Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NON-SYSTEMIC TGR5 AGONISTS
Document Type and Number:
WIPO Patent Application WO/2013/096771
Kind Code:
A1
Abstract:
Compounds of structure (I), or a stereoisomer, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R1, R2, R3, R4, R8, R9, R10, R11, R12, A1, A2, X, Y and Z are as defined herein. Uses of such compounds as TGR5 antagonists and for treatment of various indications, including Type II diabetes meletus are also provided.

Inventors:
LEWIS JASON G (US)
REICH NICHOLAS (US)
CHEN TAO (US)
JACOBS JEFFREY W (US)
CHARMOT DOMINIQUE (US)
NAVRE MARC (US)
FINN PATRICIA (US)
CARRERAS CHRISTOPHER (US)
SPENCER ANDREW (US)
Application Number:
PCT/US2012/071251
Publication Date:
June 27, 2013
Filing Date:
December 21, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ARDELYX INC (US)
International Classes:
C07D241/42; C07C235/68; C07C237/24; C07D207/16; C07D213/75; C07D215/08; C07D231/20; C07D233/90; C07D263/34; C07D265/30; C07D277/06; C07D279/10; C07D279/16
Domestic Patent References:
WO2011089099A12011-07-28
WO2003105849A12003-12-24
WO2007025709A22007-03-08
Foreign References:
US20110178089A12011-07-21
US20100105906A12010-04-29
Other References:
WAQAR NASIR ET AL: "Synthesis of novel quinoxalinone derivatives by conventional and microwave methods and assessing their biological activity", ARCHIVES OF PHARMACAL RESEARCH, PHARMACEUTICAL SOCIETY OF KOREA, HEIDELBERG, vol. 34, no. 10, 12 November 2011 (2011-11-12), pages 1605 - 1614, XP019976162, ISSN: 1976-3786, DOI: 10.1007/S12272-011-1004-4
HONGLIANG DUAN ET AL: "Design, Synthesis, and Antidiabetic Activity of 4-Phenoxynicotinamide and 4-Phenoxypyrimidine-5-carboxamide Derivatives as Potent and Orally Efficacious TGR5 Agonists", JOURNAL OF MEDICINAL CHEMISTRY, vol. 55, no. 23, 13 December 2012 (2012-12-13), pages 10475 - 10489, XP055057045, ISSN: 0022-2623, DOI: 10.1021/jm301071h
Attorney, Agent or Firm:
WAGNER, Emily, W. et al. (777 - 6th Street NWSuite 110, Washington DC, US)
Download PDF:
Claims:
CLAIMS

1. A compound having the following structure (I):

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

X is CR50R5! wherein:

R30 and R51 are the same or different and independently selected from H and C1-7-alkyl, or

R50 and R51 taken together with the C atom to which they are attached form a cyeloalkyl or heterocyclyl, wherein the cycioalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci_7~alkyi Q.rhaloalkyL C1-7-alkylcarbonyl, Cj.7-alkyloxycarbonyl, Ci_7-alkoxy, C1-7-alkoxyaikyl, (Ra)2(Rb)N- and C|-7-alkyl-S(0)o-2~, wherein, each Ra is independently, at each occurrence, hydrogen or Cj-7-alkyl and R° is an electron pair, hydrogen or Ci_7-alkyl;

Y is CR60R61, O, Nil62 or a direct bond, provided that when Y is O, Z is not O or S(0)o-2, wherein:

R60 and R61 are the same or different and independently selected from H and Cj.-7-alkyl; and

R62 is selected from H, Ci.7-alkyl, Ci-7-alkylcarbonyl, aminocarbonyl, Ci-7-alkylaminoearbonyI, Cj.7-alkyisulfone, cycloal.kylalkyl, cyeloalkyl, aralkyl. and aryi, wherein the Ci-7-aikyl, Ci-7-alkylcarbonyl, aminocarbonyl. Ci-7-a]kylaminocarbonyl, d-7-alkylsulfone, cycloalkylalkyl, cycioalkyl, aralkyl and aryi are optionally substituted with one or more substitutents selected from halogen, hydroxy, oxo, Ci_7-haloalkyl, C]-7- alkylcarboiiyl, Ci-7-alkyloxycarbonyl, C1-7-alkoxy, C1. -alkoxyalkyl, (R%(Rb)N-- and C1.7-alkyl-S(0)o-2~, wherein each Ra is independently, at each occurrence, hydrogen or Ci-7-alkyl and Rb is an electron pair, hydrogen or C1-7-a3kyl;

or X and Y taken together form a cycioalkyl, heterocyclyl, aryl or heteroaryl, wherein the cycioalkyl, heterocyclyl, aryl or heteroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C1-7- alkyl, C j .7-alkyloxycarbonyl, Ci_7-alkoxy, Ci-7-alkoxyalkyl, (R^R^N- and Ci_7-alkyl-S(0)o-2-, wherein Ra is independently,, at each occurrence, hydrogen or Ci .7-alkyl and Rb is an electron pair, hydrogen or Chalky!, and provided that when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyrimidinyl then Z is not O;

Z is CRWR7!, O, S(0)o-2 or a direct bond, wherein:

R7G and R'1 are the same or different and independently selected from H or C1-7- alkyl;

or R'"J and R?1 taken together to form oxo (=0);

or Z and R8 or R12 taken together form a cycioalkyl or heterocyclyl, wherein the cycioalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy ,οχο, f1-7-alkyL Cu7-haloalkyl, C1-7- alkykarbonyl, Cj.7-alkyioxycarbonyl, Ci-7-alkoxy, Cj-7-alkoxyalkyl, (Ra)2(R )N- and Ci.7-alkyl-S(0)o-?-, wherein each Ra is independently, at each occurrence, hydrogen or Cj.7-alkyl and Rb is an electron, pair, hydrogen or Cj.-7-alkyl;

A1 is CR o N;

A is CR!'5 or , wherein:

Rl3 and R14 are the same or different and independently selected from: hydrogen, Ci-7-alkyi, halogen, Ci_7-haloalkyl, cyano, C -alkoxy, amino and -S(0)o-2-Ci.7-alkyl;

R1 and R2 are the same or different and independently selected from: hydrogen, Ci,7-alkyl5 halogen, halogen-C1.7-alk.yl, cyano and Cj.-7-alkoxy: Ί13 is selected from: hydrogen, Cj.y-alkyL halogen, Ci-7-haSoaIkyl, Ci^-alkoxy, cyano, C3.7~cycloa.kyl, -0-C3-7-cycloalky], -0-Ci-7-alkyl-C3.7-cyc!oalkyl, -S(0)o-2-Ci-7-alkylJ N-heterocyclyl, five-membered lieteroaryl, phenyl and - NR15R16, wherein R13 and R1 are the same or different and independently selected from hydrogen, Ct.7-alkyl and. C3-7-c cloalkyl;

R4 is selected from: hydrogen., C^-alky!, halogen-Ci-7-alkyl, and C3.7-cycloalkyI; or RJ and R or R3 and ¼ together are -L1-(CR17Ri8)a- and form part of a ring, wherein:

L5 is selected from: -CR19R20-, O, S(O)0-2, C=0 and NR2';

' and R* are the same or different and independently selected from hydrogen and Ci-7-aikyl;

or R17 and R18 together with the C atom to which they are attached form an oxo moiety;

or R17 or R58 together with an adjacent R17, R18, R19 or R20 and the C atoms to which they are attached form C-C;

R19 and R20 are the same or different and independently selected from: hydrogen, hydroxy!, N(R2i)2> Ci-7-alkyl, Cw-alkoxycarbonyl, unsubstituted heterocyciyl, and heterocyciyl substituted by one or two groups selected from halogen, hydroxy and Ci.7-aikyl,

or R39 and R20 together with the C atom to which they are attached form a cyclopropyl or oxetanyf ring or together form a =C1¾ or =Cp2 group; and

R2 ! is independently, at each occurrence, selected from the group consisting of: hydrogen, d-7-aikyl, halogen-Ci-7-alkyL C3.7-cycioa1kyl and C3-7- cycloalkyl-Ci-7-alkyl, wherein C 7-eycIoalkyl is unsubstituted or substituted by carboxyl- C1-7-alkyl or Ci .y-alkoxycarbonyl, heterocyciyl, heterocyciyl - Ci-7-alkyl, heteroaryl, heteroaryl-Ci-7~alkyl, carboxyl-C'j-7-alkyl, C i.. ?- aikoxycarbony I-C 1.,7-alkyl, d-7-alkylcarbonyloxy-C 1 -7 -alky 1 , Ci .7- a!kylsulfonyl, phenyl, wherein phenyl is unsubstituted or substituted by carboxyl-Ci.7-aiky1 or Ci^-alkoxycarbonyl, phenylcarbony], wherein phenyl is unsubstituted or substituted by carboxyl-Cj-7-alkyl or Ct-7-alkoxycarbonyl, and phenylsulfonyl, wherein phenyl is unsubstituted or substituted by carboxyl.-Ci-7-alkyl or Cj.7-alkoxycarbonyl;

or R* and a ' together are -(C¾)3- and form part of a ring;

or R2i together with a pair of Rl and RI& are -CH= H-CH== and form part of a ring; and

n is 1 , 2 or 3;

R8, R9, R , Ru and are the same or different and independently selected from:

Q, hydrogen, C;..7-alkyl, C2.-7-alky.nyl, halogen, halogen-Cj _7-alkyl, Ci-7-alkoxy, halogen-C1.7-al.koxy, hydroxy, hydroxy-C1-7-alkoxy, hydroxy-Ci-7- alkyl, h droxy-C3.7-alkenyi, hydroxy-C 7-alkynyl, cyano, carboxyl, C1.7- alkoxycarbonyl, amino carbonyl, carboxyI-Ci-7-alkyl, carboxyl- C2-?-aIkenyl, carboxyl- C2-7-alkyny!, Ci^-alkoxycarbonyl-Ci^-alkyl, Ci^-alkoxycarbonyi-C^ 7-alkenyl, C^-alkoxycarbony -C^-alkynyl, carboxyl-C j.7-alkoxy, Cj .7- aikoxycarbonyl~Ci.7-aikoxy, carboxyf-Ci.7-aikyi-a.minocarbony], carboxyl-Ci.7- alkyl -(C ·..7-alkylaffii.no )-carbonyi,C 1.7-alkoxycarbonyi-C 5 ,7-alkyl- aminocarbonyl, Cj^-alkoxycarbonyl-Ci^-alkyl-CCi^-alkylanimo^carbonyl, carboxyi-Ci.7-alkyl-aminocarbonyl-Ci.7-alkyl5 carboxyl-C1.7-alkyHC1.7- alkylamino)-earbonyl-Ci-7-alkyi, Ci-7-alkoxyearbonyl~C1_7-aikyl- amiiiocarbonyl-C] ..7-alkyl, Ci.7-alkoxycarbonyl-C).7-alky]-(Ci.7-alk amino)- carbo!iyl-Ci-y-alkyk hydroxy-Ci.7-alkyl-aminocarbonyl, di-(hydroxy-Ct.7- alkyl)aminocarbonyl, aminocarbonyl-Ci.7-aikyl-amino carbonyl, hydroxysii[fony]-Ci,7-alkyl-aininocarbonyi, hydrox5¾-ulfonyl-Ci-7-alkyl-(Cj.7- alkyl-amino)-carbonyl, dH i^-alkoxycarbonyl-Ci^-alkyl)-- raethylaminocarbonyl, phenyl, wherein phenyl is ^substituted or substituted by one to three groups selected from halogen, Ci.7-alkoxy, carboxyl or Cj.7- alkoxycarbonyl, phenyl-carbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected, from halogen, C^-alkoxy, carboxyl or Ct_7- aikoxycarbonyl, phenyl-aminocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen. Q-ralkoxy, carboxyl or C j ,7-alkoxycarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen Ct.7-aikoxy, carboxyl or Ci-7-aikoxycarbonyl, phenyl-C2.7-alkyn.yl, wherein phenyl is unsubstituted. or substituted by one to three groups selected from halogen, Cj.7-alkoxy, carboxyl or Ci.7-alkoxycarbonyl, heteroaryl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen Ct_7-alkoxy, carboxyl or Ci-7-alkoxycarbonyl, heteroaryl-carbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl or Ci-7-alkoxycarbonyl, heteroaryl-amiriocarbonyi, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C1.7-alk.oxy, carboxyl or Ci_7-alkoxycarbonyl, heteroaryl-Ci-7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkyI, Ci-7-alkoxy, carboxyl or Ci-7-aikoxycarbonyl, heteroaryl- Ci^-alkyl-aniinocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Cw-alkoxy, carboxyl or C1-7- alkoxycarbonyl. hetcroaryl-carbonyl-Ci-7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C1.7- alkoxy, carboxyl or Ci-7-alk.oxycarbonyl , and cycloaikyl, wherein cycloalkyl is unsubstituted or substituted by one to three groups selected from halogen, C>,7- alkoxy, carboxyl or Ci-7-alkoxycarbonyl;

wherein:

L2 and each L3 are either the same or different and independently absent, -0-, -NR.80-, -S-, -NR80C(-O)-, -C(=0)NR80-, -NR80C(-O)NRs0-, -S02NR80-, - NR80SO2-; ~Ci..7alkylene-; -C-.-7alkylene-0-, -0-C1-7alkylene-, -C|.7alkylene~NR¾0-, - R80-Cj.7alkylene-, -Ci_7alkylene-S-, -S-Ci .7aikylene-, -Ci^alkylene-NR^C^O)-, -C(-O)NRS0Ci_7alkyiene-, -C^alkylene-CH))NR80-, -NRs0C(-O)Ci.,7alkylene-, -C1-7aikyiene-NRS0CeK))NR80-, -NR80C(-O)NR80C3-7alkylene-, -Cj^alky!ene-SOaNR80-, -S02NRaoC5 -?alkyiene-, -SO2NR80C(-O)-, -C(O)NR80S02-, - N Rs"S():NRX!1Ci 0)NRx"-. -NRS0C(=O)NRS0SO2NR80, -OC(=0)NR80-,

-NR8°C(O)0-; -Ci-7alkylene-OC(-())NR80-, -NR80C(< O-Ci-7alkylenes -CI-7alkyicne- 80C(-O)O-, -OC(=O)NRS0-Ci.7a!kylene-; -SO2NR80C,. 7alky!ene- or -C i.7alkylene- R80SO2-;

B is optionally substituted Ci.70al.k I or Chalky lene, wherein the

Ci_7oaikyl or Ci-7oaJkylene is optionally substituted with one or more functional groups selected from hydroxy!, oxo, car boxy, guanidino, amidirto, -N(R8 ):>, -N(RS0)3, phosphate, phosphonate, phospinate, sulfate, sulfonate and sulfonate, and wherein the Cj-7oaikyl or Ci.7o lk lene optionally comprises one or more moieties selected from -NR8"-, -S-; -0-, -C3-7cycloa!kyl-, ~C3-7heterocyciyi-, -C5.7aryi- and -SO2-;

I is a compound of structure (I);

R8 is independently, at each occurrence, hydrogen, C1-7alkyi or -B-(LJ- and

m is an integer ranging from 0 to 10.

The compound of claim 1, wherein X is CR50R5! and having the following structure (II):

.1 The compound of claim 2 wherein Y is () and Z is CR^R' 1 and having the following structure (III):

4. The compound of claim 2, wherein Y is NR

and having the following structure (IV):

5. The compound of claim 2, wherein Y is CR¾ R01 and Z is O and having the following structure (V):

6. The compound of claim 2, wherein R" ' and R3 taken together with the C atom to which they are attached form a. eycloaikyl or heterocycivl, wherein the eycloaikyl or heterocyciyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci-7-alkyl, Ci .7-aikylcarbonyl, C1-7-alkylQxycarbonyl, C1.7- alkox , Cj.y-alkoxyalkyl and Ci-7-aIkyl-S(())o-2-, wherein the compound has the iollowing structure (VI) and wherein W represents the cycloalkyj or heteroeycly group:

7. The compound of claim 6, wherein Y is O and Z is CR'°R' ' and having the following structure (VII):

8, The compound of claim 6, wherein Y is NR62 and Z is CR'°R and having the following structure (Vlii):

(VIII)

9. The compound of claim 6, wherein Y is CR60R61 and Z is O and having the following structure (IX):

10. The compound of claim 6, wherein the compound has one of the following structures (Via), (VIb), (Vic), (VId), (Vie), (Vlf), (VIg) or (V

(Vic) (VId)

( Ig) (Vlh) wnerem:

R'~' is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, Ci-7- alkyl, Ci-7-alkylcarbonyl, Ci-7-a!kyloxycarbonyl, C1.7-al.koxy, C;.7-alkoxyalkyl or C1.7- alkyl-S(())o-2-; and

Rd is independently, at each occurrence, an electron pair, hydrogen,

Cj-7-alkylcarbonyl, C j -7-alky ioxycarbonyi, C;..7~alkoxyalkyl or Cj.7-alkyl-S(0)o-2--

11. The compound of claim 10, wherein Y is 0 and Z is CR 70RD.7i

12. The compound of claim 10, wherein Y is NR'' and Z is CR' 'R

13. The compound of claim 10, wherein Y is OR R. ': and Z is O.

14. The compound of claim 1 wherein X and Y taken together form a cycSoalkyl. heterocyclyl, aryl or heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl or heteroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo. Ct_7-alkyl, Ci-7-alkylcarbonyl, Ci.y-alkyioxycarbonyl, C1- -alkaxy, Cj.7- alkoxyalkyl, (Ra)2(Rb)N~ and Cj.7-aikyl-S(0)o-2-, wherein R is independently, at each occurrence, hydrogen or Ci-7-alkyl and R0 is an electron pair, hydrogen or Ci-7-alkyl, and provided that when X and Y form phenyl, pyridyl, pyridyl -N-oxide or pyrimidinyl then Z is not O, wherein the compound has the following structure (X), and wherein V represents the cycloalkyl, heterocycly!, aryl or heteroaryl:

15. The compound of claim 14, wherein Z is CR'°R" and having the following structure (XI):

16. The compound of claim 14, wherein Z is CR^R'1 and R and R?t taken together form oxo (=0) and having the following structure (XII):

17. The compound of claim 14, wherein Z is O and having the following structure (XIII):

] 8. The compound of claim 14, wherein Z is S(O)0-2 and having the following structure (XIV):

19. The compound of claim 1 8 , wherein Z is ·

20. The compound of any one of claims 14-19, wherein the compound has one of the following structures (Xa), (Xb), (Xc), (Xd), (Xe), (Xfh (Xg), (Xh), (Xi), (Xj), (Xk), (XI), (Xm), ( Xn) (Xo), (Xp). (Xq), (Xr) or (Xs):

(Xe)

(Xm) (Xn) wherein:

Re is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, Ci. aikyl. Ct.7-hal0aik.yl, Ci-7-alkykarbonyl, Ci -7-aikyloxycarhonyl, C1-7-alkoxy, alkoxyalkyl, (RA)2(RB)N- and Ci_7-alkyl-S(0)o-2-; and

RF is an electron pair, hydrogen or Ci.7-alkyl.

70„7i

21. The cornpound of claim 20, wherein Z is CR' 'R

22... The compound of claim 20, wherein Z is CR'°R71 and R C and R' 1 taken together form oxo (=0). 2,3 . The compound of claim 20, wherein Z is O,

The compound of claim 20, wherein Z is -S(0)o-2-

The compound of claim 24, wherein Z is -8C½-.

26. The compound of claim 2, wherein Y is absent and Z is O and having the following structure (XV) i

27. The compound of claim 26, wherein R ' and R5i taken together with the C atom to which they are attached fonn a cycloalkyl or heterocyclyl, wherein the cycloalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci-7-a!kyl, C|.7-aikylcarbony1, C^-alkyloxycafbonyl, Cj.7- alkoxy, d-7-alkoxyalkyl and C^-alkyl-Si'QV-r, wherein the compound has the following structure (XVI) and wherein W represents the cycloalkyl or heterocycly group:

28. The compound of claim h wherein Al and A2 are both CR

29. The compound of claim 28, wherein R! is hydrogen.

30. The compound of any one of claims 1-29, wherein R"' and R4 together are -L-(CR! 7Rl8); and form part of a ring.

31. The compound of claim 30, wherein the compound has the following structure (XVII):

(XVII)

17 The compound of claim 31, wherein L3 is -C(::0)-, -S-, -S(0)2- or -N(R2lV. 3 , The compound of claim 32, wherein R is C3-7-cycloalkyl

34. The compound of claim 31 , wherein the compound has one of the

(XVIIa) (XVIIb)

(XVIIc (XV lid)

35. The compound of claim 34, wherein the compound of structure (XVIIa) has the following structure (XVIIa- i):

(XVIIa- 1 ) wherein:

Rc is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, Ci_7-alkyL Ci -7-alk lcarbonyl, Ci-7-alkoxy, C1-7- alkoxyalkyl or Ci-7-alkyi-S(0)o-2~-

36. The compound of claim 35, wherein Rc is hydrogen.

37. The compound of any one of claims 31-36, wherein Y is O and Z is CR70R71.

38. The compound of any one of claims 31-36, wherein Y is Nil' and Z is CR70R71.

39. The compound of any one of claims 31-36, wherein Y is NR6" and Z is O.

40. The compound of any one of claims 31-36, wherein Y is NRb2 and Z is S(O)0-2.

41. The compound of any one of claims 31-36, wherein Y is CR60R61 and Z is CR70R71.

42, The compound of any one of claims 31-36, wherein Y is

CRb"Rb i and Z is O.

43. The compound of any one of claims 31-36, wherein Y is

CR°¾6i and Z is S.(0)o-2.

44. The compound of claim 34, wherein the compound of structure (XVIia) has one of the following structures (XV

(XVIIa-2) "Ta-3) wherein: Re is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, Chalky!, Ci-7-haloalkyi, C<-7-dkylcaxbonyl, C 1.7-alky!oxyearbonyl, Ci.7-alkoxy, C1.7- alkoxyaikyl, (Ra)2(Rb)N- and C;-7-alkyl-S(0)o-2-; and

R1 is an electron pair, hydrogen or C<-7-alkyI.

45. The compound of any one of claims 31-34 and 44, wherein Z is

:R70„71

46. The compound of any one of claims 31-34 and 44, wherein Z is CR70R l and R70 and R taken together form oxo (=0).

47. The compound of any one of claims 31-34 and 44, wherein Z is

48. The compound of any one of claims 31-34 and 44, wherein Z is

49. The compound of claim 48, wherein Z is -S(0)2-

50. The compound of claim 30, wherein, the compound has the following structure

(xvin)

51. The compound of claim 50, wherein R*" is C 7" )2. 52, The compound of claim 50, wherein the compound has one of the following structures (XVIIIa), (XVIIIh), (XVIIIe), (XViild), (XVIIIe), (XV!Ilf), (XVII

(XVIIIe) (XVIIIf)

(XVIIIj)

Ik) (XVIIli)

53. The compound of claim 1. wherein A1 and A"' are each independently CM or N and R" is Q-7-aIkoxy, ~O~C3.7-cycloa.kyl, or -0-C1-7-alkyl- C3-7-cycloalkyl.

54. The compound of claim 53, wherein the compound has one of the following atractures (XlXa), (XlXb), (XlXc), (XlXd), (XIXe), (XlXf), or (XLXg):

(XIXe) (XlXf)

55. The compound of claim 54, wherein the compound has the structure (XIX g).

56. The compound of any one of claims 53-55, wherein X is

CRS0R51.

57. The compound of claim 56, wherein Rw and R51 taken together with the C atom to which they are attached form a cycloalkyl or heierocyclyl, wherein the cycloalkyl or heierocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C1-7-alkyl5 Ci-7-alky1earbonyl, Ci-7-alkyloxycarbonyl, C1.7- alkoxy, Ci-7-aikoxyalkyl and Ci.7-alkyl-S(0)o-2-.

58. The compound of claim 57, wherein R50 and Rji taken together with the C atom to which they are attached form a cyclopropyl.

59. 'The compound of any one of claims 56-58, wherein Y is O and Z is CR70R?i.

60. The compound of any one of claims 56-58, wherein Y is NR'2 and Z is CR70R71. 61. The compound of any one of claims 56 -58, wherein Y is NR67" and Z is O,

62. The compound of any one of claims 56-58, wherein Y is NR0Z and Z is S(0)o-2.

63. The compound, of any one of claims 56-58, wherein Y is CR60R61 and Z is CR70R71.

64. The compound of any one of claims 56-58, wherein Y is CR60R61 and Z is 0.

65. The compound of any one of claims 56-58, wherein Y is CR60R61 and Z is S(O)0.2.

66. The compound of any one of claims 53-55, wherein X and Y taken together form, a cycloalkyl, heterocyciyl, aryl or heteroaryi, wherein the cycloalkyi, heterocyciyl, aryl or heteroaryi. are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci_7-alkyl, Ci .7-alkylcarbonyi, C1-7- alkyloxycarbonyl, Ci-7-alkoxy, C^-aikoxyalkyl, (Ra)2(Rb)N- and C3-7-alkyl-S(0)o-2-, wherein Ra is independently, at each occurrence, hydrogen or Ci..7-alkyl and Rb is an electron pair, hydrogen or Ci-7~alkyi, and provided that when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyrimidinyl then Z is not ().

67. The compound of claim 66, wherein X and Y taken together form a heterocyciyl.

68. The compound of claim 67, wherein the heterocyciyl is pyrrolidinyl or thiazolidinyl.

69. The compound of any one of claims 66-68, wherein. Z is

70. The compound of any one of claims 66-68, wherei Z is CR70R?i and R'° and R'1 taken together form oxo (=0).

71. The compound of any one of claims 66-68, wherein Z is O.

72. The compound of any one of claims 66-68, wherein Z is S(0)o-2-

73. The compound of any one of claims 66-68, wherein Z is S(0)T-

74. The compound of claim 1, wherein A1 is CR1- and A" is CRl4 and wherein R13 and Rt4 are independently from each other selected from hydrogen, halogen. halogen-Ci..7-alkyl and C3.7-al.koxy.

75. The compound of claim 1, wherein A1 is CR1" and A is N, with R13 being independently from each other selected from hydrogen, halogen, halogen- Ci.7-alkyl and Cj.y-alkoxy,

76. The compound of claim 1, wherein R1 and R.2 are independently from each other selected from the group consisting of hydrogen, halogen and halogen- Cj.7-alkyi.

77. The compound, of claim i , wherein R3 and R4 together are -L3- (CR1 ;Ri8)n- and form pari of a ring; wherein

L is selected from -CR 'R* '- and -NR"-;

RJ7 and R18 are independently from each other selected from hydrogen and C] -7~alkyi; R19 and R l) are independently from each other selected from hydrogen, Cj-7alkyL Ci..?-aikoxyearbonyl, unsubsiiiuted heterocyciyi and heterocyciyi substituted, by one or two groups seiected from Ci-y-aikyl and halogen;

or R1 and R20 together with the C atom to which they are attached form a cyclopropyl or oxetanyl ring or together form a =CH2 or :;-CF2 group;

R"1 is selected from hydrogen, Cv?-alkyL C3.7- cycloalkyl and Cj-T-cycloaikyl-Ci-y-alkyl, wherein C3.7-cycioalkyl is unsubstituted or substituted by carboxyl-Ci-7-alkyj or C!-7 -aikoxycarbonyl, heterocyciyi, heterocyciyi-- C: .7-al.kyl, heteroaryl, heteroaryl-Ci-7-alkyl, carboxyl-Cj -7-alkyl, Ci-7-alkoxycarbonyl- Ci-7-alkyI, C j..7-alkylcarbonyloxy-C 1.7-alkyi, Ci.7~alkylsulfony!, phenyl, wherein phenyl is unsubstituted or substituted by carboxyl-Cj,7-alkyl or C 1.7-alkoxycarbonyl, phenylcarborryl, wherein phenyl is unsubstituted or substituted, by earboxyl-Ci.7-alkyl or Cj.y-a.lkoxycarbonyk and phenylsulfonyl, wherein phenyl is unsubstituted or substituted by earhoxyi-Ci-7-alkyl or C 1.7-alkoxycarbonyl;

or R21 and a R!7 together are -(CH );r and form part of a ring, or RJ together with a pair of R17 and R18 are -CH=CH-CH= and form part' of a ring; and

n is 1 , 2 or 3.

78. The compound of clahri 1 , wherein:

L1 is -NR21-,

R21 is seiected from hydrogen, C1-7-alkyi, C3.7-cycloai.kyl and C3-7- eycloalkyl-Ci-7-alkyl, wherein C3.7-cycloal.kyl is unsubstituted or substituted by carboxyl-Ci-7-alkyl or C^-aikoxyearbonyl, and Cj.7-a]kylsuifonyl;

R!7 and R18 are independently from each other selected from hydrogen and methyl; and

n is 2.

79. The compound of claim 1, wherein L1 is -CH2-, R1 ' and R18 are independently from each other selected from hydrogen and methyl and n is 2. 80, The compound of claim. 1 , wherein RJ and R together are - L1- (CR! ' R.,8)n~ and form part of a ring: wherein 1/ is -NR.2'- or -O- R2t is selected from hydrogen, C ;,7-alkyl and C 3.7-cycloalkyi, Ri7 and R!8 are independently from each other selected from hydrogen and methyl, and n is 2.

The compound of claim 80, wherein L1 is -(>- and the compound has the following

(XV)

The compound of claim 81 , wherein R' and R are hydrogen

83. The compound of any one of claims 80-82, wherein X is

CR50R51.

84. The compound of claim. 83, wherein R50 and R51 taken together with the C atom to which they are attached form a cycloalkyl or heterocyclyl, wherein the cycloalkyl or heteroeyelyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci-7-alkyI, C1-7-alkylcarbonvl, C!- -alkyloxyearbonyl, Ci-7- alkoxy, Ci-7-alkoxyalkyl and Ci-7-a!kyl-S(0)o-2--

85. The compound of claim 84, wherein R3° and R.51 taken together with the C atom to which they are attached form a cyclopropyl. The compound of any one of claims 83-85, wherein Y is O and Z is CR7¾7i .

87. The compound of any one of claims 83-85, wherein Y is NR. and Z is CR70R.

88, The compound of any one of claims 83-85, wherein Y is N.R.6i and Z is O.

89. The compound of any one of claims 83-85, wherein Y is NR' and Z is S(0)o.-2.

90. The compound of any one of claims 83-85, wherein Y is CR°"'R01 and Z is CR,UR71.

91. The compound of any one of claims 83-85, wherein Y is CR60R61 and Z is 0.

92. The compound of any one of claims 83-85, wherein Y is Π 'Κί ; and Z is S(O)0.2.

93. The compound of any one of claims 80-82, wherein X and Y taken together form a cyc!oalkyl, lieteroeyciyL aryl or heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl or heteroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C y-alkyl, C 1.7-a!kylcarbonyl, C1-7- alkyloxycarbonyl, (Ra)2(Rb)N- and Ci.7-alkyl-S(0)o-2-, wherein Ra is independently, at each occurrence, hydrogen or Ci-7-alkyl and Rb is an electron pair, hydrogen or Ci-7-alkyl, and provided thai when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyrimidinyi then Z is not O.

94. The compound of claim 93, wherein X and Y taken together form a heterocyciyl.

95. The compound of claim 94, wherein the heterocyclyl is pyrrolidinyl or thiazolidinyi.

96. The compound of any one of claims 93-95, wherein Z is

CR70R7\

97. The compound of any one of claims 93-95, wherein Z is CR7,JR/l and R arid R/l taken together form, oxo (=0).

98. The compound of any one of claims 93-95. wherein Z is O.

99. The compound of any one of claims 93-95, wherein Z is S(0)o-2.

100. The compound of any one of claims 93-95, wherein Z is -S(0)2-

101. The compound of claim 1, wherein RJ is selected from hydrogen, C j -alkyi, C i„7-a!koxy, N-heterocyciyl and -NRl5R16, wherein R!S and R16 are independently from each other selected from hydrogen, C 1.7-alkyl and C 3.7-cycioalkyi, and R'; is hydrogen or methyl.

102. The compound of any one of claims 1-101, wherein at least one of R R9, Ri0, Rn or R12 is halogen, C1-7-alkyl, halogen-Ci-7-alkyl. C,...7~aikoxy, halogen~C .7~al.koxy or cyano.

103. The compound of any one of claims 1-102, wherein the halogen is ch!oro.

104. The compound of claim 102 or 103, wherein the other ones of R R9, u', Rn and R12 are hydrogen.

105. The compound of any one of claims 1-104, wherein the compound has one of the following structures (XXa), (XXb), (XXc), (XXd), (XXe), (XXi

(XXc) (XXd)

(XXi) iXXj )

(XXI)

106. The compound of any one of claims 1-103, wherein at least one of R8, R9, RUI5 RN or R,2 is Q.

107. The compound of claim 106, wherein or R1U is Q.

108. The compound of claim 106 or 107, wherein the other ones of R. , R9, R30, Rn and R are selected from the group consisting of hydrogen, halogen, Cj.?- aikyl, halogen-Ci.7-aikyl, d-7-alkoxy, halogen-Ci-7-alkoxy and cyano.

.109. The compound of any one of claims 106-108, wherein the compound has one of the following structures (XXia), (XXIb), (XXIc), (XXId), (XXIe), (XXIf), (XXIg), (XXIli), (XXIi), iXXij). (XXIk) or (XXII ):

(XXIh)

iXXLk) (XXII)

1 10, The compound of any one of claims 1-103 and 106-109, wherein L2 is -0-, -Ci-7aikylene-; -C^alkylene-NR80-, -Ci-7all ylene-NR80C(=O)-, -Cl.7a[kylene-C(-0)NR8°- or -C, .7alkylene-NR80C(-O)NR80-.

1 1 1. The compound of any one of claims 1-103 and 106-110, wherein Q is -L2CR81R82(CR83R8 )fl,iG, wherein:

R81, R8 , R83 and R84 are independently, at each occurrence, hydrogen or hydroxy!;

G is -CI¾, -Cf ! ,()! 1, -C02H or -iM; and

ml is an integer ranging from 1 to 21.

112. The compound of any on of claims 1-103 and 106-111, wherein Q is wherein:

R81 , R82, R83 and. R84 are independently, at each occurrence, hydrogen or bydroxyl;

G is -CH3, -CH2OH, or -CG2H; and ml is an integer ranging from 1 to 21.

1 13. The compound of claim 1 1 1 or 112, wherein for each occurrence

> 84

of R83 and R84, one of RS3 or R84 is hydrogen and the other of R E?e8j3 o nrr R T? mS4 i ;sU h V,y:,dAr- oxyl.

114. The compound of any one of claims 1-103 and 106-113, wherein Q has one of the following structures (XXIia), (XXIib), (XXIIc), (XXTld), (XXIIe), (XXIIfL (XXIIg), (XXilh), (XXIIi), (XXIIj), (XXIIk), (XX!I!), (XXIIm), ( XXf ln ). (XXilo) or (XXIIp):

(XXIIa) (XXHb)

XXIIe)

(ΧΧΪΙΓ) (XXIIm)

ίΧΧΠη) (ΧΧΠ6)

(XX Up)

wnerem:

Rg0 is hydrogen or C1-7alkyl;

Rg is independently, at each occurrence, hydrogen or Chalky!;

Rh is an electron pair, hydrogen or Ci-7alkyl; and

xl, x2 and x3 are each independently an integer ranging from 1 to 6.

115. The compound of claim 114, wherein RX'J is hydrogen or methyl.

1 16. The compound of claim 1 14, wherein x l is 2 or 3.

1 17. The compound of any one of claims 1-103 and 106-1 10, wherein Q is wherein m2 is 2 or 3, m3 is an integer ranging from. 1 to 21 and R86 is hydrogen, hydroxy! or L"'-L

118. The compound of any one of claims 1 -103, 106-110, and 117, wherein Q is -L-i[(CH2)n O]m3(CH2)m2R.8t>> wherein ml is 2 or 3, m3 is an integer ranging from 1 to 21 and Rbo is hydrogen or hydroxyl.

119. The compound of any one of claims 1-103, 106-110, and 117, wherein Q has XIIle):

(XXIIIa)

H o

(XXIHc)

wherein T is a compound of structure f i).

120. The compound of any one of claims 1-103 and 106-1 10, wherein B has the following structure (XIV):

121. The compound of any one of claims 1-101, wherein at least two of R8, R9, R¾0, R11 and R12 are selected from:

C (.7-alkyl, C 2-7-alkenyl, C2.7-allda.yL, halogen, halogen-C j .7-alkyl, C1.7- aikoxy, halogen-Q .y-aikoxy, hydroxy. hydroxy~Ci-7-alkoxy5 hydroxy-Ci -7-alkyl, hydroxy-C3-7-alkenyl, hydtOxy-C3.7-alkynyl, cyano, carboxyl, Cj .7-alkoxycarbonyl, amino carbonyl, carboxyl-Cj.7-alkyl, carboxyJ-C2.7-alkenyl, carboxyl-C2.7-alkyny 1 , C1-7- alkoxycarbonyl-Ci-7-alkyl, Ci.7-alkoxycarbonyl-C2-7-alkenyl5 Ci -7-alkoxycarbonyl-C2-7- alkynyl, carboxyl-C [.7-alkox.y, Ci.7-alkoxycarbonyl-Ci-7-alkoxy, carboxyl-Ci-7-alkyl- aminocarbonyl, carboxyl-Ci.y-aikyl-i'Ci.T-alkylairiinol-carbonyl, C-;.7-alkoxycarbonyl - Ci-7-alkyl-aminocarbonyl, (Ci-7-alkylamino)-carbonyl, carboxyl-Ci.7-alkyl-arniiiocarbonyl-Ci.7-alkyl, carboxyl-C^-alkyHCi^-alkylam o)- carbony I -C 1 -7-alkyl, Ci.7-alkoxycarbonyl-Ci-7-alkyl-arnin.ocarboiiyl-Ci.7-alkyl, Q.7- a]koxycaitoiiyl-Ci-7-alkyl-(Ci-7-aikyla.mino)-carbony hydroxy-Ci.7-alkyi- aminocarbonyL di-(hydroxy-C _7-alkyI)aminocarbonyl, arninocarbonyl-Ci.7-alkyl- amino carbonyl, hydro xysulfonyl-Ci-7-aIkyl-aminocarbonyl, hydroxysulfo nyl-Ci-7-' alkyl-(Ci.7-a]kyl-arriino)-carbonyl, di-(C1_7-alkoxycarbonyl-Ci-7-alkyl)- methylaminocarbonyl,

phenyl, wherein phenyl is imsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and

phenyl-carbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C1_7-alkoxy, carboxyl and ^-alkoxycarbonyL phenyl-aminocarbonyl, wherein phenyl is unsubstituted or subsdtuled by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and Cj.7- alkoxycarbonyl. phenyi-Ci-7-alkyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci..7-alkoxy, carboxyl and d -7-alkoxycarbonyl, phcnyl-C2-7-a!kynyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and C1-7- alkoxycarbonyl,

heteroaryl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and ..7-alkoxycarbonyl, heteroaryl-carbonyl, wherein heteroaryl is unsubstituted or- substituted by one to three groups selected from halogen, C1.7-alk.oxy, carboxyl and C1-7- alkoxycarbonyi,

heteroaryl-aminocarbonyl. wherein heteroaryl is unsubstituted- or substituted by one to three groups selected from halogen, d-7-alkoxy, carboxyl and C i ..7-alk0xyca.rb0.nyl ,

heteroaryl-Ci-7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, d-7-alkyl. Cj .7-al.k0xy, carboxyl and C 1 -7-alkoxy carbonyl,

heteroaryl-Ci-7-a!kyl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C-alkoxy, carboxyl and C 5.7-alkoxycarbonyl, and

heteroaiyl-carbonyi-d-7-aIkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-aIkoxy, carboxyl and C .7-alkox y carbonyl ,

and the other ones of R , R9, Rl0, R1 1 and !2 are hydrogen.

122. The compound of any one of claims 1-101, wherein at least two of R8, R9, Riy, Ru and. R52 are selected from:

halogen, hydroxy, hydroxy-Ci-7-aikoxy, hydroxy-C1.7-alk.yl, cyano, carboxyl, Ci .7-alkoxycarbonyl, amino carbonyl, carboxyl-Ci.7-alkoxy, C1..7- alkoxycarbonyl-Ci.7-alkoxy, c rboxyl-Ci.7-alkyl-amii-iocarbonyl, carboxyJ-d-7-alkyi- (Ci-7-alkylamirto)-carbon.yl, Ci.7-alkoxycarbonyl-C 1.7-alkyl-aminocarbonyl, hydroxy- C3.7-alkyl-aminocarboriyl , di-(hydroxy-(^i..7-aikyI)aminocarbonyl, aminoearbonyl-Ci-7- alkyl-anaino carbonyl, hychOxysulfonyl-Ci.7-a]kyl-aminocarbonyl9 hydroxysulfonyl- Ci.7-alkyl-(Ci.7-alkyl-amino)-carbonyl, di-(C j .7-alkoxycarbonyl-Ci.7-alkyl)- niethylaminocarbonyl,

phenyl-aminocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Cj.7-alkoxy, carboxyl and C1.7- alkoxy carbonyl ,

heteroaryl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C^-alkoxy, carboxyl and C j .7-alkoxy carbonyl,

heteroaryl-Ci-7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to tliree groups selected from halogen, Ci.7-alkyl, Ci.7-alkoxy, carboxyl and C 1..7-alkoxycarhony ί ,

heteroaryi-C i -alkyl-aminocarboiiyL wherein heteroaryl is unsubstituted. or substituted by one to three groups selected from halogen, C1-7--aikoxy, carboxyl and C-alkoxycarbonyl, and

heteroaryl-carboiiyi-Ci -alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and C j -alkoxycarbonyl,

and the other ones of Rs, R9, R! , R1 1 and R12 are hydrogen.

123. The compound of any one of claims 1-101, wherein at least one of R8, R\ R10, RH and Ri2 is Q and at least one of Rs, R9, Ri0, Rn and R12 are selected from:

C i-7-aik l, C 2.7-alkenyl, C2.7-ail nyl, halogen, halogen-C^-alkyl. C i .7~ alkoxy, halogen-Ci-7-alkoxy, hydroxy, hydroxy-Cj -7-alkoxy, hydtoxy-Ci.7-aikyl, hydroxy-C^-alkenyl. hydroxy-C3.7-alkynyk cyano, carboxyl, Ci.7-alkoxycarbonyl, amino carbonyl, ca.rboxyl-C1_7-a.lkyL carboxyl-C2-7-alkenyl, carboxyl-C2-7-alkynyls C1.7- alkoxy carbonyl -C i -7-a). ky L Ct.7-alkoxycarboriyl-C2.7-alkenyl5 C 1 -7-alkoxycarbonyi-C?..7- alkynyi , car boxyl-C i .7-alkoxy, C^-alkoxycarbonyl-C j -7-alkoxy, carboxyl -C 1.7-alky 1 - aminoearbonyl. carboxyl-Ci.7-alkyl-(Ci.7-alkylamino)-carbonyt, Cj-7-aikoxycarbonyl- Ci -alkyl-aminocarbonyL Ct-7-a1koxycarbonyl-Ci-7-alkyl- (Ci-7-alkylamino)-carbonyl, carboxyl~Cu7-a!kyi-arainocarbonyl-Ci..7-alkyl, carboxyl-Ct„7-alkyl-(Ci-7-alkylaimno)- carbonyl-Cj.7-a.kyi, C1.7-alkoxycarbonyI-C1.7-aikyl-an-inocarbo.nyl-C1.7-alk.yl, C1.7- alkoxycarbonyl-Ct.7-alkyl-(Ci.7-alkyiamino)-carbonyi-Ci-7-alkyl, hydroxy-Ci.7-alkyl- aminocarbonyl, di-(hyd.roxy-C^7--alky.l)aminocarbonyl, aminocarbonyl-Cj.7-alkyl- amino carbonyl, hydro xysulfonyl-C]_7~alky{-aminocarbonyl , hydro xysulfo nyl-Ci-7- a]ky!-(Ci-7-alky].-amlTio)-carbonyl, di-(Ci-7-alkoxycarbonyl-Ci.7-alkyl)- rae4y3aminoca.rbo.ny 1 ,

phenyl, wherei phenyl is unsubstituted or subsiiioied by one to three groups selected from halogen, d-7-alkoxy, carboxyl and Ci.7-alkox carbonyl,

phenyl-carbonyl, wherei phenyl, is unsubstituted or substituted by one to three groups selected from halogen, C^-alkoxy, carboxyl and Ci.7-alkoxycarbonyl, plienyl-aminocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and C1.7- alkoxycarbonyl,

phenyi-C1.7-alk.yl, wherein plienyl is unsubstituted or substituted by one to three groups selected from halogen, d -7-alkoxy, carboxyl and Ci.7-alkoxycarbonyl, phenyl-C2-7-alkynyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci .7-alkoxy, carboxyl and C1.7- alkoxycarbonyl,

heteroaryl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Q.7-alkoxy, carboxyl and Ci.7-alkoxycarbonyl, heteroaryl-carbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected .from halogen, Ci-7-alkoxy, carboxyl and Ci-?- alkoxy carbonyl,

heteroaryi-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen. Ci -7-alkoxy, carboxyl. and Ci-7-aikoxycarbonyi.

heteroaryl-Ci -7-alkyl, wherein heteroaryi is unsubstituted or substituted by one to three groups selected from halogen, Ct.7-a.kyl, d-7-alkoxy, carboxyl and C] -7-alkoxycarbonyl, heteroaryl-Ci-7-alkyl-aminocarbon.yl, wherein heieroaryl is tmsubstituted or substituted by one to three groups selected from halogen, C-alkoxy, carboxyl and C'3 -7-alkoxycarbonyl, and

heteroaryl-carbonyl-C 1.7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Cj.7~alk.0xy, carboxyl and C i -7-alkoxycarbonyl5

and the other ones of Rs, R9, R10, R1 1 and are hydrogen.

124. The compound any of claims 1— 101 , wherein at least one of R ', R9, R!0, R1 1 and Ri2 is Q and at least one of R8, R95 Ri0, R5 1 and R12 are selected from:

halogen, hydroxy, hydroxy-C 1.7-alkoxy, hydroxy-Ci-7-alkyl, cyano, carboxyl. Ci .7-aikoxycarbonyl, amino carbonyl, carboxyl-Curalkoxy, Cj.7- alkox carbonyl-C j-7-alkoxy, carboxyi-C>..7-alkyl-arninocarbonyi, carboxyl-Ci.7-alkyl- (C|-7-alkylamlno)--earbonyl, C1.7-alkoxYcarbonyi-Ci-7-aikyl-an:unocarbonyi! hydroxy- C j .7-alkyl-arninocarbonyi, di-S¾ydroxy~Ci-7-aikyl)aminocarbonyi, aminocarbonyl-C f .7- alkyl-amino carbonyl, hydroxy sulfonyl-Ci-7-alkyl-aminocarbonyl, hydroxysulfonyl- Ci.7-alkyl-(Ci„7-alkyl-amino)-carbonyl, di-(C j ^-alkoxycarbonyl-Ci^-alkyl)- metiiylaminocarbonyl,

pheiiyl-ammocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci -7-alkoxy, carboxyl and C1.7- alkoxy carbonyl,

heteroaryl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected irom halogen, C1-7-al.koxy, carboxyl and C i -7-alkoxycarbony 1 ,

heteroaryl-Ci-7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C >.7-alkyl, Ci-7-alkoxy, carboxyl and C 1 -7-alkoxycarbonyl .

heteroaryl-Ci-7-alkyl-aniinocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C1-7-alkoxy, carboxyl and C-aikox carbon l , and heteroaiyl-carbonyl-Ci..7-alkyl, wherein heteroaryl is unsubsiituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and C Ϊ - -alkoxyc arbony 1 ,

and the other ones of Rs, R9, R10, R.1 1 and R12 axe hydrogen.

