in THF (5.0 mL) and EtOH (10 mL) was added sodium borohydride (208 mg, 5.51 mmol). The reaction mixture was stirred at rt overnight and then quenched with 1M HC1. The aqueous mixture was stirred at rt for 2.5 days. The mixture was concentrated and purified via silica gel chromatography to provide the desired product (705 mg, 85% yield) as a reddish oil: LC-MS [M+H] 303.

[00374] 88E. ((45.5R)- and (4R,55)-4-Ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)methyl methanesulfonate: To a stirred solution of ((45 * ,5R)- and (4R,55)-4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5- dihydro-lH-pyrazol-5- yl)methanol (705 mg, 2.146 mmol) in CH 2 C1 2 (10.0 mL) at 0 °C was added

methanesulfonyl chloride (0.251 mL, 3.22 mmol) and then triethylamine (0.748 mL, 5.36 mmol). The resulting mixture was stirred overnight while allowing the temperature to reach rt. The reaction mixture was diluted with CH2CI2 and washed with sat'd NaHCC^ and water. The organic layer was dried ( a2S0 4 ) and concentrated and the crude was purified via silica gel chromatography to give the desired product (678 mg, 83% yield) as a yellowish oil: LC-MS [M+H] 381.

[00375] 88F. 2-((45,55) and (4R,5R)-4-Ethyl- 1 -(4-methoxyphenyl)-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetonitrile: To a stirred solution of ((4S,5R)- and (4R,55)-4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5- dihydro-lH- pyrazol-5-yl)methyl methanesulfonate (678 mg, 1.782 mmol) was added potassium cyanide (239 mg, 3.56 mmol). The resulting mixture was heated to 50 °C and stirred at this temperature for 19 h. After this time, the reaction mixture was cooled to rt, diluted with sat. aHC03 and extracted with EtOAc. The organic extract was washed with sat'd

NaCl, dried (Na 2 S0 4 ) and concentrated. Purification via silica gel chromatography afforded the desired product (512 mg, 91% yield) as an orangish oil: LC-MS [M+H] 312.

[00376] 88G. 2-((45,55)- and (4R,45)-4-Ethyl-l-(4-hydroxyphenyl)-3-

(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetonitril e:To a stirred solution of 2- ((45,55) and (4R,5R)-4-ethyl-l-(4-methoxyphenyl)-3-(trifluoromethyl)-4,5- dihydro-lH- pyrazol-5-yl)acetonitrile (512 mg, 1.628 mmol) in CH 2 C1 2 (4.0 mL) at -78 °C under argon was added boron tribromide (0.308 mL, 3.26 mmol) dropwise. The resulting mixture was stirred at -78 °C for 10 min, then allowed to warm up to 0 °C and stirred for 1 h. At this time, the reaction mixture was quenched with MeOH (20 mL), allowed to warm up to rt and stirred for 1 h. The mixture was concentrated and the crude was purified via silica gel chromatography to provide the desired product (427 mg, 82% yield) as a yellowish solid: LC-MS [M+H] 298.

[00377] 88H. l-(2-Fluoro-5-methoxyphenyl)piperidin-4-yl 4-methylbenzenesulfonate: To a stirred solution of l-(2-fluoro-5-methoxyphenyl)piperidin-4-ol (1.25 g, 5.55 mmol) in CH2CI2 (25 mL) at rt was added 4-methylbenzene- 1 -sulfonyl chloride (1.270 g, 6.66 mmol) followed by pyridine (2.244 mL, 27.7 mmol). The resulting mixture was stirred at rt overnight and then washed with water. The organic layer was dried (T^SC^) and concentrated, and the crude was purified via silica gel chromatography to give the desired product (1.38 g, 65.5% yield) as a white solid: LC-MS [M+H] 380.

[00378] 881. 2-((45,55)- and (4R,5R)-4-Ethyl-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-3-(trifluoromethyl) -4,5-dihydro-lH-pyrazol- 5-yl)acetonitrile: To a stirred mixture of l-(2-fluoro-5-methoxyphenyl)piperidin-4-yl 4- methylbenzenesulfonate (390 mg, 1.0 mmol) and 2-((4S,5S)- and (4R,5R)-4-ethyl-l-(4- hydroxyphenyl)-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5- yl)acetonitrile (430 mg, 1.4 mmol) in DMF (10 mL) was added cesium carbonate (1.0 g, 3.1 mmol). The resulting mixture was heated to 50 °C and stirred at this temperature overnight. Then, the mixture was diluted with water and extracted with EtOAc. The organic extract was dried

( a 2 S04) and concentrated, and the crude was purified via silica gel chromatography to afford the desired product (206 mg, 36.5% yield) as a yellowish oil: LC-MS [M+H] 505.

[00379] 88J. Methyl 2-((45,55)- and (4R,5R)-4-ethyl-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-3-(trifluoromethyl) -4,5-dihydro-lH-pyrazol- 5-yl)acetate: 2-((45,55)- and (4R,5R)-4-ethyl-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-3-(trifluoromethyl) -4,5-dihydro-lH-pyrazol- 5-yl)acetonitrile was dissolved in ~3M HCl/MeOH, MeOAc solution [6.85 mL, prepared by addition of AcCl (1.45 mL) to MeOH (5.4 mL) at 0 °C and then stirring at rt for 30 min]. The resulting solution was allowed to stand at rt for 3.5 days and then evaporated. Purification via silica gel chromatography provided the desired product (102 mg, 46.5% yield) as a colorless oil: LC-MS [M+H] 538.

Example 88 (Isomers 1 2): To a solution of methyl 2-((4S,5S)- and (4R,5R)-4-ethyl-l-(4- ((l-(2-fluoro-5-methoxyphenyl)piperidin-4-yl)oxy)phenyl)-3-( trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetate (102 mg, 0.190 mmol) in THF (3.0 mL) was added a solution of lithium hydroxide (9.1 mg, 0.38 mmol) in water (0.5 mL). The resulting mixture was stirred at rt overnight. The mixture was diluted with water and the pH of the aqueous mixture was adjusted to 1 by dropwise addition of 1M HCl diluted. The aqueous mixture was extracted with CH2CI2 and, the organic extract was dried over ( a2S0 4 ) and concentrated to give the racemic product as a thick oil. The enantiomers were separated by chiral preparative SFC to afford: Example 88, Isomer 1 (white solid, 42 mg, 41% yield): LC-MS [M+H] 524. Ή NMR (500MHz, CDC1 3 ) δ 7.05 (d, J= 8.8 Hz, 2H), 6.96 (m, 1H, overlapped), 6.92 (d, J= 8.8 Hz, 2H), 6.70 - 6.35 (m, 2H), 4.53 (d, J= 10.0 Hz, 1H), 4.40 (m, 1H), 3.78 (s, 3H), 3.36 (m, 2H), 3.16 (m, 1H), 3.00 (m, 2H) 2.83 (dd, J = 16.0, 3.0 Hz, 1H), 2.46 (dd, J=16.1, 10.3 Hz, 1H), 2.24 - 1.88 (m, 4H), 1.75 (m, 1H), 1.65 (m, 1H) 1.37 (t, J= 7.0 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 7.45 min, HI: 96%. hGPR40 EC 50 = 78 nM. hGPR40 IP1 EC 50 = 18 nM. Example

88, Isomer 2 (white solid, 44 mg, 44% yield): LC-MS [M+H] 524. Ή NMR (500MHz, CDCI3) δ 7.05 (d, J= 8.8 Hz, 2H), 6.96 (m, 1H, overlapped), 6.92 (d, J= 8.8 Hz, 2H), 6.70 - 6.35 (m, 2H), 4.53 (d, J= 10.0 Hz, 1H), 4.40 (m, 1H), 3.78 (s, 3H), 3.36 (m, 2H), 3.16 (m, 1H), 3.00 (m, 2H) 2.83 (dd, J= 16.0, 3.0 Hz, 1H), 2.46 (dd, J=16.1, 10.3 Hz, 1H), 2.24 - 1.88 (m, 4H), 1.75 (m, 1H), 1.65 (m, 1H) 1.37 (t, J= 7.0 Hz, 3H). Analytical HPLC (Orthogonal method, 0% Solvent B start): RT = 7.45 min, HI: 98%. hGPR40 EC 50 = 7454 nM.

Example 89

2-((45",55)- and(4R,5R)-l-(4-((l-(2-Fluoro-5-methoxyphenyl)piperidin-4-yl )oxy)phenyl)- 4-isobutyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)a cetic acid (Isomers 1 and 2)

[00380] Example 89, Isomer 1 (white solid, 15 mg) and Isomer 2 (white solid, 16 mg) were prepared from (is)-methyl 5-methylhex-2-enoate following the procedure for Example 88. Example 89, Isomer 1 : LC-MS [M+H] 552. !H MR (500MHZ, CDCI3) δ 7.06 (d, J= 8.9 Hz, 2H), 6.93 (m, 3H), 6.53 (dd, J= 7.2, 3.1 Hz, 1H) 6.41 (dt, J= 8.8, 3.1 Hz, 1H), 4.52 (d, J= 8.7 Hz, 1H), 4.36 (m, 1H), 3.77 (s, 3H), 3.32 (m, 2H), 3.20 (m, 1H), 2.95 (m, 3H) 2.70 (m, 1H), 2.38 (m, 1H), 2.09 (m, 2H), 1.96 (m, 2H), 1.51 (m, 1H), 1.40 (m, 1H) 0.94 (t, J= 6.4 Hz, 3H), 0.92 (t, J= 6.2 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 8.18 min, HI: 96%. hGPR40 EC 50 = 7962 nM. Example 89, Isomer 2: LC-MS [M+H] 552. Ή NMR (500MHz, CDC1 3 ) δ 7.06 (d, J= 8.9 Hz, 2H), 6.93 (m, 3H), 6.53 (dd, J= 7.2, 3.1 Hz, 1H) 6.41 (dt, J= 8.8, 3.1 Hz, 1H), 4.52 (d, J= 8.7 Hz, 1H), 4.36 (m, 1H), 3.77 (s, 3H), 3.32 (m, 2H), 3.20 (m, 1H), 2.95 (m, 3H) 2.70 (m, 1H), 2.38 (m, 1H), 2.09 (m, 2H), 1.96 (m, 2H), 1.51 (m, 1H), 1.40 (m, 1H) 0.94 (t, J= 6.4 Hz, 3H), 0.92 (t, J= 6.2 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 8.17 min, HI: 98%. hGPR40 EC 50 = 390 nM.

Example 90

2-((45,55)- and (4R,5R)-4-(Cyclopropylmethyl)- 1 -(4-(( 1 -(2-fluoro-5 - methoxyphenyl)piperidin-4-yl)oxy)phenyl)-3-(trifluoromethyl) -4,5-dihydro-lH-pyrazol- -yl)acetic acid (Isomers 1 and 2)

[00381] 90A. (£)-Benzyl 4-cyclopropylbut-2-enoate: To a stirred solution of sodium hydride (0.7 g, 18 mmol) in THF (20 mL) at 0 °C under argon was added a solution of benzyl 2-(dimethoxyphosphoryl)acetate (3.8 g, 14 mmol) in THF (5.0 mL), slowly. The mixture was stirred at 0 °C for 15min until it turned into a clear solution and then, a solution of 2-cyclopropylacetaldehyde (1.0 g, 12 mmol) in THF (5.0 mL) was added dropwise. The resulting mixture was allowed to warm up to rt and was stirred at rt for 6 h. The reaction mixture was quenched with water and extracted with EtOAc. The organic extract was dried ( a 2 S04) and concentrated and the crude was purified via silica gel chromatography to provide the desired product (1.3 g, 44% yield) as a colorless oil: LC- MS [M+Na] 239.

