Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL GLP-1 RECEPTOR MODULATORS
Document Type and Number:
WIPO Patent Application WO/2013/090454
Kind Code:
A2
Abstract:
The invention relates to compounds that modulate the glucagon-like peptide 1 (GLP-1) receptor, methods of their synthesis, and methods of their therapeutic and/or prophylactic use. Such compounds are act as modulators or potentiators of GLP-1 receptor on their own, or with receptor ligands including GLP-1 peptides GLP-1(7-36) and GLP-1(9-36), or with peptide-based therapies, such as exenatide and liraglutide, and have the following general structure (where"﹋" represents either or both the R and S form of the compound): where A, B, C, Y1, Y2, Z, R1, R2, R3, R4, R5, W1, n, p and q are as defined herein.

Inventors:
BOEHM MARCUS F (US)
MARTINBOROUGH ESTHER (US)
MOORJANI MANISHA (US)
TAMIYA JUNKO (US)
HUANG LIMING (US)
YEAGER ADAM R (US)
BRAHMACHARY ENUGURTHI (US)
FOWLER THOMAS (GB)
NOVAK ANDREW (GB)
MEGHANI PREMJI (GB)
KNAGGS MICHAEL (GB)
Application Number:
PCT/US2012/069289
Publication Date:
June 20, 2013
Filing Date:
December 12, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
RECEPTOS INC (US)
International Classes:
A61K31/433; C07D211/22; A61P3/10; C07D233/54; C07D233/60; C07D239/26; C07D239/34; C07D241/08; C07D255/02; C07D263/32; C07D271/06; C07D271/107; C07D277/24; C07D285/12; C07D401/04; C07D403/04; C07D409/12; C07D413/04; C07D417/12; C07D495/04
Domestic Patent References:
WO2011094890A12011-08-11
WO1999010312A11999-03-04
WO2005077915A12005-08-25
WO2011097300A12011-08-11
Foreign References:
US5424286A1995-06-13
US6902744B12005-06-07
US7297761B22007-11-20
US7368427B12008-05-06
Other References:
KIEFFER T. J.; HABENER, J. F., ENDOCRIN. REV., vol. 20, 1999, pages 876 - 913
DRUCKER, D. J., ENDOCRINOLOGY, vol. 142, 2001, pages 521 - 7
HOLST, J. J., DIABETES METAB. RES. REV., vol. 18, 2002, pages 430 - 41
DRUCKER, D. J., ENDOCRINOLOGY, vol. 144, 2003, pages 5145 - 8
HOLZ, G. G.; CHEPURNY O. G., CURR. MED. CHEM., vol. 10, 2003, pages 2471 - 83
LIST, J. F.; HABENER, J. F., AM. J. PHYSIOL. ENDOCRINOL. METAB., vol. 286, 2004, pages E875 - 81
LIT: "Salt Selection for Basic Drugs", INTJ. PHARM., vol. 33, 1986, pages 201 - 217
ELBROND ET AL., DIABETES CARE., vol. 25, no. 8, August 2002 (2002-08-01), pages 1398404
AGERSO ET AL., DIABETOLOGIA, vol. 45, no. 2, February 2002 (2002-02-01), pages 195 - 202
RCMINGTON: "The Science and Practice of Pharmacy", 1995
"Design ofProdrugs", 1985, ELSEVIER
DUEZ, ENDOCRINOLOGY, vol. 150, no. 1, January 2009 (2009-01-01), pages 56 - 62
PEDERSON, DIABETES, vol. 47, August 1998 (1998-08-01), pages 1253 - 1258
Attorney, Agent or Firm:
HERMANNS, Karl, R. et al. (Suite 5400701 Fifth Avenu, Seattle Washington, US)
Download PDF:
Claims:
CLAIMS

We claim:

1. A compound having the structure of Formula I-R or I-S or a pharmaceutically acceptable isomer, enantiomer, racemate, salt, ester, prodrug, hydrate or solvate thereof:

I-R

I-S wherein

A is a 5-, 6- or 7-membered heterocyclyl having one, two or three heteroatoms where each such heteroatom is independently selected from O, N, and S, and where any ring atom of such heterocyclyl may be optionally substituted with one or more of R4;

B is aryl, aralkyl, heterocyclyl, or heterocyclylalkyl; C is aryl, arylalkyl, heterocyclyl or heterocyclylalkyl;

Yi and Y2 are both null, or one of Yi or Y2 is -NH- or -O- and the other Yi or Y2 is null;

Z is -C(O)- or -S(0)2-;

each Ri is independently H or Ci_4 alkyl;

R2 is -OH, -0-R8, -N(Ri)-S02-R8, -NR4iR42, -N(Ri)-(CRaRb)m-COOH, -NCRiXCRaRb -CO-NCR -heterocyclyl, -N(R1)-(CRaRb)m-CO-N(R1)(R7), or -N(Ri)- heterocyclyl;

each R3 and R4 is independently H, halo, alkyl, alkyl substituted with R3i, alkoxy, haloalkyl, perhaloalkyl, haloalkoxy, perhaloalkoxy, aryl, heterocyclyl, -OH, -OR8, -CN, -NO2, -NRiRg, -C(0)R8, -C(0)NRiR8, -NRiC(0)R8, -SR8, -S(0)R8, -S(0)2R8, -OS(0)2R8, -S(0)2NR!R8, -NR!S(0)2R8, -(CRaRb)mNRiR8,

-(CRaRb)mO(CRaRb)mR8, -(CRaRb)mNRi(CRaRb)mR8 or -(CRaRb)mNRi(CRaRb)mCOOH; or any two R3 or R4 groups on the same carbon atom taken together form oxo

each R3i is independently H, halo, hydroxyl, -NR4iR42, or alkoxy;

each R40 is independently H or alkyl;

each R4i and R42 is independently R40 or -(CH2)n-COO-R4o, -C(0)-R4o, aryl, heteroaryl, or two taken together with the N atom to which they are attached can form a 3- to 7-membered heterocyclyl;

Wi is null or -Li-(CRaRb)m-Li-R6;

each Li is independently, from the proximal to distal end of the structure of Formula I-R or I-S, null, -C(0)0-, -S(02)-, -S-, -N(Ri)-C(0)-N(Ri)-, -N(Ri)-C(0)- 0-, -C(O)- or -S(02)-NRi-;

each Ra and Rb is independently H, alkyl, alkoxy, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl, any of which alkyl, alkoxy, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl may be optionally (singly or multiply) substituted with R7, or -(CH2)mC(0)OR4o, -(CH^OR^, -(CH2)mSR40, -(CH2)mNR4iR42, - (CH2)mC(0)NR4iR42; or any two Ra and Rb taken together with the carbon to which they are attached form a cycloalkyl or heterocyclyl; or Ri and any one of Ra or Rb taken together form heterocyclyl; R5 is Ry, -(CH2)m-L2-(CH2)m-R7, or -(-L3-(CRaRb)r-)s-L3-Ry;

Re is H, alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, heterocycloalkyl, any of which may be optionally singly or multiply substituted with Ry or -(CH2)m-L2-(CH2)m-R7;

Ry is H, halo, alkyl, haloalkyl, perhaloalkyl, alkoxy, -OH, -ORg, -CN, -NRiRg, -(CRaRb)mO(CRaRb)mRg, -NRi(CRaRb)mR8, -C(0)R8, -NRi(CRaRb)mCOOH, -NRiC(0)R8, -C(0)NRiR8, -SR8, -S(0)R8, -S(0)2R8, -S(0)2NRiR8, -NRiS(0)2R8; or a ring moiety selected from cycloalkyl, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl, where such ring moiety is optionally singly or multiply substituted with halo, -OH, - CN, alkyl, alkoxy, haloalkyl or perhaloalkyl;

each R8 is independently H, alkyl, cycloalkyl or aryl;

L2 is independently, from the proximal to distal end of the structure of Formula I-R or I-S, null, -0-, -OC(O)-, -NRi- , -C(0)NRi-, -N(Ri)-C(0)-, -S(02)-, - C(O)- or -S(02)-N(R!)-;

each L3 is independently null, -0-, or -N(Ri)- each m is independently 0, 1, 2, 3, 4, 5 or 6;

each n is independently 0 or 1 or 2;

p is 0, 1, 2 or 3;

q is 0, 1, 2 or 3;

each r is independently 2, 3, or 4; and

each s is independently 1, 2, 3, or 4.

2. The compound of claim 1 wherein Yi and Y2 are null, Z is - C(O)- and A is a 5- or 6-membered heteroaryl group.

3. The compound of claim 2 wherein the the compound has the following structure:

258

I-R/S (18),

262

4. The compound of claim 1 wherein Yi and Y2 are null, Z is - C(O)- and A is a 5- or 6-membered non-aromatic heterocycyly group.

6. The compound of claim 1 wherein Yi and Y2 are null, Z is - C(O)- and C is aryl.

8. The compound of claim 1 wherein Yi and Y2 are null, Z is - C(O)- and C is heterocyclyl.

9. The compound of claim 8 wherein the the compound has the following structure:

I-R/S (41),

I-R/S (44).

10. The compound of claim 1 wherein Yi and Y2 are null, Z is - C(O)- and B is aryl or arylalkyl.

11. The compound of claim 10 wherein the the compound has the following structure:

12. The compound of claim 1 wherein Yi and Y2 are null, Z is C(O)- and B is heterocyclyl or heterocyclylalkyl.

13. The compound of claim 12 wherein the the compound has the following structure:

271

The compound of claim 1 wherein Yi and Y2 are null, Z is -

15. The compound of claim 14 wherein the the compound has the following structure:

16. The compound of claim 1 wherein where Yi is null, Y2 is -O- and Z is -C(O)-.

17. The compound of claim 16 wherein the the compound has the following structure:

18. The compound of claim 1 wherein where Yi is NH, Y2 is null and Z is -C(O)-.

19. The compound of claim 18 wherein the the compound has the following structure:

20. A pharmaceutical composition comprising a compound of claim 1 together with at least one pharmaceutically acceptable carrier, diluent or excipient.

21. A pharmaceutical combination comprising the compound of claim 1 and a second medicament.

22. The pharmaceutical combination of claim 21 wherein the second medicament is an agonist or modulator for glucagon receptor, GIP receptor, GLP-2 receptor, or PTH receptor, or glucagon-like peptide 1 (GLP-1) receptor.

23. The pharmaceutical combination of claim 21 wherein the second medicament is exenatide, liraglutide, taspoglutide, albiglutide, or lixisenatide.

24. The pharmaceutical combination of claim 21 wherein the second medicament is a DPPIV inhibitor.

25. A method of activation, potentiation, modulation or agonism of a glucagon-like peptide 1 receptor comprising contacting the receptor with an effective amount of a compound of claim 1 or a pharmaceutical composition of claim 20 or a pharmaceutical combination of claim 22.

26. A method of activation, potentiation, modulation or agonism of a glucagon-like peptide 1 (GLP-1) receptor in a subject in need thereof, said method comprising administering to said subject a compound of claim 1 or a pharmaceutical composition of claim 20 or a pharmaceutical combination of claim 22.

27. A method of treatment of a malcondition in a patient for which activation, potentiation, modulation or agonism of a glucagon-like peptide 1 receptor is medically indicated, comprising administering an effective amount of the compound of claim 1 to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient.

28. The method of claim 27 wherein the malcondition is type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety or metabolic disorder.

29. The method of claim 27 wherein the malcondition is type I diabetes or type II diabetes.

30. A compound of claims 1-19 or a pharmaceutical composition of claim 20 for use as a medicament.

31. A compound of claims 1 - 19 or a pharmaceutical composition of claim 20 for the treatment of a malcondition in a patient for which activation, potentiation, modulation or agonism of a glucagon-like peptide 1 receptor is medically indicated.

32. The compound or composition of claim 31 where the malcondition is type I diabetes, type II diabetes, gestational diabetes, obesity, appetite, insufficient satiety or metabolic disorder.

33. The compound or composition of claim 31 where the malcondition is type I diabetes or type II diabetes.

Description:
NOVEL GLP-1 RECEPTOR MODULATORS

STATEMENT REGARDING SEQUENCE LISTING

[0001] The Sequence Listing associated with this application is provided in text format in lieu of a paper copy, and is hereby incorporated by reference into the specification. The name of the text file containing the Sequence Listing

800059_407WO_SEQUENCE_LISTING.txt. The text file is about 1 KB, was created on December 12, 2012, and is being submitted electronically via EFS-Web.

FIELD OF THE INVENTION

[0002] The invention relates to compounds that bind the glucagon-like peptide 1 (GLP-1) receptor, methods of their synthesis, and methods of their therapeutic and/or prophylactic use. The present invention is directed to compounds adapted to act as modulators or potentiators of GLP-1 receptor, including peptides GLP-1 (7-36) and GLP-l(9-36), as well as peptide-based therapies such as exenatide and liraglutide.

BACKGROUND

[0003] Glucagon-like peptide 1 receptor (GLP-1R) belongs to Family Bl of the seven-transmembrane G protein-coupled receptors, and its natural agonist ligand is the peptide hormone glucagon-like peptide- 1 (GLP-1). GLP-1 is a peptide hormone arising by its alternative enzymatic cleavage from proglucagon, the prohormone precursor for GLP-1, which is highly expressed in enteroendocrine cells of the intestine, the alpha cells of the endocrine pancreas (islets of Langerhans), and the brain (Kieffer T. J. and Habener, J. F. Endocrin. Rev. 20:876-913 (1999); Drucker, D. J., Endocrinology 142:521-7 (2001); Hoist, J. J., Diabetes Metab. Res. Rev. 18:430-41 (2002)). The initial actions of GLP-1 observed were on the insulin-producing cells of the islets, where it stimulates glucose-dependent insulin secretion. Subsequently, multiple additional antidiabetogenic actions of GLP-1 were discovered including the stimulation of the growth and inhibition of the apoptosis of pancreatic beta cells (Drucker, D. J.,

Endocrinology 144:5145-8 (2003); Holz, G. G. and Chepurny O. G., Curr. Med. Chem. 10:2471-83 (2003); List, J. F. and Habener, J. F., Am. J. Physiol. Endocrinol. Metab. 286:E875-81 (2004)).

[0004] On activation, GLP-1 receptors couple to the a-subunit of G protein, with subsequent activation of adenylate cyclase and increase of cAMP levels, thereby potentiating glucose-stimulated insulin secretion. Therefore, GLP-1 is an attractive therapeutic target to lower blood glucose and preserve the ?-cells of the pancreas of diabetic patients. Glucagon has been used for decades in medical practice within diabetes and several glucagon-like peptides are being developed for various therapeutic indications. GLP-1 analogs and derivatives are being developed for the treatment for patients suffering from diabetes.

SUMMARY OF THE INVENTION

[0005] The present invention is directed to compounds adapted to act as potentiators or modulators of GLP-1 receptor; methods of their preparation and methods of their use, such as in treatment of a malcondition mediated by GLP-1 receptor activation, or when modulation or potentiation of GLP-1 receptor is medically indicated.

[0006] Certain embodiments of the present invention comprise a compound having the structure of Formula I-R or I-S or a pharmaceutically acceptable isomer, enantiomer, racemate, salt, isotope, prodrug, hydrate or solvate thereof:

I-R

I-S wherein

A is a 5-, 6- or 7-membered heterocyclyl having one, two or three heteroatoms where each such heteroatom is independently selected from O, N, and S, and where any ring atom of such heterocyclyl may be optionally substituted with one or more of R 4 ;

B is aryl, aralkyl, heterocyclyl, or heterocyclylalkyl;

C is aryl, arylalkyl, heterocyclyl or heterocyclylalkyl;

Yi and Y 2 are both null, or one of Yi or Y 2 is -NH- or -O- and the other Yi or Y 2 is null;

Z is -C(O)- or -S(0) 2 -;

each Ri is independently H or Ci_ 4 alkyl;

R 2 is -OH, -0-R 8 , -N(Ri)-S0 2 -R 8 , -NR 4 iR 42 , -N(Ri)-(CR a R b ) m -COOH, -N(Ri)-(CRaR b ) m -CO-N(Ri)-heterocyclyl, -N(Ri)-(CR a R b ) m -CO-N(Ri)(R 7 ), or -N(Ri)- heterocyclyl;

each R 3 and R 4 is independently H, halo, alkyl, alkyl substituted with R 31j alkoxy, haloalkyl, perhaloalkyl, haloalkoxy, perhaloalkoxy, aryl, heterocyclyl, -OH, -OR 8 , -CN, -N0 2 , -NRiRg, , -C(0)R 8 , -C(0)NRiR 8 , -NRiC(0)R 8 , -SR 8 , -S(0)R 8 , -S(0) 2 R 8 , -OS(0) 2 R 8 , -S(0) 2 NRiR 8 , -NRiS(0) 2 R 8 , -(CR a R b ) m NRiR 8 ,

-(CR a R b ) m O(CR a R b ) m R 8 , -(CR a R b ) m NRi (CR a R b ) m R 8 or -(CR a R b ) m NRi(CR a R b ) m COOH; or any two R 3 or R 4 groups on the same carbon atom taken together form oxo;

each R 3 i is independently H, halo, hydroxyl, -NR 4 iR 42 , or alkoxy;

each R 40 is independently H or alkyl; each R 4 i and R42 is independently R40 or -(CH 2 ) n -COO-R4o, -C(0)-R4o, aryl, heteroaryl, or two taken together with the N atom to which they are attached can form a 3- to 7-membered heterocyclyl;

Wi is null or -Li-(CR a Rb)m-Li-R 6 ;

each Li is independently, from the proximal to distal end of the structure of Formula I-R or I-S, null, -C(0)0-, -S(0 2 )-, -S-, -N(Ri)-C(0)-N(Ri)-, -N(Ri)-C(0)- 0-, -C(O)- or -S(0 2 )-NR ;

each R a and R b is independently H, alkyl, alkoxy, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl, any of which alkyl, alkoxy, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl may be optionally (singly or multiply) substituted with R 7 , or -(CH 2 ) m C(0)OR4o, -(CH 2 ) m OR4o, -(CH 2 ) m SR 40 , -(CH 2 ) m NR 4 iR4 2 ,

-(CH 2 ) m C(0)NR 4 iR 42 ; or any two R a and R b taken together with the carbon to which they are attached form a cycloalkyl or heterocyclyl; or Ri and any one of Ra or Rb taken together form heterocyclyl;

R 5 is R 7 , -(CH 2 ) m -L 2 -(CH 2 ) m -R 7 , or -(-L 3 -(CR a Rb) r -)s-L 3 -R 7 ;

Re is H, alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, heterocycloalkyl, any of which may be optionally singly or multiply substituted with R 7 or -(CH 2 ) m -L 2 -(CH 2 ) m -R 7 ;

R 7 is H, halo, alkyl, haloalkyl, perhaloalkyl, alkoxy, -OH, -OR 8 , -CN, -NRiRg, -(CR a Rb) m O(CRaRb) m R8, -NRi(CR a R b ) m R 8 , -C(0)R 8 , -NRi(CR a R b ) m COOH, -NRiC(0)R 8 , -C(0)NRiR 8 , -SR 8 , -S(0)R 8 , -S(0) 2 R 8 , -S(0) 2 NRiR 8 , -NRiS(0) 2 R 8 ; or a ring moiety selected from cycloalkyl, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl, where such ring moiety is optionally singly or multiply substituted with halo, -OH, - CN, alkyl, alkoxy, haloalkyl or perhaloalkyl;

each R 8 is independently H, alkyl, cycloalkyl or aryl;

L 2 is independently, from the proximal to distal end of the structure of Formula I-R or I-S, null, -0-, -OC(O)-, -NRi- , -C(0)NRi-, -N(Ri)-C(0)-, -S(0 2 )-, - C(O)- or -S(0 2 )-N(Ri)-;

each L is independently null, -0-, or -N(Ri)- each m is independently 0, 1, 2, 3, 4, 5 or 6; each n is independently 0 or 1 or 2;

p is 0, 1, 2 or 3;

q is 0, 1, 2 or 3;

each r is independently 2, 3, or 4; and

each s is independently 1, 2, 3, or 4.

[0007] In certain embodiments, a pharmaceutical composition comprising a compound of the invention together with at least one pharmaceutically acceptable carrier, diluent or excipient is provided.

[0008] In certain embodiments, a method of use of a compound of the invention comprising preparation of a medicament is provided.

[0009] In certain embodiments, the invention provides a pharmaceutical combination comprising a compound of the invention and a second medicament. In various such embodiments, the second medicament is an agonist or modulator for glucagon receptor, GIP receptor, GLP-2 receptor, or PTH receptor, or glucagon-like peptide 1 (GLP-1) receptor. In various such embodiments, the second medicament is exenatide, liraglutide, taspoglutide, albiglutide, or lixisenatide or other insulin regulating peptide. In various such embodiments, the second medicament is a DPPIV inhibitor. In various such embodiments, the second medicament is medically indicated for the treatment of type II diabetes.

[0010] In certain embodiments, a method of activation, potentiation or agonism of a GLP-1 receptor is provided comprising contacting the receptor with a compound, pharmaceutical composition or pharmaceutical combination of the invention.

[0011] In certain embodiments, a method is provided for treatment of a malcondition in a subject for which activation, potentiation or agonism of a GLP-1 receptor is medically indicated where such method comprises administering to such subject a compound, pharmaceutical composition or pharmaceutical combination of the invention. In various such embodiments, selective activation, potentiation or agonism of a GLP-1 receptor, is medically indicated. In various such embodiments, the malcondition comprises type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety, or metabolic disorder. [0012] In certain embodiments, the invention provides methods for synthesis of certain compounds including compounds of the invention. In certain other

embodiments, the invention provides certain intermediate compounds associated with such methods of synthesis.

DETAILED DESCRIPTION OF THE INVENTION

[0013] Certain embodiments comprise a compound having the chiral structure of Formula I-R or I-S (with the chirality as indicated) or a pharmaceutically acceptable isomer, enantiomer, racemate, salt, isotope, prodrug, hydrate or solvate thereof:

[0014] Certain embodiments of the present invention comprise a compound having the structure of Formula I-R or I-S or a pharmaceutically acceptable isomer, enantiomer, racemate, salt, isotope, prodrug, hydrate or solvate thereof:

I-S where A, B, C, Yi, Y 2 , Z, R ls R 2 , R 3 , R4, R5, Wi, n, p and q are as defined above. [0015] In certain embodiments, the compounds have the structure of Formula I- R or a pharmaceutically acceptable isomer, enantiomer, salt, isotope, prodrug, hydrate or solvate thereof. In other embodiments, the compounds have the structure of Formula I-S or a pharmaceutically acceptable isomer, enantiomer, salt, isotope, prodrug, hydrate or solvate thereof.

[0016] In certain embodiments, the compounds can be substantially

enantiomerically pure.

[0017] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi and Y 2 are null, Z is -C(O)- and A is a 5- or 6- membered heteroaryl group. Representative compounds of this embodiment include compounds of the following structures (wherein " ΛΛΛΛ ν " represents either or both the R and S form of the compound):

I-R/S (21)

I-R/S (22)

[0018] In certain embodiments, the invention provides a compound where Yi and Y 2 are null, Z is -C(O)- and A is a 5-, 6- or 7-membered non-aromatic heterocyclyl group. Representative compounds of this embodiment include compounds of the following structures (wherein " ΛΛΛΛ ν " represents either or both the R and S form of the compound):

I-R/S (23)

I-R/S (25)

[0019] In certain embodiments, the inventions provides compounds of each of structures I-R/S(l)-(29) where R 4 of the phenyl group is H.

[0020] In certain embodiments, the inventions provides compounds of each of structures I-R/S(l)-(29) where the A group (i.e., the 5-, 6- or 7-membered heterocyclyl) is not substituted by R4, or substituted by R4 where R4 is alkyl, haloalkyl, alkoxy, -NR41R42 where R41 and R 42 are independently hydrogen or alkyl, or substituted by two R4 groups which taken together form oxo.

[0021] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi and Y 2 are null, Z is -C(O)- and A is C is aryl. Representative compounds of this embodiment include compounds of the following structures (wherein " /ww " represents either or both the R and S form of the compound):

[0022] In certain embodiments, the inventions provides compounds of each of structures I-R/S(30)-(32) where q is zero.

[0023] In certain embodiments, the inventions provides compounds of each of structures I-R/S(30)-(32) where q is one, two or three.

[0024] In certain embodiments, the inventions provides compounds of structure I-R/S(30) where q is one and R 5 is -(CH 2 ) m -L 2 -(CH 2 ) m -R7 or -(-L 3 -(CR a Rb)r-)s-L 3 -R7. Representative compounds of this embodiment include compounds of the following structure (wherein " " represents either or both the R and S form of the compound):

R/S (33)

[0025] In certain embodiments, the inventions provides compounds of structure I-R/S(33) where R 7 is H or alklyl and L 2 is O. Representative compounds of this embodiment include compounds of the following structure (wherein " ΛΛΛΛ " represents either or both the R and S form of the compound):

[0026] In certain embodiments, the inventions provides compounds of structure I-R/S(30) where R 5 is R 7 . Representative compounds of this embodiment include compounds of the following structure (wherein " " represents either or both the R and S form of the compound):

I-R/S (35)

[0027] In certain embodiments, the invention provides compounds of structure I-R/S(35) where R 7 is halo, alkyl, haloalkyl, perhaloalkyl, alkoxy, -OH, -ORg, -CN, - RiRg, -(CR a Rb)mO(CR a Rb) m R8, -NRi(CR a Rb) m R8, -C(0)R 8 , -NRi(CR a Rb) m COOH, -NRiC(0)R 8 , -C(0)NRiR 8 , -SR 8 , -S(0)R 8 , -S(0) 2 R 8 , -S(0) 2 NRiR 8 or -NRiS(0) 2 R 8 .

[0028] In certain embodiments, the invention provides compounds of structure I-R/S(35) where R 7 is a ring moiety selected from cycloalkyl, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl, where such ring moiety is optionally (singly or multiply) substituted with halo, -OH, -CN, alkyl, alkoxy, haloalkyl or perhaloalkyl.

[0029] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi and Y 2 are null, Z is -C(O)- and A is C is

heterocyclyl. Representative compounds of this embodiment include compounds of the following structures (wherein " ww " represents either or both the R and S form of the compound):

[0030] In certain embodiments, the invention provides compounds of each of structures I-R/S(36)-(44) where R 7 is halo, alkyl, haloalkyl, perhaloalkyl, alkoxy, -OH, -OR 8 , -CN, -NRiRg, -(CRaRb)mO(CRaRb)mR8, -NRi(CRaRb) m R8, -C(0)R 8 ,

-NRi(CR a R b ) m COOH, -NRiC(0)R 8 , -C(0)NRiR 8 , -SR 8 , -S(0)R 8 , -S(0) 2 R 8 ,

-S(0) 2 NRiR 8 or -NRiS(0) 2 R 8 .

[0031] In certain embodiments, the invention provides compounds of each of structures I-R/S(36)-(44) where R 7 is a ring moiety selected from cycloalkyl, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl, where such ring moiety is optionally (singly or multiply) substituted with halo, -OH, -CN, alkyl, alkoxy, haloalkyl or perhaloalkyl.

[0032] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi and Y 2 are null, Z is -C(O)- and B is aryl or arylalkyl. Representative compounds of this embodiment include compounds of the following structures (wherein " ' ww " represents either or both the R and S form of the compound):



[0033] In certain embodiments, the invention provides compounds of each of structures I-R/S (45)-(48) where Wi is null.

[0034] Representative compounds of this embodiment include compounds of the following structure (wherein " ' ww " represents either or both the R and S form of the compound):

[0035] In certain embodiments, the invention provides compounds of structure I-R/S(49) where R 3 is halo, alkyl, alkoxy, haloalkyl, perhaloalkyl, haloalkoxy, perhaloalkoxy, -OH, -OR 8 , -CN, -NRiR 8 , , _C(0)R 8 , -C(0)NRiR 8 , -NRiC(0)R 8 , -SR 8 , -S(0)R 8 , -S(0) 2 R 8 , -OS(0) 2 R 8 , -S(0) 2 NRiR 8 , -NRiS(0) 2 R 8 , -(CR a R b ) m NRiR 8 or -(CRaRb) m O(CR a R b ) m R 8 .

[0036] In certain embodiments, the invention provides compounds of each of structures I-R/S (45)-(49) where R 3 is alkyl.

[0037] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi and Y 2 are null, Z is -C(O)- and B is heterocyclyl or heterocyclylalkyl. Representative compounds of this embodiment include compounds of the following structures (wherein " , ww " represents either or both the R and S form of the compound):

[0038] In certain embodiments, the invention provides compounds of each of structures I-R/S(50)-(62) where Wi is null.

[0039] In certain embodiments, the invention provides compounds of each of structures I-R/S(50)-(62) where Wi is null and R 3 is halo, alkyl, alkoxy, haloalkyl, perhaloalkyl, haloalkoxy, perhaloalkoxy, -OH, -OR 8 , -CN, -NRiR 8 , , _C(0)R 8 ,

-C(0)NRiR 8 , -NRiC(0)R 8 , -SR 8 , -S(0)R 8 , -S(0) 2 R 8 , -OS(0) 2 R 8 , -S(0) 2 NRiR 8 , -NR ! S(0) 2 R 8 , -(CR a R b ) m NRiR 8 or -(CR a R b ) m O(CR a R b ) m R 8 .

[0040] In certain embodiments, the invention provides compounds of each of structures I-R/S(50)-(62) where Wi is null, p is 1 and R 3 is alkyl.

[0041] In certain embodiments, the invention provides compounds of each of structuress I-R/S(l)-(62) where R 2 is -OH, -N(Ri)-(CR a R b ) m -COOH or -N(Ri)-S0 2 -R 8 ; where Ri is H; where R a and R b are independently H, alkyl, alkoxy,

-(CH 2 ) m C(0)NR4iR4 2 , -(CH 2 ) m C(0)OR 4 o, -(CH 2 ) m NR 4 iR4 2 , -(CH 2 ) m SR 40 , -N(Ri)- heterocyclyl, aryl optionally substituted with R 7 , or wherein Ri and any one of R a or R b taken together form heterocyclyl; R 8 is alkyl; and m is 1 or 2. [0042] In certain embodiments, the invention provides compounds of the following structures (wherein " " represents either or both the R and S form of the compound):

In certain embodiments, the invention provides compounds of structure where A is a 5-membered heteroaryl.

In certain embodiments, the invention provides compounds of structure where A is a 6-membered heteroaryl.

In certain embodiments, the invention provides compounds of structure where A is a 6-membered heteroaryl having one or two nitrogen atoms.

In certain embodiments, the invention provides compounds of structure where A is pyrimindinyl.

In certain embodiments, the invention provides compounds of structure where A is pyridinyl.

In certain embodiments, the invention provides compounds of structure where B is aryl.

In certain embodiments, the invention provides compounds of structure where B is phenyl.

In certain embodiments, the invention provides compounds of structure where B is heteroaryl.

In certain embodiments, the invention provides compounds of structure where B is thiophenyl.

In certain embodiments, the invention provides compounds of structure where R 2 is -OH. [0053] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NH(CR a R b ) m COOH.

[0054] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NHS0 2 R 8 .

[0055] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NHCH 2 COOH.

[0056] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NH(CHR b )COOH where R b is alkyl, -(CH 2 ) m OR4o,

-(CH 2 ) m SR4o, -(CH 2 ) m C(0)OR4o, -(CH^NR^R,, or -(CH 2 ) m C(0)NR4iR4 2 .

[0057] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NH(CRaR b ) m COOH where Ra and R b are independenty H, alkyl, -(CH 2 ) m OR4o, -(CH 2 ) m SR 4 o, -(CH 2 ) m C(0)OR 4 o, -(CH 2 ) m NR 41 R 42 or

[0058] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NRi(CHR b )COOH where Ri and R b taken together form heterocyclyl.

[0059] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NRi(CRaR b ) m COOH where Ri and one of R b taken together form heterocyclyl .

[0060] In certain embodiments, the invention provides compounds of structure I-R/S(63) where any two R a and R b taken together with the carbon to which they are attached form a cycloalkyl.

[0061] In certain embodiments, the invention provides compounds of structure I-R/S(63) where R 2 is -NH(CR a R b ) m COOH where one of R a and R b is H and the other Ra and R b is aryl substituted with R 7 .

[0062] In certain embodiments, the invention provides compounds of structure I-R/S(63) where p is 1 or 2 and each R 3 is independently alkyl, alkoxy, -OH, perhaloalkyl or _C(0)Rs.

[0063] In certain embodiments, the invention provides compounds of structure I-R/S(63) where p is 1 and each R 3 is alkyl. [0064] In certain embodiments, the invention provides compounds of structure I-R/S(63) where q is 1 and R 5 is -(CH 2 ) m -L2-(CH 2 ) m -R7.

[0065] In certain embodiments, the invention provides compounds of structure I-R/S(63) where q is 1 and R 5 is alkoxy.