125. The compound of any one of claims 1-101, wherein s and R! 1 are halogen and R9, R.10 and R12 are hydrogen.

126. A compound of any one of Examples 1 -291.

127. A pharmaceutical composition comprising a compound of any one of claims 1-126 and a pharmaceutically acceptable carrier or adjuvant.

128. Use of the compound of any one of claims 1-126 as a therapeutically active substance.

129. Use of a compound of any one of claims 1-126 as a therapeutic active substance for the treairaent of diseases which are associated with the modulaiion of TGR5 activity.

130. A method for the treatment of diseases which are associated with the modulation of TGR5 activity, wherein the diseases are selected from diabetes. Type II diabetes, gestational diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, hyperglycemia, obesity, metabolic syndrome, ischemia, myocardial infarction, retinopathy, vascular restenosis, hypercholesterolemia, hypertriglyceridemia, dyslipidemia or hyperlipidernia, lipid disorders such as low HDL cholesterol or high LDL cholesterol, high blood pressure, angina pectoris, coronary artery disease, atherosclerosis, cardiac hypertrophy, rheumatoid arthritis, asthma, chronic obstructive pulmonary disease (COPD), psoriasis, ulcerative colitis, Crohn's disease, disorders associated with parenteral nutrition especially during small bowel syndrome, irritable bowel syndrome (IBS), allergy diseases, fatty liver, non-alcoholic fatty liver disease (NAFLD), liver fibrosis, non-alcoholic steatohepatitts (NASH), primary sclerosing cholangitis (PSC), liver cirrhosis, primary biliary cirrhosis (PBC), kidney fibrosis, anorexia nervosa, bulimia nervosa and neurological disorders such as Alzheimer's disease, multiple sclerosis, schizophrenia and impaired cognition, the method comprising administering a therapeutically active amount of a compound of any one of claims 1-126 to a patient in need thereof.

131. The method of claim 127, wherein the disease is diabetes.

132. The method of claim 127, wherein the disease is Type II diabetes or gestational diabetes.

133. Use of the compound of any one of claims 1-127 for the preparation of medicaments for the treatment of diseases which are associated with the modulation of TGR5 activity.

134. The use according to claim 133 for the preparation of medicaments for the treatment a disease or condition selected from diabetes. Type 11 diabetes, gestational diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, hyperglycemia, obesity, metabolic syndrome, ischemia, myocardial infarction, retinopathy, vascular restenosis, hypercholesterolemia, hypertriglyceridemia, dyslipidemia or hyper!ipidemia, lipid disorders such as low HDL cholesterol or high LDL cholesterol, high blood pressure, angina pectoris, coronary artery disease, atherosclerosis, cardiac hypertrophy, rheumatoid arthritis, asthma, chronic obstructive pulmonary disease (COPD), psoriasis, ulcerative colitis, Crohn's disease, disorders associated with parenteral nutrition especially during small bowl syndrome, irritable bowl disease (IBS), allergy diseases, fatty liver, liver fibrosis, liver cirrhosis, liver colestasis, primary biliary cirrhosis, primary scleroting cholangitis, kidney fibrosis, anorexia nervosa, bulimia nervosa and neurological disorders such as Alzheimer's disease, multiple sclerosis, schizophrenia and impaired cognition.

135. The use according to claim 134, wherein, the disease is diabetes.

136. The use of claim 134, wherein the disease is Type II diabetes or gestational diabetes.

137. A pharmaceutical composition comprising a compound of any one of claims 1-126, a phanTiaceuticaliy acceptable carrier or adjuvant and one or more additional biologically active agents.

138. The pharmaceutical composition of claim 137, wherein the one or more additional biologically active agents are selected from dipeptidyl peptidase 4 (DPP-4) inhibitors, - biguanidkt.es, sulfonylureas, a-giucosidates inhibitors, thiazolidinediones, incretin mimetics CB1 antagonists, VPAC2 agonists, giucokinase activators, glucagon receptor antagonists, PEPCK inhibitors, SGLT1 inhibitors, SGLT2 inhibitors, IL-1 receptor antagonists, SIRT1 activators, SPPARMs and I ipiISDl inhibitors.

139. The pharmaceutical composition of claim 137, wherein the one or more additional biologically active agents prolong the TGR5 -mediated GLP-1 signal.

140. The pharmaceutical composition of claim 137, wherein the one or more additional biologically active agents are DPP-4 inhibitors.

141. The pharmaceutical composition of claim 137, wherein the one or more additional biologically active agents are sitag!iptin, vildagliptin, saxagliptin, linagliptin, alogliptin, gemigjiptin, omarigliptin or dutogiiptin.

142. The pharmaceutical composition of claim 137, wherein the one or more additional biologically active agents are selected from the group consisting of metformin or other biguanidine, giyburide or other sulfonyl urea, acarbose or other a- glucosidase inhibitor, rosiglitazone or other thiazolidinedionc and cxenatide or other incretin mimetic.

143. A method for treating Type II diabetes raellitus in a patient in need thereof, the method comprising administering to the patient an effective amount of a compound of any one of claims 1-126 or a pharmaceutical composition according to any one of claims 127 or 137- -142.

144. A method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR.5 agonist that is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist administration does not induce the filling of the gall bladder of the mamma! as determined by ul trasound analysis,

145. A method for stimulating GLP-i secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the patient and wherein the TGR5 agonist administration does not induce the emptying of the gall bladder of the mammal as determined by ultrasound analysis.

146. A method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the patient and. wherein the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 400% when compared to administration of a placebo.

147. The method of claim 146, wherein the change in weight of the mammal's gall bladder is determined in a mouse model.

148. The method of claim 146, wherein the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 200% when compared to administration of a placebo.

149. A method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist concentration in the gall bladder is less than about 100 μΜ

150. The method of claim 149, wherein the TGR5 asonist concentration in the gall bladder is detennined in a mouse model.

151. The method of claim 149, wherein the TGR5 agonist concentration in the gall bladder is less than about 50 uM.

152. The method of claim 149, wherein the TGR5 agonist concentration in the gall bladder is less than about 10 μΜ.

153. The method of claim 149, wherein the TGR5 agonist concentration in the gall bladder is less than about 1 μΜ.

154. The method of claim 149, wherein the TGR5 agonist concentration in the gall bladder is less than about 0.1 uM.

155. A method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist concentration in the mammal's plasma is less than the TGR5 EC50 of the TGR5 agonist.

156. The method of claim 155, wherein the TGR5 agonist concentration in the mammal's plasma is less than 50 ng/mL.

.157. The method of any of claims 144-156, wherein the TGR5 agonist is not systemically available.

158. The method of any of claims 144-154, wherein the TGR5 agonist concentration in the mammal's plasma is less than the TGR5 ECS0 of the TGR5 agonist,

159. The method of claim 158, wherein the TGR5 agonist concentration in the mammal's plasma is less than 50 rsg/raL.

160. The method of any of claims 144-156, wherein the TGR5 agonist does not modiilate TGR5~mediated suppression of cytokines.

161 . The method of any of claims 144- 456, wherein the TGR.5 agonist does not modulate the ileal bile acid tra sporter (IB AT).

162. The method of any of claims 144-156, wherein the TGR5 agonist does not modulate the Farnesoid X Receptor (FXR).

163. The method of any of claims 144-156, wherein the TGR5 agonist stimulates PYY sectretion.

164. The method of any of claims 144- 156, wherein the TGR5 agonist is a compound according to any one of claims 1-126.

165. A TGRS agonist, wherein the TGR5 agonist stimulates GLP-1 secretion i a mammal and is active in the gastrointestinal tract of the mammal and wherein administration of the TGRS agonist to the mammal does not induce filling of the gall bladder of the mammal as determined by ultrasound analysis.

166. A TGRS agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein administration of the TGR5 agonist to the mammal docs not induce emptying of the gall bladder of the mammal as determined by ultrasound analysis.

167. A TGR5 agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein administration of the TGR5 agonist to the mammal does not cause a change in weight of the mammal's gall bladder by more than 400% when compared to administration of a placebo.

168. The TGR5 agonist of claim 167, wherein the change in weight of the mammal's gall bladder is determined in a mouse model.

169. The TGR5 agonist of claim 167, wherein the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 200% when compared to administration of a placebo.

170. A TGR.5 agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mamma! and is active in the gastrointestinal tract of the mammal and wherein the TGR5 agon st is administered to the mammal, the concentration of the TGR5 agonist in the gall bladder is less than about 100 μΜ.

171. The TGR5 agonist of claim 1 0, wherein the TGR5 agonist concentration in the gall bladder is determined in a mouse model.

172. The TGR5 agonist of claim 170, wherein the TGR5 agonist concentration in the gall bladder is less than about 50 μΜ,

.173. The TGR5 agonist of claim 170, wherein the TGR5 agonist concentration in the gall bladder is less than about 10 μΜ.

174. The method of claim. 170, wherein the TGR.5 agonist concentration in the gall bladder is less than about 1 μ.Μ.

175. The method of claim 170, wherein, the TGR5 agonist concentration in the gall bladder is less than about 0.1 μΜ.

176. A TGR5 agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist is administered to the mammal, the concentration of the TGR5 agonist in the mammal's plasma is less than the TGR5 EC5.3 of the TGR5 agonist.

177. The TGR5 of claim 118, wherein the TGR5 agonist concentration in the mammal's plasma is less than 50 ng/mL.

178. The TGR5 agonist of any of claims 165-177, wherein the TGR5 agonist is not systemically available.

179. The TGR5 agonist of any of claims 165-177, wherein the TGR5 agonist concentration in the mammal's plasma is less than the TGR5 EC50 of the TGR5 agonist.

180. The TGR5 agonist of claim 179, wherein the TGR5 agonist concentration in the mammal's plasma is less than 50 ng/mL.

181. The TGR5 agonist of any of claims 165- 180, wherein the TGR5 agonist does not modulate TGR5 -mediated suppression of cytokines.

182. The TGR5 agonist of any of claims 165-180, wherein the TGR5 agonist does not modulate the ileal bile acid transporter (IB AT).

183. The TGR5 agonist of any of claims 165-180, wherein the TGR.5 agonist does not modulate the Faraesoid X Receptor (FXR).

184. The TGR5 agonist of any of claims 165-180, wherein the TGR5 agonist stimulates PYY secretion,

185. The TGR5 agonist of any of claims 165-180, wherein the TGR5 agonist is a, compound according to any one of claims 1—126.

186. A pharmaceutical composition comprising The TGR5 agonist of any of claims 165 -185 and a pharmaceutically acceptable carrier or adjuvant.

187. The pharmaceutical composition of claim 186, wherein the pharmaceutical composition further comprises one ox more additional biologically active agents.

188. The pharmaceutical composition of claim 187, wherein the one or more additional biologically active agents are DPP-4 inhibitors.

189. The pharmace tical composition of claim 187, wherein the one or more additional biologically active agents are sitagiiptin, vildagliptirx, saxagiiptin, linagliptin, alogliptin, gemigliptin, omarigliptin or dutogliptin.

1 0. A method for treating Type 1.1 diabetes meliitus in a patient in need thereof, the method comprising administering to the patient an effective amount of the TGR5 agonist according to any one of claims 165-185 or a pharmaceutical composition according to any one of claims 186-189.

Description:
NON-SYSTEMIC TGR5 AGONISTS

CRO 8 S -REFERENC E TO RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. § 1 19(e) of U.S. Provisional Patent Application No. 61/578,814 filed December 21 , 2011 and U.S. Provisional Patent Application No. 61/636,245 filed April 20, 2012. The foregoing applications are incorporated herein by reference in their entireties.

BACKGROUND Technical Field

The present invention is generally related to compounds having activity as TGR5 agonists, in particiilai- TGR5 agonists which are not systemically available. The compounds are useful for treatment of any number of TGR5 mediated diseases or conditions, including diabetes.

Description of the Related Art

Diabetes meliitus is an ever-increasing threat to human health. For example, in the United States current estimates maintain that about 16 million people suffer from diabetes meliitus. Type II diabetes accounts for approximately 90-95% of diabetes cases, killing about 193,000 U.S. residents each year. Type II diabetes is the seventh leading cause of all deaths. In Western societies, Type II diabetes currently affects 6% of the adult population with world-wide frequency expected to grow by 6% per annum. Although there are certain inheritable traits that may predispose particular individuals to developing Type II diabetes, the driving force behind the current increase in incidence of the disease is the increased sedentary life-style, diet, and obesity now prevalent in developed countries. About 80% of diabetics with Type II diabetes are significantly overweight. Also, an increasing number of young people are developing the disease. Type II diabetes is now internationally recognized as one of the major threats to human health in the 21 st century. Type II diabetes manifests as inability to adequately regulate blood- glucose levels and may be characterized by a defect in insulin secretion or by insulin resistance. Namely, those who suffer from Type Π diabetes have too little insulin or cannot use insulin effectively, insulin resistance refers to the inability of the body tissues to respond properly to endogenous insulin. Insulin resistance develops because of multiple factors, including genetics, obesity, increasing age, and having high blood sugar over long periods of time. Type ΪΙ diabetes can develop at any age, but most commonly becomes apparent during adulthood. However, the incidence of Type II diabetes in children is rising. In diabetics, glucose levels build up in the blood and urine causing excessive urination, thirst, hunger, and problems with fat and protein metabolism. If left untreated, diabetes mellitus may cause life-threatening complications, including blindness, kidney failure, and heart disease,

Type II diabetes is currently treated at several levels. A first level of therapy is through diet and/or exercise, either alone or in combination with therapeutic agents. Such agents may include insulin or pharmaceuticals that lower blood glucose levels. About 49% of individuals with Type II diabetes require oral medications, about 40% require insulin injections or a combination of insulin injections and oral medications, and 10% use diet and exercise alone.

Traditional therapies include: insulin secretagogu.es, such as sulph.onylureas, which increase insulin production from pancreatic β-cells; glucose- lowering effectors, such as metformin which reduce glucose production from the liver; activators of the peroxisome proliferator-activated receptor γ (PPARy), such as the thiazolidinediones. which enhance insulin action; and a-glucosidase inhibitors, which interfere with gut glucose production. There are, however, deficiencies associated with currently available treatments. For example sidphonylureas and insulin injections can be associated with hypoglycemic episodes and weight gain. Furthermore, patients often lose responsiveness to sulplionylureas over time. Metformin and a-glucosidase inhibitors often lead to gastrointestinal problems and PPARy agonists tend to cause increased weight gain and. edema,

More recently, new agents have been introduced to the market which prolong or mimic the effects of the naturally-secreted incredn hormones ( eiimiller, J Am Pharm Assoc. 49(suppl 1):S16~S29, 2009). Incretins are a group of gastrointestinal 2012/071251 hormones that are released from ihe beta cells of the pancreas when nutrients, especially glucose, are sensed in the gut. The two most important incretin hormones are glucose- dependent insulinotropic polypeptide (GIF) and GLP-1, which stimulate insulin secretion in a glucose-dependent manner and suppress glucagon secretion. However, GLP-1 itself is impractical as a clinical treatment for diabetes as it has a very short half- life in vivo. To address this, incretin-based agents currently available or in regulatory review for the treatment of T2DM are designed to achieve a prolonged mcretin-aetion. For example, the dipeptidyl peptidase-4 inhibitors, such as sitagliptin, inhibit the normally rapid proteolytic breakdown of endogenous incretin hormones. There are also human-derived and synthetic incretin. mimetics that are designed to he more stable and/or have a prolonged serum half-life compared to naturally secreted GLP-1, and include agents such as Hraglutide and exenatide. In either approach, the goal is to provide a sustained incretin response and thus enhance glucose-dependent insulin secretion. It is the glucose-dependence of the insulin response that provides incretin therapies with low risk of hypoglycemia. n addition, GLP-1 can also delay gastric emptying and otherwise beneficially affect satiety and hence, weigh loss (Neumiller 2009).

Although significant progress has been made, there remains a need in the art for compounds which prolong or mimic the effects of the naturally-secreted incretin hormones such as GLP-1. The present invention fulfills this need and provides further related advantages.

BRIEF SUMMARY

The present disclosure is directed to compounds having activity as TGRS agonists and are useful for treatment of any number of TGR5 related diseases or conditions, for example metabolic diseases such as diabetes. The compounds are substantially active in. the gastrointestinal (GI) tract to induce TGR5 -mediated signaling, with such interaction causing an increase in the secretion of incretins, including GLP-1. In some embodiments, the compounds are designed to be substantially non-permeable or substantially non-bioavailable in the blood stream; that is, such compounds are designed to stimulate the TGRS -mediated release of GLP-1 into the bloodstream but be substantially non-systemic (e.g., systemic exposure levels below their TGRS EC50) so as to limit their exposure to other internal organs (e.g., gall bladder, liver, heart, brain, etc.).

In accordance with one embodiment, there is provided a compound having the following structure (I):

(I)

or a stereoisomer, tautomer, pharmaceutically acceptable salt or prodrug thereof, wherein R 1 , II R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R !0 , R 11 , R i 2 , A 1 , A 2 , X, Y and Z are as defined herein.

Pharmaceutical compositions comprising a compound of structure (I), a pharmaceutically acceptable carrier or adjuvant and optionally one or more additional therapeutically active agents are also provided.

The present disclosure is further directed to a method of treatment for increasing systemic levels of GLP-1, the method comprising administering a compound as disclosed herein, and/or a pharmaceutical composition as disclosed herein, to a mammal in need thereof. Such methods may be used, in particular, to treat various metabolic disorders, including for example diabetes (e.g., Type II diabetes meilitus). In other embodiments, the methods include treatment of gestational diabetes, impaired lasting glucose, impaired glucose tolerance, insulin resistance, hyperglycemia, obesity, metabolic syndrome and/or other diseases and/or conditions.

These and other aspects of the invention will be apparent upon reference to the following detailed description. To this end, various references are set forth herein which describe in more detail certain background information, procedures, compounds ancl/or compositions, and are each hereby incorporated by reference in their entirety.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 illustrates gallbladder emptying after oral administration of Examples 176 and 178.

Figure 2 illustrates total (t)GLP~l and (t)PYY levels in mouse plasma following oral dosing of Examples 176 and 178.

DETAILED DESCRIPTION

I. Definitions

In the following description, certain specific details axe set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the invention may be practiced without these details. In other instances, well-known structures have not been shown or described in detail to avoid unnecessarily obscuring descriptions of the embodiments, Unless the context requires otherwise, throughout the specification and claims which follow, the word "comprise 5 ' and variations thereof, such as, ''comprises" and "comprising" are to be construed in an open, inclusive sense, that is, as "including, but not limited to." Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed invention.

Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment. Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily ail. referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments. Also, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. It should also be noted that the term "or" is generally employed in its sense including "and/or" unless the content clearly dictates otherwise.

The terms below, as used herein, have the following meanings, unless indicated otherwise:

"Amino" refers to the ~NH 2 radical.

"Aminocarbonyl" refers to the -C( ::=: 0)NH 2 radical.

"Carboxy" refers to the -C0 2 H radical'

"Cyano" refers to the -CN radical.

"Hydroxy" or "hydroxyi" refers to the -OH radical.

"Imino" refers to the =NH radical,

"Nitro" refers to the -N<½ radical.

"Qxo" or "carbonyl" refers to the :: ) radical.

"Tliioxo" refers to the =S radical.

"Guanidinyl" refers to the -NIIC(=NH)NH 2 radical.

"Amidinyl" refers to the -C(=NH)NH : radical.

"Phosphate" refers to the -OP(=0)(OH) 2 radical.

"Phosphonate" refers to the -P( :; =0)(OH)2 radical.

"Phosphinate" refers to the -PH(=0)OH radical.

"Sulfate" refers to the -OS(=0) 2 OH radical.

"Sulfonate" or "hydroxysulfonyl" refers to the -S(=0) 2 GH radical.

"Sulfmate" refers to the -S(=0)OH radical.

"Sulfonyl" refers to a moiety comprising a -SQ 2 ~ group. For example, "alkysulfonyl" or "alkylsulfone" refers to the -S0 2 -R a group, wherein R a is an alkyl group as defined herein.

"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated ( ,<?., 2012/071251 contains one or more double and/or triple bonds), having from one to seventy carbon atoms (Ci-C7o-alkyl), from one to twelve carbon atoms (Ci-Ci 2 -alkyl) or one to seven carbon atoms (Ci-C 7 -alkyl), and which is attached to the rest of the molecule by a single bond, e.g., methyl, ethyl, w-propyl, 1 -methylethyl (? ' ro-propyl), n-butyL n-pentyl, 1,1-dimethylethyl (/-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-l -enyl, but-l-enyl, pent-l-enyl, penta-L4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted, and an alkyl may optionally comprise one or more ether (-0-), thioether (-S-) or amine f-N<) bonds.

"Alkylate" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (/. e, , contains one or more double and/or triple bonds), and having from one to seventy carbon atoms (Ci-70-aikylene), e.g., methylene, ethylene, propylene, n-butylene, ethenylene, propenylene, «-butenyIene, propynylene, »-butynylene, and the like. The alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group ca be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the specification, an alkyl group may be optionally substituted, and an alkylene may optionally comprise one or more ether (-0-), thioether (-S-) or amine (-N<) bonds.

"Alkoxy" refers to a radical of the formula -OR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted.

"Alkylamino" refers to a radical of the formula -NHR a or -NR a R a where each R a is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted. "Alkylamiiiocarbonyl" refers to the -C(=0)NHR, or -C(=0) R a R a radical, where each R a is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, an alkylaminocarbonyl group may be optionally substituted.

"Alkoxyalkyl" refers to a radical of the formula -¾,OR a where R a is an alkyl radical as defined and where R ¾ is an alk lene radical as defined. Unless stated, otherwise specifically in the specification, an alkoxyalkyl group may be optionally substituted as described below.

"Alkylcarbonyl" refers to a radical of the formula -C( :;: 0)R a where R a is an alkyl radical as defined above. Unless stated otherwise specifically in the specification, an alkylcarbonyl group may be optionally substituted as described below.

"Alkoxycarbonyl" refers to a radical of the formula -C( ~ 0)OR a where R a is an alkyl radical as defined. Unless stated otherwise specifically in die specification, an alkyloxycarbonyl group may be optionally substituted as described below.

"Alkylcarbonyloxy" refers to a radical of the formula -OC( :::: 0)R a where a is an alkyl radical as defined above. Unless stated otherwise specifically in the specification, an alkyloxycarbonyl group may be optionally substituted as described below.

"CarhoxylalkyP refers to a radical of the formula ~-¾£(¼Η where R a is an alkyl radical as defined above. Unless stated otherwise specifically in the specification, carboxyaikyl group may be optionally substituted as described below.

"Thioaikyl" refers to a radical of the formula -SR a where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the specification, a thioaikyl group may be optionally substituted.

"Aryl" refers to a hydrocarbon ring system radical comprising hydrogen, 6 to 18 carbon atoms and at least one aromatic ring. For purposes of this invention, the aryl radical may be a monocyclic, bicyclie, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. Aryl radicals include, but are not limited to, aryl radicals derived from aceanthrylene, aeenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, iw-indacene 5 Λ'-indacene, indane, indene, naphthalene, phenalene, phenanfhrene, pleiadene, pyrene, and triphenylene. Unless stated otherwise specifically in the specification, the term. "aryl" or the prefix "ar-" (such as in "araUcy!") is meant to include aryl radicals that are optionally substituted.

"Aralkyl" refers to a radical of the formula -Rb~R c where ¾ is an alkyiene chain as defined above and R c is one or more aryl radicals as defined above, for example, benzyl, diphenylmethyl and the like. Unless stated, otherwise specifically in the specification, an araikyl group may be optionally substituted.

"Cycioalkyl" or "carbocyclic ring" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond, A "C3-7- cyeloalkyl referes to a cycioalkyl having from 3 to 7 carbon atoms in the cycioalkyl ring. Monocyclic radicals include, for example, cyciopropyl, cyclobutyl, cyc!opentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic radicals include, for example, adam.ant.yl, norbornyL decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, a cycloalkyi group may be optionally substituted,

"Cycloalkylalkyl" refers to a radical of the formula -¾,Rd where Rt, is an afkylene chain as defined above and ¾ is a cycioalkyl radical as defined above. Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted.

"Fused" refers to any ring structure described herein which is (used to an existing ring structure in the compounds of the invention. When the fused ring is a heterocyclvl ring or a heteroaryl ring, any carbon atom, on the existing ring structure which becomes part of the fused hete.roeyclyi ring or the fused heteroaryl ring may be replaced with a nitrogen atom. ' " Halo" or "halogen" refers to bromo, chloro, fluoro or iodo.

"Haloaikyl" refers to an alkyi radical, as defined above, that is substituted by one or more halo radicals, as defined above, e.g., trill uoromethyi, difiiioromethyl. trichloromethy 1 , 2,2,2-trifluoroethyl, 1 ,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1 ,2-dibromoethyl, and the like. A "C3..7 -haloaikyl refers to a haloaikyl having from 3 to 7 carbon atoms. Unless stated otherwise specifically in the specification, a haloaikyl group may be optionally substituted.

"Heterocyciyi" or "heterocyclic ring" or "heterocycle" refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyciyi radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heterocyciyi radical may be optionally oxidized; the nitrogen atom may be optionally quatemized; and the heterocyciyi radical may be partially or fully saturated. Examples of such heterocyciyi radicals include, but are not limited to, dioxoianyi, tliienyl[l ,3]dithianyl, decahydroisoquinolyl, imidazolinyl, irnidazoiidinyl, isothiazolidinyi, isoxazolidmyl. morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyL 2-oxopyrroIidinyl, oxazoiidinyi, piperidinyl, piperazinyL 4-piperidonyl, pyrrolidinyl, pyrazolidinyi, quinuclidinyl, thiazolidinyi, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiarnorpholinyl, 1-oxc-thiomorpholinyl, and 1.1-dioxo-tliiomorpholmyl. Unless stated otherwise specifically in the specification, a heterocyciyi group may be optionally substituted.

"iV-heterocyclyl" refers to a heterocyciyi radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyciyi radical to the rest of the molecule is through a nitrogen atom in the heterocyciyi radical. Unless stated otherwise specifically in the specification, a /V-heterocyclyl. group may be optionally substituted.

"Heterocyclylalkyl" refers to a radical of the formula -¾ e where ¾ is an alkylene chain as defined above and R e is a heterocyciyi. radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl. the heterocyclyl may be attached to the alkyl radical at the nitrogen atom, Unless stated otherwise specifically in the specification, a heterocyclylalkyl group may he optionally substituted.

"Heteroaryl" refers to a 5- to 14-membered ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ring. For purposes of this invention, the heteroaryl radical may be a monocyclic, bicyclic. tricyclic or tetracyclic ring system, which may include fused or bridged ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized; the nitrogen atom may be optionally quaternized. Examples include, but are not limited to, azepinyl, aeridinyl, benzimidazolyl. benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzotliia.diazoi.yL benzo [b] [ 1 ,4] dioxepiny 1 , 1 ,4-benzodioxanyl, benzonaphthof uranyl, benzoxazoly], benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl. (benzothiophenyl), benzotriazolyl, benzo[4,6]imidazo[l ,2-a]pyridinyl, carbazol l, cinnolinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyL isothiazolyl, imidazolyl. indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyL isoquinolyl, indolizinyl, isoxazolyl, napbthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyi, 1-oxidopyrimidinyl, 1-oxidopyrazinyl, 1 -oxidopyridazinyl, 1 -phenyl- lH-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyL purinyl, pyrrolyl, pyrazolyl, pyridinyl. pyrazinyl, pyrimidinyi, pyridazinyl, quinazolinyl, quinoxalinyi, quinolinyl, quinuclidinyl, isoqumolinyl, tetrahydroquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyL triazinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a. heteroaryl group may be optionally substituted.

"N-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the specification, an N-heteroaryl group may be optionally substituted. "Heteroarylaikyl" refers to a radical of the formula -¾¾ where ¾ is an alkyl en e chain as defined above and R f is a beteroaryl radical as defined above. Unless stated otherwise specifically in the specification, a heteroarylaikyl group may be optionally substituted.

The term "substituted" used herein means any of the above groups (e.g., alkyl, alkyiene, alkoxy, alkylamino, alkylaminocarbonyl, alkoxyalky!, alkylcarbonyl, alkoxycarbonyl, alkylcarbonyloxy, carboxylalkyl. thioalkyl, aryl, aralkyl, cycloalkyi, cycloalkylaikyl, haioalkyl, heterocyclyl, Λ· -heterocyclyl, heterocyclylalkyi, heteroaryi, N-heteroaryl and/or heteroarylaikyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, CI, Br, and I; an oxygen atom in groups such as hydroxy! groups, carboxyl groups, guanidine groups, imidine groups, phosphate groups, phosphmate groups, phosphonate groups, sulfate groups, sulfinate groups,alkox.y groups, ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamin.es, dialkylamines, arylamines, alkylaryS amines, diarylaniines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, diaikyiarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imin.es, oxim.es, hydrazines, and nitriies. For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced with. -NR g R h , -NR g C(=0)R h , -NR g C(=0)NR g R. h , - R g C(=0)OR h , -NR g S0 2 R h , -OC(=0)NR g R h. , -OR g , -SR g , -SOR g , -S0 2 R g , -OS0 2 R g , -S0 2 OR g , =NS0 2 R g5 and -S0 2 NR g R h . "Substituted also means any of the above groups In which one or more hydrogen atoms are replaced with -C(0)R g , -C(=0)OR g , -C(-0)NR £ R h , -CH 2 S0 2 R ¾ , -CH 2 SO 2 NR. g R. h - In the foregoing, R g and R« are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyi, cycloalkylaikyl, haioalkyl, heterocyclyl, N-heterocyclyl, heterocyclylalkyi, heteroaryi,

1 ■* > N-heteroaryi and/or heieroarylalkyl. "Substituted" further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, aikoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyi, N-heteroeyclyl, heterocyclylaiky!, heteroarvl, N-heteroaryl and/or heieroarylalkyl group. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.

Prodrugs of compounds of structure (I) are included in the scope of the invention. "Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject, in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. The prodrug compound often offers advaiitages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgaard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam)}, A discussion of prodrugs is provided in Higuchi, T., et al, Pro-drugs as Novel Drag Delivary Systems, A.C.S. Symposium Series, Vol. 14, 1975, and in Bioreversible Carriers in Drug Design, Ed, Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.

The term "prodrug" is also meant to include any covalently bonded earners, which release die active compound of the invention in vivo when such prodrug is administered to a mammalian subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mamrnaiian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol, or amide derivatives of amine functional groups in the compounds of the invention and the like.

The invention disclosed herein is also meant to encompass all pharmaceutically acceptable compounds of structure (I) being isotopically-labeied by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, °C, i C, ¾, 15 N, I5 0, 17 0, l8 0, 3i P, 32 P, 35 S, 18 F, j6 Cl, l and 5 I, respectively, ' t hese radiolabeled compounds could be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labelled compounds of stmcture (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. "' H, and carbon-14, i.e. ,4 C, are particularly useful for this purpose in view of their ease of incorporation and ready m eans of detection ,

Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.

Substitution with positron emitting isotopes, such as C, 18 F, i3 0 and 1J N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopicaily-iabeled compounds of structure (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopicaily-iabeled reagent in place of the non-labeled reagent previously employed.

The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radioiabelied compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.

"Stable compound" and "stable structure" are meant to indicate a compound that, is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.

"Manunal" includes humans and both domestic animals uch as laboratory animals and household pets (e.g. , cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.

' ptional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.

"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which, has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.

The present invention includes pharmaceutically acceptable salts of compounds of structure (I). "Pharmaceutically acceptable salt" includes both acid and base addition salts.

1.5 "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acicl, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-aeetamidobenzoie acid, camphoric acid, camphor- 10-sul fonic acid, capric acid, caproic acid, eaprylic acid, carbonic acid, cinnaniic acid, citric acid, cyciamic acid, dodecylsulfuric acid, ethane- 1 ,2-disuSfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glyceric acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, laurie acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-l,5-disulfonic acid, naphthalene-2-sulfoiiic acid. 1 -hydrox -2-nap h.oic acid, nicotinic acid, oleic acicl, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like.

"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamtne, diethanol amine, ethanolaraine, deanol, -dimethylaminoethanol,

2-diet ylaxninoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine. piperidine, W-emylpiperidine, polya ine resins and the like. Particularly preferred organic bases are isopropylamine, dietliylamine, ethanoiamirje, trimethylamine, dicyclohexylamine, choline and caffeine.

Often crystallizations produce a solvate of the compound of the invention. As used herein, the term "solvate" refers to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which, case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a monohydrale, dihydrate, liemihydrate, sesquihydrate, trihydrate. tetrahydrate and the like, as well as the corresponding solvated forms. The compomid of the invention may be true solvates, while in. other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.

A "co-crystal" of a compound of the invention can also be formed. Co- crystallization can alter the molecular interactions and composition of pharmaceutical materials, and provide unique drug properties. Co-crystals consist of a compound of the invention and a typically stoichiometric amount of a pharmaceutically acceptable co- crystal former. Pharmaceutical co-crystals are nonionic supramolecular complexes and can be used to address physical property issues such as solubility, stability and bioavailability in pharmaceutical development, w thout changing the chemical composition of the compound of the invention,

A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor. "Effective amount" or 'therapeutically effective amount" refers to that amount of a compound of the invention which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of agonizing TGR5 in the mammal, preferably a human. The amount of a compound of the invention which constitutes a "therapeutically effective amount' ' ' will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.

"Treating" or "treatment" as used herein covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes:

(i) preventing the disease or condition from occurring in a mammal, in particular, when such mammal is predisposed to the condition but Iras not yet been diagnosed, as having it;

(ii) inhibiting the disease or condition, i.e., arresting its development;

(iii) relieving the disease or condition, i.e., causing regression of the disease or condition; or

(iv) relieving the symptoms resulting from the disease or condition, i.e., relieving pain without addressing the underlying disease or condition. As used herein, the terms "disease" and "condition" may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.

The compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enanUomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (J?)~ or (SV or, as (D)~ or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)~ and (5)~, or (D and (L)~ isomers .may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivati ve) using, for example, chiral high pressure liquid chromatography (HPI.C). When the compounds described herein contain olefinic double bonds or other centres of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric !onns are also intended to be included.

A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.

A "tautome 5 refers to a proton shift from one atom of a molecule to another atom of the same molecule. The present invention includes tautomers of any said compounds.

The chemical naming protocol and structure diagrams used herein are a modified form of the I.U.P.A.C, nomenclature system, using the 'TUPAC Naming Plugin" software program (ChemAxon) and/or ChemDraw software Slruct=Name Pro 11.0 program (CambridgeSoft). For complex chemical names employed herein, a substituent group is named before the group to which it attaches. For example, cyclopropylethyl comprises an ethyl backbone with cyclopropyl substituent.

IL Compounds

As noted above, in one embodiment of the present invention, compounds having activity as TGR5 agonists are provided, the compounds having the following structure (I):

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

X is CR 50 R 51 wherein:

R 50 and R 51 are the same or different and independently selected from H and

Ci-7-alkyL or

R 50 and R 51 taken together with the C atom to which they are attached form a cycloalkyl or heterocyclyl, wherein the cycloalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C t -7-aUcyi, C1.7-haloalk.yh Ci-7-alkylcarbonyl, Ci-7-alkyloxycarbonyl,

Ci-7-alkoxy, C 1 .7-alkoxyalk.yl, (R a )2(R ¾ )N- and Ci-7-alkyl-S(0)o-2-, wherein each R a is independently, at each occurrence, hydrogen or Ci-7-alkyl and R° is an electron pair, hydrogen or C 1-7 -alkyl;

Y is CR 60 R'' 1 , O. NR. 62 or a direct bond, provided that when Y is O, Z is not O or S(0)o-2, wherein:

R 60 and O are the same or different and independently selected from H and Ci -7-alkyl; and

R 6* '' is selected from H, Ci-7-aikyl, Ci_ 7 -alkylcarbonyl, aminocarbonyl, Cj .7-alkylaminocarbonyI, Ci-7-alkyisulfone. cycloalkyl alkyl, cycloalkyl, aralkyl and aryl, wherein the Ci.. 7 -alkyl, Ct-7-aikylcarbonyl, aminocarbonyl,

Ci-7-alkylamirj.ocarbonyl, Ci. 7 -alkylsulfone, eycloalkyialkyl, cycloalkyl, aralkyl and aryl are optionally substituted with one or more substitutents selected from halogen, hydroxy, oxo, Ci-7-alkyl, Ci^-haioalkyl, C 1 .7- alkylcarbonyl. Ci -7 -aikyloxycarbonyl, Ci-7-alkoxy, d-7-alkoxyalkyl, (R a ) 2 (R b )N- and d.7-alky}-S(0)o-:r, wherein each R a is independently, at each occurrence, hydrogen or Ci-7-alkyl and R is an electron pair, hydrogen or Ci-7-alkyl;

or X and Y taken together form a. cycloalkyl, heterocyclyl, aryl or heteroarvl, wherein the cycloalkyl, heterocyclyl, aryl or heteroarvl are optionally substituted by one or two groups selected from halogen, hydroxy, 0x0, C1.7- alkyl, C 1.7-alkykarbonyl, Ci-7-alkyloxycarbonyl, Ci-7-alkoxy, C 1 .7-alkoxya.kyl, (R a )2(R°)N- and C^ -7-alkyi-S(0)o-2~, wherein R a is independently, at each occurrence, hydrogen or C|. 7 ~alkyl and R" is an electron pair, hydrogen or C h alky!, and provided that when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyrimidirsyl then Z is not O;

Z is CR'°R'\ O, S(0)o- 2 or a direct bond, wherein:

R u and R ,! are the same or different and independently selected from H or C h alky!;

or R ' " 3 and R' '3 taken together to form oxo (=0);

or Z and R 8 or R taken together form a cycloalkyl or heterocyclyl wherein the cycloalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C 1-7 -alkyl, C 1-7 -rialoalkyl, C 1-7 - alkylcarbonyl , Ci.7-alkyloxycarbonyl, Ci-ralkoxy, Ci-7-alkoxyalkyl, (R a ) 2 (R b )N- and Ci. 7 -alk.yl-S(0)o-r, wherein each R A is independently, at each occurrence, hydrogen or C 1 .7-alk.yl and R b is an electron pair, hydrogen or C.i.7~alkyl;

A/ is CR^ or N; '

A 2 is CR 14 or N, wherein:

R 13 and R' 4 are the same or different and independently selected from: hydrogen, C 1-7 -alkyl, halogen, C 1-7 -haloalkyl, cyano, Ci-j-alkoxy, amino and

-S(0)o.2-Ci.7-alkyl;

R 1 and R 2 are the same or different and independently selected from: hydrogen, d-7-alkyl, halogen, halogcn-Ci -7 -alk.y{, cyano and C1.7-alk.oxy;

R 3 is selected from: hydrogen, Q-7-aikyl, halogen, Ci-7-haioalkyl, Ci. 7 -alkoxy, cyano, C3.7-cycloal.kyl, -O-C3.7-cycloalk.yl, -O-C j .?-alky l-C 3 -7-cycloalkyl, -S(0)o-2-Ci-7-alkyl, N-heterocyclyl, five-membered heteroaryl, phenyl and -

NR i3 R 1(> , wherein R l3 and R'° are the same or different and independently selected from hydrogen, (. '' ·,. --a iky! and C3 -7 -cycloalkyl;

R 4 is selected from: hydrogen, Cj.y-alkyl, halogen-C j . 7 -alkyl, and C3- 7 -cycloalkyl; or R 3 and R 4 or R 3 and R t4 together are ~L l -(CR ,? R l8 ) n - and form part of a ring, wherein:

V is selected from: -CR 19 R 20 -, O, S(O) 0 . 2 , CO and NR 21 ;

R. 17 and R 1 " S are the same or different and independently selected from hydrogen and Ci-7-aIkyl;

or R' ' and R 1S together with the C atom to which they are attached form an oxo moiety;

or R i ? or R 1S together with an adjacent R 17 , R ts , R i9 or R 20 and the C atoms to which they are attached form C--C;

R 19 and R 2u are the same or different and independently selected from.: hydrogen, hydroxy], N(R 2l )¾ Ci-7-alkyl, Ci-7-aJkoxycarbonyl, unsubstituted heterocyclyl, and heterocyclyl substituted by one or two groups selected from halogen, hydroxy and Cs. -alkyl,

or R 19 and R 0 together with the C atom to which they are attached form a cyclopropy] or oxetanyl ring or together form a =CH 2 or OF2 group; and

R 2t is independently, at each occurrence, selected from the group consisting of: hydrogen, CV?-alkyl, halogen-Cj-7-alkyl, and C3..7- cycloalkyl-C | -7-alkyl, wherein C 3- 7-eyeloalkyl is unsubstituted or substituted by carboxyl- Cj.7-alkyl or Ci-7-alkoxycarbonyl, heterocyclyl, heterocyclyl- Ci-7-alkyl, heteroaryl, heteroaryl-Ci-7-alkyl, earboxyl-Ci-7-alkyl, C1-7- alkoxycarbonyl-Ci-7-alkyl, Ci- 7 -aIkylcarbonyloxy-Ci.7-alkyi, C 1 -7- alkylsulfonyl, phenyl, wherein phenyl is unsubstituted or substituted by carboxyl-Ct.7-alkyl or C 1-7 -alkoxycarbonyl, phenylcarbonyl, wherein phenyl is unsubstituted or substituted by carboxyl-Ci. 7 -alkyl or Ci. 7 -alkoxycarbonyl, and phenylsulfony], wherein phenyl is unsubstituted or substituted by carboxyl-C j .7-alk l or Ci. 7 -alkoxycarbonyl; or R 2 i and a 17 together are -(C¾) 3 - and form part of a ring;

or ~ together with a pair of R' ' and R are -CH=CH-CH= and form part of a ring; and

n is 1 , 2 or 3;

R 9 , R lu , R 11 and R are the same or different and independently selected from: Q, hydrogen, C3.7-aIk.yl, C2-7-alkenyl, C 2 -7-alkyiiyl, halogen, halogen-Ci.7-alkyl, Ci-7-aikoxy, halogen-Ci-7-alkoxy, hydroxy, hydroxy-Ci- 7 -alkoxy, hydrox.y-Ci-7- alkyl, hydroxy-C3.7-alke1.yl, hydroxy-C3. 7 -alkynyl, cya.no, carboxyl, C1-7- alkoxycarbonyl, amino carbonyl, carboxyl-Ci-7-alkyL carboxyl- C^-alkenyl, carboxyl- C2.7-alkyn.yl, Ci -7 -a!koxycarbonyl-Ci-7-alkyL C i. 7 -alkoxycarbonyl-C2- 7-alkeny 1 , C t ^-alkoxycarlxmyl-C^-alkynyl, carboxyl-C ^-alkoxy, C i_ 7 - alkoxycarbonyl-Ci. 7 -alkoxy, catboxyl-C^-alkyl-aminocarbonyl, carboxyl-Ci. 7 - alkyl-(C t . 7 -alkylam o)-carbonyl ? C 1.7-alkoxycarbonyl-C 1.7-alkyl - aminocarbonyl, C 1 .7-alkoxyc r onyl-C 7-aίk l-(C 1 .7-alk lamino)-carbonyl > carboxyl-Ci.7-alkyl-animocarbonyl-Ci. 7 -alkyl, carboxyi-C3-7-alkyl-(Ci. 7 - aikylamino)-carbonyl-C 1 -7-alkyl, Ci. 7 -alkoxycarbonyl-Ci-7-alkyl- ammocaxbonyI--C1.7-a.lky]., C 1- 7-alkoxycarbonyI-Ci.7-alkyi-((^. -7-alkylainino)- ' carbonyl-Ci-7-alkyl, hydrox>'-Ci- 7 -alkyl-aminocarbonyl s di-(hydroxy-Ci -7 - alkyl)ammocarbonyi, aminocarbonyl-C 1 .7-alkyl-a.mino carbonyl, hydroxysulfonyl-Ci-7-a3kyl-aminocarbonyl, hydToxysulfony]-C] .7-aIkyh(C i . 7 ~ alkyl-amino)~carbonyl, di-(Ci. 7 -alkoxycarbonyl-Ci-7-alkyl>- methylamitiocarbonyl, phenyl, wherein phenyl is unsubstituted or substituted by- one to three groups selected from halogen, C . i-7-alkoxy, carboxyl or Ci -7 - alkoxycarbonyl, phenyl-carbonyL wherein phenyl is unsubstituted or substituted, by one to three groups selected from halogen, C 1-7 -alkoxy, carboxyl or Q.?- alkoxycarbonyl, phenyl-aminocarboiiyi, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Q^-alkoxy, carboxyl or Ci.7-alkoxyearbonyl, phenyl-C 1-7 -alkyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from, halogen Cu 7 ~alkoxy, carboxyl or Ci-7-alkoxycarbonyl, phenyl -C 2 . 7 -alkynyl, wherein phenyl is unsubstituted or substitiited by one to three groups selected from halogen, carboxyl or Ci-7-alkoxycarbonyl, heteroaryi, wherein heteroaryi is unsubstituted or substituted by one to three , groups selected from halogen Ci- 7 -alkoxy s carboxyl or Ci-7-alkoxycarbonyl, heteroaryl-carbonyl, wherein heteroaryi is unsubstituted or substituted by one to three groups selected from halogen, C1.7-alk.oxy, carboxyl or Ci_ 7 -aikoxyearbonyi, heteroaryl-aminocarbonyl, wherein heteroaryi is unsubstituted or substituted by one to three groups selected from halogen, Cj-7-alkoxy, carboxyl or Ci. 7 -alkoxycarbonyl, heteroaryl-Ci _ 7 -alkyl, wherein heteroaryi is unsubstituted or substituted by one to three groups selected from halogen, C 1 . 7 -alk.yl, Ci-7-alkoxy, carboxyl or C 1.7-alkoxy carbonyl, heteroaryl- C [ .7-alkyI-aminocarbonyl, wherein heteroaryi is unsubstiiuted or substituted by one to three groups selected from halogen, C : -7 -aIkoxy, carboxyl. or C1-7- alkoxycarbonyl, heteroaryl-carbonyl-Ci. 7 -alkyl, wherein heteroaryi is unsubstituted or substituted by one to three groups selected from halogen, C1.7- alkoxy, carboxyl or Ci-7-alkoxyearbotryl, and cycloalkyl, wherein cycloaikyl is unsubstituted. or substituted by one to three groups selected from halogen, C1-7- alkoxy. carboxyl or C1.7-alkoxycarbon.yi;

wherein:

L 2 and each L 3 are either the same or different and independently absent, -0-, -NR. 80 -, -S-, -NR 80 C(=O)-, -C(=0)NR 80 -, -NR 80 C( ))NR 8C -, -SO 2 NR S0 ~, - NR S0 SO : ; -Ci. 7 alkylene-, -C 5 -7 a]kyIene-G-, -0-Ci. 7 alkylene-, -C 1-7 alkyiene-NR 80 -, -NR 80 -Ci.7alkylene-, -Ci. 7 alkylene-S-, -S-C; -7 aikylene-, -Ci. 7 alkylene-NR 80 C(=O)-, -C(-O)NR 80 Cj. 7 alkylene-, -Ci. 7 alkylene-C(=0)NR 80 -, -NR so C(-0)Ci_ 7 alkylene-, -C }-7 alkylene-NR 80 C(=O)NR 80 -, -NR s<, C(-O)NR 80 C,.7alkylene- 3 -C, -7 a.lkyiene-SO 2 NR S0 -, -SO 2 NR 80 C 1 .. 7 alkylcne-, -SO 2 NR 80 C(-O)-, -C(=O)NR 80 SO 2 -, - NR 80 SO 2 R s0 C(-O)NR 80 -, -NR 80 C(-O)NR 80 S() 2 NR 80 ! -OC(-0)NR 80 -,

~NR 80 C(-O)O-; -Ci. 7 alkylene-OC(-0)NR 80 -, 1 7 alkylene- or -Ci - 7 alkylene-NR ao S0 2 -;

B is optionally substituted C ! -7 oalkyi or C 1-7 oalkylene, wherein the Ci-7oaikyl or Ci-7oalkylene is optionally substituted with one or more functional groups selected from hydroxy!, oxo, carboxy, gua idino, arnidino, -N(R 80 ) 2 , -N(R 8 ) 3 , phosphate, phosphonate, phospinate, sulfate, sulfonate and sulfonate, mid wherein the Ci-7oalkyl or Ci.. 7 oalkylene optionally comprises one or more moieties selected from -NR 8J -, -S-; -0-, -C3 -7 cycloalkyl-, and -SO2-;

I is a compound of structure (I);

R so is independently, at each occurrence, hydrogen, C 1-7 alkyi or -B-(L 3 -

I) m ; and

m is an integer ranging from 0 to 10.