[00382] Example 90, Isomer 1 (white solid, 39 mg) and Isomer 2 (white solid, 45 mg) were prepared from (is)-benzyl 4-cyclopropylbut-2-enoate following the procedure for Example 88. Example 90, Isomer 1 : LC-MS [M+H] 550. l R NMR (500MHz, CDC1 3 ) δ 7.06 (d, J= 9.1 Hz, 2H), 6.92 (m, 3H), 6.54 (dd, J=7.2, 3.0 Hz, 1H), 6.41 (dt, J=8.7, 3.0 Hz, 1H), 4.77 (d, J= 9.9 Hz, 1H), 4.37 (m, 1H), 3.77 (s, 3H), 3.34 (m, 2H), 3.28 (m, 1H), 2.98 (m, 2H) 2.82 (dd, J= 16.0, 2.5 Hz, 1H), 2.46 (dd, J=16.0, 10.3 Hz, 1H), 2.10 (m, 2H), 1.98 (m, 2H), 1.66 (m, 1H), 1.48 (m, 1H) 0.76 (m, 1H), 052 (m, 1H), 0.44 (m, 1H), 0.19 - 0.08 (m, 2H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 7.89 min, HI: 98%. hGPR40 EC 50 = 1626 nM. Example 90, Isomer 2: LC-MS [M+H] 550. l R NMR (500MHz, CDC1 3 ) δ 7.06 (d, J= 9.1 Hz, 2H), 6.92 (m, 3H), 6.54 (dd, J=7.2, 3.0 Hz, 1H), 6.41 (dt, J=8.7, 3.0 Hz, 1H), 4.77 (d, J= 9.9 Hz, 1H), 4.37 (m, 1H), 3.77 (s, 3H), 3.34 (m, 2H), 3.28 (m, 1H), 2.98 (m, 2H) 2.82 (dd, J= 16.0, 2.5 Hz, 1H), 2.46 (dd, J=16.0, 10.3 Hz, 1H), 2.10 (m, 2H), 1.98 (m, 2H), 1.66 (m, 1H), 1.48 (m, 1H) 0.76 (m, 1H), 052 (m, 1H), 0.44 (m, 1H), 0.19 - 0.08 (m, 2H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 7.87 min, HI: 97%. hGPR40 EC 50 = 156 nM.

Example 91

2-((45 , ,55)-3-(4-Cyanophenyl)-l-(4-((l-(2-fluoro-5-methoxyphe nyl)piperidin-4- yl)oxy)phenyl)-4-methyl-4,5-dihydro- lH-pyrazol-5-yl)acetic acid, TFA

[00383] 91 A. 4-((2-(4-Methoxyphenyl)hydrazono)methyl)benzonitrile: To a solution of 4-methoxyphenylhydrazine hydrochloride (626 mg, 3.51 mmol) and 4- cyanobenzaldehyde (421 mg, 3.21 mmol) in DMF (3.5 mL) was added triethylamine (1.0 mL, 7.14 mmol). The mixture was stirred at rt under argon for 47 h, water (6.8 mL) was added and stirring was continued for 1 h. The solid that resulted was filtered, rinsed with water (6 mL) and dissolved in CH 2 Ci2 (100 mL). The CH 2 Ci2 solution was dried ( a 2 S04, overnight) and concentrated to provide the desired product (857 mg, 97% yield) as a brownish solid: LC-MS [M+H] 252.

[00384] 9 IB. 4-Cyano-N'-(4-methoxyphenyl)benzohydrazonoyl bromide: N- Bromosuccinimide (612 mg, 3.41 mmol) was added to a stirred solution of 4-((2-(4- methoxyphenyl)hydrazono)methyl)benzonitrile (855 mg, 3.10 mmol) in THF (5.1 mL) at 0 °C under argon. The mixture was stirred at 0 °C for 2.5 h and then evaporated under reduced pressure. Purification via silica gel chromatography afforded the desired product (650 mg, 56.6% yield) as a brownish solid: LC-MS [M+H] 330, 332.

[00385] 91C. 4-((45,5R)-l-(4-Methoxyphenyl)-4-methyl-5-((5)-2-oxo-4- phenyloxazolidine-3-carbonyl)-4,5-dihydro-lH-pyrazol-3-yl)be nzonitrile and 4-((4R,55)- l-(4-methoxyphenyl)-4-methyl-5-((5)-2-oxo-4-phenyloxazolidin e-3-carbonyl)-4,5- dihydro-lH-pyrazol-3-yl)benzonitrile: A flask containing a solution of 4-cyano-N'-(4- methoxyphenyl)benzohydrazonoyl bromide (416 mg, 1.121 mmol) and (S,E)-3-(but-2- enoyl)-4-phenyloxazolidin-2-one (300 mg, 1.271 mmol) in dioxane (16 mL) was evacuated and backfilled with argon. Silver carbonate (877 mg, 3.15 mmol) was added to the solution and the resulting suspension was stirred and heated to 50 °C. After stirring at this temperature for 14.5 h, the reaction mixture was cooled to rt and filtered through CELITE®. The filter cake was rinsed with EtOAc (120 mL) and, the combined filtrate and rinse were concentrated. The crude was chromatographed to afford 4-((45',5R)-l-(4- methoxyphenyl)-4-methyl-5-((5)-2-oxo-4-phenyloxazolidine-3-c arbonyl)-4,5-dihydro- lH-pyrazol-3-yl)benzonitrile (152 mg, 28% yield) as a yellow solid: LC-MS [Μ+Η] 481. Further elution of the silica column gave a mixture of the diastereomers (270 mg, 50% yield) followed by 4-((4R,55)-l-(4-methoxyphenyl)-4-methyl-5-((5)-2-oxo-4- phenyloxazolidine-3-carbonyl)-4,5-dihydro-lH-pyrazol-3-yl)be nzonitrile (18 mg, 3% yield) as a yellow solid: LC-MS [Μ+Η] 481. [00386] 91D. 4-((45,5R)-5-(Hydroxymethyl)-l-(4-methoxyphenyl)-4-methyl-4, 5- dihydro-lH-pyrazol-3-yl)benzonitrile: To a solution of 4-((45',5R)-l-(4-methoxyphenyl)- 4-methyl-5-((5')-2-oxo-4-phenyloxazolidine-3-carbonyl)-4,5-d ihydro-lH-pyrazol-3- yl)benzonitrile (171 mg, 0.356 mmol) in THF (6.0 mL) at rt was added a solution of sodium borohydride (83 mg, 2.172 mmol) in water (1.2 mL). After stirring at rt for 4.4 h, the reaction mixture was cooled to 0 °C and quenched with 10% KHSO4 (10 mL). The resulting aqueous mixture was allowed to warm up to rt and stirred for 2 h. The organic solvent was mostly evaporated under reduced pressure and the remaining aqueous mixture was extracted with CH2CI2 (3x30 mL). The combined organic extracts were dried ( a 2 S04) and concentrated. Chromatography of the crude afforded the desired product (138 mg, 99% yield) as a yellow oil: LC-MS [M+H] 322.

[00387] 91E. ((45,5R)-3-(4-Cyanophenyl)-l-(4-methoxyphenyl)-4-methyl-4,5- dihydro-lH-pyrazol-5-yl)methyl methanesulfonate: To a solution of 4-((45',5R)-5- (hydroxymethyl)- 1 -(4-methoxyphenyl)-4-methyl-4,5 -dihydro- lH-pyrazol-3 - yl)benzonitrile (138 mg, 0.35 mmol) and methanesulfonyl chloride (0.030 mL, 0.386 mmol) in CH 2 C1 2 (3.1 mL) at 0 °C was added triethylamine (0.078 mL, 0.557 mmol). The mixture was stirred for 30 min at 0 °C and for 3.2 h while warming to rt. Then, the mixture was diluted with EtOAc (50 mL) and washed with sat'd NaHCC^ (2x30 mL) and sat'd NaCl (20 mL). The EtOAc solution was dried ( a 2 S04) and evaporated. Drying under vacuum gave the desired product (154 mg, 91% yield) as a yellow oil: LC-MS [M+H] 400.

[00388] 9 IF. 4-((45,55)-5-(Cyanomethyl)- 1 -(4-methoxyphenyl)-4-methyl-4,5-dihydro- lH-pyrazol-3-yl)benzonitrile: To a solution of ((45',5R)-3-(4-cyanophenyl)-l-(4- methoxyphenyl)-4-methyl-4,5-dihydro- lH-pyrazol-5-yl)methyl methanesulfonate (154 mg, 0.320 mmol) in DMSO (1.4 mL) was added potassium cyanide (25 mg, 0.372 mmol). The mixture was heated to 40 °C and stirred at this temperature for 12 h, the mixture was cooled to rt, diluted with 4/1 EtOAc/Hex (60 mL) and, washed with sat'd NaHC0 3 (2x30 mL), water (2 x 30 mL) and sat'd NaCl (30 mL). The organic layer was dried ( a 2 S0 4 ) and concentrated. Purification via silica gel chromatography gave the desired product (94 mg, 87% yield) as a yellow oil: LC-MS [M+H] 331. [00389] 91G. 4-((45,55)-5-(Cyanomethyl)-l-(4-hydroxyphenyl)-4-methyl-4,5- dihydro- lH-pyrazol-3-yl)benzonitrile: To a solution of 4-((4S,55)-5-(cyanomethyl)-l-(4- methoxyphenyl)-4-methyl-4,5-dihydro-lH-pyrazol-3-yl)benzonit rile (93 mg, 0.28 mmol) in CH 2 Cl2(0.8 mL) at 0 °C was added boron trifluoride-methyl sulfide complex (0.18 mL, 1.7 mmol). The reaction mixture was stirred at 0 °C for 20 min and then at rt for 2.7 h. The reaction mixture was cooled to 0 °C and the reaction was quenched with MeOH (6.0 mL) followed by AcCl (0.2 mL). The mixture was allowed to warm up to rt, stirred for 1.5 h and evaporated. The residue was taken up in EtOAc (50 mL) and, washed with 5% aHC03 (2x25 mL) and sat'd NaCl (20 mL). The organic layer was dried ( a2S0 4 ) and concentrated. Purification via silica gel chromatography provided the desired product (87 mg, 97% yield) as a yellow oil: LCMS [M+H] 317.

[00390] 91H. l-(2-Fluoro-5-methoxyphenyl)piperidin-4-yl methanesulfonate: To a solution of l-(2-fluoro-5-methoxyphenyl)piperidin-4-ol (146 mg, 0.62 mmol) and methanesulfonyl chloride (0.067 mL, 0.86 mmol) in CH2CI2 (6.2 mL) at 0 °C was added triethylamine (0.18 mL, 1.285 mmol). The mixture was stirred for 30 min at 0 °C and for 5.0 h while warming to rt. Then, the mixture was diluted with EtOAc (60 mL) and washed with sat'd NaHC0 3 (2x30 mL) and sat'd NaCl (20 mL). The EtOAc solution was dried (Na 2 S0 4 ) and evaporated. Drying under vacuum gave the desired product (180 mg, 93% yield) as a yellow solid: LC MS [M+H] 304.