[0066] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi and Y 2 are null and Z is -S(0) 2 -. Representative compounds of this embodiment include compounds of the following structures (wherein " ''ww " represents either or both the R and S form of the compound):

[0067] In certain embodiments, the invention provides compounds of structure I-R/S(64) where A is pyrimidinyl, B is phenyl and C is phenyl. Representative compounds of this embodiment include compounds of the following structure (wherein " >/ww " represents either or both the R and S form of the compound):

[0068] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi is null, Y 2 is -O- and Z is -C(O)-. Representative compounds of this embodiment include compounds of the following structures (wherein " /ww " represents either or both the R and S form of the compound):

[0069] In certain embodiments, the invention provides a compound of Formula I-R and/or Formula I-S where Yi is NH, Y 2 is null and Z is -C(O)-. Representative compounds of this embodiment include compounds of the following structures (wherein " /ww " represents either or both the R and S form of the compound):

[0070] In certain embodiments, the invention provides a pharmaceutical composition comprising a compound of the invention together with at least one pharmaceutically acceptable carrier, diluent or excipient.

[0071] In certain embodiments, the invention provides a pharmaceutical composition comprising a compound of the invention and a second medicament. In certain of such embodiments, the second medicament is a GLP-1 agonist or a DPPIV inhibitor.

[0072] In certain embodiments, the invention provides a method of use of compounds of the invention for preparation of a medicament.

[0073] In certain embodiments, the invention provides a pharmaceutical combination comprising a compound of the invention and a second medicament. In various such embodiments, the second medicament is an agonist or modulator for glucagon receptor, GIP receptor, GLP-2 receptor, or PTH receptor, or glucagon-like peptide 1 (GLP-1) receptor. In various such embodiments, the second medicament is exenatide, liraglutide, taspoglutide, albiglutide, or lixisenatide or other insulin regulating peptide. In various such embodiments, the second medicament is a DPPIV inhibitor. In various such embodiments, the second medicament is medically indicated for the treatment of type II diabetes.

[0074] In certain embodiments, a method is provided for activation, potentiation or agonism of a glucagon-like peptide 1 comprising contacting the receptor with an effective amount of a compound, pharmaceutical composition or pharmaceutical combination of the invention.

[0075] In further embodiments, a method is provided for activation or agonism of a GLP-1 receptor by contacting the receptor with an effective amount of an invention compound and GLP-1 peptides GLP-1 (9-36) and GLP-1 (7-36), pharmaceutical composition or pharmaceutical combination, wherein the GLP-1 receptor is disposed within a living mammal; in certain embodiments wherein such mammal is a human.

[0076] In certain embodiments, a method is provided for treatment of a malcondition in a subject for which activation, potentiation or agonism of a GLP-1 receptor is medically indicated, by administering an effective amount of an invention compound to the subject at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient. In yet further embodiments, a method is provided for treatment of a malcondition in a patient for which activation, potentiation, or agonism of a GLP-1 receptor is medically indicated, by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient, wherein the malcondition comprises type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety, or metabolic disorder. In certain embodiments, the subject is a patient or a human being. In certain embodiments, the human being is afflicted with, or at risk of developing, a disease or condition selected from the group consisting of type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety, and metabolic disorder. In certain of such embodiments, said disease is type I diabetes or type II diabetes.

[0077] In certain embodiments, the invention provides methods for synthesis of certain compounds including compounds of the invention as more fully illustrated herein. In certain other embodiments, the invention provides certain intermediate compounds associated with such methods of synthesis as illustrated herein.

[0078] In certain embodiments, methods are provided for use of an invention compound for preparation of a medicament adapted for treatment of a disorder or a malcondition wherein activation or inhibition of a GLP-1 receptor is medically indicated. In certain embodiments, the malcondition comprises type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety, and metabolic disorder. Preferably said disease is type I diabetes or type II diabetes.

[0079] In certain embodiments, the method additionally comprises

administering to the subject a second medicament selected from the group of peptidic GLP-1 agonists and DPPIV inhibitors, wherein such second medicament is either a component of the pharmaceutical composition or a second pharmaceutical composition. In certain of such embodiments, the second medicament can be exenatide or sitagliptin.

[0080] As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise.

[0081] As used herein, "individual" (as in the subject of the treatment) means both mammals and non-mammals. Mammals include, for example, humans;

non-human primates, e.g., apes and monkeys; cattle; horses; sheep; and goats.

Non-mammals include, for example, fish and birds. [0082] A "receptor", as is well known in the art, is a biomolecular entity usually comprising a protein that specifically binds a structural class of ligands or a single native ligand in a living organism, the binding of which causes the receptor to transduce the binding signal into another kind of biological action, such as signaling a cell that a binding event has occurred, which causes the cell to alter its function in some manner. An example of transduction is receptor binding of a ligand causing alteration of the activity of a "G-protein" in the cytoplasm of a living cell. Any molecule, naturally occurring or not, that binds to a receptor and activates it for signal transduction, is referred to as an "agonist" or "activator." Any molecule, naturally occurring or not, that binds to a receptor, but does not cause signal transduction to occur, and which can block the binding of an agonist and its consequent signal transduction, is referred to as an "antagonist." Certain molecules bind to receptors at locations other than the binding sites of their natural ligands and such allosteric binding molecules may potentiate, activate or agonize the receptor and may enhance the effect of a natural ligand or a coadministered ligand.

[0083] An "GLP-1 compound" or "GLP-1 agonist" or "GLP-1 activator" or "GLP-1 inhibitor" or "GLP-1 antagonist" or "GLP-1 potentiator" or "GLP-1 modulator" as the terms are used herein refer to compounds that interact in some way with the GLP-1 receptor. They can be agonists, potentiators, or activators, or they can be antagonists or inhibitors. An "GLP-1 compound" of the invention can be selective for action of the GLP-1 receptor family.

[0084] "Substantially" as the term is used herein means completely or almost completely; for example, a composition that is "substantially free" of a component either has none of the component or contains such a trace amount that any relevant functional property of the composition is unaffected by the presence of the trace amount, or a compound is "substantially pure" is there are only negligible traces of impurities present.

[0085] Substantially enantiomerically or diasteromerically pure means a level of enantiomeric or diasteromeric enrichment of one enantiomer with respect to the other enantiomer or diasteromer of at least 90%, 95%, 98%, 99%, 99.5% or 99.9%. [0086] "Treating" or "treatment" within the meaning herein refers to an alleviation of symptoms associated with a disorder or disease, or inhibition of further progression or worsening of those symptoms, or prevention or prophylaxis of the disease or disorder.

[0087] The expression "effective amount", when used to describe use of a compound of the invention in providing therapy to a patient suffering from a disorder or malcondition mediated by GLP-1 refers to the amount of a compound of the invention that is effective to bind to as an agonist or as an antagonist a GLP-1 receptor in the individual's tissues, wherein the GLP-1 is implicated in the disorder, wherein such binding occurs to an extent sufficient to produce a beneficial therapeutic effect on the patient. Similarly, as used herein, an "effective amount" or a "therapeutically effective amount" of a compound of the invention refers to an amount of the compound that alleviates, in whole or in part, symptoms associated with the disorder or condition, or halts or slows further progression or worsening of those symptoms, or prevents or provides prophylaxis for the disorder or condition. In particular, a "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result by acting as an agonist of GLP-1 activity. A therapeutically effective amount is also one in which any toxic or detrimental effects of compounds of the invention are outweighed by the therapeutically beneficial effects. For example, in the context of treating a malcondition mediated by activation of a GLP-1 receptor, a therapeutically effective amount of a GLP-1 receptor agonist of the invention is an amount sufficient to control the malcondition, to mitigate the progress of the malcondition, or to relieve the symptoms of the malcondition.

Examples of malconditions that can be so treated include, but not limited to, type II diabetes.

[0088] All chiral, diastereomeric, racemic forms of a structure are intended, unless a particular stereochemistry or isomeric form is specifically indicated.

Compounds used in the present invention can include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions, at any degree of enrichment. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these are all within the scope of certain embodiments of the invention.

[0089] The isomers resulting from the presence of a chiral center comprise a pair of non-superimposable isomers that are called "enantiomers." Single enantiomers of a pure compound are optically active, i.e., they are capable of rotating the plane of plane polarized light. Single enantiomers are designated according to the

Cahn-Ingold-Prelog system. Once the priority ranking of the four groups is

determined, the molecule is oriented so that the lowest ranking group is pointed away from the viewer. Then, if the descending rank order of the other groups proceeds clockwise, the molecule is designated (R) and if the descending rank of the other groups proceeds counterclockwise, the molecule is designated (S). In the example in Scheme 14, the Cahn-Ingold-Prelog ranking is A > B > C > D. The lowest ranking atom, D is oriented away from the viewer.

(R) configuration (S) configuration

[0090] "Isolated optical isomer" means a compound which has been

substantially purified from the corresponding optical isomer(s) of the same formula. Preferably, the isolated isomer is at least about 80%, more preferably at least 90%> pure, even more preferably at least 98%> pure, most preferably at least about 99% pure, by weight.

[0091] Enantiomers are sometimes called optical isomers because a pure enantiomer rotates plane-polarized light in a particular direction. If the light rotates clockwise, then that enantiomer is labeled "(+)" or "d" for dextrorotatory, its counterpart will rotate the light counterclockwise and is labeled "(-)" or "1" for levorotatory. [0092] The terms "racemate" and "racemic mixture" are frequently used interchangeably. A racemate is an equal mixture of two enantiomers. A racemate is labeled "(±)" because it is not optically active (i.e., will not rotate plane-polarized light in either direction since its constituent enantiomers cancel each other out).

[0093] It is understood that due to chemical properties (i.e., resonance lending some double bond character to the C-N bond) of restricted rotation about the amide bond linkage (as illustrated below) it is possible to observe separate rotamer species and even, under some circumstances, to isolate such species, example shown below. It is further understood that certain structural elements, including steric bulk or substituents on the amide nitrogen, may enhance the stability of a rotamer to the extent that a compound may be isolated as, and exist indefinitely, as a single stable rotamer. The present invention therefore includes any possible stable rotamers of compounds of the invention which are biologically active in the treatment of type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety, or metabolic disorder.

[0094] The preferred compounds of the present invention have a particular spatial arrangement of substituents on the aromatic rings, which is related to the structure activity relationship demonstrated by the compound class. Often such substitution arrangement is denoted by a numbering system; however, numbering systems are often not consistent between different ring systems. In six-membered aromatic systems, the spatial arrangements are specified by the common nomenclature "para" for 1 ,4-substitution, "meta" for 1,3-substitution and "ortho" for 1 ,2-substitution as shown below.

[0095] All structures encompassed within a claim are "chemically feasible", by which is meant that the structure depicted by any combination or subcombination of optional substituents meant to be recited by the claim is physically capable of existence with at least some stability as can be determined by the laws of structural chemistry and by experimentation. Structures that are not chemically feasible are not within a claimed set of compounds. Further, isotopes of the atoms depicted (such as deuterium and tritium in the case of hydrogen) are encompassed within the scope of this invention.

[0096] In general, "substituted" refers to an organic group as defined herein in which one or more bonds to a hydrogen atom contained therein are replaced by one or more bonds to a non-hydrogen atom such as, but not limited to, a halogen (i.e., F, CI, Br, and I); an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, aralkyloxy groups, oxo(carbonyl) groups, carboxyl groups including carboxylic acids, carboxylates, and carboyxlate esters; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfoxide groups, sulfone groups, sulfonyl groups, and sulfonamide groups; a nitrogen atom in groups such as amines, hydroxylamines, nitriles, nitro groups, N-oxides, hydrazides, azides, and enamines; and other

heteroatoms in various other groups. Non-limiting examples of substituents that can be bonded to a substituted carbon (or other) atom include F, CI, Br, I, OR', OC(0)N(R) 2 ,

CN, CF 3 , OCF 3 , R', O, S, C(O), S(O), methylenedioxy, ethylenedioxy, N(R) 2 , SR, SOR, S0 2 R, S0 2 N(R * ) 2 , S0 3 R, C(0)R, C(0)C(0)R, C(0)CH 2 C(0)R * , C(S)R * , C(0)OR, OC(0)R, C(0)N(R') 2 , OC(0)N(R) 2 , C(S)N(R) 2 , (CH 2 ) 0 _ 2 NHC(O)R, (CH 2 ) 0 _ 2 N(R)N(R) 2 , N(R)N(R)C(0)R, N(R)N(R * )C(0)OR, N(R)N(R)CON(R * ) 2 ,

N(R)S0 2 R*, N(R*)S0 2 N(R) 2 , N(R*)C(0)OR*, N(R)C(0)R, N(R)C(S)R*,

N(R)C(0)N(R) 2 , N(R)C(S)N(R) 2 , N(COR)COR, N(OR * )R * , C(=NH)N(R) 2 ,

C(0)N(OR)R, or C(=NOR')R' wherein R' can be hydrogen or a carbon-based moiety, and wherein the carbon-based moiety can itself be further substituted. [0097] Substituted alkyl, alkenyl, alkynyl, cycloalkyl, and cycloalkenyl groups as well as other substituted groups also include groups in which one or more bonds to a hydrogen atom are replaced by one or more bonds, including double or triple bonds, to a carbon atom, or to a heteroatom such as, but not limited to, oxygen in carbonyl (oxo), carboxyl, ester, amide, imide, urethane, and urea groups; and nitrogen in imines, hydroxyimines, oximes, hydrazones, amidines, guanidines, and nitriles.

[0098] Substituted ring groups includes substituted aryl, heterocyclyl and heteroaryl groups. Substituted ring groups can be substituted by one or more substituents at any available ring position. In some embodiments, two substituents on a substituted ring group may taken together with the ring to which they are attached to form a ring, such that the two rings are fused together. For example, benzodioxolyl is a fused ring system formed by two substituents taken together on a phenyl group.

[0099] Such substituted ring groups also include rings and fused ring systems in which a bond to a hydrogen atom is replaced with a bond to a carbon atom. Therefore, substituted aryl, heterocyclyl and heteroaryl groups can also be substituted with alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, and alkynyl groups as defined herein, which can themselves be further substituted.

[00100] The linking groups (e.g., Li and L 2 ) of Formula I-R or I-S are partial structures which may be represented by a formula, say, for example, -N(Ri)-C(0)-, which is read from left-to-right. Accordingly, the nitrogen atom of the -N(Ri)-C(0)- linker will be attached to the proximal end of the structure of Formula I-R or I-S, and the carbonyl carbon atom of the -N(Ri)-C(0)- linker will be attached to the distal end of the structure of Formula I-R or I-S.

[00101] The term "heteroatoms" as used herein refers to non-carbon and non- hydrogen atoms, capable of forming covalent bonds with carbon, and is not otherwise limited. Typical heteroatoms are N, O, and S. When sulfur (S) is referred to, it is understood that the sulfur can be in any of the oxidation states in which it is found, thus including sulfoxides (R-S(O)-R') and sulfones (R-S(0) 2 -R'), unless the oxidation state is specified; thus, the term "sulfone" encompasses only the sulfone form of sulfur; the term "sulfide" encompasses only the sulfide (R-S-R) form of sulfur. When the phrases such as "heteroatoms selected from the group consisting of O, NH, NR' and S," or "[variable] is O, S . . ." are used, they are understood to encompass all of the sulfide, sulfoxide and sulfone oxidation states of sulfur.

[00102] Alkyl groups include straight chain and branched alkyl groups and cycloalkyl groups having from 1 to about 20 carbon atoms, and typically from 1 to 12 carbons (C 1 -C 12 alkyl), or, in some embodiments, from 1 to 8 carbon atoms (Ci-Cg alkyl), or, in some embodiments, from 1 to 4 carbon atoms (C 1 -C4 alkyl). Examples of straight chain alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n- butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl groups. Examples of branched alkyl groups include, but are not limited to, isopropyl, iso-butyl, sec-butyl, t-butyl, neopentyl, isopentyl, and 2,2-dimethylpropyl groups. Alkyl groups as used herein may optionally include one or more further substituent groups. Representative substituted alkyl groups can be substituted one or more times with any of the groups listed above, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.

[00103] Cycloalkyl groups are alkyl groups forming a ring structure, which can be substituted or unsubstituted, wherein the ring is either completely saturated, partially unsaturated, or fully unsaturated, wherein if there is unsaturation, the conjugation of the pi-electrons in the ring do not give rise to aromaticity. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 5, 3 to 6, or 3 to 7. Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbornyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like. Cycloalkyl groups also include rings that are substituted with straight or branched chain alkyl groups as defined above. Representative substituted cycloalkyl groups can be mono-substituted or substituted one or more times with any of the groups listed above, for example, but not limited to, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups. [00104] The terms "carbocyclic" and "carbocycle" denote a ring structure wherein the atoms of the ring are carbon. In some embodiments, the carbocycle has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms is 4, 5, 6, or 7. Unless specifically indicated to the contrary, the carbocyclic ring can be substituted with as many as N substituents wherein N is the size of the carbocyclic ring with for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.

[00105] (Cycloalkyl)alkyl groups, also denoted cycloalkylalkyl, are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkyl group as defined above.

[00106] Alkenyl groups include straight and branched chain and cyclic alkyl groups as defined above, except that at least one double bond exists between two carbon atoms. Thus, alkenyl groups have from 2 to about 20 carbon atoms, and typically from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms. Examples include, but are not limited to -CH=CH(CH 3 ), -CH=C(CH 3 ) 2 , -C(CH 3 )=CH 2 , -C(CH 3 )=CH(CH 3 ), -C(CH 2 CH 3 )=CH 2 , vinyl, cyclohexenyl, cyclopentenyl,

cyclohexadienyl, butadienyl, pentadienyl, and hexadienyl among others.

[00107] The term "cycloalkenyl" alone or in combination denotes a cyclic alkenyl group wherein at least one double bond is present in the ring structure.

Cycloalkenyl groups include cycloalkyl groups having at least one double bond between two adjacent carbon atoms. Thus for example, cycloalkenyl groups include but are not limited to cyclohexenyl, cyclopentenyl, and cyclohexadienyl groups.

[00108] (Cycloalkenyl)alkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkenyl group as defined above.

[00109] Alkynyl groups include straight and branched chain alkyl groups, except that at least one triple bond exists between two carbon atoms. Thus, alkynyl groups have from 2 to about 20 carbon atoms, and typically from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms. Examples include, but are not limited to - C≡CH, -C≡C(CH 3 ), -C≡C(CH 2 CH 3 ), -CH 2 C≡CH, -CH 2 C≡C(CH 3 ), and

-CH 2 C≡C(CH 2 CH 3 ), among others.

[00110] Aryl groups are cyclic aromatic hydrocarbons that do not contain heteroatoms. Thus aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups. In some embodiments, aryl groups contain 6-14 carbons in the ring portions of the groups. The phrase "aryl groups" includes groups containing fused rings, such as fused aromatic- aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like), and also includes substituted aryl groups that have other groups, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring atoms. Representative substituted aryl groups can be mono-substituted or substituted more than once, such as, but not limited to, 2-, 3-, 4-, 5-, or 6-substituted phenyl or naphthyl groups, which can be substituted with groups including but not limited to those listed above.

[00111] Aralkyl groups are alkyl groups as defined above in which a hydrogen atom of an alkyl group is replaced with an aryl group as defined above. Representative aralkyl groups include benzyl and phenylethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl. The aryl moiety or the alkyl moiety or both are optionally substituted with other groups, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups. Aralkenyl group are alkenyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined above.

[00112] Heterocyclyl or heterocyclic groups include aromatic and non-aromatic ring moieties containing 3 or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, S, or P. In some embodiments, heterocyclyl groups include 3 to 20 ring members, whereas other such groups have 3 to 15 ring members, including for example single ring systems containing 5, 6 or 7 ring members. At least one ring contains a heteroatom, but every ring in a polycyclic system need not contain a heteroatom. For example, a dioxolanyl ring and a benzdioxolanyl ring system (methylenedioxyphenyl ring system) are both heterocyclyl groups within the meaning herein. A heterocyclyl group designated as a C2-heterocyclyl can be a 5 -ring with two carbon atoms and three heteroatoms, a 6-ring with two carbon atoms and four heteroatoms, and so forth. Likewise a C4-heterocyclyl can be a 5 -ring with one heteroatom, a 6-ring with two heteroatoms, and so forth. The number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms.

[00113] The term "heterocyclyl" includes fused ring species including those having fused aromatic and non-aromatic groups. The phrase also includes poly cyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl and also includes heterocyclyl groups that have substituents, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring members. A heterocyclyl group as defined herein can be a heteroaryl group or a partially or completely saturated cyclic group including at least one ring heteroatom. Heterocyclyl groups include, but are not limited to, pyrazinyl, pyrimidinyl, pyridazinyl, thiadiazolyl, oxadiazolyl, imidazolinyl, hexahydropyrimidinyl, diazepanyl, triazinyl, imidazolyl, pyrrolidinyl, furanyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl,

dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups.

Heterocyclyl groups can be substituted. Representative substituted heterocyclyl groups can be mono-substituted or substituted more than once, including but not limited to, rings containing at least one heteroatom which are mono, di, tri, tetra, penta, hexa, or higher-substituted with substituents such as those listed above, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, and alkoxy groups.

[00114] Heteroaryl groups are aromatic ring moieties containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S. A heteroaryl group designated as a C 2 -heteroaryl can be a 5 -ring with two carbon atoms and three heteroatoms, a 6-ring with two carbon atoms and four heteroatoms and so forth. Likewise a C4-heteroaryl can be a 5 -ring with one heteroatom, a 6-ring with two heteroatoms, and so forth. The number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms. Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, pyrazinyl, pyrimidinyl, thiadiazolyl, imidazolyl, oxadiazolyl, thienyl, triazolyl, tetrazolyl, triazinyl, thiazolyl, thiophenyl, oxazolyl, isoxazolyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl,

benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, and quinazolinyl groups. The terms "heteroaryl" and "heteroaryl groups" include fused ring compounds such as wherein at least one ring, but not necessarily all rings, are aromatic, including tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolyl and 2,3-dihydro indolyl. The term also includes heteroaryl groups that have other groups bonded to one of the ring members, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups. Representative substituted heteroaryl groups can be substituted one or more times with groups such as those listed above.

[00115] Additional examples of aryl and heteroaryl groups include but are not limited to phenyl, biphenyl, indenyl, naphthyl (1-naphthyl, 2-naphthyl), N- hydroxytetrazolyl, N-hydroxytriazolyl, N-hydroxyimidazolyl, anthracenyl (1- anthracenyl, 2-anthracenyl, 3-anthracenyl), thiophenyl (2 -thienyl, 3-thienyl), furyl (2- furyl, 3-furyl), indolyl, oxadiazolyl (1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl), thiadiazolyl (1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl), isoxazolyl, quinazolinyl, fluorenyl, xanthenyl, isoindanyl, benzhydryl, acridinyl, thiazolyl, pyrrolyl (2 -pyrrolyl), pyrazolyl (3- pyrazolyl), imidazolyl (1 -imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl), triazolyl (1,2,3-triazol-l-yl, l,2,3-triazol-2-yl l,2,3-triazol-4-yl, l,2,4-triazol-3-yl), oxazolyl (2 -oxazolyl, 4-oxazolyl, 5-oxazolyl), thiazolyl (2-thiazolyl, 4-thiazolyl, 5- thiazolyl), pyridyl (2-pyridyl, 3-pyridyl, 4-pyridyl), pyrimidinyl (2 -pyrimidinyl, 4- pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl), pyrazinyl, pyridazinyl (3- pyridazinyl, 4- pyridazinyl, 5 -pyridazinyl), pyrazolo[l,5-a]pyridinyl, quinolyl (2-quinolyl, 3-quinolyl,

4- quinolyl, 5-quinolyl, 6-quinolyl, 7-quinolyl, 8-quinolyl), isoquinolyl (1-isoquinolyl, 3-isoquinolyl, 4-isoquinolyl, 5-isoquinolyl, 6-isoquinolyl, 7-isoquinolyl, 8-isoquinolyl), benzo[b]furanyl (2-benzo[b]furanyl, 3-benzo[b]furanyl, 4-benzo[b]furanyl, 5- benzo[b]furanyl, 6-benzo[b]furanyl, 7-benzo[b]furanyl), isobenzofuranyl, 2,3-dihydro- benzo[b]furanyl (2-(2,3-dihydro-benzo[b]furanyl), 3-(2,3-dihydro-benzo[b]furanyl), 4- (2,3-dihydro-benzo[b]furanyl), 5-(2,3-dihydro-benzo[b]furanyl), 6-(2,3-dihydro- benzo[b]furanyl), 7-(2,3-dihydro-benzo[b]furanyl), benzo[b]thiophenyl (2- benzo[b]thiophenyl, 3-benzo[b]thiophenyl, 4-benzo[b]thiophenyl,

5- benzo[b]thiophenyl, 6-benzo[b]thiophenyl, 7-benzo[b]thiophenyl), 2,3-dihydro- benzo[b]thiophenyl, (2-(2,3-dihydro-benzo[b]thiophenyl), 3-(2,3-dihydro- benzo[b]thiophenyl), 4-(2,3-dihydro-benzo[b]thiophenyl), 5-(2,3-dihydro- benzo[b]thiophenyl), 6-(2,3-dihydro-benzo[b]thiophenyl), 7-(2,3-dihydro- benzo[b]thiophenyl), indolyl (1-indolyl, 2-indolyl, 3-indolyl, 4-indolyl, 5-indolyl, 6- indolyl, 7-indolyl), indazole (1-indazolyl, 3-indazolyl, 4-indazolyl, 5-indazolyl, 6- indazolyl, 7-indazolyl), benzimidazolyl (1-benzimidazolyl, 2-benzimidazolyl, 4- benzimidazolyl, 5 -benzimidazolyl, 6-benzimidazolyl, 7-benzimidazolyl,

8-benzimidazolyl), benzoxazolyl (1-benzoxazolyl, 2-benzoxazolyl), benzothiazolyl (1- benzothiazolyl, 2-benzothiazolyl, 4-benzothiazolyl, 5-benzothiazolyl, 6-benzothiazolyl, 7-benzothiazolyl), benzo[d]isoxazolyl, carbazolyl (1-carbazolyl, 2-carbazolyl, 3- carbazolyl, 4-carbazolyl), 5H-dibenz[b,f]azepine (5H-dibenz[b,f]azepin-l-yl, 5H- dibenz[b,f]azepine-2-yl, 5H-dibenz[b,f]azepine-3-yl, 5H-dibenz[b,f]azepine-4-yl, 5H- dibenz[b,f]azepine-5-yl), 10,1 l-dihydro-5H-dibenz[b,f]azepine (10,1 l-dihydro-5H- dibenz[b,f]azepine-l-yl, 10,1 l-dihydro-5H-dibenz[b,f]azepine-2-yl, 10,1 l-dihydro-5H- dibenz[b,f]azepine-3-yl, 10,1 l-dihydro-5H-dibenz[b,f]azepine-4-yl, 10,1 l-dihydro-5H- dibenz[b,f]azepine-5-yl), and the like.

[00116] Heterocyclylalkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heterocyclyl group as defined above. Representative heterocyclyl alkyl groups include, but are not limited to, furan-2-yl methyl, furan-3-yl methyl, pyridine-2-yl methyl (a-picolyl), pyridine-3-yl methyl (β-picolyl), pyridine-4-yl methyl (γ-picolyl), tetrahydrofuran-2-yl ethyl, and indol-2-yl propyl. Heterocyclylalkyl groups can be substituted on the heterocyclyl moiety, the alkyl moiety, or both.

[00117] Heteroarylalkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heteroaryl group as defined above. Heteroarylalkyl groups can be substituted on the heteroaryl moiety, the alkyl moiety, or both.

[00118] By a "ring system" as the term is used herein is meant a moiety comprising one, two, three or more rings, which can be substituted with non-ring groups or with other ring systems, or both, which can be fully saturated, partially unsaturated, fully unsaturated, or aromatic, and when the ring system includes more than a single ring, the rings can be fused, bridging, or spirocyclic. By "spirocyclic" is meant the class of structures wherein two rings are fused at a single tetrahedral carbon atom, as is well known in the art.

[00119] A "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" as the term is used herein refers to a ring system including an unsaturated ring possessing 4n+2 pi electrons, or a partially reduced (hydrogenated) form thereof. The aromatic or partially aromatic ring can include additional fused, bridged, or spiro rings that are not themselves aromatic or partially aromatic. For example, naphthalene and tetrahydronaphthalene are both a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" within the meaning herein. Also, for example, a benzo-[2.2.2]- bicyclooctane is also a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" within the meaning herein, containing a phenyl ring fused to a bridged bicyclic system. A fully saturated ring has no double bonds therein, and is carbocyclic or heterocyclic depending on the presence of heteroatoms within the meaning herein.

[00120] When two "R" groups are said to be joined together or taken together to form a ring, it is meant that together with the carbon atom or a non-carbon atom (e.g., nitrogen atom), to which they are bonded, they may form a ring system. In general, they are bonded to one another to form a 3- to 7-membered ring, or a 5- to 7-membered ring. Non-limiting specific examples are the cyclopentyl, cyclohexyl, cycloheptyl, piperidinyl, piperazinyl, pyrolidinyl, pyrrolyl, pyridinyl.

[00121] The term "alkoxy" refers to an oxygen atom connected to an alkyl group, including a cycloalkyl group, as are defined above. Examples of linear alkoxy groups include but are not limited to methoxy, ethoxy, n-propoxy, n-butoxy, n-pentyloxy, n- hexyloxy, n-heptyloxy, n-octyloxy n-nonyloxy, and the like. Examples of branched alkoxy include but are not limited to isopropoxy, sec-butoxy, tert-butoxy, isopentyloxy, isohexyloxy, and the like. Examples of cyclic alkoxy include but are not limited to cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.

[00122] The terms "aryloxy" and "arylalkoxy" refer to, respectively, an aryl group bonded to an oxygen atom and an aralkyl group bonded to the oxygen atom at the alkyl moiety. Examples include but are not limited to phenoxy, naphthyloxy, and benzyloxy.

[00123] An "acyl" group as the term is used herein refers to a group containing a carbonyl moiety wherein the group is bonded via the carbonyl carbon atom. The carbonyl carbon atom is also bonded to another carbon atom, which can be part of an alkyl, aryl, aralkyl cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl group or the like. In the special case wherein the carbonyl carbon atom is bonded to a hydrogen, the group is a "formyl" group, an acyl group as the term is defined herein. An acyl group can include 0 to about 12-20 additional carbon atoms bonded to the carbonyl group. An acyl group can include double or triple bonds within the meaning herein. An acryloyl group is an example of an acyl group. An acyl group can also include heteroatoms within the meaning here. A nicotinoyl group (pyridyl-3 -carbonyl) group is an example of an acyl group within the meaning herein. Other examples include acetyl, benzoyl, phenylacetyl, pyridylacetyl, cinnamoyl, and acryloyl groups and the like. When the group containing the carbon atom that is bonded to the carbonyl carbon atom contains a halogen, the group is termed a "haloacyl" group. An example is a trifluoroacetyl group.

[00124] The term "amine" includes primary, secondary, and tertiary amines having, e.g., the formula N(group) 3 wherein each group can independently be H or non- H, such as alkyl, aryl, and the like. Amines include but are not limited to R-NH 2 , for example, alkylamines, arylamines, alkylarylamines; R 2 NH wherein each R is independently selected, such as dialkylamines, diarylamines, aralkylamines, heterocyclylamines and the like; and R 3 N wherein each R is independently selected, such as trialkylamines, dialkylarylamines, alkyldiarylamines, triarylamines, and the like. The term "amine" also includes ammonium ions as used herein.

[00125] An "amino" group is a substituent of the form -NH 2 , -NHR, -NR 2 , - NR 3 + , wherein each R is independently selected, and protonated forms of each.

Accordingly, any compound substituted with an amino group can be viewed as an amine.

[00126] An "ammonium" ion includes the unsubstituted ammonium ion N¾ + , but unless otherwise specified, it also includes any protonated or quaternarized forms of amines. Thus, trimethylammonium hydrochloride and tetramethylammonium chloride are both ammonium ions, and amines, within the meaning herein.

[00127] The term "amide" (or "amido") includes C- and N-amide groups, i.e., -C(0)NR 2 , and -NRC(0)R groups, respectively. Amide groups therefore include but are not limited to carbamoyl groups (-C(0)NH 2 ) and formamide groups (-NHC(O)H). A "carboxamido" group is a group of the formula C(0)NR 2 , wherein R can be H, alkyl, aryl, etc.

[00128] The term "carbonyl," refers to a -C(O)- group.

[00129] "Halo," "halogen," and "halide" include fluorine, chlorine, bromine and iodine.

[00130] The term "perhaloalkyl" refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms. Perhaloalkyl groups include, but are not limited to, -CF 3 and -C(CF 3 ) 3 . The term "haloalkyl" refers to an alkyl group where some but not necessarily all of the hydrogen atoms are replaced by halogen atoms. Haloalkyl groups include but are not limited to -CHF 2 and -CH 2 F.

[00131] The term "perhaloalkoxy" refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms. Perhaloalkoxy groups include, but are not limited to, -OCF 3 and -OC(CF 3 ) 3 . The term "haloalkoxy" refers to an alkoxy group where some but not necessarily all of the hydrogen atoms are replaced by halogen atoms. Haloalkoxy groups include but are not limited to -OCHF 2 and -OCH 2 F.