In some other embodiments of the foregoing compound, R 3 is selected from: hydrogen. Cj . .7-alkyl, halogen, C1.7-haloalk.yl, Ci-7-alkoxy, cyano, Qvrcycloalkyl, -O-C3.7-cycloa.kyl, -S(0)o- 2 -Ci.7-alkyl, N-heierocyclyl, five-raembered heteroaryl, phenyl and -NR 1 s R i6 , wherein R 15 and R ! & are the same or different and independently selected from hydrogen, Ci_ 7 -alkyl and C3. 7 -cycloalkyl;

In some other embodiments of the foregoing compound, R 8 , R 9 , R'°, R a and R 12 are the same or different and independently selected from: Q, hydrogen, C1-7- alkyl, C 2 .7-alk.enyl, C2- 7 ~alkynyl, halogen, halogen-Ci-7-alkyl, Ci- 7 -alkoxy, halogen-Ci.7- alkoxy, hydroxy, hydroxy .7-aik.oxy, hydroxy-Ci- 7 -alkyl, hydroxy-C3.7-alke1.yl, hydroxy-C3.7-alkyn.yl, cyano, earboxyi, C1.7-alkoxycarbor1.yl, amino carbonyi, carboxyl- Ci-7-aikyl, earboxyi- C 2 -7-a.lke.ny!, earboxyi- C 2 -7-alkynyi, C1.7-alkoxycarbonyl-C1.7- alkyl, C1.7-aikoxycarbonyl-C2.7-alke.nyl, Ci.7-alkoxycarbony -C2- 7 -aikyny3 , carboxyl- C i .7-alkoxy, C l- 7-aIkoxycarbonyl-Ci. 7 -alkoxy ; ca.rboxyl-C>.7-alkyl-aminocarbonyl, carboxyl-C 1 -7-aikyl-(C^.7-alkyIamino)-carbonyl,Cj . . 7 -alk^

aminocarbonyl, Ci.7-alkoxycarbonyl-C^.7-alkyl-(Ci. 7 -alkylarrtino)-carbonyl, carboxyl- C } .7-alkyl-aminocarbonyl-Ci .7-alkyl, carboxyl-Ci. 7 -alkyl-(Ci. 7 -all^larrdno)-carbonyl- C 1.7-alkyl, C1.7- a3koxycarbonyl-Ci_7-alkyl-(Ci-7-alk}darnino)-carbonyl"Ci-7-a lkyL hydroxy-C .. 7 -alky]- aminocarbonyl, di-(¾ydroxy-Ci.7-alkyi)ammocarbonyl, aminocarbonyl-Ci- 7 -alkyl- amino carbonyl, hydroxy sulfonyl-C 1.7-alkyl-aminocarbonyl, bydroxysulfonyl-C 1.7- alkyl-(Cj. 7 -alkyl -amino )-carbonyl, di-(Ci.7-alkoxycarbonyl-Cj. 7 ~alkyl)~ methylarainocarbonyL phenyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C5.7~aj.koxy, carboxyl or C 1.--alkoxycarbonyl, phenyl-carbonyl, wherein phenyl is imsubstiiuted or substituted by one to three groups selected from halogen, C 1-7 -alkoxy, carboxyl or C1-7-a.lkoxycarbon.yl, phenyl- aminocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Cw-alkoxy, carboxyl or Ci_ 7 -alkoxycarbonyl. phenyl-Ci.--alkyi, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen Ci. 7 -alkoxy, carboxyl or C{. 7 -alkoxycarbonyl, phenyl -C2.7-alk.ynyi, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci-r-alkoxy, carboxyl or C 1.7 -alkoxycarbonyl, heteroaryl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen. Ci. 7 -alkoxy, carboxyl or Ci-7-alkoxycarboayl, heteroaryl-carbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci- 7 -alkoxy, carboxyl or Ct-7-alkoxycarbonyl, heteroaryl-aminocarbonyl. wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Cj. 7 -alkoxy, carboxyl or Ci-7-alkoxycarbonyl, heteroaryl-Ci-7-alkyl, wherein heieroaryl is unsubstituted or substituted by one to three groups selected from halogen, C| -7 -alkyL C j .7-alkoxy, carboxyl or C 1 ,. 7 -al koxyc arbonyl, hetero ary 1-C 1 -7 - alkyl-aniinocarbonyl, wherein, heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C1.7-alko.xy, carboxyl or Ci. 7 -alkoxycarbonyl, and heteroaryl-carbonyl-Ci. 7 - alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from, halogen, Ci- 7 ~alkoxy, carboxyl or Ci_ 7 -alkoxycarbonyl.

As one of skill in the art will appreciate, each of the substituents of compounds as described herein may also be optionally substituted with one or more of the substituents defined above and below. In some other embodiments of the foregoing compound, X is C. ' R ' ' C' R ' and the compound has the following structure (II):

In other embodiments, Y is O and Z is CR. ' " l 71 and the compound has the following structure III):

In yet other embodiments, Y is NR &2 and Z is CR 70 R 71 and the compound has the followin structure (IV):

In some other embodiments, Y is CR 0 R 61 and Z is O and the compound has the following structure (V):

?7

In even other embodiments, R 5 and IV' taken together with the C atom to which they are attached form a cycloalkvi or eterocyclyl, wherein the cycloalk i or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Cj-7-alkyl, Ci_ 7 -alkylcarbonyl, C 1 .7-alkyloxycarboD.yl, Ci-7-alkoxy,€\.η- alkoxyalkyl and C i- 7 -alkyl-S(0)o.2-, wherein the compound has the following structure (VI) and wherein W represents the cycloalkvi or heteroeye!y group:

In even other embodiments, Y is O and Z is CR /0 R' i and the compound has the following structure (VII):

Tn still other embodiments, Y is NR 62 and Z is CR /0 R 7i and the compound has the following structure (VIII):

In other embodiments, Y is CR b0 R 6i and Z is O and the compound has the following structure IX):

In still other embodiments, the compound has one of the following structures (Via), (VIb), (Vic), (Vld), (Vie), (Vi (VIg) or (Vlh):

(Vic)

lf)

(VIg) (VTh) wherein:

R° is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, C1.7-alk.yl, Ci -7 -alkylcarbonyl , Ci-7-alkyloxycarbonyl, C1-7- alkoxyalkyl or C^-atkyl-SCO^ ¾»d

R d is independently, at each occurrence, an electron pair, hydrogen, Cj-y-alk !, C 1-7 -alkylcarbonyl, Ci-7-alkyioxycarbonyl, or Ci.-7-alkyl-S(C))o-2-. For example, in some embodiments Y is C ) and Z is C ' '¾ ' . In other embodiments Y is NR 62 and Z is CR 70 R 7! , and in other embodiments Y is CR 60 R 61 and Z is 0.

in even other embodiments, X and Y taken together form a. cycloalkyi, heterocyclyl, aryl or heieroaryl, wherein the cycloalkyi, heterocyclyl, aryl or heieroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C1.7-alk.yl, C1.7-alkylcarbon.yl, C j -7-alkyloxycarbonyl, Cj-y-alkoxy, Ci-7-alkoxyalkyl, (R a ) 2 (R°)N- and Ci-7-aikyl-S(0)o-r, wherein R is independently, at each occurrence, hydrogen or Ci-7-alkyl and R b is an electron pair, hydrogen or Cj- ? ~alkyl, and provided that when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyrimidinyl then Z is not O, wherein the compound has the following structure (X), and. wherem V represents the cycloalkyi, heterocyclyl, aryl or heieroaryl:

In some embodiments of the compound of structure (X), Z is CR' R and the compound has the following structure (XI):

In other embodiments of the compound of structure (X), Z is CR' 0 R.' 1 and R 70 and R. 7i taken together form oxo (=0) and the compound has the following structure (XII): ϊη still other embodiments of the compound of structure (X), Z is 0 and the compound has the following structure (ΧΤΠ):

in even more embodiments of the compound of structur S(0)o-2 and the com ound has the following structure (XIV):

For example, in some embodiments of the compounds of structure Z is -SO?-.

In even more embodiments of the compound of structure (X), the compound has one of the following structures (Xa). (Xb), (Xe), (Xd), (Xe), (Xf), (Xg), (Xh), (Xi), (Xj), (Xk), (XI), (Xm), (Xn), (Xo), (Xp), (Xq), (Xr) or (Xs):

(Xh)

Xk) (XI)

(Xo) (Xp)

(Xq) (Xri

(Xs)

wherein:

R e is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo. Cj-7-alkyL Q-7-haloalkyl, Cj .7-alkylcarbonyL Ci .7-alky loxycarbonyl, Ci-7-alkoxy, Cw-aikoxyalkyl, (R a ) 2 (R b )N- and Ci- 7 -alkyl-S(0)o- 2 -; and

R 1 is an electron pair, hydrogen or Ci^-alkyl.

For example, in certain embodiments of the foregoing 7 is CR H ' , In other embodiments, Z is CR 0 R S and R /u and R" taken together form oxo (=0). In still other embodiments Z is O. In yet more embodiments Z is -S(0)o-2-, for example in some embodiments Z is ~80 2 ~.

In some other embodiments, Y is absent and Z is O and the compound has the following structure (XV):

In some embodiments of the compound of structure (XV), R J ° and R 51 taken together with the C atom to which they are attached form a cycloalkyl or heterocyclyl, wherein the cycloallcyi or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C^-alkyL Ci-7-alkylcarbonyl, C 1 .7- alkyloxycarbonyl, C^-alkoxy, Ci. 7 -alkoxyalkyl and Q -7-alkyI-S(0)o-2- > wherein the compound has the following structure (X ' Vi) and wherein W represents the cycloalkyl or heterocyclyl group:

In still other embodiments, Al and A2 are both CR For example, m some embodiments of the foregoing R.' 3 is hydrogen.

In other embodiments, R 3 and R 4 together are -L~(CR 17 R !8 ) n - and form part of a ring. For example, in some embodiments the compound has the following structure (XVII):

In some embodiments of the compound of structure (XVil), L is - C( ::; 0)~, -S-, -S(0) 2 - or -N(R )-. J- ' or example, in some embodiments R" is C3.7- cycloalkyl.

other embodiments of the compound of structure (XVII). the compound has one of the following structures (XVIIa). (XVIIb), (XVIIc) or (XV

(XVTia) (XVIIb)

(XVIIc) (XVIld)

In other embodiments of the compound of structure XVIia, X is CR 50 R , R 50 and R.' " 1 taken together with the C atom to which they are attached form a eycloalkyl or heterocyclyl, wherein the eycloalkyl or heterocyclyi are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C1.7- aikyicarbonyl, Ci-7-alkyIoxycarbonyi, Ci. 7 -alkoxy, C 1-7 -aikoxyalkyl and C1.7- alkyl-S(0)o-2-. In still other embodiments, R 50 and R 51 taken together with the C atom to which they are attached form a eycloalkyl according to structure (Via), and the compound of structure (XXVIla) has the following structure (XVIIa-1):

(XVIIa-1) wherein:

R c is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, Cj -aikyl, Ci-7-alkylcarbonyl, Ci-7-alkyloxycarbonyl, Cj.7~alkoxy, C 1 .7- alkoxyalkyi or Ci. 7 -alkyl-S(0)o-2-. In some other embodiments, R° is hydrogen.

For example, in certain embodiments of a compound, of formula (XVIIa- 1), Y is O and Z is CR 70 R 71 . In other embodiments, Y is O and Z is CR. 70 R 7i . In other embodiments, Y is NR 62 and Z is CR^'R 7 '. In other embodiments, Y is NR 0 and Z is O, In other embodiments, Y is NR 6i and Z is S(O) 0- 2. In other embodiments, Y is CR 60 R. 61 and Z is CR 7ft R 7i . Tn other embodiments, Y is CR 60 R 6i and Z is O. In other embodiments, Y is CR 60 R 61 and Z is S(O) 0 - 2 .

In other embodiments of the compound of structure XVIIa, X and Y taken together form a eycloalkyl, heterocyclyl, aryi or heteroaryl, wherein the eycloalkyl, heterocyclyl, aryl or heteroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci -7 -alkyI, Ci_ 7 -alkylcarbonyl, C 1-7 - alkyloxycarbonyl, Ci-7-alkoxy, Ci -7 -alkoxyalkyl, (R a ) (R b )N- and Ci. 7 -alkyi-S(0) 0-2-, wherein R d is independently, at eac occurrence, hydrogen or Ci-7-alkyl and R b is an electron pair, hydrogen or Ci-7-alkyl, and provided that when X and Y form phenyl, pyridyl, pyridyl-N -oxide or pyrimidinyl then Z is not O. In still other embodiments, X and Y taken together form a heterocvclyl according structure (Xb) or structure (Xg), and the compound of structure (XXVTIa) has the following structure (XVIIa-2) or

(XVIIa-2)

wherein:

R e is independently, at each occurrence, hydrogen, halogen, hydroxy, oxo, C 1 .7- alkyl, C 1 .7~haioa.lkyl, Ci.7-alkylcarbonyl, Ci- 7 -alkyloxycarbonyi, Ci_ 7 -alkoxy, C1.7- aikoxyalkyl, (R j 2 (R b )N~ and Ci. 7 -alkyl-S(0)o_ 2 -; and R* is an electron pair, hydrogen or Ci-7-alkyl.

For example, in certain embodiments of the foregoing Z is CR' R . In. other embodiments, Z is CR^R' 1 and R ?0 and. R 71 taken together form oxo (=0). In still other embodiments Z is (), in yet more embodiments Z is -S(0)o-2-, for example in some embodiments Z is -SO?.-.

In still other embodiinents, the compound has the following structure

(XVIII):

(Χνΐϊϊ)

In other embodiments of the compound of structure (XVIII), R 2 is

N(R " ) 2 . For example, in some embodiments the compound has one of the following structures iXYUkii. (XVIIIb), (XVIIIe), (XVIIId), (XVIIIe), (XVIIIf), (XVIIIg), (XVIIIh), (XVII!i . (XVlUj). (XVHIk) or (XVIIIl):

(XVIIIa) (XVIIIb)

(XVIIIe) and

(XVIIIe) (XVIIif)

(XVIllli)

(XVIIIi) (XVfflj)

(XVIIIk) (XV! ill) in still other embodiments, A* and A 2 are each independently CH or N and R 3 is C ]-7 -alkoxy, -O-C3.7-cycloaUi.yl, or -0-C 1-7 -alkyl-C 3 . 7 -cycloalkyl. For example, in some embodiments the compound has one of the following structures

(XIXc) (XiXd) U 2012/071251

(XLXg)

In other embodiments, the compound has the structure (XlXg).

In certain embodiments of the foregoing, X is CR 50 R 51 . In other embodiments of the compound of structure ( lXg). R 50 and R 51 taken together with the C atom to which they are attached form a cycloalkyl or heterocyclyl, wherein the cycloalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C ;-7 -alkyk Ci-7-alkylcarbonyl, d-7-alkoxy, C1.7- alkoxyalkyl and Ch-y-alkyl-SiOjo--?-, For example, in certain embodiments, R-' and ' taken together with the C atom to vvhich they are attached form a cyclopropyl.

In still other embodiments, Y is O and Z is CR 70 R 71 ; Y is NR 62 and Z is

62 , 60n 61

CR 70 R 71; Y is R E and Z is O; Y is R^ and Z is S(O) 0 - 2 ; Y is CR^R 01 and Z is

, 60T> 6I

CR 70 R 71 ; Y is CR W R C 1 and Z is O; and Y is CR W R 01 and Z is S(O) 0 . 2 .

In certain embodiments of the foregoing, X and Y taken together form a cycloalkyl, heterocyclyl, aryl or heteroaryt, wherein the cycloalkyl, heterocyclyl, aryl or heleroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci-7-alkyl, C^-alkylcarbonyl, Ci-7-aIkyloxycarbonyl, Ci-7-aikoxy, C1.7- alkoxyalkyl, (R a )2(R°)N- and Ci. 7 -alkyl-S(0)o-2-, wherein R. d is independently, at each occurrence, hydrogen or Cj . . 7 -alkyl and R b is an electron pair, hydrogen, or Ci-7-alkyl, and provided that when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyrimidinyl then Z is not O. For example, in certain embodiments, X and Y taken, together form a. heterocyclyl. in still other embodiments, the heterocyclyl is pyrrolidinyl or thiazolidinyl.

For example, in certain embodiments of the foregoing Z is CR'°R 7i . In other embodiments, Z is CR ?0 R' 1 and R'° and R 71 taken together form oxo (=0). In still other embodiments Z is O. In yet more embodiments Z is -S(0)o- 2 ~, for example in some embodiments Z is -SO2-.

In even more embodiments, A 1 is CR 13 and A 2 is CR 14 and wherein R u and R 1 are independently from each other selected from hydrogen, halogen, halogen- Ci-7-alkyl and Ci-7-alkoxy.

In other embodiments, A 1 is CR 13 and A 4 is N, with R li being independentl from each other selected from hydrogen, halogen, haiogen-Ci, 7 -alkyl and

In yet more embodiments, R l and R " are independently from each other selected from the group consisting of hydrogen, halogen and haiogen-Ci_ 7 -alkyl.

In other embodiments, R J and R 4 together are -L' -(CR I 7 R l8 ) n - and form part of a ring; wherein

L 1 is selected from -CR 59 R 20 - and -NR 21 -;

R l / and R 18 are independently from each other selected from hydrogen and C 1-7 -alkyl:

R 19 and R 2,J are independently from, each other selected from hydrogen, Ci -7 alkyl, Ci. 7 -alkoxycarbonyl, unsubstituted heterocyclyl and heterocyclyl substituted by one or two groups selected, from Ci-7-alkyl and halogen;

or R 1 and R 20 together with the C atom to which they are attached form a cyclopropyl or oxetanyl ring or together form a =C¾ or =CF 2 group; R 21 is selected, froni hydrogen, C 1-7 ~alkyl, Cj -7 ~ cycloalkyl and C3.7-cycloalkyl-Ci.7-alk.yl, wherein C's^-cyeloalkyl is urisubstituted or substituted by carhoxyl-Cj.y-alkyl or Cj-y-alkoxycarbonyl, heterocyclyl, heterocyclyl- Ci-7-alkyl, heteroaryL heteroaryl-Ci . 7 ~alkyl, carboxyl-Ci-7-alkyl, Ci.y-alkoxyearboi yl- C 1 .7-alkyl, C 1.7-alkylcarbonyloxy-C {.y-alkyl, Ci .7-alkylsulf nyl, phenyl, wherein phenyl is urisubstituted or substituted by carboxyl-Ci- 7 -alkyl or C ^-alkoxycarbonyl, phenylcarbonyl, wherein phenyl is urisubstituted or substituted by carboxyl-Ci. 7 -aIkyl or d-7-alkoxycarbonyl, and plienylsulfonyl. wherein phenyl is unsubstituted or substituted by carboxyl-Ci^-alkyl or Ci.7-alkoxycarbonyl ;

or R 2i and a R 17 together are -(CH 2 )3- and form part of a ring, or R 2i together with a pair of R 17 and R 18 are -CH=CH-CH= and form part of a ring; and

n is 1 , 2 or 3.

In still other embodiments, the compound has stmcture (I), wherein: V is -NR 21 -,

R 2i is selected from hydrogen. C 7 -alkyi, Cs-T-cycloalkyl and C3.7- cycloalkyl-Ci-7-alkyl, wherein C3_7-cycloalkyl is unsubstituted or substituted by carboxyl-Ci.7-alkyl or Ci-7-alkoxycarbonyl, and C^-alkylsulfonyl;

R 17 and R 18 are independently from each other selected from hydrogen and methyl; and

n is 2.

In still other embodiments, L 1 is -CH 2 -, R 1 '' and R 18 are independently from each other selected from hydrogen and methyl and n is 2.

In other embodiments. R 3 and R l4 together are - lA(CR"R J,8 )n~ and form part of a ring; wherein i, 1 is -NR 2i - or -0--, R 21 is selected from hydrogen, C1.7- alkyl and C 3 . 7 -cycloalkyl, R 17 and R 18 are independently from each other selected from hydrogen and methyl, and n is 2.

For example, in some embodiments, L 1 is -O- and the compound has the following structure (XV): 12 071251

(XV)

In certain embodiments of the foregoing structure (XV), R 1 '' and R 18 are hydrogen. In other embodiments, X is CR^ ' R 51 . In other embodiments of the compound of structure (XV), R 50 and R 51 taken together vvith the C atom to which they are attached form a cycloalkyl or heterocyclyl, wherein the cycloalkyl or heterocyclyl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, C1.7- alkyl, C ι-7-alkylcarbonyl, Ci.7-alkyloxycarbonyl, d^-alkoxy, C1.7-aikoxya.kyl and C1-7- alkyl-S(0)o-2-, For example, in certain embodiments, R 5 and R 5i taken together with the C atom to which they are attached form a c clopropyl.

In still other embodiments, Y is O and Z is CR 70 R 71 ; Y is NR 62 and Z is CR 7o R 7i; Y is NR 2 md∑ h Q. γ is R 62 md ? is $( Ο ) 0 . 2 ; Y is CR 6 °R 65 and Z is

CR 70 R 71 Y is CR 60 R 63 and Z is O; or Y is CR 6n R 61 and Z is S(O) 0 . 2 .

In certain embodimen ts of the foregoing, X and Y taken together form a cycloalkyl heterocyclyl, aryi or heteroaiyl, wherein the cycloalkyl, heterocyclyl, aryi or heteroaryl are optionally substituted by one or two groups selected from halogen, hydroxy, oxo, Ci-7-alkyl, C 6-7 -alkyIcarbonyl, Ci-7-alkyloxycarbonyl, C { . 7 -alkoxy 5 C1.7- alkoxyalkyl, (R^CR^N- and Cj.7-a].kyl-S(0)o- 2 - 5 wherein R a is independently, at each occurrence, hydrogen or Ci- 7 -alkyl and R b is an electron pair, hydrogen or Cj.7-aik.yl, and provided that when X and Y form phenyl, pyridyl, pyridyl-N-oxide or pyriniidinyl then Z is not O. For example, in certain embodiments, X. and Y taken together form a heterocyclyl. In still other embodiments, the heterocyclyl is pyrrolidinyl or thiazoiidinyl. For example, in certain embodiments of the foregoing Z is CR'°R 7 i . In other embodiments. Z is CR R and R' and R' taken together form, oxo (=0). In still other embodiments Z is O, in yet more embodiments Z is -S((J)o.2-» for example in some embodiments Z is -S0 2 ~.

In other embodiments, R:' is selected from hydrogen, C i_ 7 -alkyl, C j. 7 - alkoxy, N-heterocycIyl and ~NR 15 R lb , wherein R 15 and R l6 are independently from each other selected from hydrogen, C 1.7-aikyl and C 3.7-cycloaikyl, and R 4 is hydrogen or methyl.

In other embodiments, at least one of R 8 , R , R u ' s R ! l or R 12 is halogen,

10 Cj-7-alkyl, halogen -C 1-7 -alkyl, Ci-7-alkoxy, halogen-Ci. 7 -alkoxy or cyano. For example, in some embodiments the halogen is chloro. In other embodiments, the other ones of R 8 , R 9 , R 10 , R n or R i2 are hydrogen.

In even more embodiments, the compound has one of the following structures (XXa), (XXb), (XXe), (XXd), (XXe), (XXf), (XXg), (XXh), (XXi), (XX i t> i XXk ) or XXI):

(XXa)

(XXe) (XXd)

(XXI) In still other embodiments, at least one of R 8 , R 9 , R 10 , R u or R 12 is Q. For example, in some embodiments R 9 or R 10 is Q. In other embodiments, the other ones of R 8 , R 9 , R 10 , R n or R 12 are selected from the group consisting of hydrogen, halogen, Ci-7-alkyl, Ci-7-alkoxy, halogen-Ci-7-alkoxy and cyano.

In other embodim.ents, the compound has one of the following structures (XXIa), (XXib), (XXIe), (XXId), (XXIe), (XXIf), (XXlg). (XXlh), (XXIi), (XXij), (XXIk) or (XXII):

XXIa) (XXIb)

(XXIe) (XXIf)

(XXIk) in even more embodiments, I is -0-, -Ci^alkylene-;

or

hi still other embodiments, Q is -L 2 CR 81 R 82 (CR s -'R S4 ) ml G, wherein:

R , R , R 5_i and are independently, at each occurrence, hydrogen or hydroxy!:

G is -CS E . ;. -Ci i Oi l. -CO :; ! I or ·! /··!: and

ml is an integer ranging from ί to 21 . In still other embodiments, G is -Cl¾. -C¾OH, or -CO 2 H.

For example, in some embodiments of the foregoing, for each occurrence of R 83 and R 84 , one of R Si or R 84 is hydrogen and the other of R 83 or R* 4 is hydroxy!.

n other embodiments, Q has one of tlae following structures (XXIIa), (XXIib), i X i !e). (XXIId), (XXIIe), (ΧΧΪΪί), (XXIIg), ( XX !I n ). (XXlli). (XXIIj

(XXl !k), {XXl li}. (XXIIm), (XXiln),

XXIIe) (XXIId) e)

(ΧΧΪΙί)

(XXIIg)

XXITh)

XXIIi)

(XXIII) (XXI Lrn)

(XXilo)

(XXIIp) wherein:

R s0 is hydrogen or Ci -7 alkyl; R s is idenpendently, at each occurrence, hydrogen or Ci-yalkyi;

R h is an electron pair, hydrogen or Cj. 7 alk.yi; and

xl, x2 and x3 are each independently an integer ranging from. 1 to 6.

in other embodiments of the foregoing, R 80 is 1 drogen or methyl, and in other embodiments l is 2 or 3.

In even more embodiments, Q is -I [(CH 2 )m2Qlny {CH 2 ) m2 R wherein ml is 2 or 3, m3 is an integer ranging from 1 to 21 and R 86 is hydrogen, hydroxy! or L 3 - 1.

In even more embodiment ' s, Q is -L 2 [(CH?.) m 20] m 3(CH2) m 2R !s6 J wherein m2 is 2 or 3, m3 is an integer ranging from 1 to 21 and R Sfi is hydrogen or hydroxyl.

In some other embodiments, Q has one of the following structures (XXIIIa), (XXIIIb) or (XXIIIc):

XIIIa)

(XXIIIb)

wherein I is a compound of structure (I) In some embodiments, B has the followin structure (XIV):

(XIV)

In certain embodiments, at least two of R s , R*, R 10 , R n and R 12 are selected from:

C i-7-alkyl, C 2-7-alkenyl, C 2 .7-alkin.yl, halogen, halogen-C^-alky!, C 1 -7- alkoxy, halogen-Ci. 7 -alkoxy, hydroxy, hydroxy-Ci -7 -aIkoxy, hydroxy-Ci-7-alkyi, cyano, carboxyl, Ci-7-alkoxycarbonyl, amino earbonyl, carboxyl-Ci .7-alkyi, carboxyl-C 2 - 7 -alkenyl, carboxyl-C2-7-alkynyl, C1-7- alkoxycarbonyl-Ci.?-alkyl, Ci_7-alkoxycarbonyl-C 2- 7~alkenyl, Ci.7-alkoxycarbonyl-C2.7- alkyrtyl, carboxyl-Ci-7-alkQxy, Ci-7-alkoxycarboayi-Ci. 7 -alkoxy, carboxyl-Cj .7-alkyl- ammocarbonyl, carboxyl-Ci.7-alkyl-(Ci.7-alkyiamino)-carbonyl, Ci-7-alkoxycarbonyl- Ci-7-alkyl-aminocarbonyl, Ci_ 7 -aikoxycarbonyl-C 1- 7-alkyl- (Q .7-aJkylammo)-carbonyl, carboxyl-Ci.7-alkyl-ammocarbonyl-Ci-7-alkyl, cai¾oxyl--C! -7-alkyl-(Ci.7-alkylamino)- carbonyi-C 1.7-alkyl, Ci^-alkoxycarbonyl-Ci^-alkyl-aminocarbonyl-Ci^-alkyl, C1.7- alkoxycarbonyl-C f ..7-alky]-(Ci-7-dkylamino)-catbony b.ydroxy-C).. 7 -alkyl- aminocarbonyl, di-(hydroxy-Ci- 7 -alkyl)amiaocarbonyl, aminocarbonyl-C 1.7-alkyl - amino earbonyl, hydro xysulfonyI-Ci -7 -alkyl-aminocarbor 1 yl, hydro xysulfo nyl-Ci-7- alkyl-(Ci. 7 -alkyl-amino)-carbonyl, di-iCi^-alkoxycarbonyl-Ci^-alkyl)- methylaminocarbonyl,

phenyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C 1-7 -alkoxy, carboxyl and C 1 .7-alkoxycarbon. l,

phenyl -earbonyl. wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and C 1-7 -alkoxycarbonyL phenyl-aminocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C^-alkoxy, carboxyl and C1.7- alko yearbonyl, phenyl-Ci-7-aB yl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Cj.7-al.k0xy. carboxyl and C^-alkoxycarbonyl, phenyl-C 2 -7-aIkynyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C^-alkoxy, carboxyl and C F -7 - alkoxycarbonyl,

heteroaryl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C 1-7 -alkoxy, carboxyl and C 1. 7 -alkGxycarbonyl, heteroaryl-carbonyi, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ct.7-alk.0xy, carboxyl and C1-7- alkoxy carbon yl .

heteroaryl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and Ci .7-alkoxycarbonyl,

heteroaryl-Ci-7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkyL Ci-7-aIkoxy, carboxyl and C1.7-alkoxycarbon.yl,

heteroaryl-Ci-T-aikyl-aininocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C-alkoxy, carboxyl and Ci-7-alkoxycarbonyl, and

heteroaryl-carbonyl-C 1 .7-a.lkyl, wherein heteroaryl is unsubstituted or substituted, by one to three groups selected from halogen, Ci. 7 -aikoxy, carboxyl and C 1.7-alkoxycarbonyl,

and the other ones of R s , R 9 , R 10 , R 1 1 and R 12 are hydrogen.

In even more embodiments, at least two of I s , R 9 , R. 10 , R i l and R 12 are selected from:

halogen, hydroxy, hydroxy-C3.7-al.koxy, hydroxy-Ci. 7 -alkyl, cyano, carboxyl, CJ . 7 -alkoxycarbonyl, amino carbonyL carboxyl-Ci-7-alkoxy, C1.7- alkoxycarbonyl-C 1 .7-alkoxy, carboxyl-C i. 7 ~alky S -aminocarbonyl, carboxyl-C s .7-aikyl- (C 1 ,7-aIky] amitio) -carbonyl , Ci. 7 -aIkoxycarbonyl -C 1. 7 -alkyl-aminocarbonyl, hydroxy- Ci .7-alkyl-aminocarbonyl, di-(hydroxy-Ci-7-alky])aminoearbonyl, arninocarbonyl-Ci-7- 1251

alkyl-amino carbonyl, hydroxysulfonyl-C1.7-aikyl-aminocarbon.yl, hydroxysulfonyl- Ci^-alkyl-iCi^-alJ yl-amino^carbonyl, di-(Ci-7-alkoxycarbonyl-Ci-7-alkyl)- methylaminocarbonyl,

phenyl-aminocarbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Ci_7-alkoxy, carboxyl and C1.7- alkoxycarbony ί ,

heteroaryl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci -7 -alkoxy, carboxyl and -7-alkoxycarbonyl,

heteroar>l-Ci.7-alkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkyl, Ci„7-alkoxy, carboxyl and

C ] .7-alkoxycarbony 1 ,

heteroaryl-Ci-7-alkyl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and C-alkoxycarbonyl, and

heteroaryl-carbonyl-Ci-7-aikyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected, from halogen, C 1 - 7 -alkoxy, carboxyl and C 1 -7-alkoxycarbonyl,

and the other ones of R 3 , R 9 , R 1 , R 5 5 and R 12 are hydrogen. In still other embodiments, at least one of R 8 , R 9 , R > , R u and R 3 "' is Q and at least one of R s , R 9 , R 10 , R. 11 and R u are selected from:

Ci-7-alkyl, C 2.7-alkenyl, C2- 7 -alkinyl, halogen, haJogeii-Ci -7 -aSkyl, Ci.. 7 - alkoxy, halogen-C j -7 -alkoxy, hydroxy, hydroxy-Ci- 7 -alkoxy, hydroxy-Ci-ralkyl, hydroxy-Cs-y-alkenyi, bydroxy-C^-alkynyl, cyano, carboxyl, C 1.7-alkoxycarbonyl, amino carbonyl, carboxyl -Ci -7 -alkyl, carboxyl-C2. 7 -alkenyl, carboxyl-0 ;-7 -alkynyi C1-7- alkoxycarbonyl-C 1.. 7 -alky ί , C j .7-alkoxycarbonyl-C 2 -7-alkenyl, C 1.7-alkoxycarbonyl-C 2 -7- alkynyl, earboxyl-C^-alkoxy, C1.7-aikoxycarbonyl-C 1 .7-alk.oxy, carboxyl-Ci. 7 -aIkyl- aminocarbonyl, carboxyl-Ci. 7 -alkyl-(Ci.7-aikylarnino)-carbonyl, Ci-7-alkoxycarbonyl- C i. 7 -alkyl-aminocarbonyi, C j .7-alkoxycarbonyl-C 1 .7-alkyl- (Cj . 7 -alkylamino)-carbonyi, carboxy ί -C i .7-alkyl-aminocarbonyl-C 1 -7-alkyl, carboxyl-C 1. 7 -alkyi-(C j .. 7 -alkylamino)~ carbony!-Ci_ -aikyl 5 Ci. 7 -aIkoxycarbonyl-Ci -7 -alkyl-ammocarbonyl-Ci-7-alkyl, Cj. 7 - alkoxycarbonyl-C 1-7 -alkyl-(C! -7 -alkylammo)-carbony3-Ci-7 hydroxy-Ci. 7 -alkyl- aminocarbony 1 , di-(hydroxy-C i . 7 -alkyl)aminocarbonyl , aminocarbonyl-Ci. 7 -alkyl- amino earbonyl, hydro xysuifonyl-Ci^-alkyl-amiriocarborryL hydro xysulfo nyl-Ci.7- alkyl-(Ci -7-alkyl-amino)-carbonyi, di -(C; .--alkoxyearbonyl-C j . 7 -a1kyl)- methylaminocarbonyl,

phenyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C 1-7 -alkoxy, carboxyl and Ci. 7 -aikoxycarbonyl,

phenyl-carbonyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, Cj-7-alkoxy, carboxyl and C 1-7 -alkoxycarbonyl, phenyl-aminocarboiiyl, wherein phenyl is un substituted or substituted by one to three groups selected from halogen, Ci. 7 -alkoxy, carboxyl and C1.7- alkoxycarbony!,

phenyl- Cj -7 -alkyl, wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C . i-7-alkoxy, carboxyl and Ci_ 7 -alkoxycarbonyl, ph.enyl-C 2 .7-alkynyl 9 wherein phenyl is unsubstituted or substituted by one to three groups selected from halogen, C; ..7~a1koxy, carboxyl and Ci- 7 - alkoxycarbonyL

heteroaryl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Q -7 -aIkoxy, carboxyl and Ci. 7 -alkoxycarbonyl, heteroaryl-carbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Cj. 7 -alkoxy, carboxyl and C1-7- alkoxycarbonyl,

heteroaryl -aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C f . 7 -alkoxy, carboxyl and C j . -7 -alkoxy earbonyl,

heteroaryl-Ci-7-aikyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen. C 1-7 -alkyl, carboxyl and Ci-7-alkoxycarbonyl, heteroaryl-C j .7-alkyi-aminQcarbonyl, wherein heteroary! is nsubstituted or substituted by one to three groups selected from halogen, C-alkoxy, carboxyl and Ci-7-alkoxycarbonyl, and

li teroaryi-carbonyl-C'i-7--alkyl. wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, C 1-7 -alkoxy, carboxyl and Ci-7-alkoxycarbonyl,

and the other ones of R 8 , R^, R 10 , R ! ' and R l are hydrogen. In still more embodiments, at least one of R 8 , R :'J , R 10 , R 1 ' and R 12 is Q and at least one of R 8 , R 9 , R 10 , R 11 and R ! " are selected from:

halogen, hydroxy, hydroxy-Ct^-alkoxy, hydroxy-d^-alkyl, cyano. carboxyl, _7-alkoxycatbonyl, amino carbonyl, carboxyl-Ci-7-aikoxy, C1.7- aikoxycarboayl-Ci .7-alkoxy, carboxyl~Ci.7-alkyI-aminocarboiiyl, earboxyl-Cj. 7 -alkyl- (C j - 7 -alkylami.no)-carbonyl, Ct-7-alkoxycarbonyl-Ci -7-alk 1-aminocarbonyl , hydroxy- C] ,7~alkyl~ammoearboiiyl, di-{liydr()xy-Ci -7 -alkyl)aminocarbonyl, aminocarbonyl-Ci .7- alkyi-amino carbonyl, hydroxysulfonyl-Ci-7-al!cyI-aminocarbonyl, hydroxysulfbnyl- Ci. 7 -alkyl-(Ci.7-alkyl-araino)-carbonyl, di-(Ci..7-a!koxycarbonyi-Ci. 7 -alkyl)- methylaminocarbonyl,

phenyl-aminoearbonyL wherein phenyl is unsubstituted or subsiituted by one to three groups selected from halogen. Cj.7-alkoxy, carboxyl and C1-7- alkoxycarbonyl,

heteroaryl-aminocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from, halogen, Ci-7-alkoxy, carboxyl and C i..7-aikoxycarbonyi,

heteroaryl-C_1.7-a.lkyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci_7-alkyl, d .7-alkoxy., carboxyl and C i .7-alkoxycarbonyl,

heteroaryl-C|. 7 -alkyl-a.minocarbonyl, wherein heteroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-aikoxy, carboxyl and C-alkoxycarbonyl, and 12 071251

lieteroaryl-carbonyl-Cj.j-alkyl, wherein heleroaryl is unsubstituted or substituted by one to three groups selected from halogen, Ci-7-alkoxy, carboxyl and Ci .7-alkoxycarbonyl,

and the other ones of R.\ R , R l0 , R l 1 and R 12 are hydrogen. in other embodiments, R 8 and R il are halogen and R y , R 10 and R 12 are hydrogen.

In certain embodiments, the compound is any one of Examples 1-291. in other embodiments, the disclosure provides a compound which is a TGR5 agonist, wherein the TGR.5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein administration of the TGR.5 agonist to the mammal does not induce filling of the gall bladder of the mammal as determined by ultrasound analysis.

In still other embodiments, the disclosure provides a compound which is TGR5 agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein administration of the TGR5 agonist to the mammal does not induce emptying of the gall bladder of the mammal as determined by ultrasound analysis.

In yet embodiments, the disclosure provides a compound which is TGR5 agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein administration of the TGR5 agonist, to the mammal does not cause a change in weight of the mammal's gall bladder by more than 400% when compared to administration of a placebo. The change in weight of the mammal's gall bladder can be determined, by any number of techniques known in the art. For example, in some embodiments change in weight, of the mammal.' s gall bladder is determined in a mouse model.

In other embodiments of the forgoing, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 300% when compared to administration of a placebo. In other embodiments of the forgoing, the TGR5 agonist administration does not cause a. change in weight of the mammal's gall bladder by more than 200% when, compared to administration, of a placebo. In. other embodiments of the forgoing, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 100% when compared to administration of a placebo. In other embodiments of the forgoing, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 50% when compared to administration of a placebo. In other embodiments of the forgoing, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 10% when compared to administration of a placebo.

In some other embodiments, the disclosure provides a compound which is a TGR5 agonist, wherein the TGR.5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist is administered to the mammal, the concentration of the TGR5 agonist in the gall bladder is less than about 100 μΜ. The amount of the TGR5 agonist in the mammal's gall bladder can be determined by any number of techniques known in the art. For example, in some embodiments the amount of the TGR5 agonist in the mammal's gall bladder is determined in a mouse model .

In still other embodiments, the TGR5 agonist concentration in the gall bladder is less than about 50 μΜ. In some other embodiments, the TGR5 agonist concentration in the gall bladder is less tha about 25 μΜ. In other embodiments, the TGR5 agonist concentration in the gall bladder is less than about 10 μΜ. In still other embodiments, the TGR5 agonist concentration in the gall bladder is less than about 5 μΜ. In still other embodiments, the TGR5 agonist concentration in the gall bladder is less than about 1 μΜ. In still other embodiments, the TGR5 agonist concentration in the gall bladder is less than about 0.1 μΜ.

In some embodiments, the compounds have systemic exposure levels below their TGR5 EC50, yet they are still able to elicit a significant increase in plasma GLP-1 levels. For example. In some embodiments the disclosure provides a TGR5 agonist, wherein the TGR5 agonist stimulates GLP-1 secretion in a mammal and is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist is administered to the mammal, the concentration of the TGR5 agonist in the mammal's plasma is less than the TGRS EC 50 of the TGRS agonist. For example in some embodiments, the TGRS agonist concentration in the marnmai's plasma is less than about 50 ng/mL. In some other embodiments the TGRS agonist concentration in the mammal's plasma is less than about 25 iig/rnL. in some other embodiments the TGRS agonist concentration in the mammal's plasma is less than about 10 ng/mL. In some other embodiments the TGRS agonist concentration in the mammal's plasma is less than about 5 ng/mL. In yet other embodiments the TGRS agonist, concentration in the marnmai's plasma is less than about 1 ng/mL.

In some other embodiments of any of the foregoing TGR5 agonists, the TGRS agonist is not systemically available. In other embodiments of any of the foregoing TGRS agonists, the TGRS agonist concentration in the mammal's plasma is less than the TGRS EC50 of the TGRS agonist, For example, in some embodiments the TGRS agonist concentration in the mammal's plasma is less than about 50 ng/mL. In some other embodiments the TGRS agonist concentration in the mammal's plasma is less than about 25 ng mL. In some other embodiments the TGRS agonist concentration in the mammal's plasma is less than abou 10 ng/mL. In some other embodiments the TGRS agonist concentration in the mammal's plasma is less than about 5 ng/mL, in yet other embodiments the TGRS agonist concentration in the mammal's plasma is less than about 1 ng/mL.

' In other embodiments of any of the forgoing TGRS agonists, the TGRS agonist does not modulate TGRS-mediated suppression of cytokines, in some other embodiments, the TGRS agonist does not modulate the ileal bile acid transporter (IBAT). In yet other embodiments, the TGR5 agonist does not modulate the Famesoid X Receptor (FXR),

In other embodiments of any of the foregoing TGR S agonists, the TGRS agonist stimulates PYY secretion.

In some embodiments of the foregoing, the TGRS agonist is a com pound of structure (I).

The compounds described herein are meant to include all racemic mixtures and all Individual cnantiomers or combinations thereof, whether or not they are specifically depicted herein. Further, the compounds are also i tended to include ail tautomeric forms, even if not specifically depicted. Tautomers are compounds which result from the formal migration of a hydrogen atom or proton, accompanied by a switch of a single bond and adjacent double bond.

Compounds as described herein may be in the tree form or in the form of a salt thereof, in some embodiments, compounds as described herein, may be in the form of a phannaceutically acceptable salt, which are known in the art (Berge el a!,, J. Pharm, Set 1977, 66. 1). Pharmaceutically acceptable salt as used herein includes, for example, salts that have the desired pharmacological activity of the parent compound (salts which retain the biological effectiveness and/or properties of the parent compound and which are not biologically and/or otherwise undesirable). Compounds as described herein having one or more functional groups capable of forming a salt may be, for example, formed as a pharmaceutically acceptable salt. Compounds containing one or more basic functional groups may be capable of forming a pharmaceutically acceptable salt with, for example, a- pharmaceutically acceptable organic or inorganic acid. Pharmaceutically acceptable salts may be derived from, for example, and without limitation, acetic acid, adipic acid, aiginic acid, aspartic acid, ascorbic acid, benzoic acid, benzenesuifonic acid, butyric acid, cinnamic acid, citric acid, camphoric acid, camphorsulfonic acid, cyclopentanepropionic acid, dicihylacetic acid, digluconie acid, dodecylsuifonic acid, ethanesulfonic acid, formic acid, fumaric acid, gluco eptanoic acid, gluconic acid, glycerophosphoric acid, glycolic acid., hemisulfonic acid, heptanoic acid, hexanoic acid, hydrochloric acid, hydrobromic acid, hydriodic acid, 2- hydroxyethanesulfonic acid, isonicotinic acid, lactic acid, malic acid, maleic acid, malonic acid, mandelic acid, methanes uifonic acid, 2-napthalenesulfonic acid, naphthalenedisulphonic acid, p-toluenesulfonic acid, nicotinic acid, nitric acid, oxalic acid, pamoic acid, pectinic acid, 3-ph.enylpropionic acid, phosphoric acid, picric acid, pimeiic acid, pivalic acid, propionic acid, pyruvic acid, salicylic acid, succinic acid, sulfuric acid, sulfamic acid, tartaric acid, thiocyanic acid or tmdecanoic acid. Compounds containing one or more acidic functional groups may be capable of forming pharmaceutically acceptable salts with a pharmaceutically acceptable base, for example, 2012/071251

and without limitation, inorganic bases based on alkaline metals or alkaline earth metals or organic bases such as primary amine compounds, secondary amine compounds, tertiary amine compounds, quaternary amine compounds, substituted amines, naturally occurring substituted amines, cyclic amines or basic ion-exchange resins. Pharmaceutically acceptable salts may be derived from, for example, and without limitation, a hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation such as ammonium, sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese or aluminum, ammonia, benzathine, meglumine, metbylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylaniine, tripropylamine, tributylamine, ethanoiamme, diethanolamine, 2- dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, ai-ginine, histidine, caffeine, hydrabamine, choline, betaine, ethyl enediamine, glucosamine, giucamine, methylg!ucamine, theobromine, purines, piperazine, piperidine, procaine, N- ethylpiperidine, theobromine, tetramethylammoniuni compounds, tetraethylammoni am compounds, pyridine, N,N--dimethyianiline, N-memyipiperidine, morplioline, N- methylmorpholine, -et ylmorpholine, dicyclohexylamine, dibenzylamine, N,N- dsbenzylphenethy [amine, 1 -ephenamine, Ν,Ν'-dibenzylethyIcnediamine or polyamine resins. In some embodiments, compounds as described herein may contain both acidic and basic groups and may be in the form of inner salts or zwitterions. for example, and without limitation, betaines. Salts as described herein may be prepared by conventional processes known io a person skilled in the art, for example, and without limitation, by- reacting the free form with an organic acid or inorganic acid or base, or by anion exchange or cation exchange from other salts. Those skilled in tire art will appreciate that preparation of salts may occur in situ during isolation and purification of the compounds or preparation of salts may occur by separately reacting an isolated and purified compound.