[00391] 911. 4-((45,55)-5-(Cyanomethyl)-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-4,5-dihydr o-lH-pyrazol-3- yl)benzonitrile: To a solution of 4-((4S,55)-5-(cyanomethyl)-l-(4-hydroxyphenyl)-4- methyl-4,5-dihydro-lH-pyrazol-3-yl)benzonitrile (41 mg, 0.128 mmol) in DMF (0.25 mL) were added potassium carbonate (powder, 30 mg, 0.215 mmol) and l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl methanesulfonate (61 mg, 0.195 mmol). The mixture was heated to 85 °C and stirred at this temperature for 24 h. Then, the mixture was cooled to rt, diluted with 1/1 EtO Ac/Hex (60 mL) and, washed with 5% NaHC0 3 (2x30 mL), water (30 mL) and sat'd NaCl (20 mL). The organic layer was dried (Na 2 S0 4 ) and evaporated. Purification by two consecutive chromatographies (S1O2, first 95/5 CH^C^/Ether and then 7/3 Hex/EtOAc) afforded the desired product (23 mg, 32.5% yield) as a yellow oil: LCMS [M+H] 524. [00392] 91J. Methyl 2-((45,55)-3-(4-cyanophenyl)-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-4,5-dihydr o-lH-pyrazol-5- yl)acetate and Methyl 4-((45',55 , )-l-(4-((l-(2-fluoro-5-methoxyphenyl)piperidin-4- yl)oxy)phenyl)-5-(2-methoxy-2-oxoethyl)-4-methyl-4,5-dihydro -lH-pyrazol-3- yl)benzoate: 4-((45',55 , )-5-(Cyanomethyl)-l-(4-((l-(2-fluoro-5-methoxyphenyl)p iperidin- 4-yl)oxy)phenyl)-4-methyl-4,5-dihydro-lH-pyrazol-3-yl)benzon itrile: (22 mg, 0.040 mmol) was dissolved in ~3M HCl/MeOH, MeOAc, CH 2 C1 2 solution [6.3 mL, prepared by addition of AcCl (1.3 mL) to 3/2 CH 2 Cl 2 /MeOH (5.0 mL) at 0 °C and then stirring at rt for 30 min]. The resulting solution was stirred at rt for 12.0 h, then diluted with MeCN (6 mL) and evaporated. The residue was taken up in EtOAc (50 mL) and, washed with 5% aHC03 (2x30 mL) and sat'd NaCl (20 mL). The crude was purified by preparative RP-HPLC. Fractions containing the slower moving component were combined, basified with solid NaHCC^ and partially evaporated at rt to remove most of the MeCN. The remaining aqueous mixture was extracted with CH 2 C1 2 (3x30 mL) and, the combined extracts were dried (Na 2 S0 4 ) and concentrated. Drying under vacuum provided methyl 2- ((45 , ,55)-3-(4-cyanophenyl)-l-(4-((l-(2-fluoro-5-methoxyphe nyl)piperidin-4- yl)oxy)phenyl)-4-methyl-4,5-dihydro-lH-pyrazol-5-yl)acetate (6 mg, 27% yield) as a yellow solid: LC-MS [Μ+Η] 557. Fractions containing the faster moving component were combined, basified with solid NaHCC^ and partially evaporated at rt to remove most of the MeCN. The remaining aqueous mixture was extracted with CH 2 C1 2 (3x30 mL) and, the combined extracts were dried (Na 2 S0 4 ) and evaporated. The concentrate was dissolved in 1/1 CH 2 Cl 2 /MeOH (4.0 mL) and allowed to stand at rt for 7 days to enable conversion to a slower moving derivative. The solution was concentrated to give methyl 4-((45',55)-l-(4-((l-(2-fluoro-5-methoxyphenyl)piperidin-4-y l)oxy)phenyl)-5-(2- methoxy-2-oxoethyl)-4-methyl-4,5-dihydro-lH-pyrazol-3-yl)ben zoate (10 mg, 0.015 mmol, 36.3% yield) as a yellow solid: LC-MS [Μ+Η] 590.

[00393] Example 91 : To a stirred solution of methyl 2-((45',55)-3-(4-cyanophenyl)-l- (4-((l-(2-fluoro-5-methoxyphenyl)piperidin-4-yl)oxy)phenyl)- 4-methyl-4,5-dihydro-lH- pyrazol-5-yl)acetate (6.0 mg, 10 μιηοΐ) in THF (0.4 mL) and water (0.04 mL) at rt was added l.OM aqueous lithium hydroxide (0.03 mL, 0.030 mmol). After stirring at rt for 5.0 h, the reaction mixture was partially evaporated to remove most of the THF. The remaining solution was partitioned between water (30 mL) and Hex (10 mL). The aqueous layer was acidified to pH 2 by dropwise addition of IM HCl, and extracted with CH 2 Cl2 (3x30 mL). The combined organic extracts were dried ( a 2 S04) and

concentrated. The crude (5.5 mg) was purified by preparative RP-HPLC. Fractions containing the desired acid were combined and partially evaporated at rt to remove most of the MeCN. The remaining aqueous mixture was extracted with CH2CI2 (3x30 mL) and, the combined extracts were dried ( a 2 S04) and evaporated. Drying under vacuum afforded the desired product (3.5 mg, 51.5% yield) as a yellow solid: HPLC-50% start B (RT 6.75, Area% 100); LC-MS [M+H] 543. ¾ NMR (400MHz, CD 2 C1 2 ) δ 7.82 (d, J= 8.0 Hz, 2H),7.66 (d, J= 8.0 Hz, 2H), 7.14 (m, 2H), 6,96 (m, 3H), 6.69 (m, 1H), 6.50 (dt, J = 8.9, 3.0 Hz, 1H), 4.44 (m, 2H), 3.77 (s, 3H), 3.43 (m, 3H), 3.05 (m, 2H), 2.83 (d, J = 16.2 Hz, 1H), 2.41 (dd, J=16.2, 10.6 Hz, 1H), 2.17 (m, 2H), 1.99 (m, 2H), 1.32 (d, J= 7.2 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 6.75 min, HI: 100%. hGPR40 EC50 = 62 nM.

Example 92

2-((45 , ,55)-l-(4-((l-(2-Fluoro-5-methoxyphenyl)piperidin-4-yl )oxy)phenyl)-3-(4- (methoxycarbonyl)phenyl)-4-methyl-4,5-dihydro- lH-pyrazol-5-yl)acetic acid, TFA

[00394] To a stirred solution of methyl 4-((45,55)-l-(4-((l-(2-fluoro-5- methoxyphenyl)piperidin-4-yl)oxy)phenyl)-5-(2-methoxy-2-oxoe thyl)-4-methyl-4,5- dihydro-lH-pyrazol-3-yl)benzoate (10 mg, 0.015 mmol) in THF (0.5 mL) and water (0.04 mL) at rt was added 1.0M aqueous lithium hydroxide (0.03 mL, 0.03 mmol). After stirring at rt for 2.4 h, the mixture was cooled to 0 °C and acidified with IM HCl (0.04 mL). The aqueous mixture was partitioned between CH2Q2 (40 mL) and water (15 mL).

The aqueous layer pH was adjusted to 2 by dropwise addition of IM HCl and the biphasic mixture was shaken. The organic layer was collected and the aqueous layer was extracted with CH 2 Cl2 (2x20 mL). The combined organic layers were dried ( a 2 S04) and evaporated. The crude (8 mg) was purified by preparative RP-HPLC. Fractions containing the desired acid were combined and partially evaporated at rt to remove most of the MeCN. The remaining aqueous mixture was extracted with CH2CI2 (3x30 mL) and, the combined extracts were dried ( a2S0 4 ) and evaporated. Drying under vacuum afforded 2-((45 , ,55)-l-(4-((l-(2-fluoro-5-methoxyphenyl)piperidin-4-yl )oxy)phenyl)-3-(4- (methoxycarbonyl)phenyl)-4-methyl-4,5-dihydro-lH-pyrazol-5-y l)acetic acid, TFA (5.0 mg, 49.0% yield) as a yellow solid: LC-MS [Μ+Η] 576. !pi MR (500MHz, CD 2 C1 2 ) δ

8.03 (d, J= 8.6 Hz, 2H),7.81 (d, J= 8.6 Hz, 2H), 7.15 (d, J= 8.9 Hz, 2H), 7,01 (dd, J=12.1, 9.0 Hz, 1H), 6.96 (d, J= 8.9 Hz, 2H), 6.81 (m, 1H), 6.58 (dt, J= 9.0, 3.0 Hz, 1H), 4.45 (m, 2H), 3.90 (s, 3H), 3.77 (s, 3H), 3.55 (m, 2H), 3.47 (m, 1H), 3.14 (m, 2H), 2.83 (dd, J= 16.3, 2.8 Hz, 1H), 2.40 (dd, J=16.3, 10.4 Hz, 1H), 2.23 (m, 2H), 2.03 (m, 2H), 1.33 (d, J= 7.1 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 7.28 min, HI: 99%. hGPR40 EC 50 = 139 nM. Example 93

2-((45 , ,55)-3-(4-Cyanophenyl)-l-(4-((l-(5-ethoxy-2-fluorophen yl)piperidin-4- yl)oxy)phenyl)-4-methyl-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00395] 93 A. l-(5-Ethoxy-2-fluorophenyl)piperidin-4-ol: A mixture of piperidin-4-ol (607 mg, 5.88 mmol), 4-ethoxy-l,2-difluorobenzene (0.4 mL, 2.83 mmol), pyridine (0.6 mL) and DMSO (1.2 mL) was heated at 160 °C in a microwave reactor for 20 h. Then, the mixture was partitioned between 1/4 Hex/EtOAc (50 mL) and 2% NaHCC^ (30 mL). The organic layer was washed with 2% aHC03 (2x20 mL) and sat'd NaCl (20 mL), dried ( a2S0 4 ) and evaporated. Purification via silica gel chromatography (3/2 Hex/EtOAc) gave the desired product (222 mg, 32.1% yield) as a colorless oil: LCMS [M+H] 240.

[00396] 93B. 4-((45,55)-5-(Cyanomethyl)-l-(4-((l-(5-ethoxy-2- fluorophenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-4,5-dihydro -lH-pyrazol-3- yl)benzonitrile: To a stirred solution of 4-((45,5S)-5-(cyanomethyl)-l-(4-hydroxyphenyl)- 4-methyl-4,5-dihydro-lH-pyrazol-3-yl)benzonitrile (310 mg, 0.970 mmol), l-(5-ethoxy- 2-fluorophenyl)piperidin-4-ol (368 mg, 1.492 mmol) and triphenylphosphine (420 mg, 1.585 mmol) in THF (5.8 mL) at rt was added di-tert-butylazodicarboxylate (375 mg, 1.596 mmol). The resulting solution was stirred at rt for 25 h and then evaporated. The residue was purified by two consecutive chromatographies (Si0 2 , first 95/5 CHC^/Ether and then 4/1 to 3/2 Hex/EtOAc) to give the desired product (390 mg, 74.0% yield) as a yellow oil: LC-MS [M+H] 538.

[00397] 93C. Methyl 2-((45,55)-3-(4-cyanophenyl)-l-(4-((l-(5-ethoxy-2- fluorophenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-4,5-dihydro -lH-pyrazol-5-yl)acetate: 4-((45 , ,55)-5-(Cyanomethyl)-l-(4-((l-(5-ethoxy-2-fluorophenyl )piperidin-4- yl)oxy)phenyl)-4-methyl-4,5-dihydro-lH-pyrazol-3-yl)benzonit rile (389 mg, 0.716 mmol) was dissolved in -2.5M HCl/MeOH, MeOAc, CH 2 C1 2 solution [36.6 mL, prepared by addition of AcCl (6.6 mL) to 3/2 CH 2 Cl 2 /MeOH (30.0 mL) at 0 °C and then stirring at rt for 30 min]. The resulting solution was stirred at rt for 6.5 h and then evaporated to a volume of about 5 mL. The oily remnant was stripped from MeOH (2x20 mL), taken up in EtOAc (100 mL) and, washed with 5% NaHC0 3 (2x50 mL) and sat'd

NaCl (40 mL). The organic layer was dried (Na 2 S0 4 ) and concentrated. Purification via silica gel chromatography provided the desired (75 mg, 18% yield) as a yellow oil: LC- MS [M+H] 571.