[00132] The terms "comprising," "including," "having," "composed of," are open-ended terms as used herein, and do not preclude the existence of additional elements or components. In a claim element, use of the forms "comprising,"

"including," "having," or "composed of means that whatever element is comprised, had, included, or composes is not necessarily the only element encompassed by the subject of the clause that contains that word.

[00133] A "salt" as is well known in the art includes an organic compound such as a carboxylic acid, a sulfonic acid, or an amine, in ionic form, in combination with a counterion. For example, acids in their anionic form can form salts with cations such as metal cations, for example sodium, potassium, and the like; with ammonium salts such as NH 4 + or the cations of various amines, including tetraalkyl ammonium salts such as tetramethylammonium, or other cations such as trimethylsulfonium, and the like. A "pharmaceutically acceptable" or "pharmacologically acceptable" salt is a salt formed from an ion that has been approved for human consumption and is generally non-toxic, such as a chloride salt or a sodium salt. A "zwitterion" is an internal salt such as can be formed in a molecule that has at least two ionizable groups, one forming an anion and the other a cation, which serve to balance each other. For example, amino acids such as glycine can exist in a zwitterionic form. A "zwitterion" is a salt within the meaning herein. The compounds of the present invention may take the form of salts. The term "salts" embraces addition salts of free acids or free bases which are compounds of the invention. Salts can be "pharmaceutically-acceptable salts." The term

"pharmaceutically-acceptable salt" refers to salts which possess toxicity profiles within a range that affords utility in pharmaceutical applications. Pharmaceutically

unacceptable salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present invention, such as for example utility in process of synthesis, purification or formulation of compounds of the invention.

[00134] Suitable pharmaceutically-acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic,

trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, β-hydroxybutyric, salicylic, galactaric and galacturonic acid. Examples of pharmaceutically unacceptable acid addition salts include, for example, perchlorates and tetrafluoroborates.

[00135] Suitable pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example,

N^-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine,

ethylenediamine, meglumine (N-methylglucamine) and procaine. Examples of pharmaceutically unacceptable base addition salts include lithium salts and cyanate salts. Although pharmaceutically unacceptable salts are not generally useful as medicaments, such salts may be useful, for example as intermediates in the synthesis of Formula I compounds, for example in their purification by recrystallization. All of these salts may be prepared by conventional means from the corresponding compound according to Formula I by reacting, for example, the appropriate acid or base with the compound according to Formula I. The term "pharmaceutically acceptable salts" refers to nontoxic inorganic or organic acid and/or base addition salts, see, for example, Lit et al, Salt Selection for Basic Drugs (1986), IntJ. Pharm., 33, 201-217, incorporated by reference herein.

[00136] A "hydrate" is a compound that exists in a composition with water molecules. The composition can include water in stoichiometric quantities, such as a monohydrate or a dihydrate, or can include water in random amounts. As the term is used herein a "hydrate" refers to a solid form, i.e., a compound in water solution, while it may be hydrated, is not a hydrate as the term is used herein.

[00137] A "solvate" is a similar composition except that a solvent other that water replaces the water. For example, methanol or ethanol can form an "alcoholate", which can again be stoichiometric or non-stoichiometric. As the term is used herein a "solvate" refers to a solid form, i.e., a compound in solution in a solvent, while it may be solvated, is not a solvate as the term is used herein.

[00138] A "prodrug" as is well known in the art is a substance that can be administered to a patient where the substance is converted in vivo by the action of biochemicals within the patient's body, such as enzymes, to the active pharmaceutical ingredient. Examples of prodrugs include esters of carboxylic acid groups, which can be hydro lyzed by endogenous esterases as are found in the bloodstream of humans and other mammals.

[00139] "Isotopes" are well know in the art and refer to atoms with the same number of protons but different number of neutrons. For example, carbon 12, the most common form of carbon, has six protons and six neutrons, whereas carbon 14 has six protons and eight neutrons.

[00140] In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group. For example, if X is described as selected from the group consisting of bromine, chlorine, and iodine, claims for X being bromine and claims for X being bromine and chlorine are fully described. Moreover, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any combination of individual members or subgroups of members of Markush groups. Thus, for example, if X is described as selected from the group consisting of bromine, chlorine, and iodine, and Y is described as selected from the group consisting of methyl, ethyl, and propyl, claims for X being bromine and Y being methyl are fully described. COMPOSITIONS AND COMBINATION TREATMENTS

[00141] The GLP-1 compounds, their pharmaceutically acceptable salts or hydrolyzable esters of the present invention may be combined with a pharmaceutically acceptable carrier to provide pharmaceutical compositions useful for treating the biological conditions or disorders noted herein in mammalian species, and more preferably, in humans. The particular carrier employed in these pharmaceutical compositions may vary depending upon the type of administration desired (e.g., intravenous, oral, topical, suppository, or parenteral).

[00142] In preparing the compositions in oral liquid dosage forms (e.g., suspensions, elixirs and solutions), typical pharmaceutical media, such as water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like can be employed. Similarly, when preparing oral solid dosage forms (e.g., powders, tablets and capsules), carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like can be employed.

[00143] Another aspect of an embodiment of the invention provides

compositions of the compounds of the invention, alone or in combination with another GLP-1 agonist or another type of therapeutic agent, or both. Non- limiting examples of the GLP-1 receptor agonists include exenatide, liraglutide, taspoglutide, albiglutide, lixisenatide, and mixtures thereof.

[00144] In one embodiment, the GLP-1 agonist is exenatide (Byetta®) or Byetta LAR®. Exenatide is described, for example, in U.S. Pat. Nos. 5,424,286; 6,902,744; 7,297,761, and others, the contents of each of which is herein incorporated by reference in its entirety.

[00145] In one embodiment, the GLP-1 agonist is liraglutide (VICTOZA®) (also called NN-2211 and [Arg34, Lys26]-(N-epsilon-(gamma-Glu(N-alpha-hexadecanoyl))- GLP-1 (7-37)), includes the sequence

HAEGTFTSDVSSYLEGQAAKEFIAWKVRGRG (SEQ ID NO: l) and is available from Novo Nordisk (Denmark) or Scios (Fremont, Calif. USA). See, e.g., Elbrond et al, 2002, Diabetes Care. August; 25(8):1398404; Agerso et al, 2002, Diabetologia. February; 45(2): 195-202). [00146] In one embodiment, the GLP-1 agonist is taspoglutide (CAS Registry No. 275371-94-3) and is available from Hoffman La-Roche. See, for example, U.S. Pat. No. 7,368,427, the contents of which are herein incorporated by reference in its entirety.

[00147] In one embodiment, the GLP-1 agonist is albiglutide (SYNCRIA® from Glaxo SmithKline) .

[00148] In another embodiment, the GLP-1 agonist is lixisenatide (Lyxumia® from Sanofi-Aventis/Zealand Pharma)

[00149] As set forth herein, compounds of the invention include stereoisomers, tautomers, solvates, hydrates, salts including pharmaceutically acceptable salts, and mixtures thereof. Compositions containing a compound of the invention can be prepared by conventional techniques, e.g., as described in Remington: The Science and Practice of Pharmacy, 19th Ed., 1995, incorporated by reference herein. The compositions can appear in conventional forms, for example capsules, tablets, aerosols, solutions, suspensions or topical applications.

[00150] Typical compositions include a compound of the invention and a pharmaceutically acceptable excipient which can be a carrier or a diluent. For example, the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which can be in the form of an ampoule, capsule, sachet, paper, or other container. When the active compound is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound. The active compound can be adsorbed on a granular solid carrier, for example contained in a sachet. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols,

polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone. Similarly, the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.

[00151] The formulations can be mixed with auxiliary agents which do not deleteriously react with the active compounds. Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances preserving agents, sweetening agents or flavoring agents. The compositions can also be sterilized if desired.

[00152] The route of administration can be any route which effectively transports the active compound of the invention to the appropriate or desired site of action, such as oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal or parenteral, e.g., rectal, depot, subcutaneous, intravenous, intraurethral, intramuscular, intranasal, ophthalmic solution or an ointment, the oral route being preferred.

[00153] For parenteral administration, the carrier will typically comprise sterile water, although other ingredients that aid solubility or serve as preservatives can also be included. Furthermore, injectable suspensions can also be prepared, in which case appropriate liquid carriers, suspending agents and the like can be employed.

[00154] For topical administration, the compounds of the present invention can be formulated using bland, moisturizing bases such as ointments or creams.

[00155] If a solid carrier is used for oral administration, the preparation can be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a troche or lozenge. If a liquid carrier is used, the preparation can be in the form of a syrup, emulsion, soft gelatin capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution.

[00156] Injectable dosage forms generally include aqueous suspensions or oil suspensions which can be prepared using a suitable dispersant or wetting agent and a suspending agent Injectable forms can be in solution phase or in the form of a suspension, which is prepared with a solvent or diluent. Acceptable solvents or vehicles include sterilized water, Ringer's solution, or an isotonic aqueous saline solution. Alternatively, sterile oils can be employed as solvents or suspending agents. Preferably, the oil or fatty acid is non-volatile, including natural or synthetic oils, fatty acids, mono-, di- or tri-glycerides.

[00157] For injection, the formulation can also be a powder suitable for reconstitution with an appropriate solution as described above. Examples of these include, but are not limited to, freeze dried, rotary dried or spray dried powders, amorphous powders, granules, precipitates, or particulates. For injection, the formulations can optionally contain stabilizers, pH modifiers, surfactants,

bioavailability modifiers and combinations of these. The compounds can be formulated for parenteral administration by injection such as by bolus injection or continuous infusion. A unit dosage form for injection can be in ampoules or in multi-dose containers.

[00158] The formulations of the invention can be designed to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art. Thus, the formulations can also be formulated for controlled release or for slow release.

[00159] Compositions contemplated by the present invention can include, for example, micelles or liposomes, or some other encapsulated form, or can be

administered in an extended release form to provide a prolonged storage and/or delivery effect. Therefore, the formulations can be compressed into pellets or cylinders and implanted intramuscularly or subcutaneously as depot injections. Such implants can employ known inert materials such as silicones and biodegradable polymers, e.g., polylactide-polyglycolide. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).

[00160] For nasal administration, the preparation can contain a compound of the invention, dissolved or suspended in a liquid carrier, preferably an aqueous carrier, for aerosol application. The carrier can contain additives such as solubilizing agents, e.g., propylene glycol, surfactants, absorption enhancers such as lecithin

(phosphatidylcholine) or cyclodextrin, or preservatives such as parabens. [00161] For parenteral application, particularly suitable are injectable solutions or suspensions, preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil.

[00162] Dosage forms can be administered daily, or more than once a day, such as twice or thrice daily. Alternatively dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician.

[00163] An embodiment of the invention also encompasses prodrugs of a compound of the invention which on administration undergo chemical conversion by metabolic or other physiological processes before becoming active pharmacological substances. Conversion by metabolic or other physiological processes includes without limitation enzymatic (e.g, specific enzymatically catalyzed) and non-enzymatic (e.g., general or specific acid or base induced) chemical transformation of the prodrug into the active pharmacological substance. In general, such prodrugs will be functional derivatives of a compound of the invention which are readily convertible in vivo into a compound of the invention. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, ed. H. Bundgaard, Elsevier, 1985.

[00164] In another embodiment, there are provided methods of making a composition of a compound described herein including formulating a compound of the invention with a pharmaceutically acceptable carrier or diluent. In some embodiments, the pharmaceutically acceptable carrier or diluent is suitable for oral administration. In some such embodiments, the methods can further include the step of formulating the composition into a tablet or capsule. In other embodiments, the pharmaceutically acceptable carrier or diluent is suitable for parenteral administration. In some such embodiments, the methods further include the step of lyophilizing the composition to form a lyophilized preparation.

[00165] The compounds of the invention can be used therapeutically in combination with i) one or more other GLP-1 modulators and/or ii) one or more other types of therapeutic agents which can be administered orally in the same dosage form, in a separate oral dosage form (e.g., sequentially or non-sequentially) or by injection together or separately (e.g., sequentially or non-sequentially). Examples of combination therapeutic agents include Sitagliptin (MK-0431 , Januvia) an oral antihyperglycemic (antidiabetic drug) of the dipeptidyl peptidase-4 (DPP-4) inhibitor class and Exenatide (Byetta) an incretin mimetic.

[00166] Combinations of the invention include mixtures of compounds from (a) and (b) in a single formulation and compounds from (a) and (b) as separate formulations. Some combinations of the invention can be packaged as separate formulations in a kit. In some embodiments, two or more compounds from (b) are formulated together while a compound of the invention is formulated separately.

[00167] The dosages and formulations for the other agents to be employed, where applicable, will be as set out in the latest edition of the Physicians' Desk Reference, incorporated herein by reference.

METHODS OF TREATMENT

[00168] In certain embodiments, the present invention encompasses compounds that bind with high affinity and specificity to the GLP-1 receptor in an agonist manner or as an activator or a potentiator. In certain embodiments a compound of the invention acts as a positive allosteric modulator of GLP-1 receptor.

[00169] In certain embodiments, the present invention provides a method for activating, potentiating, or agonizing (i.e., to have an agonic effect, to act as an agonist) a GLP-1 receptor, with a compound of the invention. The method involves contacting the receptor with a suitable concentration of an inventive compound to bring about activation of the receptor. The contacting can take place in vitro, for example in carrying out an assay to determine the GLP-1 receptor activation activity of an inventive compound undergoing experimentation related to a submission for regulatory approval.

[00170] In certain embodiments, the method for activating an GLP-1 receptor, can also be carried out in vivo, that is, within the living body of a mammal, such as a human patient or a test animal. The inventive compound can be supplied to the living organism via one of the routes as described above, e.g. , orally, or can be provided locally within the body tissues. In the presence of the inventive compound, activation of the receptor takes place, and the effect thereof can be studied.

[00171] An embodiment of the present invention provides a method of treatment of a malcondition in a patient for which activation of an GLP-1 receptor is medically indicated, wherein the patient is administered the inventive compound in a dosage, at a frequency, and for a duration to produce a beneficial effect on the patient. The inventive compound can be administered by any suitable means, examples of which are described above.

[00172] In certain embodiments, the present invention is directed to compounds adapted to act as modulators or potentiators of Class B GPCRs. These compounds may have activity on their own or in the presence of receptor ligands. Receptors include incretin peptides including GLP-1 (7-36) and GLP-1 (9-36). [00173] Methods of treatments provided by the invention include administration of a compound of the invention, alone or in combination with another

pharmacologically active agent to a subject or patient having a malcondition for which activation, potentiation or agonism of a glucagon-like peptide 1 receptor is medically indicated such as type I diabetes, type II diabetes, gestational diabetes, obesity, excessive appetite, insufficient satiety, or metabolic disorder.

PREPARATION OF CERTAIN EMBODIMENTS

General Synthetic Methods for Preparing Compounds

[00174] Molecular embodiments of the present invention can be synthesized using standard synthetic techniques known to those of skill in the art. Compounds of the present invention can be synthesized using the general synthetic procedures set forth in Schemes 1-21.

Scheme 1:

[00175] Reagents: PGi and PG 2 are protecting groups: (i) If Z = CO then Amide coupling with acid-Cl: DIEA, DCM or amide coupling with acid: EDC, HOBt, DMF or HATU, DMF; If Z=S0 2 , then coupling with sulfonyl-chloride: DIEA or NEt 3 , DCM or DMF; (ii) Deprotection of PGi e.g., methyl ester deprotection: LiOH, dioxane, water.

[00176] The other enantiomer can be prepared in a similar manner using Scheme

1. Scheme 2:

[00177] Reagents: (i) Zn(CN) 2 , Pd(PPh 3 ) 4 , NMP; (ii) NH 2 OH HCl, TEA, EtOH.

Scheme 3:

[00178] Reagents: PG is a protecting group (i) EDC, HOBt, DMF then heat; (ii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water.

[00179] The other enantiomer can be prepared in a similar manner using Scheme

3.

Scheme 4:

[00180] Reagents: PG is a protecting group (i) NH 2 OH, TEA, water or EtOH; (ii) EDC, HOBt, DMF then heat; (iii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water.

[00181] The other enantiomer can be prepared in a similar manner using Scheme

4. Scheme 5:

[00182] Reagents: X 1 = O or S; (i) N-Methylmorpholine, isobutyl chloroformate, THF, DMF; (ii) For Xi = oxygen, then 2-Chloro-l,3-dimethylimidazolinium chloride, TEA, DCM; For Xi = sulfur then 2,4-bis(4-methoxyphenyl)-l,3,2,4- dithiadiphosphetane 2,4-disulfide, THF; (iii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water.

[00183] The other enantiomer can be prepared in a similar manner using Scheme

5.

Scheme 6:

[00184] Reagents: PG is a protecting group (i) For Z=CO, then Amide coupling with acid-Cl: DIEA, DCM or amide coupling with acid: EDC, HOBt, DMF or HATU, DMF; For Z=S0 2 , then coupling with sulfonyl chloride DIEA or NEt 3 , DCM or DMF (ii) DIEA, 1,1,1 -trifluoro-N-phenyl-N-((trifluoromethyl)sulfonyl)methanesulf onamide, DCM; (iii) KOAc, bis-pinacolatoborane, PdCl 2 (dppf) or Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (iv) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (v) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (vi) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water. Each occurance of XA and X B is independently CR 4 or N.

[00185] The other enantiomer can be prepared in a similar manner using Scheme

6. Scheme 7:

[00186] Reagents: PG is a protecting group; (i) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (ii) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (iii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water. Each occurance of XA and X B is independently CP or N.

[00187] The other enantiomer can be prepared in a similar manner using Scheme

7.

Scheme 8:

[00188] Reagents: PG is a protecting group (i) DIEA or TEA, acetonitrile; (ii) Acetamide, boron trifluoride etherate, DCM; (iii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water.

[00189] The other enantiomer can be prepared in a similar manner using Scheme

8.

Scheme 9:

[00190] Reagents: PG is a protecting group (i) Boron trifluoride etherate, acetamide, DCM; (ii) Zn, I 2 , Pd 2 (dba) 3 , dicyclohexyl(2',6'-dimethoxy-[l, -biphenyl]-2- yl)phosphine, DMF; (iii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water.

[00191] The other enantiomer can be prepared in a similar manner using Scheme

9.

Scheme 10:

[00192] Reagents: PG is a protecting group (i) Pd(dppf)Cl 2 , Na 2 C0 3 , THF,

MeCN, water; (ii) Deprotection e.g., methyl ester deprotection: NaOH, MeOH, water.

The other enantiomer can be prepared in a similar manner using Scheme 10. Scheme 11:

[00193] Reagents: PG is a protecting group (i) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (ii) Deprotection e.g., tert- vXy\ ester deprotection: DCM, TFA

[00194] The other enantiomer can be prepared in a similar manner using Scheme

Scheme 12:

[00195] Reagents: PGi, PG 2 , and PG 3 are protecting groups (i) Zn, I 2 , Pd 2 (dba) 3 , dicyclohexyl(2',6'-dimethoxy-[l, -biphenyl]-2-yl)phosphine, DMF; (ii) Deprotection of PG 2 e.g., tert-butyl carbonate and PG 3 , e.g., SEM deprotection: DCM, TFA; (iii) If Z=CO then coupling with acd: base (DIEA, TEA, or NMM), coupling reagents (EDC, HOBt or DCC, HOBt, or DCC, DMAP or HATU), solvent (DMF or DCM); If Z=S0 2 then coupling with sulfonyl-Cl: DIEA or TEA, DCM or DMF; (iv) Deprotection of PG, e.g., tert-bvXy\ ester deprotection: DCM, TFA

[00196] The other enantiomer can be prepared in a similar manner using Scheme

12.

Scheme 13:

[00197] Reagents: PGi and PG 2 are protecting groups (i) 2,4-bis(4- phenoxyphenyl)-l,3,2,4-dithiadiphosphetane 2,4-disulfide, DME, THF; (ii)

isopropanol; (iii) Zn, I 2 , Pd 2 (dba) 3 , dicyclohexyl(2',6'-dimethoxy-[l,l'-biphenyl]-2- yl)phosphine, DMF; (iv) Deprotection of PG 2 , e.g., tert-butyl ester deprotection: DCM, TFA; (v) If Z=CO then coupling with acd: base (DIEA, TEA, or NMM), coupling reagents (EDC, HOBt or DCC, HOBt, or DCC, DMAP or HATU), solvent (DMF or DCM); If Z=S0 2 then coupling with sulfonyl-Cl: DIEA or TEA, DCM or DMF;

(vi) Deprotection of PGi, e.g., methyl ester deprotection: NaOH, MeOH, water.

[00198] The other enantiomer can be prepared in a similar manner using Scheme

13.

Scheme 14:

[00199] Reagents: PGi and PG 2 are protecting groups: (i) Zn, I 2 , Pd 2 (dba) 3 , dicyclohexyl(2',6'-dimethoxy-[l, -biphenyl]-2-yl)phosphine, DMF; (ii) Deprotection of PG 2 , e.g., tert-bvXy\ carbonate deprotection: DCM, TFA; (iii) If Z=CO then coupling with acd: base (DIEA, TEA, or NMM), coupling reagents (EDC, HOBt or DCC, HOBt, or DCC, DMAP or HATU), solvent (DMF or DCM); If Z=S0 2 then coupling with sulfonyl-Cl: DIEA or TEA, DCM or DMF; (iv) Deprotection of PGi, e.g., tert- butyl ester deprotection: DCM, TFA.

[00200] The other enantiomer can be prepared in a similar manner using Scheme

14. Scheme 15:

[00201] Reagents: PGi and PG 2 are a protecting group (i) 1,1,3,3- tetramethylguanidine, THF; (ii) H 2 , Dioxane; (iii) Deprotection of PGi e.g., boc-amine deprotection: DCM, TFA; (iv) If Z=CO then coupling with acd: base (DIEA, TEA, or NMM), coupling reagents (EDC, HOBt or DCC, HOBt, or DCC, DMAP or HATU), solvent (DMF or DCM); If Z=S0 2 then coupling with sulfonyl-Cl: DIEA or TEA, DCM or DMF; (v) Deprotection of PG 2 , e.g., tert-butyl ester deprotection: DCM, TFA.

Scheme 16:

[00202] Reagents: PGi and PG 2 are protecting groups (i) 1,1,3,3- tetramethylguanidine, THF; (ii) H 2 , Dioxane; (iii) Deprotection of PGi e.g., boc amine deprotection: DCM, TFA; (iv) If Z=CO then coupling with acid: base (DIEA, TEA, or NMM), coupling reagents (EDC, HOBt or DCC, HOBt, or DCC, DMAP or HATU), solvent (DMF or DCM); If Z=S0 2 then coupling with sulfonyl-Cl: DIEA or TEA, DCM or DMF; (v) Deprotection of PG 2 , e.g., tert- vXy\ ester deprotection: DCM, TFA.

Scheme 17:

[00203] Reagents: PG is a protecting group (i) 3-bromo-5-chloro-l,2,4- thiadiazole, NaHC0 3 , Pd(dppf)Cl 2 , water and THF, ACN or dioxane; (ii) NaHC0 3 , Pd(dppf)Cl 2 , water and THF, ACN or dioxane; (iii) Deprotection of PG, e.g., tert-butyl ester deprotection: DCM, TFA.

[00204] The other enantiomer can be prepared in a similar manner using Scheme

17. Scheme 18:

[00205] Reagents: PG is a protecting group (i) NaO'Bu or Cs 2 C0 3 , Pd(dppf)Cl 2 or Pd 2 (dba) 3 , 2-dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl, water and THF, ACN or dioxane; (ii) Deprotection of PG, e.g., tert-bvXy\ ester deprotection: DCM, TFA.

[00206] The other enantiomer can be prepared in a similar manner using Scheme

18.

Scheme 19:

[00207] Reagents: PG is a protecting group (i) NaO'Bu or Cs 2 C0 3 , Pd 2 (dppf)Cl 2 or Pd 2 (dba) 3 , 2-dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl, water and THF, ACN or dioxane; (ii) Deprotection of PG, e.g., tert- vXy\ ester deprotection: DCM, TFA.

[00208] The other enantiomer can be prepared in a similar manner using Scheme

19. Scheme 20:

[00209] Reagents: PG is a protecting group (i) NaO'Bu or Cs 2 C0 3 , Pd(dppf)Cl 2 or Pd 2 (dba) 3 , 2-dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl, water and THF, ACN or dioxane; (ii) Deprotection of PG, e.g., tert-bvXy\ ester deprotection:

DCM, TFA.

[00210] The other enantiomer can be prepared in a similar manner using Scheme

20.

Scheme 21:

[00211] Reagents: PG is a protecting group (i) (a) where R 2 is NH-(CR a Rb) m - COOH: NH 2 -(CR a R b ) m -COOPG, HATU, DMF then deprotection e.g., tert-butyl ester deprotection: DCM, TFA;(b) where R 2 is NH-S0 2 -R 8 : R 8 S0 2 NH 2 , DCC, DMAP, DCM (c) where R 2 is NR 4 iR4 2 : HNR 4 iR 42, HATU, DMF then deprotection e.g., tert-butyl ester deprotection: DCM, TFA: (d) where R 2 is N(Ri)-(CRaR b )m-CO-N(Ri)- heterocyclyl: HN(Ri)-(CR a R b )m-CO-N(Ri)-heterocyclyl, HATU, DMF then

deprotection e.g., tert-bvXy\ ester deprotection: DCM, TFA; (e) where R 2 is -N(Ri)- (CRaRb) m -CO-N(Ri)(R 7 ): NH 2 -(CR a R b ) m -COOPG, HATU, DMF then deprotection e.g., tert-butyl ester deprotection: DCM, TFA then HN(Ri)(R 7 ), HATU, DMAP, DCM (f) where R 2 is N(Ri)-heterocyclyl: HN(Ri)-heterocyclyl, HATU, DMF then deprotection e.g., tert- vXy\ ester deprotection: DCM, TFA.

[00212] The other enantiomer and diasteroisomer can be prepared in a similar manner using Scheme 21.

Scheme 22:

[00213] Reagents: PGi and PG 2 are protecting groups (i) DIEA, 1,1,1-trifluoro- N-phenyl-N-((trifluoromethyl)sulfonyl)methanesulfonamide, DCM; (ii) KOAc, bis- pinacolatoborane, PdCl 2 (dppf); (iii) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water;

(iv) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (v) Deprotection of PG 2 , eg CBZ: Pd/C, H 2 , EA; (vi) If Z= CO then Amide coupling with acid-Cl: DIEA, DCM or amide coupling with acid: EDC, HOBt, DMF or HATU, DMF; If Z=S0 2 , then coupling with sulfonyl chloride: DIEA or NEt 3 , DCM or DMF; (vii) Deprotection of PGi, e.g., tert- butyl ester deprotection: DCM, TFA. [00214] The other enantiomer can be prepared in a similar manner using Scheme

Scheme 23:

[00215] Reagents: PG is a protecting group (i) Pd(dppf)Cl 2 , Na 2 C0 3 , THF, MeCN, water; (ii) Deprotection of PG, e.g., tert-butyl ester deprotection: DCM, TFA.

[00216] The other enantiomer can be prepared in a similar manner using Scheme

23.

Scheme 24:

[00217] Reagents: PG is a protecting group (i) Zn(CN) 2 , Pd(Ph 3 ) 4 , NMP;

(ii) hydroxylamine, NEt 3 , EtOH; (iii) EDC, HOBt, DMF then heat; (iv) Deprotection of PG, e.g., tert-bvXy\ ester deprotection: DCM, TFA.

[00218] The other enantiomer can be prepared in a similar manner using Scheme

24.

Scheme 25:

[00219] Reagents: PG is a protecting group (i) NH 4 C1, NaN 3 , DMF; (ii) CsC0 3 , or K 2 C0 3 , DMF, acetone or acetonitrile; (iii) Deprotection of PG, e.g., tert- vXy\ ester deprotection: DCM, TFA.

[00220] The other enantiomer can be prepared in a similar manner using Scheme

25.

Scheme 26:

[00221] Reagents: PG is a protecting group (i) sodium tert-butoxide, Pd 2 (dba) 3 , dioxane; (ii) Deprotection of PG, e.g., tert- vXy\ ester deprotection: DCM, TFA.

[00222] The other enantiomer can be prepared in a similar manner using Scheme

26.

[00223] Reagents: PG is a protecting group (i) NaO'Bu or Cs 2 C0 3 , Pd 2 (dppf)Cl 2 or Pd 2 (dba) 3 , 2-dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl, water and THF, ACN or dioxane; (ii) Pd/C, H 2 , EtOH, (iii) Deprotection of PG, e.g., tert-butyl ester deprotection: DCM, TFA.

[00224] The other enantiomer can be prepared in a similar manner using Scheme

27.

EXAMPLES

[00225] The invention is further illustrated by the following examples. The examples below are non-limiting are merely representative of various aspects of the invention. General Methods

NMR spectra

[00226] 1H NMR (400 MHz) and 13 C NMR (100 MHz) were obtained in solution of deuteriochloroform (CDCI3) or dimethyl sulfoxide (d 6 -DMSO). NMR spectra were processed using MestReNova 6.0.3-5604.

LCMS data

[00227] Mass spectra (LCMS) were obtained using one of 5 systems. System 1: Agilent 1100/6110 HPLC system equipped with a Thompson ODS-A, 100A, 5 μ (50 X 4.6 mm) column using water with 0.1% formic acid as the mobile phase A, and acetonitrile with 0.1% formic acid as the mobile phase B. Method 1: 20-100% mobile phase B over 2.5 min then held at 100% for 2.5 min with a flow rate of 1 mL/min. Method 2: 5% mobile phase B for 1 min, 5-95% over 9 min, then held at 95% for 10 min, with a flow rate of 1 mL/min. Method 3: 20-100% mobile phase B over 2.5 min then held at 100% for 4.5 min with a flow rate of 1 mL/min. System 2: Agilent 1200 LCMS equipped with an Agilent Zorbax Extend RRHT 1.8 μιη (4.6 x 30 mm) column using water with 0.1% formic acid as mobile phase A and acetonitrile with 0.1% formic acid as mobile phase B. Method 4: 5-95% mobile phase B over 3.0 min with a flow rate of 2.5 mL/min, then held at 95% for 0.5 min with a flow rate of 4.5 mL/min. Method 5: 5-95% mobile phase B over 14 min with a flow rate of 2.5 mL/min, then held at 95% for 0.5 min with an flow rate of 4.5 mL/min. System 3: Waters Fractionlynx LCMS system equipped with an Agilent Zorbax Extend RRHT 1.8 μιη, (4.6 x 30 mm) column using water with 0.1% formic acid as mobile phase A and acetonitrile with 0.1% formic acid as mobile phase B. Method 6: 5-95% mobile phase B over 3.0 min with a flow rate of 2.5 mL/min, then held at 95% for 0.5 min with a flow rate of 4.5 mL/min. Method 7: 5-95% mobile phase B over 14 min with a flow rate of 2.5 mL/min, then held at 95% for 0.5 min with an flow rate of 4.5 mL/min. System 4: Agilent 1260 LCMS equipped with an Agilent Zorbax Extend RRHT 1.8 μιη (4.6 x 30 mm) column using water with 0.1% formic acid as mobile phase A and acetonitrile with 0.1% formic acid as mobile phase B. Method 8: 5-95% mobile phase B over 3.0 min with a flow rate of 2.5 mL/min, then held at 95% for 0.5 min with a flow rate of 4.5 mL/min. Method 9: 5- 95% mobile phase B over 14 min with a flow rate of 2.5 mL/min, then held at 95% for 0.5 min with an flow rate of 4.5 mL/min. System 5: Agilent 1260 LCMS equipped with a Waters Xselect CSH CI 8 3.5 μιη (4.6x50 mm) column using water with 0.1% formic acid as mobile phase A and acetonitrile with 0.1% formic acid as mobile phase B. Method 10: The gradient was 5-95% mobile phase B over 13.0 min with a flow rate of 2.5 mL/min, then held at 95% for 1.0 min with an flow rate of 4.5 mL/min. Method 11: The gradient was 5-95% mobile phase B over 3.0 min with a flow rate of 2.5 mL/min, then held at 95% for 0.6 min with an flow rate of 4.5 mL/min.

Reaction Conditions and Abbreviations

[00228] Pyridine, dichloromethane (DCM), tetrahydrofuran (THF), and toluene used in the procedures were from Aldrich Sure-Seal bottles or Acros AcroSeal dry solvent and kept under nitrogen (N 2 ). All reactions were stirred magnetically and temperatures are external reaction temperatures. Compounds with salt-able centers were presumed to be the trifluoroacetic acid (TFA) salt. The following abbreviations are used: ethyl acetate (EA), l-methy-2-pyrrolidinone (NMP), triethylamine (TEA), N- hydroxybenzotriazole (HOBt), l-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC), N,N-dimethylformamide (DMF), dimethyl acetamide (DMA), Di-tert-butyl dicarbonate (Boc 2 0), N,N-Diisopropylethylamine (DIEA), acetic acid (AcOH), hydrochloric acid (HC1), O-(7-azabenzotriazol-l-yl)-N,N,N',N'- tetramethyluronium hexafluorophosphate (HATU), 4-dimethylaminopyridine (DMAP), tert-butanol (t-BuOH), sodium hydride (NaH), sodium triacetoxyborohydride

(Na(OAc) 3 BH), ethanol (EtOH), methanol (MeOH), acetonitrile (ACN).