Furthermore, all compounds of the invention which exist in free base or acid form can be converted to their pharmaceutically acceptable sails by treatment with the appropriate inorganic or organic base or acid by methods known to one skilled in the art. Salts of the compounds of the invention can be converted to their free base or acid form by standard techniques. In some embodiments, compounds and all different forms thereof (e.g. free forms, salts, polymorphs, isomeric forms) as described herein may be in the solvent addition form, for example, solvates. Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent in physical association the compound or salt thereof. The solvent may be, for example, and without limitation, a pharmaceutically acceptable solvent. For example, hydrates are formed when the solvent is water or alcoholates are formed when the solvent is an alcohol.

In some embodiments, compounds and all different forms thereof (e.g. free forms, salts, solvates, isomeric forms) as described herein may include crystalline and amorphous forms, for example, polymorphs, pseudopolymorp s, conformational polymorphs, amorphous forms, or a combination thereof. Polymorphs include different, crystal packing arrangements of the same elemental composition of a compound.. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability and/or solubility. Those skilled in the axt will appreciate that various factors including recrystallization solvent, rate of crystallization and storage temperature may cause a single crystal form to dominate.

In some embodiments, compounds and all different forms thereof (e.g. free forms, salts, solvates, polymorphs) as described herein include isomers such as geometrical isomers, optical isomers based on asymmetric carbon, stereoisomers, tautomers, individual enantiomers, individual diastereomers, racemates, diastereomeric mixtures and combinations thereof, and are not limited by the description of the formula illustrated for the sake of convenience.

In some embodiments, pharmaceutical compositions in accordance with this invention may comprise a salt of such a compound, preferably a pharmaceutically or physiologically acceptable salt. Pharmaceutical preparations will typically comprise one or more carriers, excipients or diluents acceptable for the mode of administration of the preparation, be it by injection, inhalation, topical administration, lavage, or other modes suitable for the selected treatment. Suitable carriers, excipients or diluents are those known in the art for use in such modes of administration. Pharmaceutical compositions are described in more detail below. It is understood that any embodiment of the compounds of structure (I), as set forth above, and any specific substituent set forth herein for a R 1 , R 2 , R 3 , R 4 , R 8 , R 9 . R 1G , R 11 , R !i , A 1 , A 2 , X, Y and Z group in the compounds of structure (I), as set forth above, may be independently combined with other embodiments and/or substituents of compounds of structure (1) to form embodiments of the inventions not specifically set forth above. In addition, in the event that a list of substitutents is listed for any particular R group in a particular embodiment and/or claim, it is understood that each individual substituent may be deleted from the particular embodment and/or claim and that the remaining list of substituents will be considered to be within the scope of the invention. It is understood that in the present description, combinations of substituents and/or variables of the depicted formulae are permissible only if such contributions result in stable compounds.

The present disclosure also provides a pharmaceutical composition, comprising an one or more of the compounds disclosed herein and a pharmaceutically acceptable carrier as described below.

I I. Preparation of Compounds

Compounds for use in the present invention ma be obtained from commercial sources, prepared synthetically, obiained from naturally occurring sources or combinations thereof. Methods of preparing or synthesizing compounds of the present invention will be understood by a person of skill in the art having reference to known chemical synthesis principles.

The following Reaction Schemes I-IV illustrate methods for making compounds of this invention, ie., compounds of structure (I):

(I)

or a stereoisomer, tautomer, pharmaceutically acceptable salt or prodrug thereof. wherein R 1 , R 2 , R 3 , R 4 , R 8 , R 9 , R i0 , R n , R 12 , A 1 , A 2 , X, Y and Z are as defined above. It is understood that one skilled in the ait may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other compounds of structure (I) not specifically illustrated below by using the appropriate stalling components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example. Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th edition (Wiley, December 2000)) or prepared, as described in this invention.

General Reaction Scheme 1

Referring to General Reaction Scheme I, an appropriate aromatic amine of structure ia can be purchased or prepared according to methods known in the art and combined with an optional carboxyl activation reagent and/or acylation catalyst and a compound of structure lb containing either a protected or tree nucleophile (e.g., Y), to form compounds of structure ic. Ic may then be reacted with either compounds of structure Id (LG is an appropriate leaving group) or le to form various compounds of structure (I). One skilled in the art will recognize that the methods may optionally include deprotection of PG and use of a hydride reducing agent where ie comprises an arylaldehyde or arylkeione.

General. Reaction Scheme II

Alternatively, compounds of structure (!) may be prepared according to General Reaction Scheme li. wherein la is an appropriate aromatic amine and Ila is a carboxylate containing an electro hy!ic center Y, LG is a leaving group and Z is a nucleophile. Reaction of la with Ila may be performed in the presence of a carboxylate activation reagent, a base and an optional acyiation catalyst. Hb can then be combined with a compound of structure lie in the presence of an appropriate base to form various compounds of structure (I).

General Reaction Scheme III

In another embodiment, compounds of structure (I) are prepared according to General Reaction Scheme IK, wherein la is an appropriate aromatic amine, LG is a leaving group and X is a nucleophile. Reaction of ia with a phosgene equivalent (LG-CO-LG, wherein LG is a leaving group) and an appropriate base results in compounds of structure Ilia, ilia is then treated with an appropriate base, and an optional acylation catalyst to produce various compounds of structure (I).

General Reaction Scheme IV

Other various compounds of structure (I) may be preared according to General Reaction Scheme IV, wherein la is an appropriate aromatic amine and Ila is a carboxylate containing aryl group with appropriate linking elements X,Y, 7. Reaction of la and Ila in the presence of an appropriate carboxy late activation reagent, a base and an optional acylation catalyst results in various compounds of structure (I).

With regard to General Reaction Schemes I-IV, typical carboxylate activation reagents include DCC, EDO, HATU, oxalyl chloride and the like. Typical bases include TEA, DIE A, pyridine, K 2 C0 3 , NaH and the like. Typical acylation catalysts include HOBt, HOAt, 4-dimethylaniinopyridine and the like. Typical hydride reducing agents include NaB¾, NaBH(GAc)3 > NaB¾CN and the like. Typical phosgene equivalents include phosgene, triphosgene, carbonyldiirnidazole, 4- nitrophenylchloroformate and the like.

One skilled in the art will recognize that variations to the order of the steps and reagents discussed in reference to General Reaction Scheme I are possible. Methodologies for preparation of compounds of structure (I) are described in more detail in the following non-limiting exemplary schemes. It will also be appreciated by those skilled in the art that in the process described herein the functional groups of intermediate compounds may need, to be protected by suitable protecting groups. Such functional groups include hydroxy, amino, mercapto and carboxylic acid. Suitable protecting groups for hydroxy include trialkyisilyJ or diary lalkylsilyl (for example, /-butyl dimetliylsilyi, /-butyldiphemisilyl or trimethylsilyl), tetra ydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include /-butoxycarbonvl, benzyloxycarbonyl, and the like. Suitable protecting groups for mercapto include -C(0)-R" (where R" is alkyl, aryl or aryialkyl), p-niethoxybenzyl, trityl and the like. Suitable protecting groups for carboxylic acid include alkyl, aryl or aryialkyl esters. Protecting groups may be added or removed in accordance with standard, techniques, which are known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Green, T.W. and P.G.M. Wutz, Protective Groups in Organic Synthesis (199.9), 3rd Ed„ Wiley. As one of skill in the art would, appreciate, the protecting group may also be a polymer resin such, as a Wang resin. Rink resin or a 2-chlorotrityl-chloride resin.

it will also be appreciated by those skilled in the art, although such protected derivatives of compounds of this invention may not possess pharmacological activity as such, they may be administered to a mammal and thereafter metabolized in the bod to form compounds of the invention which are pharmacologically active. Such derivatives may therefore be described as " 'prodrugs ' ", All prodrugs of compounds of this invention are included within the scope of the invention.

By the methods described above, the representative compounds set forth in Examples 1-2 1 may be made, as well as by the more detailed procecures disclosed in the Examples. IV. TGR5 Methods

As mentioned above, new agents have recently been introduced to the market which prolong or mimic the effects of the naturally-secreted incretin hormones (Neumiller, J Am Pharm Assoc. 49(suppl 1):S16-S29, 2009). Another approach to initiating an incretin response involves the activation of TGR5, a bile acid, sensitive G- protein coupled receptor (GPCR). TGR5 activation induces the secretion of incretins such as GLP-1 from the enteroendocrine L cells of the distal gut, thus providing the benefits of incretin therapy through an alternative mechanism, Activation of TGR5 might therefore be beneficial for the treatment of diabetes, obesity, metabolic syndrome, and related disorders. However, a key challenge remains in discovering how TGR5 agonism could generate a prolonged GLP-1 response, which would be necessary to achieve therapeutic benefit.

Bile acids (BA) are amphipathic molecules which are synthesized in the liver from cholesterol and stored in. the gall bladder until secretion into the duodenum to play an important role in the solubilization and absorption of dietary fat and lipid- solub!e vitamins. Approx. 99% of BA are absorbed again by passive diffusion and active transport in the terminal ileum and transported back to the liver via the portal vein (enterohepatic circulation), in the liver, BA decrease their own biosynthesis from cholesterol through the activation of the farnesoid X receptor alpha (FXRct) and small heterodimer partner (SHP), leading to the transcriptional repression of cholesterol 7a- hydroxylase, the rate-limiting step of BA biosynthesis from cholesterol. A. G protein- coupled receptor responsive to bile acids, called TGR5, was independently identified by two investigators (Maruyama et al, "Identification of membrane-type receptor for bile acids (M-BAR)" Biochem. Biophys. Res. Comm. 298, 714-719, 2002; Kawamata et al,, "A G Protein-coupled Receptor Responsive to Bile Acids" J. Biological Chem. 278, Mo, 1 1, 9435-9440, 2003), marking the first identification of ceil surface receptors for fliis class of molecules. TGR5, in the literature also termed GPBARI, M-BAR or BG37, is expressed in inflammation-mediating cells (e.g. macrophages), as well as a number of eirteroendocrine derived cells lines such as GLUTtag, STC-1 and NCI-H716. Katsuma and colleagues demonstrated that bile acids could mediate the secretion of GLP-1 via TGR5 in STC-1 cells (Katsuma et al., Biochemical and Biophysical Research Communications 329:386-390, 2005).

TGR5 mRNA and protein have been reported to be expressed hi a wide variety of tissues, although agreement on the sites of predominant expression appears to vary depending on the investigating group. It is clear that TGR5 mediates sensing of bile acids in. for example, brown fat, macrophages, gall bladder, and intestinal neurons; however, the function of this signaling is still being elucidated. While TGR5 has been found to be expressed in liver, it is not expressed in hepatocytes. but rathe in liver sinusoidal endothelial cells and cholangiocytes (epithelial cells of the bile duct). This has implications for the role of TG 5 in bile acid regulation.

The compounds of the present invention are impermeable but still capable of inducing a TGR5 -stimulated GLIM response, indicating that the TGR5 receptor may be present on the apical surface of the enteroendocrine L-cell in the GI tract. The development of methods to isolate primary L cells from mouse intestine (Reimann et ah, Cell Metabolism. 8:532-539, 2C08) allowed confirmation that TGR5 was expressed in these GLP-1 secreting cells. In another study, a modestly active agonist of TGR5 was used to demonstrate a role for TGR5 in glucose homeostasis (Thomas et al.. Cell Metabolism 10:167-477, 2009). In particular, they demonstrated that, oral admini tration of INT-777 (EC 50 of ~1 μΜ vs. human TGR5; mouse potency not reported) to wild type mice resulted in an increase of plasma levels of GLP-1. When the experiment was performed in TGR5-/- mice, the response to INT-777 was not observed. Using INT-777 in a chronic diet-induced obesity model in mice, the investigators showed that the TGR5 agonist would improve glucose tolerance, an effect that was lost in the TGR5-/- mice. However, since this systemic TGR5 agonist also has significant effects on energy metabolism in mice due to its effects on brown fat and other tissues, it. was miclear what contribution the enhanced GLP-1 expression had on the improvement of diet induced obesity.

TGR5 is also expressed in the gall bladder, and appears to modulate the filling and emptying of this organ. Vassiieva and coworkers performed in situ hybridization experiments in TG.R.5 knockout mice and determined that, there is significant TGR.5 expression in the epithelial ceils of the mouse gall bladder (Vassiieva et al, Biochem, J. 398:423-430, 2006). They also demonstrated that TGR5 null mice are resistant to cholesterol gallstone disease when fed a lithogenic diet. In investigating the mechanism of resistance, they noted that the level of phospholipids was reduced in the total bile pool, indicating that the bile had a reduced cholesterol saturation index. They attributed this change to significantly higher hepatic expression levels of genes involved in bile acid synthesis (Cyp7al and Cyp27al), and in hepatocellular uptake. (Ntc l and Oalpl) in mice on the lithogenic diet, which suggests that the loss of TGR5 function impairs the negati ve feedback regulation of bile acid synthesis. TGR5 protein is also expressed in human gallbladder epithelium (Keitel et a!., Hepatology 50(3), 861 -870, 2009). Keitel and coworkers examined 19 human gall bladder samples and detected TGR5 m NA and protein, in all samples tested. And although TGR5 mRNA was elevated in the presence of gallstones, no such relation was found for TGR5 protein levels. In addition, they found that TGR5 also localized in apical recycling endosomes, indicating thai the receptor is regulated through translocation, The authors noted the significance of this finding, as in both cholangiocy es and gallbladder epithelium (which are exposed to millknolar bile acid concentrations) TG 5 is mainly localized in a subapical compartment and only to a smaller extent in the plasma membrane. In contrast, in sinusoidal endothelial cells and Kupffer cells (cells normally exposed to low bile acid concentrations) the receptor was predominantly detected within the plasma membrane.

It has also been reported that TGR5 mediated cAMP elevation can result in fluid and electrolyte secretion via activation and translocation of the cystic fibrosis transmembrane conductance regulator (CFTR). In addition to the presence of TGR5 in gallbladder epithelium, (Lavoie et al, J Physiol 588(17):3295~3305 2010) demonstrated via PGR and immuriohistoehemistry that TGR5 is also expressed in gallbladder smooth muscle cells in the mouse. Functionally, they showed that bile acid TGR5 agonists could disrupt gallbladder smooth muscle function ex vivo, and that this disruption did not occur for tissues removed from TGR5-/- mice.

Additional functional confirmation of the role of TGR5 activation in gallbladder function came from the Mangelsdorf group, who used. TGR5 knockout mice to demonstrate that TGR5 activation stimulates gallbladder filling (Li et al, Mol Endocrinol 25(6), 1066-71 , 201.1). They demonstrated that i.p. injections of TGR5 agonists lithocholic acid (LCA) or l ' NT-777 resulted in an approximately two-fold doubling of gallbladder volume in 30 minutes. The effect was completely blunted in the knockout mice. In further experiments examining direct effects on gallbladder smooth muscle in ex vivo tensiometry experiments, the investigators showed that both. LCA and INT-777 markedly relaxed gallbladders from wild-type but not knockout mice, supporting the model that TGR5 acts directly on gallbladder to cause smooth muscle relaxation via induction of secondary messengers. In aggregate, these studies indicate that TGR5 stimulation elicits gallbladder relaxation most likely via epithelial and/or smooth muscle TGR5 activation. The findings described, above suggest that a TGR5 agonist being developed for diabetes should most preferably cause little or no activation of TGR5 in the biliary tree, as evidenced by lack of gallbladder filling during short or Song term dosing.

In the small intestine, stimulation of TGR5 on enteroendocrine cells (L cells) by bile acids results in activation of adenylate cyclase (AC), thereby stimulating cAMP production and calcium influx. Increases in intracellular calcium and cAMP both lead to increased secretion of GLP-1 from L cells. Secreted GLP-1 has a number of effects, It augments glucose-dependent insulin release from β cells, it promotes β cell development, and it stimulates afferent nerves. GLP-1 also induces transcription of the insulin gene, thereby replenishing insulin stores. GLP-1 directly stimulates anorectic pathways in tire hypothalamus and brain stem, resulting in a reduction in food intake.

While specific activation of TGR5 on the enteroendocrine cells of the GI tract offers distinct benefits to a diabetic population, activation of TGR5 receptors o tissues outside the GI tract, such as macrophages, liver sinusoidal endothelial ceils (SECs), cholangiocytes (epithelial cells of the bile duct), and the like, can have unknown effects. For example, Kawamata and coworkers showed that bile acid treatment suppressed cytokine production in rabbit alveolar macrophages and TGR5- expressing monocytic cell line ΊΉΡ-1 (Kawamata, Journal of Biological Chemistry, 278(1 l):9435-9440, 2003) in macrophages, monocytes and upffer cells (liver resident macrophages) TGR5 activation iniiibits cytokine release (interleukins (ILs) and tumor necrosis factor (TNF)-cc). In liver SECs, TGR.5 activation increases endothelial nitric oxide synthase (eNOS) activity, leading to nitric oxide production and vasodilation. Therefore, a preferred TGR5 agonist should ideally be capable of the bile-acid like stimulation of Gi-resident L cells from the GI luminal side, but possess minimal to no systemic exposure and thereby avoid or minimize interactions with TGR5 receptors present on macrophages, cholangiocytes, tissues of the gall bladder, and the like. Although the compounds of the present invention are, in certain embodiments, impermeable, they are still capable of inducing a TGR5-stimulated GLP-1 response, indicating that the TGR5 receptor may be present on the apical surface of the enteroendocrine L-cell in the GI tract. Accordingly, and in some embodiments, the present compounds find utility as TGR5 agonists and may be employed in methods for treating various conditions or diseases, including diabetes. Advantageously, some embodiments include compounds which are substantially non-svstemically available. In certain embodiments, such compounds do not modulate filling or emptying of the gall bladder and in some embodiments may be present in the gall bladder in concentrations less than about 10 μΜ. While not wishing to be bound by theory, Applicants believe that certain functional groups on the compounds may contribute to the non-systemic availability of the compounds. For example, compounds of structure (T) which comprise polar functionality (e.g., a "Q" substituent having hydroxyl, guanidinyl, carboxyl, etc. substitutions) may be particularity useful as non-systemic TGR5 agonists.

In one embodiment the present disclosure provides the use of the disclosed compounds (compounds of structure (I)) as a therapeutically active substance, for example as a therapeutic active substance for the treatment of diseases which are associated with the modulation of TGK5 activity.

in other embodiments, the disclosure is directed to a method for the treatment of diseases which are associated with the modulation of TGR5 activity, wherein the diseases are selected from diabetes. Type II diabetes, gestational diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, hyperglycemia, obesity, metabolic syndrome, ischemia, myocardial infarction, retinopathy, vascular restenosis, hypercholesterolemia, hypertriglyceridemia, dyslipidemia or hyperlipidemia, lipid disorders such as low HDL cholesterol or high LDL cholesterol, high blood pressure, angina pectoris, coronary artery disease, atherosclerosis, cardiac hypertrophy, rheumatoid arthritis, asthma, chronic obstructive pulmonary disease (COPD), psoriasis, ulcerative colitis, Crohn's disease, disorders associated with parenteral nutrition especially during small bovvel syndrome, irritable bovvel syndrome (IBS), allergy diseases, fatty liver, non-alcoholic fatty liver disease (NAFLD), liver fibrosis, nonalcoholic steatohepatitis (NASH), primary sclerosing cholangitis (PSC), liver cirrhosis, primary biliary cirrhosis (PBC), kidney fibrosis, anorexia nervosa, bulimia nervosa and neurological disorders such as Alzheimer's disease, multiple sclerosis, schizophrenia and impaired cognition, the method comprising administering a therapeutically active amount of a compound of any one of claims 1 -68 to a patient in need thereof.

In certain embodiments the disease is diabetes, and in other embodiments the disease is is Type II diabetes or gestational diabetes.

The disclosure also provides use of the disclosed compounds (i.e.. any compound of structure (I)) for the preparation of medicaments for the treatment of diseases which are associated with the modulation of TGRS activity. For example, in certain embodiments die use is for the preparation of medicaments for the treatment a disease or condition selected from, diabetes, Type II diabetes, gestational diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, hyperglycemia, obesity, metabolic syndrome, ischemia, myocardial infarction,, retinopathy, vascular restenosis, hypercholesterolemia, hypertriglyceridemia, dvslipidemia or hyperlipidemia, lipid disorders such as low HDL cholesterol or high LDL cholesterol, high blood pressure, angina pectoris, coronary artery disease, atherosclerosis, cardiac hypertrophy, rheumatoid arthritis, asthma, chronic obstructive pulmonary disease (COPD), psoriasis, ulcerative colitis, Crohn's disease, disorders associated with parenteral nutrition especially during small bowl syndrome, irritable bowl disease (IBS), allergy diseases, fatty liver, liver fibrosis, liver cirrhosis, liver .cholestasis, primary biliary cirrhosis, primary scleroting cholangitis, kidney fibrosis, anorexia nervosa, bulimia nervosa and neurological disorders such as Alzheimer's disease, multiple sclerosis, schizophrenia and impaired cognition. In even other embodiments the disease is diabetes, and in other embodiments disease is Type II diabetes or gestational diabetes.

In still other embodiments, the disclosure provides a method for treating Type II diabetes mellitus in a patient in need thereof, the method comprising administering to the patient an effective amount of a compound of structure (I) or a pharmaceutical composition comprising the same.

In still other embodiments, the disclosure provides a method for treating inflammation of the GI tract in a patient, in need thereof, the method comprising administering to the patient an effective amount of a compound of structure (I) or a pharmaceutical composition comprising the same. In certain embodiments, the use is for the preparation of medicaments for the treatment of a disease or condition selected from ulcerative colitis and Crohn's disease, conditions generally referred to in the aggregate as inflammatory bowel disease (IBD). In IBD, suppression of proinflammatory cytokine production within, the Gi tissues surrounding the lumen of the GI is a desirable attribute. Therefore, a preferred TGR5 agonist for the treatment of IBD should ideally be capable of the bile-acid like stimulation of Coresident L cells from the GI luminal side as well as macrophages, monocytes and other ceils resident in tissues surrounding the GI lumen but possess minimal to no systemic plasma exposure and thereby avoid or minimize interactions with TGR5 receptors present on eholangiocytes, tissues of the gall bladder, and the like.

in still other embodiments, the disclosure provides a method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist administration does not induce the filling of the gall bladder of the mammal as determined by ultrasound analysis.

In yet other embodiments, the disclosure provides a method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the patient and wherein the TGR5 agonist administration does not induce the emptying of the gall bladder of the mammal as determined by ultrasound analysis.

In some other embodiments, the disclosure provides a method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the patient and wherein the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than. 400% when compared to administration of a placebo. For example, in some embodiments the change in weight of the mammal's gall bladder is determined in a mouse model.

In certain embodiments of the forgoing, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 300% when compared to administration of a placebo. In other embodiments, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 200% when compared to administration of a placebo. In some other embodiments, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 100% when compared to administration of a placebo. In other embodiments, the TGR5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 50% when compared to administration of a placebo. In certain other embodiments, the TG 5 agonist administration does not cause a change in weight of the mammal's gall bladder by more than 10% when compared to administration of a placebo.

Another embodiment is directed to a method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the mammal and wherein the TGR5 agonist concentration, in the gall bladder is less than about 100 uM. The concentration of the TGR5 agonist in the gall bladder may be determined by any number of methods kno wn in the art. For example, in some embodiments the TGR5 agonist concentration in the gall bladder is determined in a mouse model.

In other embodiments of the foregoing, the TGR5 agonist concentration in the gall bladder is less than about 50 μΜ, les than, about 25 μΜ, less than about 10 μΜ. less than about 5 μΜ, less than about 1 μΜ or even less than about 0.1 μΜ.

In another embodiment, the present disclosure provides a method for stimulating GLP-1 secretion in a mammal, the method comprising administering a TGR5 agonist that is active in the gastrointestinal tract of the mammal and. wherein the TGR5 agonist concentration in the mammal's plasma is less than the TGR5 EC 5 o of the TGR5 agonist. For example, in some embodiments the TGR5 agonist concentration in the mammal's plasma is less than 50 ng/mL. In some other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 25 ng/mL. in some other embodiments the TGR5 agonist concentration in the mammal's plasma is less than, about 1 ng/mL. In some other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 5 ng/mL. In yet other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 1 ng/mL. In still other embodiments the disclosure orovides a method for treating Type II diabetes mellitus in a patient in need thereof, the method comprising administering to the patient an effective amount of any of the disclosed TGR5 agonists or a pharmaceutical composition comprising the same, In some embodiments, the phannaceutical composition comprises an additional therapeutic agent selected from the additional therapeutic agents described above.

in some other embodiments of any of the foregoing methods, the TGR5 agonist is not systemically available. In other embodiments of any of the foregoing TGR5 agonists, the TGR5 agonist concentration in the mammal's plasma is less than the TGR.5 EC50 of the TGR5 agonist. For example, in some embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 50 ng/mL. In some other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 25 ng/mL. in some other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 10 ng/mL. In some other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 5 ng/mL. In yet other embodiments the TGR5 agonist concentration in the mammal's plasma is less than about 1 ng/mL.

In other embodiments of any of the forgoing methods, the TGR5 agonist- does not modulate TGR5-mediated suppression of cytokines. In some other embodiments, the TGR5 agonist does not modulate the ileal bile acid transporter (IB AT). In yet other embodiments, the TGR5 agonist does not modulate the Farnesoid X Receptor (FXR).

In other embodiments of any of the foregoing methods, the TGR5 agonist stimulates ΡΥΎ secretion, Enteroendocrine L-cells ca be stimulated by nutrients and/or bile acids to co-secrete PYY and GLP-1. PYY plays an integral role in appetite control and energy homeostasis, and thus its co-release with GLP-1 in response to a TGR.5 agonist could provide an added beneficial effect.

In other embodiments of any of the foregoing methods, the TGR5 agonist stimulates GLP-2 secretion. Enteroendocrine L-cells can be stimulated by nutrients and/or bile acids to co-secrete GLP-1 and GLP-2. GLP-2 plays an integral role in mainiainance of the gastrointestinal mucosal epithelium and thus its co-release with GLP-1 in response to a TGR5 agonist could provide an added beneficial effect in conditions associated with disruption of the gastrointestinal mucosal epithelium. Pharmacological intervention with a GLP-2 agonist reduces the severity of damage in a rodent models of ulcerative colitis (Daniel J, Drucker et al., Am. J. Physiol. GastrointesL Liver Physiol. 276, G79-G91, 1999 "Human [Gly2]GLP-2 Reduces the Severity of Colonic Injury in a Murine Model of Experimental Colitis" and Marie- Claude L'Heureux ct al, J. Pharmacol Exp. Ther. 306, 347-354, 2003 "Glucagon-Like Peptide-2 and Common Therapeutics in a Murine Model of Ulcerative Colitis' " ). For example, in certain embodiments the use is for the preparation of medicaments for the treatment a disease or condition selected from ulcerative colitis, Crohn's disease and disorders associated with parenteral nutrition especially during small bowel syndrome.

In certain other embodiments of any of the foregoing methods, the TGR5 agonist is a compound of structure (I).

In mammals such as mice, gallbladder phenotype (e.g. filled or empty) can be assessed surgically, by excising and weighing the gallbladder at a defined interval in an experiment in humans and other higher mamamals, there are also convenient and non-invasive ways to assess gallbladder phenotype. For example, Liddle and coworkers used abdominal ultrasonography to assess gallbladder volumes, wall thickening and the presence of gallstones or other pathology in human subjects taking a cholecystokinin (CCK) receptor antagonist (which blocks gallbladder emptying) (Liddle, J. Clin. Invest. 84:1220-1225, 1989). Such techniques can be used in the present invention to determine if a TGR5 agonist is affecting the filling or emptying of the gallbladder. V. Compositions and Administration

For the purposes of administration, the compounds of the present invention may be administered as a raw chemical or may be formulated as pharmaceutical compositions. Pharmaceutical compositions of the present invention comprise a compound of structure (I) and a pharmaceutically acceptable carrier, diluent or excipient. The compound of structure (I) is present in the composition in an amount which is effective to trea a particular disease or condition of interest - that is, in an amount sufficient to agonize TGR5, and preferably with acceptable toxicity to the patient, TGR5 activity of compounds of structure (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art.

Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out ia any of the accepted modes of admini tration of agents for serving similar utilities. The pharmaceutical compositions of the invention can be prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. Typical routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal, and intranasal. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, mtrasternal injection or infusion techniques. Pharmaceutical compositions of the invention are formulated so as io allow the active ingredients contained therein to be bioavaiiable upon administration of the composition, to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aerosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms arc known, or will be apparent, to those skilled in this art; for example, see Remington; The Science and Practice of Pharmacy, 20ih Edition (Philadelphia College of Pharmacy and Science, 2000). The composition to he administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this invention. In some embodiments, the disclosure provides a pharmaceutical composition comprising any of the foregoing compounds (I.e., a compound of structure (I)) and a pharmaceutically acceptable carrier or adjuvant.

In some embodiments, the disclosure provides a pharmaceutical composition comprising any of the foregoing compounds (i.e., a compound, of structure (I)), a pharmaceutically acceptable carrier or adjuvant and one or more additional biologically active agents. For example, in some embodiments the one or more additional biologically active agents are selected from dipeptidyl peptidase 4 (DPP-4) inhibitors, biguanidines, sulfonylureas, oc-glucosidates inhibitors, thiazoHdinediones, incretin mimetics, CBl antagonists, VPAC2 agonists, glucokinase activators, glucagon receptor antagonists, PEPC . inhibitors, SGLT1 inhibitors, SGLT2 inhibitors, IL-1 receptor antagonists, SIRT1 activators. SPPARMs and 1 ΙβΙ ISD1 inhibitors.

In some other embodiments, the one or more additional biologically active agents prolong the TGR5 -mediated GLP-1 signal. In other embodiments, the one or more additional biologically active agents are DPP-4 inhibitors. In still other embodiments, the one or more additional biologically active agents are sitagliptm, vildagliptin, saxagliptin, Iinagliptin, alogliptin, gemigliptin or dutogliptin. In. even other embodiments, the one or more additional biologically active agents are selected from the group consis ting of metformin or other biguanidine, glyburide or other s ulfonyl urea, acarbose or other a-glucosidase inhibitor, rosiglitazone or other thiazolidiriedioae and exenatide or other incretin mimetic.

Tn some other embodiments, the present disclosure is directed to a pharmaceutical composition comprising any of TGR5 agonists described herein and a pharmaceutically acceptable carrier or adjuvant. For example, in some further embodiments of the foregoing, the pharmaceutical compositiosn further comprises one or more additional biologically active agents. In some embodiments, the one or more additional biologically active agents are DPP-4 inhibitors. In other embodiments, the one or more additional biologically active agenta are sitagliptin, vildagliptin, saxagliptin, Iinagliptin, alogliptin, gemigliptin or duiogliptirt. A pharmaceutical composition of the invention may be in the form of a solid or liquid. In one aspect, the carrier(s) are particulate, so that the compositions are, for example, in tablet or powder form. The carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inbalatory administration.

When intended for oral administration, the pharmaceutical composition is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.

As a solid composition for oral administration, the pharmaceutical composition may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form. Such a solid composition will typically contain one or more inert diluents or edible carriers. In addition,, one or more of the following may be present: binders such as carboxymethylcellulose, ethyl cellulose, rnicrocrystailine cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as aiginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.

When the pharmaceutical composition is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.

The pharmaceutical composition may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension, The liquid may be for oral administration or for delivery by injection, as two examples. When intended for oral administration, preferred composition contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer. In a composition intended to be administered by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included. The liquid pharmaceutical compositions of the invention, whether they be solutions, suspensions or other like form, may include one or more of she following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline. Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Physiological saline is a preferred adjuvant. An injectable pharmaceutical composition is preferably sterile.

A liquid pharmaceutical composition of the invention intended for either parenteral or oral administration should contain an amount of a compound of the invention such that a suitable dosage will be obtained.

The pharmaceutical composition of the invention may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base. The base, for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and. eniulsiiiers and stabilizers. Thickening agents may be present in a pharmaceutical composition for topical administration. If intended for transdermal administration, the composition may include a transdermal patch or iontophoresis device.

The pharmaceutical composition of the invention may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug, The composition for rectal administration may contain an oleaginous base as a suitable nonirritating excipient. Such bases include, without limitation, lanolin, cocoa butter and poly ethylene glycol.

The pharmaceutical composition of the invention, may include various materials, which modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating shell around the active ingredients. The materials thai form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents. Alternatively, the active ingredients may be encased in a gelatin capsule.

The pharmaceutical composition of the invention in solid or liquid form may include an agent that binds to the compound of the invention and thereby assists in the delivery of the compound. Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.

The pharmaceutical composition of the invention may consist of dosage units that can be administered as an aerosol. The term aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the invention may be delivered in single phase, bi-phasie, or tri-phasic systems in order to deliver the active ingredieni(s). Delivery of the aerosol includes the necessary container, activators, valves, subcontainers, and the like, which together may form a kit. One skilled in the art, without undue experimentation may determine preferred aerosols.

The pharmaceutical compositions of the invention may be prepared by methodology well known in the pharmaceutical art. For example, a pharmaceutical composition intended to be administered by injection can be prepared by combining a compound of the invention with sterile, distilled water so as to form a solution. A surfactant may be added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalendy interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.

The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed: the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy,

Compounds of the invention, or pharmaceutically acceptable derivatives thereof, may also be administered, simultaneously with, prior to, or after administration of one or more other therapeutic agents. For example, the compounds of the present invention may be administered with other therapeutically active compounds. Such methods are describe in more detail below. Such combination therapy includes administration of a. single pharmaceutical dosage formulation which contains a compound of the invention and one or more additional active agents, as well as administration of the compound of the invention and each active agent in its own separate pharmaceutical dosage formulation. For example, a compound of the invention and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations. Where separate dosage formulations are used, the compounds of the invention and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens,

Suitable pharmaceutical compositions may be formulated by means known in the art and their mode of administration and dose determined by the skilled practitioner. For parenteral administration, a compound may be dissolved in sterile water or saline or a pharmaceutically acceptable vehicle used for administration of non-water soluble compounds such as those used for vitamin K. For enteral administration, the compound may be administered in a tablet, capsule or dissolved in liquid form. The tablet or capsule may be enteric coated, or in a formulation for sustained release. Many suitable formulations are known, including, polymeric or protein microparticies encapsulating a compound to be released, ointments, pastes, gels, hydro gels, or solutions which, can be used topically or locally to administer a compound. A sustained release patch or implant may be employed to provide release over a prolonged period of time. Many techniques known, to one of skill in the art axe described in Remington: the Science & Practice of Pharmacy by Alfonso Gennaro, 20 i ed., Lippencott- Williams & llkins, (2000). Formulations for parenteral administration may, for example, contain excipients, polyalkvlene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated naphthalenes. Biocompatible;, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Other potentially useful parenteral delivery systems for modulatory compounds include eihyiene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel,

Compounds or pharmaceutical compositions in accordance with this invention or for use in this invention may be administered by means of a medical device or appliance such as a implant, graft, prosthesis, stent, etc. Also, implants may be devised which are intended to contain and release such compounds or compositions. An example would be an implant made of a polymeric material adapted to release the compound over a period of time.

It is to be noted that dosage values may vary with the severity of the condition to be alleviated. For any particular subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions. Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners. The amount, of active compound(s) in the composition may vary according to factors such as the disease state, age, sex, and weight of the subject, Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. In general, compounds of the invention should, be used without causing substantial toxicity. Toxicity of the compounds of the invention can be determined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index, i.e., the ratio between the LD50 (the dose lethal to 50% of the population) and the 1,1)100 (the dose lethal to 1.00% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the compositions. Some compounds of this invention may he toxic at some concentrations. Titration studies may be used to determine toxic and non-toxic concentrations.

Compounds as described herein may be administered to a subject or patient. As used herein, a "subject" or "patient" may be a human, non-human primate, mamma!, rat, mouse, cow, horse, pig, sheep, goat, dog, cat and the like.

Various alternative embodiments and examples of the invention are described herein. These embodiments and examples are illustrative and should not be construed as limiting the scope of the invention.

EXAMPLES

i~(4-cyclopropyl ,2,3,4 eirahvdroquffl^

Scheme 1 : 5. Primary amine R 4 NH2 (R4=cyclopropyl); 2. methyl 2-chloro-2- oxoacetate, TEA, DCM; 3. ¾, Pd/C, MeOH; 4, PPh 3 , DMF 5. BH 3 *THF, THF; 6. 2- hydroxy-2-meihyIpropanoic acid, HATU, DIE A, DMF; 7. 2-(bromomethyl)- 1 ,4- dichlorobenzene, NaH, DMF.

hitermediate la: N-cyclopropyl-2-nitroanUine. To cyelopropylamine (100 mL) was added l~rluoro-2-nii.robenzene (30.0 g, 0.213 inol, LOO equiv) drop-wise with, stirring. The reaction mixture was stirred overnight at 30 °C then diluted with water (1 00 mL), extracted with ethyl acetate (2x100 mL) and the organic layers combined. The combined organic extract was washed with brine (3x100 mL) dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 45 g (crude) iV-cyclopropyi-2-nitroaraline as a yellow solid which was used without further purification.

Intermediate lb: methyl [cyclopropyl(2-nitrophenyl)carbamoyl]forniate. To a stirred 0 °C solution of A-cyclopropyl-2-nitroaniline (60 g, 0.337 mol, 1.00 equiv) and triethylamme (97.0 g, 0.959 mmol, 2.85 equiv) in dichloromethane (600 mL) was added methyl 2-chloro-2-oxoacetate (97.0 g, 0.792 mol, 2.35 equiv) drop-wise. The resulting reaction mixture was stirred for 3 h at 0-10 °C then diluted with of water (300 mL) and extracted with dichloromeihaiie (600 mL). The organic phase was washed with of aqueous sodium carbonate (3x200 mL) and brine (2x200 mL). dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide (88 g, 99%) of lb as red oil, MS (ES, rn/z): 265 j .Yl : H |

Intermediate 1 c : 1 -cyclopropyl-4-hy droxy- 1 ,2,3 ,4- tetrahydToquinoxaline-2,3-dione. Hydrogen gas was introduced into a stirred solution of [cyclopropyl(2-nitrophenyl)carbamoyl]formate (45,0 g, 0.170 niol, 1.00 equiv) and palladium on carbon (13 gj in methanol (400 mL). The resulting suspension was stirred for 3 h at 40 °C then solids were removed by filtration. The filter cake was washed with N, Λ-dimethyiformainide, the combined filtrate was concentrated under reduced pressure to provide (31 g, 83%) of lc as a white solid, MS (ES, m/z): 219 [M+Hf .

Intermediate Id: 1 -cyclopropyl- 1 ,2 ,44etrahydroqmnoxalme-2.3-dione. A stirred solution of l-cyclopropyl-4-hy<fcoxy-l,2 ,4 etrahydioquinoxaiine-2,3-dione

(31.0 g, 0.142 niol, 1 ,00 equiv) and ttiphenylphosphine (56.0 g, 0.2.14 mol, 1.50 equiv) in A V-dimethyiform amide (250 mL) was purged and maintained under an atmosphere of nitrogen,. The resulting solution was stirred for 2 h at 135 °C in an oil bath. The reaction mixture was cooled to 0 °C with an ice/water bath. Then diluted with of dichioromethane (300 mL) the solids were collected by filtration to provide (20 g, 70%) of Id as a brown solid. MS (ES, m/z): 203 [M+H] ÷ ,

Intermediate le: l-cyclopropyl-L2,3,4-tetrahydroquinoxaline. To a solution of l-cyclopropyl-l,2,3,4-tetrahydroquinoxaIine-2,3-dione (20.0 g, 0.989 mol, 1.00 equiv) in tetrahydrofuran (100 mL) was added B1¾ « THF (250 mL) the resulting solution was stirred for 4 h at 50 °C. The reaction mixture was then quenched by addition of aqueous sodium carbonate (100 mL) then concentrated under reduced pressure, diluted with of water (200 mL) and extracted with ethyl acetate (2x200 mL). The combine organic extract was washed with brine (2x200 mL), dried over sodium sulfate and concentrated under reduced pressure to provide crude product residue. The residue was purified by silica gel column chromatography with and eluent gradient of petroleum etherrethyl acetate (45:1 to 30:1) to furnish (11 g, 64%) of l e as a white solid. MS (ES, m/z): 175 [M+H] +

Intermediate If: l-(4-cyciopropyl-l,2,3,4-tetrahydroquinoxalin-l-yl)-2- hydroxy-2-methylpropan- 1 -one. A. solution of 1 -cyclopropyl- 1 ,2,3,4- tetrahydroquinoxaline (100 mg, 0.57 mmol, 1.0 equiv), 2-hydroxy-2-methylpropanoic acid (66 mg, 0.63 mmol, 1.10 equiv), HATU (262 nig, 0.69 mmol, 1 ,2 equiv) and DIEA (89 mg, 0.69 niniol, 1.2 equiv) in i^N-dimethylfomamide (2 mL) was stirred overnight at room temperature. The resulting solution was diluted with of Ι¾ϋ (5 mL) and extracted with of ethyl acetate (2x5 mL). The combined organic extract was washed with brine (1x10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative TLC (ethyl acetaterpetroleum ether 1 :5) to provide (30 mg, 20%) of If as yellow oil. MS (ES, m/z): 261 (M+H] +

Example 1 : 1 -(4 -cyclopropyl- 1,2,3 ,4-tetrahydroqumoxalin- 1 -y!)-2- [(2.5- dichlorophenyl)methoxy]-2-oiethylpropan-l-one. To a solution of l-(4-cyclopropyl-

1 ,2,3 ,4-terrahydroquinoxa! in- 1 -yl)-2-h ydrox -2-rnethylpropan- 1 -one (30 mg, 0.12 mmol, 1.0 equiv) in N, N-dimethylformamide (2 mL) was added sodium hydride (15 mg, 0.62 mmol, 5.4 equiv) at 0 °C and the reaction mixture was stirred at this temperature for 15 min then 2-(bromomethyl)-l ,4-dichloro benzene (30 mg, 0, 13 mmol, 1, 1 equiv) was added. The. reaction mixture was stirred overnight at room temperature then quenched by the addition of 5 ml. of water. The resulting solution was extracted with ethyl acetate (2x5 mL) and the combined organic extract was washed with brine (1x10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative TLC with ethyl acetate : petroleum ether (1 : 1). The crude product (20 mg) was purified by preparative HPLC : Column, SunFire Prep-CI S, 19* 150mm 5μτη; mobile phase gradient, water 0.05% TFA : C¾CN (35% to 50% CH 3 CN over 10 min; detector Waters 2545 UV detector 254/220nm) to furnish (2.7 mg, 3%) of the title compound TFA salt as brown oil .MS (ES, m/z): 419 [M-MTf . ! Ii-NMR (400 MHz, CD 3 OD) 5 7.27-7.37 (m, 4H), 7, 10-7.12 (m, 1H), 7.03 (t, J = 7.6 Hz, 1H), 6.68 (t, J = 7.6 Hz, 1H), 4.53 (s, 211), 4,09 (s, 2H), 3.35-3.38 (m, 2H), 2.34-2.39 (m, 1H), 1 .65 (s, 6H), 0.75-0.80 (m, 2H), 0.49 (m, 211).

Example 2

(4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)((^

Example 2: (4^yclopropyl-3,4~dihydroquinoxalm-l(2H)-yl)((2S s 4R)-l- (2,5-dic lorobenzyl)^-hydroxypyrrolidin-2-yl)metb.anone bis TFA salt. Example 2 was prepared using the procedures described in Example 6. MS (ES, m/z): 446 [M÷HJ + . Ή- NMR (400 MHz, CI¾OD) δ 7.71 (s, IH), 7.55 (s, 2H), 7.26 (s, 2H), 6.97 (d, J = 8 Hz, IH), 6.79 (m, 1H), 5.05 (t, J - 8 Hz, IH), 4.85-4.75 (in, IH), 4.60 (ra, IH), 4.52 (m, 1H), 3.99 (m, 1H), 3,81 (m, IH), 3.55 (m, IH), 3.40-3.32 (m, 2H), 3.19-3.15 (m, IH), 2.49 (s, IH), 2.04-1.94 (m, 211), 0.92-0.84 (m, 2H), 0.71-0.53 (m, 2H), 0.52 (d, J = 8

Example 3

(4-cyclopropyl-3 ,4-dihydroquinoxalin- 1 (2H)-yl)((2S,4S)- 1 -(2 < 5-dichiorobenzyl)-4- fluoiOpyrrolidin-2-yi)nietha.n.o.ae

Scheme 3: 1. dieftylaminosulfur Ixifluoride, ethyl acetate

Example 3 : (4-cyclopropyi-3 ,4-dihydroquinoxalin- 1 (2 H)-yl)((2S,4S)-l- (2,5-dichlorobenzyl)^-fluoropyrroMdin-2-yl)methajione. To Example 2 (40 mg, 0.090 mmol, 1.0 equiv) in ethyl acetate (6 mL) at 0 °C was added dropwise an ethyl acetate solution of diethyl aminosulfuf trifluoride (DAST; 36 mg, 0.22 mmol, 2.5 equiv) and the resulting solution was stirred overnight at room temperature, The mixture was diluted with 30 mL of ethyl acetate, washed with 1x20 mL of saturated aqueous sodium bicarbonate and 3x20 mL of brine, dried over sodium sulfate, concentrated and then purified by preparative reverse-phase HPLC to afford 40 mg (100%) of Example 3 bis TFA salt as a grey semi-solid. MS (ES, m/z): 448 ( i I I] . 1 H-NMR (400 MHz, CD 3 OD) δ 7.81 (s, 1H), 7.49 (s, 2H), 7.27 (s, 2H), 7.05 (s, 1H), 6.81 (s, 1H), 5.44-5.31 (m, 1H), 5.05 (t, J = 8 Hz, 1H), 4.57 (m, 1 H), 3.99-3.83 (m, 20). 3.83-3.62 (in, 2H), 3.46-3,40 (m, 1H), 3.27-3.23 (m, 1H), 2.48 (s, 2H), 2.30-2.13 (m, 2H), 0.89 (t, J= 4 Hz 5 2H), 0.66-0.54 (m, 2H).

Example 4

1 -(4-cyclopropyl-3 ,4-< ihydroquiiioxalin- 1 (2H -yl)-2-(2,5-dichlorophen.oxy )ethanone

Scheme 4: I . t-butyl bromoacetate, potassium t-butoxide, THF; 2. HC1 (g), DCM 3. 1 -cyclopropyl- 1 ,2,3 ,4-tetranydroquinoxaline, HAITI, DIEA, DMF.

Intermediate 4a: tert-butyl 2~(2.5-dichloroph.en.oxy)acetate, To 2,5- dichlorophenol (300 mg, 1.84 mmol, 1.00 equiv) in THF (10 mL) was added potassium t-butoxide (400 mg, 3.56 mmol, 1.94 equiv) and the mixture was stirred for 20 min. To this was added t-butyl 2-bromoacetate (700 mg, 3.59 mmol, .1.95 equiv) and the reaction was stirred for 1 h at room temperature. The mixture was diluted with 10 mL of water, extracted, with 2x20 mL of ethyl acetate, the organic layers combined and then washed with 2x15 mL of brine. The organic layer was dried, concentrated and then purified via silica gel chromatography, eluting with petroleum ether/ethyl acetate (30:1) to afford 300 mg (59%) of inteniiediate 4a as a colorless solid.

Intermediate 4b: 2-(2,5-dichlorophenoxy)acetic acid. To intermediate 4a (300 mg, 1.08 rnmol, 1.00 equiv) in dichlorornethane (10 mL) was bubbled hydrogen chloride gas and the solution then stirred for 5 h at 5 °C. The reaction was concentrated to afford 350 mg (95%, purity -65%) of crude intermediate 4b as a white solid.