[00398] Example 93: To a stirred solution of methyl 2-((45",55)-3-(4-cyanophenyl)-l- (4-((l-(5-ethoxy-2-fluorophenyl)piperidin-4-yl)oxy)phenyl)-4 -methyl-4,5-dihydro-lH- pyrazol-5-yl)acetate (17 mg, 0.03 mmol) in THF (0.9 mL) and water (0.09 mL) at rt was added 1.0M aqueous lithium hydroxide (0.09 mL, 0.09 mmol). After stirring at rt for 4.5 h, the reaction mixture was partially evaporated to remove most of the THF. The remaining mixture was partitioned between water (50 mL) and Hex (15 mL). The aqueous layer was acidified to pH 2 by dropwise addition of 1M HCl, and then extracted with CH 2 C1 2 (3x30 mL). The combined organic extracts were dried (Na 2 S0 4 ) and concentrated. The residue was dried under vacuum to afford the desired product (16.7 mg, 92% yield) as a yellow solid: LC-MS [M+H] 557. l H NMR (500MHz, CD 2 C1 2 ) δ 7.82 (d, J= 8.2 Hz, 2H),7.66 (d, J= 8.2 Hz, 2H), 7.14 (d, J= 8.8 Hz, 2H), 6.95 (d, J= 8.8 Hz, 2H), 6,91 (dd, J=12.1, 8.8 Hz, 1H), 6.55 (m, 1H), 6.41 (m, 1H), 4.48 (d, J= 9.6 Hz, 1H), 4.38 (m, 1H), 3.97 (q, J= 7.0 Hz, 2H), 3.44 (m, 1H), 3.35 (m, 2H), 2.96 (m, 2H), 2.85 (dd, J= 16.3, 2.8 Hz, 1H), 2.41 (dd, J=16.3, 10.5 Hz, 1H), 2.11 (m, 2H), 1.95 (m, 2H), 1.37 (t, J= 7.0 Hz, 3H), 1.32 (d, J= 7.2 Hz, 3H). Analytical HPLC (Zorbax method, 0% Solvent B start): RT = 8.23 min, HI: 98%. hGPR40 EC 50 = 82 nM. hGPR40 IP1 EC 5 o = 7 nM. Acute oral glucose tolerance: -56% @ 0.3 mg/kg.

Example 94

2-((4S,5S)-3-(4-Cyano-2-fluorophenyl)-l-(4-((l-(5-ethoxy-2-f luorophenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00399] 94A. 4-((45',55)-5-(Cyanomethyl)-l-(4-hydroxyphenyl)-4-methyl-4,5 -dihydro- lH-pyrazol-3-yl)-3-fluorobenzonitrile was prepared from 3-fluoro-4-formylbenzonitrile following the procedure for the preparation of 4-((4S,55)-5-(cyanomethyl)-l-(4- hydroxyphenyl)-4-methyl-4,5-dihydro-lH-pyrazol-3-yl)benzonit rile (91G): LC-MS

[Μ+Η] 335.

[00400] Example 94 (yellow solid, 11.7 mg) was prepared from 4-((45',55)-5- (cyanomethyl)-l-(4-hydroxyphenyl)-4-methyl-4,5-dihydro-lH-py razol-3-yl)-3- fluorobenzonitrile following the procedure for Example 93 but purification of the final acid was achieved by acidification of the reaction mixture, extraction with CH2CI2 and chromatography (Si0 2 , 95/5 CHCl 3 /MeOH). LC-MS [M+H] 575. ¾ NMR (500MHz, CD 2 Cl 2 ) 5 8.16 (t, J= 8.0 Hz, 1H),7.49 (dd, J= 8.2, 1.6 Hz, 1H), 7.44 (dd, J= 1 1.2, 1.6

Hz, 1H), 7.18 (d, J= 9.0 Hz, 2H), 6.99 (d, J= 9.0 Hz, 2H), 6,95 (dd, J=12.4, 8.8 Hz, 1H), 6.55 (dd, J=7.4, 3.0 Hz, 1H), 6.44 (dt, J= 8.8, 3.0 Hz, 1H), 4.53 (d, J= 10.3, 2.6 Hz, 1H), 4.42 (m, 1H), 4.01 (q, J= 7.0 Hz, 2H), 3.58 (m, 1H), 3.37 (m, 2H), 2.99 (m, 2H), 2.89 (dd, J= 16.3, 2.9 Hz, 1H), 2.49 (dd, J=16.3, 10.3 Hz, 1H), 2.14 (m, 2H), 1.97 (m, 2H), 1.41 (t, J= 7.0 Hz, 3H). 1.32 (d, J= 7.2 Hz, 3H). Analytical HPLC (Zorbax method, 0% Solvent B start): RT = 8.16 min, HI: 100%. hGPR40 EC 50 = 59 nM. Example 95

2-((45 , ,55)-l-(4-((l-(5-Ethoxy-2-fluorophenyl)piperidin-4-yl) oxy)phenyl)-3-(4- fluorophenyl)-4-methyl-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00401] 95A. 2-((45,55)-l-(4-Bromophenyl)-3-(4-fluorophenyl)-4-methyl-4,5 - dihydro-lH-pyrazol-5-yl)acetonitrile was prepared from 4-fluorobenzaldehyde and (4- bromophenyl)hydrazine following the procedure for the preparation of 4-((4,S,55)-5- (cyanomethyl)-l-(4-methoxyphenyl)-4-methyl-4,5-dihydro-lH-py razol-3-yl)benzonitrile (example 91G). LC-MS [Μ+Η] 372, 374.

[00402] 95B. 2-((45,55')-3-(4-Fluorophenyl)-4-methyl-l-(4-(4,4,5,5-tetram ethyl-l,3,2- dioxaborolan-2-yl)phenyl)-4,5-dihydro-lH-pyrazol-5-yl)aceton itrile: To a stirred solution of 2-((45 , ,55)-l-(4-bromophenyl)-3-(4-fluorophenyl)-4-methyl-4,5 -dihydro-lH-pyrazol-5- yl)acetonitrile (511 mg, 1.373 mmol) in DMF (15 mL) was added 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(l,3,2-dioxaborolane) (558 mg, 2.196 mmol), potassium acetate (404 mg, 4.12 mmol) and 1 , l'-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane complex, (56.5 mg, 0.069 mmol). The resulting reaction mixture was purged with argon three times and then heated to 80 °C. After stirring at this temperature overnight, the reaction mixture was cooled to rt, diluted with water and extracted with CH 2 Cl2. The combined organic extracts were washed with sat'd NaCl, dried ( a 2 S04) and purified via silica gel chromatography to afford the desired product (447 mg, 72.2% yield) as a yellowish solid.: LC-MS [M+H] 420.

[00403] 95C. 2-((45,55)-3-(4-Fluorophenyl)-l-(4-hydroxyphenyl)-4-methyl-4 ,5- dihydro-lH-pyrazol-5-yl)acetonitrile: To a stirred solution of 2-((45',55)-3-(4- fluorophenyl)-4-methyl-l-(4-(4,4,5,5-tetramethyl-l,3,2-dioxa borolan-2-yl)phenyl)-4,5- dihydro-lH-pyrazol-5-yl)acetonitrile (447 mg, 0.991 mmol) in ethyl acetate (10.0 mL) at rt was added 30% hydrogen peroxide (3.04 mL, 29.7 mmol) over 2 min. The resulting solution was stirred at rt for 5 h. At this time, the reaction mixture was cooled to 0 °C and quenched by slow addition of aqueous sodium sulfite solution. The aqueous mixture was diluted with water and extracted with CH2CI2. The combined organic extract was dried ( a 2 S04), concentrated and purified via silica gel chromatography (0 to 60%

EtO Ac/Hex) to provide the desired product (226 mg, 73.7% yield) as a dark solid: LC- MS [M+H] 310.

[00404] Example 95 (yellowish solid, 10 mg) was prepared from 2-((45,55)-3-(4- fluorophenyl)-l-(4-hydroxyphenyl)-4-methyl-4,5-dihydro-lH-py razol-5-yl)acetonitrile following the procedure for Example 93. The resulting TFA salt was treated with an equivalent amount of LiOH, diluted with water and the pH of the aqueous mixture was adjusted to 2 by dropwise addition of 1M HC1. The aqueous mixture was extracted with CH 2 C1 2 and, the organic extract was dried ( a 2 S0 4 ) and concentrated. LC-MS [M+H]

550. !H MR (400MHZ, CDCI3) δ 7.72 (m, 2H),7.09 (m, 4H), 6.92 (m, 3H), 6,55 (m,

1H), 6.40 (m, 1H), 4.38 (m, 2H), 3.99 (q, J= 7.0 Hz, 2H), 3.36 (m, 3H), 2..97 (m, 2H), 2.84 (dd, J= 16.0, 2.9 Hz, 1H), 2.36 (dd, J=16.0, 10.5 Hz, 1H), 2.11 (m, 2H), 1.98 (m, 2H), 1.40 (t, J= 7.0 Hz, 3H), 1.32 (d, J= 7.1 Hz, 3H). Analytical HPLC (Zorbax method, 0% Solvent B start): RT = 8.81 min, HI: 97%. hGPR40 EC 50 = 108 nM.

Example 99

2-((45,55)-3-(4-Cyanophenyl)-l-(4-(((3R,4R)-l-(5-ethoxy-2-fl uorophenyl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-ethyl-4,5-dihydro-lH-pyra zol-5-yl)acetic acid

[00405] 99A. (£)-Pent-2-enoic anhydride: To a stirred solution of (is)-pent-2-enoic acid (5 g, 48.9 mmol) in CH 2 C1 2 (50 mL) was added triethylamine (6.82 mL, 48.9 mmol).

The resulting solution was purged with argon and then cooled to 0 °C. Triphosgene (2.90 g, 9.79 mmol) was slowly added in portions and after completion of the addition, the resulting mixture was allowed to warm up to rt and stirred under argon overnight. The solvent was removed by blowing nitrogen onto the reaction mixture and the residual white solid was taken up in EtO Ac (20 mL). The resulting suspension was filtered and the filter cake was rinsed with Et20. The combined filtrates were concentrated to afford the desired product (5.10 g, quantitative yield) as an orange liquid: LC-MS [M+Na] 205.

[00406] 99B. (SJ^-3-(Pent-2-enoyl)-4-phenyloxazolidin-2-one: To a light suspension of (5)-4-phenyloxazolidin-2-one (3.8 g, 23 mmol), lithium chloride (1.017 g, 24 mmol) and triethylamine (4.32 mL, 31 mmol) in THF (50 mL) at -20 °C was added (is)-pent-2- enoic anhydride (5.09 g, 27.9 mmol) dropwise. After completion of addition, the cold bath was removed and the mixture was allowed to warm to rt. The mixture was stirred at rt for 2.5 days and the resulting thick suspension was diluted with EtOAc. The mixture was washed sequentially with 0.2M HC1, sat'd NaHCC^, water and sat'd NaCl. The organic layer was dried ( a 2 S04) and concentrated to give an orange liquid which was purified via silica gel chromatography to afford (the desired product (4.37 g, 76% yield) as a white solid: LC-MS [M+H] 246.

[00407] 99C. 4-((45,55)-5-(Cyanomethyl)-4-ethyl-l-(4-hydroxyphenyl)-4,5-d ihydro- lH-pyrazol-3-yl)benzonitrile was prepared from (5',£)-3-(pent-2-enoyl)-4- phenyloxazolidin-2-one following the procedure for the preparation of 4-((45',55)-5-

(cyanomethyl)-l-(4-hydroxyphenyl)-4-methyl-4,5-dihydro-lH -pyrazol-3-yl)benzonitrile (91G): LC-MS [Μ+Η] 331.