Purifications

[00229] Chromatographies were carried out using either a Combiflash Rf flash purification system (Teledyne Isco) equipped with Redisep (Teledyne Isco), Telos (Kinesis) or GraceResolv (Grace Davison Discovery Sciences) silica gel (Si0 2 ) columns. Preparative HPLC purifications were performed using one of two systems. System 1 : Varian Pro Star/Prep Star system equipped with a Waters SunFire Prep CI 8 OBD, 5 μιη (19 x 150 mm) column using water containing 0.05% trifluoroacetic acid as mobile phase A, and acetonitrile with 0.05% trifluoroacetic acid as mobile phase B. The gradient was 40-95% mobile phase B over 10 min, held at 95% for 5-10 min, and then return to 40% over 2 min with flow rate of 18 mL/min. Fractions were collected using a Varian Prostar fraction collector by UV detection at 254 nm and were evaporated using a Savant SpeedVac Plus vacuum pump or a Genevac EZ-2. System 2: Waters Fractionlynx system equipped with an Agilent Prep-C18, 5 μιη (21.2 x 50 mm) column using water containing 0.1% formic acid as mobile phase A, and acetonitrile with 0.1% formic acid as mobile phase B. The gradient was 45-95% mobile phase B over 7.5 min, held at 95% for 1 min, and then returned to 45% over 1.5 min with a flow rate of 28 mL/min. Fractions were collected by UV detection at 254 nm or by mass and evaporated using a Genevac EZ-2.

Chiral Methods

[00230] Enantiomeric excess was determined by integration of peaks that were separated on a Diacel Chiralpak IA, 4.6 x 250 mm column, 5 μιη particle size. The solvents used were "Solvent A": 4: 1 (hexanes with 0.2% TFA): DCM, and "Solvent B": EtOH. The flow rate was held at 1.0 mL / min with the following gradient: Increase Solvent B from 2-10% over 30 min, hold Solvent B at 10% for 15 min.

Experimental Procedures

General Procedures

General Procedure 1: Preparation ofNitriles.

[00231] A stirred a solution of bromide or triflate (1 eq), zinc cyanide (2 eq) and tetrakis (triphenylphosphine) palladium (1 - 5 mol%) in dry NMP (0.5 - 1 M) was degassed with N 2 . The reaction was heated to 100°C for 18 h while stirring under N 2 . The reaction mixture was cooled and poured into water and DCM. The solid material was removed by filtration and the filtrate was extracted with water. The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by

chromatography.

General Procedure 2: Preparation of Amidoximes.

[00232] To a stirring solution of nitrile (1 eq) in EtOH was added hydroxylamine (50% solution in H 2 0, 5 eq) and TEA (1.1 eq). The mixture was heated for 2 - 12 h at 80 - 85°C then concentrated. The resulting solid was dissolved in EA, washed with water, then dried with Na 2 S0 4 , concentrated and used without further purification. Alternatively, to a stirring solution of nitrile (1 eq) and TEA (2-3 eq) in DMF or EtOH was added hydroxylamine hydrochloride (2-3 eq). The mixture was stirred at RT up to 80°C for up to 24 h then concentrated. The resulting solid was dissolved in EA or DCM, washed with water or brine, then dried with Na 2 S0 4 , concentrated, and used without further purification.

General Procedure 3: Preparation of Amides via Acid Chlorides.

[00233] To a solution of amine (1 eq) and base (either DIE A or TEA) (2 - 3 eq) in DCM (0.06 - 0.30 M) was treated with the appropriate acid chloride (1.0 - 1.5 eq). The reaction mixture was stirred until the reaction was complete. The reaction was diluted with DCM and washed with saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and concentrated. The product was purified by chromatography. Alternatively, the crude reaction mixture can be carried on to the next step without further purification.

General Procedure 4: Hydrolysis of Esters to Acids.

[00234] To a stirring solution of ester (1 eq) in THF or dioxane and water, was added NaOH or LiOH (1 - 3 eq). The reaction mixture was stirred at up to 60°C for up to 18 h. The reaction mixture was neutralized with AcOH or HC1 and either diluted with water or concentrated. If the reaction mixture was diluted with water, then HC1 was added to acidify the reaction mixture to a pH of approximately 2. The resulting precipitate was isolated by filtration to yield product which can be purified by chromatography, preparative HPLC, or used without purification. If the reaction mixture was concentrated, the crude material was diluted with DCM or EA and washed with brine. The organic layer was concentrated and purified by chromatography or preparative HPLC to give final product. Alternatively, the crude material can be carried forward without purification.

General Procedure 5: Preparation of Oxadiazoles via Acids or Acid Chlorides. Oxadiazoles via Acids:

[00235] To a solution of acid (1 eq) in DMF was added HOBt (2 eq) and EDC (2 eq). After stirring for 2 h, amidoxime (2 eq) was added and the mixture was stirred at RT for up to 12 h. The reaction mixture was then heated to 100°C for up to 12 h.

Alternatively, after stirring at RT, the reaction mixture was diluted with DCM, washed with NaHC0 3 , then dried with Na 2 S0 4 and concentrated. The resulting residue was dissolved in EtOH and heated in a microwave for 35 min at 110°C. The solvent was removed and the final product was purified by preparative HPLC.

[00236] Oxadiazoles via Acid Chlorides: To synthesize oxadiazoles via acid chlorides, dioxanes and DIEA (1.5 eq) were added to a stirred solution of amidoxime (1 eq) followed by an acid chloride (1.1 eq). The reaction mixture was stirred at RT for 30 min then at 120°C for up to 6 h. The reaction mixture was allowed to cool to RT, diluted with EA and washed with brine. The organics were concentrated and the residue purified by chromatography.

General Procedure 6: Removal of tert-Butyl Carbamate.

[00237] A solution of the tert-butyl carbamate (1 eq) in DCM (0.06 M) was treated with TFA (0.16 - 0.33 M). The reaction mixture was stirred at either RT or 30°C until complete. The solvent was removed and the product was purified by

chromatography or preparative HPLC. General Procedure 7: Preparation of Amides via Peptide Coupling.

[00238] A solution of amine (1.0 eq) and base (DIEA, TEA or NMM) (0 - 3.0 eq) in DCM or DMF (0.08 - 0.10 M) was treated with the appropriate carboxylic acid (1.0 - 1.5 eq). To this mixture was added the coupling reagent. The coupling reagent could be HATU (1.05 - 2.5 eq), EDC (1.5 eq) with HOBt (1.5 eq), DCC (1.1 eq) with HOBt (1.1 eq) or DCC (1.5 eq) with DMAP (2.0 eq). The reaction mixture was stirred until the reaction was complete. The reaction was diluted with EA and washed with saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and

concentrated. The product was purified by chromatography or alternatively can be carried on to the next step without further purification.

General Procedure 8: Deprotection of t-Butyl Esters to Acids or Deprotection of Boc-Amines

[00239] A solution of the tert-butyl ester or Boc-amine (1.00 eq) in DCM (0.06 M) was treated with TFA (0.16 - 0.33 M). The reaction mixture was stirred at either RT or 30°C until complete. The solvent was removed and the product was purified by chromatography or preparative HPLC.

General Procedure 9: Formation ofTriflate.

[00240] A solution of the phenol (1.0 eq) in DCM (0.25 M) was treated with 1,1- trifluoro-N-phenyl-N-((trifluoromethyl)sulfonyl)methanesulfo namide (1.1 eq). The reaction mixture was stirred at RT until complete. The reaction was stirred with water and saturated aqueous NaHC0 3 . The organic layers was dried and concentrated. The material was purified by chromatography or alternatively used without purification.

Procedure 10: Palladium-catalyzed Coupling Reactions.

[00241] A solution of boronic acid or boronate ester (1.0 - 1.3 eq), halide (1.0 - 1.3 eq), sodium bicarbonate or sodium carbonate decahydrate (2.0 - 2.5 eq), and Pd(dppf)Cl 2 were combined in THF, acetonitrile, or dioxane (0.1 - 0.2 M) and water (0.25 - 0.50 M). The reaction was heated at 80 to 100°C until complete. The reaction was diluted with EA and washed with saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and concentrated. The product can be purified by

chromatography, preparative HPLC, or carried on to the next step without further purification.

General Procedure 11: Palladium-catalyzed aryl Amidation.

[00242] A solution of aryl bromide or triflate (1.00 eq), sodium tert-butoxide or cesium carbonate (1-2 eq) and amine (1.0-1.5 eq) in dioxane or THF (0.05M) was degassed using N 2 bubbling for 10 min. Pd 2 (dba) 3 (0.10 eq) and 2- dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl (0.15 eq) are added and the reaction mixture was heated for 45-60 min at 100-120°C in a microwave reactor or up to 80°C with conventional heating for up to 18 h. The reaction was diluted with EA and washed with saturated aqueous NaHC0 3 . The organic layer was dried over Na 2 S0 4 and concentrated. The product can be purified by chromatography, preparative HPLC, or carried on to the next step without further purification.

General Procedure 12: Alklation of Phenols, Imidazoles, and Lactams.

[00243] To a solution of a phenolic intermediate in DMF, acetone or ACN (0.1 M) were added the appropriate bromoalkane (1.5 eq) and either CsC0 3 (1.5 -2.0 eq) or K 2 C0 3 (1.5 -2.0 eq). The reaction mixture was heated at 40-70°C for 18 h, then diluted with DCM and washed with H 2 0. The organic layer was dried over Na 2 S0 4 and concentrated. The product can be purified by chromatography, preparative HPLC, or carried on to the next step without further purification.

General Procedure 13: Sulfonate or Sulfonamide formation.

[00244] To a solution of alcohol or amine in DCM (0.02 M) was added the sulfonyl chloride (2 eq) and triethylamine (3 eq). The reaction was stirred at RT until complete. The reaction was diluted with DCM and washed with saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and concentrated. The product can be purified by chromatography, preparative HPLC, or carried on to the next step without further purification.

General Procedure 14: Reduction of Aryl Nitro to an Aryl Amine.

[00245] To a stirring solution of aryl nitro (1 eq) in THF purged with N 2 was added palladium on carbon. The reaction mixture was subjected to an H 2 atmosphere for up to 4 h. The reaction mixture can be filtered over a pad of celite and solvent concentrated. The crude material was carried forward without further purification.

General Procedure 15: Preparation of a Secondary or Tertiary Amine via Reductive Amination.

[00246] To a stirring solution of aldehyde or ketone (0.9-1.0 eq) in DCM or 1,2- dichloroethane or THF was added an amine (0.9-1.1 eq). After stirring at RT for up to 2 h, one drop of acetic acid (optional) was added followed by sodium

triacetoxyborohydride (1.5-2.0 eq) and the reaction mixture was stirred overnight. In some cases it is necessary to filter the reaction mixture, redissolve and add additional reducing agent to drive the reaction to completion. The crude reaction mixture was quenched with NaHC0 3 and stirred for 5 min. The aqueous layer was extracted with DCM and the organic layer was dried over MgS0 4 and concentrated. The final product was isolated by chromatography.

General Procedure 16: Preparation of 2-Iodopyrimidines

[00247] To a stirred solution of a 2-chloro pyrimidine (1 eq) in 57% aqueous hydrogen iodide (1 mL) was added sodium iodide (2 eq). The reaction mixture was stirred at ambient temperature until the starting material was consumed. The reaction mixture was quenched with NaHC0 3 (5 mL) then extracted with EA (3 x 5 mL). The combined organic layer was washed with brine (10 mL), dried over MgS0 4 and concentrated. The crude product was used in the subsequent step without purification. General Procedure 17. Preparation of 2-Iodopyridines

[00248] To a stirred solution of a 2-chloropyridine (1 eq) in acetonitrile (2 mL) was added sodium iodide (6 eq). The reaction mixture was heated to 40°C and acetyl chloride (0.6 eq) was added. The reaction mixture was stirred until the staring material was consumed. The reaction was quenched with NaHC0 3 (5 mL) and extracted with EA (3 x 5 mL). The combined organic layer was washed with brine (10 mL), dried over MgS0 4 and concentrated. The crude product was used in the subsequent step without purification.

Synthesis of Representative Compounds

4-(heptyloxy)benzonitrile

[00249] Prepared using General Procedure 1: A stirred a solution of l-bromo-4- (heptyloxy)benzene (2.0 g, 7.37 mmol), zinc cyanide (1.73 g, 14.74 mmol) and tetrakis (triphenylphosphine) palladium (76.12 mg, 0.07 mol) in dry NMP (20 mL) was degassed with N 2 . The reaction was heated to 100°C for 18 h while stirring under nitrogen. The reaction mixture was cooled and poured into water (100 mL) and DCM (20 mL). The solid material was removed by filtration and the filtrate was extracted with water (3 x 20 mL). The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by chromatography (EA / hexanes) to afford 1.15 g (73%) of 4-(heptyloxy)benzonitrile as a light yellow solid. LCMS-ESI (m/z) calculated for Ci 4 Hi 9 NO: 217.1; found 218.1 [M+H] + , t R = 11.14 min (Method 2). 1H NMR (400 MHz, CDC1 3 ) δ 7.64 - 7.50 (m, 2H), 7.05 - 6.83 (m, 2H), 3.99 (t, J= 6.5 Hz, 2H), 1.89 - 1.69 (m, 2H), 1.58 - 1.12 (m, 8H), 0.90 (dd, J= 9.1, 4.5 Hz, 3H). 13 C NMR (101 MHz CDC1 3 ) δ 162.47, 133.91, 132.78, 132.12, 129.13, 119.31, 115.18, 103.58, 68.41, 31.73, 28.98, 25.89, 22.58, 14.07. (Z)-4-(heptyloxy)-N'-hydroxybenzimidamide

[00250] Prepared using General Procedure 2: To a stirring solution of 4- (heptyloxy)benzonitrile (1.0 g, 4.6 mmol) in EtOH (15 mL) were added hydroxylamine hydrochloride (0.96 g, 13.8 mmol) and TEA (2.22 g, 23.0 mmol). The reaction was heated to 85°C for 2 h. The solvent was removed under reduced pressure and the residue was diluted with water (20 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were concentrated under reduced pressure. The crude material was crystallized from isopropanol (20 mL) to afford 1.05 g (91%) of (Z)-4-(heptyloxy)- N'-hydroxybenzimidamide as a white solid. LCMS-ESI (m/z) calculated for

C14H22N2O2: 250.2; found 251.3 [M+H] + , t R = 1.70 min (Method 1). 1H NMR (400 MHz, CDCI 3 ) δ 9.45 (s, 1H), 7.59 (d, J= 8.6 Hz, 2H), 6.93 (t, J= 14.7 Hz, 2H), 5.82 - 5.48 (m, 2H), 3.97 (t, J= 6.5 Hz, 2H), 1.83 - 1.55 (m, 2H), 1.56 - 1.05 (m, 8H), 0.87 (t, J= 6.7 Hz, 3H). 13 C NMR (101 MHz CDC1 3 ) δ 159.19, 150.53, 126.64, 125.55, 113.87, 67.40, 31.21, 28.62, 28.40, 25.44, 22.02, 13.92.

(S)-methyl 4-(2-amino-3-methoxy-3-oxopropyl)benzoate

[00251 ] To a solution of (S)-2-amino-3-(4-(tert- butoxycarbonyl)phenyl)propanoic acid (500.0 mg, 1.88 mmol) in MeOH (20 mL) at 0°C was slowly added thionyl chloride (447.64 mg, 3.77 mmol). The reaction was stirred for 1 h at 0°C then warmed to RT and stirred for 1 h. The solvent was removed under reduced pressure. The reaction mixture was washed with saturated aqueous NaHCC"3 (20 ml) and extracted with DCM (3 x 10 ml). The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by chromatography (EA / hexanes) to afford 425 mg (95%) of (5)-methyl 4-(2-amino-3-methoxy-3- oxopropyl)benzoate as the HCl salt. LCMS-ESI (m/z) calculated for C 12 H 15 NO 4 : 237.1; found 238.0 [M+H] + , t R =1.01 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 8.55 (s, 3H), 7.94 (d, J= 8.3 Hz, 2H), 7.41(d, J= 8.3 Hz, 2H), 4.37 (t, J= 6.8 Hz, 1H), 3.86 (s, 3H), 3.68 (s, 3H), 3.20 (dd, J= 11.8, 6.8 Hz, 2H).

(S)-methyl 4-(2-(4-(tert-butyl)benzamido)-3-methox -3-oxopropyl)benzoate

[00252] Prepared using General Procedure 3: To the solution of (5)-methyl 4-(2- amino-3-methoxy-3-oxopropyl)benzoate (425.0 mg, 1.79 mmol) in DCM (10 mL) and DIEA (463.0 mg, 3.58 mmol) was added 4-(tert-butyl)benzoyl chloride (556.6 mg, 2.83 mmol) at RT. The reaction was stirred for 2 h and the reaction was partitioned between DCM and saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by chromatography (EA / hexanes) to afford 317 mg (45%) of (5)-methyl 4-(2-(4-(fert-butyl)benzamido)-3-methoxy-3- oxopropyl)benzoate. LCMS-ESI (m/z) calculated for C23H27NO5 : 397.2; found 398.1 [M+H] + , t R =2.31 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 7.97 - 7.75 (m, 2H), 7.67 - 7.51 (m, 2H), 7.46 - 7.26 (m, 2H), 7.14 (d, J= 8.3 Hz, 2H), 6.60 (d, J= 7.4 Hz, 1H), 5.03 (dt, J= 7.4, 5.7 Hz, 1H), 3.82 (s, 3H), 3.68 (s, 3H), 3.28 (dd, J = 13.7, 5.8 Hz, 1H), 3.18 (dd, J = 13.7, 5.5 Hz, 1H), 1.24 (s, 9H). S)-4-(2-(4-(tert-butyl)benzamido)-3-methoxy-3-oxo ropyl)benzoic acid (INT-1)

[00253] Prepared using General Procedure 4: To a stirred solution of (5)-methyl 4-(2-(4-(tert-butyl)benzamido)-3-methoxy-3-oxopropyl)benzoat e (316.6 mg, 0.79 mmol) in dioxane (15 mL) and water (1 mL) at 0°C was added lithium hydroxide monohydrate (93.52 mg, 2.23 mmol). After 2 h, the solution was neutralized with 1 M HC1 to pH 7.0. The mixture was partitioned between DCM (15 mL) and saturated aqueous NaHC0 3 (10 mL). The organic layer was washed with saturated aqueous NaHC0 3 (3 x 10 mL) and brine (10 mL). The organic layer was dried over MgS0 4 and concentrated to afford 208 mg (69%) of (5)-4-(2-(4-(fert-butyl)benzamido)-3-methoxy- 3-oxopropyl)benzoic acid INT-1. LCMS-ESI (m/z) calculated for C22H25NO5 : 383.2; found 384.1 [M+H] + , t R = 2.13 min. (Method 1). 1H NMR (400 MHz, DMSO) δ 12.86 (s, 1H), 8.80 (d, J= 8.0 Hz, 1H), 7.87 - 7.78 (m, 2H), 7.75 - 7.65 (m, 2H), 7.50 - 7.35 (m, 4H), 4.72 (ddd, J= 10.3, 8.0, 5.1 Hz, 1H), 3.65 (s, 3H), 3.28 - 3.05 (m, 2H), 1.29 (s, 9H). 13 C NMR (101 MHz, DMSO) δ 173.00, 167.21, 166.29, 154.39, 143.10, 130.85, 129.34, 129.27, 129.21, 129.03, 127.21, 125.39, 125.10, 53.75, 52.04, 34.64, 30.92, 30.88.

(S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(3-(4-(heptyloxy)phenyl)-l, 2,4- oxadiazol-5- l)phenyl)propanoate

[00254] Prepared using General Procedure 5: To a solution of (S)-4-(2-(4-(tert- butyl)benzamido)-3-methoxy-3-oxopropyl)benzoic acid, INT-1 (10.0 mg, 0.026 mmol) in anhydrous DMF (1 mL) was added HOBt (5.27 mg, 0.39 mmol) and EDC (7.48 mg, 0.39 mmol). After stirring for 2 h, (Z)-4-(heptyloxy)-N'-hydroxybenzimidamide (9.76 mg, 0.39 mmol) was added. The reaction mixture was stirred at RT for 2 h, partitioned between saturated aqueous NaHCC (5 ml) and EA (5 mL), and concentrated under reduced pressure to afford the intermediate (5)-methyl 2-(4-(tert-butyl)benzamido)-3- (4-(((4-(heptyloxy)benzimidamido) oxy)carbonyl) phenyl) propanoate. The

intermediate was dissolved in DMF (ImL) and heated to 100°C for 18 h. The reaction mixture was cooled to RT and partitioned between EA (5 mL) and saturated aqueous NaHC03 (5 mL). The organic layer was extracted with water (2 x 5 mL) and brine (5 mL). The organic layer was dried over MgS0 4 and concentrated. The brown oil was purified by preparative HPLC to afford 4.5 mg (29%) of (5)-methyl 2-(4-(tert- butyl)benzamido)-3-(4-(3-(4-(heptyloxy)phenyl)-l,2,4-oxadiaz ol-5-yl) phenyl) propanoate. LCMS-ESI (m/z) calculated for C 36 FL 3 N 3 O 5 : 597.3; no m/z observed, fa = 12.75 min (Method 2). 1H NMR (400 MHz, DMSO) δ 8.85 (d, J= 8.0 Hz, 1H), 8.09 (d, J= 8.3 Hz, 2H), 8.00 (d, J= 8.9 Hz, 2H), 7.74 (d, J= 8.5 Hz, 2H), 7.59 (d, J= 8.4 Hz, 2H), 7.48 (d, J= 8.6 Hz, 2H), 7.12 (d, J= 8.9 Hz, 2H), 4.87 - 4.56 (m, 1H), 4.06 (t, J = 6.5 Hz, 2H), 3.67 (s, 3H), 3.32 - 3.13 (m, 4H), 1.74 (dd, J= 14.2, 6.5 Hz, 2H), 1.51 - 1.37 (m, 2H), 1.33 (s, 4H), 1.26 (d, J = 20.2 Hz, 9H), 0.88 (t, J = 6.9 Hz, 3H). 13 C NMR (101 MHz, DMSO) δ 175.00, 171.91, 167.89, 166.27, 161.21, 154.37, 143.68, 130.78, 130.30, 128.76, 127.80, 127.18, 125.07, 121.69, 118.21, 115.07, 67.72, 53.61, 52.05, 36.15, 34.60, 31.20, 30.87, 28.54, 28.39, 25.40, 22.02, 13.93.

(S)-2-(4-(tert-butyl)benzamido)-3-(4-(3-(4-(heptyloxy)phe nyl)-l,2,4-oxadiazol- 5- l)phenyl)propanoic acid (Compound 1)

[00255] Prepared using General Procedure 4: To a solution of (5)-methyl 2-(4- (tert-butyl)benzamido)-3-(4-(3-(4-(heptyloxy)phenyl)-l,2,4-o xadiazol-5-yl)phenyl) propanoate (4.52 mg, 0.008 mmol) in MeOH (2 mL) was added of 1 N NaOH (1 mL). The reaction mixture was stirred at 50°C for 3 h. The resulting mixture was purified by preparative HPLC to afford 0.36 mg (8%) of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(3- (4-(heptyloxy)phenyl)-l,2,4-oxadiazol-5-yl)phenyl) propanoic acid. LCMS-ESI (m/z) calculated for C35H41N3O5 : 583.7; no m/z observed, fa = 12.59 min {Method 2).

(S)-methyl 2-amino-3-(4-c anophenyl)propanoate

[00256] To a solution of (5)-2-((fert-butoxycarbonyl)amino)-3-(4- cyanophenyl)propanoic acid (1.0 g, 3.44 mmol) in MeOH (20 mL) at 0°C was slowly added thionyl chloride (818.1 mg, 6.89 mmol) over 1 h. The reaction was warmed to RT and stirred for 1 h. The solvent was removed under reduced pressure. The reaction mixture was washed with saturated aqueous NaHC0 3 (20 ml) and extracted with DCM (3 x 10 ml). The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by chromatography (EA / hexanes) to afford 789 mg (97%) of (S)- methyl 2-amino-3-(4-cyanophenyl)propanoate as the HC1 salt. LCMS-ESI (m/z) calculated for C11H12N2O2: 204.1; found 205.0 [M+H] + , t R = 3.25 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 8.69 (s, 3H), 7.83 (d, J= 8.3 Hz, 2H), 7.51 (t, J= 8.8 Hz, 2H), 4.37 (t, J= 6.7 Hz, 1H), 3.68 (s, 3H), 3.23 (qd, J= 14.4, 7.7 Hz, 2H).

(S)-meth l 2-(4-(tert-butyl)benzamido)-3-(4-cyanophenyl)propanoate

[00257] Prepared using General Procedure 3: To the solution of (S)-methyl 2- amino-3-(4-cyanophenyl)propanoate (789.2 mg, 3.32 mmol) in DCM (15 mL) and DIEA (1.29 g, 9.96 mmol) was added 4-(tert-butyl)benzoyl chloride (981.3 mg, 4.99 mmol) at RT. The reaction was stirred for 2 h and the reaction was partitioned between DCM and saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by chromatography (EA / hexanes) to afford 1.06 g (88%) of (5)-methyl 2-(4-(tert-butyl)benzamido)-3-(4- cyanophenyl)propanoate. LCMS-ESI (m/z) calculated for C 22 H 24 N 2 0 3 : 364.2; found 365.3 [M+H] + , t R = 3.55 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 8.81 (d, J = 8.0 Hz, 1H), 7.85 - 7.60 (m, 4H), 7.49 (dd, J= 15.1, 8.4 Hz, 4H), 4.85 - 4.60 (m, 1H), 3.65 (s, 3H), 3.30 - 3.23 (m, 1H), 3.18 (dd, J= 13.7, 10.6 Hz, 1H), 1.29 (s, 9H). (S,Z)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(N'-hydroxycarbamimidoyl) phen l) propanoate (INT-2)

[00258] Prepared using General Procedure 2: To a stirring solution (5)-methyl 2- (4-(tert-butyl)benzamido)-3-(4-cyanophenyl)propanoate (1.0 g, 2.74 mmol) in EtOH (15 mL) were added hydroxylamine hydrochloride (572.2 mg, 8.22 mmol) and TEA (1.38 g, 13.7 mmol). The reaction was heated to 85°C for 2 h. The solvent was removed under reduced pressure and the residue was diluted with water (20 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were concentrated under reduced pressure. The crude material was crystallized from isopropanol (20 mL) to afford 1.04 g (95%) of (£Z)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(N'- hydroxycarbamimidoyl)phenyl)propanoate INT-2 as a white solid. LCMS-ESI (m/z): calcd for: C22H27N3O4, 397.2; found 398.1 [M+l] + , t R = 2.26 min (Method 1). 1H NMR (400 MHz, DMSO) δ 10.19 (s, 1H), 9.57 (s, 1H), 8.78 (d, J= 7.9 Hz, 1H), 7.74 (d, J= 8.4 Hz, 2H), 7.58 (d, J= 8.2 Hz, 2H), 7.48 (d, J= 8.4 Hz, 2H), 7.30 (d, J= 8.3 Hz, 2H), 4.79 - 4.49 (m, 1H), 3.65 (s, 3H), 3.15 (dt, J = 13.6, 6.0 Hz, 2H), 1.75 (d, J = 13.6 Hz, 1H), 1.29 (s, 9H).

(S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)- 1,2,4- oxadiazol-3- l)phenyl)propanoate

[00259] Prepared using General Procedure 5: To a solution of 4- (heptyloxy)benzoic acid (400.0 mg, 1.54 mmol) in anhydrous DMF (6 mL) were added HOBt (312.3 mg, 2.31 mmol) and EDC (442.75 mg, 2.31 mmol). After stirring for 2 h, (5 * ,Z)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(N'-hydroxycarbamimidoyl)ph enyl)- propanoate, INT-2 (673.3 mg, 1.69 mmol) was added. The reaction mixture was stirred at RT for 2 h, partitioned between saturated aqueous NaHC0 3 (15 mL) and EA (15 mL), and concentrated under reduced pressure to afford the intermediate (5)-methyl 2- (4-(tert-butyl) benzamido)-3-(4-(N-((4-(heptyloxy) benzoyl)oxy)

carbamimidoyl)phenyl)propanoate. The intermediate was dissolved in DMF (10 mL) and heated to 100°C for 18 h. The reaction mixture was cooled to RT and partitioned between EA (10 mL) and saturated aqueous NaHC0 3 (50 mL). The organic layer was extracted with water (2 x 10 mL) and brine (10 mL). The organic layer was dried over MgS0 4 and concentrated. The brown oil was purified by chromatography (EA / hexanes) to afford 710 mg (77%) of (5)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(5-(4- (heptyloxy)phenyl)-l,2,4-oxadiazol-3-yl)phenyl)-propanoate as a white solid. LCMS- ESI (m/z) calculated for C 3 6H4 3 N 3 0 5 : 597.3; no m/z observed, fa = 12.80 min {Method 2). 1H NMR (400 MHz, DMSO) δ 8.84 (d, J= 8.0 Hz, 1H), 8.08 (t, J= 17.2 Hz, 2H), 7.97 (dd, J= 18.2, 8.5 Hz, 2H), 7.74 (d, J= 8.5 Hz, 2H), 7.50 (dd, J= 18.6, 8.3 Hz, 4H),7.18 (d, J= 8.9 Hz, 2H), 4.85 - 4.63 (m, 1H), 4.09 (dd, J= 13.8, 7.3 Hz, 2H),3.67 (s, 3H), 3.24 (ddd, J= 23.8, 15.7, 7.3 Hz, 4H), 2.08 (s, 4H), 1.74 (dd, J= 14.1, 6.9 Hz, 2H), 1.42 (dd, J= 13.6, 6.3 Hz, 2H), 1.30 (d, J= 14.5 Hz, 9H), 0.88 (t, J= 6.8 Hz, 3H). 13 C NMR (101 MHz, DMSO) δ 174.05, 170.87, 133.81, 165.14, 161.43, 153.21, 140.51, 129.70, 128.85, 128.78, 126.06, 125.84, 123.93, 123.39, 114.36, 114.25, 66.86, 52.66, 50.88, 34.32, 33.47, 30.06, 29.74, 27.33, 27.24, 24.23, 20.89, 12.80. (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl )-l,2,4-oxadiazol- 3- l)phenyl)propanoic acid (Compound 2)

[00260] Prepared using General Procedure 4: To a solution of (5)-methyl 2-(4- (tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l,2,4-o xadiazol-3-yl)phenyl) propanoate (710.0 mg, 1.19 mmol) in MeOH (20 mL) was added 1 N NaOH (10 mL). The reaction mixture was stirred at 50°C for 3 h. The resulting mixture was purified by chromatography (DCM / MeOH) to afford 218 mg (31%) of (5)-2-(4-(tert- butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l,2,4-oxadiaz ol-3- yl)phenyl)propanoic acid as a white solid. LCMS-ESI (m/z) calculated for C35H41N3O5 : 583.3; no m z observed, t R = 12.16 min (Method 2). 1H NMR (400 MHz, DMSO) δ

8.69 (d, J= 8.3 Hz, 1H), 8.16 - 8.02 (m, 2H), 7.98 (d, J= 8.3 Hz, 2H), 7.74 (d, J= 8.5 Hz, 2H), 7.53 (d, J= 8.3 Hz, 2H), 7.47 (d, J= 8.5 Hz, 2H), 7.18 (d, J= 9.0 Hz, 2H),

4.70 (ddd, J= 10.8, 8.4, 4.5 Hz, 1H), 4.09 (t, J= 6.5 Hz, 2H), 3.30 (dd, J= 13.8, 4.2 Hz, 1H), 3.17 (dd, J= 13.8, 10.7 Hz, 1H), 1.74 (dd, J= 14.5, 6.7 Hz, 2H), 1.42 (dd, J = 13.8, 6.1 Hz, 2H), 1.37 - 1.14 (m, 14H), 0.87 (t, J= 6.9 Hz, 3H). 13 C NMR (101 MHz, DMSO) δ 175.16, 173.00, 167.96, 166.19, 162.55, 154.18, 142.11, 131.08, 129.95, 129.89, 127.14, 126.92, 125.01, 124.39, 115.49, 115.37, 67.98, 53.72, 36.19, 34.58, 31.19, 30.89, 28.46, 28.37, 25.36, 22.01, 13.92.

[00261] Compounds 3 - 11 and 13 - 61 were prepared from ( Z)-methyl 2-(4- (tert-butyl)benzamido)-3-(4-(N'-hydroxycarbamimidoyl)phenyl) propanoate INT-2 using General Procedures 5 and 4 sequentially. [00262] Compounds 62 - 66 were prepared from (S,Z)-mst y\ 2-(4-(tert- butyl)benzamido)-3 -(4-(N'-hydroxycarbamimidoyl)phenyl)propanoate INT-2 using General Procedures 5, 6, and 4 sequentially.