Example 4: i-(4-cyck)propyl-3,4-dihydroquinoxa!in-l(2ITj-yl)-2-(2,5- dichlorophenoxy)ethanone. To i-eyclopropyi-1.,2 5 3,4 etrahydroqu.inoxaline (200 mg, 1.15 rnmol, 1.00 equiv) in DMF (10 mL) was added intermediate 4b (350 mg, 1.58 mmol, 1.40 equiv), HATU (655 mg, 1.72 rnmol, 1.50 equiv) and DIEA (222 mg, 1.72 rnmol, .1.50 equiv) and the resulting solution stirred for .2 h at 25 °C, The reaction was diluted with 50 mL of water, extracted with 3x25 mL of ethyl acetate, the organic layers then combined, washed with 2x25 mL of brine and then dried over anhydrous sodium, sulfate. The solution, was concentrated and the residue purified via preparative reverse- phase HPLC to afford 76.9 mg of Example 4 TFA salt as a white solid. MS (ES, m/z): 377 ; M - i l j . ^I-NM (400 MHz, DMS(W 6 ) δ 7.45 (m, 211), 7.01 (m, 4H), 6.66 (t, J = 6.8 Ηζ, ΙΗ), 5.13 (s, 2H), 3.74 (s, 2H), 3.36 (s, 2H), 2.45 (m, 1H), 0.84 (d, J = 6 Hz, 2H

Scheme 5: 1. a. oxalyl chloride, DM80, DCM; b. TEA; 2. DAST, DCM; 3. TFA, DCM; 4. 2-(bromometliyl) ,4-diehlorobenzene, 2 C0 3 , C¾CN; 5. Li OH, 1 ,4- dioxane, methanol, water; 6. 1 -cyclopropyl-1 ,23,4-tetrahydroqumoxalirie, HATU, DIEA, DMF. Intermediate 5a: (S)-l-tert-butyl 2-methyl 4-oxopyrrolidine-l,2- dicarboxyiate. To a solution of DMSO ( 1.90 g, 24.3 mmoL 3.00 equiv) in diehlorornethane (20 mL) at -78 °C was added oxalyl chloride (1.54 g, 12.1 mmol, 1.50 equiv) and the mixture was stirred for 15 min. To this was added dropwise a solution of 1-tert-butyi 2-methyl (2S ; 4R)~44iydroxypym)lidine-l ,2-dicarboxylate (2.00 g, 8.15 mmol, 1.00 equi v) in diehloromethane (8 ml,) and the mixture was stirred for 60 min at -78—60 °C. The solution was allowed to warm to RT and triethylamine (4.90 g, 48.4 mmol, 6.00 equiv) was added, The mixture was then diluted with 50 mL of diehloromethane, washed with 2x30 mL of brine, dried over anhydrous sodium sulfate, concentrated under reduced pressure and then purified via silica gel chromatography (dichloromethane/metbanol, 10:1) to afford 1 g (50%) of intermediate 5a as a yellow oil.

Intermediate 5b: (S)-l-tert-butyl 2-methyl 4.4-difluoropyrrolidine-I,2- dicarboxylate. To intermediate 5a (300 rag, 1.23 mmol, 1.00 equiv) in diehloromethane (30 mL) at 0 °C was added dropwise a solution of DAST (1.80 g, 11.2 mmol, 9.00 equiv) in diehloromethane (10 mL) and the resulting solution was stirred overnight at room temperature. The mixture was then washed with 1x30 mL of saturated aqueous sodium bicarbonate and 3x30 mL of brine, the organic layer was dried over anhydrous sodium sulfate and then concentrated under reduced pressure to afford 300 mg (92%) of intermediate 5b as yellow oil.

Intermediate 5c: (S)-methyl 4,4-difluoropyrrolidine-2-carboxylate. To intermediate 5b (300 mg, 1.13 mmol, 1.00 equiv) in diehloromethane (1 mL) was added trifluoroacetic acid (1 mL) and the resulting solution was stirred for 1 h at room temperature. The mixture was then concentrated under reduced pressure to afford 200 mg (crude) of intermediate 5c as a brown oil.

Intermediate Sd: (S)-methyl l-(2,5-dichiorobeiizyl)-4 ; 4- difluoropyrrolidine-2-carboxylate. To intermediate 5c (200 mg, 1 ,21 mmol. 1.00 equiv) in CH 3 CN (5 mL) was added 2-(bromomeihyl)-l,4-dichlorobenzene (288 mg, 1.20 mmol, 1.00 equiv) and potassium carbonate (502 mg, 3.63 mmol, 3.00 equiv) and the resulting solution was stirred overnight at room temperature. The mixture was diluted with 50 mL of ethyl acetate, washed with 2x30 mL of brine, the organic layer dried over anhydrous sodium sulfate, concentrated, and then purified via silica gel chromatography (petroleum ether/ethyl acetate, 50:1) to afford 200 mg (51%) of intermediate 5d as a yellow oil.

intermediate 5e: (S)- 1 -(2,5-dich]orobenzyl)--4,4-difluoropyrroIidine-2- carboxylic acid. To intermediate 5d (170 mg, 0.52 mmol, 1.0 equiv) in 1,4- dioxane/CH 3 OH/H 2 0 (3:2: 1 niL) was added LiOH*¾0 (44.0 mg, 1.05 mmol, 2.00 equiv) and the resulting solution was stirred for 60 mm at 80 °C. The pH value of the solution was adjusted to 6 with aqueous 2M HCl and the resulting mixture concentrated under reduced pressure to afford 120 mg (74%) of intermediate 5e as yellow oil.

Example 5: (S)-(4-cyclopropyl-3 5 4-dihydroqumoxdin-l(2H)-yl)(l -(2,5- dichkircmenzyr)-4,4-difluoropyrroiidin-2- i)methaiione. To intermediate 5e (120 mg, 0.39 mmol, 1.0 equiv) in DMF (5 mL) was added l-cyclopropyl-1 ,2,3,4- tetrahydroquinox aline (67.2 mg, 0.39 mmol, 1.00 equiv), HATU (294 mg, 0.77 mmol, 2.0 equiv) and D1EA (96.6 mg, 0.75 mmol, 2.0 equiv) and the resulting solution stirred overnight at room temperature. The mixture was diluted with 30 mL of ethyl acetate, washed with 3x20 mL of brine, and the organic layer dried over sodium sulfate. The crude product was purified by preparative reverse-phase HPLC to afford 20 mg (11%) of Example 5 bis TFA salt as a white solid.

Example 6

( 4-cyclopropyl-3 ,4-dmv&oqumoxaliri- 1 (2H>yl)((2S,4SV 1 -(2,5-dichloro¾enzyl)-4- hydroxvpyrroli.din-2"Vl)methanone

Scheme 6: 1. I -cyclopropyl-ί ,2,3 ,4-ietrahydroquinoxaline, HATU. DIE A, DMF; 2. Hydrochloric acid, 1 ,2-dicbloroethane; 3. DiAD, PPh 3 , PhC0 2 H, THF; 4. Piperidme, DMF; 5. 2-(bromomethyl)-l,4-dichlorobenzene, 2 C0 3 , CH 3 CN; 6. K 2 C0 3 , MeOFl. intermediate 6a: (2S,4R)-(9H-fluoren-9-yl)me yi 4-teit-b toxy-2-(4- cyclopropyl- 1 ,2,3,4-letrahydroquinoxaline- 1 -carbonyl)pyrroiidine~ 1 -carboxylate. To 1 - cyclopropyl-l,2,3,4-tetrahydroquinoxaline (400 nig, 2.30 mnioi, LOO equiv) in N,N- dimethylformamide (8 mi.) was added (2S,4R)-4-(tert-butoxy)-l-[(9H-fluoren-9- ylmethoxy)carbonyl]pyrrolidme-2-carboxylic acid (940 mg, 2.30 mmol. 1.00 equiv), HATU (1.30 g, 3.42 mmol, 1.50 equiv) and DIEA (444 nig, 3.44 mmol, 1.50 equiv) and the mixture was stirred overnight at room temperature. The resulting solution was diluted with 40 mL of ethyl acetate, washed with 4x30 mL of brine, dried over sodium sulfate, filtered and then concentrated under reduced pressure to afford 1.5 g (crude) of intermediate 6a as a blue solid.

Intermediate 6b: (2S,4R)-(9H-fluoren-9-yl)methyl 2-(4-cyclopropyl-

1 ,2,3,4-terrahydroquinoxaline-l -carbonyl)-4-hydroxypyrrolidine-l -carboxylate. To intermediate 6a (300 mg, 0.53 mmol, 1.00 equiv) in 1 ,2-dichloroethane (10 mL) was added concentrated hydrochloric acid (1 mL) and the resulting solution was stirred overnight at room temperature. The pH value of the solution was adjusted to 9 with saturated aqueous sodium carbonate then extracted with 3x20 mL of dichloromethane. The organic layers were combined, washed with 3x20 mi, of brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to afford 180 mg (67%) of intermediate 6b as a blue oil.

Intermediate 6c: (2S,4S)-(9H-fluoren-9-yl)me%! 4-(benzoyloxy)-2-(4- cyclopropyl - 1 ,2,3 ,4-tetrahydroqirinoxaline- -carbonyi)pyrrolidine- 1 -carboxylate. To intermediate lb (120 mg, 0,24 mmol, 1.0 equiv) in tetrahydrofuran (8 mL) at 0 °C was added PPh 3 (144 mg, 0.55 mmol, 2.4 equiv) and benzoic acid (72 n g, 0,59 mmol, 2.4 equiv) followed by the dropwise addition of a solution of DIAD (120 mg, 0.59 mmol, 2,4 equiv) in tetrahydrofuran (1 mL). The resulting solution was stirred for 2. h at room temperature then diluted with 40 mL of ethyl acetate, washed with 2x30 mL of brine and then dried over anhydrous sodium sulfate. The mixture was concentrated then applied onto a silica gel column, eiutmg with petroleum, ether/ethyl acetate (5: 1) to afford 200 mg (crude) of intermediate 6c as yellow oil.

Intermediate 6d: (3S,5S)-5-(4-cyclopropyl-l,2,3,4- tetrahydroq inoxaline-l-carbonyi)pyrrolidin-3-yl benzoate. To intermediate lc (200 mg, 0.33 nxmo!, 1.00 equiv) in. DMF (5 mL) was added piperidine (1 mL) and the resulting solution was stirred overnight at room temperature. The mixture was diluted with 30 mL of ethyl acetate, washed with 4x20 mL of brine, dried over sodium sulfate, filtered and then concentrated under reduced pressure to afford 150 mg (crude) intermediate 6d.

Intermedi ate 6e : (3 S , 5 S )-5 ·- (4 -cyclopropyl- 1,2,3,4- tetraliydroqumoxaline-- 1 -carbonyl)- 1 -(2,5-dichlorobenzyl)pyrrolidin-3-y 1 bertzoate. To intermediate lc (300 mg, 0.77 mmol, 1.0 equiv) in C¾CN (10 mL) was added 2- (bromomethyl)-l,4-dichlorobenzenc (180 mg, 0.75 mrnol, 1.0 equiv) and potassium carbonate (300 mg, 2.15 mrnol, 3.00 equiv) and the resulting suspension was stirred for 2 h at room temperature. The solids were filtered out and the filtrate was concentrated under reduced pressure to afford. 300 mg (71%) of intermediate 6e as a yellow oil.

Example 6:

(2.5-dieb1oroberizyl)-4~h.ydroxypy To intermediate le (300 mg,

0.54 mmol, 1.0 equiv) in. methanol (8 mL) was added potassium carbonate (226 mg, 1 .64 mmol, 3.00 equiv.) and the resulting solution was stirred for 60 min at room temperature. The mixture was concentrated, the residue was dissolved in 30 mL of ethyl acetate, washed with 3x20 ml, of brine, dried over anhydrous sodium sulfate, then filtered and concentrated under reduced pressure. The crude product (200 rag) was purified by preparative reverse -phase HPLC to afford 30.7 mg of the title compound bis TFA salt as a light yellow solid. MS (ES, m/z): 446 [M+Hf. ' f i-K ' MR (400 MHz, CD 3 OD) δ 7.81 (s, 1H), 7.55 (d, J = 8 Hz, 2H), 7.27 (m, 2H), 7.07 (d, J = 8 Hz, 1H), 6.79 (m, lH), 4.97 (t, J = 8 Hz, 1H), 4.67-4.58 (m, 2H), 4.42 (m, 1H), 4.14 (m, I B), 3.80-3.72 (m, lH), 3.63 (m, 1H), 3.49-3.43 (m s 3H), 3.18 (m, lH), 2.51 (t J = 4 Hz, H i }. 2.38 (m, 1H), 1.72 (d, J = 8 Hz, I H), 0.93-0.87 (m, 2H), 0.75-0.65 (m, 1H), 0.53 ( J= 4 Hz, 1H). >k < 7

1 -(4-cvcloprop yl-3.4-dihydroquinoxalin- 1 f 2H -vl)-2-(2.5 -dic orober!zylamino)propan-

1-one

Scheme 7: 1. 2-bromopropionyl chloride, TEA, DCM; 2. (2,5- di chiorophenyl)me anamme, K 2 CO 3 , DMF .

Intermediate 7a: 2-bromo-l-(4-cyclopropyl-3,4-dihy<koquinoxaliii- l(2H)-yl.)propan- 1 -one. To a solution of l-cyclopropyi-l,2,3,4-tetrahydroquinoxaline (600 mg, 3.44 mmol, 1.00 equiv) in DCM (20 mL) at 0 °C was added triethyiamine (697 mg, 6.89 rrumol, 2.00 equiv) followed by the dropwise addition of 2- bromopropanoyl chloride (1.17 g, 6.84 mmol, 2.00 equiv) and the resulting solution was allowed to warm to room temperature and then stirred for 3 h. The mixture was diluted with dichlorom ethane (50 mL), washed with 2x50 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 690 mg (65%) of intermediate 7a as a yellow oil.

Example 7: l-(4-cyclopropyl-3,4-dmydroqmnoxalin-i(2H)-yI)-2-(2,5- dic hi orobenzylamino)propan- 1-one. To intermediate 7a (600 mg, 1.94 mmol, 1.00 equiv) in DMF (10 mL) was added (2,5-dichlorophenyi)metbanamine (341 mg. 1.94 mmol, 1.00 equiv) and potassium carbonate (542 mg, 3.92 mmol, 2.00 equiv) and the reaction was stirred for 3 h at 60 °C. The reaction was diluted with. 50 mL of ethyl acetate, washed with water (2x50 mL), brine (2x50 mL), dried over anhydrous sodium sulfate and then concentrated. The residue was purified by preparative TLC (petroleum, ether/ethyl acetate (4:1)) followed by preparative reverse-phase HPLC to afford 30.2 mg (4%) of Example 7 bis TFA salt as a pink oil. MS (ES, m/z): 404 [M+H] \ 1H-NMR (300 MHz, CD 3 OD) 6 7,70 (s, IH), 7.56-7.47 (m, 2H), 7.29-7.22 (m, 211), 7.14-7.12 (m, 1H), 7.56-7.47 (m, 2H), 6.81 -6.70 (m, 1H), 4.73-4.59 (m, 1H), 4.46-4.30 (m, 3H), 3.83 (s, 1H), 3.54-3.31 (m, 3H), 2.51 (brs, 1H), 1 .63 (s, 1H), 1.24 (d, 6.9 Hz, 2H), 0.96- 0.82 (m, 2H), 0.73-0,62 (m, 1H), 0.52-0.48 (m, 3H).

Example 8

(4-eyclopropyl-3 ,4-dih vdroquinoxalin- 1 (2H )- yl)( 1 -

Scheme 8: 1. TMSCN, Znl 2 , DCM; 2. HC1, AcOH; 3. le, EDCI, HOAT, DMF; 4. 2-(bromometliyl)-l ,4-dichlorobenzene, NaH, DMF.

Intermediate Sa 1 -(trimetliylsilyloxyjcyelopentanecarbomtrile: Cyclopentanoiie (2 g, 23.78 mmol), TMSCN (3.53 g, 35.66 mmol). and Znl 2 (890 mg, 2.79 mmol) were dissolved in dichlorometliane (20 mL). The resulting solution was stirred for 6 h at room temperature, then diluted with 20 mL of ¾0 and extracted with twice with dicliloromethane. The combined organic layers were washed, with brine. The mixture was dried over anhydrous sodium sulfate and. concentrated under reduced pressure to give 8a (4 g, 92%) as brown oil, which was used without further purification.

Intermediate 8b 1-hydroxycyclopentatiecarboxylic acid: 8a (3 g, 16.36 mmol 1 .00 equiv) was dissolved in acetic acid (4 mL) and concentrated hydrogen chloride (4 mL). The resulting solution was stirred for 4 h at 80 °C. The mixture was then concentrated under reduced pressure to give 8b (2 g, 94%) as a white solid, which was used without further purification.

Intermediate 8c (4-cyclopropy i-3 ,4-dihydroquinoxalin- 1 (2H)- !)( 1 - hydrQxycyclopentyl)methanone: 8b (170 mg, 0.98 mmol), 1-hydroxycyclopentane-l- carboxylic acid (260 mg, 2.00 mmol), EDCi (288 mg, 1.50 mmol), and HO AT (204 mg, 1.50 rrmioi) were dissolved in /V A'-dimethylfomiamide (3 mL) and stirred, overnight at room temperature. The resulting solution was diluted with 10 ml, of ¾0 and extracted twice with ethyl acetate and the combined organic layers washed with brine. The organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. The resulting residue was purified by flash-column chromatography using ethyl acetate/petroleum ether (1 :5) as eluent to give 8c (40 mg, 14%) as a yellow solid. MS (ES, m/z) 287 [M+H] .

Example 8 (4-cy clopropy 1-3 ,4-dihydroqumoxalin- 1 (2H)~yl)( 1 - (2,5dichlorobenzyloxy)cyclopentyl)methanone: 8c (30 mg, 0.10 mmol was dissolved in (1 mL) and the resulting solution cooled to 0 °C. To the stirring solution was added sodium hydride (10 mg, 0.25 mmol) and the resulting mixture was stirred- for 10 rmn at 0 °C, A solution of 2-(bromometh i)- 1 ,4- dichloro benzene (40 mg, 0.17 mmol) in N,N-dimethyiformamide (1 mL) was then added and the resulting solution was stirred for 4 h at room temperature. The crade mixture was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 6 min gradient CI¾CN : ¾0 0.05% TFA (72 : 28 to 84 : 16) and detection by UV at 254 nm to provide the title compound TFA salt (25.7 mg. 55%) as a yellow semi-solid. MS (ES, m/z) 445 [M+l] + . Ή-NMR (300 MHz, CD 3 OD) δ 7.25-7.38 (m, 4H), 7.01-7.07 (m, 2H), 6.71 (t, J ~ 7.8 Hz, i l l). 4.44 (s, 2H), 4.01 (s, 2H), 3.33 (m, 2H), 2.33-2.41 (m. 3H), 2.04-2.19 (m, 2H), 1.72-1.82 (m, 4H), 0.76 i m. 2H), 0.52 (m, 2H).

Example 9

(4-cvclopropYl-3.4-dihvdi quinoxdin-l (2H)-yl){T -(2,5

U 2012/071251

Scheme 9: 1 . 1 -bydroxycyclopropanecarboxylic acid, HATU, DIEA, DMF; 2. 2- (bromomethyl)- 1.4-dichiorobenzene, K 2 C0 3 , DMF. intermediate a (4-cyclopropyl-3,4-dihydroquinoxalin-l(2ff)-yl)(l- hydroxycyelopropyl)methanone: 1-Hydroxycyclopropane-l-carboxylic acid (100 mg, 0.98 mmol), l-cyclopropyl-l,2,3,4-tetraliydroquinoxaiine (100 mg, 0.57 mmol, 1.0 equiv). HATU (262 mg, 0.69 mmol, 1.2 equiv), DIEA (90 mg, 0.70 mmol) was dissolved in N,N-dimethylform amide (2 mL). The resulting solution was stirred overnight at room temperature, then diluted with 10 mL of ¾0 and extracted twice with ethyl acetate. The organic layers were combined and washed with brine, then dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by preparative TLC with ethyl acetate/petroleum ether (1 : 1 ) to give 9a (100 mg, 67%) as a light yellow solid. MS (ES, m/z) 259 [M+i/ .

Example 9 (4-cyciopropyl-3,4-dihydroquinoxalm-l(2H)-y3.)(l-(2,5- dic orobenzyloxy)cyclopropyl)methanorie: 9a (85 mg, 0.33 mmol), 2-(bromomethyl)- 1 ,4-die orobenzene (85 mg, 0.35 mmol), and potassium carbonate (85 rng, 0.62 mmol) were dissolved in N,N-dimethyjformamide (2 mL). The resulting solution was stirred overnight at room temperature, then diluted with 20 mL of H 2 0 and extracted twice with ethyl acetate. The organic layers were combined and washed with brine, then dried over sodium sulfate and concentrated under reduced pressure. The crude mixture was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 7 rnirt gradient (CH 3 CN : H 2 0 0.05% TFA 60 : 40 to 80 : 20%) and detection by UV at 254 nm to provide the title compound (25, 1 mg, 18%) as the TFA salt. MS (ES, m/z): 417 [M+H] + . 'H- MR (400 MHz, CD 3 OD) 6 7.33 (d, J = 8.0 Hz, 1 H), 7.27 (d, J = 8.4 Hz, 1H), 7.19 (d, J= 8.4 Hz, HI), 7.08 (t, J = 8.0 Hz, IH), 6.99 (d, J = 7.6 Hz, HI), 6.74 (1 J = 8.0 Hz, IH), 6.57 (m, IH), 4.37 (s, 2H), 3.92 (s, 2H), 3.38-3.41 (m, 2H), 2.25-2.27 (m, IH), 1.46 (m, 2H), 1.18-1.22. (m, 2H), 0.66-0.67 (m, 2H), 0.19 (m, 2H). 71251

Example 10

Scheme 10: 1. HATU, DIEA, DMF; 2, TFA, DCM; 3. 2-(bromome l)-l.,4- dichlorobenzene, K2CO 3 , MeCN.

Intermediate 10a (S)- er/-butyi 2-( 1 ,2,3 ,4-tetraliydroquinollne- 1 - carbonyl)pyrrolidine- 1 -carboxylate: (2S)- 1 -[(Tert-butoxy)carbonyl]pyrrolidine-2- carboxylic acid (500 mg, 2.32 mmol), L2,3,4 etrahydroqumoline (620 rag, 4.65 mmol), HATU (1.77 g, 4.66 mmol), and DIEA (600 mg. 4.64 mmol) were dissolved in Ν,Ν-dimethylformamide (5 mL). The resulting solution was stirred for 2 h at room temperature, then quenched by the addition of water. The resulting solution was extracted with thrice with ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 10a (0.57 g, 74%) as yellow oil, which was used directly without further purification.

Intermediate 10b (S)-(3 ,4-dihydroqumolin- 1 (2H)-yl)(pyrroiidin-2- yi)mefhanone: 10a (500 mg, 1.51 mmol) was dissolved in dichloromethane (10 mL) and trifiuoroacetic acid (0.5 mL). The resulting solution was stirred overnight at room temperature, then concentrated under reduced pressure to give 10b (304 mg, 87%) as yellow oil, which was used directly without further purification.

Example 10 (S)-(l -(2,5-dichlorobenzyl)pyrrolidin-2-yl)(3 ,4- dmydroqumolin- 1 (2H)-yi)methanone: 10b (200 mg, 0.87 mmol), 2-(bromomethy])-l,4- dichlorobenzene (208 mg,.0.87 mmol). and potassium carbonate (360 mg, 2.60 mmol) were dissolved, in acetonitrile (5 mL) and the resulting solution was stirred overnight at room temperature. The mixture was then diluted with H 2 0 and extracted trice with ethyl acetate. The organic layers were combined and dried over anhydrous sodium sulfate, then concentrated under reduced pressure. The crude mixture was purified by preparative HPLC with a CI S silica gel stationary phase using a 8 rain gradient (CH 3 CN : H 2 0 0.05% TFA 23 : 77 to 41 : 59) and. detection by UV at 254 nm to provide the title compound as TFA salt (140 mg, 41%) as a yellow semi-solid. MS (ES, m/z); 389 [M+H] \ ! H-NMR (400 MHz, CD 3 OD) δ 7.75 (d, ,/= 26.1 Hz, 1.H), 7.54 (d, J = 4.5 Hz, 2H), 7.33 (s, 2H), 7.14-7.23 (m, 2H), 4.55-4.72 (m, 2H), 3.79-3.90 (m, 1H), 3.70 (d, J ~ 4.5 Hz, 2H), 3.50 (s, III), 3.37-3.42 (m, IH), 2,84 (d, J = 18 Hz, IH), 2.68 (d, ./ 3.6 Hz, U S ... 2.21 -2.33 (rn. IH), 2.01 -2.15 (m, IH ), 1.74 (s, IH).

Example 11

fS)-(l-(2,5-dicMorobenzyl)piperi

Example 11 (S)-(l -(2.5-dicb.lorobeiizyl)piperidtri-2-yl)(3,4~ dihydroqiiinolin- .1 (2H)-yl)methanone. 11 was synthesized in an analogous fashion to Example 10, using (S)-l-(tert-butoxycarbonyl)piperidine-2-carboxylic acid in place of (S)-lert-butyl 2-(l 5 2,3,4 etrahydroqu ioline- l -caA

Isolated as the TFA salt. MS (ES, m/z): 403 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) 5 7.80-7.86 (m, IH), 7.54-7,61 (m, 2H), 7,37-7.41 (ra, 3H), 7.19-7.22 (m, IH), 4.76-4.81 (m, HI), 4.66 (d, J= 13.2 Hz, IH), 4.51 (d, J = 12.9 Hz, I H), 4.36-4.40 (m, IH), 3,46- 3.55 (m, I H), 2.82-2,94 (m, IH), 2.12-2.19 (m, IH), 1.88- 1.99 (m, 2H), 1.68-1.77 (m, 4H), 1.33 (d, J= 6.6 Hz, I H). Example 12

-(4-cyclopropy[-3,4-c hy&^

-yl)methanone

Scheme 12: 1. (C0C1) 2 , DMF (cat.), DCM. 2. le, TEA, DCM. 3, 33% IIBr in HOAc.

4. 2-(broniomethyl)~l : ,4-dic " h1orobeiizene, K2CO3. CH 3 CN.

Intermediate 12a: (S)-ber.zyl 2-(chlorocarbonyl)pyirolidkie-l- carboxylate. To a solution of (S)- 1 -[(beazyloxy)carbonyl]pyrrolidine-2-carboxylic acid (214 nig, 0.86 mmol, 1.00 equiv) and DMF (cat) in DCM (10 mL) was added oxalyl chloride (324 mg, 2,55 mmol, 2,97 equiv) dropwise. The reaction mixture was stirred at room temperature for 2 h, and concentrated under reduced pressure to give 250 mg (crude) of (S)-benz i 2-(chlorocarbonyl)pyiTolidine-l-carboxylate as yellow oil.

Intermediate 12b: (S)-benzyi 2-(4-cyclopropyl-l ,2,3,4- tetrahydroquinoxaline- 1 -carbonyl) pyrrolidine-1 -carboxylate. To a solution of le (210 mg, 1.21 mmol, 1.00 equiv) in DCM (20 mL), were added (S)-benzyl 2- (chiorocarbonyl)pyrrolidine-l -carboxylate (320 mg, 1 ,20 mmol, 1.00 equiv) and triethylamine (126 mg, 1,25 mmol, 1.00 equiv). The resulting solution was stirred for 4 h at room temperature. The resulting mixture was concentrated under reduced pressure to provide 400 mg (82%) of (S)-benzyi 2-(4-cyclopropyl- 1 5 2 ,4 etrahydroq inoxaline- 1 -carbonyl) pyrrolidine- 1 -carboxylate as a yellow solid. MS (ES, m/z): 406 [M+H]' " .

Intermediate 12c: (S)-(4-cyclopropyl-3,4-dihydroquinoxalin- 1 (2H)~ yl)(pyrrolidin-2-yi)meiharione. To (S)-benzyl 2-(4-cyclopropyl- 1 ,2,3,4- tetrahydroquinoxaline-l-carbonyl) pyrrolidine- 1-carboxylate (300 mg, 0.74 mmol, 1.00 equiv) was added hydrogen bromide (33 wt% solution in glacial acetic acid, 5 mL). The mixture was stirred for 0.5 h at room temperature. The resulting mixture was concentrated under reduced pressure to give 300 mg (crude) of (S)-(4-cyclopropyl-3,4- cB ydroquinoxalin- 1 (2H)-yl)(pyrrolidin-2-yl)rnethariorie as a light yellow solid. MS (ES, m/z): 272 [M+H] +

Example 12: (S)-(4-cyclopropyl-3,4-diliydroquinoxalm-l(2H)-yl)(l- (2,5-dichlorobenzyl) pyrro]idin-2-yl)methanone. To a solution of (S)-(4~cycl opropy 1- 3,4-dihydroq inoxalin-l(2ir-yl)(pyrrolidin-2-yl)met^ (50 mg, 0.18 mmol, 1.0 equiv) in C¾CN (2 mL.) were added 2-(bromomethyl)- 1 ,4-dichlorobenzene (50 mg, 0.21 mmol, 1.2 equiv) and potassium carbonate (54 mg, 0.39 mmol, 2.0 equiv). The resulting solution was stirred for 2 h at room temperature. The resulting mixture was diluted with ethyl acetate, washed with brine (2x20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product (80 mg) was purified by Prep-HPLC : Column, SunFire Prep-C18, 19*150mro Sum; mobile phase gradient, water 0.05%TFA : CH 3 CN (35% to 55% C¾C over 10 min; detector. Waters 2.545 UV detector 254/220rmi) to provide 50 mg (63%) of the title compound as a white solid. MS (ES, m/z): 430 [M+H , 1H-NMR (400 MHz, CD 3 OD) δ 7.76 (s, 0.3H), 7.71 (s, 0.7H), 7.57 - 7.46 (m, 2H), 7.41 (d, J = 8.9 Hz, 0.3H), 7.30 - 7.20 (m, 1.711), 7.08 (d, J= 7.8 Hz, 1H), 6.79 (t, J= 7.1 Hz, 0.711), 6.70 - 6.61 (m, 0.3H), 4.67 - 4.44 (m, 2H), 4.03 - 3.92 (m, 1H), 3.84 - 3.71 (m, 1H), 3.71 - 3.53 (m, 2H), 3.52 - 3.35 (m, 2H), 3.22 - 3.10 (m, 1H), 2.53 - 1.74 (m, 5H), 0.99 -· 0.81 (m, -2H), 0.74 ■■■ · 0.46 (m. 2H).

Example 13

fS)-(2-(4-cyciopropy - 23,4 etm^

dichlorophenyl)methanone

Scheme 13: 1 , (S)-methyl piperidine-2-carboxylate, HATU, DIEA, DMF; 2. LiOH, THF, H 2 0; 3. le, HATU, DIEA, DMF.

Intermediate 13a: (S)-metliyl l-(2,5-dichlorobetiz.o l)piperidine-2- carboxylate. To a solution of 2,5-dichlorobeiizoic acid (1.00 g, 5.24 mmol, LOO equiv) in DMF (10 mL)were added (S)-methyl piperidine-2-carboxylate (750 rag, 5.24 mraol, 1.00 equiv), HATU (4.00 g, 10.5 mmol, 2.00 equiv), DIEA (2.74 g, 21.20 mmol 4.00 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated, under reduced pressure. The residue was purified by flash- co.lu.tnr: chromatography with petroleum ether/ethyl acetate (1 :1.) to give 1.45 g (88%) of (S)-methyl l-(2.5-dichlorobeiizoyl)pIperidine-2--carboxylate as yellow oil, MS (ES, m/z) 316 [M+Hj + .

Intermediate 13b: (S)-l-(2 s 5-dic orobenzoyl)piperidine-2-carboxylic acid. To a solution of (S)-methyl l-(2,5-dichlorobenzoyl)piperidine-2-carboxylate (450 mg, 1.42 mmol, 1.00 equiv) in TIIF/water (10/10 mL) was added LiOH ½0 (300 mg, 7.15 mmoK 5.00 equiv). The resulting solution was stirred, overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The pH of the solution was adjusted to 2~3 with hydrogen chloride (1 M). The resulting solution was extracted with ethyl acetate (3x30 tnL). The organic layers were combined, dried over anhydrous sodium sulfate and. concentrated under reduced pressure to give 0.4 g (93%) of (S)- 1 -(2,5-dicUorx)benzoyl)piperidiiie-2-carboxyiic acid as light-yellow oil, MS (ES, m/z): 302 [M+i-]J + .

Example 13: (S)-(2-(4-cyclopropyl-l,2,3,4-teixahydroquinoxaline-l- carbonyl)piperidin- 1 -yl)(2,5-dichloroph.enyl)methanone. To a solutio of (S)~l-(2,5- dichloiObenzoyl)piperidine-2-carboxy].ic acid (130 mg, 0,43 mmol, 1.5 equiv) in DMF (5 tnL) were added le (50 mg, 0.29 mmol, 1.0 equiv), HATU (218 mg, 0.57 mmol, 2.0 equiv) and DIEA (149 mg, 1.15 mmol, 4.00 equiv). The resulting solution was stirred overnight at room temperature, The resulting mixture was concentrated under reduced pressure. The crude product ( 100 mg) was purified by Prep-HPLC : Column, SunFire Prep-Ci8, 19* 150mm Sum; mobile phase gradient, water 0.05%TFA : CI¾CN (75% to 78% CH 3 C over 10 min; detector, Waters 2545 IJV detector 254/220nm) to provide 20 mg (15%) of (S)-(2-(4-cyclopropyl- 1,2,3 , 4-tetrahydroquinoxaline- 1- carbonyl)piperi<mi-l-yl)(2,5-<HcUorophenyl)memanone TFA salt as a light yellow solid. MS (ES, m/zy. 458 | M-H [] . 1H-NMR (300 MHz, CD 3 OD) δ 7.20-7.42 (m, 2H), 6.96-7.17 (m, 3H), 6.65 (t, .7= 7.5 Hz, 1H), 5.70-5.80 (m, 1H), 4.19-4.75 (m, 2H), 3.21- 3.76 (m, 3H), 2,39 (d, J - 3.6 Hz, 1 H), 1.20-1.75 (in, 7H), 0.70-0.79 (m, 2H), 0.53-0,58 (m, 2H).

Example 14

(S -(4-cyciopropyl-3,4-dihYdroqmrioxalme-l(2H)-yl)(l^

2- l) methaoone

Example 14 : (S)-(4-cyclopropyl-3 ,4-dihydroquinoxaiine- 1 (2H)- l)(l - (2,5-dichlorobenzyi) piperidin-2-yl)methanorie. Example 14 was prepared using the procedure described for the preparation of Example 12, except that (S)-l -(t- butoxycarbonyl)piperidme-2-carboxylic acid was used in place of (S)-l- [(benzyloxy)carbor.iyl]pyrrolidine-2-carboxylic acid. Isolated as the bis TFA salt. MS (ES, /z : 444 fM+Hf. 1H-NMR (300 MHz, CD 3 OD) δ 7.81 (s, I E), 7.60-7.53 (m, 2H). 7.30-7.27 (m, 3H), 6.87-6.71 (m, 1 H), 4.85-4.41 (m, 4H), 3.51-3.25 (m, 5H), 2.56- 2,42 (m, 1H), 1.98-1.60 (m, 5H), 1.40-1.20 (m, 1 H), 0.97-0.82 (m, 2H), 0.66-0.45 (m, 2H).

Example IS

( )-(4-cvclopropyl-3,4-dihydroquinoxalin-l(2H)-yl^

2-yi)methaiione

Example 15: (R)-(4-cyclopropyl-3,4-dihydroquinoxalm-l (2H)-yl)(i- (2,5-dicbJorobenzyl) pyrrolidin-2-yl)metharj.one. Example 15 was prepared using the procedure described for the preparation of Example 12 except that (R)-l-(t- butoxycarbonyl)pyrrolidine-2-carboxylic acid was used in place of (S)-l- [(benzyloxy)carbonyl]pyrrolidine-2-carboxylic acid. Isolated as the bis TFA salt. MS (ES, m/z): 430 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.70-7.73 (m, 1H), 7.50-7.54 (m, 2H), 7.24-7.27 (m, 2H), 7.07-7.10 (m, III), 6.79-6.81 (m, 1H), 4.58 (dd, J-25, 13Hz ; 2H), 3.96-4.01 (m, 1H), 3.37-3.84 (ra, 5H), 3.13-3.18 (m, 1H), 1.75-2.58 (m, 5H), 0.53-0.92 (m, 4H). Example 16

(Ry{ 1-cye¾opropyl-3 4-dihydroq

2-yD methanone

Example 16: (R)-(4-cyclopropyl-3 5 4-dihydroquinoxaline-l(2H)-yl)(l- (2,5-dichlorobenzyl) piperidin-2-yl) methanone. Example 16 was prepared using the procedure described for the preparation, of Example 12, except that (R)-l-(t- butoxycarbonyl)piperidine-2-carboxylic acid was used in place of (S)-l- [(benzyloxy)carbonyl]pyrrolidins~2~carboxyIic acid. Isolated as the bis TEA salt. MS (ES, m/z): 444 [M+H] + . Ή-NMR (400 MHz, CD 3 OD) δ 7.77-7.79 (m, IH), 7.53-7.58 (m, 2H), 7.23-7.31 (m, 3H), 6.75-6.90 (m, 1 H , 4.49-4.54 (m, 1H), 4.34-4.39 (m, 3H), 3.47-3.56 (m, 4H), 3.12-3.31 (m, IH), 2.51 (m, I H), 1 .68-1.78(m, 5H), 1.25-1.40 (m, IH), 0.88-0.91 (m, 2H), 0.62-0.65.

Example 17

S)-(4-cyclopropYi-3 ,4-dihydroquinoxaline- 1 (2H)-y 1 )( 1 -(2,5 -dichlorobenzyl)azetidin-2 yl) methanone

Example 17: (S)-(4-cyclopropyl-3 5 4-dibydroquinoxaline-l(2H)-yl)(l- (2,5-dichlorobenzyl) azetidin-2-yl)methan.one. Example 17 was prepared using the procedure described for the preparation of Example 12 except that (S)-l-(t- butoxycarbonyl)azetidine-2-carboxylic acid was used in place of (S)-l- [(benzyloxy)carbonyl]pyrrolidin.e-2-carboxyiic acid. Isolated as the bis TFA salt. MS (ES, m/zy. 416 [M+H] + . ! H-NMR (400 MHz, CD 3 OD) δ 7.61 (s, IH), 7.54 (m.2H). 7,28-7.24 (m, 2H), 6.94 (d, ./ 8S i .. I I I). 6.80 (m, 1H), 5.60 (t, J=9Hz, 1H), 4.53 (d l J= >\, 14Hz, 2H), 4.18 (m, IH), 3.95 (m, IH), 3.83-3.74 (m, 2H), 3.44-3.38 (m, HI), 3.20-3.14 (m, IH), 2.51 (s, I H), 2.43-2.36 (m, 2H), 0.93 (d, J=6Hz, 2H), 0.68-0.60 (m, 2H).

Example 18

(S)-(4-cycio ropyl-3.4-dih droquinoxdine-l(2H)-vl)(l -(2,5- dicMorophenyl)sulfonyl)pyrroMia-2-yl)metfaanone

Example 18: (S)-(4-cyclopropy1-3,4-dihydroquinoxaline-l (2H)-yl)(l-

(2,5-dichloroph.enyl) sulfonyl)pyirolidin-2-yl)methanone. To a solution of 2,5- dicMorobenzene-l-s lfonyl chloride (200 mg, 0.81 inmol, 1.00 equiv) in DCM (10 xnL) were added 12c (220 mg, 0.81 minoL 1.00 equiv) and triethylamme (180 mg, 1.78 mmoL 2, 18 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure, The crude product (320 mg) was purified by Prep-HPLC : Column, SunFire Prep-C18, 19* 150mm Sum; mobile phase gradient, water 0.05%TFA : CH 3 CN (46% to 61% CH 3 CN over 7 min; detector, Waters 2545 UV detector 254/220nm) to provide 191.8 nig (49%) of the title compound TFA salt as a brown solid, MS (ES, m/z): 480 [M+Hf . l H~NMR. (300 MHz, CD 3 OD) δ 7.72 (s, IH), 7.54-7.53 (m, 2H), 7.25-7.16 (m, 2H), 6.98 (d, J-7.5Hz, IH), 6.68-6.62 (m, IH), 5.09-4.96 (m, IH), 4.30-4.19 (m, IH), 3.70-3.64 (m, IH), 3.50-3.35 (m, 4H), 2,53 (s, IH), 2.18-2.01 (m, 3H), 1.88-1 .74 (m, I H), 0.88-0.84 (m, 2H), () .72-0.62(m, 2H). Example 19

(S)-(4-eyclopropyl-3,4-di¼^

dimethylpyrrolidin-2-yl)methanone

Example 19: (S)-(4-cyclopropyl-3 ,4-dihydroqulnoxaline~ 1 (2H)-yl)(l- (2,5-dichlorobenzyl)-4,4-dimethylp>TTo m-2-yl)methanone. Example 19 was prepared using the procedure described for the preparation of Example 12 except that (S)-l-(t- butoxycarbonyl)-4,4~dimethylpyrroIidine-2-carboxyiic acid was used in place of (S)-l- [(benzyloxy) cai'bonyljpyrrolidiiie-2-carboxyiic acid. Isolated as the bis TFA salt. MS (ES, m/z); 458 [M+H] . ; H- MR (300 MHz, CD 3 OD) δ 7.77 (m, IH), 7.53 (m, 2H), 7.27 (m, 2H), 7.05 (m, I H), 6.80 (m, IH), 4.97 (m, IH), 4.59 (m, 2H), 3.70 (m, 2H), 3.56 (m, I H). 3.42 (m, 2H), 3.05 (m, IH), 2.51 (in, IH), 2.13 (rn . I H), 1.75 (m, IH), 1 ,17 (m, 6H), 0.93 (m, 2H), 0.68 (m, H), 0.55 (m, IH).

(S)-(4-cyclopropyl-3,4-dihydroquinoxdine-l(2H)-yl)(l 2,5-dic

methylpyrrolidin-2-yl)niethanone

Example 21): (S)-(4-cyclopropyl-3,4-dihydroquinoxalirie-l(2H)-yl)(l-

(2,5-dichlorobenzyl)-2"methylpyrrolidin-2-yl)methanone. Example 20 was prepared using the procedure described for the preparation of Example 12 except that (S)-l- ((benzyk ) xy)carbonyl)-2-methylpyrrolidine-2-carboxyiic acid was used in place of (S)- l-[(benzyloxy) carbonyl]pyrrolidine-2-carboxylic acid.. Isolated as the bis TFA salt, MS (ES, m/zY. 444 [M+Fff. 1H-NMR (300 MHz, CD 3 OD) δ 7,96 (s, I E), 7.59-7.51 (m, 2H), 7.38 (s, IH), 7.28-7.23 (m, 2H), 6.82-6.76 (m, 1H), 4.72-4.62 (m, IH), 4.41-4.02 (m, 2H), 3.80-3.32 (m, 5H , 2.53-2.14 (m, 4H), 2.10-1 ,95 (m, 111), 1.80-1.38 (m, 3H), 0.95-0.84 (m, 2H), 0.72-0.50 (m, 2H).

Example 21

-(4-cYciopropyl-3,4-dihydroqi inoxalme-l(2H)-ylX3-(2,5- dichk)robenzvi)thi3zolidiR-4-y3)iriethanone

Example 21 ; (R)-(4-cyclopropyl-3,4-dihydroqumoxaime-l (2H)-yl)(3- (2,5-dieMorobenzyl) thiazolidin-4-yl)methanone. Example 21 was prepared using the procedure described for the preparation of Example 12 except that (R)-3-(t- butoxyearbon l)thiazolidine-4-carboxylie acid was used in place of (S)-l- [(benzyloxy)caTbonyl]pyrrolidine-2-carboxylic acid, isolated as the bis TFA salt. MS (ES, m/z): 448 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.40-7.03 (m, 611), 6.70-6.63 (m, 1H), 4.74 (bs, I H), 4.11 (d, J==9.9Hz, 2H), 3.98-3.76 (m, 3H), 3.60-3.36 (111, 4H), 3.22-3.09 (m, IH), 2.49-2.40 (m, IH), 0.85-0,81 (m, 2H), 0.61-0.49 (m, 2H). Example 22

(RH4-cyclopropvl-3,4-d-hydroqumoxalme-l(2H)-yl)(4-(2,

dicMorobenzyl)thiomorpholin-3-yl)methanone

Example 22 : (R)-(4-eycl opropyl-3 ,4-dihydroquinoxaline- 1 (2H)-yl)(4- (2,5-dichlorobenzyl) thiomorpholin-3-yl)metihian.one. Example 22 was prepared using the procedure described for the preparation of Eixampie 12 except that (R)-4-(t- batoxycarbonyi)iliiomorpholine-3-earboxylic acid was used in place of (S)-l - [(benzyloxy) carbonyi]pyrrolidine-2-carboxyiic acid. MS (ES, m/z) 462 [M+H] + . 1H- NMR. (400 MHz, CDCW) δ 7,51 (s, 1H), 7.28-7.25 (m, 2H), 7. Ϊ 9-7.15 (m, 3H), 6.76- 6.72 (m, 1H), 4.21-4.10 (m, 2H), 3.88-3.77 (m, 1H), 3.70-3.62 (m, 1H), 3.61-3.48 (rn, lH), 3.47-3.32 (m, 3H), 2.94-2.60 (m, 2H), 2.53-2.36 (m, 311), 1.60-1.54 (m, 1H), 0.91- 0.80 (m, 2H), 0.68-0.50 (m, 2H).

Example 23

(SV(4-cyeiopropyl-3,4-<l ydroquinoxa^^

3-yi)rnethanone

Example 23 : (S)-(4-cycloprop 1-3 ,4-dihydroquinoxaline- 1 (2 (I- (2,5-dic orobenzyi)-2-metbylp>Trolidin-2-yl)methanone. Example 23 was prepared using the procedure described for the preparation of Example 12 except that (S)-4-(t- Ηιαΐο γο Λοην^ΐΐΐοφΙιοΙίηε-Β-θΕΛοχίΊίο acid was used in place of (S)-l-[(benzyloxy) carbonyljpyiTolidine-2-carboxylic acid. Isolated as the bis TFA salt, MS (ES, m/z): 446 [M+H] + . ¾ H-NMR (400 MHz, CIX¾) δ 7.71 (s, 1H), 7.43-7.35 (in, 211). 7.21 (s, 2H), 6.72 (s, 2111 4.66-4.62 (m, 2H), 4.47 (s, 1 H), 4.39-4.34 (m, 1H), 4.10-3.30 (m, 8H), 3.02 (s, 1H), 2.48-2.45 (m, 1H), 0.95-0.82 (m, 2H), 0.74-0.67 (m, 1H), 0.55-0.46 (m, 1 H).