[00408] Example 99 (yellow solid, 5.0 mg) was prepared from 4-((45,55)-5- (cyanomethyl)-4-ethyl-l-(4-hydroxyphenyl)-4,5-dihydro-lH-pyr azol-3-yl)benzonitrile and (3R,45)-l-(5-ethoxy-2-fluorophenyl)-3-methylpiperidin-4-ol following the procedure for Example 93 but no purification by preparative RP-HPLC was required: LC-MS

[M+H] 585. !H NMR (400MHZ, CDC1 3 , selected peaks) δ 7.80 (d, J= 8.5 Hz, 2H),7.64

(d, J= 8.5 Hz, 2H), 7.13 (d, J= 8.8 Hz, 2H), 6.94 (d, J= 8.8 Hz, 2H), 6,91, 1H, overlapped), 6.53 (m, 1H), 6.40 (m, 1H), 4.58 (m, 1H), 3.98 (q, J= 7.0 Hz, 2H), 3.83 (m, 1H), 3.44 (m, 2H), 3.32 (m, 1H), 2.87 (dd, J= 15.8, 3.0 Hz, 1H), 2.81 (m, 1H, overlapped), 2.39 (dd, J=15.8, 10.4 Hz, 1H), 1.40 (t, J= 7.0 Hz, 3H), 1.13 (d, J= 6.7 Hz, 3H), 0.97 (t, J= 7.3 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 8.47 min, HI: 98%. hGPR40 EC 50 = 25 nM. hGPR40 IP1 EC 50 = 20 nM. Example 102 2-((4S,5S)-l-(4-((l-(2-Fluoro-5-hydroxyphenyl)piperidin-4-yl )oxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid

[00409] 102A. Ethyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5-hydroxyphenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetate: To a solution of EtOH (0.86 niL) was added acetyl chloride (0.18 niL, 2.6 mmol) to generate anhydrous HC1. After addition, the mixture was warmed to room temp, and 2-((4S,5S)-l- (4-(( 1 -(2-fluoro-5-methoxyphenyl)piperidin-4-yl)oxy)phenyl)-4-meth yl-3 - (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid (Example 1, 0.044 g, 0.086 mmol) was added. The mixture was stirred at rt for 1.5h. The reaction mixture was evaporated to dryness. The residue was dissolved in CH 2 Ci2 (0.6 mL) and was cooled to

-78 °C, and BBr3 (0.8 mL, 8.46 mmol) was added. The reaction mixture was stirred for 2h, and was warmed to -40 °C overnight. The reaction solution was poured over ice and, after 30 min agitation, the mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over MgSC^, filtered and evaporated to give 85 mg of a yellow oil that was purified via silica chromatography to give 25 mg of desired product as a clear oil. Anal. Calc'd for C26H29F4 3O4: 523.21, LCMS obs. [M+H] = 524.1.

[00410] Example 102: To a solution of ethyl 2-((4S,5S)-l-(4-((l-(2-fluoro-5- hydroxyphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluo romethyl)-4,5-dihydro-lH- pyrazol-5-yl)acetate (0.01 g, 0.019 mmol) in acetonitrile (0.3 mL) and water (0.2 mL) was added lithium hydroxide (3 mg, 0.1 mmol), and the mixture was stirred for lh. The reaction mixture was diluted with acetonitrile and IN HC1 and the mixture was purified via preparative reverse-phase HPLC. Product-containing fractions were evaporated to remove acetonitrile. The water layer was extracted 2x with CH2CI2. The combined organic layers were dried over a 2 S04, and the organic layer was decanted. The solution was evaporated to give 8 mg of a white foam that was re-dissolved in acetonitrile and 3M HC1 (aq). The mixture was stirred for 5 min, then evaporated and placed on the pump overnight to give 8 mg of 2-((4S,5S)-l-(4-((l-(2-fluoro-5-hydroxyphenyl)piperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid as a white solid. LCMS Anal. Calc'd for C 2 4H 2 5F4 3 04: 495.18; found [M+H] = 496.1. l H NMR (400MHz, CD 3 CN) δ 7.61 (br. s., 1H), 7.12 - 6.89 (m, 2H), 6.84 (d, J=9.1 Hz, 2H),

6.70 (br. s., 1H), 4.50 (br. s., 1H), 4.28 (d, J=9.6 Hz, 1H), 3.67 (br. s., 2H), 3.33 (br. s., 2H), 3.18 (br. s., 1H), 2.58 (dd, J=16.4, 3.3 Hz, 2H), 2.34 (dd, J=16.4, 9.6 Hz, 4H), 1.15 (d, J=7.3 Hz, 3H). Analytical HPLC (Orthogonal method): RT=10.2 min, HI: 98%.

hGPR40 EC 50 = 190 nM.

Example Al (Isomer 1) and (Isomer 2)

2-((4S,5S)-l-(4-((l-(5-Chloro-2-methoxypyridin-4-yl)-3-fl uoropiperidin-4- yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-p yrazol-5-yl)acetic acid, HCl

[00411] A1A. Benzyl 4-((trimethylsilyl)oxy)-5,6-dihydropyridine-l(2H)-carboxylat e: To a solution of benzyl 4-oxopiperidine-l-carboxylate (1.3 g, 5.4 mmol) in DMF (4 mL) was added chlorotrimethylsilane (0.8 mL, 6.53 mmol) followed by triethylamine (1.5 mL, 10.89 mmol). The resulting heterogeneous mixture was warmed to 80 °C and stirred for 16 h. The cooled mixture was diluted with hexanes (50 mL), washed with sat. NaHCC^ (30 mL) and brine (30 mL), then dried over MgSC^, filtered and concentrated to afford benzyl 4-((trimethylsilyl)oxy)-5,6-dihydropyridine-l(2H)-carboxylat e (1.5 g, 4.91 mmol, 90% yield) as a clear oil. l H NMR (400MHz, CDC1 3 ) δ 7.22 - 7.08 (m, 5H), 4.98 - 4.94 (m, 2H), 4.60 (br. s., 1H), 3.77 (q, J=2.3 Hz, 2H), 3.42 (t, J=5.6 Hz, 2H), 1.94 (br. s., 2H), 0.04 -0.04 (m, 9H).

[00412] A1B. Benzyl 3-fluoro-4-oxopiperidine-l-carboxylate: To a solution of benzyl 4-((trimethylsilyl)oxy)-5,6-dihydropyridine-l(2H)-carboxylat e (1.5 g, 4.98 mmol) in acetonitrile (31 mL) at rt was added SELECTFLUOR® (2.1 g, 5.97 mmol) portionwise over 10 min. The mixture was stirred for 2 h, then concentrated to dryness and partitioned between EtOAc and brine. The aq layer was extracted with EtOAc and the combined organic phases were washed with brine, dried over MgSC^, filtered and concentrated to afford benzyl 3-fluoro-4-oxopiperidine-l-carboxylate (1.2 g, 4.78 mmol, 96% yield) as a solid. !H MR (400MHZ, CDC1 3 ) δ 7.51 - 7.30 (m, 5H), 5.27 - 5.16 (m, 2H), 4.97 - 4.70 (m, 1H), 4.47 (br. s., 1H), 4.33 - 4.18 (m, 1H), 3.59 - 3.28 (m, 2H), 2.72 - 2.41 (m, 2H).

[00413] AIC. Benzyl 3-fluoro-4-hydroxypiperidine-l-carboxylate: To a solution of benzyl 3-fluoro-4-oxopiperidine-l-carboxylate (880 mg, 3.50 mmol) in MeOH (7 mLl) was added sodium borohydride (130 mg, 3.50 mmol) slowly. After 1 h, 10% KHSO4 (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x30 mL), and the combined organic extracts were dried ( a 2 S04), filtered, and concentrated to afford benzyl 3-fluoro-4-hydroxypiperidine-l-carboxylate (870 mg, 3.44 mmol, 98% yield). LC-MS Anal. Calc'd for C 13 H 16 FN0 3 , found [M+H] 253.26 254. Ή NMR (400MHz, CDC1 3 ) δ 7.47 - 7.27 (m, 5H), 5.13 (s, 2H), 4.75 - 4.52 (m, 1H), 4.07 - 3.68 (m, 3H), 3.61 - 3.04 (m, 2H), 2.22 (d, J=5.0 Hz, 1H), 1.93 - 1.63 (m, 2H).

[00414] AID. Benzyl 4-((tert4outyldimethylsilyl)oxy)-3-fluoropiperidine-l- carboxylate: To a solution of benzyl 3-fluoro-4-hydroxypiperidine-l-carboxylate (410 mg, 1.62 mmol) in DCM (1.6 mL) and triethylamine (670 μί, 4.86 mmol) was added tert-butyldimethylsilyl trifluoromethanesulfonate (390 μί, 1.70 mmol) at 0 °C. After 1 h, sat. aq. NaHCC^ (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x30 mL), and the combined organic extracts were dried

( a 2 S04), filtered, and concentrated to afford benzyl 4-((tert-butyldimethylsilyl)oxy)-3- fluoropiperidine-l-carboxylate (450 mg, 1.224 mmol, 76% yield). !H MR (400MHZ, CDCI3) δ 7.36 - 7.20 (m, 5H), 5.05 (s, 2H), 4.43 - 4.22 (m, 1H), 4.07 - 3.96 (m, 1H), 3.88 - 3.62 (m, 2H), 3.59 - 3.24 (m, 2H), 1.75 - 1.50 (m, 2H), 0.84 (s, 9H), 0.02-0.05 (m, 6H).

[00415] A IE. 4-((tert-Butyldimethylsilyl)oxy)-3-fluoropiperidine: A mixture of benzyl 4-((tert-butyldimethylsilyl)oxy)-3-fluoropiperidine-l-carbox ylate (440 mg, 1.20 mmol) and Pd on activated carbon (127 mg, 0.120 mmol) in MeOH (12 mL) was purged with H 2 for 30 min and stirred under atm H 2 at rt for 1 h. The mixture was filtered through CELITE®, washed with EtOAc (30 mL) and MeOH (30 mL) and concentrated to afford 4-((tert-butyldimethylsilyl)oxy)-3-fluoropiperidine (270 mg, 1.157 mmol, 97% yield) as a clear oil. l H NMR (400MHz, CDC1 3 ) δ 4.50 - 4.27 (m, 1H), 3.97 - 3.77 (m, 1H), 3.19 - 3.05 (m, 1H), 2.93 (ddd, J=13.1, 6.3, 4.1 Hz, 1H), 2.82 - 2.65 (m, 2H), 2.62 - 2.46 (m, 1H), 1.73 - 1.50 (m, 2H), 0.88 (s, 9H), 0.00 (d, J=3.8 Hz, 6H).

[00416] A1F. 4-(4-((tert-Butyldimethylsilyl)oxy)-3-fluoropiperidin-l-yl)- 5-chloro-2- methoxypyridine: A mixture of 4-((tert-butyldimethylsilyl)oxy)-3-fluoropiperidine (194 mg, 0.83 mmol), 4-bromo-5-chloro-2-methoxypyridine (185 mg, 0.83 mmol) and Sphos precatalyst (6 mg, 8.3 μιηοΐ) in THF (1.7 mL) was purged with argon and lithium bis(trimethylsilyl)amide (1M solution in THF, 1 mL, 1.0 mmol) was added. The reaction was heated at 70 °C for 2 h, and sat. aq. NaHCC^ (10 mL) was added slowly to the reaction mixture. The mixture was extracted with EtOAc (2x10 mL), and the combined organic extracts were washed with water (20 mL) and brine (20 mL), and dried

( a 2 S0 4 ), filtered, and concentrated. Purification by chromatography gave 4-(4-((tert- butyldimethylsilyl)oxy)-3-fluoropiperidin-l-yl)-5-chloro-2 -methoxypyridine (182 mg, 0.49 mmol, 58% yield). LC-MS Anal. Calc'd for C 17 H 28 ClFN 2 0 2 Si 374.16, found

[M+H] 374.9.