(S)-2-(2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy) phenyl)- 1,2,4- oxadiazol-3-yl)phenyl)propanamido)acetic acid (Compound 67)

[00263] Prepared using General Procedures 7 and 8: To a solution of (S)-2-(4- (tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l,2,4-o xadiazol-3-yl)phenyl) propanoic acid, Compound 2 (10.0 mg, 0.017 mmol) in anhydrous DMF (1 mL) was added HOBt (3.52 mg, 0.027 mmol) and EDCI (4.88 mg, 0.027 mmol) at RT. After 2 h, tert-butyl 2-aminoacetate (3.49 mg, 0.027 mmol) was added and the reaction mixture stirred at RT for 2 h. LCMS analysis showed complete conversion to the intermediate. The reaction mixture was partitioned between NaHCOs aqueous (5 ml) and DCM (1 mL), the organic layer was collected and concentrated by vacuum and then was re- dissolved in 1 mL of DCM and 0.1 mL of TFA. The mixture was heated to 30°C for 3 h. The final compound was purified by HPLC to afford 9.6 mg (88%) of (S)-2-(2-(4- (tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l,2,4-o xadiazol-3- yl)phenyl)propanamido)acetic acid. LCMS-ESI (m/z) calculated for C37H44N4O6:

640.3; no m/z observed, t R = 11.51 min (Method 2). 1H NMR (400 MHz, DMSO) δ: 8.60 (d, J= 8.4 Hz, 1H), 8.47 (t, J= 5.7 Hz, 1H), 8.10 (d, J= 8.8 Hz, 2H), 7.96 (d, J = 8.2 Hz, 2H), 7.75 (d, J= 8.4 Hz, 2H), 7.57 (d, J= 8.0 Hz, 2H), 7.45 (d, J= 8.4 Hz, 2H), 7.17 (d, J= 8.8 Hz, 2H), 4.83 (d, J= 8.1 Hz, 1H), 4.09 (t, J= 6.4 Hz, 2H), 3.94 - 3.69 (m, 2H), 3.34 (s, 2H), 3.26 (d, J= 13.5 Hz, 1H), 3.15 - 3.01 (m, 1H), 1.83 - 1.65 (m, 2H), 1.50 - 1.15 (m, 16H), 0.87 (t, J= 6.7 Hz, 3H). C NMR (101 MHz, DMSO) δ: 175.12, 171.58, 171.13, 167.99, 166.02, 162.54, 154.10, 142.44, 131.16, 130.02, 129.89, 127.23, 126.81, 124.91, 124.25, 115.50, 115.36, 67.97, 54.23, 40.10, 37.12, 34.57, 31.19, 30.88, 28.46, 28.37, 25.36, 22.02, 13.93.

[00264] Compound 68 was prepared from Compound 5 using General

Procedures 7, and 8 sequentially.

[00265] Compounds 69 and 70 were prepared from (S,Z)-methyl 2-(4-(tert- butyl)benzamido)-3 -(4-(N'-hydroxycarbamimidoyl)phenyl)propanoate Compound 2 using General Procedures 7, and 8 sequentially.

[00266] Compounds 71 and 72 were prepared from Methyl 2-amino-2-(4- bromophenyl)acetate hydrochloride using General Procedures 7, 1, 2, 5, and 4 sequentially.

[00267] Compounds 73 and 74 were prepared from (S)-methyl 2-amino-4-(4- hydroxyphenyl)butanoate hydrobromide using General Procedures 7, 9, 1, 2, 5, and 4 sequentially.

[00268] Compound 75 was prepared from (5)-methyl 3-amino-4-(4- hydroxyphenyl)butanoate hydrochloride using General Procedures 7, 9, 1, 2, 5, and 4 sequentially.

4-(heptyloxy)benzohydrazide

[00269] To a stirred solution of 4-(heptyloxy)benzoic acid (679 mg, 2.87 mmol) in THF (5 mL) was added Ι,Γ-carbonyldiimidazole (559 mg, 3.45 mmol). After stirring at room temperature for 2 h, the solution was added to a stirred mixture of hydrazine hydrate (0.729 mL, 5.75 mmol) in THF (2 mL) and stirred a further 2 h. The reaction mixture was poured onto water (20 mL) and stirred for 30 min. The resulting precipitate was collected by filtration, washed with water (2 x 10 mL) then acetonitrile (3 mL) to afford 0.54 g (71%) of 4-(heptyloxy)benzohydrazide as a white solid. LCMS-ESI (m/z) calculated for C14H22N2O2: 250.3 found 251.0 [M+H] + , ¾ = 2.05 min. (Method 4).

(S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(2-(4-(heptyloxy)benzoyl)hy drazine- carbon l)phenyl)propanoate (INT-3)

[00270] To a stirring solution of (5)-4-(2-(4-(tert-butyl)benzamido)-3-methoxy- 3-oxopropyl)benzoic acid INT-1 (260 mg, 0.68 mmol) in THF (5 mL) were added 4- methylmorpholine (0.15 mL, 1.36 mmol) and isobutyl carbonochloridate (0.09 mL, 0.71 mmol). After stirring at room temperature for 2 h, 4-(heptyloxy)benzohydrazide (187 mg, 0.75 mmol) was added and stirring continued for another 2 h. The reaction mixture was poured onto NaHC0 3 (50 mL) and extracted with DCM (3 x 20 mL). The combined organics were dried over MgS0 4 and evaporated. The crude product was purified by column chromatography (100% EA in z ' so-hexanes) to afford 297 mg (71%) of (5)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(2-(4-

(heptyloxy)benzoyl)hydrazinecarbonyl)phenyl) propanoate INT-3 as an off-white foam. LCMS-ESI (m/z) calculated for C 3 6H 45 N 3 0 6 : 615.8 found 616.0 [M+H] + , t R = 2.89 min. (Method 4).

(S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l, 3,4- oxadiazol-2- l)phenyl)propanoate

[00271 ] To a stirring solution S)-methyl 2-(4-(fert-butyl)benzamido)-3-(4-(2-(4- (heptyloxy)benzoyl)hydrazinecarbonyl)phenyl)propanoate INT-3 (127 mg, 0.21 mmol) and TEA (0.09 mL, 0.62 mmol) in DCM (4 mL) was added 2-chloro-l,3- dimethylimidazolidinium chloride (41.8 mg, 0.25 mmol). The reaction mixture was stirred at room temperature for 18 h then warmed to 40°C for 1 h. The reaction mixture was cooled to room temperature, diluted with NaHC0 3 (15 mL), shaken, split through a hydrophobic frit and evaporated to afford 120 mg (95%) of (S)-methyl 2-(4-(fert- butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l,3,4-oxadiaz ol-2- yl)phenyl)propanoate as a white solid. LCMS-ESI (m/z) calculated for C 3 6H43N 3 0 5 : 597.8; found 598.0 [M+H] + , t R = 3.25 min. (Method 4).

[00272] Compound 76 was prepared using (S)-methyl 2-(4-(tert- butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l,3,4-oxadiaz ol-2- yl)phenyl)propanoate and General Procedure 4.

2-bromo-l-(4-(heptyloxy)phenyl)ethanone (INT-4)

[00273] To a stirring solution of l-(4-(heptyloxy)phenyl)ethanone (500 mg, 2.13 mmol) in THF (8.5 mL) under nitrogen was added phenyltrimethylammonium tribromide (842 mg, 2.24 mmol). The reaction mixture was stirred at RT for 2 h, filtered under vacuum and the captured solid washed with THF. The combined liquors were concentrated to afford 919 mg (100%) of 2-bromo-l-(4-(heptyloxy)phenyl)ethanone INT-4 as a yellow oil. LCMS-ESI (m/z) calculated for Ci 5 H 2 iBr0 2 : 313.2; found 313.0 [M+H] + , t R = 2.12 min. (Method 4).

(S)-2-(4-(heptyloxy)phenyl)-2-oxoethyl 4-(2-(4-(tert-butyl)benzamido)-3- methox -3-oxopropyl)benzoate

[00274] A solution of 2-bromo-l-(4-(heptyloxy)phenyl)ethanone, INT-4 (166 mg, 0.45 mmol) in acetonitrile (1 mL) was added to a solution of (S)-4-(2-(4-(tert- butyl)benzamido)-3-methoxy-3-oxopropyl)benzoic acid INT-1 (190 mg, 0.50 mmol) and TEA (75.0 μΐ, 0.54 mmol) in acetonitrile (4 mL). The reaction mixture was stirred at RT for 18 h then poured onto 0.5 M citric acid (30 mL) and extracted with EA (3 x 25 mL). The combined organics were dried over MgSC^, filtered and concentrated. The residue was triturated with Et 2 0 (10 mL) and the filtrate concentrated to afford 159 mg (49%) of (5)-2-(4-(heptyloxy)phenyl)-2-oxoethyl 4-(2-(4-(tert-butyl)benzamido) -3- methoxy-3-oxopropyl) benzoate as a white solid. LCMS-ESI (m/z) calculated for

CsvFLsNOy: 615.8; found 616.0 [M+H] + , t R = 2.76 min. (Method 4). (S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(4-(4-(heptyloxy)phenyl)oxa zol-2- yl)phenyl)propanoate

[00275] To borontrifluoride diethyl etherate (33.3 μΐ, 0.27 mmol) was added a mixture of acetamide (763 mg, 12.9 mmol) and (5)-2-(4-(heptyloxy)phenyl)-2-oxoethyl 4-(2-(4-(tert-butyl)benzamido)-3-methoxy-3-oxopropyl)benzoat e (159 mg, 0.26 mmol). The reaction mixture was stirred at 140°C for 1 h. The reaction mixture was allowed to cool to RT, diluted with EA (15 mL) and extracted with NaHC0 3 (3 x 15 mL) and brine (15 mL). The combined organics were dried over MgS0 4 , filtered and concentrated. The residue was recrystallised from Et 2 0 (5 mL), filtered and rinsed with Et 2 0. The filtrate was concentrated to afford 55 mg (16%) of (5)-methyl 2-(4-(tert- butyl)benzamido)-3-(4-(4-(4-(heptyloxy)phenyl)oxazol-2-yl)ph enyl)propanoate as an orange oil. LCMS-ESI (m/z) calculated for C 37 H 44 N 2 0 5 : 596.8; found 597.0 [M+H] + , tR = 3.11 min. (Method 4).

[00276] Compound 77 was prepared from (5)-methyl 2-(4-(tert- butyl)benzamido)-3-(4-(4-(4-(heptyloxy)phenyl)oxazol-2-yl)ph enyl)propanoate using General Procedure 4.

2- 4-bromophenyl)-2-oxoethyl 4-(heptyloxy)benzoate

[00277] To stirring mixture of 4-(heptyloxy)benzoic acid (2.0 g, 8.46 mmol) in acetonitrile (30 mL) at RT was added TEA (1.24 mL, 8.87 mmol) drop wise. The reaction mixture was stirred at RT for 1 h, poured onto 0.05 M citric acid (100 mL) and EA (10 mL) then stirred for 10 min. The precipitate was isolated by filtration, washed with water (30 mL) and zso-hexanes (2 x 10 mL) then dried in air to afford 3.8 g (98%) of 2-(4-bromophenyl)-2-oxoethyl 4-(heptyloxy)benzoate. LCMS-ESI (m/z) calculated for C22H 25 Br0 4 : 433.3; found 455.0/457.0 [M+Na] + , t R = 3.21 min. (Method 4).

4-(4-bromophenyl)-2-(4-(heptyloxy)phenyl)oxazole

r

[00278] To boron trifluoride etherate (0.322 mL, 2.5 mmol) was added 2-(4- bromophenyl)-2-oxoethyl 4-(heptyloxy)benzoate (1.0 g, 2.3 mmol) and acetamide (4.91 g, 83.0 mmol) in DCM (10 mL). The reaction mixture was heated to 50°C then 140°C for 16 h, DCM was distilled off. The reaction mixture was cooled, diluted with acetonitrile and stirred at RT for 1 h. The precipitate was isolated by filtration to afford 273 mg (23%) of 4-(4-bromophenyl)-2-(4-(heptyloxy)phenyl)oxazole as a brown solid. LCMS-ESI (m/z) calculated for C 2 2H2 4 BrN0 2 : 414.3; found 414.0 [M+H] + , t R = 3.00 min. (Method 4).

(S)-methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(2-(4-(heptyloxy)phenyl) oxazol- 4-yl)phenyl)propanoate

[00279] To zinc (104 mg, 1.59 mmol) stirring in DMF (1.5 mL) was added iodine (20.2 mg, 0.08 mmol). After the color disappeared, (i?)-methyl 2-((tert- butoxycarbonyl)amino)-3-iodopropanoate (175 mg, 0.53 mmol) and further iodine (20.2 mg, 0.08 mmol) were added. After 30 min, the mixture was de-gassed by bubbling through N 2 then treated with 4-(4-bromophenyl)-2-(4- (heptyloxy)phenyl)oxazole (220 mg, 0.53 mmol), Pd 2 dba 3 (12.2 mg, 0.01 mmol) and dicyclohexyl(2',6'-dimethoxy-[l, -biphenyl]-2-yl)phosphine (10.9 mg, 0.03 mmol) followed by THF (1 mL). The reaction mixture was heated to 50°C for 2 h, cooled to RT and purified by column chromatography (gradient of 15-95% EA in z ' so-hexanes) to afford 188 mg (65%) of (S)-methyl 2-((fert-butoxycarbonyl)amino)-3-(4-(2-(4- (heptyloxy)phenyl)oxazol-4-yl)phenyl)propanoate. LCMS-ESI (m/z) calculated for C 3 iH 4 oN 2 0 6 : 536.6; found 537.0 [M+H] + , t R = 3.72 min. (Method 11).

[00280] Compound 78 was prepared from (5)-methyl 2-((tert- butoxycarbonyl)amino)-3-(4-(2-(4-(heptyloxy)phenyl)oxazol-4- yl)phenyl)propanoate and 4-(tert-butyl)benzoic acid using General Procedures 8, 7 then 4.

2-(4-bromophenyl)-4-(4-(heptyloxy)phenyl)thiazole

[00281] To a stirring solution of 2-bromo-l-(4-(heptyloxy)phenyl)ethanone INT- 4 (1.37 g, 4.38 mmol) in EtOH (10 mL) were added 4-bromobenzothioamide (0.95 g, 4.38 mmol) and isopropanol (10 mL). The reaction mixture was stirred at RT for 16 h. The solid was isolated by filtration, washed with EtOH (5 mL) then taken up in DCM (10 mL) and NaHC0 3 (20 mL) and stirred for 1 h at RT. The solid was isolated by filtration, washed with water (2 x 10 mL) and acetonitrile ( 2 x 4 mL) then dried to afford 1.02 g (52%) of 2-(4-bromophenyl)-4-(4-(heptyloxy)phenyl)thiazole as a white micro-crystalline solid. LCMS-ESI (m/z) calculated for C 22 H 24 BrNOs: 429.1; found 430.0 [M+H] + , t R = 3.20 min. (Method 4). (S)-methyl 2-(( tert-butoxycarbonyl)amino)-3-(4-(4-(4-(heptyloxy)phenyl)thia zol- 2-yl)phenyl)propanoate

[00282] To a stirring suspension of zinc (228 mg, 3.49 mmol) in DMF (2 mL) was added diiodine (44 mg, 0.17 mmol). When the color was discharged, (i?)-methyl 2- ((fert-butoxycarbonyl)amino)-3-iodopropanoate (382 mg, 1.16 mmol) and further diiodine (44.2 mg, 0.17 mmol) were added. After stirring at RT for 30 min, the reaction mixture was de-gassed by bubbling through N 2 then 2-(4-bromophenyl)-4-(4- (heptyloxy)phenyl)thiazole (500 mg, 1.16 mmol), dicyclohexyl(2',6'-dimethoxy-[l , l'- biphenyl]-2-yl)phosphine (23.8 mg, 0.06 mmol), Pd 2 dba 3 (26 mg, 0.03 mmol) and DMF (2 mL) were added. The reaction mixture was heated to 50 °C for 3 h, cooled and purified by column chromatography (10-80% EA in z ' so-hexanes) to afford 620 mg (96%) of (iS)-methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(4-(4-(heptyloxy) phenyl) thiazol-2-yl) phenyl propanoate. LCMS-ESI (m/z) calculated for C 3 iH 40 N 2 O 5 S: 552.3; no ion observed, t R = 3.37 min. {Method 4).

[00283] Compound 79 was prepared from (5)-methyl 2-((tert- butoxycarbonyl)amino)-3-(4-(4-(4-(heptyloxy) phenyl) thiazol-2-yl) phenyl propanoate and 4-(tert-butyl)benzoic acid using General Procedures 8, 7 then 4.

4-(he tyloxy)benzothioamide

[00284] To stirring suspension of 4-(heptyloxy)benzamide (1.24 g, 5.29 mmol) in DME (20 mL) and THF (10 mL) was added 2,4-bis(4-phenoxyphenyl)-l ,3,2,4- dithiadiphosphetane 2,4-disulfide (2.80 g, 5.29 mmol). The reaction mixture was stirred at RT for 16 h. The reaction mixture was concentrated onto silica and purified by column chromatography (0-60% EA in z ' so-hexanes) to afford 1.4 g (62%) of 4- (heptyloxy)benzothioamide as a yellow waxy solid. LCMS-ESI (m/z) calculated for C 14 H 21 NOS: 251.4; found 252.0 [M+H] + , t R = 3.13 min. (Method 6).

4-(4-bromo henyl)-2-(4-(heptyloxy)phenyl)thiazole

[00285] To a stirring mixture of 4-(heptyloxy)benzothioamide (1.30 g, 5.17 mmol) in isopropanol (20 mL) was added 2-bromo-l-(4-bromophenyl)ethanone (1.44 g, 5.17 mmol). The precipitate was collected by filtration and washed with EtOH (2 x 5 mL). The filter cake was slurried with NaHC0 3 (2 x 20 mL), water (2 x 20 mL) then EtOH (2 x 5 mL) and dried to afford 926 mg (41%) of 4-(4-bromophenyl)-2-(4- (heptyloxy)phenyl)thiazole as a pale yellow powder. LCMS-ESI (m/z) calculated for C 2 2H 24 BrNOS: 429.1; found 430.0 [M+H] + , t R = 3.41 min. (Method 4).

(S)-methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(2-(4-(heptyloxy)phenyl) thiazol- 4- l)phenyl)propanoate

[00286] To a stirring mixture of zinc (182 mg, 2.79 mmol) in DMF (2 mL) was added diiodine (35.4 mg, 0.14 mmol). When the color was discharged, further diiodine (35.4 mg, 0.14 mmol) and (i?)-methyl 2-((tert-butoxycarbonyl)amino)-3- iodopropanoate (306 mg, 0.93 mmol) were added. After 30 min, DMF (1 mL) was added and the mixture de-gassed by bubbling through N 2 . To the reaction mixture were added 4-(4-bromophenyl)-2-(4-(heptyloxy)phenyl)thiazole (400 mg, 0.93 mmol), Pd 2 dba 3 (21 mg, 0.02 mmol) and dicyclohexyl(2',6'-dimethoxy-[l, -biphenyl]-2- yl)phosphine (19 mg, 0.05 mmol), the mixture was further de-gassed then heated to 50 °C for 3 h. The reaction mixture was cooled and purified by column chromatography (10-80% EA in z ' so -hexanes). The product obtained was taken into DCM (4 mL) and washed with water (20 mL) and dried through a hydrophobic frit. The organics were suspended in ACN (4 mL) and concentrated to afford 432 mg (83%) of (5)-methyl 2- ((fert-butoxycarbonyl)amino)-3-(4-(2-(4-(heptyloxy)phenyl)th iazol-4- yl)phenyl)propanoate as a yellow foam. LCMS-ESI (m/z) calculated for C 3 iH 40 N 2 O5S: 552.7; no ion observed, fa = 3.36 min. {Method 4).

[00287] Compound 80 was prepared from (5)-methyl 2-((tert- butoxycarbonyl)amino)-3-(4-(2-(4-(heptyloxy)phenyl)thiazol-4 -yl)phenyl)propanoate and 4-(tert-butyl)benzoic acid using General Procedures 8, 7 then 4.

4-(5-( 4-(heptyloxy)phenyl) thiazol-2-yl)benzaldehyde

[00288] To a stirring suspension of 4-(thiazol-2-yl)benzaldehyde (349 mg, 1.84 mmol), tricyclohexylphosphine (27 mg, 0.07 mmol), pivalic acid (64.2 μΐ, 0.55 mmol), potassium carbonate (382 mg, 2.77 mmol) and palladium (II) acetate (8 mg, 0.04 mmol) in DMA (5.15 mL) under nitrogen was added a solution of l-bromo-4- (heptyloxy)benzene (500 mg, 1.84 mmol) in DMA (1 mL). The reaction mixture was evacuated and purged with nitrogen 3 times then heated at 100°C for 6 h. Once cooled, the reaction mixture was diluted with EA (40 mL), washed with water (3 x 40 mL) and brine (40 mL). The organic phase was dried over MgS0 4 , filtered and concentrated in vacuo to afford a brown-green solid. The crude product was purified by

chromatography (0-50% EA in hexanes) to afford 270 mg (37%) of 4-(5-(4- (heptyloxy)phenyl)thiazol-2-yl)benzaldehyde as an iridescent yellow solid. LCMS-ESI (m/z) calculated for C23H25NO2S : 379.5; found 380.0 [M+H] + , t R = 2.99 min. (Method

Methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(5-(4-(heptyloxy)phenyl) thiazol-2- l)phenyl) acrylate

[00289] A stirring mixture of 1,1,3,3-tetramethylguanidine (86 μΐ, 0.69 mmol) was added to a suspension of 4-(5-(4-(heptyloxy)phenyl)thiazol-2-yl)benzaldehyde (260 mg, 0.685 mmol) and methyl 2-((tert-butoxycarbonyl)amino)-2- (dimethoxyphosphoryl)acetate (185 mg, 0.62 mmol) in anhydrous THF (10 mL) under nitrogen, at -70°C. The reaction mixture was stirred at -70°C for 1 h then at RT for 18 h. The reaction mixture was diluted with DCM (50 mL), washed with water (50 mL), passed through a phase separation cartridge and the organic phase concentrated in vacuo to afford a yellow solid. The solid was triturated with EA/EtOH (20 mL) and the collected solid washed with EtOH (10 mL) and Et 2 0 to afford 284 mg (79%) of methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(5-(4-(heptyloxy)phenyl) thiazol-2-yl)phenyl) acrylate as a yellow solid. LCMS-ESI (m/z) calculated for C 3 iH 38 N 2 0 5 S: 550.7; found 551.0 [M+H] + , t R = 3.11 min. (Method 8).

Methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(5-(4-(heptyloxy)phenyl) thiazol-2- l)phenyl) propanoate

[00290] A stirring mixture of Methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(5-(4- (heptyloxy)phenyl) thiazol-2-yl) phenyl)acrylate (50 mg, 0.091 mmol) dissolved in dioxane (5 mL) was hydrogenated using an H-Cube hydrogenator (10% Pd/C, 30 x 4 mm, full hydrogen, 40°C, 1 mL/min). The reaction mixture was concentrated in vacuo to afford 21 mg (29%) of methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(5-(4- (heptyloxy) phenyl) thiazol-2-yl) phenyl) propanoate as a yellow solid. LCMS-ESI (m/z) calculated for C31H40N2O5S: 552.7; found 553.0 [M+H] + , t R = 1.85 min. (Method 8).

[00291] Compound 81 was prepared from Methyl 2-((tert- butoxycarbonyl)amino)-3-(4-(5-(4-(heptyloxy) phenyl) thiazol-2-yl) phenyl) propanoate and 4-(tert-butyl)benzoyl chloride using General Procedures 8, 3 then 4.

[00292] Compound 82 was prepared in a similar fashion to Compound 81 using 4-(2-(4-(heptyloxy) phenyl) thiazol-5-yl)benzaldehyde in place of 4-(5-(4- (heptyloxy)phenyl)thiazol-2-yl) benzaldehyde.

(S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)-l, 3,4- thiadiazol-2- l)phenyl)propanoate

[00293] Prepared using INT-3 : To a stirring solution of 2,4-bis(4- methoxyphenyl)-l,3,2,4-dithiadiphosphetane 2,4-disulfide (65.7 mg, 0.16 mmol) in THF (3 mL) was added (5)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(2-(4- (heptyloxy)benzoyl) hydrazinecarbonyl) phenyl) propanoate INT-3 (100.0 mg, 0.16 mmol) and the mixture heated to 65°C. After 1 h, the reaction mixture was

concentrated and purified by column chromatography (10-100% EA in z ' so-hexanes) to afford 37.0 mg (29%) of (S)-methyl 2-(4-(tert-butyl)benzamido)-3-(4-(5-(4- (heptyloxy)phenyl)-l,3,4-thiadiazol-2-yl)phenyl)propanoate as a yellow solid. LCMS- ESI (m/z) calculated for C36H43N3O4S : 613.8; no ion observed, fa = 3.31 min. {Method 4).

[00294] Compound 83 was prepared from (5)-methyl 2- (4-(tert-butyl) benzamido) -3-(4-(5-(4-(heptyloxy) phenyl)- 1, 3, 4-thiadiazol-2-yl)phenyl)propanoate using General Procedure 4.

[00295] Compound 84 was prepared using 3-bromo-5-chloro- 1 ,2,4-thiadiazole, (4-(heptyloxy)phenyl)boronic acid and INT-13 using General Procedures 10, 10, and 8 sequentially.

(S)-tert-butyl 2-( ( (benzyloxy)carbonyl)amino)-3-(4- (((tri uoromethyl)sulfonyl)oxy)-phenyl) propanoate (INT-5)

[00296] Prepared using General Procedure 9: A stirred solution of (S)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-hydroxyphenyl)propanoate hydrate (25 g, 64.2 mmol) in DCM (100 mL) was treated with magnesium sulfate (4.01 g, 33.7 mmol). After 15 min, the mixture was filtered and washed with DCM (2 x 20 mL). The organics were treated with N-ethyl-N-isopropylpropan-2-amine (17.41 g, 134.7 mmol) and stirred. This solution was treated with 1,1,1-trifluoro-N-phenyl-N- ((trifluoromethyl)sulfonyl)methanesulfonamide (26.44 g, 74.01 mmol) and the mixture was allowed to stir overnight at RT. The mixture was treated with water (50 mL) and saturated aqueous NaHC0 3 (20 mL) and stirred vigorously for 10 min. The layers were separated and the organic layer was further washed with saturated aqueous NaHC0 3 (2 x 50 mL), water (50 mL), and saturated aqueous NaHC0 3 (50 mL) and concentrated. The compound was purified by chromatography (EA / hexanes) to afford 26.85 g (79%) of {S)-tert- vXy\ 2-(((benzyloxy)carbonyl)amino)-3-(4- (((trifluoromethyl)sulfonyl)oxy)phenyl) propanoate INT-5. LCMS-ESI (m/z) calculated for C 22 H 24 F 3 NO 7 S: 503.1; found 526.1 [M + Na] + , ¾ = 4.12 min (Method 3).

(S)-tert-butyl 2-(( (benzyloxy)carbonyl)amino)-3-(4-(4, 4, 5, 5-tetramethyl-l, 3, 2- dioxaborolan-2- l)phenyl)propanoate (INT-6)

[00297] A solution of (S)-tert-buty\ 2-(((benzyloxy) carbonyl)amino)-3-(4- (((trifluoromethyl)sulfonyl)oxy) phenyl) propanoate INT-5 (26.85 g, 53.4 mmol), potassium acetate (15.71 g, 160.1 mmol), bis-pinacolatoborane (27.1 g, 106.7 mmol) and DMSO (100 mL) was degassed with a steady flow of nitrogen gas for 5 minutes. To this solution was added PdCi 2 (dppf) (1.95 g, 2.67 mmol) and the solution further degassed and kept under an atmosphere of nitrogen. The mixture was heated at 100°C for 18 h then cooled to RT and diluted with EA (50 mL) and washed with saturated aqueous NaHC0 3 (20 mL), water (3 x 30 mL), dried over MgSC^, filtered, and the solvent removed under reduced pressure. The compound was purified by column chromatography to give 11.10 g (41 %) of (S)-tert- vXy\ 2- (((benzyloxy)carbonyl)amino)-3-(4-(4,4,5,5-tetramethyl-l,3,2 -dioxaborolan-2-yl) phenyl) propanoate INT-6 as a oil. LCMS-ESI (m/z) calculated for C 27 H 36 BNO 6 :

481.3; found 504.3 [M+Na] + , t R = 4.21 min (Method 3). 1H NMR (400 MHz, DMSO) δ 7.72 (d, J= 8.3 Hz, 1H), 7.60 (d, J= 8.0 Hz, 2H), 7.42 - 7.11 (m, 6H), 4.98 (s, 2H), 4.22 - 4.08 (m, 1H), 3.03 (dd, J= 13.7, 5.2 Hz, 1H), 2.85 (dd, J= 13.6, 10.1 Hz, 1H), 1.36 (s, 6H), 1.30 (s, 9H), 1.22 - 1.13 (m, 6H).

I l l (S)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-(5-bromopyrimidin-2- l)phenyl) propanoate (INT-7)

[00298] Prepared using General Procedure 10: A stirred mixture of (S)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-(4,4,5,5-tetramethyl-l,3 ,2-dioxaborolan-2-yl) phenyl) propanoate INT-6 (21.7 g, 45.0 mmol) and 5-bromo-2-iodopyrimidine (15.4 g, 54.0 mmol) in dioxane (400 mL) with sodium carbonate decahydrate (25.7 g, 90 mmol) in water (100 mL) was de-gassed. PdCl 2 (dppf) (0.99 g, 1.4 mmol) was added and the mixture further de-gassed then heated to reflux for 5 h. The mixture was allowed to cool while stirring overnight. The mixture was poured onto water (1 L) and EA (300 mL) and stirred for 30 min. The mixture was filtered and the layers were separated. The aqueous layer was further extracted with EA (2 x 200 mL) and the combined organic layers were washed with water (2 x 100 mL) then brine (50 mL), dried over MgS0 4 and concentrated. Column chromatography (EA / hexanes) gave 14.84 g (63 %) of (S)-tert- butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-(5-bromopyrimidin-2-yl)p henyl) propanoate INT-7. LCMS-ESI (m/z) calculated for C^fteBrNsC 511.1; found 534.0 [M + Na] + , t R = 2.97 min (Method 11).

(S)-tert-butyl 2-( ( (benzyloxy)carbonyl)amino)-3-(4-(5-(4- heptyloxy)phenyl)pyrimidin-2-yl) phenyl)pro anoate (INT-8)

[00299] Prepared using General Procedure 10: A stirred solution of (S)-tert- butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-(5-bromopyrimidin-2-yl)p henyl)propanoate INT-7 (759 mg, 1.48 mmol), (4-(heptyloxy)phenyl)boronic acid (455 mg, 1.93 mmol) and sodium bicarbonate (311 mg, 3.70 mmol) in acetonitrile (5 ml), THF (5 ml), and water (4 ml) was degassed with N 2 for 5 min. Pd(dppf)Cl 2 (108 mg, 0.15 mmol) was added and the reaction was heated to 110°C in the microwave for 50 min. The reaction was diluted with EA and water then filtered. The organic phase was dried over MgS04, filtered, and concentrated. The crude product was purified by chromatography on silica gel (EA / hexanes) to afford 591 mg (62 %) of (S)-ieri-butyl 2- (((benzyloxy)carbonyl)amino)-3-(4-(5-(4-(heptyloxy)phenyl)py rimidin-2-yl)phenyl) propanoate INT-8 as a yellow solid. LCMS-ESI (m/z) calculated for C38H45N3O5 : 623.8; no m/z observed, t R = 3.42 min (Method 8).

(S)-tert-butyl 2-amino-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2-yl)phenyl) ropanoate (INT-9)

[00300] To a stirred solution of (S)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3- (4-(5-(4-(heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoate INT-8 (591 mg, 0.95 mmol) in EA (25 ml) was added Pd/C (101 mg, 0.09 mmol) and the suspension degassed with H 2 . The mixture was stirred vigorously under an atmosphere of H 2 overnight then filtered through celite and the filtrate was concentrated to give 405 mg (83%) of (S)-tert-bvXy\ 2-amino-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2- yl)phenyl)propanoate INT-9. LCMS-ESI (m/z) calculated for C30H39N3O3: 489.3; found: 490.2 [M+H] + , t R = 2.35 min (Method 8). (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl )pyrimidin-2- yl)phenyl)propanoic acid (Compound 85)

[00301 ] A stirred solution of (S)-ierf-butyl 2-amino-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoate INT-9 (1.34 g, 2.74 mmol) and 4- (tert-butyl)benzoic acid (0.54 g, 3.01 mmol) in DMF (5 mL) and N-ethyl-N- isopropylpropan-2-amine (1.01 ml, 5.47 mmol) was treated with HATU (1.09 g, 2.87 mmol). After stirring for 1 h, the mixture was treated with water (60 mL) and iso- hexanes (20 mL) and stirred for 1 h. The product was collected by filtration, washed with water (3 x 10 mL) then z ' so-hexanes (10 mL) and dried in the vacuum oven. The ester was taken up in DCM (5 mL) and treated with TFA (5 mL). After 2 h, the mixture was treated with toluene (5 mL) and evaporated. The residue was taken up in DMSO (6 mL) then treated with water (20 mL) and stirred for 1 h. The product was collected by filtration, washed with water (3 x 15 mL) then acetonitrile (2 x 5 mL), and dried in the vacuum oven to give 1.40 g (85 %) of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4- (heptyloxy) phenyl) pyrimidin-2-yl) phenyl)propanoic acid Compound 85 as a white solid. LCMS-ESI (m/z) calculated for C37H43N3O4: 593.3; found: 594.0 [M+H] + , t R = 11.18 min (Method 9) and 97% e.e. (Chiral Method). 1 H NMR (400 MHz, DMSO-d6) δ 12.79 (br, s, 1H), 9.16 (s, 2H), 8.66 (d, J= 8.2 Hz, 1H), 8.45 - 8.27 (m, 2H), 7.89 - 7.69 (m, 4H), 7.57 - 7.38 (m, 4H), 7.18 - 7.02 (m, 2H), 4.77 - 4.62 (m, 1H), 4.03 (t, J = 6.5 Hz, 2H), 3.30 - 3.24 (m, 1H), 3.22 - 3.12 (m, 1H), 1.80 - 1.68 (m, 2H), 1.48 - 1.20 (m, 17H), 0.96 - 0.82 (m, 3H).