Exam le 24

(4-cyclopropYl~3,4-dihydroquinoxa3ine-l(2H)-yl)( ' 2S)

azabicycio[3.1.0 lhexan-2- vi imcthanorsc

Example 24 : (4-cyclopropyl-3 5 4-dihydroquinoxalme- 1 (2H)-yl)((2S)-3-

(2,5-dichlorobenzyl)-3-azabicyclo[3.1.0]hexan-2-yi)methan one. Example 24 was prepared using the procedure described for the preparation of Example 12 except that (2S)-3-((benzyloxy)carbonyi)-3- azabicyclo[3.1.0]hexane-2-carboxylic acid (prepared from commercial (2S)-3-azabicyclo[3 J .0]hexane-2-carboxylic acid by the acton of benzyl chlorofomiate under typical Schotten~Bauman.ii conditions) was used, in place of (S)-l-[(benzyloxy) carbonyl]pyrroUdine-2-carboxyiic acid. MS (ES, m/z): 442 [M+H] T . 1 H-NM (300 MHz, CD 3 OD) δ 7.78-7.69 (m, IB), 7.57-7.43 (m, 2H), 7.31-7.20 (m,

2.7H), 7.11-7.04 (m, 0.3H), 6.88-6.78 (m, 0.7H), 6.65-6.56 (m, 0.3H), 5.25 (d, J=4.5Hz, 1H), 4.72-4.54 (m, 2H), 4.37-4.29 (m, 0.7H), 4.08-3.97 (m, 0.3H), 3.95-3.24 (m, 5H), 2.57-2.2S (m, I.3H), 2.08-1.70 (m, 0.3H), 1 .81 -1.70 (m, 0.7H), 1.43-1.32 (m, 0.7H), 1.01-0.42 (ra, 6H). Example 25

(S)-5-(4-cyclopropvl~l,2 ,4-tetra^

dichlorobenzvDpyrrolidin-2-one

Scheme 25: I . 4. 2~(bromomethyl)- l,4-dichlorobenzene, NaH, THF.

Intermediate 25a: (S)-5-(4-cyclopropyl- 1 ,2, ,4-tetrahydroquinoxaline- 1 - carbonyl)-pyrrolidin-2-one. Intermediate 25a was prepared using the procedure described for the preparation of Intermediate 12c except, that (S)- 1 -(t-b toxycarbonyl)- 5-oxopyrrolidine-2-carboxylic acid was used in place of (S)- 1 - [(benzyloxy) carbony]]pyrrolidine-2-carboxylic acid.

Example 25 : (S)-5-(4-cyclopropyl- 1.2,3 ,4-tetrahydroquinoxaline- 1 - carbotiyl)- 1 -(2,5-dicMorobenzyl)pyrrolidin-2-orie. To a mixture of (S)-5-(4- cyclopropyl-1 ,2,3,4-tetrahydroquinoxaline- 1 -carbonyl)-pyrrolidin-2-one 25a (80 mg, 0.28 mmol, 1.0 equiv) in THF (5 mL) was added sodium hydride (33 mg, 0.82 mmol, 3,00 equiv, 60%), followed by addition of 2-(bromomethyl)-l ,4-dichlorobenzene (67 mg, 0.28 mmol, 1 ,00 equiv). The resulting solution was stirred for 2 h at room temperature. The resuitmg solution was diluted with ethyl acetate (10 mL), washed with brine (2x10 mL) and concentrated under reduced pressure. The crude product (50 mg) was purified by Prep-HPLC : Column, SunFire Prep-C18, 19* 150mm Sum; mobile phase gradient, water 0.05%TFA : CH 3 CN (56% to 70% CH 3 CN over 10 mm; detector, Waters 2545 U detector 254 and 220nm) to provide 20 mg (1.6 %) of the title compound TFA salt as a white solid. MS (ES, m/z): 444 [M+H] + . 1H- MR (300 MHz, CD3OD) δ 7.18(m, 51 i h 6.67 (m, 1H), 6.52 (m, 1H), 4.65 (m, 2H), 4.09 (m, 1H), 3.89 (m, Hi), 3.32 (m, 3H), 2.52 (m, III), 2.38 (m, 2H), 2.13 (m, 1H), 1.98 (m, 1 H), 0.76 (m, 2H), 0.51 (m, 211). Example 26

(4 ; yclopropyl-3.4-dihYdroquinoxalin- 1 (-2HVyl)(l -((2,5- dichlorobenzvi)animo)cyclopropyl)inethanone

Scheme 26: 1. le, HATU, DIEA, DMF, rt to 50 °C. 2. 2,5-dichlorobenzyl chloride, NaH, KI, DMF; 3. 4 M HC1 in. 1 ,4-dioxane.

Intermediate 26a: t-Butyl (l-(4-cyclopropyl~ 1,2,3,4- tetrahydroqumoxaline-l -carbonyl) cyclopropyl)carbaraate. To a mixture of Boc-1 - aminocyclopropane-l-carboxylic acid (40 mg, 0.20 mmoi, 1 equiv) and le (34,6 mg, 0.20 mraol, 1 equiv) in DMF (0.5 mL) were added DIEA (173 uL, 1.0 mmol, 5 equiv) and HATU (90.8 mg, 0.24 mmol, 1.2 equiv). The mixture was stirred at room temperature for 1 hr and at 50 °C overnight. The mixture was diluted with ethyl acetate, washed with ¾0 (2x) and brine (Ix), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was purified by flash-column chromatography to give 40 mg (56 %) t-Butyl (1 -(4-cyelopropyl- ί ,2,3,4- tetrahydroquinoxaline-1 -carbonyl) cyclopropyl)carbamate as a clear syrup. MS (E8, m/z): 357.9 [M+H]\

Intermediate 26b: t-Butyl (l-(4-cyclopropyl-i,2,3,4- tetrahydroquinox.aline-1 -carbonyl) cyclopropyl)(2,5-dichlorobenzyl)carbamate. To a solution of t-Butyl (1 -(4-cyclopropyl- 1 ,2,3.4-tetrahydroquinoxaiine-l - carbonyl)cyclopropyl)carbamate (83.7 nig, 0.234 mmol, 1.00 equiv) in DMF (1 ml,) at 0 °C was added 60 % sodium hydride (103 mg, 0.258 mm.ol, 1 .1 equiv). The mixture was stirred at room temperature for 30 minutes and cooled to 0 °C. To ihe mixture was added, a solution of 2,5-dichlorobenzyl chloride (49 uL, 0.35 mmol, 1.3 equiv) in DMF (0.4 mL) and KI (cat.). The mixture was stirred at room temperature for 2 h and 45 °C for 1 h. The reation was quenched with water, extracted with ethyl acetate. The organic layer was washed with brine (1 x)„ dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by flash- column chromatography to give 82 mg (68%) t-Butyl (l-(4-cyclopropyl-l ,2,3,4-tetrahydroquinoxaline-l-carbonyl) cyciopropyl)(2,5-dichiorobenz>'l)carbamate as a yellow syrup. MS (ES, m/z) 515.9 | ··! [] " .

Example 26 : (4-cyclopropyl-3 ,4-dihydroqumoxaim- 1 (2H)-yl)(l -((2,5- dicWorobenzyl)amino) c clopropyl)methanone. To t-Butyl (l~(4~cyejopropyl--l,2,3,4~ tetrahydroquinoxalme- 1-carbonyl) cyc]opropyl)(2,5-dichlorobenzyl)carbamate (82 mg, 0.16 mmol) was added hydrochloric acid (4 M in 1,4-dioxane). The mixture was stirred at room temperature for 1 hour and concentrated. The residue was diluted with ethyl acetate, washed with saturated aqueous NaHCC (lx) and brine (Ix), dried over anhydrous sodium sulfate, concentrated under reduced, pressure, and purified by flash- column chromatography to give 32 mg (48 %) (4-cyclopropyi-3 } 4-dihydroquinoxalin- l(2H)-yl)(l -((2,5-dichloroben¾yl)amino)cyclopropyl)methanone as a clear syrup. MS (ES, m/z): 416 [M÷Hf . 1H-NMR (400 MHz, CD 3 OD) δ 7.33 (dd, J = 7.9, 1 .5 Hz, 1H), 7.26 (d, J - 8.5 Hz, 1H), 7.20 (dd, J ------ 8.3, 1.4 Hz, IE), 7.1 7 (dd, J = 8.5, 2.6 Hz, 1H),

7.12 (ddd, J- 8.4, 7.3, 1 .5 Hz, 1H), 6.87 (s, lH), 6.78-6.71 (m, Hi), 3.89 (t, J = 5.8 Hz, 2H), 3.61 (s, 2I-T), 3.44 (t, J = 5,9 Hz, 2H), 2.46-2.35 (m, 1H), 1.46-1.38 (m, 2H), 0.95 (q, J= 4.3 Hz, 2H), 0.82-0.74 (m, 2H), 0.49-0.41 (m, 2H).

Example 27

(4-cyclopropyl-3.4-dih^^

dich1orobei¾zyl)(methyl)amino)cyclopropyl)met^

Exampl e 27: (4-cyclopropyl-3,4-dih " droquinoxalin- 1 (2H)-yl)(l -((2,5- dichlorobenzyl)(metbyl) ammo)cyclopropyl)methanojie. To a mixture of (4- cyc]opropyl-3 ; 4-dihydroquinox

cyclopropyl)methanone (14.5 mg, 0.035 mmol) in DMF (0.2 ml.,) were added iodomethane (1 .4 uL, 0.23 mmol) and 2CO3 (12.8 mg,.0.093 mmol). The mixture was stirred at 50 °C for 64 hours and purified by Prep-HPLC to give 8 rag (4- cyc!opropyl-3,4 -dihydroquinoxalin-l(2H)~yl)(1 -((2,5-dichiorobenzyl)(methyl)amino) cyclopropyl)methanone bis TFA salt as a yellow syrup. MS (ES, m/z): 430 [M+H] + . *H- NMR. (400 MHz, CDiOD) δ 7,35-7.29 (m, 2H), 7.27-7.22 (m, 2H), 7.18 (d, J = 2.5 Hz, ΓΗ), 7.05 (ddd, J = 8.6, 7.3, 1.5 Hz, IH), 6.70-6.63 (m, 1H), 3.99 (t, J ~ 5.4 Hz, 2H), 3.80 (s, 2H), 3.4.1 (i, J = 5.6 Hz, 2H), 2.49-2.33 (m, 1H), 2.19 (s, 3H), 1.29 (dd, J = 7.6, 5.3 Hz, 2H), 1.15 (dd, J= 7.7, 5.2 Hz, 2H), 0.88-0.77 (ra, 2H), 0.64-0.53 (m, 2H).

Example 28

2-(2-cMoroben-ryl)pyrrolidm-l-y yl-3,4-dihy

Example 28: 2 -(2-chlorobenzy Dpyrrolidin- 1 -y i)(4-cyclopropyl-3 ,4- dihydroqumoxal.in (2H)-yI)me anone. To a solution of triphosgene (22.7 mg, 0.(577 mmol, 1 equiv) in DCM (1.3 mL) at 0 °C was added a solution of le (40 mg, 0.23 mmol, 3 equiv) and triethylaniine (40 uL, 0.29 mraol 3.7 equiv) in DCM (1 mL). The mixture was stirred at room temperature for 2 h. To the mixture were added 2-(2- chloro-benzyl)-pyrrolidine (54 nig, 0.276 mmol, 3.6 equiv) and triethylamine (42 iiL, 0.299 mmol, 3.9 equiv). The mixture was stirred at room temperature for 1 h, coneentrated under reduced pressure, and purified by flash-colun chromatography to give 62.8 mg (69 %) as yellow syrup. MS (ES, m/z): 396 [M+Hf . 1H- MR (400 MHz, CDCi 3 ) δ 7.33 (dd, J = 7.4, 1,8 Hz, 1 H), 7.28-7.20 (m, 1H), 7.1 7-7.12 (m, 3H), 6.94 (dd, J = 12.1, 4.6 Hz, 2H), 6.68 (td, J= 7.5, 1 ,4 Hz, 1H), 4.35-4.26 (m, 1H), 4.20-4.06 (m, 1H), 3.49-3.31 (m, 3H), 3.32-3.05 (m, 3H), 2.80 (dd, J = 12.9, 9.5 Hz, 1H), 2.47- 2.31 (m, 1H), 1.82-1.74 (m, 2H), 1.70-1.56 (m, 2H), 0.86-0.76 (m, 2H), 0.76-0.66 (m, 1H), 0.57-0.43 (m, 1 1 i).

exam le l

(4-cyclopropyl-3.4-dihydroquin.oxaliit-1 (2H)-yl)(2-(2,5-dichlorophenoxy)cyclohex-l- erryOmethanone

Scheme 29: 1. Tf 2 0, DIEA, DCM; 2. K 2 C0 3 , DMF; 3. LiOH»¾0, 1,4-dioxane, H 2 0; 4. le, HATU, DIEA, MeC .

Intermediate 29a: ethyl 2-(trifluoromethylsulfonyl.oxy)eyclohex-l- enecarboxylate. Ethyl 2-oxocyclohexanecarboxyla.te (170 mg, 1 .00 mmol) and DIEA (417 jjJL, 2,40 mmol) were dissolved in DCM (2 mL) and cooled to -78 °C. To the stirring solution was added dropwise trifluoromethanesuiionic anhydride (202 μΤ, 1.20 mmol), then the resulting solution was allowed to warm to room temperature and stirred for 16 h. The solution was then diluted with DCM and washed with I M aqueous HCl and the solvent removed under reduced pressure. The crude residue was purified by flash column ehroniatograpliy using a gradient of hexanes : EtOAc (9 : 1 to 1 : 1) to give 29a (231 mg, 76%) as a clear oil. 1 H~NMR (400 MHz, CDC1 3 ) δ 4.23 (q, J = 7.1 Hz, 2H), 2.51-2.40 (m, 2H), 2.40-2.31 (m, 2H), 1.81-1.70 (m, 2H), 1.70-1.57 (m, 2H), L28 (t, J= 7.1 Hz, 3H).

intermediate 29b: ethyl 2-(2,5-dic orophenoxy)eyclohex- 1 - enecarboxylate. 29a (174 mg, 0.576 mmol), 2 C0 3 (279 mg, 2.02 mmol). and DMF (2 mL) were combined and the resulting suspension stirred at 120 °C for 2 h. The suspension was diluted with MeOH and filtered, then the solvent removed under reduced pressure. The crude residue was purified by flash column chromatography using a gradient of hexanes : EtOAc (95 : 5 to 75 : 25) to give 29b (108 mg, 59%). ! H- NMR (400 MHz, CDC1 3 , mixture of rotamers) δ 7.29 (d, J= 8.5 Hz, 1H), 7.19 (d, J = 2.4 Hz, 0.33H), 7.00 (dd, J = 8.5, 2.4 Hz, 0.33H), 6.94 (dd, J = 8.5, 2.3 Hz, 0.67H), 6.85 (d, J= 2.3 Hz, 0.67H), 4.18 (m, 0.67H), 4.07 (q. J = 7.1 Hz, 0.33H), 2.46 (m, 2H), 2.22 (m, 2H), 2.19-1.92 (m, 2H), 1.80-1.55 (m, 4H), 1.26 (t, J = 7.1 Hz, 1H), 1.08 (t J = 7.1 Hz, 2H).

Intermediate 29c: 2-(2,5-dichlorophenoxy)cyclohex-l-enecarboxylic acid. 29b (108 mg, 0.343 mmol) and LiOH*H 2 0 (115 mg, 2.74 mmol) were dissolved in EtOH (2 mL) and H 2 .0 (1 mL) and stirred at 80 °C for 1 h. The solvent was removed under reduced pressure and the resulting residue dissolved in DCM and washed with 5% aqueous HCl, then the sol vent removed to give 29c (23 mg, 23%) as a clear oil. ¾ NMR (400 MHz. CDCL , mixture of rotamers) δ 7.34 (d, J = 8.6 Hz, 0.67H), 7.30 (d, J - 8.6 Hz, 0.33H), 7.19 (d, j = 2.4 Hz, 0.33H), 7.09 (dd, J= 8.6, 2.4 Hz, 0.67H), 7.04 (d, J= 2.4 Hz, 0.33H), 7.03 - 6.99 (m, 0.67H), 2.47 (t, J= 6.1 Hz, 1.34H), 2.14 (m, 2.68H), 1.79 - 1.59 (m, 4.02H).

Example 29: (4-cyclopropyi-3,4-dihydroquinoxalin-l(2H)-yl)(2-(2,5- dichlorophenoxy)cyclohex-l-enyl)methanone. 29c (23 mg, 0.080 mmol), le (17 mg. 0.096), HATU (34 mg, 0.088 mmol), and DIEA (56 μΐ,, 0.32 mmol) were dissolved in MeCN (1 mL). The solution was stirred at room temperature for 1 h, then a single crystal of DMAP added and the solution stirred and additional 2 h at room temperature. The solution was then heated for 2 h at 60 °C, then purified by preparative HPLC with a C18 silica gel stationary phase using a gradient of H 2 0 0.05% TFA : CH 3 CN 0.05% TFA (50 : 50 to 5 : 95) and detection by UV at 254 nm to give the title compound (3.2 mg, 9%) TFA salt as a yellow powder. MS (ES, m/z): 443 [M+l] + . 1H-NMR (400 MHz, ( Ί he ') ! ) ) δ 7.29 (d, J - 8.4 Hz, 1H), 7.1 1 (t, J= 7.1 Hz, 1H), 7.03-6.84 (m, 4H), 6.69 (t, J= 7.1 Hz, 1H), 4.63 (s, 1H), 3.02 (s, 1H), 2.66 (s, 1H), 2.43 (s, 1H), 2.11 (s, 1H), 1.84- 1.57 (m, 4H), 1.36-1.27 (m, 2H), 0.94-0,83 (m, 2.H), 0.73-0.38 ( n, 4H).

Example 30

(SH2H-benzorbiri.41tMaziti-4(3^

yijmethanorte

Scheme 30: L a. 3,4-dihydro-2FI-benzo[b][l ,4]thiazine, DIEA, DCM; b. HBr/AcOH; c. L4-dichloro-2-(chloromethyl)benzene, DIEA, MeCN.

Example 30: (S)-(2H-berizo[bj[L4]thiazin-4(3H)-yl)(l-(2,5- dicWorobenz l)pyirolidin-2-yl)methanone. 3,4-dihydro-2H-benzo[b][l ,4]thiazine (67 mg, 0.44 mmol) and DIEA (209 Ε, 1.21 mmol) were dissolved, in DCM (1 mL) and cooled to 0 °C. To the stirring solution was added drop wise, a solution of (S)-benzyl 2- (chlorocarbonyl)pyrrolidme-l-carboxylate (108 mg, 0.402 mmol) in DCM (1 mL). The resulting solution was then allowed to warm to room temperature and stirred for 1 h, The solvent was removed under reduced pressure and then further dried in vacuo. The crude residue was dissolved in 33 wt % hydrobromic acid in acetic acid solution and left at room temperature for 1 h, then triturated with Et 2 0 and the solvent decanted to give an oil. The crude oil was dissolved in MeCN, to which l,4~dichioro-2- (chloromethyl)benzene(94 mg, 0.48 mmol) and DiEA (349 μί, 2.01 mmol). The solution was stirred at room temperature for 16 h, then purified by flash column chromatography using a gradient of hexan.es : EtOAc (9 : 1 to 3 : 2) to give the title compound (72 mg, 44%). MS (ES, m/z): 407 [M+lf , 5 H NMR (400 MHz, CDC1 3 ) δ 7,34 (d, J = 8.6 Hz, 0.67H), 7.30 (d, J = 8.6 Hz, 0.33H), 7.19 ( t J = 2.4 Hz, 0.33H), 7.09 (del, J = 8.6, 2.4 Hz, 0.67H), 7.04 (d, J = 2.4 Hz, 0.33H), 7.03-6.99 (m, 0.67H), 2.47 (t, J = 6.1 IIz, 1.34H), 2.14 (m, 2.68H), 1.79-1.59 (m, 4.02H).

Example 31

l-f4-CYclopro yl-l,2 ,4-tetrahyckoqi hioxalin-l-vl)

dimethylpropan- 1 -one

Scheme 31 : 1. a. 3-bromo-2,2-dimethylpropanoic COC% DCM: b. TEA, DCM: 2. 2.5-dichlorophenol K 2 C0 3 , DCM,

Intermediate 31a: 3-bromo-l-(4-cyclopropyl-l,2 ; 3,4- tetrahydroquinoxalin-l-yi)-2 ; 2-dimethyipropan~l-orie. To a stirred 0 °C solution of 3- bromo-2,2~dimetbylpfopanoic acid (120 mg, 0.660 mmol, 1.16 equiv) in dichloromethane (10 mL) was added oxalyl chloride (2.0 niL) drop-wise. The resulting solution was stirred, for 2 h at room temperature then concentrated under reduced pressure to provide a residue of the acid chloride used in the next step without additional purification. The acid chloride residue in dichloromethane (10 mL) was added to a stirred 0 °C solution of 1-cyclopropyi-l ,2,3,4-tetrahydroquinoxaline (100 mg, 0.57 mmol, 1.0 equiv) and triethylamine (87 mg, 0.86 mmol, 1.5 equiv) in dichloromethane (10 mL). The resulting solution, was stirred for 4 h at room temperature then diluted with. ¾0 (20 mL) and extracted with dichloromethane (3x20 mL). The combined organic extraci was washed, with brine (2x20 mL) and dried over anhydrous sodium sulfate then concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography using an eluent of ethyl acetate/petroleum ether (Ϊ i5) to provide the product (150 mg, 77%) as pink oil. MS (ES.

Example 31: l-(4-cycloprppyl-l,2 ,4 e1xahydroquinoxalin-l-yl)-3-(2,5- dichIorophenoxy)-2,2-diinethylpropan-l-one. A solution of 2,5-dichlorophenol (60 mg, 0.37 mmol, 1.0 equiv), 3 -bromo- 1 -(4-eyclopropyl- 1 ,2,3 ,4-te1rahy<koquinoxalin- 1 - I)- 2,2-dimethyipropan- 1 -one (80 mg, 0.24 mmol, 0.64 equiv) and potassium carbonate (64 mg, 0.46 mmol, 1.26 equiv) in N.N-dimethylformamide (4 mL) was stirred for 5 h at 50 °C. The resulting solution was diluted with H 2 0 (10 mL) and extracted with ethyl acetate (3x20 mL). The combined organic extract was washed with brine (1x20 mL) and dried over anhydrous sodium .sulfate then concentrated under reduced pressure. The crude product (100 nig) was purified by preparative HPLC with a Cjg silica gel stationary phase using a 40 min gradient (¾0 0.05% TFA: C¾CN 0.05% TFA 95 : 5 to 0 : 100%) and detection by UV at 254 nm to provide TFA salt of the title compound (53 mg, 34%) as a yellow solid. MS (ES, m/z) 419 [M+H . 1H-NMR (300 MHz, CD 3 OD) 8 7.35 (d, J 8.4 Hz, I Fi), 7.20-7.23 (m, 1H), 7.08-7.14 (m, 2H), 6.91-6.95 (m, 1H), 6.67-6.76 (m, 2H), 3.90 (s, 3H), 3.83 (t, ./ - 5.7 Hz, 2H), 3.43 (t, J = 6.0 Hz, 2H), 2.31-2.35 (m, 1H), 1.40 (s, 3H). 0.70-0.76 (m, 2H), 0.33-0.38 (m, 2H).

Example 32

l-cyclQpropyi-4 il~f2. -dic

ol, K.2CO 3 , Ki, DMF.

Interrnediate 32a: 1 -(hydroxymet yl)cyclopropane- 1 -carboxyiic acid. A solution of potassium hydroxide (1.90 g, 33.9 mmol, 2.00 equiv) and ethyl 1- bromocyciobutane-l-carboxylate (3.50 g, 16.9 mmol, 1.00 equiv) in water (60 mL) was stirred overnight at 30 °C. The reaction mixture was cooled on ice and the pH value of the solution was adjusted to 1 with concentrated HCl, then concentrated under reduced pressure. The resulting residue was dissolved, in methanol (50 mL), solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The resulting residue was applied onto a silica gel column and eluted with a dichloromethane/methanol mobile phase gradient (100:1 to 20:1) to provide 1.8 g

(92%) of the product as a white solid.

Intermediate 32b: l-(chloromethyl)cyciopropane-l -carbon yl chloride. 1 -

(hydroxymethyl)cycIopiOpane-l-carboxylic acid (650 mg, 5.60 mmol, 1 ,00 equiv) was dissolved in ihionyl chloride (8 mL) and stirred for 5 h at 80 °C in an oil bath. The resulting reaction mixture was concentrated under reduced pressure to provide 680 mg (79%) of the product as a light yellow oil

mtermediate 32c: !-[[] -(chloromethyl)cyclopropyl]carbonylj-4 cyclopropyl-l ,2,3,4-tetrahydroq«inoxaline. To a stirred solution of 1-cyclopropyl-

125 1,2,3,4-tetrahydroquinoxaline (850 mg, 4.88 mmol, 1.1.0 equiv) and triethylamine (900 mg, 8.89 mmol, 2.00 equiv) in dichloromethane (10 mL) was added dropwise a solution of l-(chloromethyl)cyclopropane-] -carbonyi chloride (680 mg, 4.44 mmol, 1.00 equiv) in dichloromethane (2 mL). The resulting reaction mixture was stirred overnight at room temperature and then concentrated under reduced pressure. The resulting residue was applied to a silica gel column and etuted with a mobile phase gradient of ethyl acetate/petroleum ether (1 :15-1:1) to provide 500 mg (39%) of the product as a light yellow oil.

Example 32: l -cyclopropyi-4-f[l-(2,5- dichlorophenoxymethyl)cyciopropyl]carbonyl3-l ,2,3,4-tetrahydroquinoxaIine. A solution of 2,5-diehiorophenoi (60 mg, 0.37 mmol, 1.2 equiv). l-[[i- (cWoromethyl)cyclopropyl]carto

(89.2 mg, 0.31 mmol, 1.0 equiv), potassium carbonate (85.6 mg, 0.62 mmol, 2.0 equiv) and Kl (5.0 mg, 0.03 mmol, 0.10 equiv) in t V,N-dimethylform amide (3.0 mL) was stirred overnight at 65 °C in an oil bath. Solids were removed from the reaction mixture by filtration and the filtrate concentrated under reduced pressure. The crude product (50 mg) was purified by preparative HPLC with the following conditions: Column, SunFire preparative CIS. 19* 150mm 5μοι; Mobile phase gradient, water containing 0.05% TFA : CH 3 CN (30:70 to 15:85 over 10 min then to 100% over 1 min); Detector, Waters 2545 UV detector at 254 and 220nm to provide 12.4 mg (10%) of the title compound trifluoroacetate salt as a white solid. MS (ES, m/z): 417 [M+H] + ; 1H- MR (300 MHz, CD 3 OD) 6 7. 1-7.41 (m, 2H), 7.06-7.14 (m, 2H), 6.90-6.94 (m, 1H), 6.68-6.73 (m. 3H), 6.60-6.61 (m, 11T), 3.86-3.90 (m, 2H), 3.68 (s, 2H), 3.32-3.39 (m, 2H), 2.20-2.29 (m, 1H), 1.36-1.40 (m, 2H) ; 0.96-0.99 (m, 2H), 0.63-0.69 (m. 2H), 0.16-0.21 (m, 2H).

Example 33

3-[2,5-dichloro-4-(ri-r(4-cyclopropvl-1 3,4 efaahvdroqumo alin-l- vl)carbonyl]cvclopropvl m.ethoxy phenyi ropanoic acid

Scheme 33: 1. tert-butyl aerylate, PdCPPh 3 ) 4? TEA, DMF; 2. Rh/C, H 2 , EtOAc; 3. ,¾rt-butyi 3-(2,5-dichloro-4-hydroxyphenyl)propanoate, K 2 C0 3 , i, DMF; 4, TMSBr, DCM.

Intermediate 33a: tent-butyl (2E)-3-(2,5-dichloro-4-hydroxypheiiyl)prop- 2-enoate. A stirred solution of 4-bromo-2,5-dichJ.orophenol (10.0 g, 41.3 mmol, 1.00 equiv), tert-but l. prop-2-enoate (5.00 g, 39.0 mmol 1 .00 equiv), triethylanime (8.30 g, 82.0 mmol, 2.00 equiv) and Pd(PP]¾)4 (2.00 g, 1.73 mmol, 0.05 equiv) was purged and maintained under an inert atmosphere of nitrogen then heated overnight at 1 10 °C in an oil bath. The resulting reaction mixture was diluted with. 200 mL of dichloromethane washed with brine (2x100 mL) and the combined organic phase concentrated under reduced pressure. The resulting residue was applied to a silica gel column and eiuted with ethyl acetate/petroleum ether (1 : 10) to provide 8 g (67%) of the product as a yellow solid.

Intermediate 33b: terf-butyl 3-(2,5-dichloro-4- hydroxyphenyl)propanoate. To a stirred solution of tert-butyl (2E)-3-(2,5-dichloro-4- hydroxyphenyl)prop-2-enoate (8.0 g, 27.7 mmol, 1.00 equiv) and Rhodium on Carbon (8,0 g) in ethyl acetate (50 mL) was introduced hydrogen gas. The resulting reaction mixture was stirred overnight at 25 °C solids were removed by fitration and the titrate concentrated under reduced pressure. The residue was applied onto a silica gel column with and. eiuted with a mobile phase of ethyl acetate/petroleum ether (1 :10) to provide 7 g (87%) of the product as a white solid. MS (ES, m/z): 289 [M-H] " ; 1H-NMR (300 MHz, CDCI3) 5 7.20 (s, IE), 5,69 (d, J= 18 Hz, 1H), 2.92 (m, 2H), 2.52 (m, 2H), 1.43 (s, 9H).

Intermediate 33e: ½r -butyl 3-[2,5 » dicWoro-4-([l- (4-cycloprcpyl- l,2,3.4-tetrahydroquinoxalin-l-yl)carhon^ propanoate. A solution of ferZ-butyl 3-(2.5-dichloro-4-h droxyphenyl)propanoatc (539 mg, 1.85 mmol, 1.20 equiv), 1 -[[1 -(chloromethy eyclopropy!jcarbony 1] -4-cyclopropyl- 1 ,2,3,4- tetrahydroqu ttoxaline (450 mg, 1.55 mmol, 1.00 equiv), potassium carbonate (426 mg, 3.08 mmol, 2.00 equiv), Kl (24.9 mg, 0.15 mmol, 0.10 equiv) in N,N~ dimethylformamide (8 mL) was stirred overnight at 70 °C in an oil bath. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (4x20 mL) and the organic layers combined. The combined organic phase was washed with brine (2x20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was applied to a silica gel column and eluted with a mobile phase of ethyl acetate/petroleum ether (1 :20-1: 1) to provide 710 mg (84%) of the product as a yellow solid.

Example 33: 3-[2,5-dichloro-4-([l-[(4-cyclopropyl-l ,2,3,4-tetrahydro- quinoxalin-l-yl)carbonyl]cyclopropyl] methoxy)phenyl]propanoic acid. To a stirred 0 °C solution of iert-butyl 3-[2,5-dichloro-4-([l-[(4-cyclopropyM tetrahydroquinoxalin-l~> )carfao (120 mg, 0.22 mmol, 1.00 equiv) in dichloromethane (5.0 mL) was added TMSBr (4.0 mL) dropwise. ' " Hie resulting reaction mixture was allowed to warm to room temperature, stirred for 1.5 h, then was quenched by the addition of dichloromethane/methanol (10:1). The resulting mixture was concentrated under reduced pressure, the crude residue (100 mg) was purified by Preparative HPLC under the following conditions: Column, SiinFire preparative C18, 1 * 150mm 5μιη; Mobile phase gradient, water containing 0.05% TFA : C1¾CN (26:74 to 9:91 over 6 min then to 100% over 1 min); Detector, Waters 2545 UV detector at 254 and 220nm. This resulted in 13.5 mg (13%) of the title compound trill uoroacetate salt as a white solid. MS (ES, m/z): 489 [M+H] + ; Ή-NMR (300 MHz, CD 3 OD) δ 7.35-7.39 (m, 2H), 7.04-7.14 (m, 2H), 6.67-6.73 (m, 1H), 6.58 (s, 1H), 3.85-3.89 (m, 2H), 3.67 (s, 2H), 3.36-3.40 (in, 2H), 2.90-2.95 (m, 211), 2.54-2.59 (m, 2H), 2.19-2.23 (m, 1H), 1.37-1.40 (m, 2H), 0.94-0.98 (m, 2H), 0.63- 0,69 (m, 2H), 0.11-0.16 (m, 2H),

Example 34

3-f 2.5-diehloro-4-f [1 - ( ' 4-cyclopro yl- 12 ,4 ettahydroqiiinoxalin-l -

pentahydroxyhexyljpropanaixxide

Scheme 34: 1. (2R3R,4R,5S)-6-(meihyk¾iiino)hexa.ne-] .2 ,4,5-peiitaol 5 HATU DIEA, DMF.

Example 34: l-Cyclopropyl-4-([l-[(isoquinolm-5-yloxy)metb.yl]cyclo- propyl]caxbonyl)-l ,2,3,4-tetrahydroquinoxaline. A solution of 3-[2,5-dichioro-4-([l-

[ (4-cyclopropyl -1,2,3 ,4-tetrahydroqumoxaIin- 1 - yl)cai¾onyl]cyclopropyl]meihoxy)ph.eiiyl]~propanoic acid (200 nig, 0,41 mmol, 1.0 equiv), HATU (232 mg, 0.61 mmol, 1.5 equiv), DIEA (78. S rng, 0.61 mmol, 1.50 equiv) and (2R,3R J 4R 5 5S)-6-(methy]amino)hexane-l 5 2,3,4,5-peiitol ( 119.1 mg, 0,61 mmol, 1.50 equiv) in ¾A<~dimemyliomiamide (10 mL) was stirred overnight at room temperature. The resulting reaction mixture was diluted with brine (30 mL) and extracted with ethyl acetate (4x30 mL) and the organic layers combined. The combined organic phase was washed, with brine (20 mL), dried over sodium sulfate and concentrated under reduced pressure. The crude product (200 mg) was purified by- preparative HPLC under the following conditions: Column, SunFire preparative CI 8, 19* 150mm 5pm; Mobile phase gradient, water containing 0.05% TFA : CFI3CN (56:44 to 38:62 over 6 min then to 100% over 1 min); Detector, Waters 2545 UV detector at 254 and 220nm to provide 55.3 mg (20%) of the title compound, trifluoroacetate salt as a white solid. MS (ES, m/z): 666 [M+H] + ; ' l l-.N R (300 MHz, CD 3 OD) 5 7,36-7.40 (m, 2H), 7.06-7.1 1 (m, 2H), 6.68-6.70 (m, 1H), 6.60-6.61 (m, ].H), 3.60-4.00 (m, 1 1FI), 3,31-3.38 (m, 3H), 3.01-3.31 (m, 2H), 2.87-2.97 (m, 4H), 2.63-2.81 (m, 2H), 2,24 (s, 1H), 1.37 (m 5 2H), 0.95-0.99 (m, 2H) 5 0.64-0.69 (m, 21 U. 0.16 (m, 21 E).

((S)- 1 -(2.5-dichlorobenzyl pyrro1idin-2-v (4-methyl-3.4-dihYdroqranolin- 1 (2JD- vDmethanonc

Example 35: ((S)-l-(2,5-dich[orobenzyl)pyrrolidiii-2-yl)(4-metJ yi-3,4- ' dihydroquinolin- 1 (2H)-yl)methanone. 35 was synthesized in an analogous fashion to Example 10, using 4-met yl-l ? 2,3 !( 4-tetrahydroquinoline in place of 1,2,3,4- tetrahydtoquinoline. Isolated as the TFA salt. MS (ES, fn/z): 403 M+H] + .

pie 3Ό

H2,5-dicMorobenzyloxy)-N-^-^

38a

Scheme 36: 1. a. TMSCH 2 N 2 , DCM, MeOH; b. 1 ,4-Dichloro-2- (chloromethyl)benzene, Nail, .DMF: c. LiOH*H 2 0, ¾0, 1,4-dioxane; 2. 2-Methoxy-N- methylaniline, HATU, DIE A, MeCN. intermediate 36a: 1 -(2,5-dichlorobeiizy[oxy)cyclopropa.nccarboxyIic acid. 1-hydroxyeyclopropanecarboxylic acid (204 mg, 2.00 mniol) was dissolved in DCM (2.5 mL) and MeOH (0.5 mL). To the stirring solution was added 2.0 M TMSCH 2 N 2 in Et 2 0 (1.1 mL, 2.2 mmol) dropwise, the resulting solution stirred for 10 mm at room temperature. The solvent was removed under reduced pressure and the resulting residue dissolved in DMF (1 mL), followed by addition of 60% NaH in mineral oil dispersion (120 mg) and the resulting suspension stirred for 5 min at room temperature. l ,4-Dichioro-2-(chlorometh S)henzene (583 mg, 3.00 mmol) was added and the suspension stirred at room temperature for 16 h. The suspension was then quenched with 5% a.q, HQ and extracted with EtOAc. The organic layer was washed with HbO and brine, then dried over N S0 4 and. the solvent removed under reduced pressure. The crude residue was purified by flash column chromatography using a gradient of hexanes : EtOAc (100 : 0 to 80 : 20). The resulting oil was dissolved in ¾0 (2 mL) and 1,4-dioxane (4 mL). then LiOH*H 2 0 (133 mg, 3.18 mmol) added, and the solution stirred for 1 h at room temperature, then 1 h at 50 °C. The solutio was concentrated under reduced pressure, then diluted with 5% aq. HC1 and extracted with EtOAc. The organic layer was then washed with brine and dried over Na?.S04, then the solvent removed under reduced pressure to give 36a (99 mg, 19%) as a white solid. l H NMR (400 MHz, CDCI 3 ) 7.52 (d, J= 2.5 Hz, IH), 7.29 - 7.27 (m, Iff), 7.22 - 7.19 (m, 1H), 4.77 (s, 3H), 1.53 - 1 ,48 (m, 211). !..41 - .1.36 (ra. 2H).

Example 36: 1 -(2,5-dichlorobenzyloxy)- -(2-methoxyphenyi)-N- methylcyelopropanecarboxamide. Intermediate 36a (24 mg. 0.092 mmol), 2-methoxy- N-methylaniline (16 mg, 0.11 mmol), HAITI (38 mg, 0.10 mmol), and DIEA (64 \sL, 0.37 mmol) were all combined in MeCN (1 mL) and stirred for 24 h at room temperature. The solution was then purified by preparative HPLC with a CIS silica gel stationary phase using a gradient of H 2 0 0.05% TFA : CH3CN 0.05% TFA (70 :30 to 5 : 95) and detection by U V at 254 nm to give the title compound (18 mg, 52%) as a while solid. MS (ES, m/zy. 380 [M+if. 1H-NMR (400 MHz, CDCI 3 ) δ 7.30-7.22 (m, 1H), 7.22-7.13 (m, 2H), 7.09 (d, J = 8.3 Hz, 1H), 6.88 (t, J- 7.5 Hz, I H), 6.68 (<L J = 8.5 Hz, 1H), 6.40 (s, IH), 4.49 (d, J = 13.6 Hz, 1H), 4.31 (d, J = 14.0 Hz, 1H), 3.43 (s, 3H), 3.21 (s, 3H), 1.58-1.46 (m, IH), 1.36-1.24 (m, 2Π). 1.09-0.96 (m, IH), 0.92-0.81 (m, IH)..

Example 37

i2,5-diehlorobenzy^

methylcvclopropanecar oxamide

36a 37 36a. 2-itiethoxy-N-TnethylpyrIdin-3-a.mine, HATU, DIEA, MeCN.

Example 37: l-(2,5-dichlorobenzyloxy)-N-(3-methox.ypyridiii-2-yl)--N- methylcyclopropanecarboxamide. The title compound was prepared in the same manner as Example 36, using 2-methoxy-N-methylpyridm-3-amine in place of 2-meihoxy-N- racthylaniiine to give 37 (8%) as the TFA salt. MS (ES, m/z): 3 1 [M+l] + . 1H-NMR (400 MHz, CDCI 3 ) δ 7.94 (dd, J= 4.9, 1.4 Hz, 1H), 7.24 (dd, J= 8.2, 4.9 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 7.10 (td, J = 8.8, 2.0 Hz, 2H), 6.39 (s, IE), 4.40 (s, 2H), 3.64 ( s , 3H), 3.30 (s, 3H), 1.52 (s, 2H), 1.02 (s, 2H).

1 -(2,5 -di . chl orobenzylamino )-N-(2-methox vphenyl) -N- methy lc y clopro panecar boxamide

Example 38: 1 -(2,5-dichlorobenz> aiiimo)-N-(2-metlioxyphenyl)-N- me ylcyclopropanecarboxamide. The title compound was prepared in the same manner as 27. using 2-methoxy-N-methylani ine in place of le. Isolated as the TFA salt. MS (ES, m/z): 378 [M+l] + . Ή-NMR. (400 MHz, CDCI 3 ) δ 7.44-7.40 (m, lH), 7.39 (d, j = 2.2 Hz, 1H), 7.37-7.33 (m, 1H), 7.31-7.27 (m, 1H), 7.14-7.11 (m, 1H), 7.06-6.99 (m, 2H), 4.23 (s, 2H), 3.83 (s, 3H), 3.21 (s, 3H), 1.41 (s, 2H), 1.03 (s. 1H), 0.97-0.85 (m, 1.H). 2 071251

Example 39

( ' 8)-N~(2-icyelopropvim

carboxamide

Scheme 39: I . Cyclopropylmethanol, NaH, THF; 2. Pd/C, H 2 , EtOAc; 3. (Boc) EtOH; 4. Mel, NaH, DMF; 5. IF A, DCM; 6. (S)-benzyl 2-(chlorocarbonyi)pyrrolidme. 1-carboxylate, Ei. 3 N, DCM; 7. Pd/C, ¾, MeOH; 8. 2-(broniomethyl)-l,4 dichlorobenzene, K?C€>3, DMF. Intermediate 39a: l-(cyciopropylmethoxy)-2-nitrobenzene. Cyclopropylmethanol (2.55 g. 35.4 mraol) was dissolved in tetrahydrofiiran (50 mL). NaH 60% dispersion in mineral oil (1 .70 g) was added t to the stirring solution in several batches at 0 °C and the mixture was stirred for 1 h. l-Fluoro-2 -nitrobenzene (5.00 g, 35.4 mmol) was then added and the resulting mixture stirred for 2 h at 80 °C. The reaction was then quenched by the addition of 30 mL of water and then extracted thrice with ethyl acetate. The organic layers were combined and dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure to give 39s (6.5 g, 95%) as a brown oil. MS (ES, rn/z): 193 I M r .

Intermediate 39b: 2-(cyciopropylmemoxy)aniline. 39a (6.50 g, . 33.6 mmol) and palladium on carbon (6.5 g) was dissolved in ethyl acetate (50 mL). The suspension was stirred, overnight at room temperature under an atmosphere of ¾. The suspension was filtered, and the filtrate concentrated under reduced pressure to give 39b (5.68 g), as a red oil, which was used without further purification. MS (ES, m z): 164 [M+H .

Intermediate 39c: tert-butyl N-[2-

(cyclopropylmethoxy)phenyl] carbamate. 39b (5.68 g, 34.8 mmol) was dissolved in in ethanol (50 mL), followed by addition of di-tert-butyl dicarbonate (9.12 g, 41.8 mmol) in several batches. The resulting solution was stirred for 3 h at room temperature, then the solvent removed under reduced pressure to give 39c (9 g, 98%) as a red oil.

Intermediate 39d: tert-butyl N-[2-(cyclopropylmethoxy)phenyi]-N- methylcarbamate. 39d (9.00 g, 34.2 mmol) was dissolved in N,N-dimethylformamide (150 mL), followed by the addition of NaH 60% dispersion in mineral oil (2.1 g) in several batches at 0 °C. The mixture was stirred for 1 h at room temperature, then iodomethane (9.70 g, 68.3 mmol) added and the resulting solution stirred overnight at room temperature. The solvent was then removed under reduced pressure and the resulting residue dissolved in ethyl acetate and washed with H 2 0 and brine. The organic layer was then dried over anhydrous sodium sulfate and concentrated under reduced pressure and the residue purified by normal-phase flash column chromatography, using ethyl acetate : petroleum ether (1 : 50) as eluent to give 39d 8.0 g (84%) as a red oil.

Intermediate 39e: 2-(cyclopropylmethoxy)-N-methylaniline. 39d (2.00 g, 7.21 mmol) was dissolved in dichloromethane (3 mL) and trifluoroacetic acid (3 mL). The was stirred for 1 h at room temperature, then the solvent removed under reduced pressure. The resulting residue was dissolved in DCM and washed with saturated aqueous NaHC0 3 . The aqueous layer was then extracted thrice with DCM and the organic layers combined and dried over anhydrous sodium sulfate solvent removed under reduced pressure to give 39e (870 mg, 68%) as a red oil. MS (ES, m/z): 178 [M+H] + .

Intermediate 39f: Benzyl (2S)-2-(carbonochloridoyl)pyrrolidine-l- carboxylate (260 mg, 0.970 mmol) and triethylamine (202 mg, 2.00 mmol) were dissolved in DCM (4 mL). To this solution was added 39e (177 mg, 1.00 mmol) and the resulting solution stirred for 3 h at room temperature. The reaction was then quenched by the addition of 10 mL of water and the mixture extracted thrice with DCM and the organic layers combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 39f (400 mg, 99%) as a yellow oil. MS (ES, m/z): 409 [M+H] + .

Intermeditate 39g: (2S)-N-[2-(cyclopropylmethoxy)phenyl]-N-methyl- pyrrolidine-2-carboxamide. 39f (380 mg, 0.93 mmol) and palladium on carbone (400 mg) was added to methanol (5 mL). The resulting suspension was stirred for 2 h at room temperature under an atmosphere of H 2 . The suspension was filtered and the filtrate concentrated under reduced pressure to give 39g (200 mg, 78%) as a colorless oil. MS (ES, m/z): 275 [M+H] + .

Example 39: (S)-N-(2-(cycl opropylmethoxy)phenyl)- 1-(2,5- dichlorobenzyl)-N-methylpyrrolidine-2-carboxamide. 39g (200 mg, 0.73 mmol), 2- (bromomethyl)-l,4-dichlorobenzene (176 mg, 0.73 mmol), and potassium carbonate (203 mg, 1.47 mmol) were dissolved in N,N-dimethylformamide (4 mL). The resulting solution was stirred overnight at room temperature, then diluted with 20 mL of water and extracted thrice with ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate, and the solvent removed under reduced pressure. The crude residue was purified by normal-phase flash column chromatography, using ethyl acetate : petroleum ether (1 : 8) to give Example 39 (100 mg. 32%) as a yellow oil. MS (ES, m/z): 433 [M+H] + . 1H-NMR (300MHz, CD 3 OD, ppm): 7.46 (d, J = 2.4Hz, 0.6H), 7.34 (d, J = 2.7Hz, 0.4H), 7.15-6.97 (m, 3.6H), 6.88-6.78 (m, 1.6H), 6.67-6.61 (m, 0.8H), 3.71-3.59 (m, 3H), 3.41 (d, J= 14.6Hz, 0.4H), 3.22 (d, J= 14.6Hz, 0.6H), 3.02-2.96 (m, 2.2H), 2.93-2.80 (m, 2H), 2.72 (m, 0.6H), 2.13 (m, 0.4H), 1.87 (m, 0.6H), 1.73-1.52 (m, 2.6H), 1.52-1.35 (m, 1.4H), 1.10-0.84 (1.2H), 0.42-0.32 (m, 0.8H), 0.32-0.18 (m, 1.2H), 0.14-Ό.06.