[00417] A1G. l-(5-Chloro-2-methoxypyridin-4-yl)-3-fluoropiperidin-4-ol: To a mixture of 4-(4-((ter?-butyldimethylsilyl)oxy)-3-fluoropiperidin- 1 -yl)-5-chloro-2- methoxypyridine (192 mg, 0.51 mmol) in THF (1.0 mL) was added TBAF (0.6 mL, 0.61 mmol). The reaction mixture was stirred at 23 °C for 2 h and sat. aq. NaHCC^ (10 mL) was added slowly to the reaction mixture. The mixture was then extracted with EtOAc

(2x10 mL), and the combined organic extracts were washed with water (20 mL) and brine (20 mL), and dried (Na 2 S0 4 ), filtered, and concentrated. Purification by chromatography gave l-(5-chloro-2-methoxypyridin-4-yl)-3-fluoropiperidin-4-ol (1 10 mg, 0.42 mmol, 82% yield) as a white foam. LC-MS Anal. Calc'd for C U H 24 C1FN 2 0 2 260.07, found [M+H] 261.0.

[00418] A1H. Methyl 2-((4S,5S)-l-(4-((l-(5-chloro-2-methoxypyridin-4-yl)-3- fluoropiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl )-4,5-dihydro-lH-pyrazol- 5-yl)acetate: To a solution of methyl 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetate (86 mg, 0.27 mmol) and l-(5- chloro-2-methoxypyridin-4-yl)-3-fluoropiperidin-4-ol (47 mg, 0.18 mmol) in toluene (2.2 mL) was added tributylphosphine (75 μί, 0.29 mmol). While stirring, (E)-diazene-l,2- diylbis(piperidin-l-ylmethanone) (73 mg, 0.29 mmol) was added portionwise to the reaction mixture, and the reaction mixture was heated at 50 °C for 120 min. The mixture was cooled to rt and 6 mL hexanes was added to the mixture and white solid was formed. The mixture was filtered, and the filtrate was concentrated. Purification by

chromatography yielded methyl 2-((4S,5S)-l-(4-((l-(5-chloro-2-methoxypyridin-4-yl)-3- fluoropiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl )-4,5-dihydro-lH-pyrazol- 5-yl)acetate (20 mg, 0.036 mmol, 20% yield) as a white foam. LC-MS Anal. Calc'd for C 25 H 27 C1F 4 N 4°4 558 · 2 > found [ M+H ] 559 · 2 · lfi NMR (400MHz, CDC1 3 ) δ 7.97 - 7.87 (m, 1H), 7.04 - 6.97 (m, 2H), 6.93 - 6.86 (m, 2H), 6.23 (s, 1H), 4.86 - 4.61 (m, 1H), 4.31 (ddd, J=10.6, 7.5, 3.6 Hz, 2H), 3.87 - 3.79 (m, 3H), 3.70 - 3.58 (m, 4H), 3.41 - 3.26 (m, 1H), 3.20 - 3.09 (m, 2H), 2.95 - 2.87 (m, 1H), 2.72 (dd, J=16.1, 3.1 Hz, 1H), 2.34 (dd, J=16.1, 10.1 Hz, 1H), 2.25 - 2.13 (m, 1H), 1.91 - 1.82 (m, 1H), 1.27 (d, J=7.0 Hz, 3H).

[00419] Example Al (Isomer 1) and (Isomer 2): To a stirred solution 0 f methyl 2- ((4S,5 S)- 1 -(4-(( 1 -(5 -chloro-2-methoxypyridin-4-yl)-3 -fluoropiperidin-4-yl)oxy)phenyl)- 4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)ace tate (18 mg, 0.032 mmol) in THF (0.3 mL) and water (30 μΓ) at rt was added 2N LiOH solution (40 μί, 0.081 mmol). The reaction mixture was stirred at rt for 16 h. Then, IN HC1 (80 μί, 0.081 mmol) was added at 0 °C to pH = 4-5, followed by warming to rt. The solvent was evaporated and the residue was extracted with EtOAc 3 times. The organic portions were dried over a 2 S04, filtered and concentrated. The diastereomers were separated by chiral Prep. SFC to provide Example Al, Isomer 1 and Isomer 2 as single stereoisomers.

Example Al, Isomer 1 : (white solid, 5 mg). LC-MS Anal. Calc'd for C24H25CIF4 4O4

544.2, found [M+H] 545.2. !H MR (400MHZ, CD 3 CN) δ 8.03 (s, 1H), 7.20 - 6.97 (m, 4H), 6.43 (s, 1H), 4.94 - 4.73 (m, 1H), 4.58 - 4.41 (m, 2H), 3.88 (s, 3H), 3.80 - 3.69 (m, 1H), 3.48 - 3.30 (m, 2H), 3.19 (dt, J=12.4, 7.3 Hz, 1H), 3.11 - 3.02 (m, 2H), 2.31 - 2.22 (m, 2H), 1.91 - 1.79 (m, 2H), 1.36 - 1.30 (m, 3H). HPLC (Orthogonal method, 50% Solvent B start): RT = 9.2 min, HI: 100%. hGPR40 EC 50 = 184 nM. Example Al, Isomer

2: (white solid, 5 mg). LC-MS Anal. Calc'd for C24H25CIF4N4O4 544.2, found [M+H]

545.2. !H MR (400MHZ, CD 3 CN) δ 8.03 (s, 1H), 7.15 - 6.92 (m, 4H), 6.43 (s, 1H), 4.92 - 4.58 (m, 1H), 4.47 (br. s., 2H), 3.87 (s, 3H), 3.81 - 3.66 (m, 1H), 3.46 - 3.30 (m, 2H), 3.24 - 3.13 (m, 2H), 3.10 - 3.00 (m, 2H), 2.08 (d, J=7.0 Hz, 2H), 1.84 (dd, J=8.7, 3.9 Hz, 1H), 1.35 - 1.30 (m, 3H). HPLC (Orthogonal method, 50% Solvent B start): RT = 9.1 min, HI: 100%. hGPR40 EC 50 = 218 nM.

Example A2

2-((4S,5S)-l-(4-(l-(5-Methoxy-2-methylphenyl)piperidin-4- yloxy)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetic acid

[00420] A2A. l-(5-Methoxy-2-methylphenyl)piperidin-4-one. To a round bottom flask was added 5-methoxy-2-methylaniline (265 mg, 1.93 mmol), K2CO3 (40 mg, 0.29 mmol) and ethanol (2.5 mL). To this mixture at 100 °C was added a slurry of 1-benzyl-l- methyl-4-oxopiperidin- 1 -ium, iodide salt (960 mg, 2.9 mmol) in water (1 mL) over 20 min, and stirring and heating were continued for 1 h. The solution was cooled to rt, and then it was extracted with CH2CI2 (2 x 20 mL). The combined organic extracts were washed with brine (20 mL), dried over MgS0 4 , filtered and concentrated. The residue was purified via silica chromatography to give l-(5-methoxy-2-methylphenyl)piperidin- 4-one (120 mg, 0.55 mmol, 28% yield) as a clear oil. LC-MS Anal. Calc'd for

C 13 H 17 N0 2 : 219.1, found [M+H] 220.1. !H MR (400MHZ, CDCI3) δ 7.15 (d, J=8.1 Hz, 1H), 6.69 - 6.54 (m, 2H), 3.81 (s, 3H), 3.23 (t, J=6.1 Hz, 4H), 2.63 (t, J=6.1 Hz, 4H), 2.33 (s, 3H). [00421] A2B. l-(5-Methoxy-2-methylphenyl)piperidin-4-ol. To a round bottom flask was added l-(5-methoxy-2-methylphenyl)piperidin-4-one (1 10 mg, 0.49 mmol), THF (2 mL) and NaBH 4 (19 mg, 0.49 mmol), and the solution was stirred at rt for 30 min. The reaction was quenched with water (1 mL), and the mixture was partitioned between EtOAc (30 mL) and water (15 mL). The organic layer was separated, washed with brine (10 mL), dried over MgS0 4 , filtered and concentrated. The crude product was purified via silica chromatography to give l-(5-methoxy-2-methylphenyl)piperidin-4-ol (95 mg, 0.43 mmol, 87% yield) as a clear oil. LC-MS Anal. Calc'd for C 13 H 19 N0 2 : 221.3, found [M+H] 222.2. !H MR (500MHZ, chloroform-d) δ 7.09 (d, J=8.3 Hz, 1H), 6.62 (d, J=2.8 Hz, 1H), 6.55 (dd, J=8.3, 2.5 Hz, 1H), 3.86 (d, J=3.3 Hz, 1H), 3.18 - 3.06 (m, 2H), 2.79 - 2.65 (m, 2H), 2.27 - 2.23 (m, 3H), 2.10 - 1.99 (m, 2H), 1.75 (dtd, J=12.7, 9.3, 3.7 Hz, 2H), 1.48 (br. s., 1H).

[00422] A2C. Methyl 2-((4S,5S)-l-(4-(l-(5-methoxy-2-methylphenyl)piperidin-4- yloxy)phenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-py razol-5-yl)acetate. To a round bottom flask was added l-(5-methoxy-2-methylphenyl)piperidin-4-ol (35 mg, 0.16 mmol), methyl 2-((4S,5 S)- 1 -(4-hydroxyphenyl)-4-methyl-3 -(trifluoromethyl)-4,5- dihydro-lH-pyrazol-5-yl)acetate (50 mg, 0.16 mmol), toluene (1 mL), BU3P (0.062 mL, 0.25 mmol) and l, l '-(azodicarbonyl)dipiperidine (63.8 mg, 0.253 mmol). The mixture was stirred at 65 °C for 1 hr. The reaction solution was cooled and diluted with heptane. The mixture was filtered and the filtrate was concentrated. The residue was purified via silica gel chromatography to give methyl 2-((4S,5S)-l-(4-((l-(5-methoxy-2- methylphenyl)piperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H- pyrazol-5-yl)acetate (45 mg, 0.087 mmol, 55% yield) as a clear oil. LC-MS Anal. Calc'd for C 27 H32F3 30 4 : 519.2, found [M+H] 520.2.

[00423] Example A2: To a round bottom flask was added methyl 2-((4S,5S)-l-(4-((l- (5-methoxy-2-methylphenyl)piperidin-4-yl)oxy)phenyl)-4-methy l-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetate (45 mg, 0.087 mmol), 2M LiOH (0.26 mL, 0.52 mmol) and THF (1 mL). The reaction solution was stirred at rt for 1.5 h, and the solution was acidified with IN HC1 to pH <4. The mixture was extracted with EtOAc (10 mL). The organic layer was washed with water (10 mL) and brine (10 mL), and then it was dried over MgS0 4 , filtered and concentrated. The residue was purified by Prep-HPLC to give 2-((4S,5S)-l-(4-(l-(5-methoxy-2-methylphenyl)piperidin-4-ylo xy)phenyl)-4-methyl- 3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetic acid (31 mg, 0.062 mmol, 71% yield). LC-MS Anal. Calc'd for C26H30F3 3O4: 505.2, found [M+H] 506.1. !H MR (500MHz, DMSO-d 6 ) δ 7.12 - 7.03 (m, 3H), 6.99 (d, J=9.1 Hz, 2H), 6.59 (br. s., 1H),

6.55 (dd, J=8.3, 1.9 Hz, 1H), 4.45 (d, J=3.9 Hz, 2H), 3.72 (s, 4H), 3.39 - 3.33 (m, 2H), 3.05 (d, J=4.4 Hz, 2H), 2.78 (t, J=9.9 Hz, 2H), 2.66 (dd, J=16.2, 3.0 Hz, 1H), 2.18 (s, 3H), 2.05 (br. s., 2H), 1.85 - 1.72 (m, 2H), 1.27 (d, J=6.9 Hz, 3H). hGPR40 EC 50 = 161 nM. Example A3

2-((4S,5S)-l-(2-Bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyrid in-4-yl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- -yl)acetic acid

[00424] A3 A. (4S,5R)-l-(4-Bromophenyl)-5-(((tert-butyldimethylsilyl)oxy)m ethyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazole: To a solution of ((4S,5R)-l-(4- bromophenyl)-4-methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyr azol-5-yl)methanol (1G, 285 mg, 0.845 mmol) and imidazole (151 mg, 2.198 mmol) in dichloromethane (2.7 mL) at rt was added a solution of tert-butyldimethylsilyl chloride (171 mg, 1.10 mmol) in CH2CI2 (1.4 mL) dropwise. The reaction mixture was stirred at rt for 19 h. After this time, the mixture was diluted with EtOAc (70 mL) and washed with sat'd NaHCC^ (2x30 mL) and sat'd NaCl (20 mL). The organic layer was dried ( a2S0 4 ) and evaporated. Chromatography (Si0 2 , 9/lHex/Ether) of the crude provided (4S,5R)-l-(4- bromophenyl)-5-(((ter?-butyldimethylsilyl)oxy)methyl)-4-meth yl-3-(trifluoromethyl)-4,5- dihydro-lH-pyrazole (351 mg, 92% yield) as a colorless oil: LC-MS [M+H] 451, 453.