[00302] Compounds 86 - 102, 104 - 158 and 296 were prepared from (S)-tert- butyl 2-amino-3-(4-(5-(4-(heptyloxy) phenyl) pyrimidin-2-yl)phenyl)propanoate INT-8 using General Procedures 3 ox 7 followed by 4 or 8. (S)-tert-butyl 2-( ( (benzyloxy)carbonyl)amino)-3-(4-(5-(4-(tert- but l)phenyl)pyrimidin-2-yl) phenyl)propanoate (INT-10)

[00303] Prepared using General Procedure 10: A stirred solution of (S)-tert- butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-(5-bromopyrimidin-2-yl)p henyl)propanoate INT-7 (0.96 g, 1.86 mmol), (4-(tert-butyl)phenyl)boronic acid (0.43 g, 2.42 mmol) and sodium bicarbonate (0.39 g, 4.66 mmol) in acetonitrile (5 ml), THF (5 ml) and water (5 ml) was degassed with N 2 for 5 min. Pd(dppf)Cl 2 (0.136 g, 0.186 mmol) was added and the reaction was heated to 110°C in the microwave for 45 min. The reaction was diluted with EA (50mL) and filtered over celite. The organic phase was washed with water (100 mL) and concentrated. The crude product was purified by chromatography on silica gel (EA / isohexanes) to afford 757 mg (70 %) of {S)-tert-bvXy\ 2- (((benzyloxy)carbonyl)amino)-3 -(4-(5 -(4-(tert-butyl)phenyl)pyrimidin-2-yl) phenyl) propanoate INT-10 as a white powder. LCMS-ESI (m/z) calculated for C35H39N3O4: 565.3; no m/z observed, = 3.39 min {Method 8).

(S)-tert-butyl 2-amino-3-(4-(5-(4-(tert-butyl)phenyl)pyrimidin-2-yl)phenyl) - ropanoate (INT-11)

[00304] To a stirred solution of (S)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3- (4-(5-(4-(tert-butyl)phenyl)pyrimidin-2-yl)phenyl)propanoate INT-10 (757 mg, 1.34 mmol) in EA (100 ml) was added Pd/C (142 mg, 0.13 mmol) and the suspension degassed with H 2 . The mixture was stirred vigorously under an atmosphere of H 2 overnight then filtered through celite and the filtrate was concentrated to give 532 mg (88%) of (S)-tert-butyl 2-amino-3-(4-(5-(4-(fert-butyl)phenyl)pyrimidin-2- yl)phenyl)propanoate INT-11. LCMS-ESI (m/z) calculated for C27H33N3O2: 431.3; found: 432.0 [M+H] + , t R = 2.01 min (Method 4).

[00305] Compounds 159 - 181 were prepared from (S)-tert- vXy\ 2-amino-3-(4- (5-(4-(tert-butyl) phenyl)pyrimidin-2-yl)phenyl)propanoate INT-11 using General Procedures 3 ox 7 followed by 4 or 8.

[00306] Compound 182 can be prepared in a manner analogous to 165 starting from (R)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-hydroxyphenyl)propanoate .

[00307] Compound 183 was prepared from (5)-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)-2-(4-hydroxybenzami do)propanoic acid, Compound 114 using General Procedure 13.

[00308] Compounds 184 - 190 were prepared from (S)-tert- vXy\ 2-amino-3-(4- (5-(4-(heptyloxy) phenyl)pyrimidin-2-yl)phenyl)propanoate INT-9 using General Procedures 13 and 8 sequentially.

[00309] Compound 191 can be prepared in a manner analogous to 85 starting from (R)-tert-butyl 2-(((benzyloxy)carbonyl)amino)-3-(4-hydroxyphenyl)propanoate .

(S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4- (heptyloxy)phenyl)- pyrimidin-2-yl)phenyl)propanoic acid (Compound 192)

[00310] A stirring solution of (S)-ierf-butyl 2-amino-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoate INT-9 (5.50 g, 11.23 mmol) and 5-(fert-butyl)thiophene-2-carboxylic acid (2.13 g, 11.57 mmol) in DMF (50 mL) and DIEA (6.22 ml, 33.70 mmol) was treated portion wise with HATU (4.48 g, 11.79 mmol). After stirring for 1 h, the mixture was treated with water (200 mL) and iso- hexanes (20 mL) and stirred for 10 min. The product was collected by filtration, washed with z ' so-hexanes (2 x 30 mL), water (2 x 50 mL) then MeOH (20 mL) and zso-hexanes (30 mL). The ester was taken up in DCM (50 mL) and treated with TFA (10 mL). After 1 h, additional TFA (15 mL) was added. After a further 5 h, the mixture was treated with toluene (20 mL) and concentrated. The residue was washed with acetonitrile (25 mL) then taken up in DMSO (20 mL) then treated with water (100 mL) and stirred for 1 h. The product was collected by filtration, washed with water (4 x 50 mL) then acetonitrile (3 x 30 mL), and dried in a vacuum oven to give 5.30 g (75 %) of (S)-2-(5- (tert-butyl)thiophene-2-carboxamido)-3 -(4-(5 -(4-(heptyloxy)phenyl) pyrimidin-2-yl) phenyl)propanoic acid, Compound 192 as an off-white solid. LCMS-ESI (m/z) calculated for C35H41N3O4S: 599.3; no m/z observed, t R = 11.10 min (Method 10). The chiral purity was 98% e.e. (Chiral Method). 1H NMR (400 MHz, DMSO-d6) δ 12.87 (s, 1H), 9.17 (s, 2H), 8.68 (d, J= 8.3 Hz, 1H), 8.47 - 8.17 (m, 2H), 7.96 - 7.71 (m, 2H), 7.64 (d, J= 3.8 Hz, 1H), 7.55 - 7.29 (m, 2H), 7.26 - 7.02 (m, 2H), 6.93 (d, J= 3.8 Hz, 1H), 4.79 - 4.48 (m, 1H), 4.03 (t, J= 6.5 Hz, 2H), 3.27 (dd, J= 13.9, 4.5 Hz, 1H), 3.12 (dd, J= 13.9, 10.6 Hz, 1H), 1.90 - 1.58 (m, 2H), 1.58 - 1.01 (m, 17H), 1.01 - 0.69 (m, 3H).

[00311] Compound 193 was be prepared in a manner analogous to 192 starting from (R)-tert-buty\ 2-(((benzyloxy)carbonyl)amino)-3-(4-hydroxyphenyl)propanoate . Tert-butyl (4-( tert-butyl) benzoyl)-L-tyrosinate

[00312] Prepared using General Procedure 7. Into a solution of 4-(tert- butyl)benzoic acid (8.3 g, 46.4 mmol) in DMF (100 mL) were added HATU (19.2 g, 50.6 mmol), TEA (17.6 mL, 126.4 mmol) and (S)-tert-butyl 2-amino-3-(4- hydroxyphenyl) propanoate (10.0 g, 42.1 mmol). After 5 h, the reaction mixture was diluted with EA, washed with saturated aqueous NaHC0 3 and brine, then dried

(Na 2 S0 4 ), concentrated, and purified by chromatography (EA/ hexanes) to provide 12.9 g (69%) of tert-butyl (4-(tert-butyl)benzoyl)-L-tyrosinate. LCMS-ESI (m/z) calculated for C 24 H 3 iN0 4 : 397.5; no m/z observed, t R = 3.59 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 7.71 - 7.65 (m, 2H), 7.47 - 7.39 (m, 2H), 7.04 (t, J = 5.7 Hz, 2H), 6.78 - 6.70 (m, 2H), 6.59 (d, J = 7.5 Hz, 1H), 4.91 (dt, J = 7.5, 5.6 Hz, 1H), 3.15 (qd, J = 14.0, 5.6 Hz, 2H), 1.45 (s, 9H), 1.33 (s, 9H).

Tert-butyl (S)-2-(4-(tert-butyl)benzamido)-3-(4-( ((trifluoromethyl)sulfonyl)oxy) phenylpropanoate (INT-12)

[00313] Prepared using General Procedure 9. Into a solution of tert-butyl (4- (tert-butyl)benzoyl)-L-tyrosinate (8.0 g, 17.9 mmol) were added DIEA (3.7 mL, 1.2 mmol) and N-Phenylbis(trifluoromethanesulfonimide) (7.0 g, 19.7 mmol). After stirring for 36 h, the reaction mixture was diluted with DCM then washed with 10% aqueous citric acid and saturated aqueous NaHC0 3 . The organic layer was dried over Na 2 S0 4 , and concentrated to provide 9.5 g (100%) tert-butyl (S)-2-(4-(tert- butyl)benzamido)-3-(4-(((trifluoromethyl)sulfonyl)oxy) phenyl) propanoate INT-12, which was used without further purification. LCMS-ESI (m/z) calculated for

C 2 5H 3 oF 3 N0 6 S: 529.6; no m/z observed, t R = 4.42 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 7.71 - 7.65 (m, 2H), 7.49 - 7.43 (m, 2H), 7.32 - 7.26 (m, 2H), 7.22 - 7.16 (m, 2H), 6.69 (d, J = 7.0 Hz, 1H), 4.94 (dt, J = 6.9, 5.9 Hz, 1H), 3.24 (t, J = 7.1 Hz, 2H), 1.41 (s, 9H), 1.33 (s, 9H).

Tert-butyl (S)-2-(4-(tert-butyl)benzamido)-3-(4-(4, 4, 5, 5-tetramethyl-l, 3, 2- dioxaborolan-2-yl)phenyl)propanoate (INT-13)

[00314] Into a degassed solution of (5)-2-(4-(tert-butyl)benzamido)-3-(4- (((trifluoromethyl)sulfonyl)oxy) phenyl) propanoate INT-12 (9.5 g, 24 mmol), KOAc (7.0 g, 72 mmol), and bis-pinacolatoborane (9.1 g, 36 mmol) in DMSO (20 mL) was added Pd(dppf)Cl 2 (0.87 g, 1 mmol). The reaction mixture was heated at 100°C for 12 h under an atmosphere of N 2 . The reaction mixture was diluted with EA then washed with saturated aqueous NaHC0 3 and H 2 0. The organic layer was dried over Na 2 S0 4 , concentrated, and purified by chromatography (EA / hexanes) to provide 7.2 g (60%>) of tert-butyl (5)-2-(4-(tert-butyl)benzamido)-3-(4-(4,4,5,5-tetramethyl-l, 3,2-dioxaborolan- 2-yl)phenyl) propanoate INT-13. LCMS-ESI (m/z) calculated for C 30 H 42 BNO 5 : 507.5; no m/z observed, t R = 4.53 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 7.74 (d, J = 8.0 Hz, 2H), 7.72 - 7.67 (m, 2H), 7.48 - 7.43 (m, 2H), 7.21 (d, J = 8.0 Hz, 2H), 6.59 (d, J = 7.4 Hz, 1H), 5.05 - 4.92 (m, 1H), 3.27 (qd, J = 13.7, 5.4 Hz, 2H), 1.47 (s, 9H), 1.36 (m, 21H).

Tert-butyl (S)-3-(4-( 5-bromopyrimidin-2-yl)phenyl)-2- (4-( tert- butyl)benzamido)propanoate (INT-14)

[00315] Prepared using General Procedure 10. Into a degassed solution of (5)-2- (4-(tert-butyl)benzamido)-3-(4-(4,4,5,5-tetramethyl-l,3,2-di oxaborolan-2- yl)phenyl)propanoate INT-13 (1.0 g, 2.0 mmol), Na 2 HC03 (420 mg, 3.9 mmol), and 5- bromo-2-iodopyrimidine (615 mg, 2.2 mmol) in 2/2/1 MeCN/THF/H 2 0 was added Pd(dppf)Cl 2 (140 mg, 0.2 mmol). The reaction mixture was heated at 110°C for 1 h in a microwave reactor. The reaction mixture was concentrated, dissolved in DCM and washed with H 2 0. The organic layer was dried over Na 2 S0 4 , concentrated, and purified by chromatography (EA / hexanes) to provide 630 mg (58%) of tert-butyl (S)-3-(4-(5- bromopyrimidin-2-yl)phenyl)-2-(4-(tert-butyl) benzamido) propanoate INT-14. LCMS- ESI (m/z) calculated for C 2 8H3 2 BrN 4 0 3 : 538.5; no m/z observed, tR = 4.66 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 8.84 - 8.78 (s, 2H), 8.31 (t, J = 7.0 Hz, 2H), 7.75 - 7.64 (m, 2H), 7.46 - 7.38 (m, 2H), 7.30 (dd, J = 12.9, 7.1 Hz, 2H), 6.65 (d, J = 7.2 Hz, 1H), 5.10 - 4.94 (m, 1H), 3.43 - 3.20 (m, 2H), 1.45 (s, 9H), 1.32 (s, 9H).

[00316] Compounds 194 - 236 were prepared from tert-butyl (5)-3-(4-(5- bromopyrimidin-2-yl) phenyl)-2-(4-(tert-butyl)benzamido)propanoate INT-14 using General Procedures 10 and 8 sequentially. Tert-butyl (5-(tert-butyl)thiophene-2-carbonyl)-L-tyrosinate

[00317] Prepared using General Procedure 7. Into a solution of 5-(tert- butyl)thiophene-2-carboxylic acid (1.93 g, 10.0 mmol) in DMF (20 mL) were added HATU (4.56 g, 12.0 mmol) and TEA (4.18 mL, 30.0 mmol). The mixture was stirred at room temperature for 30 min and (S)-tert-butyl 2-amino-3-(4-hydroxyphenyl) propanoate (2.37 g, 10.0 mmol) was added. After 1 h, the reaction mixture was poured into 400 mL of ice-water and the solid was filtered. The solid was dissolved in DCM and EA, dried over MgS0 4 , concentrated, and purified by chromatography (EA / hexanes) to provide 3.6 g (89%) of tert-butyl (5-(tert-butyl)thiophene-2-carbonyl)-L- tyrosinate. LCMS-ESI (m/z) calculated for C 2 2H 29 N0 4 S: 403.2; found: 426.1 [M+Na] + , t R = 9.07 min (Method 2).

Tert-butyl (S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4- (((trifluoromethyl)sulfonyl) oxy) phenyl) propanoate (INT-15)

[00318] Prepared using General Procedure 9. Into a solution of tert-butyl (5- (tert-butyl)thiophene-2-carbonyl)-L-tyrosinate (3.52 g, 8.72 mmol) were added DIEA (4.56 mL, 26.17 mmol) and N-phenylbis(trifluoromethanesulfonimide) (3.27 g, 9.16 mmol). After stirring for 18 h, the reaction mixture was diluted with DCM then washed with saturated aqueous NaHC0 3 . The organic layer was dried over MgS0 4 and concentrated. The crude product was purified by chromatography to provide 4.10 g (87.6 %) of tert-butyl (S)-2-(5-(tert-butyl)thiophene-2-carboxamido) -3-(4- (((trifluoromethyl) sulfonyl)oxy)phenyl) propanoate INT-15. LCMS-ESI (m/z) calculated for C 23 H 28 F 3 N0 6 S 2 : 535.1; no m z observed, tR = 4.22 min {Method 3).

Tert-butyl (S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(4,4,5,5- tetramethyl-1, 3,2-dioxaborolan-2-yl)phenyl)propanoate (INT-16)

[00319] Into a degassed solution of tert-butyl (5)-2-(5-(tert-butyl)thiophene-2- carboxamido)-3-(4-(((trifluoromethyl)sulfonyl)oxy)phenyl)pro panoate INT-15 (3.89 g, 7.26 mmol), KOAc (2.14 g, 21.79 mmol), and bis-pinacolatoborane (2.40 g, 9.44 mmol) in DMSO (50 mL) was added Pd(dppf)Cl 2 (0.27 g, 0.36 mmol). The reaction mixture was heated at 100°C for 18 h under an atmosphere of N 2 . The reaction mixture was poured into 600 mL of ice-water and the solid was filtered. The precipitate was diluted with EA, dried over MgS0 4 , concentrated, and purified by chromatography (EA / hexanes) to provide 3.68 g (99%) of tert-butyl (5)-2-(5-(tert-butyl)thiophene-2- carboxamido)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl) phenyl)propanoate INT-16. LCMS-ESI (m/z) calculated for C 2 8H 4 oBN0 5 S: 513.3; no m/z observed, t R = 4.51 min (Method 3). Tert-butyl (S)-3-(4-(5-bromopyrimidin-2-yl)phenyl)-2-(5-(tert-butyl)thi ophene- 2 -carboxamidojpropanoate (INT-17)

[00320] Prepared using General Procedure 10. Into a degassed solution of tert- butyl (5)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3 -(4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl) phenyl)propanoate INT-16 (510 mg, 1.0 mmol) and 5-bromo-2- iodopyrimidine (570 mg, 2.0 mmol) in 2/2/1 MeCN/THF/saturated aqueous NaHC0 3 (10 mL) was added Pd(dppf)Cl 2 (30 mg, 0.4 mmol). The reaction mixture was heated at 120°C for 1 h in a microwave reactor. The reaction mixture was diluted with water (100 mL) and EA (50 mL) and filtered over Celite. The aqueous layer was extracted with EA (3 x 30 mL) and the combined organic layer was dried over MgSC^, concentrated, and purified by chromatography (EA / hexanes) to provide 342 mg (63%) of tert-butyl (S)-3 -(4-(5 -bromopyrimidin-2-yl)phenyl)-2-(5 -(tert-butyl)thiophene-2- carboxamido)propanoate INT-17. LCMS-ESI (m/z) calculated for C26H 30 BrN 3 O 3 S: 543.1; found: 488.0 [M-tBu+H] + , t R = 10.95 min (Method 2).

[00321] Compounds 237 - 247 were prepared from tert-butyl (5)-3-(4-(5- bromopyrimidin-2-yl)phenyl)-2-(5-(fert-butyl)thiophene-2-car boxamido)propanoate INT-17 using General Procedures 10 and 8 sequentially.

Tert-butyl (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-cyanopyrimidin

propanoate (INT-18)

[00322] Prepared using General Procedure 1. Into a degassed solution of (5)-3- (4-(5-bromopyrimidin-2-yl)phenyl)-2-(4-(tert-butyl)benzamido )propanoate INT-14 (100 mg, 0.190 mmol), and Zn(CN) 2 (44mg, 0.370 mmol) in NMP (5 niL) was added Pd(Ph 3 ) 4 (2 mg, 0.002 mmol). The mixture was heated for 45 min at 80°C in a microwave reactor then partitioned between DCM and H 2 0. The organic layer was dried over Na 2 S0 4 , concentrated, and purified by chromatography (EA/ hexanes) to provide 75 mg (84%) of tert-butyl (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5- cyanopyrimidin-2-yl)phenyl)propanoate INT-18. LCMS-ESI (m/z) calculated for C 29 H 32 N 4 0 3 : 484.60; no m/z observed, t R = 4.17 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 8.97 (s, 2H), 8.38 (d, J = 7.9 Hz, 2H), 7.67 (d, J = 8.0 Hz, 2H), 7.46 - 7.35 (m, 2H), 7.33 (d, J = 7.9 Hz, 2H), 6.77 (d, J = 6.8 Hz, 1H), 4.96 (d, J = 6.1 Hz, 1H), 3.27 (dd, J = 13.1, 8.0 Hz, 2H), 1.37 (d, J = 34.5 Hz, 9H), 1.26 (d, J = 21.0 Hz, 9H).

Tert-butyl (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(N-hydroxycarbamimid oyl)- pyrimidin-2-yl)phenyl)propanoate

[00323] Prepared using General Procedure 2. A solution of (S)-2-(4-(tert- butyl)benzamido)-3-(4-(5-cyanopyrimidin-2-yl)phenyl)propanoa te INT-18 (35 mg, 0.07 mmol), hydroxylamine (25 \L, 0.36 mmol, 50% solution in H 2 0), and NEt 3 (11xL, 0.08 mmol) in EtOH (5 mL) was heated at 80°C for 1.5 h. The reaction mixture was concentrated, dissolved in DCM and washed with H 2 0 to provide 22 mg of tert- butyl (S)-2-(4-(tert-butyl)benzamido)-3 -(4-(5 -(N-hydroxycarbamimidoyl) pyrimidin-2- yl)phenyl)propanoate. LCMS-ESI (m/z) calculated for C 29 H 35 N 5 0 4 : 517.6; found 462.2 [M-'Bu+H] + , t R = 3.72 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 9.19 (s, 2H), 8.42 (d, J = 8.2 Hz, 2H), 7.67 (dd, J = 8.5, 2.1 Hz, 2H), 7.40 (dd, J = 9.2, 8.0 Hz, 2H), 7.34 (dd, J = 10.3, 8.4 Hz, 2H), 6.74 (dd, J = 7.1, 4.7 Hz, 1H), 5.00 (q, J = 5.6 Hz, 1H), 2.83 (d, J = 5.3 Hz, 2H), 1.44 (s, 9H), 1.28 (d, J = 22.0 Hz, 9H).

Tert-butyl (S)-2- (4-( tert-butyl)benzamido)-3- (4-(5-( 5-hexyl-l, 2, 4-oxadiazol-3- yl)pyrimidin-2-yl)phenyl)propanoate (Compound 248)

[00324] Prepared using General Procedure 5. A solution of heptanoic acid (7 mg, 0.05 mmol), HOBt (12 mg, 0.09 mmol) and EDC (13 mg, 0.09 mmol) was heated at 80°C for 2 h. The reaction mixture was diluted with EtOAc and washed with

NaHC0 3 . The organic layer was dried over Na 2 S0 4 and concentrated. The resulting mixture was dissolved in EtOH (2 mL) and heated for 45 min at 80°C in a microwave reactor. The mixture was concentrated and purified by preparatory HPLC to provide 1.5 mg of tert-butyl (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(5-hexyl-l,2,4-oxadi azol-3- yl)pyrimidin-2-yl)phenyl)propanoate. LCMS-ESI (m/z) calculated for C 3 6H 45 N50 4 : 611.8; no m/z observed, t R = 5.5 min (Method 1). 1H NMR (400 MHz, CDC1 3 ) δ 9.45 (s, 2H), 8.44 (d, J = 8.3 Hz, 2H), 7.71 (d, J = 8.5 Hz, 2H), 7.48 (d, J = 8.5 Hz, 2H), 7.38 (d, J = 8.3 Hz, 2H), 6.80 (d, J = 7.3 Hz, 1H), 5.04 (dd, J = 12.7, 5.5 Hz, 1H), 3.37 (ddd, J = 18.9, 13.8, 5.5 Hz, 2H), 3.02 (t, J = 7.6 Hz, 2H), 1.92 (dt, J = 15.3, 7.5 Hz, 2H), 1.49 (s, 9H), 1.44 - 1.28 (m, 15H), 0.93 (t, J = 7.1 Hz, 3H).

[00325] (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(5-hexyl-l,2,4-oxadi azol-3-yl) pyrimidin-2-yl) phenyl)propanoate was deprotected using General Procedure 8 to provide 1.4 mg (6% overall) of (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(5-hexyl- 1,2,4- oxadiazol-3-yl)pyrimidin-2-yl)phenyl) propanoic acid Compound 248. LCMS-ESI (m/z) calculated for C32H37N5O4: 555.68; no m/z observed, fa = 11.03 min {Method 2). 1H NMR (400 MHz, CDC1 3 ) δ 9.41 (s, 2H), 8.47 (d, J = 8.2 Hz, 2H), 7.66 (d, J = 8.4 Hz, 2H), 7.42 (dd, J = 15.1, 8.4 Hz, 4H), 6.60 (d, J = 6.8 Hz, 1H), 5.21 - 4.95 (m, 1H), 3.43 (ddd, J = 20.0, 14.0, 5.6 Hz, 2H), 3.05 - 2.90 (m, 2H), 1.98 - 1.76 (m, 2H), 1.55 - 1.22 (m, 15H), 0.91 (t, J = 7.0 Hz, 3H).

Tert-butyl (S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4- h droxyphenyl)pyrimidin-2-yl)phenyl)propanoate

[00326] Prepared using General Procedure 10. To a degassed solution of tert- butyl (S)-3 -(4-(5 -bromopyrimidin-2-yl) phenyl)-2-(5 -(tert-butyl)tfiiophene-2- carboxamido) propanoate INT-17 (180 mg, 0.3 mmol), sodium carbonate (70 mg, 0.7 mmol) and 4-hydroxyphenylboronic acid (55 mg, 0.4 mmol) in 5 mL of 2/2/1 MeCN/ THF/ H 2 0 was added Pd(dppf)Cl 2 (24 mg, 0.03mmol). The reaction mixture was heated at 110°C for 45 min in a microwave reactor. The mixture was filtered through celite, concentrated, then dissolved in DCM and washed with H 2 0. The organic layer was concentrated and purified by prep HPLC to provide 131 mg (78%) of tert-butyl (S)-2-(5-(tert-butyl)thiophene-2-carboxamido) -3-(4-(5-(4-hydroxyphenyl) pyrimidin-2- yl)phenyl) propanoate. LCMS-ESI (m/z) calculated for C 32 H 35 N 3 O 4 S: 557.7; no m/z observed, t R = 4.08 min (Method 1). 1H NMR (400 MHz, CDCI 3 ) δ 8.98 (s, 2H), 8.35 (d, J= 8.1 Hz, 2H), 7.49 (d, J= 8.6 Hz, 2H), 7.40 - 7.31 (m, 3H), 6.94 (d, J= 8.5 Hz, 2H), 6.81 (d, J= 3.8 Hz, 1H), 6.51 (d, J= 7.5 Hz, 1H), 5.00 (dd, J= 12.9, 5.8 Hz, 1H), 3.28 (qd, J= 13.8, 5.6 Hz, 2H), 1.47 (s, 9H), 1.39 (s, 9H).

(S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4- (decyloxy)phenyl)- rimidin-2-yl)phenyl)propanoic acid (Compound 249)

[00327] Prepared using General Procedure 12. To a solution of tert-butyl (S)-2- (5 -(tert-butyl)thiophene-2-carboxamido) -3 -(4-(5 -(4-hydroxyphenyl) pyrimidin-2- yl)phenyl) propanoate (20 mg, 0.04 mmol) in DMF (0.5 mL) were added 1- bromodecane (8 ί, 0.05 mmol) and K 2 CO 3 (8 mg, 0.05 mmol). The reaction mixture was heated at 40°C for 18 h, then diluted with DCM and washed with H 2 0. The organic layer was dried over Na2S0 4 and concentrated. The crude material was deprotected using General Procedure 8 then purified by preparatory HPLC to provide 3.9 mg (17%) of (5)-2-(5-(fert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4- (decyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoic acid Compound 249. LCMS-ESI (m/z) calculated for C 38 H 47 N 3 O 4 S: 641.9; no m/z observed, fa = 13.49 min (Method 2). 1H NMR (400 MHz, CDCI 3 ) δ 9.01 (s, 2H), 8.36 (d, J = 8.1 Hz, 2H), 7.56 (d, J = 8.7 Hz, 2H), 7.44 (d, J = 8.2 Hz, 2H), 7.33 (d, J = 3.8 Hz, 1H), 7.03 (d, J = 8.8 Hz, 2H), 6.80 (d, J = 3.8 Hz, 1H), 6.54 (d, J = 6.8 Hz, 1H), 5.13 (d, J = 6.8 Hz, 1H), 4.01 (t, J = 6.6 Hz, 2H), 3.44 (d, J = 4.9 Hz, 2H), 1.91 - 1.72 (m, 2H), 1.47 (dd, J = 15.0, 7.3 Hz, 2H), 1.38 (s, 9H), 1.28 (s, 12H), 0.88 (t, J = 6.8 Hz, 3H). [00328] Compounds 250 - 252 were prepared from tert-butyl (5)-2-(5-(tert- butyl)thiophene-2-carboxamido) -3 -(4-(5 -(4-hydroxyphenyl) pyrimidin-2-yl)phenyl) propanoate using General Procedure 12 followed by General Procedure 8.

(S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(tert-butyl)pi peridin-l-yl)pyrimidm^ 2-yl)phenyl)propanoic acid (Compound 253)

[00329] Prepared using General Procedure 11. Into a degassed solution of INT- 14 (50 mg, 0.09 mmol), sodium tert-butoxide (18 mg, 0.19 mmol) and 4-tert- butylpiperidine HC1 (23 mg, 0.11 mmol) in dioxane (2.5 mL) were added Pd 2 (dba) 3 (9 mg, 0.01 mmol) and 2-dicyclohexylphosphino-2'-(N,N-dimethylamino)biphenyl (6 mg, 0.015 mmol). The reaction mixture was heated for 45 min at 120°C in a microwave reactor. The mixture was diluted with EA and washed with NaHC0 3 . The organic layer was dried over Na 2 S0 4 , concentrated, and purified by preparatory HPLC. The isolated intermediate was deprotected using General Procedure 8 to provide 2.9 mg (6%) of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(tert-butyl)piper idin-l-yl)pyrimidin- 2-yl)phenyl)propanoic acid Compound 253. LCMS-ESI (m/z) calculated for

C 33 H 42 N 4 0 3 : 542.7; found 543.3 [M+H] + , t R = 10.79 min (Purity). 1H NMR (400 MHz, CDC1 3 ) δ 8.52 (s, 2H), 8.23 (d, J= 8.0 Hz, 2H), 7.72 (d, J= 8.4 Hz, 2H), 7.44 (dd, J = 11.3, 8.4 Hz, 4H), 6.79 (d, J= 6.8 Hz, 1H), 5.18 (d, J= 6.5 Hz, 1H), 3.89 (d, J= 11.9 Hz, 2H), 3.47 (d, J= 5.2 Hz, 2H), 2.83 (t, J= 11.5 Hz, 2H), 1.88 (d, J= 12.0 Hz, 2H), 1.52 - 1.37 (m, 2H), 1.34 (s, 9H), 1.24 (dd, J= 24.7, 12.8 Hz, 1H), 0.92 (s, 9H).

[00330] Compound 254 was prepared from INT-14 using General Procedure 11 then General Procedure 8. Tert-butyl (S)-3-(4-( 5-(2H-tetrazol-5-yl)pyrimidin-2-yl)phenyl)-2-(4- ( tert-butyl)- benzamido) propanoate

[00331 ] Into a solution of tert-butyl (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5- cyanopyrimidin-2-yl)phenyl)propanoate INT-18 (34 mg, 0.07mmol) in DMF (2 mL) were added NH 4 C1 (7.5 mg, 1.4 mmol) and NaN 3 (7 mg, 0.1 mmol). The reaction mixture was heated at 100°C for 3 h then diluted with EA and washed with NaHC0 3 . The organic layer was dried over Na 2 S0 4 , concentrated, and purified by preparatory HPLC to provide 4.6 mg (12%) of tert-butyl (5)-3-(4-(5-(2H-tetrazol-5-yl)pyrimidin-2- yl)phenyl)-2-(4-(tert-butyl)benzamido)propanoate. LCMS-ESI (m/z) calculated for C 29 H 33 N 7 0 3 : 527.6; no m/z observed, t R = 3.83 min (Method 1). l U NMR (400 MHz, CDC1 3 ) δ 9.35 (s, 2H), 8.42 (d, J = 8.1 Hz, 2H), 7.75 (d, J = 8.4 Hz, 2H), 7.47 (d, J = 8.5 Hz, 2H), 7.43 (d, J = 8.2 Hz, 2H), 7.11 (d, J = 7.8 Hz, 1H), 5.13 (dd, J = 14.4, 7.1 Hz, 1H), 3.28 (ddd, J = 21.0, 13.6, 6.7 Hz, 2H), 1.47 (d, J = 6.8 Hz, 9H), 1.33 (s, 9H).

[00332] Compound 255 was prepared from tert-butyl (5)-3-(4-(5-(2H-tetrazol-5- yl)pyrimidin-2-yl)phenyl)-2-(4-(tert-butyl)benzamido)propano ate using General Procedure 12 then General Procedure 8.