Example 40

N-(2-(cvclopropylmethoxy phenyl)-l-( ' 2,5-dichlorobenzyloxy)-N- methylcvclopropanecarboxamide

40 Example 40: N-(2-(cyclopropylmethoxy)phenyl)-l-(2,5- dichlorobenzyloxy)-N-methylcyclopropanecarboxamide. 40 was synthesized in an analogous fashion to Example 36, using 39e in place of 2-Methoxy-N-methylaniline. MS (ES, m/z): 420 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.30-7.18 (m, 4H), 6.92 (t, J= 7.5 Hz, 1H), 6.75 (d, J= 7.8 Hz, 1H), 6.43 (s, 1H), 4.44 (dd, J= 13.8, 26.7 Hz, 2H), 3.56-3.50 (m, 1H), 3.32-3.22 (m, 4H), 1.51 -1.50 (m, 1H), 1.49-1.46 (m, 1H), 1.32-1.04 (m, 2H), 0.95-0.89 (m, 1H), 0.55-0.52 (m, 2H), 0.24-0.16 (m, 2H).

Example 41

1 -cyclopropyl -4-( F ΐ - [ (2 , 5 -dichlorophenvDmethoxyl c yclobutyl] carbonylV 1,2,3,4- tetrahydroquinoxaline

41

Example 41: l-cyclopropyl-4-([l-[(2,5-dichlorophenyl)methoxy]- cyclobutyl]carbonyl)-l,2,3,4-tetrahydroquinoxaline. 41 was synthesized in an analogous fashion to Example 8, using cyclobutanone in place of cyclopentanone. Isolated as the TFA salt. MS (ES, m/z): 431 [M+H] + . 1H-NMR (400 MHz, CD 3 OD) δ 6.56-7.55 (m, 7H), 4.02-4.46 (m, 2H), 3.86 (t, J= 5.6 Hz, 2H), 3.36 (t, J= 5.6 Hz, 2H), 2.75-2.81 (m, 2H), 2.38 (m, 3H), 1.82-2.19 (m, 2H), 0.04-0.92 (m, 4H).

Example 42

(l-f5-chlQro-2-(trifluoromethyl)benzyloxy)cyclopropyl (4-cvclopropyl-3,4- dihydroquinoxalin- 1 f 2H)-yl)methanone

Example 42: (l-(5-chloro-2-(trifluoromethyl)benzyloxy)cyclopropyl)(4- cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)methanone. Example 42 was synthesized in an analogous fashion to Example 9, using 2-(bromomethyl)-4-chloro-l- (trifluoromethyl)benzene in place of 2-(bromomethyl)-l,4-dichlorobenzene. Isolated as the TFA salt. MS (ES, m/z): 451 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.53 (d, J= 9 Hz, 1H), 7.32 (m, 2H), 7.07 (m, 1H), 6.95 (d, J= 9 Hz, 1H), 6.75 (m, 1H), 6.59 (s, 1H), 4.42 (s, 2H), 3.85 (t, J = 6 Hz, 2H), 3.33 (t, J= 6 Hz, 2H), 2.24-2.17 (m, 1H), 1.45 (m, 2H), 1.16-1.12 (m, 2H), 0.61-0.56 (m, 2H), 0.01 (m, 2H).

Example 43

l- (l-[[2-chloro-5-(trifluoromethyl)phenyl1methoxylcyclopropync arbonyl1-4- cyclopropyl- 1 ,2,3 ,4-tetrahydroquinoxaline

Example 43: (l-(5-chloro-2-(trifluoromethyl)benzyloxy)cyclopropyl)(4- cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)methanone. 43 was synthesized in an analogous fashion to Example 9, using 2-(bromomethyl)-l-chloro-4- (trifluoromethyl)benzene in place of 2-(bromomethyl)-l,4-dichlorobenzene. Isolated as the TFA salt. MS (ES, m/z): 451 [M+H] + . 1H-NMR (400 MHz, CD 3 OD) δ 7.64 (s, 2H), 7.33 (d, J= 4 Hz, 1H), 7.01-6.92 (m, 3H), 6.72 (t, J= 8 Hz, 1H), 4.45 (s, 2H), 3.93 (t, J = 4 Hz, 2H), 3.41 (t, J= 4 Hz, 2H), 2.27-2.25 (m, 1H), 1.54-1.48 (m, 2H), 1.31-1.21 (m, 2H), 0.65 (m, 2H).

Example 44

(4-cyclopropyl-3 ,4-dihydroquinoxalin-l (2H)-yl)( 1 -(2,6- dichlorobenzyloxy)cyclopropyl)methanone

Example 44: (4-cyclopropyl-3,4-dihydroquinoxalin-l (2H)-yl)(l-(2,6- dichlorobenzyloxy)cyclopropyl)methanone. Example 44 was synthesized in analogous fashion to Example 9, using 2-(bromomethyl)-1.3-dichlorobenzene place of 2-(bromomethyl)-l,4-dichlorobenzene. Isolated as the TFA salt. MS (ES, m/z): 417 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.41-7.32 (m, 3H), 7.29 (m, 1H), 7.28-7.22 (m, 1H), 7.05-6.99 (m, 1H), 6.68-6.62 (m, 1H), 4.90 (s, 2H), 4.10 (t, J= 6 Hz, 2H), 3.41 (t, J = 6 Hz, 2H), 2.43-2.38 (m, 1H), 1.20 (s, 4H), 0.86-0.81 (m, 2H), 0.65-0.60 (m, 2H).

Example 45

3-(Yl -f4-cyclopropyl- 1 ,2,3.4-tetrahvdroquinoxaline-l - carbonyl)cvclopropoxy methy benzonitrile

Example 45: 3-((l-(4-cyclopropyl-l,2,3,4-tetrahydroquinoxaline-l- carbonyl)cyclopropoxy)methyl)benzonitrile. Example 45 was synthesized in an analogous fashion to Example 9, using 3-(bromomethyl)benzonitrile in place of 2- (bromomethyl)-l,4-dichlorobenzene. Isolated as the TFA salt. MS (ES, m/z): 374 [M+H] + . 1H-NMR (400 MHz, CD 3 OD) δ 7.55 (d, J = 8 Hz, 1H), 7.37 (t, J = 8 Hz, 211), 7.23 (s, 1H), 7.18-7.10 (m, 2H), 6.92 (m, 1H), 6.77-6.73 (m, 1 H), 4.41 (s, 2H), 3.94 (t, J = 4 Hz, 2H), 3.40 (t, J = 8 Hz, 2H), 2.34-2.29 (m, 1H), 1.44 (m, 2H), 1.19- 1.17 (m, 2H), 0.73-0.65 (m, 2H).

Example 46

(S -(l- 5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-2-yl)(4-cyclop ropyl-3,4- dihydroquinoxaline- 1 (2H)-yl)methanonc

Example 46: (S)-(l-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-2- yl)(4-cyclopropyl-3,4-dihydroquinoxaline-l(2H)-yl)methanone. Example 46 was prepared using the procedure described for the preparation of Example 12 except that 2- (bromomethyl)-4-chloro-l-(trifluoromethyl)benzene was used in place of 2- (bromomethyl)-l,4-dichlorobenzene. Isolated as the bis TFA salt. MS (ES, m/z): 464 [M+H] + . Ή-NMR (300 MHz, CD 3 OD) δ 8.01 (s, 1H), 7.69-7.81 (m, 2H), 7.24 (s, 2H), 7.04-7.07 (m, 1H), 6.78-6.81 (m, 1H), 4.56-4.87 (m, 2H), 3.99 (m, 1H), 3.17-3.77 (m, 6H), 2.47 (t, J= 4.8 Hz, 1H), 1.83-2.18 (m, 4H), 0.87-0.91 (m, 2H), 0.50-0.66 (m, 2H).

Example 47

fS (4-cvclopropyl-3,4-dihydroquinoxaline-l(2H)-yl)( " l-(2,6-dich1orobenzyl s )pyrrolidin-

2-yl) methanone

Example 47: (S)(4-cyclopropyl-3,4-dihydroquinoxaline-l(2H)-yl)(l- (2,6-dichlorobenzyl) pyrrolidin-2-yl) methanone. Example 47 was prepared using the procedure described for the preparation of Example 12 except that 2-(bromomethyl)- 1,3-dichlorobenzene was used in place of 2-(bromomethyl)-l,4-dichlorobenzene. MS (ES, m/z): 430 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.50-7.56 (m, 3H), 7.12 (m, 2H), 7.10-7.12 (m, 1H), 6.60-6.81 (m, 1H), 4.98 (m, 1H), 4.78 (m, lH), 3.33-3.85 (m, 6H), 3.12-3.14 (m, 1H), 1.90-2.51 (m, 5H), 0.88-0.92 (m, 2H), 0.56-0.69 (m, 2H).

Example 48

3-(2,5-dichloro-4-((( ' S)-2-(4-cvclopropyl- 1.2.3, 4-tetrahvdroquinoxaline- lcarbonvn yrrolidin-l-vnmethvnphenv -N-metliyl-N-f S.SR^R.SR^^J^.S.e- pentahydroxyhexyDpropanamide

Scheme 48: 1. Cr0 3 , HO Ac, Ac 2 0, H 2 S0 4 ; 2. (2-(t-butoxy)-2- oxoethy1)triphenylphosphonium bromide, NaOH, H 2 0, DCM; 3. NaBH 4 , MeOH; 4. Rh/C, H 2 , EtOAc; 5. PPh 3 , NBS, DCM, THF; 6. 12c, K 2 C0 3 , CH 3 CN; 7. TMSBr, DCM; 8. N-methylglucamine, HATU, DIEA, DMF.

Intermediate 48a: 2,5-Dichloroterephthalaldehyde. To a mixture of acetic acid (300 g, 5.00 mol, 25.0 equiv), acetic anhydride (600 g, 5.88 mol, 29.4 equiv) and sulfuric acid (90.0 g, 899 mmol, 4.50 equiv, 98%) at 0-10 °C was added 1,4- dichloro-2,5-dimethylbenzene (35.0 g. 200 mmol, 1.00 equiv), and followed by addition of chromium trioxide (60.0 g, 600 mmol, 3.00 equiv) in several batches over 2 h. The resulting solution was stirred for 4 h at room temperature, and then quenched by the addition of 2000 mL of crushed ice. The resulting solution was extracted with 3x1000 mL of ethyl acetate and the organic layers were combined and concentrated under reduced pressure to give a solid. The solid was added to a mixture of ethanol (300 mL), water (300 mL), and sulfuric acid (30 mL), and the mixture was heated to reflux for 3 h and then cooled. The solids were collected by filtration. The solid was purified by column with ethyl acetate/petroleum ether (1 :30) to give 12 g (30%) of 2,5- dichloroterephthalaldehyde as a white solid.

Intermediate 48b: tert-Butyl 3-(2,5-dichloro-4-formylphenyl)acrylate. To a solution of 2,5-dichloroterephthalaldehyde (10.0 g, 49.3 mmol, 1.00 equiv) in dichloromethane (200 mL) at 0 °C was added (2-(tert-butoxy)-2- oxoethyl)triphenylphosphanium bromide (15.9 g, 34.8 mmol, 0.70 equiv), and followed by addition of a solution of sodium hydroxide (9.9 g, 0.25 mol, 5.00 equiv) in water (14.5 mL) dropwise with stirring. The resulting solution was stirred for 1 h at 0 °C. The resulting mixture was concentratedconcentrated under reduced pressure. The residue was purified by column with ethyl acetate/petroleum ether (1 :200-1:30) to give 9.8 g (66%) of tert-butyl 3-(2,5-dichloro-4-formylphenyl)acrylate as a white solid.

Intermediate 48c: tert-butyl 3-[2,5-dichloro-4-(hydroxymethyl)- phenyljacrylate. To a solution of tert-butyl 3-(2,5-dichloro-4-formylphenyl)prop-2- enoate (6.9 g, 22.9 mmol, 1.00 equiv) in methanol (100 mL) was added NaBH 4 (1.60 g, 42.3 mmol, 2.00 equiv). The resulting solution was stirred for 1 h at room temperature. The reaction was then quenched by the addition of 10 mL of water and concentratedconcentrated under reduced pressure. The resulting mixture was diluted with 100 mL of brine, extracted with ethyl acetate (2x200 mL). The organic layers were combined, dried over sodium sulfate and concentratedconcentrated under reduced pressure. The residue was purified by column with ethyl acetate/petroleum ether (1 :200- l :30to provide 6.3 g (91%) of tert-butyl 3-[2,5-dichloro-4- (hydroxymethyl)phenyl]acrylate as colorless oil.

Intermediate 48d: tert-butyl 3-[2,5-dichloro-4-(hydroxymethyl)phenyl]- propanoate. To a solution of tert-butyl 3-[2,5-dichloro-4- (hydroxymethyl)phenyl] aery late (600 mg, 1.00 equiv) in ethyl acetate (20 mL) was added Rh/C (600 mg). The resulting solution was stirred overnight under H 2 at room 1251 temperature. The solids were filtered out. The resulting mixture was concentratedconcentrated under reduced pressure. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :30) to give 500 mg (82%) of tert-butyl 3-[2,5-dichloro-4-(hydroxymethyl)phenyl]propanoate as light brown oil. Ή NM (300 MHz, DMSO) δ 7.52 (s, 1H), 7.40 (s, 1H), 5.51 (t, J = 5.7 Hz, 1H), 4.51 (d, J= 5.7 Hz, 2H), 2.90 (t, J= 7.4 Hz, 2H), 2.59 - 2.51 (m, 2H), 1.37 (s, 9H).

Intermediate 48e: tert-butyl 3-[4-(bromomethyl)-2,5-dichlorophenyi]- propanoate. To a solution of tert-butyl 3-[2,5-dichloro-4- (hydroxymethyl)phenyl]propanoate (800 mg, 2.62 mmol, 1.00 equiv) in dichloromethane/ tetrahydrofuran (5/5 mL) at 0 °C were added NBS (1034 mg, 5.81 mmol, 2.00 equiv) and PPh 3 (888 mg, 3.39 mmol, 1.20 equiv). The resulting solution was stirred for 2 h at room temperature. The resulting solution was diluted with ethyl acetate (30 mL), washed with brine (2x20 mL), dried over sodium sulfate, and concentratedconcentrated under reduced pressure. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :200-1:20) to give 600 mg (62%) of tert-butyl 3-[4-(bromomethyl)-2,5-dichlorophenyl]propanoate as a white solid.

Intermediate 48f: (S)-tert-butyl 3-(2,5-dichloro-4-((2-(4-cyclopropyl- 1 ,2,3,4-tetrahydroquinoxaline-l -carbonyl)pyrrolidin-l -yl)methyl)phenyl)propanoate. To a solution of tert-butyl 3-[4-(bromomethyl)-2,5-dichlorophenyl]propanoate (100 mg, 0.27 mmol, 1.00 equiv) in C¾CN (2 mL) were added 12c (110 mg, 0.41 mmol, 1.50 equiv) and potassium carbonate (75 mg, 0.54 mmol, 2.00 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparative TLC with ethyl acetate/petroleum ether (1 :3) to give 50 mg (33%) of (S)-tert-butyl 3-(2,5-dichloro-4- ((2-(4-cyclopropyl - 1 ,2,3 ,4-tetrahydroquinoxaline- 1 -carbonyl)pyrrolidin- 1 - yl)methyl)phenyl)propanoate as light brown oil. MS (ES, m/z): 558 [M+Hj + .

Intermediate 48g: (S)-3-(2,5-dichloro-4-((2-(4-cyclopropyl-l ,2,3,4- tetrahydroquinoxaline-l -carbonyl)pyrrolidin-l-yl)methyl)phenyl)propanoic acid. To a solution of (S)-tert-butyl 3-(2,5-dichloro-4-((2-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline-l-carbonyl)pyrrolidin-l-yl)methyl)phen yl)propanoate (50 mg, 0.090 mmol, 1.00 equiv) in dichloromethane (2 mL) was added TMSBr (1 mL). The resulting solution was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in 20 mL of ethyl acetate, washed with brine (2x10 mL), and concentrated under reduced pressure to provide 50 mg (crude) of (S)-3-(2,5-dichloro-4-((2-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline- 1 -carbonyl)pyrrolidin- 1 -yl)methyl)phenyl)propanoic acid as light brown oil

Example 48: 3-(2,5-dichloro-4-(((S)-2-(4-cyclopropyl-l,2,3,4- tetrahydro-quinoxaline- 1 carbonyl)pyrrolidin- 1 -yl)methyl)phenyl)-N-methyl-N- ((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl)propanamide. To a solution of (S)-3-(2,5- dichloro-4-((2-(4-cyclopropyl-l,2,3,4-tetrahydroquinoxaline- l-carbonyl)pyrrolidin-l- yl)methyl)phenyl)-propanoic acid (50 mg, 0.10 mmol, 1.0 equiv) in DMF (2 mL) were added (2R,3R,4R,5S)-6-(methylamino)hexane-l,2,3,4,5-pentol (26 mg, 0.13 mmol, 1.5 equiv), HATU (50 mg, 0.13 mmol, 1.50 equiv), and DIEA (23 mg, 0.18 mmol, 2.0 equiv). The resulting solution was stirred overnight at room temperature. The solids were filtered out. The crude product (50 mg) was purified by Prep-HPLC : Column, SunFire Prep-Cl 8, 19* 150mm 5um; mobile phase gradient, water 0.05%TFA : CH 3 CN (38% to 50% CH 3 CN over 8 min; detector, Waters 2545 UV detector 254/220nm) to provide 8.7 mg (13%) of 3-(2,5-dichloro-4-(((S)-2-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline- 1 carbonyl)pyrrolidin- 1 -yl)methyl)phenyl)-N-methyl-N- ((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl)propanamide bis TFA salt as a white solid. MS (ES, m/z): 679 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.72 (dd, J = 1 1.8, 5.2 Hz, 1H), 7.56 (s, 1H), 7.41 - 7.19 (m, 2H), 7.15 - 7.01 (m, 1H), 6.86 - 6.75 (m, 0.6H), 6.72 - 6.59 (m, 0.4H), 4.53 (dd, J = 34.6, 14.2 Hz, 2H), 4.24 - 4.06 (m, 0.8H), 4.04 - 3.89 (m, 1.2H), 3.87 - 3.56 (m, 7H), 3.54 - 3.36 (m, 3H), 3.25 - 3.16 (m, 1H), 3.16 - 2.89 (m, 5H), 2.86 - 2.64 (m, 2H), 2.63 - 2.22 (m, 2H), 2.19 - 1.83 (m, 3H), 1.81 - 1.56 (m, 1.2H), 1.44 - 1.23 (m, 0.8H), 1.02 - 0.80 (m, 2H), 0.75 - 0.42 (m, 2H).

2012/071251

Example 49

(2S -N-i2-cyclobutoxyphenylVl-r(2,5-(^chlorophenyl metliyl1-N-metliylpyrrolidine-2- carboxamide

Example 49: (2S)-N-(2-cyclobutoxyphenyl)-l-[(2,5- dich1orophenyl)methyl]-N-methylpyrrolidine-2-carboxamide. 49 was synthesized in an analogous fashion to Example 39, using cyclobutanol in place of cyclopropylmethanol. MS (ES, m/z): 433 [M+H] + . Ή-NMR (400 MHz, DMSO-<¾) δ 7.63 (s, 1H), 7.26-7.46 (m, 4H), 6.84-7.00 (m, 2H), 4.70-4.72 (m, 1H), 3.53-3.80 (m, 2H), 3.16-3.32 (m, 1H), 3.01-3.16 (m, 4H), 2.37-2.51 (m, 3H), 1.60-1.82 (m, 8H).

Example 50

f 4-cyclopropyl-3 ,4-dihydroquinoxalin- 1 (2H)-yl (3 -f 2,5-dichlorobenzyloxy¼zetidin-3 - ypmethanone

Scheme 50: 1. TMSCN, Znl 2 , THF; 2. AcOH, HC1; 3. (Boc) 2 0, K 2 C0 3 , THF, H 2 0; 4. le, HOAT, EDCI, DFM; 5. 2-(bromomethyl)-l,4-dichlorobenzene, NaH, DMF; 6. HC1, 1,4-dioxane.

Intermediate 50a: tert-butyl 3-cyano-3-(trimethylsilyloxy)azetidine-l- carboxylate. tert-Butyl 3-oxoazetidine-l-carboxylate (10.0 g, 58.4 mmol), trimethylsilanecarbonitrile (8.68 g, 87.5 mmol, and diiodozinc (1.86 g, 5.83 mmol) were dissolved in tetrahydrofuran (100 mL). The resulting solution was stirred overnight at room temperature, then concentrated under reduced pressure. The residue was dissolved in 300 mL of ethyl acetate and washed twice with 5% sodium bicarbonate and once with H 2 0. The organic layer was dried over anhydrous sodium sulfate and solvent removed under reduced pressure to give the title compound (11.8 g, 75%) as yellow oil, which was used without further purification.

Intermediate 50b: 3-hydroxyazetidine-3-carboxylic acid hydrochloride. 50a (11.8 g, 43.6 mmol) was added to acetic acid (20 mL), then concentrated hydrogen chloride (20 mL) was added dropwise with stirring at 0 °C. The resulting solution was stirred for 4 h at 1 10 °C, then the solvent removed under reduced pressure to give the title compound (6.6 g, 98%) as a yellow solid. MS (ES, m/z): 118 [M+H] + .

Intermediate 50c: l-(tert-butoxycarbonyl)-3-hydroxyazetidine-3- carboxylic acid. 50b (6.06 g, 43.0 mmol) was dissolved in tetrahydrofuran / H20 (60 / 60 mL), followed by the addition of potassium carbonate (18.0 g, 129 mmol) at 0 °C and di-tert-butyl dicarbonate (10.3 g, 47.2 mmol. The resulting mixture was stirred overnight at room temperature, then concentrated under reduced pressure. The pH value of the solution was adjusted to between 3 and 4 with aqueous HC1 (3 M). The resulting solution was extracted four times with ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude residue was re-crystallized from petroleum ether / ethyl acetate (10 / 1) to give the title compound (4 g, 43%) as a white solid.

Intermediate 50d: tert-butyl 3-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline-l-carbonyl)-3-hydroxyazetidine-l-carbo xylate. 50c (900 mg, 4.14 mmol), le (150 mg, 0.86 mmol, 1.00 equiv), EDCI (248 mg, 1.29 mmol), and HO AT (176 mg, 1.29 mmol) were dissolved in N,N-dimethylformamide (3 mL) and the resulting solution was stirred for 4 h at room temperature. The soluution was then diluted with of ethyl acetate (20 mL) and washed thrice with brine, dried over sodium sulfate, and the solvent removed under reduced pressure. The crude residue was purified by normal-phase flash column chromatography, using a gradient of ethyl acetate : petroleum ether (1 : 50 to 5 : 1) to give the title compound (150 mg, 47%) as a brown solid. MS (ES, m/z): 374 [M+H] + .

Intermediate 50e: tert-butyl 3-(4-cyclopropyl-l, 2,3,4- tetrahydroquinoxaline-l-carbonyl)-3-(2,5-dichlorobenzyloxy)a zetidine-l-carboxylate. 50d (160 mg, 0.43 mmol) was dissolved in N,N-dimethylformamide (5 niL). To the stirring solution was added 60% dispersion of sodium hydride in mineral oil (34.2 mg) in several batches at 0 °C. The mixture was stirred for 20 min at 0 °C, then 2- (bromomethyl)-l,4-dichlorobenzene (124 mg, 0.52 mmol) was added and the resulting mixture was stirred for 1 h at room temperature. The mixture was diluted with ethyl acetate (10 mL) and washed thrice with brine, then dried over anhydrous sodium sulfate and the solvent removed under reduced pressure. The crude residue was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 10 min gradient (CH 3 CN : ¾0 0.05% TFA 50 : 50 to 70 : 30) and detection by UV at 254 nm to provide the title compound TFA salt (18.1 mg, 8%) as a white solid. 1H-NMR (400MHz, CO 3 OO,ppm): 7.61-7.59(m, 1H), 7.35-7.15(m, 2H), 7.09-6.90(m, 2H), 6.85- 6.73(m, 2H), 4.58-4.17(m, 4H), 4.10-4.06(m, 1H), 3.88-3.81(m, 2H), 3.77-3.73(m, 1H), 3.50-3.42 (m, 2H), 2.50-2.35(m, 1H), 1.46(s, 9H), 0.81-0.72(m, 2H), 0.57-0.50(m, 1H), 0.25-0. l l(m, 1H). MS (ES, m/z): 532 [M+H] + .

Example 50: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(3-(2,5- dichlorobenzyloxy)azetidin-3-yl)methanone. 50e (100 mg, 0.19 mmol) was dissolved in 1,4-1,4-dioxane (1.5 mL), then concentrated HC1 (0.5 mL) was added at 0 °C. The resulting solution was stirred for 1 h at room temperature, then the pH value of the solution was adjusted to 9 with sodium carbonate. The resulting solution was extracted thrice with ethyl acetate and the organic layers combined and washed with brine. The organic layer was dried over anhydrous sodium sulfate and solvent removed under reduced pressure. The crude residue was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 10 min gradient (CH 3 CN : H 2 0 0.05%o TFA 52 : 48 to 100 : 0) and detection by UV at 254 nm to provide the title compound bis TFA salt (9.6 mg, 12%) as a white solid. MS (ES, m/z): All [M+H] + . TI-NMR (400 MHz, CD 3 OD) δ 7.62-7.56 (m, 1H), 7.40-6.93 (m, 5H), 6.73-6.69 (m, 1H), 4.58-4.36 (m, 2H), 4.22-4.11 (m, 2H), 3.87-3.65 (m, 4H), 3.50-3.41 (m, 2H), 2.40-2.30 (m, 1H), 0.81-0.72 (m, 2H), 0.60-0.40 (m, 1H), 0.30-0.20 (m, 1H).

Example 51

(4-cvc1opropyl-3,4-dihvdroquinoxalin-K2H -yl¥3-(2.5-dichlorobenzyloxy)-l- methylazetidin-3 -vDmethanone

Scheme 51: 1. Formaldehyde, NaBH 3 CN, AcOH, MeOH.

Example 51: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(3-(2,5- dichlorobenzyloxy)-l-methylazetidin-3-yl)methanone. 50 (50 mg, 0.12 mmol) and acetic acid (70 mg, 1.17 mmol) were dissolved in methanol (2 mL), followed by addition of 37% aqueous formaldehyde (30 mg, 0.37 mmol). The mixture was stirred for 1 h at room temperature, then NaBH 3 CN (21 mg, 0.33 mmol was added and the solution was stirred for 1 h at room temperature. The solvent was removed under reduced pressure, then the residue dissolved in dichloromethane (10 mL) and washed with washed saturated aqueous NaHC0 3 . The organic phase was dried over anhydrous sodium sulfate and the solvent removed under reduced pressure. The crude residue was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 10 min gradient (CH 3 CN : H 2 0 0.05% TFA 44 : 56 to 100 : 0) and detection by UV at 254 nm to provide the title compound bis TFA salt (19.6 mg, 38%) as a brown solid. MS (ES, m/z): 446 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.73-7.58 (m, 0.6H), 7.48-7.26 (m, 2H), 7.25-7.14 (m, 1H), 7.12-6.96 (m, 1.7H), 6.92 (s, 0.7H), 6.75 (t, J = 7.4 Hz, 1H), 4.65 (s, 1H), 4.44 (s, 2H), 3.89 (t, J= 5.9 Hz, 1.4H), 3.69 (s, 0.6H), 3.45 (t, J= 6.1 Hz, 1.4H), 3.11-2.86 (m, 3.6H), 2.48-2.23 (m, 1H), 0.90-0.78 (m, 0.6H), 0.79-0.65 (m, 1.4H), 0.63-0.49 (m, 0.6H), 0.24 (s, 1.4H). Example 52

(4-cyclopropyl-3.4-dihydroquinoxalin-lf2H^^

vDmethanone

Example 52: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(3-(2,5- dichlorobenzyloxy)-l-ethylazetidin-3-yl)methanone. 52 can be synthesized in a similar manner as 51, substituting acetaldehyde for formaldehyde. Isolated as a bis-TFA salt. MS (ES, m/z): 460 [M+H] + ; 1H-NMR (300 MHz, CD 3 OD) δ 7.71-7.59 (m, 0.6H), 7.48- 7.14 (m, 3H), 7.14-6.93 (m, 1.7H), 6.92-6.65 (m, 1.7H), 4.74-4.09 (m, 5H), 3.89 (t, J = 5.7 Hz, 1.3II), 3.71 (s, 0.7H), 3.51-3.24 (m, 2H), 2.46-2.23 (m, 1H), 1.37-1.13 (m, 3H), 0.91-0.77 (m, 0.7H), 0.70 (d, J= 5.3 Hz, 1.3H), 0.55 (s, 0.7H), 0.19 (s, 1.3H).

Example 53

r4-cvclopropyl-3^-dihvdroquinoxalin-l( " 2H)-yl r3-(2,5-dichlorobenzyloxy)-l- iso ropylazetidin-3-yl)methanone

Example 53 : (4-cyclopropyl-3 ,4-dihydroquinoxalin- 1 (2H)-yl)(3 -(2,5- dichlorobenzyloxy)-l-isopropylazetidin-3-yI)methanone. 53 can be synthesized in a similar manner as 51, substituting acetone for formaldehyde. Isolated as a bis TFA salt. MS (ES, m/z): 474 [M+H] + . Ή-NMR (300 MHz, CD 3 OD) δ 7.73-7.60 (m, 0.6II), 7.48- 7.14 (m, 3H), 7.13-6.90 (m, 1.7H), 6.88-6.60 (m, 1.7H), 4.79-4.51 (m, 3H), 4.51-4.20 (m, 3H), 3.89 (s, 1.3H), 3.72 (s, 0.7H), 3.56-3.39 (m, 2H), 2.48-2.20 (m, 1H), 1.43-1.15 (m, 6H), 0.92-0.77 (m, 0.7H), 0.67 (d, J= 5.5 Hz, 1.3H), 0.56 (s 5 0.7H), 0.14 (s, 1.3H). Example 54

1 -(3 -(4-cyclopropyl- 1 ,2,3 ,4-tetrahydroquinoxaline- 1 -carbonyl)-3 -(2,5- dichlorobenzyloxy azetidin- 1 -vDethanone

Scheme 54: 1. Acetic anhydride, triethylamine, DCM.

Example 54: l-(3-(4-cyclopropyl-l ,2,3,4-tetrahydroquinoxaline-l- carbonyl)-3-(2,5-dichlorobenzyloxy)azetidin-l-yl)ethanone. 50 (70 mg, 0.16 mmol), acetic anhydride (18 mg, 0.18 mmol), and triethylamine (49 mg, 0.48 mmol) were dissolved in DCM (2 mL) and stirred for 1 h at room temperature. The solution was then diluted with DCM and washed with brine, then dried over anhydrous sodium sulfate and the solvent removed under reduced pressure. The crude residue was purified by preparative HPLC with a C 18 silica gel stationary phase using a 7 min gradient (CH 3 CN : H 2 0 0.03% NH 4 OH 51 : 49 to 68 : 32) and detection by UV at 254 nm to provide the title compound (14.5 mg, 19%) as an off white solid. MS (ES, m/z): 474 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.45-7.32 (m, IH), 7.31-7.12 (m, 2H), 7.06- 6.82 (m, 2H), 6.60-6.52 (m, 2H), 4.57-4.39 (m, 2H), 4.39-4.16 (m, 2H), 4.16-4.03 (m, IH), 3.73-3.53 (m, 3H), 3.34-3.21 (m, 2H), 2.18-2.1 1 (m, IH), 1.73-1.68 (m, 3H), 0.73- 0.60 (m, IH), 0.48-0.33 (m, IH), 0.20-0.10 (m, IH).

Example 55

(4-cvclopropyl-3^-dihvdroquinoxalin-U2H)-vn(3-( ' 2,5-dichlorobenzyloxy - 1 - (methylsulfonyl¼zetidin-3-yl methanone

Scheme 55: 1. MsCl, triethylamine, THF.

Example 55: (4-cyclopropyl-3,4-dihydroquinoxalin-l (2H)-yl)(3-(2,5- dichlorobenzyloxy)-l -(methylsulfonyl)azetidin-3-yl)methanone. 50 (50 mg, 0.12 mmol) and triethylamine (35 mg, 0.35 mmol) were dissolved in tetrahydrofuran (3 HiL), followed by the addition of methanesulfonyl chloride (16 mg, 0.14 mmol) dropwise with stirring at 0 °C. The resulting solution was stirred for 10 min at 0 °C and for an additional 1 h at room temperature. The solution was then diluted with saturated aqueous sodium bicarbonate and extracted thrice with ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate and then the solvent removed under reduced pressure. The crude residue was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 7 min gradient (CH 3 CN : H 2 0 0.03% NH 4 OH 51 : 49 to 68 : 32) and detection by UV at 254 nm to provide the title compound TFA salt (8.9 mg, 15%) as a white solid. MS (ES, m/z): 510 [M+H] + . 1H- NM (400 MHz, CD 3 OD) δ 7.75-7.62 (m, 1H), 7.37-7.04 (m, 4H), 6.90-6.74 (m, 2H), 4.62-4.26 (m, 5H), 3.97-3.74 (m, 3H), 3.50-3.34 (m, 2H), 3.20-2.96 (m, 3H), 2.40-2.34 (m, 1H), 0.83-0.72 (m, 1H), 0.72-0.57 (m, 1H), 0.57-0.50 (m, 1H), 0.20-0.10 (m, 1H). Example 56

(4-cvclopropyl-3,4-dihvdroquinoxalin-lf2H)-yl)(l-((2,5-

Example 56: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(l -((2,5- dichlorobenzyl)amino)cyclobutyl)methanone. Example 56 was prepared using the procedure described for the preparation of Example 26 except that l-((t- butoxycarbonyl)amino)cyclobutanecarboxylic acid was used in place of Boc-1- aminocyclopropane-l-carboxylic acid. Isolated as the bis TFA salt. MS (ES, m/z): 430 [M+H . 1H-NMR (400 MHz, CD 3 OD) δ 7.60 (d, J= 2.5 Hz, 1H), 7.48 (d, J = 8.6 Hz, 1H), 7.44-7.36 (m, 2H), 7.24 (dd, J= 8.3, 1.4 Hz, 1H), 7.19-7.11 (m, 1H), 6.73 (td, J = 7.9, 1.4 Hz, 1H), 4.1 1 (s, 2H), 3.90 (t, J = 5.5 Hz, 2H), 3.47 (t, J = 5.8 Hz, 2H), 2.83- 2.68 (m, 2H), 2.49-2.33 (m, 3H), 2.17-2.00 (m, 1H), 1.88-1.68 (m, 1H), 0.91-0.78 (m, 2H), 0.62-0.49 (m, 2H).

Example 57

(4-cyclopropyl-3,4-dihvdroquinoxalm-l(2H)-yl fl-(2,5-dichlorobeiizyl pyrrolidin-3-yl methanone

Example 57: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(l-(2,5- dichlorobenzyl)pyrrolidin-3-yl) methanone. Example 57 was prepared using the procedure described for the preparation of Example 12 except that l-(t-butoxycarbonyl) pyrrolidine-3-carboxylic acid was used in place of (S)-l- [(benzyloxy)carbonyl]pyrrolidine-2-carboxylic acid. Isolated as the bis TFA salt. MS (ES, m/z): 430 [M+H] + . 1H-NMR (400 MHz, CD 3 OD) δ 7.72 (s, 1H), 7.61-7.47 (m, 2H), 7.22 (dd, J = 8.3, 1.5 Hz, 1H), 7.18 (t, J = 7.6 Hz, 1H), 7.08 (d, J= 7.6 Hz, 1H), 6.75 (t, J = 7.2 Hz, 1H), 4.59 (s, 2H), 4.11-3.35 (m, 9H), 2.59-2.36 (m, 1H), 2.30-1.91 (m, 2H), 0.95-0.78 (m, 2H), 0.57 (s, 2H).

Example 58

(4-cyclopropyl-3,4-dihvdroquinoxalin-l(2HVyl)2-(3-rnethylben zyl)pyrrolidin-l- yPmethanone

Example 58: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)2-(3- methylbenzyl)pyrrolidin-l-yl)methanone. Example 58 was prepared using the procedure described for the preparation of Example 28, except that 2-(3- methylbenzyl)pyrrolidine was used in place of 2-(2-chlorobenzyl)pyrrolidine. MS (ES, m/z): 376 [M+H] + ; 1H-NMR (400 MHz, CDC1 3 ) δ 7.21-7.10 (m, 2H), 7.05-6.88 (m, 5H), 6.71-6.60 (m, 1H), 4.25-4.07 (m, 2H), 3.53-3.34 (m, 2H), 3.33-3.20 (m, 2H), 3.15- 3.06 (m, 2H), 2.52 (dd, J= 12.4, 9.9 Hz, 1H), 2.45-2.36 (m, 1H), 2.30 (s, 3H), 1.93-1.45 (m, 4H), 0.87-0.76 (m, 2H), 0.73 (dd, J = 9.5, 3.9 Hz, 1H), 0.51 (dd, J = 10.0, 3.4 Hz, 1H).

Scheme 59: 1. t-butyl acrylate, Pd(OAc) 2 , PPh 3 , TEA; 2. 5% Rh/Al 2 0 3 , H 2 , EtOAc; 3. PPh 3 Br 2 , DCM; 4. DIEA, CH 3 CN, KI; 5. 4 M HC1 in 1,4-dioxane.

Intermediate 59a: t-Butyl 3-(4-chloro-3-

(hydroxymethyl)phenyl)acrylate. To a mixture of (5-bromo-2-chlorophenyl)methanol (1.0 g, 4.51 mmol, 1 equiv.) and t-butyl acrylate (1.86 mL) in TEA (7.6 mL) were added palladium acetate (51 mg, 0.23 mmol, 0.05 equiv.) and PPh 3 (1 18 mg, 0.451 mmol, 0.1 equiv.). The mixture was stirred under N 2 at 90 °C overnight. The reaction mixture was concentrated under reduced pressure and purified by flash-column chromatography to give 1.03 g (85 %) of t-butyl 3-(4-chloro-3- (hydroxymethyl)phenyl)acrylate as a clear syrup.

Intermediate 59b: t-butyl 3-(4-chloro-3-

(hydroxymethyl)phenyl)propanoate. To a solution of t-butyl 3-(4-chloro-3- (hydroxymethyl)phenyl)acrylate (1.03 g, 3.84 mmol) in ethyl acetate (20 mL) was added Rh Al 2 0 3 (5 %, 300 mg). The mixture was stirred at room temperature under H 2 for 3 h. More Rh/Al 2 0 3 (5 %, 150 mg) was added and the mixture was stirred at room temperature under H 2 overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure to give lg (96 %) of t-butyl 3-(4-chloro-3- (hydroxymcthyl)phenyl)propanoate as a clear syrup.

Intermediate 59c: t-butyl 3-(3-(bromomethyl)-4- chlorophenyl)propanoate. To a solution of t-butyl 3-(4-chloro-3- (hydroxymethyl)phenyl)propanoate (460 mg, 1.7 mmol, 1 equiv.) in DCM (12 mL) was added dibromo triphenylphosphorane (863 mg, 2.0 mmol, 1.2 equiv). The mixture was stirred at room temperature for 30 minutes, quenched with water, extracted with ether. The organic layer was washed with brine, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by flash-column chromatography to give 263 mg (46 %) of t-butyl 3-(3-(bromomethyl)-4-chlorophenyl)propanoate as clear oil.

Intermediate 59d: t-butyl 3-(4-chloro-3-(((l-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline- 1 -carbonyl)cyclopropyl)amino)methyl)phenyl)propanoate. To a mixture of (l-aminocyclopropyl)(4-cyclopropyl-3,4-dihydroquinoxalin-l-( 2H)- yl)methanone HCI salt (26.4 mg, 0.08 mmol, 1 equiv), prepared form 26a by treating it with 4 M hydrochloric acid in 1 ,4-dioxane) and t-butyl 3-(3-(bromomethyl)-4- chlorophenyl)propanoate (32 mg, 0.096 mmol, 1.2 equiv) in acetonitrile (0.3 mL) were added DIEA (55.7 μί, 0.32 mmol, 4 equiv.) and KI (cat.). The mixture was stirred at 50 °C overnight, concentrated under reduced pressure, and purified by flash-column chromatography to give 31 mg (76%) of t-butyl 3-(4-chloro-3-(((l -(4-cyclopropyl- l,2,3,4-tetrahydroquinoxaline-l-carbonyl)cyclo- propyl)amino)methyl)phenyl)propanoate as yellow syrup. MS (ES, m/∑): 510 [M+H] + .

Example 59: 3-(4-chloro-3-(((l-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline- 1 -carbonyl)cyclopropyl)amino)methyl)phenyl)propanoic acid. To t-butyl 3 -(4-chloro-3-((( 1 -(4-cyclopropyl- 1 ,2,3 ,4-tetrahydroquinoxaline- 1 - carbonyl)cyclopropyl)amino)methyl)phenyl) propanoate (31 mg, 0.06 mmol) was added 4 M hydrochloric acid in 1 ,4-dioxane (1 mL). The mixture was stirred at room temperature for 3 h and concentrated to give 32 mg (crude) of 3-(4-chloro-3-(((l-(4- cyclopropyl- 1 ,2,3 ,4-tctrahydroquinoxaline- 1 -carbonyl)cyclopropyl)- amino)methyl)phenyl)propanoic acid as a red solid. Some of the red solid (8 mg) was purified by preparative HPLC to give 5.9 mg of 3-(4-chloro-3-(((l-(4-cyclopropyl- 1 ,2,3 ,4-tetrahydroquinoxaline- 1 -carbonyl)cyclopropyl)amino) methyl)phenyl)propanoic acid bis TFA salt as a yellow solid. MS (ES, m/z): 454 [M+H] + . Ή-NMR (400 MHz, CDC1 3 ) δ 7.34-7.26 (m, 2H), 7.26-7.14 (m, 3H), 7.02 (d, J= 2.0 Hz, 1H), 6.76 (td, J = 7.6, 1 .4 Hz, 1 H), 4.07 (s, 2H), 3.89 (t, J= 5.8 Hz, 2H), 3.43 (t, J= 5.8 Hz, 2H), 2.86 (t, J= 7.6 Hz, 2H), 2.57 (t, J = 7.6 Hz, 2H), 2.48-2.36 (m, 1H), 1.39 (dd, J = 7.8, 5.3 Hz, 2H), 1.22 (dd, J= 7.8, 5.3 Hz, 2H), 0.89-0.77 (m, 2H), 0.57-0.44 (m, 2H).

Example 60

3-(4-chloro-3-(( ' (l-r4-cvclopropyl-l,2.3,4-tetrahydroquinoxaline-l- carbonyl)cvclopropyl)amino methvDphenyl -N-methyl-N-((2S,3R,4R,5R)-2,3,4,5 1 6- pentahvdroxyhexyDpropanamide

Example 60: 3-(4-chloro-3-(((l-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline-l-carbonyl)cyclopropyl)amino)methyl)ph enyl)-N-methyl-N- ((2S,3R,4R,5R)-2,3,4,5,6-penta-hydroxyhexyl)propanamide. To a mixture of Example 59 HC1 salt (8.2 mg, 0.016 mmol, 1 equiv.) and N-methyl glucamine (3.9 mg, 0.02 mmol) in DMF (0.1 mL) were added HATU (7.6 mg, 0.02 mmol) and DIEA (17 uL, 0.1 mmol). The mixture was stirred at room temperature for 1 h and purified by preparative HPLC to give 7.4 mg (54 %) of 3-(4-chloro-3-(((l-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline-l-carbonyl)cyclopropyl)amino)methyl)-p henyl)-N-methyl-N- ((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl)propanamide bis TFA salt as an off-white solid. MS (ES, m/z): 631 [M+H] + . H-NMR (400 MHz, CD 3 OD) 6 7.34-7.26 (m, 2H), 7.26-7.20 (m, 2H), 7.20-7.14 (m, 1H), 7.09 (dd, J = 12.2, 1.9 Hz, 1H), 6.76 (ddd, J = 9.2, 3.4, 1.7 Hz, 1H), 4.1 1 (d, J = 8.8 Hz, 2H), 3.99-3.84 (m, 3H), 3.81-3.54 (m, 6H), 3.48-3.34 (m, 3H), 3.09 (s, 1.4H), 2.96 (s, 1.6H), 2.87 (t, J= 7.5 Hz, 2H), 2.84-2.70 (m, 1H), 2.67 (t, J= 7.5 Hz, 1H), 2.49-2.36 (m, 1H), 1.44-1.33 (m, 2H), 1.28-1.20 (m, 2H), 0.83 (dt, J= 6.6, 1.6 Hz, 2H), 0.57-0.45 (m, 2H).

Example 61

(4-cyclopropyl-3.4-dihydroquinoxalin- 1 (2H)-vD(l -(2,5-dichlorobenzyl)- 1 H-pyrrol-2- vDniethanone

Scheme 61 : 1. 2-(bromomethyl)-l,4-dichlorobenzene, NaH, THF; 2. NaOH, EtOH, H 2 0; 3. le, EDCI, DMAP, DCM.

Intermediate 61a: ethyl l-(2,5-dichlorobenzyl)-lH-pyrrole-2- carboxylate. 2-(Bromomethyl)-l,4-dichlorobenzene (517 mg, 2.15 mmol) and ethyl 1H- pyrrole-2-carboxylate (300 mg, 2.16 mmol) were dissolved in tetrahydrofuran (20 mL). To the stirring solution was added sodiu hydride dispersion in mineral oil (174 mg, 4.35 mmol) in several batches at 0-5 °C. The resulting suspension was stirred overnight at room temperature then quenched by the addition of 5 mL of methanol. The solvent was removed under reduced pressure and the resulting residue purified by preparative TLC (ethyl acetate/petroleum ether 1 :10) to give 61a (270 mg, 42%) as a white solid. MS (ES, m/∑): 298 [M+H] + .

Intermediate 61b: l-(2,5-dichlorobcnzyl)-lH-pyrrole-2-carboxylic acid. 61a (200 mg, 0.67 mmol) and sodium hydroxide (539 mg, 13.47 mmol) were dissolved in ethanol/H 2 0 (8/4 mL) and stirred for 3 h at 85 °C. The mixture was concentrated under reduced pressure and then diluted with 50 mL of dichloromethane. The pH value of the solution was adjusted to 3-4 with aqueous HC1 (1 M). The organic layer was then washed twice with brine and dried over sodium sulfate, then solvent removed under reduced pressure to give 61b (150 mg, 83%) as a yellow solid, which was used directly without further purification.

Example 61 : (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(l -(2,5- dichlorobenzyl)-lH-pyrrol-2-yl)methanone. 61b (80 mg, 0.30 mmol, le (47 mg, 0.27 mmol), EDCI (77 mg, 0.40 mmol), and 4-dimethylaminopyridine (49 mg, 0.40 mmol) where dissolved in dichloromethane (3 mL). The resulting solution was stirred for 4 h at room temperature, then diluted with 20 mL of dichloromethane. The resulting mixture was washed twice with brine and dried over sodium sulfate, then concentrated under reduced pressure. The crude residue was purified by preparative HPLC with a CI 8 silica gel stationary phase using a 6 min gradient (CH 3 CN : ¾0 0.05% TFA 32 : 68 to 50 : 50) and detection by UV at 254 nm to provide the title compound (23.7 mg, 19%) bis TFA salt as an off-white solid. MS (ES, m/z): 426 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.41 (d, J = 8.7 Hz, 1H), 7.29 (dd, J = 8.7,2.4 Hz, 1H), 7.23 (d, J = 8.4 Hz, 1H), 6.77-6.51 (m, 2H), 6.23 (d, J = 3.9 Hz, 1H), 6.14 (t, J = 3.3 Hz, 1H), 5.45 (s, 2H), 3.90 (t, J = 5.7 Hz, 1H), 2.45-2.41 (m, 1H), 0.90-0.84 (m, 2H), 0.66-0.61 (m, 2H).