[00425] A3B. (4S,5R)-5-(((tert-Butyldimethylsilyl)oxy)methyl)-4-methyl- 1 -(4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)-3-(trif luoromethyl)-4,5-dihydro- lH-pyrazole: A flask containing a suspension of (4S,5R)-l-(4-bromophenyl)-5-(((tert- butyldimethylsilyl)oxy)methyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazole (351 mg, 0.778 mmol), bis(pinacolato)diboron (230 mg, 0.897 mmol) and potassium acetate (232 mg, 2.340 mmol) in DMF (1.4 mL) was evacuated and backfilled with argon. [l, l-bis(Diphenylphosphino)ferrocene]dichloropalladium(II), complex with

dichloromethane (32 mg, 0.039 mmol) was added to the suspension and the mixture was degassed by ultrasound irradiation under argon for 5 min. The mixture was heated to 80 °C and stirred at this temperature for 6.0 h. At this time, the reaction mixture was allowed to cool to rt while stirring for an additional 12 h. The reaction mixture was filtered through CELITE® and the filter cake was rinsed with EtOAc (80 mL). The combined filtrate and rinse were washed with 10% a 2 C03 (2x40 mL), water (40 mL) and sat'd NaCl (40 mL), dried ( a 2 S04) and evaporated. To a solution of the residue and imidazole (1 16 mg, 1.69 mmol) in CH 2 C1 2 (2.1 mL) at rt was added a solution of tert- butyldimethylsilyl chloride (132 mg, 0.85 mmol) in CH 2 C1 2 (1.1 mL) dropwise. The reaction mixture was stirred at rt for 14 h and then, diluted with EtOAc (80 mL) and washed with sat'd NaHCC^ (2x40 mL) and sat'd NaCl (40 mL). The organic layer was dried ( a 2 S04) and evaporated. The crude was chromatographed (Si0 2 , 95/5 to 9/1 Hex/Ether) to provide (4S,5R)-5-(((tert-Butyldimethylsilyl)oxy)methyl)-4-methyl-l- (4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)-3-(trif luoromethyl)-4,5-dihydro- lH-pyrazole (318 mg, 82% yield) as a colorless oil: LC-MS [M+H] 499.

[00426] A3C. 4-((4S,5R)-5-(((½rt-Butyldimethylsilyl)oxy)methyl)-4-methyl -3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-l-yl)phenol: To a stirred solution of (4S,5R)-5- (((tert-butyldimethylsilyl)oxy)methyl)-4-methyl- 1 -(4-(4,4,5,5-tetramethyl- 1,3,2- dioxaborolan-2-yl)phenyl)-3-(trifluoromethyl)-4,5-dihydro-lH -pyrazole (227 mg, 0.455 mmol) in EtOAc (2.0 mL) at rt was added 30% hydrogen peroxide (0.7 mL, 6.85 mmol) dropwise. The reaction mixture was stirred at rt for 21 h. After this time, the mixture was cooled to 0 °C and treated with 10% a 2 S203 (1 1 mL). The resulting aqueous mixture was stirred at rt for 2 h and then extracted with EtOAc (3x40 mL). The organic extracts were washed with sat'd NaCl (20 mL), dried ( a 2 S04) and concentrated.

Chromatography (Si0 2 , 7/3 Hex/EtOAc) of the crude residue afforded 4-((4S,5R)-5-

(((tert-butyldimethylsilyl)oxy)methyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H- pyrazol-l-yl)phenol (156 mg, 88% yield) as a white solid: LC-MS [M+H] 389.

[00427] A3D. 3-Bromo-4-((4S,5R)-5-(((½rt-butyldimethylsilyl)oxy)methyl)- 4-methyl- 3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-l-yl)phenol: To a stirred solution of 4- ((4S,5R)-5-(((ter?-butyldimethylsilyl)oxy)methyl)-4-methyl-3 -(trifluoromethyl)-4,5- dihydro-lH-pyrazol-l-yl)phenol (44.5 mg, 0.115 mmol) in CH 2 C1 2 (2.3 mL) at 0 °C was added pyridinium bromide perbromide (41 mg, 0.115 mmol) in three portions. The solution turned purple after portion addition and was stirred, until it turned back to clear, before addition of the next portion. After addition of the last portion, the reaction mixture was stirred until decoloration and then quenched with 10% a2S2C>3 (4.0 mL). The aqueous mixture was allowed to warm to rt and stirred for 30 min. The final mixture was partitioned between EtOAc (50 mL) and water (10 mL). The organic layer was washed with 5% NaHC0 3 (2x30 mL) and sat'd NaCl (20 mL), dried (Na 2 S0 4 ) and evaporated. To a solution of the residue in CH 2 Ci2 (0.5 mL) at rt was added a solution oi tert- butyldimethylsilyl chloride (32 mg, 0.206 mmol) in CH2CI2 (0.25 mL), dropwise. The reaction mixture was stirred at rt for 7.3 h. At this time, additional imidazole (28 mg, 0.407 mmol) and solution of tert-butyldimethylsilyl chloride (32 mg, 0.206 mmol) in dichloromethane (0.25 mL) were added and stirring at rt was continued for an additional 11 h. The mixture was diluted with EtOAc (50 mL) and washed with sat'd NaHCC^ (2x20 mL) and sat'd NaCl (20 mL). The organic layer was dried ( a 2 S0 4 ) and evaporated. To a solution of the resulting residue in DMF (0.45 mL) was added water (0.05 mL) followed by cesium carbonate (30 mg, 0.092 mmol). The resulting yellow solution was stirred at rt for 26 h. After this time, the solution was diluted with ether (40 mL) and washed with sat'd NaCl (20 mL). The organic layer was dried (Na 2 S0 4 ) and evaporated, and the crude was chromatographed (SiC^, 4/lHex/EtOAc) to give a mixture of isomeric aryl bromides (30 mg). This isomeric mixture was separated by

chromatography (Si0 2 , 96/4 CHC1 3 /Ether) to afford 3-bromo-4-((4S,5R)-5-(((tert- butyldimethylsilyl)oxy)methyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro- lH-pyrazol- 1 - yl)phenol (1 1 mg, 14.21% yield) as a colorless oil: LC-MS [M+H] 467, 469.

[00428] A3E. 4-((3R,4R)-4-(3-Bromo-4-((4S,5R)-5-(((tert- butyldimethylsilyl)oxy)methyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro- lH-pyrazol- 1 - yl)phenoxy)-3-methylpiperidin-l-yl)-5-chloro-2-methoxypyridi ne: To a stirred solution of 3-bromo-4-((4S,5R)-5-(((/er/-butyldimethylsilyl)oxy)methyl)- 4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-l-yl)phenol (10.2 mg, 0.022 mmol), (3R,4S)-1- (5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-ol (811, 1 1 mg, 0.042 mmol) and triphenylphosphine (13 mg, 0.049 mmol) in THF (0.2 mL) at rt was added <Xi-tert- butylazodicarboxylate (11 mg, 0.047 mmol). The resulting solution was stirred at rt for 59 h and then evaporated The residue was chromatographed (SiC^, 96/4 CHC^/Ether) to afford 4-((3R,4R)-4-(3-bromo-4-((4S,5R)-5-(((tert-butyldimethylsily l)oxy)methyl)-4- methyl-3 -(trifluoromethyl)-4,5 -dihydro- 1 H-pyrazol- 1 -yl)phenoxy)-3 -methylpiperidin- 1 - yl)-5-chloro-2-methoxypyridine (14 mg, 66% yield) as a colorless oil: LC-MS [M+H] 705, 707, 709.

[00429] A3F. ((4S,5R)-l-(2-Bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyridin -4-yl)- 3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluorometh yl)-4,5-dihydro-lH- pyrazol-5-yl)methanol: 4-((3R,4R)-4-(3-Bromo-4-((4S,5R)-5-(((tert- butyldimethylsilyl)oxy)methyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro- lH-pyrazol- 1 - yl)phenoxy)-3-methylpiperidin-l-yl)-5-chloro-2-methoxypyridi ne was dissolved in -1.1M HCl/MeOH, MeOAc solution [3.25 mL, prepared by addition of AcCl (0.25 mL) to MeOH (3.0 mL) at 0 °C and then stirring at rt for 30 min]. The resulting solution was allowed to stand at rt for 12 h. After this time, the solution was diluted with MeCN (4 mL) and evaporated. The residue was taken up in EtOAc (40 mL) and, washed with 5% aHC03 (2x20 mL) and sat'd NaCl (20 mL). The organic layer was dried ( a2S0 4 ) and concentrated. Chromatography (SiC^, 4/lto 7/3 Hex/EtOAc) of the crude afforded

((4S,5R)-l-(2-bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyri din-4-yl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)methanol (9 mg, 82% yield) as a colorless oil: LC-MS [M+H] 591, 593.

[00430] A3G. ((4S,5R)-l-(2-Bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyridin -4-yl)- 3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluorometh yl)-4,5-dihydro-lH- pyrazol-5-yl)methyl methanesulfonate: To a solution of ((4S,5R)-l-(2-bromo-4- (((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperid in-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)metha nol (9 mg, 0.016 mmol) and methanesulfonyl chloride (2 μϊ ^ , 0.026 mmol) in CH2CI2 (0.3 mL) at 0 °C was added triethylamine (5 μΐ, 0.036 mmol). The mixture was stirred for 30 min at 0 °C and for 3.0 h while warming to rt. Then, the mixture was diluted with EtOAc (30 mL) and washed with sat'd NaHC0 3 (2x15 mL) and sat'd NaCl (15 mL). The EtOAc solution was dried

( a2S0 4 ) and evaporated. Drying under vacuum gave ((4S,5R)-l-(2-bromo-4-(((3R,4R)- l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl)ox y)phenyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)methyl methanesulfonate (10 mg, 97% yield) as a yellowish oil: LC-MS [M+H] 669, 671.

[00431] A3H. 2-((4S,5S)-l-(2-Bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyrid in-4- yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoro methyl)-4,5-dihydro-lH- pyrazol-5-yl)acetonitrile: To a solution of ((4S,5R)-l-(2-bromo-4-(((3R,4R)-l-(5-chloro- 2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4- methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)methyl methanesulfonate (10 mg, 0.015 mmol) in DMSO (0.2 mL) was added potassium cyanide (2.0 mg, 0.030 mmol). The mixture was heated to 40 °C and stirred at this temperature under argon for 8.5 h. At this time, the mixture was cooled to rt and stirred for an additional 12 h. The mixture was diluted with EtOAc (40 mL) and, washed with sat'd NaHC0 3 (2x15 mL), water (2x15 mL) and sat'd NaCl (10 mL). The organic layer was dried (Na 2 S0 4 ) and concentrated. The crude was chromatographed (Si0 2 , 7/3 Hex/EtOAc) to give 2-((4S,5S)-l-(2-bromo- 4-(((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiper idin-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceto nitrile (7 mg, 78% yield) as a colorless oil: LC-MS [M+H] 600, 602.