[00333] Compound 256 was prepared from INT-14 and 5-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)isoindolin-l-one using General Procedures 10, 12 and 8.

[00334] Compound 257 was prepared from INT-14 and 6-Hydroxypyridine-3- boronic acid pinacol ester using General Procedures 10, 12 and 8.

[00335] Compound 258 was prepared from INT-13 and 5-(benzyloxy)-2- chloropyrimidine using General Procedure 10, followed by General Procedure 8.

[00336] Compounds 259 and 260 were prepared from INT-14 and the appropriate boronic acid using General Procedures 10 then 8. Tert-butyl 4-(4-(heptyloxy)phenyl)-3-oxopiperazine-l-carboxylate

[00337] To a stirring solution of l-bromo-4-(heptyloxy)benzene (447 mg, 1.65 mmol) in dioxane (5 mL) were added tert-butyl 3-oxopiperazine-l-carboxylate (330 mg, 1.65 mmol), copper I iodide (31.4 mg, 0.17 mmol), (\R,2R)-N\,N2- dimethylcyclohexane-l,2-diamine (234 mg, 1.65 mmol) and potassium carbonate (456 mg, 3.30 mmol). The reaction mixture was heated at 120°C for 16 h. The reaction mixture was passed through a plug of celite, eluted with EA (50 mL). The organics were washed with ammonium chloride (25 mL), water (25 mL) and brine (25 mL) then dried over MgS0 4 and concentrated to afford 602 mg (89%) of tert-butyl 4-(4- (heptyloxy)phenyl)-3-oxopiperazine-l-carboxylate. LCMS-ESI (m/z) calculated for C22H 34 N 2 0 4 : 390.5; found 319.0 [M+H] + , t R = 2.90 min. (Method 4). l-(4-(heptyloxy)phenyl)piperazin-2-one

[00338] To tert-butyl 4-(4-(heptyloxy)phenyl)-3-oxopiperazine-l-carboxylate (540 mg, 1.38 mmol) was added 4M HC1 in dioxane (2.07 mL, 8.30 mmol). The reaction mixture was stirred at room temperature for 2 h. The precipitate was filtered, washed with hexane (5 mL) and dried. The crude product was purified by column chromatography (79/20/1 DCM/MeOH/NH 4 ) to afford 325 mg (80%) of l-(4- (heptyloxy)phenyl)piperazin-2-one as a colorless solid. LCMS-ESI (m z) calculated for C17H26N2O2: 290.4; found 291.0 [M+H] + , t R = 1.49 min. (Method 4).

[00339] Compound 261 was prepared from INT-12 and l-(4- (heptyloxy)phenyl)piperazin-2-one using General Procedures 11 and 8. [00340] Compound 262 was prepared in a similar fashion from INT-12 and l-(4- (heptyloxy)phenyl)imidazolidin-2-one using General Procedures 11 and 8.

[00341] Compound 263 was prepared using (5)-methyl 2-amino-3-(4- nitrophenyl)propanoate hydrochloride, 4-(tert-butyl)benzoic acid and l-(4- (heptyloxy)phenyl)piperidin-4-one using General Procedures 7, 14, 15 then 4.

Tert-but l 4-(4-(heptyloxy)phenyl)-4-hydroxypiperidine-l-carboxylate

[00342] To a stirring solution of l-bromo-4-(heptyloxy)benzene (668 mg, 2.46 mmol) in THF (5 mL) at -78°C was added butyllithium (985 μΐ, 2.46 mmol). After 30 min, a solution of tert-butyl 4-oxopiperidine-l-carboxylate (491 mg, 2.46 mmol) in THF (2 mL) was added. After 10 min, the cooling bath was removed and the reaction mixture stirred for 16 h. The reaction mixture was poured onto NH 4 C1 (50 mL) and extracted with Et 2 0 (3 x 20 mL). The combined organics were washed with water (20 mL), dried over MgSC^ and evaporated. The crude product was purified by column chromatography (5-70% AcMe in z ' so-hexanes) to afford 0.4 g (33%) of tert-butyl 4-(4- (heptyloxy)phenyl)-4-hydroxypiperidine-l-carboxylate. LCMS-ESI (m/z) calculated for C23H37NO4: 391.5; found 414.0 [M+Na] + , t R = 2.24 min. (Method 4).

4-(4-(heptyloxy)phenyl)piperidine (INT-19)

[00343] To a stirring solution of tert-butyl 4-(4-(heptyloxy)phenyl)-4- hydroxypiperidine-l-carboxylate (388 mg, 0.99 mmol) and triethylsilane (791 μΐ, 4.95 mmol) in DCM (2 mL) cooled to -30 °C was slowly added 2,2,2-trifluoroacetic acid (379 μΐ, 4.95 mmol) in a drop- wise fashion. The reaction mixture was allowed to warm slowly and stirring continued for 16 h. The reaction mixture was poured onto ice- water/NaOH (50 mL/5 mL, 2 M) and extracted with DCM (3 x 20 mL). The combined organic extracts were washed successively with water (50 mL) and NaHC0 3 (20 mL), dried over MgSC^ and evaporated to afford 166 mg (58%) of 4-(4- (heptyloxy)phenyl)piperidine INT-19 as a white, waxy solid. LCMS-ESI (m/z) calculated for Ci 8 H 29 NO: 275.4; found 276.0 [M+H] + , t R = 2.88 min. (Method 11).

[00344] Compound 264 was prepared using INT-12 and INT-19 using General Procedures 11 then 8.

[00345] Compound 265 was prepared in a similar fashion to 264 using INT-12 and 3-(4-(heptyloxy)phenyl)pyrrolidine using General Procedures 11 then 8.

[00346] Compound 266 can be prepared using INT-12 and l-([l,l'-biphenyl]-4- yl)piperazine using General Procedures 11 then 8.

[00347] Compound 267 was prepared using INT-12, tert-butyl 4-(4- hydroxyphenyl)piperazine-l-carboxylate and 1-bromoheptane using General

Procedures 12, 8, 11 then 8.

[00348] Compound 268 was prepared using INT-12, tert-butyl 1,4-diazepane-l- carboxylate and l-bromo-4-(heptyloxy)benzene using General Procedures 11, 8, 11 then 8.

[00349] Compound 269 was prepared using 5-bromo-2-iodopyridine, INT-13 and (4-(heptyloxy)phenyl)boronic acid using General Procedures 10, 10, and 8 sequentially.

[00350] Compound 270 was prepared using 5-bromo-2-iodopyridine, (4- (heptyloxy)phenyl)boronic acid and INT-13 using General Procedures 10, 10, and 8 sequentially.

[00351] Compound 271 was prepared using 5-bromo-2-iodopyrimidine, (4- (heptyloxy)phenyl)boronic acid and INT-13 using General Procedures 10, 10, and 8 sequentially.

[00352] Compound 272 was prepared using 2-bromo-5-iodopyrazine, (4- (heptyloxy)phenyl)boronic acid and INT-13 using General Procedures 10, 10, and 8 sequentially. [00353] Compound 273 was prepared using 3-chloro-6-iodopyridazine, (4- (heptyloxy)phenyl)boronic acid and INT-13 using General Procedures 10, 10, and 8 sequentially.

3-(4-bromo henyl)-6-(4-(heptyloxy)phenyl)-l,2,4-triazine (INT-20)

[00354] To a stirring solution of 4-bromobenzohydrazide (1.85 g, 8.62 mmol) in ethanol (10 mL) was added acetic acid (1 mL). The reaction mixture was stirred at 60°C for 30 min then 2-bromo-l-(4-(heptyloxy)phenyl)ethanone (1.35 g, 4.31 mmol) INT-4 and sodium acetate (0.389 g, 4.74 mmol) were added and the mixture heated to reflux for 30 min. The reaction mixture was cooled to RT and the resultant precipitate was filtered and washed with z ' so-hexanes (20 mL) then dried. The solid was dissolved in NMP and heated to 120 °C for 16 h. The crude material was cooled to RT, diluted with Et 2 0 (4 mL), filtered, triturated with ethanol (3 x 2 mL), filtered and dried to afford 241 mg (13%) of 3-(4-bromophenyl)-6-(4-(heptyloxy)phenyl)-l,2,4-triazine INT-20 as an orange solid. LCMS-ESI (m/z) calculated for C 22 H 24 BrN 3 0: 425.1; found 426.3

[M+H] + , t R = 3.40 min (Method 8).

[00355] Compound 274 was prepared in a similar fashion to 79 using 3-(4- bromophenyl)-6-(4-(heptyloxy)phenyl)-l,2,4-triazine INT-20 in place of 2-(4- bromophenyl)-4-(4-(heptyloxy)phenyl)thiazole.

6- ( 4-bromophenyl)-3- ( 4-(heptyloxy)phenyl)-l , 2, 4-triazine (INT-21 )

[00356] To a stirring solution of 4-(heptyloxy)benzohydrazide (400 mg, 1.60 mmol) in ethanol (15 mL) was added acetic acid (1 mL). The reaction mixture was stirred at 60°C for 30 min then 2-bromo-l-(4-bromophenyl)ethanone (222 mg, 0.80 mmol) and sodium acetate (72.1 mg, 0.88 mmol) were added and the solution heated to reflux for 2 h. The reaction mixture was cooled to RT and the resultant crystals were filtered, washed with z ' so-hexanes (20 mL) then dried to afford 108 mg (31%) of 6-(4- bromophenyl)-3-(4-(heptyloxy)phenyl)-l,2,4-triazine INT-21. LCMS-ESI (m/z) calculated for C 22 H 24 BrN 3 0: 425.1; found 426.1 [M+H] + , t R = 3.38 min (Method 8).

[00357] Compound 275 was prepared in a similar fashion to 274 using 6-(4- bromophenyl)-3-(4-(heptyloxy)phenyl)-l,2,4-triazine INT-21 in place of 3-(4- bromophenyl)-6-(4-(heptyloxy)phenyl)- 1 ,2,4-triazine.

[00358] Compound 276 was prepared using 274 using General Procedures 7 and

8.

[00359] Compounds 277 and 278 were prepared using INT-16 and 5-bromo-2- iodopyridine using General Procedures 10, 10, and 8 sequentially.

[00360] Compounds 279 and 280 were prepared using INT-16 and 3-chloro-6- iodopyridazine using General Procedures 10, 10, and 8 sequentially.

[00361] Compounds 281 and 282 were prepared using INT-16 and 2-bromo-5- iodopyrazine using General Procedures 10, 10, and 8 sequentially.

[00362] Compound 283 was prepared from Compound 279 and tert-butyl glycinate using General Procedures 7 and 8 sequentially.

[00363] Compound 284 was prepared from Compound 281 and tert-butyl glycinate using General Procedures 7 and 8 sequentially.

[00364] Compound 285 was prepared from Compound 277 and tert-butyl glycinate using General Procedures 7 and 8 sequentially.

2-(4-(he tyloxy)phenyl)-2-oxoethyl 4-bromobenzoate

[00365] To a solution of 2-bromo-l-(4-(heptyloxy)phenyl)ethanone INT-4 (1.3 g, 4.2 mmol) and 4-bromobenzoic acid (0.70 g, 3.5 mmol) in ACN (30 mL) was added TEA (0.72 ml, 5.2 mmol). After stirring overnight, the mixture was poured onto aq. citric acid and EA then stirred for 10 min before the solid was collected by filtration. The cake was washed with water and iso-hexanes then dried to provide 905 mg (57%) of 2-(4-(heptyloxy)phenyl)-2-oxoethyl 4-bromobenzoate. LCMS-ESI (m/z) calculated for C 22 H 25 Br0 4 : 432.1; found 433.2 [M+H] + , t R = 3.24 min (Method 8).

2-(4-bromo henyl)-5-(4-(heptyloxy)phenyl)-lH-imidazole

[00366] To a solution of 2-(4-(heptyloxy)phenyl)-2-oxoethyl 4-bromobenzoate (905 mg, 2.09 mmol) in toluene (6 ml) was added CH 3 COONH 4 (1600 mg, 20.9 mmol). After heating overnight at 115°C, the reaction mixture was diluted with aq. NaHC0 3 and extracted into DCM. The organic layers were combined, dried over MgS0 4 , filtered, and the solvent was removed under reduced pressure. The crude reaction mixture was purified by chromatography (EA/ hexanes) to provide 370 mg (33%) of 2- (4-bromophenyl)-5-(4-(heptyloxy)phenyl)-lH-imidazole. LCMS-ESI (m/z) calculated for C 22 H 25 BrN 2 0: 412.1; found 413.2 [M+H] + , t R = 2.33 min (Method 8).

2-(4-bromophenyl)-5-(4-(heptyloxy)phenyl)-l-((2-(trimethy lsilyl)ethoxy)methyl)- lH-imidazole

[00367] To a solution of 2-(4-bromophenyl)-5-(4-(heptyloxy)phenyl)-lH- imidazole (370 g, 900 mmol) in DMF (4 ml) was added NaH (40 mg, 980 mmol). After 2 h, 2-(trimethylsilyl)ethoxymethyl chloride (160 g, 990 mmol) in THF (2 ml) was added dropwise and reaction mixture was stirred overnight. The reaction mixture was diluted with EA and washed with aq. NaHC0 3 . The organics were dried over MgSC^, filtered, and the solvent was removed under reduced pressure. The crude product was purified by chromatography (EA / hexane) to afford 32 mg (65%) of 2-(4- bromophenyl)-5 -(4-(heptyloxy)phenyl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H- imidazole as a tan solid. LCMS-ESI (m/z) calculated for C 2 8H 3 9BrN 2 0 2 Si: 542.2; found 543.3 [M+H] + , t R = 3.35 min (Method 8).

(S)-methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(4-(4-(heptyloxy)phenyl) -l-((2- trimethylsilyl)ethoxy)methyl)-lH-imidazol-2-yl)phenyl)propan oate

[00368] A stirred suspension of zinc (68 mg, 1.03 mmol) in DMF (2 mL) was treated with I 2 (12 mg, 0.05 mmol). After the color disappeared, ((i?)-methyl 2-((tert- butoxycarbonyl)amino)-3-iodopropanoate (110 mg, 0.34 mmol) and further I 2 (12 mg, 0.05 mmol) were added. After 30 min, the mixture was de-gassed then 2-(4- bromophenyl)-5 -(4-(heptyloxy)phenyl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H- imidazole (170 mg, 0.31 mmol), dicyclohexyl(2',6'-dimethoxy-[l,l'-biphenyl]-2- yl)phosphine (7 mg, 0.02 mmol) and Pd 2 (dba) 3 (8 mg, 7.8 μιηοΐ) were added. After further de-gassing, DMF (2 mL) was added and the reaction mixture was heated at 50°C overnight. The reaction mixture purified by column chromatography (EA / hexane) to provide 55 mg (25%) of (5)-methyl 2-((tert-butoxycarbonyl)amino)-3-(4-(4-(4- (heptyloxy)phenyl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H-imidazol-2- yl)phenyl)propanoate as a colorless oil. LCMS-ESI (m/z) calculated for C 37 H 55 N 3 O 6 S1: 665.9; found 666.4 [M+H] + , t R = 3.10 min (Method 8).

(S)-methyl 2-amino-3-(4-(4-(4-(heptyloxy)phenyl)-lH-imidazol-2-yl)pheny l)- ropanoate

[00369] (5)-methyl 2-amino-3-(4-(4-(4-(heptyloxy)phenyl)-lH-imidazol-2- yl)phenyl)propanoate was prepared from (S)-methyl 2-((tert-butoxycarbonyl)amino)-3- (4-(4-(4-(heptyloxy)phenyl) - 1 -((2-(trimethylsilyl) ethoxy)methyl)- 1 H-imidazol-2-yl) phenyl) propanoate using General Procedure 8. LCMS-ESI (m/z) calculated for C26H33N3O3 : 435.6; found 436.3 [M+H] + , t R = 1.43 min (Method 8).

(S)-2-(4-(tert-butyl)benzamido)-3-(4-(4-(4-(heptyloxy)phe nyl)-lH-imidazol-2- l)phenyl)propanoic acid hydrochloride (Compound 286)

[00370] To a solution of 4-(tert-butyl)benzoic acid (25 mg, 0.14 mmol), (S)- methyl 2-amino-3 -(4-(4-(4-(heptyloxy)phenyl)- 1 H-imidazol-2-yl)phenyl)propanoate (55 mg, 0.13 mmol), and TEA (53 μΐ, 0.38 mmol) in DMF (1 mL) was added HATU (53 mg, 0.14 mmol). The reaction mixture was stirred at RT for 2 h, diluted in DCM, and washed aq. NaHCC^. The organic layer was dried, concentrated and purified by chromatography (EA / hexane) to provide 14 mg (17%) of methyl (S)-2-(4-(tert- butyl)benzamido)-3-(4-(4-(4-(heptyloxy)phenyl)-lH-imidazol-2 -yl)phenyl)propanoate. LCMS-ESI (m/z) calculated for C37H45N3O4: 595.8; found 596.4 [M+H] + , t R = 2.33 min. (Method 8).

[00371] The isolated ester intermediate was deprotected using General

Procedure 4 to provide 14 mg (17.5%) of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(4-(4- (heptyloxy)phenyl)-lH-imidazol-2-yl)phenyl)propanoic acid hydrochloride Compound 286 as a light tan solid. LCMS-ESI (m/z) calculated for C36H43N3O4: 581.8; found 582.4 [M+H] + , t R = 6.56 min (Method 9).

4-bromo-l- 4-(heptyloxy)phenyl)-lH-imidazole

[00372] Into a vial was charged (4-(heptyloxy)phenyl)boronic acid (1.00 g, 4.24 mmol), 4-bromo-lH-imidazole (0.31 g, 2.1 mmol), Cu-(TMEDA) 2 (0H) 2 C1 2 (0.10 g, 0.21 mmol) and DCM (12 ml). After stirring at RT for 42 h, the mixture was purified by chromatography (EA / hexane) to provide 80 mg of impure product. Further purification by chromatography (CAN / DCM) provided 42 mg (6%) of 4-bromo-l-(4- (heptyloxy)phenyl)-lH-imidazole as a colourless oil. LCMS-ESI (m/z) calculated for Ci 6 H 2 iBrN 2 0: 336.1; found 337.1 [M+H] + , t R = 2.71 min (Method 8).

(S)-2-(4-(tert-butyl)benzamido)-3-(4-(l-(4-(heptyloxy)phe nyl)-lH-imidazol-4- l)phenyl)propanoic acid (Compound 287)

[00373] Prepared using General Procedure 10. Into a vial containing INT-13 (96 mg, 0.19 mmol) and 4-bromo-l-(4-(heptyloxy)phenyl)-lH-imidazole (64 mg, 0.19 mmol) in 2/2/1 THF/CAN/H 2 0 (3 niL) was added Na 2 C0 3 (40 mg, 0.38 mmol). The reaction mixture was degassed and Pd(dppf)Cl 2 (14 mg, 0.02 mmol) was added. After heating at 120°C for 30 min in a microwave reactor, the mixture was diluted with EA, washed with aq. NaHC0 3 , dried over MgS0 4 and concentrated. Purification by chromatography (EA/ hexanes) provided 14 mg (12%) of the intermediate tert-butyl (5)-2-(4-(tert-butyl)benzamido)-3-(4-(l-(4-(heptyloxy)phenyl )-lH-imidazol-4- yl)phenyl)propanoate as a white solid.

[00374] The intermediate was deprotected according to General Procedure 8 to provide 9 mg (8%) of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(l-(4-(heptyloxy)phenyl )- lH-imidazol-4-yl)phenyl)propanoic acid, Compound 287 as a white solid. LCMS-ESI (m/z) calculated for C 36 H 43 N 3 0 4 : 581.3; found 582.2 [M+H] + , t R = 8.33 min (Method 9).

(S)-2-(4-(tert-butyl)benzamido)-3-(4-(l-(4'-methyl-[l,l '-biphenyl]-4-yl)-lH- razol-4-yl)phenyl)propanoic acid (Compound 288)

[00375] Prepared using General Procedure 10. Into a vial containing INT-13 (100 mg, 0.20 mmol) and 4-bromo-l-(4'-methyl-[l, -biphenyl]-4-yl)-lH-pyrazole (63 mg, 0.201 mmol) in 2/1 ACN/H 2 0 (3 mL) was added sat aq. NaHC0 3 (670 i , 0.60 mmol). The reaction mixture was degassed and Pd(dppf)Cl 2 (15 mg, 0.02 mmol) was added. After heating at 120°C for 60 min in a microwave reactor, the mixture was diluted with DCM, washed with aq. NaHC0 3 , passed through a phase separation cartridge, and concentrated. Purification by chromatography (EA / hexane) provided 58 mg (47%) of the intermediate tert-butyl (S)-2-(4-(tert-butyl)benzamido) -3-(4-(l-(4'- methyl-[l,l'-biphenyl]-4-yl) -lH-pyrazol-4-yl)phenyl) propanoate as a white solid. LCMS-ESI (m/z) calculated for C 40 H 43 N 3 O 3 : 613.8; found 614.0 [M+H] + , t R = 3.02 min (Method 8). The intermediate was stirred in 4M HC1 / dioxane for 132 h and filtered. The resulting solid was washed with hexane to provide 13 mg of solid product. The filtrate was loaded onto a strong anion exchange (SAX) column, washed with MeOH, and eluted with 5% AcOH in MeOH. The elution liquors were combined with the trituration solid and concentrated in vacuo to afford 18 mg (32%) of (S)-2-(4-(tert- butyl)benzamido)-3-(4-( 1 -(4'-methyl-[ 1 , 1 '-biphenyl]-4-yl)- 1 H-pyrazol-4- yl)phenyl)propanoic acid 288 as a white solid. LCMS-ESI (m/z) calculated for C 36 H 35 N 3 0 3 : 557.3; found 558.0 [M+H] + , t R = 9.37 min (Method 9). Methyl 2-(4-bromophenyl)-2-(5-(tert-but l)thiophene-2-carboxamido)acetate

[00376] Prepared using General Procedure 7. To a solution of methyl 2-amino- 2-(4-bromophenyl)acetate, HC1 (730 mg, 2.6 mmol), 5-(tert-butyl)thiophene-2- carboxylic acid (480 mg, 2.6 mmol) and TEA (1090 μΐ, 7.8 mmol) in DMF (lOmL) was added HATU (1090 mg, 2.9 mmol). After stirring overnight, the reaction mixture was diluted with EA (lOOmL) and washed with 1M HC1 (100 mL) and brine. The organic layer was dried over Mg 2 S0 4 , concentrated, and purified by chromatography (EA /hexane) to provide 900 mg (76%) of methyl 2-(4-bromophenyl)-2-(5-(tert- butyl)thiophene-2-carboxamido)acetate as a white powder. LCMS-ESI (m/z) calculated for Ci 8 H 2 oBrN0 3 S: 410.3; found 412.0 [M+2] + , t R = 2.71 min (Method 8).

Methyl 2-(5-(tert-butyl)thiophene-2-carboxamido)-2-(4-(4,4,5,5-tetr amethyl- l,3,2-dioxaborolan-2- l)phenyl)acetate

[00377] Prepared using General Procedure 10. A solution of 2-(4- bromophenyl)-2-(5-(tert-butyl)thiophene-2-carboxamido)acetat e (900 mg, 2.2

KOAc (650 mg, 6.6 mmol) and 4,4,4 * ,4 * ,5,5,5 * ,5 * -octamethyl-2,2 , -bi(l,3,2- dioxaborolane) (670 mg, 2.6 mmol) in DMSO (10 mL) at 40°C was de-gassed. PdCl 2 dppf (80 mg, 0.11 mmol) was added and the mixture was heated at 100°C for 3 h. The reaction mixture was purified by chromatography (EA / hexane with 1% TEA) to provide 491 mg (41%) of methyl 2-(5-(tert-butyl)thiophene-2-carboxamido) -2-(4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl) phenyl) acetate. LCMS-ESI (m/z) calculated for C 2 4H 32 BN0 5 S: 457.4; found 458.0 [M+H] + , t R = 2.89 min (Method 8).

2-(4-(5-bromopyrimidin-2-yl)phenyl)-2-(5-(tert-butyl)thio phene-2- carboxamido) acetic acid

[00378] Prepared using General Procedure 10. A mixture of methyl 2-(5-(tert- butyl)thiophene-2-carboxamido) -2-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl) phenyl) acetate (320 mg, 0.71 mmol) and 5-bromo-2-iodopyrimidine (220 mg, 0.78 mmol) in THF (2 mL) and MeCN (2 mL) was treated with saturated aq. NaHC0 3 (1600 μΐ, 1.40 mmol) and de-gassed (N 2 bubbling). PdCl 2 dppf (26 mg, 0.04 mmol) was added and the mixture was heated at 120°C for 30 min in a microwave reactor. The mixture was poured onto H 2 0 (30 mL), acidified with AcOH and extracted with EA (3 x 15 mL). The combined organics were dried over MgS0 4 , evaporated, and purified by chromatography (EA / hexane with 1% AcOH) to provide 160 mg (46%) of 2-(4-(5- bromopyrimidin-2-yl)phenyl)-2-(5 -(fert-butyl)thiophene-2-carboxamido)acetic acid as a white solid. LCMS-ESI (m z) calculated for C 2 iH 20 BrN 3 O 3 S: 473.0; found 474.0 [M+H] + , t R = 2.68 min (Method 8). (S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4-(he ptyloxy)phenyl)- rimidin-2-yl)phenyl)propanoic acid (Compound 289)

[00379] Prepared using General Procedure 10. A solution of 2-(4-(5- bromopyrimidin-2-yl)phenyl)-2-(5-(tert-butyl)thiophene-2-car boxamido)acetic acid (160 mg, 0.34 mmol), (4-(heptyloxy)phenyl)boronic acid (94 mg, 0.40 mmol) and sat aq. NaHC0 3 (930 μΐ, 0.84 mmol) in ACN (1.5 mL) and THF (1.5 mL) was degassed (N 2 bubbling). PdCl 2 (dppf) (262 mg, 0.34 mmol) was added and the reaction mixture was heated at 110°C in a microwave reactor for 50 min. The reaction was partitioned between EA and H 2 0. The organic layer was dried over MgSC^, filtered, concentrated and purified by chromatography (EA / hexane with 1% AcOH) to afford 113 mg (55%) of 2-(5-(tert-butyl)thiophene-2-carboxamido)-2-(4-(5-(4-(heptyl oxy)phenyl)pyrimidin- 2-yl)phenyl)acetic acid Compound 289 as a white solid. LCMS-ESI (m/z) calculated for C34H39N3O4S: 585.3; found 586.0 [M+H] + , t R = 3.37 min (Method 9).

(S)-N-(l-amino-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2-y l)phenyl)-l- oxopropan-2-yl)-4-(tert-butyl)benzamide

[00380] A solution of Compound 85 (245 mg, 0.413 mmol) in DMF (5 mL) was treated with NH 4 C1 (180 mg, 3.3 mmol), DIEA (760 μΐ, 4.1 mmol) and HATU (170 mg, 0.4 mmol). After stirring overnight, the reaction mixture was diluted with EA (50 mL), washed with aq. 0.5 M HC1 (100 mL) and brine (20 mL), then dried over MgS0 4 and concentrated. The residue was re-slurried from ACN (4 mL) to afford 204 mg (77%) of (S)-N-( 1 -amino-3 -(4-(5 -(4-(heptyloxy)phenyl)pyrimidin-2-yl)phenyl)- 1 -oxopropan-2- yl)-4-(tert-butyl)benzamide as a fine white solid. LCMS-ESI (m/z) calculated for C 37 H 44 N 4 0 3 : 592.3; found 593.0 [M+H] + , t R = 3.43 min (Method 6).

(S)-methyl 3-(4-(tert-butyl)benzamido)-4-(4-hydroxyphenyl)butanoate

[00381] Prepared using General Procedure 7. A solution of (5)-methyl 3-amino- 4-(4-hydroxyphenyl)butanoate hydrochloride (2.1 g, 8.7 mmol), 4-(tert-butyl)benzoic acid (1.6 g, 9.0 mmol) and DIEA (3.5 ml, 18.8 mmol) in DMF (20 mL) and DCM (20 mL) was treated with HATU (3.3 g, 8.5 mmol). After 1 h, the mixture was poured onto 1M HC1 (100 mL) and extracted with EA (3 x 50 mL). The combined organic extracts were washed successively with 1M HC1 (50 mL), water (50 mL) and brine (20 mL), then dried over MgS0 4 and concentrated. The resulting residue was purified by chromatography (EA/ hexane) to provide 2.3 g (72%) of (S)-methyl 3-(4-(tert- butyl)benzamido)-4-(4-hydroxyphenyl) butanoate as white needles. LCMS-ESI (m/z) calculated for C 22 H 27 NO: 369.4, found 370.0 [M+H] + , t R = 2.52 min (Method 6). (S)-methyl 3-(4-(tert-butyl)benzamido)-4-(4-(((trifluoromethyl)sulfonyl )oxy)- phenyl)butanoate

[00382] Prepared using General Procedure 9. A stirred solution of (5)-methyl 3- (4-(tert-butyl)benzamido)-4-(4-hydroxyphenyl) butanoate (2.30 g, 6.3 mmol) in DCM (25 mL) was treated with DIEA (1.4 ml, 7.6 mmol) then 1,1,1-trifluoro-N-phenyl-N- ((trifluoromethyl)sulfonyl)methanesulfonamide (2.5 g, 6.9 mmol). After 18 h, the reaction mixture was diluted with DCM (100 mL), H 2 0 (50 mL) and NaHC0 3 (75 mL) and stirred for 1 h. The organic layer was isolated, washed with NaHC0 3 (100 mL), dried over MgS0 4 , concentrated, and purified by chromatography (EA/ hexane) to provide 2.5 g (75%) of (S)-methyl 3-(4-(tert-butyl)benzamido)-4-(4- (((trifluoromethyl)sulfonyl)oxy)phenyl)butanoate as a thick oil. LCMS-ESI (m/z) calculated for C 23 H 26 F 3 N0 6 S: 501.5, found 502 [M+H] + , t R = 3.20 min (Method 6).

(S)-methyl 3-(4-(tert-butyl)benzamido)-4-(4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl)butanoate

[00383] To a vial under a N 2 atmosphere were added 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(l,3,2-dioxaborolane) (530 mg, 2.1 mmol), (5)-methyl 3-(4-(tert- butyl)benzamido)-4-(4-(((trifluoromethyl)sulfonyl)oxy)phenyl )butanoate (810 mg, 1.6 mmol), KOAc (280 mg, 4.8 mmol) and DMSO (14 mL). The solution was degassed. Pd(dppf)Cl 2 (59 mg, 0.08 mmol) was added and the solution was heated to 80°C for 6 h. The reaction mixture was cooled to RT, diluted with EA (100 mL) and washed with sat aq. NaHC0 3 (50 ml) and brine (50 mL). The organic layer was dried over MgSC^, concentrated and purified by chromatography (EA / hexane) to afford 446 mg (57%) of (iS)-methyl 3-(4-(tert-butyl)benzamido)-4-(4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)phenyl) as a colorless crystalline solid. LCMS-ESI (m/z) calculated for

C 28 H 38 BN0 5 : 479.4, found 480.3 [M+H] + , t R = 2.86 min (Method 6).

(S)-methyl 4-(4-(5-bromopyrimidin-2-yl)phenyl)-3-(4-(tert-butyl)benzami do)- butanoate

[00384] Prepared using General Procedure 10. Into a vial were added (S)- methyl 3-(4-(tert-butyl)benzamido)-4-(4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)phenyl) (390 mg, 0.81 mmol), 5-bromo-2-iodopyrimidine (240 mg, 0.85 mmol), Na 2 C0 3 (170 mg, 1.6 mmol), THF (1.5 mL), ACN (1.5 mL) and H 2 0 (0.75 mL). The solution was degassed and PdCl 2 (dppf) (60 mg, 0.08 mmol) was added. The reaction mixture was heated in a microwave reactor at 1 10°C for 60 min. The sample was cooled, diluted with EA (50 mL), and washed with sat aq.NaHC0 3 (30 mL). The organic layers were dried over MgSC^, filtered, concentrated, and purified by chromatography (EA / hexane) to afford 205 mg (49%) of (5)-methyl 4-(4-(5- bromopyrimidin-2-yl)phenyl)-3-(4-(fert-butyl)benzamido)butan oate as a colourless solid. LCMS-ESI (m/z) calculated for C 26 H 28 BrN 3 0 3 : 510.4, found 512.2 [M+H] + , t R = 2.77 min (Method 6). (S)-3-(4-(tert-butyl)benzamido)-4-(4-(5-(4-(heptyloxy)phenyl )pyrimidin-2- yl)phenyl)butanoic acid (Compound 291)

[00385] Prepared using General Procedures 10 and 4. Into a vial were added (iS)-methyl 4-(4-(5 -bromopyrimidin-2-yl)phenyl)-3 -(4-(tert-butyl)benzamido)butanoate (180 mg, 0.35 mmol), (4-(heptyloxy)phenyl)boronic acid (98 mg, 0.41 mmol), Na 2 C0 3 (73 mg, 0.69 mmol), ACN (1.2 mL), THF (1.2 mL) and H 2 0 (0.7 mL). The solution was degassed, Pd(dppf)Cl 2 (25 mg, 0.03 mmol) was added, and the reaction mixture was heated in a microwave reactor at 110°C for 80 min. The reaction mixture was diluted with EA (50 mL) and washed with sat aq. NaHC0 3 (30 mL). The organics layer was dried over MgS0 4 , concentrated, and purified by chromatography (EA / hexane) to afford 44 mg of methyl ester intermediate. The solid was dissolved in THF (1 mL) and 1M LiOH (1 mL). The solution was stirred at ambient temperature for 1 h,

concentrated, and 1M HC1 (1.5 mL) was added. The solid was collected by filtration, washing with water (2 x 5 mL) and hexane (2 x 5 mL) to provide 19 mg (9%) of (5)-3- (4-(tert-butyl)benzamido)-4-(4-(5-(4-(heptyloxy)phenyl)pyrim idin-2- yl)phenyl)butanoic acid Compound 291 as a colorless solid. LCMS-ESI (m/z) calculated for C 3 8H 45 N 3 0 4 : 607.8, found 608.4 [M+H] + , t R = 10.99 min (Method 10).

5-bromo-2-chloro-4-methoxypyrimidine

[00386] To a stirred solution of 5-bromo-2,4-dichloropyrimidine (500 mg, 2.19 mmol) in MeOH (5 mL) was added a 30% solution of sodium methoxide (0.40 mL, 2.26 mmol). The reaction mixture was stirred at RT for 2 h then concentrated. The residue was dissolved in water (5 mL) and extracted with EA (3 x 5 mL). The combined organic layer was washed with brine, dried over MgS0 4 and concentrated to afford 432 mg (88%) of 5-bromo-2-chloro-4-methoxypyrimidine as white solid. LCMS- ESI (m/z) calculated for C 5 H 4 BrClN 2 0: 223.4; found 224.2 [M+H] + , t R = 7.66 min. (Method 2).

5-bromo-2-iodo-4-methoxypyrimidine

[00387] Prepared using General Procedure 16: To a stirred solution of 5-bromo- 2-chloro-4-methoxypyrimidine (100 mg, 0.447 mmol) in 57 % aq. HI (1.0 mL) was added sodium iodide (125 mg, 0.838 mmol). The reaction mixture was stirred at 40°C for 16 h, cooled, then quenched with NaHC0 3 (5 mL) and extracted with EA (3 x 5 mL). The combined organics were washed with brine, dried over MgS0 4 and concentrated to afford 22.0 mg (16%) of 5-bromo-2-iodo-4-methoxypyrimidine as an off-white solid. LCMS-ESI (m/z) calculated for C 5 H 4 BrIN 2 0: 314.9; found 315.9

[M+H] + , t R = 8.22 min. (Method 2). 1H NMR (400 MHz, DMSO) δ 8.25 (s, 1H), 4.07 (s, 3H).

Tert-butyl (S)-3-(4-( 5-bromo-4-methoxypyrimidin-2-yl)phenyl)-2- (4-( tert- butyl)benzamido)propanoate

[00388] Prepared using General Procedure 10: A mixture of tert-butyl (5)-2-(4- (tert-butyl)benzamido)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxa borolan-2- yl)phenyl)propanoate INT-13 (30.0 mg, 0.06 mmol), 5-bromo-2-iodo-4- methoxypyrimidine (22.3 mg, 0.07 mmol), and sodium carbonate (12.5 mg, 0.12 mmol) in acetonitrile (0.80 mL), THF (0.80 mL) and H 2 0 (0.40 mL) was degassed for 10 min. Pd(dppf)Cl 2 :CH 2 Cl 2 (5 mg, 0.005 mmol) was added and the reaction mixture heated at 110°C in a microwave for 30 min. Once cooled, the reaction was diluted with NaHC0 3 (5 mL), extracted with EA (3 x 5 mL) and the combined organics dried over MgS0 4 and concentrated. The residue was purified by column chromatography (EA/hexanes) to afford 20.0 mg (60%) of tert-butyl (5)-3-(4-(5-bromo-4-methoxypyrimidin-2- yl)phenyl)-2-(4-(tert-butyl)benzamido)propanoate as a white solid. LCMS-ESI (m/z) calculated for C 29 H 34 BrN 3 0 4 : 568.5; found 514.2 [M-tBu+H] + , i R = 11.0 min. (Method 2).

Tert-butyl (S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl )-4- methoxypyrimidin-2-yl)phenyl)propanoate

[00389] Prepared using General Procedure 10: A mixture of tert-butyl (5)-3-(4- (5-bromo-4-methoxypyrimidin-2-yl)phenyl)-2-(4-(tert-butyl)be nzamido)propanoate (18.0 mg, 0.031 mmol), (4-(heptyloxy)phenyl)boronic acid (10.0 mg, 0.042 mmol) and sodium carbonate (8.97 mg, 0.084 mmol) in acetonitrile (0.80 mL), THF (0.80 mL) and H 2 0 (0.40 mL) was degassed for 10 min. Pd(dppf)Cl 2 :CH 2 Cl 2 (3.09 mg, 0.003 mmol) was added and the reaction mixture heated at 110°C in a microwave for 30 min. Once cooled, the reaction was diluted with NaHC0 3 (5 mL) and extracted with EA (3 x 5 mL). The combined organics were dried over MgS0 4 and concentrated. The residue was purified by column chromatography (EA:hexanes) to afford 20.0 mg (60%) of tert- butyl (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl )-4- methoxypyrimidin-2-yl)phenyl)propanoate as pale yellow solid. LCMS-ESI (m/z) calculated for C42H53N3O5 : 679.8; no ion observed, fa = 13.83 min. {Method 2).

(S)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phe nyl)-4- methoxypyrimidin-2-yl)phenyl)propanoic acid (Compound 292)

[00390] Prepared using General Procedure 8: A solution of tert-butyl (S)-2-(4- (tert-butyl)benzamido)-3 -(4-(5 -(4-(heptyloxy)pheny 1 -4-methoxypyrimidin-2- yl)phenyl) propanoate (20.0 mg, 0.029 mmol) in DCM (1 mL) was treated with TFA (0.350 mL). The reaction mixture was stirred at RT for 12 h. The solvent was concentrated and the product was purified preparative HPLC to yield 15.0 mg (82%) of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl )-4-methoxypyrimidin-2- yl)phenyl)propanoic acid, Compound 292 as pale yellow solid. LCMS-ESI (m/z) calculated for C38H45N3O5 : 623.8; no ion observed, fa = 12.17 min. (Method 2).

[00391] Compound 293 was prepared using tert-butyl (S)-2-(4-(tert- butyl)benzamido)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborola n-2-yl)phenyl)propanoate INT-13 and 5-bromo-2-chloro-N,N-dimethylpyrimidin-4-amine using General Procedures 10, 10 and 8 sequentially.

[00392] Compound 294 was prepared using tert-butyl (S)-2-(4-(tert- butyl)benzamido)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborola n-2-yl)phenyl)propanoate INT-13 and 5-bromo-2-iodo-4-methylpyridine using General Procedures 10, 10 and 8 sequentially 5-bromo-2-iodo-4-(trifluoromethyl)pyridine

[00393] Prepared using General Procedure 17: To a stirred a solution of 5- bromo-2-chloro-4-(trifluoromethyl)pyridine (150 mg, 0.576 mmol) in acetonitrile (2 mL) was added sodium iodide (518 mg, 3.45 mmol). The reaction mixture was heated to 40°C and acetyl chloride (26.0 mg, 0.345 mmol) was added. The reaction mixture was stirred at 40°C for 90 min. Once cooled, the reaction was quenched with NaHC0 3 (5 mL) and extracted with EA (3 x 5 mL). The combined organics were washed with brine (10 mL), dried over MgS0 4 and concentrated to give 80.0 mg (40%) of 5-bromo- 2-iodo-4-(trifluoromethyl)pyridine as a white crystalline solid which was used in the subsequent step without purification. LCMS-ESI (m/z) calculated for C 6 H 2 BrF 3 IN: 351.9; found 352.5 [M+H] + , t R = 3.91 min. (Method 1).

[00394] Compound 295 was prepared by employing tert-bvXy\ (S)-2-(4-(tert- butyl)benzamido)-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborola n-2-yl)phenyl)propanoate INT-13 and 5-bromo-2-iodo-4-(trifluoromethyl)pyridine using General Procedures 10, 10 and 8 sequentially.

(S)-(2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)ph enyl)pyrimidin-2- yl)phenyl)propanoyl)glycine (Compound 297)

[00395] Prepared using General Procedures 7 and 8: To a solution of (5)-2-(4- (tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidi n-2-yl)phenyl)propanoic acid Compound 85 (185 mg, 0.312 mmol), tert-butyl 2-aminoacetate hydrochloride (52.2 mg, 0.312 mmol), and DIEA (163 μΐ, 0.935 mmol) in DMF (3 mL) was added HATU (124 mg, 0.327 mmol). The mixture was stirred for 1 h at RT. The crude material was diluted in EA (50 mL), washed with saturated aqueous sodium bicarbonate (20 mL) and brine (20 mL). The organic layer was dried over MgS0 4 , filtered, and the solvent removed under reduced pressure. The crude product was purified by

chromatography (EA / hexanes) to afford the intermediate tert-butyl ester (110 mg).

[00396] The tert-butyl ester was dissolved in DCM (1 mL) and TFA (2 mL) was added. The solution was stirred at RT for 3 h and the solvent was removed under reduced pressure. The crude mixture was dissolved in DMSO (0.8 mL) and precipitated by the addition of water (3 mL). The precipitate was filtered, washed with water (3 mL) and hexane (2 x 2 mL) to yield 58 mg (28%) of (S)-(2-(4-(tert-butyl)benzamido)- 3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2-yl) phenyl) propanoyl)glycine, Compound 297 as a colorless solid. LCMS-ESI (m/z) calculated for C 39 H 46 N 4 0 5 : 650.4; found 651.4 [M+H] + , t R = 10.43 min (Method 10). The chiral purity was calculated at 92% e.e. (Chiral Method). 1H NMR (400 MHz, DMSO-d6) δ 12.62 (s, 1H), 9.15 (s, 2H), 8.60 (d, J= 8.6 Hz, 1H), 8.49 - 8.40 (m, 1H), 8.35 - 8.25 (m, 2H), 7.84 - 7.70 (m, 4H), 7.58 - 7.49 (m, 2H), 7.48 - 7.41 (m, 2H), 7.16 - 7.02 (m, 2H), 4.90 - 4.75 (m, 1H), 4.03 (t, J = 6.5 Hz, 2H), 3.93 - 3.75 (m, 2H), 3.25 (dd, J= 13.8, 3.8 Hz, 1H), 3.09 (dd, J= 13.7, 11.2 Hz, 1H), 1.79 - 1.68 (m, 2H), 1.51 - 1.21 (m, 17H), 0.94 - 0.80 (m, 3H).

(S)-3-(2-(4-(tert-butyl)benzamido)-3-(4-(5-(4-(heptyloxy) phenyl)pyrimidin-2- yl)phenyl)propanamido)propanoic acid (Compound 298)

[00397] Prepared using General Procedures 7 and 8: HATU (116 mg, 0.31 mmol) was added to a stirring solution of (5)-2-(4-(fert-butyl)benzamido)-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoic acid Compound 85 (173 mg, 0.29 mmol), tert-butyl 3-aminopropanoate hydrochloride (53 mg, 0.29 mmol) and DIE A (153 μΐ, 0.87 mmol) in DMF (3 mL). The crude material was diluted in EA (50 mL), washed with saturated aqueous sodium bicarbonate (20 mL) and brine (20 mL). The organic layer was dried over MgS0 4 , filtered, and the solvent removed under reduced pressure. The crude product was purified by chromatography (EA / hexanes) to afford the intermediate tert-butyl ester (122 mg).

[00398] The tert-butyl ester was dissolved in DCM (1 mL) and TFA (2 mL) was added. The reaction mixture was stirred at RT for 3 h and the solvent was removed under reduced pressure. The crude mixture was dissolved in DMSO (0.8 mL) and precipitated by the addition of water (3 mL). The precipitate was filtered, washed with water (3 mL) and hexane (2 x 2 mL) to yield 48 mg (25%) of (5)-3-(2-(4-(tert- butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2- yl)phenyl)propanamido)propanoic acid, Compound 298 as a colorless solid. LCMS- ESI (m/z) calculated for C4oH 48 N 4 0 5 : 664.4; found 665.4 [M+H] + , t R = 10.36 min (Method 10). 1H NMR (400 MHz, DMSO-d6) δ 12.26 (s, 1H), 9.15 (s, 2H), 8.51 (d, J = 8.5 Hz, 1H), 8.40 - 8.25 (m, 2H), 8.25 - 8.14 (m, 1H), 7.96 - 7.65 (m, 4H), 7.65 - 7.36 (m, 4H), 7.28 - 6.99 (m, 2H), 4.84 - 4.64 (m, 1H), 4.03 (t, J= 6.5 Hz, 2H), 3.32 - 3.24 (m, 2H), 3.17 (dd, J= 13.7, 4.4 Hz, 1H), 3.06 (dd, J= 13.7, 10.4 Hz, 1H), 2.41 (t, J= 6.9 Hz, 2H), 1.81 - 1.68 (m, 2H), 1.50 - 1.20 (m, 17H), 0.88 (t, J= 6.7 Hz, 3H).

(S)-4-(tert-butyl)-N-(3-(4-(5-(4-(heptyloxy)phenyl)pyrimi din-2-yl)phenyl)-l- (methylsulfonamido)-l-oxopropan-2-yl)benzamide (Compound 299)

[00399] To a solution of (5)-2-(4-(tert-butyl)benzamido)-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoic acid Compound 85 (78.0 mg, 0.13 mmol), methanesulfonamide (20.0 mg, 0.21 mmol), and DMAP (16.1 mg, 0.13 mmol) in DMF (1.5 mL) was added EDC (40.3 mg, 0.21 mmol) and the solution stirred overnight at RT. The reaction mixture was diluted in EA (50 mL), washed with aqueous saturated sodium bicarbonate (2 x 20 mL) and brine (20 mL). The organic layer was dried over MgSC^, filtered, and the solvent removed under reduced pressure. The crude product was purified by chromatography (hexane / EA) to afford 36 mg (40%) of (5)-4-(tert-butyl)-N-(3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin -2-yl)phenyl)-l- (methylsulfonamido)-l-oxopropan-2-yl)benzamide, Compound 299 as a colorless solid. LCMS-ESI (m/z) calculated for 670.3; found 671.3 [M+H] + , t R = 11.01 min (Method 10).

[00400] Compounds 300 - 304 were prepared from (S)-2-(4-(tert- butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2-yl )phenyl)propanoic acid Compound 85 using General Procedures 3 ox 7 followed by 4 or 8.

[00401] Compounds 305 - 317 were prepared from (5)-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)-2-(4-isopropylbenza mido)propanoic acid Compound 94 using General Procedures 3 ox 7 followed by 4 or 8.

[00402] Compound 318 was prepared from (5)-2-(4-(tert-butyl)benzamido)-3-(4- (5-(4-(hexyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoic acid Compound 225 using General Procedures 7 followed by 8.

(S)-(2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4 - (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoyl)glycine ( Compound 319)

[00403] Prepared using General Procedures 7 and 4: TEA (93 μΐ, 0.67 mmol) was added to a solution of (5)-2-(5-(fert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoic acid Compound 192 (100 mg, 0.167 mmol), methyl 2-aminoacetate hydrochloride (23.03 mg, 0.18 mmol) and HATU (76 mg, 0.20 mmol) in DMF (2 mL). The solution was stirred at RT for 18 h. The reaction mixture was diluted with EA (25 mL) and washed with saturated aqueous NaHC0 3 (2 x 25 mL) and 1 M HC1 (2 x 25 mL). The organic phase was dried over MgS0 4 , filtered, and concentrated. The solid was purified by chromatography (EA / hexanes) to afford the methyl ester intermediate as a colorless solid.

[00404] The solid was dissolved in THF (3 mL) and 1 M LiOH (333 μΐ, 0.33 mmol) was added. The resultant yellow solution was stirred at RT for 1 h. The reaction mixture was acidified to pH 1 using 1M HC1 and the THF removed in vacuo. The residue was suspended in water and the mixture filtered under vacuum. The solid was azeotroped with MeOH and dried in a vacuum oven to afford 48 mg (44 %) of ( )-(2- (5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4-(heptylox y)phenyl)pyrimidin-2- yl)phenyl)propanoyl)glycine, Compound 319 as a yellow solid. LCMS-ESI (m/z) calculated for C 3 7H 44 N 4 0 5 S: 656.3; found 657.0 [M+H] + , t R = 10.34 min (Method 10). The chiral purity was calculated at 95% e.e. (Chiral Method). 1H NMR (400 MHz, DMSO-d6) δ 12.61 (s, 1H), 9.16 (s, 2H), 8.62 (d, J= 8.7 Hz, 1H), 8.51 - 8.41 (m, 1H), 8.36 - 8.26 (m, 2H), 7.84 - 7.75 (m, 2H), 7.68 (d, J= 3.8 Hz, 1H), 7.55 - 7.43 (m, 2H), 7.14 - 7.05 (m, 2H), 6.92 (d, J= 3.8 Hz, 1H), 4.84 - 4.72 (m, 1H), 4.03 (t, J= 6.5 Hz, 2H), 3.89 - 3.73 (m, 2H), 3.22 (dd, J= 13.9, 3.7 Hz, 1H), 3.10 - 2.96 (m, 1H), 1.78 - 1.66 (m, 2H), 1.31 (s, 17H), 0.94 - 0.81 (m, 3H).

((S)-2-(5-(tert-butyl)thiophene-2-carboxamido)-3-(4-(5-(4 - (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)propanoyl)-L-glutami ne (Compound 320)

[00405] Prepared using General Procedures 7 and 8: To a stirred solution of (S)- 2-(5-(tert-butyl)thiophene-2-carboxamido) -3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2- yl)phenyl) propanoic acid Compound 192 (250 mg, 0.42 mmol), {S)-tert-bvXy\ 2,5- diamino-5-oxopentanoate hydrochloride (109 mg, 0.46 mmol) and TEA (145 μΐ, 1.04 mmol) in DMF (4 mL) was added HATU (190 mg, 0.50 mmol) and the reaction mixture was stirred at RT for 2 h. The reaction mixture was diluted with EA (50 mL), washed with 1M HC1 (50 mL) and brine (100 mL), dried over magnesium sulfate, and concentrated.

[00406] The crude product was dissolved in DCM (5 mL) and TFA (3 mL) was added. After 3 h, toluene (10 mL) was added and the solvent removed. The compound was purified by preparative HPLC to afford 78 mg (25%) of ((5)-2-(5-(tert- butyl)thiophene-2-carboxamido)-3-(4-(5-(4-(heptyloxy)phenyl) pyrimidin-2- yl)phenyl)propanoyl)-L-glutamine, Compound 320 as a white powder. LCMS-ESI (m/z) calculated for C40H49N5O6S: 727.3; found 728.0 [M+H] + , t R = 10.71 min (Method 10). The chiral purity was 90% d.e. (Chiral Method). 1H NMR (400 MHz, DMSO-d6) δ 9.15 (s, 2H), 8.56 (d, J= 8.6 Hz, 1H), 8.42 - 8.34 (m, 1H), 8.34 - 8.27 (m, 2H), 7.84 - 7.75 (m, 2H), 7.66 (d, J= 3.9 Hz, 1H), 7.54 - 7.48 (m, 2H), 7.32 (s, 1H), 7.12 - 7.04 (m, 2H), 6.90 (d, J= 3.8 Hz, 1H), 6.77 (s, 1H), 4.81 - 4.65 (m, 1H), 4.19 - 4.11 (m, 1H), 4.03 (t, J= 6.5 Hz, 2H), 3.20 (dd, J= 14.1, 3.5 Hz, 1H), 3.07 - 2.96 (m, 1H), 2.24 - 2.09 (m, 2H), 2.06 - 1.93 (m, 1H), 1.90 - 1.79 (m, 1H), 1.78 - 1.68 (m, 2H), 1.47 - 1.20 (m, 17H), 0.93 - 0.82 (m, 3H).

[00407] Compounds 321— 350 were prepared from Compound 192 using General Procedures 3 ox 7 followed by 4 or 8.

[00408] Compounds 351— 368 were prepared from Compound 165 using General Procedures 7 followed by 4 or 8.

[00409] Compound 369 was prepared from Compound 139 using General Procedures 7 followed by 8.

[00410] Compound 370 was prepared from Compound 167 using General Procedures 7 followed by 8.

[00411] Compound 371 was prepared from Compound 142 using General Procedures 7 followed by 8.

[00412] Compound 372 was prepared from Compound 143 using General Procedures 7 followed by 8.

[00413] Compound 373 was prepared from Compound 182 using General Procedures 7 followed by 8.

[00414] Compounds 374— 379 were prepared from Compound 193 using General Procedures 3 or 7 followed by 4 or 8.

[00415] Compound 380 was prepared from Compound 191 using General Procedures 7 followed by 8.

(S -4-(tert-butvn-N-(3-(4-(5-(4-(heptyloxy phenvnpyrimidin-2-vnphenvn-l- ((2-(methylsulfonamido)-2-oxoethyl)amino)-l-oxopropan-2-yl)b enzamide (Compound 381)

[00416] TEA (32.1 μΐ, 0.23 mmol) was added to a suspension οΐ (S)-(2-(4-(tert- butyl)benzamido)-3-(4-(5-(4-(heptyloxy)phenyl)pyrimidin-2-yl )phenyl)propanoyl) glycine Compound 297 (75.0 mg, 0.11 mmol), methanesulfonamide (12.1 mg, 0.13 mmol), HATU (52.6 mg, 0.14 mmol) and DMAP (1.41 mg, 0.01 mmol) in DCM (2 mL). The resultant yellow suspension was stirred at RT for 3 h. The reaction mixture was washed with saturated aqueous NaHC0 3 (2 mL) and the mixture passed through a phase separation cartridge. The organic phase was concentrated in vacuo to afford a yellow solid. The crude product was purified by chromatography (EA / 1% AcOH in hexanes) to afford 9 mg (11%) (5)-4-(tert-butyl)-N-(3-(4-(5-(4- (heptyloxy)phenyl)pyrimidin-2-yl)phenyl)- 1 -((2- (methylsulfonamido)-2- oxoethyl)amino)-l-oxopropan-2-yl)benzamide, Compound 381 as a yellow solid. LCMS-ESI (m/z) calculated for C 4 oH 4 9N 5 0 6 S: 727.3; found 728.0 [M+H] + , t R = 10.51 min (Method 10).

[00417] Selected compounds and their corresponding analytical data are shown in Table 1, where the LCMS data was collected using the method indicated.

Table 1

ı75

ı99

200

201

202

211

215

BIOLOGICAL ASSAYS

Assay Procedures

GLP-1 P AM shift cAMP assay: dose response of peptide ligand in presence of fixed concentration of compound.

[00418] A GLP-1 R expressing CRE-bla CHO-Kl cell line was purchased from Invitrogen. Cells were seeded into 384-well white flat bottom plates at 5000 cells/well/20 growth media (DMEM-High glucose, 10% dialyzed FBS, 0.1 mM NEAA, 25 mM Hepes, lOOU/mL penicillin/ 10(^g/mL streptomycin, 5 μ§/ητΙ,

Blasticidin, 600 μ§/ι Ι, Hygromycin) and incubated for 18 h at 37°C in 5% C0 2 .

Growth medium was replaced with 12 μΐ, assay buffer (Hanks Balanced Salt solution, 10 mM Hepes, 0.1% BSA, pH 7.4). A 5x peptide dose response curve (12-point) was generated in assay buffer containing 1.5 mM IBMX, 12.5% DMSO, and 50 μΜ compound. Peptide ligand was GLP- 1(9-36). The 5x peptide dose response plus compound mix was added (3 μί) and cells were incubated for 30 min at 37°C. Direct detection of cAMP was carried out using DiscoveRx HitHunter cAMP kit according to manufacturer's instructions and luminescence was read using a SpectraMax M5 plate reader. Luminescence was analyzed by non-linear regression to determine the EC 50 and Emax. A GLP- 1(7-36) dose response was included to determine maximum efficacy.

EC 20 GLP- 1(9-36) PAM cAMP assay: dose response of compound in the presence of fixed concentration of GLP-1 (9-36).

[00419] GLP-1R CRE-bla CHO-K1 cells cultured in growth medium (DMEM- High glucose, 10% dialyzed FBS, 0.1 mM NEAA, 25 mM Hepes, lOOU/mL

penicillin/ 100μg/mL streptomycin, 5 μg/mL Blasticidin, 600 μg/mL Hygromycin) were tryspsinized and plated in suspension into 384 well white flat bottom plates at 5000 cells/well in 12 μΐ ^ assay buffer (Hanks Balanced Salt solution, 10 mM Hepes, 0.1%> BSA, pH 7.4). A 5x compound dose response curve (12-point) was generated in assay buffer containing 1.5 mM IBMX, 12.5% DMSO. GLP- 1(9-36) was diluted to 4.2 μΜ in assay buffer containing 1.5 mM IBMX and 12.5% DMSO. The 5x compound dose response was added (3 μί), followed by 0.5 μί of GLP- 1(9-36) and cells were incubated for 30 min at 37°C. Direct detection of cAMP was carried out using

DiscoveRx HitHunter cAMP kit according to manufacturer's instructions and luminescence was read using a SpectraMax M5 plate reader. Luminescence was converted to total cAMP using a cAMP standard curve and data was analyzed by nonlinear regression to determine the EC 50 and Emax. Peptide sequences

[00420] GLP-l(7-36) (SEQ ID NO:2):

HAEGTFTSDVSSYLEGQAAKEFIAWLVKGR-NH2. GLP-l(9-36) (SEQ ID NO:3): EGTFTSDVSSYLEGQAAKEFIAWLVKGR-NH 2 . GLP-1 (7-36) was purchased from GenScript. GLP-1 (9-36) was purchased from Biopeptide Co., Inc.

Reported GLP-1 Activity

[00421] Activity data for selected GLP-1 modulators are displayed in Table 2. The EC 20 GLP-1 (9-36) PAM Activity range is denoted as follows: + denotes activity < 0.8 μΜ, ++ denotes activity between 0.8 and 2.5 μΜ, +++ denotes activity between 2.5 and 5 μΜ, and ++++ denotes activity 5 to 10 μΜ.

Table 2

COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

13 ++ 203 ++++

14 +++ 204 +++

15 +++ 205 ++

16 ++++ 206 +++

17 +++ 207 +++

18 ++ 208 +++

19 ++ 209 ++

20 ++ 210 +++

21 +++ 211 ++++

22 +++ 212 ++++

23 +++ 213 +++

24 ++ 214 +++

25 ++ 215 +++

26 +++ 216 +++

27 ++ 217 ++++

28 +++ 218 +++

29 ++ 219 ++

30 ++ 220 +++

31 ++ 221 +++

32 ++++ 222 ++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

33 +++ 223 ++

34 +++ 224 ++

35 +++ 225 +

36 +++ 226 ++

37 +++ 227 +++

38 ++ 228 ++

39 ++ 229 ++

40 +++ 230 ++

41 +++ 231 ++++

42 +++ 232 ++

43 +++ 233 +++

44 +++ 234 ++++

45 ++ 235 +++

46 ++ 236 ++++

47 +++ 237 ++

48 ++++ 238 ++

49 ++++ 239 ++

50 +++ 240 +++

51 +++ 241 ++

52 ++ 242 ++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

53 +++ 243 ++

54 +++ 244 +++

55 +++ 245 ++

56 +++ 246 ++

57 +++ 247 ++

58 +++ 248 +++

59 +++ 249 ++++

60 +++ 250 ++

61 +++ 251 ++++

62 +++ 252 ++++

63 ++ 253 +++

64 ++ 254 ++

65 +++ 255 ++++

66 +++ 256 ++++

67 ++ 257 ++

68 +++ 258 ++++

69 ++++ 259 ++

70 ++++ 260 +

71 +++ 261 +++

72 ++++ 262 +++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

73 ++ 263 ++

74 +++ 264 ++

75 +++ 265 ++++

76 ++ 266 ++++

77 ++ 267 ++

78 ++ 268 ++

79 ++ 269 ++

80 ++ 270 +++

81 ++++ 271 ++

82 ++ 272 ++

83 ++ 273 ++

84 ++ 274 ++

85 ++ 275 ++

86 ++ 276 ++

87 ++ 277 +

88 ++++ 278 +++

89 ++ 279 ++++

90 +++ 280 +++

91 ++ 281 +

92 ++ 282 ++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

93 ++++ 283 ++++

94 ++ 284 ++

95 ++ 285 ++

96 +++ 286 +++

97 ++ 287 ++++

98 ++ 288 +++

99 ++ 289 +++

100 ++ 291 ++

101 ++ 292 ++

102 ++ 293 +++

104 ++ 294 ++

105 ++++ 295 ++

106 ++ 296 ++

107 ++ 297 +

108 ++ 298 +

109 ++ 299 +

110 ++ 300 +

111 ++ 301 ++

112 ++ 302 +++

113 ++ 303 ++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

114 ++ 304 ++

115 ++ 305 ++

116 ++ 306 +

117 + 307 ++

118 + 308 ++

119 + 309 +

120 ++ 310 +

121 ++ 311 ++

122 ++ 312 ++

123 ++ 313 ++

124 ++ 314 ++

125 ++ 315 ++

126 ++ 316 +

127 ++ 317 +

128 ++ 318 +

129 ++ 319 +

130 + 320 +

131 ++ 321 +

132 ++ 322 ++

133 ++ 323 + COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

134 ++ 324 ++

135 ++ 325 ++++

136 ++++ 326 +

137 ++ 327 +

138 ++ 328 ++

139 ++ 329 ++

140 +++ 330 ++

141 ++ 331 ++

142 + 332 ++

143 + 333 +

144 ++ 334 ++

145 ++ 335 ++

146 +++ 336 +

147 +++ 337 ++

148 ++ 338 ++

149 ++ 339 ++

150 ++ 340 ++

151 ++ 341 +

152 ++ 342 ++

153 ++ 343 ++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

154 + 344 +

155 ++ 345 +

156 ++ 346 ++

157 ++ 347 +

158 + 348 ++

159 ++ 349 +

160 ++ 350 +

161 ++ 351 +

162 ++ 352 +

163 ++ 353 ++

164 ++ 354 +

165 ++ 355 ++

166 ++ 356 ++

167 ++ 357 ++

168 ++ 358 ++

169 ++++ 359 ++

170 ++ 360 ++

171 ++ 361 ++

172 ++ 362 +++

173 ++ 363 ++ COMPOUND EC 20 GLP- 1(9-36) COMPOUND EC 20 GLP- 1(9-36) NUMBER PAM ECso NUMBER PAM ECso

174 + 364 ++

175 + 365 +++

176 ++++ 366 +++

177 ++ 367 ++

178 ++ 368 +

179 ++++ 369 ++

180 + 370 ++

181 ++ 371 +

182 ++ 372 +

183 ++ 373 ++

184 ++ 374 ++

185 ++ 375 +

186 ++ 376 ++

187 +++ 377 +

188 +++ 378 +

189 ++ 379 +

190 +++ 380 ++

191 ++ 381 ++ IN VIVO ASSAYS

In Vivo Procedures

The oral glucose tolerance test in C57BI/6 mice.

[00422] The use of these compounds to lower glucose can be evaluated in mice using an oral glucose tolerance test (oGTT). The protocol is described by Duez et. al (Endocrinology, January 2009, 150(l):56-62). C57BL/6 male mice are chronically cannulated. After a 5 day recovery, mice are fasted for 4 h. A baseline blood sample is collected before the iv administration of a bolus of either saline or exendin-4, or administration of the compound. Immediately after, mice receive a bolus of glucose (1.5 g/kg) by oral gavage (time 0). Blood samples are collected at frequent time intervals from the tail tip for glucose measurement (BD glucometer; Becton-Dickinson, Lincoln Park, NJ). For plasma insulin determinations, a blood sample is removed from the tail vein at 5 min after glucose administration.

The oral glucose tolerance test in fa/fa rats.

[00423] The use of these compounds to lower glucose can be evaluated in rats using an oral glucose tolerance test (oGTT). The protocol is described by Pederson et. al. (Diabetes, Vol. 47, August 1998, 1253-1258). After an overnight fast, lean or obese animals are administered oral glucose by syringe and feeding tube (1 g/kg) as a 40% solution (wt/vol). Compound is dissolved and administered along with the glucose. In control experiments, vehicle is administered along with oral glucose. Blood samples are collected from the tail veins of conscious unrestrained rats into heparinized capillary tubes at 0 and 5, 10, 20, 30, and 60 min after glucose administration. Blood samples are centrifuged at 4°C, and plasma is stored at -20°C until analysis for glucose and insulin measurement. Glucose levels are measured using the glucose oxidase procedure (Beckman glucose analyzer; Fullerton, CA).

[00424] The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments. These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.




 
Previous Patent: MULTI-RADIO COEXISTENCE

Next Patent: CONCRETE BUILDING PANEL