[00432] A3I. Methyl 2-((4S,5S)-l-(2-bromo-4-(((3R,4R)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-me thyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetate: 2-((4S,5S)-l-(2-Bromo-4-(((3R,4R)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-me thyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (7 mg, 0.012 mmol) was dissolved in ~3M HCl/MeOH, CH 2 C1 2 , MeOAc solution [3.8 mL, prepared by addition of AcCl (0.8 mL) to a 3/2 CH 2 Cl 2 /MeOH solution (3.0 mL) at 0 °C and then stirring at rt for 20 min]. The resulting solution was allowed to stand at rt for 29 h. After this time, the solution was evaporated and the remaining oily material was stripped from MeOH (2x4 mL). The residue was dissolved in ~3M HCl/MeOH, MeOAc solution [3.8 mL, prepared by addition of AcCl (0.8 mL) to MeOH (3.0 mL) at 0 °C and then stirring at rt for 30 min]. The resulting solution was heated to 40 °C and allowed to stand at this temperature for 23.0 h. After this time, the solution was cooled to rt, diluted with MeCN (4 mL) and evaporated. The residue was taken up in EtOAc (40 mL) and, washed with sat'd aHC03 (2x35 mL) and sat'd NaCl (30 mL). The organic layer was dried ( a 2 S0 4 ) and concentrated. The crude was chromatographed (Si0 2 , 4/1 Hex/EtOAc) to afford methyl 2-((4S,5S)-l-(2-bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyrid in-4-yl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoromethyl )-4,5-dihydro-lH-pyrazol- 5-yl)acetate (7 mg, 89% yield) as a colorless oil: LC-MS [M+H] 633, 635. [00433] Example A3 : To a stirred solution of methyl 2-((4S,5S)-l-(2-bromo-4- (((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperid in-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceta te (6.5 mg, 10.25 μιηοΐ) in THF (0.5 mL) and water (0.04 mL) at rt was added 1.0M aqueous lithium hydroxide (0.04 mL, 0.040 mmol). The mixture was stirred at rt for 13.5 h and then partially evaporated to remove most of the THF. The remaining solution was partitioned between water (40 mL) and Hex (15 mL). The aqueous layer was acidified to pH 2 by dropwise addition of 1M HC1, and then extracted with CH2CI2 (3x30 mL). The combined organic extracts were dried ( a 2 S04) and concentrated. The residue was dried under vacuum to afford 2-((4S,5 S)- 1 -(2-bromo-4-(((3 R,4R)- 1 -(5 -chloro-2-methoxypyridin-4-yl)-3 - methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetic acid (6 mg, 89% yield) as a white solid: LC-MS [M+H] 619, 621. Ή NMR (400MHz, CD 2 C1 2 ) δ 7.96 (s, 1H), 7.21 (d, J= 8.8 Hz, 1H), 7.19 (d, J= 8.8 Hz, 1H), 6.91 (dd, J= 8.8, 2.8 Hz, 1H), 6.29 (s, 1H), 4.63 (m, 1H), 3.99 (dt, J= 4.1, 8.8 Hz, 1H), 3.86 (s, 3H), 3.52 (m, 2H), 3.27 (m, 1H), 2.89 (m, 1H), 2.66 (dd, J=12.2, 9.3 Hz, 1H), 2.41 (m, 2H), 2.18 (m, 2H), 1.79 (m, 1H), 1.41 (d, J= 7.1 Hz, 3H), 1.10 (d, J= 6.8 Hz, 3H).

Analytical HPLC (Zorbax method, 0% Solvent B start): RT = 8.69 min, HI: 97%.

hGPR40 EC50 = 844 nM. Example A4

2-((4S,5S)-l-(3-Bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyrid in-4-yl)-3- methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- -yl)acetic acid

[00434] A4A. 2-((4S,5S)-4-Methyl-l-(4-(4,4,5,5-tetramethyl-l,3,2-dioxabor olan-2- yl)phenyl)-3-(trifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetonitrile: A flask containing a suspension of 2-((4S,5S)-l-(4-bromophenyl)-4-methyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (II, 645 mg, 1.863 mmol), bis(pinacolato)diboron (560 mg, 2.183 mmol) and potassium acetate (573 mg, 5.78 mmol) in DMF (3.4 mL) was evacuated and backfilled with argon. [1, 1- Bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (76 mg, 0.093 mmol) was added to the suspension and the mixture was degassed by ultrasound irradiation under argon for 5 min. The mixture was heated to 78 °C and stirred at this temperature for 10.8 h. After this time, the reaction mixture was filtered through CELITE® and the filter cake was rinsed with EtOAc (130 mL). The combined filtrate and rinse were washed with 10% Na 2 C0 3 (50 mL), sat'd NaHC0 3 (70 mL), water (70 mL) and sat'd NaCl (50 mL). The organic layer was dried ( a 2 S04) and evaporated. The crude was chromatographed (Si<¾, 4/1 to 7/3 Hex/EtOAc) to provide 2-((4S,5S)-4- methyl-l-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phe nyl)-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (642 mg, 88% yield) as a colorless oil: LC-MS [M+H] 394.

[00435] A4B. 2-((4S,5S)-l-(4-Hydroxyphenyl)-4-methyl-3-(trifluoromethyl)- 4,5- dihydro-lH-pyrazol-5-yl)acetonitrile: To a stirred solution of the 2-((4S,5S)-4-methyl-l- (4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)-3-(t rifluoromethyl)-4,5-dihydro- lH-pyrazol-5-yl)acetonitrile (572 mg, 1.455 mmol) in EtOAc (6.4 mL) at rt was added 30% hydrogen peroxide (2.3 mL, 22.52 mmol) dropwise. The reaction mixture was stirred at rt for 22 h. After this time, the mixture was cooled to 0 °C and treated with 10% a2S2C>3 (35 mL). The resulting aqueous mixture was stirred at rt for 2 h and then extracted with EtOAc (3x50 mL). The organic extracts were washed with sat'd NaCl (40 mL), dried ( a 2 S04) and concentrated. Chromatography (SiC^, 3/2 Hex/EtOAc) of the crude afforded 2-((4S,5S)-l-(4-hydroxyphenyl)-4-methyl-3-(trifluoromethyl)- 4,5- dihydro-lH-pyrazol-5-yl)acetonitrile (391 mg, 94% yield) as a yellowish oil: LC-MS [M+H] 284.

[00436] A4C. 2-((4S,5S)-l-(3-Bromo-4-(((3R,4R)-l-(5-chloro-2-methoxypyrid in-4- yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3-(trifluoro methyl)-4,5-dihydro-lH- pyrazol-5-yl)acetonitrile: To a stirred solution of the 2-((4S,5S)-l-(4-hydroxyphenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceto nitrile (179 mg, 0.489 mmol), (3R,4S)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperidin -4-ol (811, 228 mg, 0.881 mmol) and triphenylphosphine (262 mg, 0.988 mmol) in THF (2.9 mL) at rt was added di-tert-butylazodicarboxylate (231 mg, 0.984 mmol). The resulting solution was stirred at rt for 24 h and then evaporated. The residue was chromatographed (Si0 2

230-400 mesh, 96/4 CHC1 3 /Ether) to afford 2-((4S,5S)-l-(3-bromo-4-(((3R,4R)-l-(5- chloro-2-methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phe nyl)-4-methyl-3- (trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (257 mg, 87% yield) as a colorless oil: LC-MS [M+H] 600, 602.

[00437] A4D. Methyl 2-((4S,5S)-l-(3-bromo-4-(((3R,4R)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-me thyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetate: 2-((4S,5S)-l-(3-Bromo-4-(((3R,4R)-l-(5-chloro-2- methoxypyridin-4-yl)-3-methylpiperidin-4-yl)oxy)phenyl)-4-me thyl-3-(trifluoromethyl)- 4,5-dihydro-lH-pyrazol-5-yl)acetonitrile (258 mg, 0.425 mmol) was dissolved in ~3M HCl/MeOH, CH 2 C1 2 , MeOAc solution [12.6 mL, prepared by addition of AcCl (2.6 mL) to a 3/2 CH 2 Cl 2 /MeOH solution (10.0 mL) at 0 °C and then stirring at rt for 20 min]. The resulting solution was allowed to stand at rt for 18 h. The solution was evaporated and the remaining oily material was stripped from MeOH (2x10 mL). The residue was dissolved in ~3M HCl/MeOH, MeOAc solution [12.6 mL, prepared by addition of AcCl (2.6 mL) to MeOH (10.0 mL) at 0 °C and then stirring at rt for 30 min]. The resulting solution was heated to 40 °C and allowed to stand at this temperature for 23.0 h. After this time, the solution was cooled to rt, diluted with MeCN (10 mL) and evaporated. The residue was taken up in EtOAc (60 mL) and, washed with sat'd NaHCC^ (2x35 mL) and sat'd NaCl

(30 mL). The organic layer was dried (Na 2 S0 4 ) and concentrated. The crude was chromatographed (Si0 2 , 4/1 Hex/EtOAc) to afford methyl 2-((4S,5S)-l-(3-bromo-4- (((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperid in-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceta te: 2-((4S,5S)-l-(3-Bromo- 4-(((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiper idin-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceto nitrile (247 mg, 92% yield) as a white solid: LC-MS [M+H] 633, 635.

[00438] Example A4: To a stirred solution of methyl 2-((4S,5S)-l-(3-bromo-4- (((3R,4R)-l-(5-chloro-2-methoxypyridin-4-yl)-3-methylpiperid in-4-yl)oxy)phenyl)-4- methyl-3-(trifluoromethyl)-4,5-dihydro-lH-pyrazol-5-yl)aceta te (44 mg, 0.069 mmol) in THF (2.0 mL) and water (0.2 mL) at rt was added l.OM aqueous lithium hydroxide (0.2 mL, 0.200 mmol). After stirring at rt for 4.5 h, the reaction mixture was partially evaporated to remove most of the THF. The remaining solution was partitioned between water (50 mL) and Hex (15 mL). The aqueous layer was acidified to pH 2 by dropwise addition of 1M HC1, and then extracted with CH2CI2 (3x30 mL). The combined organic extracts were dried ( a 2 S04) and concentrated. The residue was dried under vacuum to afford 2-((4S,5 S)-l-(3 -bromo-4-(((3 R,4R)- 1 -(5 -chloro-2-methoxypyridin-4-yl)-3 - methylpiperidin-4-yl)oxy)phenyl)-4-methyl-3 -(trifluoromethyl)-4,5-dihydro- 1 H-pyrazol- 5-yl)acetic acid (43 mg, 100% yield) as a white solid: LC-MS [M+H] 619, 621. Ή NMR (500MHz, CD 2 C1 2 ) δ 7.97 (s, 1H), 7.36 (d, J= 2.8 Hz, 1H), 7.02 (dd, J= 9.0, 2.8 Hz, 1H), 6.95 (d, J= 9.0 Hz, 1H), 6.30 (s, 1H), 4.41 (m, 1H), 4.02 (dt, J= 3.9, 8.3 Hz, 1H), 3.86 (s, 3H), 3.54 (m, 2H), 3.28 (m, 1H), 2.90 (m, 1H), 2.84 (dd, J=16.5, 3.1 Hz, 1H), 2.18 (m, 2H), 1.86 (m, 2H), 1.35 (d, J= 7.1 Hz, 3H), 1.17 (d, J= 6.7 Hz, 3H). Analytical HPLC (Zorbax method, 50% Solvent B start): RT = 8.55 min, HI: 100%. hGPR40 EC 50 = 45 nM.