Example 62

(4-cvclopropyl-3.4-dihvdroqumoxalin-l (2H -yl (l-(2,5-dichlorobenzyl -lH-imidazol-

2-vi)methanone

Scheme 62: 1. K 2 C0 3 , DMF; 2. a. LiOH«H 2 0, THF, H 2 0; b. le, HATU, DIEA, DMF.

Intermediate 62a: 2,5-dichlorobenzyl l-(2,5-dichlorobenzyl)-lH- imidazole-2-carboxylate. lH-Imidazole-2-carboxylic acid (100 mg, 0.892 mmol), 1,4- dichloro-2-(chloromethyl)benzene (382 mg, 1.96 mmol), and K 2 C0 3 (370 mg, 2.68 mmol) were combined in DMF. The suspension was stirred at 100 °C for 1 h, then added to 5% aqueous HC1 and extracted with EtOAc. The organic phase was washed with saturated aqueous NaHC0 3 , H 2 0, and brine, then dried over Na 2 S0 4 and the solvent removed under reduced pressure. The crude residue was purified by flash- column chromatography using a gradient of DCM : MeOH (100 : 0 to 98 : 2) to give 62a (290 mg, 76%) as a yellow oil.

Example 62: (4-cyclopropyl-3,4-dihydroquinoxalin-l (2H)-yl)(l -(2,5- dichlorobenzyl)-lH-imidazol-2-yl)methanone. 62a (290 mg, 0.674 mmol) and LiOH»H 2 0 (113 mg, 2.70 mmol) were dissolved in THF (3 mL) and H 2 0 (2 mL) and stirred at room temperature for 3 h. The solvent was removed under reduced pressure and the residue dissolved in EtOAc and MeOH and filtered, then solvent removed under reduced pressure. The crude residue was then dissolved in DMF, to which le (19 mg, 0.11 mmol), HATU (42 mg, 0.11 mmol), and DIEA (80 μΕ, 0.461 mmol) were added. The solution was stirred at room temperature for 1 h, then purified by preparative HPLC with a C18 silica gel stationary phase using a gradient of H20 0.05% TFA : CH 3 CN 0.05% TFA (30 : 70 to 5 : 95) and detection by UV at 254 nm to give the title compound (23 mg, 5%) as the bis TFA salt. MS (ES, m/z): 427 [M+l] + . ¾-NMR (400 MHz, CDC1 3 ) δ 7.40 (d, J - 8.6 Hz, 1H), 7.37-7.29 (m, 2H), 7.24-7.17 (m, 2H), 7.11-7.02 (m, 2H), 6.78-6.33 (m, 2H), 5.41 (s, 2FI), 4.02 (t, J= 5.2 Hz, 2H), 3.50 (s, 2H), 2.48 (s, 1H), 0.87-0.81 (m, 2H), 0.68-0.62 (m, 2H).

Example 63

l-cyclopropyl-4-(r6-[f2,5-dichlorophenyl)methoxylpyridin-2-y llcarbonyl)-l , 2,3,4-

DME, LiBr, NaH; 3. LiOH, THF, H 2 0; 4. le, HATU, D1LA, DMF.

Intermediate 63a: methyl 6-hydroxypyridine-2-carboxylate. A solution of 6-hydroxypyridine-2-carboxylic acid (5.00 g, 35.94 mmol, 1.00 equiv) in methanol (100 mL) and sulfuric acid (20 mL). was stirred overnight at 65 °C. The resulting reaction mixture was concentrated under reduced pressure, diluted with water (200 mL) and the solid precipitate was collected by filtration and washed with water and aqueous NaHC0 3 . The filter cake was dissolved in ethyl acetate (20 mL) dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 2 g (36%) of the product as a white solid.

Intermediate 63b and 63c: methyl l-(2,5-dichlorobenzyl)-6-oxo-l,6- dihydropyridine-2-carboxylate and methyl 6-(2,5-dichlorobenzyloxy)picolinate respectively. To a solution of methyl 6-hydroxypyridine-2-carboxylate (300 mg, 1.96 mmol, 1.00 equiv) in DMF (1 mL) and DME (3 mL) was added of sodium hydride (90 mg, 2.25 mmol, 1.15 equiv, 60%) at 0 ° C followed by LiBr (339 mg, 3.90 mmol, 1.99 equiv) after a few minutes. The mixture was stirred for 15 min at room temperature then 2-(bromomethyl)-l,4-dichlorobenzene (900 mg, 3.75 mmol, 1.91 equiv) was added. The resulting solution was stirred overnight at 65 °C then quenched by the addition of 2 mL of H 2 0. The resulting solution was extracted with ethyl acetate (2x10 mL) the organic layers combined and concentrated under reduced pressure. The resulting residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 :2) to furnish 80 mg (13%) of the product 63b as a light yellow solid and 150 mg (25%) of the product 63c as a light yellow solid.

Intermediate 63d: 6-[(2,5-dichlorophenyl)methoxy]pyridine-2- carboxylic acid. A solution of methyl 6-[(2,5-dichlorophenyl)methoxy]pyridine-2- carboxylate 63c (150 mg, 0.48 mmol, 1.00 equiv), LiOH (10 mg, 0.42 mmol, 1.00 equiv) in tetrahydrofuran/H 2 0 (2:1 mL). The resulting solution was stirred for 2 h at room temperature then diluted with water (10 mL) and extracted with ethyl acetate (2x10 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 110 mg (77%) of the product as a light yellow solid.

Example 63 : 1 -cyclopropyl-4-([6- [(2,5-dichlorophenyl)methoxy]pyridin- 2-yl]carbonyl)-l,2,3,4-tetrahydroquinoxaline. A solution of 6- [(2,5- dichlorophenyl)methoxy]-pyridine-2-carboxylic acid (110 mg, 0.37 mmol, 1.50 equiv), l-cyclopropyl-l,2,3,4-tetrahydroquinoxaline (43 mg, 0.25 mmol, 1.00 equiv), HATU (187 mg, 0.49 mmol, 2.00 equiv), DIEA (64 mg, 0.50 mmol, 2.00 equiv) in N,N- dimethylformamide (2 mL) was stirred for 2 h at 40 °C. The resulting solution was diluted with of ethyl acetate (20 mL), washed with brine (2x10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product (100 mg) was purified by preparative HPLC with the following conditions: Column, SunFire Preparative CI 8, 19* 150mm 5μιη; Mobile phase gradient, water containing 0.05% TFA : CH 3 CN (48 : 52 to 25 : 75 over 6 min then up to 100% over 1 min); Detector, Waters 2545 UV detector at 254 and 220nm. This resulted in 18 mg (16%) of the title compound ditrifluoroacetate salt as a yellow semi-solid. MS (ES, m/z) 454 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.80 (m, 1H), 7.37 (m, 2H), 7.30 (m, 2H), 7.20 (d, J = 8.1 Hz, 1H), 6.95 (m, 2H), 6.40 (d, J = 1.8 Hz, 2H), 3.94 (s, 1H), 3.49 (s, 2H), 2.45 (m, 1H), 1.19 (m, 3H), 0.87 (m, 2H), 0.69 (m, 2H).

Example 64

l-cvclopropyl-4-( 6-r(2,5-dichlorophenyl)memoxylpyridin-2-yllcarbonyl)-L23,4- tetrahydroquinoxaline

Scheme 64: 1. a. (COCl) 2 , cat. DMF, DCM b. TEA, DCM.

Intermediate 64a: l-[(2,5-dichlorophenyl)methyl]-6-oxo-l,6- dihydropyridine-2-carboxylic acid. A solution of methyl l-[(2,5- dichlorophenyl)methyl]-6-oxo-l,6-dihydropyridine-2-carboxyla te 63b (80 mg, 0.26 mmol, 1.00 equiv), LiOH (5 mg, 0.21 mmol, 0.81 equiv) in tetrahydrofuran: water (2.T mL) was stirred for 2 h at room temperature then diluted with of water (5 mL). The resulting solution was extracted with ethyl acetate (2x10 mL) and the organic layers combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 50 mg (65%) of the product as a light yellow solid.

Example 64: 6-[(4-cyclopropyl- 1,2,3, 4-tetrahydroquinoxalin-l- yl)carbonyl]-l-[(2,5-dichlorophenyl)methyl]-l,2-dihydropyrid in-2-one. To a solution of l-[(2,5-dichlorophenyl)methyl]-6-oxo-l,6-dihydropyridine-2-c arboxylic acid (50 mg, 0.17 mmol, 1.00 equiv) N.N-dimethylformamide (a catalytic amount), in dichloromethane (10 mL) was added oxalyl dichloride (1 mL). The resulting solution was stined for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure to provide 60 mg of the crude l-[(2,5-dichlorophenyl)methyl]-6-oxo- l,6-dihydropyridine-2-carbonyl chloride as a yellow oil used without further purification. To a stirred at 0 °C solution of l-cyclopropyl-1,2,3,4- tetrahydroquinoxaline (30 mg, 0.17 mmol, 1.00 equiv), triethylamine (0.5 mL) in dichloromethane (5 mL) was added l-[(2,5-dichlorophenyl)methyl]-6-oxo-l,6- dihydropyridine-2-carbonyl chloride (60 mg, 1.00 equiv) in DCM. The resulting solution was stirred for 2 h at room temperature, concentrated under reduced pressure and the crude product (50 mg) was purified by preparative HPLC with the following conditions: Column, SunFire Preparative CI 8, 19* 150mm 5μηι; Mobile phase gradient, water containing 0.05% TFA: CH 3 CN (65:35 to 48:52% over 10 min then to 100% in 1 min); Detector, Waters 2545 UV detector at 254 and 220nm. This resulted in 13 mg (17%) of title compound trifluoroacetate salt as a yellow solid. MS (ES, m/z): 454 [M+H] + ; 1H-NM (400 MHz, CD 3 OD) d 7.53 (s, 1H), 7.41 (δ, J = 6.6 Hz, 1H), 7.19 (m, 2H), 7.08 (m, 1H), 6.65 (m, 2H), 6.50 (m, 2H), 6.36 (s, 1H), 5.41 (m, 2H), 3.95 (s, 2H), 3.50 (s, 2H), 2.42 (s, 1H), 0.88 (d, J= 3.6 Hz, 2H), 0.63 (s, 2H).

Example 65

1 -cyclopropyl-4- [ [5-(2,5-dichlorophenoxy)- 1 ,3 -dimethyl- 1 H-pyrazol-4-yl]carbonyll -

1 2,3 ,4-tetrahydroquinoxaline.

Scheme 65 : 1. 2,5-dichlorophenol, Cul, K 2 C0 3 , DMF; 2. NaC10 2 , NaH 2 P0 4 , ¾0 t- BuOH, 2-methylbut-2-ene; 3. a. (COCl) 2 ; b. le, TEA, DCM.

Intermediate 65a: 5 -(2,5 -dichlorophenoxy)- 1,3 -dimethyl- lH-pyrazole-4- carbaldehyde. A solution of 2,5-dichlorophenol (200 mg, 1.23 mmol, 1.95 equiv), 5- chloro-l,3-dimethyl-lH-pyrazole-4-carbaldehyde (100 mg, 0.63 mmol, 1.00 equiv), potassium carbonate (350 mg, 2.53 mmol, 4.02 equiv), Cu (25 mg, 0.39 mmol, 0.62 equiv), Cul (25 mg, 0.13 mmol, 0.21 equiv) in N N-dimethylformamide (4 mL) was stirred overnight at 100 °C in an oil bath. The resulting reaction mixture was diluted with H 2 0 (20 mL) and extracted with ethyl acetate (3x20 mL) the combined organic layers were washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to furnish 120 mg (67%) of the product as brown oil.

Intermediate 65b: 5-(2,5-dichlorophenoxy)-l ,3 -dimethyl- lH-pyrazole-4- carboxylic acid. A solution of 5-(2,5-dichlorophenoxy)- 1,3 -dimethyl- lH-pyrazole-4- carbaldehyde (120 mg, 0.42 mmol, 1.00 equiv) NaH 2 P0 4 (420 mg, 3.50 mmol, 8.32 equiv), NaC10 2 (360 mg, 4.00 mmol, 9.50 equiv) in rt-Butanol (6 mL), H 2 0 (6 mL) and 2-methylbut-2-ene (1 mL) was stirred overnight at room temperature. The resulting reaction mixture was diluted with H 2 0 (10 mL), extracted with ethyl acetate (3x20 mL) and the organic layers combined washed with brine (1x20 mL), dried over sodium sulfate and concentrated under reduced pressure to provide 110 mg (87%) of the product as a colorless oil.

Example 65: 1 -cyclopropyl-4-[[5-(2,5-dichlorophenoxy)-l ,3-dimethyl- lH-pyrazol-4-yl]carbonyl]-l,2,3,4-tetrahydroquinoxaline. To a stirred solution of 5- (2,5-dichlorophenoxy)-l,3-dimethyl-lH-pyrazole-4-carboxylic acid (100 mg, 0.33 mmol, 1.00 equiv) in dichloromethane (10 mL) was added oxalyl dichloride (10 mL) dropwise. The reaction mixture was stirred for 2 h at room temperature then concentrated under reduced pressure. The crude residue was dissolved in dichloromethane (5 mL) and added to a stirred 0 °C solution of l-cyclopropyl-1,2,3,4- tetrahydroquinoxaline (80 mg, 0.46 mmol, 1.40 equiv), triethylamine (70 mg, 0.69 mmol, 2.00 equiv) in dichloromethane (10 mL). The resulting reaction mixture was allowed to warm to room temperature and stirred for 4 h then was diluted with H 2 0 (10 mL) and extracted with dichloromethane (3x10 mL) and the combined organic layers washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product (60 mg) was purified by Flash-Preparative- HPLC with the following conditions: Column, CI 8 silica gel; Mobile phase gradient CH 3 CN in H 2 0 (containing 0.05% TFA) 5%-100% over 40 min; Detector, UV at 254 nm. This resulted in 16.1 mg (11%) of title compound trifluoroacetate salt as a light yellow solid. MS (ES, m/z): 457 [M+H] + . 1H-NMR (300 MHz, CD 3 OD) δ 7.44 (d, J = 8.1 Hz, 1H), 7.10-7.18 (m, 2H), 6.96-7.04 (m, 1H), 6.79 (d, J= 2.4 Hz, 1H), 6.66 (d, J = 7.2 Hz, 1H), 6.51 (t, J = 7.2 Hz, 1H), 3.78 (s, 2H), 3.56 (s, 3H), 3.19 (t, J = 5.1 Hz, 2H), 2.31-2.38 (m, 1H), 2.15 (s, 3H), 0.73-0.83 (m, 2H), 0.53 (m, 2H).

Example 66

l-cvclopropyl-4-(| " 5-r(2,5-dichlorophenyl)methyl1-l,3-oxazol-4-yl1carbony l)-l,2.3.4- tetrahydroq ui nox aline

Scheme 66: 1. H 2 S0 4 H 2 0; 2. 2-isocyanoacetate, CDI, tBuOK; 3. LiOH,THF, H 2 0; 4. le, EDCI, HOAT, DMF.

Intermediate 66a: 2-(2,5-dichlorophenyl)acetic acid. To a stirred solution of 2-(2,5-dichlorophenyl)acetonitrile (700 mg, 3.76 mmol, 1.00 equiv) in water (6 mL) was added sulfuric acid (8 mL) dropwise. The resulting solution was stirred for 3 h at 110 °C in an oil bath, diluted with H 2 0 (100 mL), extracted with dichloromethane (3x50 mL) and the combined organic layers washed with brine (3x100 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 700 mg (91%) of the product as a white solid.

Intermediate 66b: ethyl 5-r(2,5-dichlorophenyl)methyl]-l,3-oxazole-4- carboxylate. A stirred solution of 2-(2,5-dichlorophenyl)acetic acid (1 g, 4.88 mmol, 1.00 equiv) and (2-ethoxy-2-oxocthyl)(methylidyne)azanium (560 mg, 4.91 mmol, 1.01 equiv), in N,N-dimethylformamide (10 mL) was treated with CDI (800 mg, 4.93 mmol, 1.01 equiv) followed by i-BuOK (55 mg, 0.49 mmol, 0.10 equiv). The resulting reaction mixture was stirred overnight at room temperature then diluted with H 2 0 (30 mL) and extracted with ethyl acetate (3x30 mL) and the organic layers combined. The combined organic layer was washed with brine (2x30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 :5) to furnish 540 mg (37%) of the product as a yellow oil.

Intermediate 66c: 5-[(2,5-dichlorophenyl)methyl]-l ,3-oxazole-4- carboxylic acid. A stirred solution of ethyl 5-[(2,5-dichlorophenyl)methyl]-l,3-oxazole- 4-carboxylate (200 mg, 0.67 mmol, 1.00 equiv) and LiOH (50 mg, 2.09 mmol, 3.13 equiv) in tetrahydrofuran/H 2 0 (50/20 mL) was stirred overnight at 80 °C in an oil bath. The pH value of the resulting reaction mixture was adjusted to 3 with 1 M HC1 extracted with ethyl acetate (3x20 mL) and the organic layers combined. The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 130 mg (72%) of the product as a white solid.

Example 66: l-cyclopropyl-4-([5-[(2,5-dichlorophenyl)methyl]-l,3- oxazol-4-yl]carbonyl)-l,2,3,4-tetrahydroquinoxaline. A solution of 5-[(2,5- dichlorophenyl)methyl]-l ,3-oxazole-4-carboxylic acid (100 mg, 0.37 mmol, 1.00 equiv), l-cyclopropyl-l,2,3,4-tetrahydroquinoxaline (60 mg, 0.34 mmol, 0.94 equiv), EDCI (75 mg, 0.39 mmol, 1.06 equiv) and HO AT (55 mg, 0.40 mmol, 1.10 equiv) in N,N-dimethylformamide (4 mL) was stirred overnight at room temperature. The reaction mixture was diluted with H 2 0 (20 mL), extracted with ethyl acetate (3x20 mL) and the organic layers combined. The combined organic phase was washed with brine (1x20 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide crude product (150 mg) which was purified by preparative HPLC with the following conditions: Column, SunFire Preparative C18, 19*1 50mm 5μπι; Mobile phase gradient, water containing 0.05% TFA : CH 3 CN (48:52 to 32:68 over 10 min then to 100.0% in 1 min); Detector, Waters 2545 UV detector at 254 and 220nm to provide 77.8 mg (49%) of title compound trifluoroacetate salt as a brown solid. MS 12 071251

(ES, m/z) 428 [M+H] + . ! H-NMR (300 MHz, CDC1 3 ) δ 7.30 (s, 1H), 7.02 (m, 2H), 6.59 (t, J= 6.9 Hz, 1H), 5.49 (s, 1H), 4.16 (s, 1H), 4.07 (t, J= 5.7 Hz, 2H), 3.47 (t, J = 5.7 Hz, 2H), 2.41-2.48 (m, 1H), 0.81-0.87 (m, 2H), 0.62-0.67 (m, 2H).

Example 67

(4-cvclopropyl-3 ,4-dihydroquinoxalin- 1 (2HV yl)f (2S 1 -( 1 - (2.5- dichlorophenyl)ethyl)pyrrolidin-2-yl)methanone

Example 67: (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)((2S)-l-(l- (2,5-dichlorophenyl)ethyl)pyrrolidin-2-yl)methanone. Example 67 was prepared using the procedure described for the preparation of Example 12, except that l-(2,5- dichlorophenyl)-ethyl methanesulfonate (prepared from l-(2,5-dichlorophenyl)ethanol by standard mesylation methods) was used in place of 2-(bromomethyl)-l,4- dichlorobenzene. Two isomers were separated by preparative HPLC. Isomer 1 : MS (ES, m/z): 444 [M+H] + . 1H-NMR (400 MHz, CD 3 OD) δ 7.67 - 7.59 (m, 1H), 7.49 (d, J= 8.6 Hz, 0.2H), 7.42 (dd, J = 8.6, 2.5 Hz, 0.8H), 7.39 - 7.34 (m, 0.3H), 7.32 (d, J = 8.6 Hz, 0.7H), 7.28 - 7.15 (m, 1.7H), 7.08 - 6.98 (m, 0.3H), 6.77 - 6.67 (m, 1.5H), 6.63 - 6.53 (m, 0.5H), 5.21 - 5.07 (m, 0.3H), 5.05 - 4.92 (m, 0.7H), 4.58 (dd, J= 10.6, 3.2 Hz, 1H), 4.08 - 3.90 (m, 2H), 3.74 - 3.38 (m, 2H), 3.26 - 3.18 (m, 1H), 2.90 - 2.78 (m, 1H), 2.63 - 1.98 (m, 5H), 1.80 (d, J = 7.0 Hz, 0.6H), 1.64 (d, J = 6.8 Hz, 2.4H), 0.97 - 0.80 (m, 2H), 0.69 - 0.49 (m, 2H).Isomer II 5 7.89 - 7.71 (m, 1H), 7.68 - 7.46 (m, 2.4H), 7.27 (d, J = 4.2 Hz, 1.6H), 7.19 - 7.04 (m, 1H), 6.88 - 6.77 (m, 0.7H), 6.75 - 6.64 (m, 0.3H), 5.34 - 5.19 (tn. 0.3H), 5.16 - 5.03 (m, 0.7H), 4.80 - 4.74 (m, 1H), 4.01 - 3.38 (m, 511), 3.22 - 3.08 (m, 1H), 2.59 - 2.40 (m, 1H), 2.34 - 1.87 (m, 4H), 1.74 (d, J= 6.8 Hz, 0.9H), 1.56 (d, J = 6.9 Hz, 2.1H), 0.89 (dd, J = 6.5, 2.0 Hz, 2H), 0.72 - 0.46 (m, 2H). Example 68

(l-((5-(3-aminopropyl -2-chlorobenzyl amino cyclopropyl)( ' 4-cvclopropyl-3,4- - 1 (2H)-yl)methanone

Scheme 68: 1. t-butyl prop-2-yn-l-ylcarbamate, Pd(PPh 3 ) 2 Cl 2 , Cul, TEA, DMF; 2. 5% Rh/Al 2 0 3; H 2 , EtOAc; 3. PPh 3 Br 2 , DCM. 4. DIPEA, CH 3 CN, KI; 5. 4 M HC1 in 1,4-dioxane.

Intermediate 68a: t-Butyl (3-(4-chloro-3-(hydroxymethyl)phenyl)prop-2- yn-l-yl)cabamate. To a mixture of (5-bromo-2-chlorophenyl)methanol (1.00 g, 4.51 mmol), t-butyl prop-2-yn-l-ylcabamate (0.84 g, 5.4 mmol) and TEA (5.2 niL) in DMF (3.2 mL) were added Pd(PPh 3 ) 2 Cl 2 (158 mg, 0.226 mmol) and Cul (86 mg, 0.45 mmol). The mixture was stirred under N 2 at 50 °C overnight. The reaction mixture was diluted with EtOAc, washed with water (2x) and brine (lx), dried over anhydrous sodium sulfate, concentrated under reduced pressure and purified by flash-column chromatography to give 477 mg (36 %) of 68a as a pale yellow syrup.

Intermediate 68b: t-butyl (3-(4-chloro-3-

(hydroxymethyl)phenyl)propyl)-carbamate. To a solution of t-butyl (3-(4-chloro-3- (hydroxymethy])phenyl)prop-2-yn-l-yl)cabamate (477 mg, 1.61 mmol) in ethyl acetate (15 mL) was added Rh/Al 2 0 3 (5 %, 160 mg). The mixture was stirred at room temperature under H 2 for 6 h. More Rh/Al (5 %, 160 mg) was added and the mixture was stirred at room temperature under an atmosphere of H 2 overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure to give 463 mg (96 %) of 68b as a brown syrup.

Intermediate 68c: t-butyl (3-(3-(bromomethyl)-4-chlorophenyl)propyI)- carbamate. To a solution of t-butyl (3-(4-chloro-3- (hydroxymethyl)phenyl)propyl)carbamate (190 mg, 0.63 mmol) in DCM (4.5 mL) was added dibromo triphenylphosphorane (295 mg, 0.7 mmol). The mixture was stirred at room temperature for 30 minutes, quenched with water, and extracted with ether. The organic layer was washed with brine (lx), dried with anhydrous sodium sulfate, concentrated under reduced pressure, and purified by flash-column chromatography to give 45 mg (20 %) of 68c as a yellow syrup.

Intermediate 68d: t-butyl (3-(4-chloro-3-(((l-(4-cyclopropyl-l,2,3,4- tetra-hydroquinoxaline- 1 - carbonyl)cyclopropyl)amino)methyl)phenyl)propyl)carbamate. To a mixture of (1- aminocyclopropyl)(4-cyclopropyl-3,4-dihydroquinoxalin-l-(2H) -yl)methanone HC1 salt (20 mg, 0.060 mmol, prepared from 26a by treating it with 4 M hydrochloric acid in 1 ,4-dioxane) and t-butyl (3-(3-(bromomethyl)-4-chlorophenyl)propyl)carbamate (22 mg, 0.06 mmol) in acetonitrile (0.25 mL) were added DIPEA (43 uL, 0.25 mmol) and KI (cat.). The mixture was stirred at 50 °C overnight, concentrated under reduced pressure, and purified by flash-column chromatography to give 21 mg (64 %) of 68d as a yellow syrup. MS (ES, m/z): 539 [M+H] +

Example 68: (l-((5-(3-aminopropyl)-2- chlorobenzyl)amino)cyclopropyl)(4-cyclopropyl-3,4-dihydroqui noxalin-l(2H)- yl)methanone. To t-butyl (3-(4-chloro-3-(((l-(4-cyclopropyl-l,2,3,4- tetrahydroquinoxaline-l-carbonyl)cyclopropyl)amino)methyl)ph enyl)-propyl)carbamate (21 mg, 0.039 mmol) was added a 4 M hydrochloric acid solution in 1,4-dioxane (1 mL). The mixture was stirred at room temperature for 30 min and concentrated under reduced pressure to give 21 mg (crude) of (l -((5-(3-aminopropyl)-2-chlorobenzyl)- amino)cyclopropyl)(4-cyclopropyl-3,4-dihydroquinoxalin-l(2H) -yl)methanon as a red solid. Some of the red solid (4 mg) was purified by preparative HPLC to give 3 mg of the title compound TFA salt as a yellow solid. MS (ES, m/z): 439 [M+H] + . 1H-NMR (400 MHz, CDCI 3 ) δ 7,30 (dd, J= 7.9, 1.4 Hz, IH), 7.26 (d, J= 8.1 Hz, IH), 7.20 (dd, J = 8.3, 1.4 Hz, IH), 7.16-7.07 (m, 2H), 6.87 (d, J = 1.8 Hz, IH), 6.73 (td, J = 7.7, 1.4 Hz, IH), 3.88 (t, J= 5.6 Hz, 2H), 3.76 (s, 2H), 3.40 (t, J= 5.8 Hz, 2H), 2.90 (t, J= 8.0 Hz, 2H), 2.62 (t, J ~- 8.0 Hz, 2H), 2.47-2.35 (m, IH), 1.99-1.81 (m, 2H), 1.40 (q, J= 4.6 Hz, 2H), 1.01 (q, J= 4.6 Hz, 2H), 0.84-0.72 (m, 2H), 0.53-0.37 (m, 2H). s!e 69

3-(3-(4-chloro-3-(((l-(4-cyclopropyl-L2,3 ^ ^

carbonyl)cyclopropyl)amino)methyl)phenyl)propyl)- 1 -methyl- 1 -((2S,3 ,4R,5R)- ,4,5,6-pentahydroxyhexyl)iirea

Example 69 : 3-(3 -(4-chl oro-3 -(((1 -(4-cycl opropyl- 1 ,2,3 ,4-tetrahydro- quinoxaline- 1 -earbony !)cyc I opropy l)amino )methyl)phenyl )propyl)- 1 -methyl- 1 - ((2S,3R,4R 5 5R)-2,3,4 5 5,6-pentahydroxyhexyl)urea. To a mixture of Example 68 (16.2 mg, 0.037 mmol) in THF (0.2 mL) was added Ν,Ν'-disuccinimidyI carbonate (10.4 nig, 0.041 mmol). The mixture was stirred at room temperature for lh. To the mixture was added N-methyl-D-glucamine (10.8 mg, 0.055 mmol). The reaction mixture was stirred at 60 °C for 4 h and more Ν,Ν' -disuccinirnidyl carbonate (10.4 mg, 0.041 mmol) was added. The mixture was stirred at 60 °C overnight, concentrated under reduced pressure, and purified by preparative HPLC to give 8.8 mg (27 %) of the title compound TFA salt as a yellow syrup. MS (ES, m/z) 660 [M+H] + . 'i l- R (400 MHz, CD3OD) δ 7.39 (d, J = 8.1 Hz, I H), 7.30-7.23 (m, 4H), 7.22-7.15 (m, IH), 6.76 (td, J - 7.5, 1.4 Hz, I H), 4.37 (s, 2H), 3.99-3.87 (m, 3H), 3.81-3.59 (m, 5H), 3.50-3.41 (m, 3H), 3.40-3.33 (m, IH), 3.14 (t, J = 6.9 Hz, 211). 2.95 (s, 3H), 2.65 (t, J = 7.6 Hz, 2H), 2.49-2.38 (m, IH), 1.86-1 .73 (m, 2H), 1.44-1.32 (m, 4H), 0.92-0.77 (m, 2H), 0.63-0.50 (m, 2H). 3n 4-chloro-3n(C l~i4-cyelo^

;arbonyl)cyclopropyl amino)meth^

pentahydroxyhexyl)propanamide

Example 70 : 3 ~(4-chloro-3 -((( 1 -(4-cyclopropyl- 1,2,3,4- tetrahydroqmnoxa]ine-l-carbonyi)cyclopropyl)airdno)methyl)ph enyI)-N- ((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxy-hexyl)propanaiaide. Example 70 was prepared using the procedure described for the preparation of Example 60, except that D- glucamine was used in place of N-methyl-D-glucamine. MS (ES. m/z) 617 M+H]*

Example 71

pentahydroxyhexYl ipropanarriide

Example 71 : 3-(4-chloro-3-(((l-(4-cyc!opropyl-l,2,3,4- tetrahyckoquinoxaHne-l-^

((2S,3R,4S 3 5R)-2.,3,4,5 ! 6-penta-hydroxyhexyl)propanamide. Example 71 was prepared using the procedure described for the preparation of Example 60, except that 1-Deoxy- 1 -(methylamino)-D-galactitol was used in place of -methyi-D-gl camine. MS (ES, ): 631 [M+H]

Example 72

N 2-cyclobutoxvphenyl)-1 -(2,5-dicMorobenzyloxy)-N- methylcvciopropanecarboxamide

Example 72: N-(2-cyclobtitoxyphenyl)-l-(2,5-diclilorobenzyloxy)-N- methylcyclopropattecarboxamide. Example 72 was synthesized in an analogous fashion to Example 36 using 2-cyciobutoxy-N-methylaniline (which was made in an analogous fashion to 39e, substituting cyclobutatiol for cyolopropylmethanol) in place of 2- methoxy-N-methylanilitte. MS (ES, m/z) 420 [M+H] + .

Example 73

l-cyclopropyl-4-[[5-(2,5-dichl.orophenoxy -l J-diim^t]:ryi-lH-pyrazo¾-4-yl]carbonyi]

1.2 ,3 ,4-tetrahydroQ " uinoxaline .

Scheme 73: 1 ,2-dibromoethane, K 2 C0 3> CH 3 CN; 2. NaH, DMF; 3. Fe/NFLCL MeOH, H 2 0; 4. (Boc) 2 0, EtOH; 5. Mel, NaH, DMF; 6. C oroiodomethane, Et 2 Zn, 2012/071251

DCE; 7.TFA, DCM; 8. HO At, EDCI, DMF; 9, 2-0>roraomethyl -l,4-dicMorobenzene,

Nail, DMF, intermediate 73a 1 -(2-bromoethoxy)-2-riitrobenzene: A solution of 2- nitrophenol (1.00 g, 7.19 mmol, 1 .00 equiv) 1,2-dibromoethan.e (4.00 g, 21.3 mmol, 3,00 equiv), potassium carbonate (1.90 g, 13.8 mmol, 2.00 equiv) in CH 3 CN (30 mL) was stirred for 3 h at 90 °C. The resulting reaction mixture was concentrated under reduced pressure, dissolved in of ethyl acetate (200 mL) and washed with brine (3x50 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 1 g (57%) of 73a as a green oil.

intermediate 73b 1 ~(ethenyloxy)-2-nitrobenzene: To a stirred 0-5 °C solution of 1 -(2-bromoetihoxy)-2-nitrobenzene (550 mg, 2.24 mmol, 1.00 equiv) in DMF ' (6 mL) was added sodium hydride (180 mg, 4,50 mmol, 2.00 equiv) in portions. The resulting reaction, mixture was stirred overnight at room temperature, diluted with of ethyl acetate (50 mL) arid washed with, brine (3x20 mL), dried, over anhydrous sodium sulfate, and concentrated under reduced pressure to provide 200 mg (54%) of 73b as a yellow oil.

Intermediate 73c 2-(ethenyloxy)aniline: To a 60 °C solution of 1- (e1henyloxy)-2-mtrobenzene (300 mg, 1 .82 mmol, 1.00 equiv) in methanol (10 mL) was added a solution of ammonium chloride (970 mg, 18.1 mmol, 10.0 equiv) in water (3 mL) followed by the addition of iron powder (1 g, 17,91 mmol, 10.00 equiv) in portions. The resulting reaction mixture was stirred for 2 h, solids were removed by filtration and the filtrate concentrated under reduced pressure. The residue was dissolved in 20 mL of ethyl acetate, washed with brine (2x20 mL), the organic phase dried over anhydrous sodium sulfate, and concentrated under reduced pressure to provide 200 mg (81%) of 73c as a brown oil.

Intermediate 73d tert-butyl N-[2-(cthenyIoxy)phenyl]carbamate: A solution of 2"(ethenyloxy)aniline (230 mg, 1.70 mmol, .1.00 equiv) in ethanol (2 mi .) and. di-tcrt-butyi dicarbonate (446 mg, 2.04 mmol, 1 .20 equiv) was stirred overnight at room temperature. The resulting reaction mixture was concentrated under reduced pressure and the residue was purified by silica, gel chromatography eluting with ethyl acetate/petroleum ether (1:200) to provide 200 mg (50%) of 73d as a yellow oil. Intermediate 73 e tert-butyl N-[2-(ethenyloxy)phenyl]~N- methylcarbamate: To a stirred 0 °C solution of tert-butyl M-[2~ (emenyloxy)phenyl]carbamate (190 mg, 0.81 mmol, 1.00 equiv) in DMF (2 mL) was added sodium hydride (49 mg, 1.2 mmol, 1.5 equiv) in several batches. The reaction mixture was stirred for 0.5 h at 0 °C and iodomethane (230 mg, 1.62 mmol, 2.00 equiv) was added dropwise with stirring. The resulting reaction mixture was allowed to warm to room temperature and stirred for 0.5 h then diluted with 50 mL of ethyl acetate. The resulting organic solution was washed, with brine (3x20 mL) dried over anhydrous sodium sulfate, and concentrated under reduced pressure to provide 150 mg (75%) of 73e as a brown oil.

Intermediate 73f tert-butyl N-(2-cyclopropoxyphenyl)-N- methylcarbamate: To a stirred 0 °C solution, of tert-butyl N-[2-(cthenyloxy)phenyl]~N- methylcarbamate (150 mg, 0.60 mmol, 1.00 equiv) in 1 ,2-dichloroethane (1.0 mL) was added chloro(iodo)methane (382 mg, 2.17 mmol, 3.60 equiv) followed by dropwise addition of diethylzinc (1.5 mL, 2,40 equiv. 1.0 M). The resulting reaction mixture was allowed to warm to 25 °C, stirred overnight, then quenched by the addition of 20 mL of aqueous NH 4 CI. The resulting reaction mixture was extracted with dichloromethane (2x20 mL) and the combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 150 mg of 73 f as a brown oil used without further purification.

Intermediate 73g 2-eyclopropoxy-N--methylanilH.ie: A stirred solution of tert-butyl N-(2-cyelopropoxyphenyl)-N-methyicarbamate (30 mg, 0.11 mmol, 1.0 equiv) in 1,4-dioxane (1.5 mL) and concentrated hydrogen chloride (0.5 mL) was stirred for 1 h at 25 °C. The pH value of the reaction mixture was adjusted to 9 with sodium carbonate then extracted with ethyl acetate (2x50). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 10 mg of 73g as a brown oil, which was used without further purification.

intermediate 73h N-(2~cyclopropoxyphenyl)- 1 -hydroxy-N- methylcyclopropane-l-carboxamide: A stirred solution of 1 -hydroxycyclopropane- 1 - carboxylic acid (100 mg, 0.98 mmol, LOO equiv), 2-cycJopropoxy-N-methylaniline (176 mg, 1.08 mmol. 1.10 equiv), EDO (283 mg, 1.48 mmol, 1.50 equiv) and HO At (200 mg, 1.47 mmol, 1.50 equiv) in DMF (2 mL) was stirred overnight at room temperature. The resulting reaction mixture was diluted with ethyl acetate (50 mL) washed with brine (4x2.0 mL), dried over anhydrous sodium svdfate, and concentrated under reduced pressure to provide 80 mg (33%) of 73h as a white solid. MS (ES, m/z) 248 I M i J I i .

Example 73 l-cycIopropyI-4-[[5-(2,5-dichlorophenoxy)-l,3-dimeth.yl- lH~pyrazoi-4-yl]carbonyl]-i,2,3.4 ^ ^ Example 73 was prepared as described for example 8, substituting 73h for 8c. MS (ES, m/z): 406 jM+H ; 1 H- NMR (300 MHz, CD 3 OD) δ 7.26-7.10 (m, 5H), 6.89-6.84 (m, IH), 6.40 (na, 2H), 4.43-4.39 (m, IH), 4.32-4.27 (m, IH), 3.10 (s, 3H), 1.40-1.36 (m, IH), 1.30-1.10 (m, IH), 1.10-0.90 (m, IH), 0.89-0.88 (m, IH), 0.67-0.50 (m, 3H), 0.35-0.32 (m, IH).

l-[ (2S)-l-r -cMoro-5-(trifluoromethoxy)phenyl3methy

cyclo propyl- 1 ,2,3 ,4-tetrahydroquinoxaline

Example 74 l-[[(2S)-l-[[3-chloro-5-(trifluoromethoxy)phenyl]meth.yl]- pyrrolidin-2-yl]earbonyl]-4-cyclopropyl- 1 ,2,3,4-tetrahydroqukox.alme: Example 74 was prepared as described for example 12, substituting 1 -(bromomethyl)-3-chloro-5- (trifluoromethoxy)benzene for 2-(bromomethyl)-l,4-dichloro benzene. This resulted in 33.4 mg (31%) of the title compound as colorless oil. MS (ES, m/z) 480 [M+H] + ; ¾- MR (400 MHz, CD 3 OD) δ 7.20 (s, Hi), 7.15-7.18 (m, 4H), 6.90 (s, IH), 6.70-6.72 (m s IH), 4.88 (s, I H), 4.60 (m, IH), 3.88 (m, Ϊ Η), 3.66-3.69 (m, IH), 3.32-3.50 (m, 3H), 3.03-3.16 (m, I H), 2.37-2.45 (m, 2H), 1.81 -1 .95 (m, 4H), 0.82-0.84 (m, 2H), 0.51 (s, 2H). Example 75

3-[2,5-dicMoro-4-([l-[( -cyclopro^^

vl carbonyllcyclopropyl]meth^^^

pentahydroxyhexyljpropanamide

Example 75 3 ■■ [2,5-dichloro-4-([ 1 - [(4-cyclopropyl- 1 ,2,3 ,4-tetrahydro- quinoxaHn-l-yl)carbonyl]cyclopropyl]methoxy)phenyl]-N-[(2S,3 R,4R,5R)-2,3,4,5,6- pentahydroxyliexyl]propaaamide: Example 75 was prepared as described for example 34 substituting (2R,3R,4R 3 5S)-6-aminohexane-l;2,3,4 3 5-peritoI for (2R,3R,4R,5S)-6- (ineth}damino)hexa.ne-l,2,3,4,5-pentaol. This resulted in 83.3 nig (31%) of the title compound trifluoroacetaie salt, as an off-white solid, MS (ES, m/z): 652 [M÷H] + ; 1H- NMR (300 MHz, CD 3 OD) δ 7.38-7.42 (d, J ~ 7.8 Hz, 1H), 7.33 (s, 1H), 7.08-7.1 1 (ni, 2H), 6.70-6.73 (m, 1H), 6.61 (s, 1H), 3.61-3.90 (m, 11H), 3.37-3.44 (m, 3H), 2.92-2.96 (m, 2H), 2.46-2.48 (m, 2H), 2.23-2.25 (m, 1H), 1.35-1.37 (m, 2H), 0.95-0,99 (m, 2H), 0.64-0.68 (m, 2H), 0.17-0.18 (m, 2H).

Example 76

yl carbonvl]-l,23,4-tetrahydroqui«oxaline

Scheme 76: 1. 2-(bromomethyl)-l,4-dichlorobenzene, K 2 CO3, CH 3 CN; 2. LiOH, 1,4- dioxane, MeOH, ¾0; 3. 1-cyclopropyl-l ,2,3,4-tetrahydroquinoxaline, HATU, DIEA, DMT. Intermediate 76a methyl (2S,4R)~ 1 -[(2,5-dichlorophenyl)methyl]-4- methoxypyrrolidine-2-carboxylate: A solution of methyl (2S,4R)-4- methoxypyrrolidine-2-carboxylate ( 150 mg, 0.94 mmol, 1.00 equiv), 2-(bromomethyl)- 1 ,4-dichlorobenzene (243 mg, 1.01 mmol, 1 .07 equiv), and potassium carbonate (390 mg, 2.82 mmol, 2.99 equiv) in C3¾CN (5 mL) was stirred overnight at room temperature. The resulting solution was diluted with 30 mi of ethyl acetate then washed with 2x20 mL of brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was applied onto a silica gel column and eiuted with a mobile phase of petroleum ether/ethyl acetate (20: 1) to provide 260 mg (87%) of 76a as a colorless oil. MS (ES, m/z): 318 [M+Hf. 1H-NMR (300 MHz, CDCI 3 ) 6 7.52-7.49 (m, 1H), 7.27-7.19 (m, 1H), 7.15-7.11 (m, I II), 4.02-3.91 (m, 2H), 3.82-3.77 (m, 1 I i ). 3.67 (s, 3H), 3.60 (t, J 7.8 Hz, iff), 338-3.32 (m, IH), 3.26 (s, 3H), 2.54-2,49 (in, 1H), 2.19-2.15 (m, 2H).

Intermediate 76b (2S,4R)-l-[(2,5-dichlorophenyl)methyl]-4-methoxy- pyrrolidine-2-carboxylic acid: A solution of methyl (2S,4R)-l-[(2,5- dicMoropheny1)memyl]-4-methoxypyrrolidine-2-carboxylate (260 mg, 0.82 mmol, 1.00 equiv) in .1 ,4-dioxane MeOH/¾0 (6 mL) was added lithium hydroxide (69 mg, 1.6 mmol, 2.0 equiv). The resulting solution was stirred for 1 h at 80 °C in an oil bath. The pH value of the solution was adjusted to 6 with hydrogen chloride (2 M). The resulting mixture was concentrated under reduced pressure to furnish 300 mg (crude) of 76b as a colorless oil, which was used without further purification. MS (ES, m/z): 304 [M+H] r .

Example 76 l-cyclopropyl-4-[[(2S,4R)-l-[(2,5-dichlorophenyl)methyl]- 4-memoxypvrrolidm-2-yl]carbonyl]-l 5 2,3,4-tetohydroqxunoxalme: A solution of (2S,4R)-l-[(2,5-dicWorophenyl)memyl]-4-memoxypyiToHdme-2-ear boxylic acid (104 mg, 0.34 mmol, 1 .00 equiv), l-cyclopropyl-l,2,3,4-tetrahydroquinoxaline (60 mg, 0.34 mmol, 1.0 equiv), HATU (262 mg, 0.69 mmol, 2.00 equiv), and DIEA (89 mg, 0.69 mmol, 2.0 equiv) in DMF (3 mL) was stirred overnight at room temperature. The resulting reaction mixture was diluted with ethyl acetate (30 mL). The resulting mixture was washed with brine (3x20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude product (100 mg) was purified by preparative HPLC with the following conditions: Column, SunFire preparative CI 8, 19* 150mm 5μ η ι; Column, SunFire preparative CI S, 19* 150mm 5μπι; mobile phase gradient, water containing 0.05% TEA : CH 3 CN (40% CH 3 CN up to 56% in 6 min); detector, Waters 2545 UV detector at 254 and 220nm to provide 44.2 mg (28%) of the title compound bis-trifluoroacetate salt as a white solid. MS (ES, m/z); 460 [M + H] + ; l E~ NMR (300 MHz, CD 3 OD) δ 7.69 (s, IH), 7.47 (s, 2H), 7.23 (d„ J ~ 3 Hz, 2H), 6.98 (d, J = 9 Hz, IH), 6.76 (m, IH), 4.83 (m, IH), 4.68 (d, J = 12 Hz, IH), 4.55 (d, J - 15 Hz, I H), 4.09 (s, I f I ). 3.96 (t, J = 6 Hz, IH), 3.79 (d, J = 6 Hz, IH), 3.58-3.49 (m, 2H), 3.43-3.33 (m, 2H), 3.27 (s, 3H), 3.14 (t, J = 6 Hz, IH), 2.44 (t, J= 3 Hz, 2H), 2.12-2.07 (m, IH), 1.86 (m, IH), 0.88-0.84 (m, 2H), 0.66-0.63 (m, IH), 0.50-0.46 (m, IH).

Exampl 77

l-cyclopropyl-4-[[(2S,4S)-l-[(2,5-dic oroph^

yl] carbonyl] - 1 ,2,3 A -tetrahydroquinoxalme

Example 77 l-cyclopropyl-4-[[(2S,4S)-l-L(2,5-dichioroplienyl)ine11iyl]-

4-methoxypyrrolidin-2-yl]carbonyl]-l ,2,3,4-tetrahydroquinoxaline: Example 77 was prepared as described for example 76, substituting (2S,4S)-i-[(2,5- dicWorophenyl)methyl]-4-methoxypyrroiidine-2-c rboxylate for methyl (2S,4R)- 1 - [(2,5-dichlorophenyl)methyl]-4-methoxyp Trolidine-2-carboxylate. This resulted in 55.7 mg (35%) of the title compound bis-trifluoroacetate salt as a white solid. MS (ES, m/z): 460 [M+Hf : ! ! I-NMR (400 MHz, CD 3 OD) δ 7.79 (s, IH), 7.54 (s, 2.H), 7.28 (d, J = 8 Hz, 211). 7.06 (d, J ~ 8 Hz, IH), 6.80 (m, I H), 4.95 (m, IH). 4.67-4.56 (m, 2H), 4.07 (d, J - 12 Hz, 2H), 3.87 (d, J = 12 Hz, IH), 3.47-3.41 (m, 3H), 3.28 (s, 3H ) , 3.24- 3.15 (m, IH), 2.52 (m, 2H), 1.92 (m, I H), 0.94-0.89 (m, 2H), 0.70 (m, IH), 0.51 (m, IH). 71251

-cyc opropy - - j (2S,4 n-r(2,5- ic ^

Example 78 1 -cyclopropyl-4-[[(2S,4R)-l -[(2,5-dichlorophenyl)methyl]- 4-.fiuoropyrrolidin-2-y]]carbonyl]-l ,2,3,4-tetrahydroqumoxaline: Example 78 was prepared as described, for example 76, substituting methyl

2-carboxyIate for methyl (2S,4R)- 1 -[(2,5-dichlorophenyl)me l]-4- mefhoxypyrrolidine-2-carboxylate. This resulted in 19.3 mg (13%) of the title compound bis-trifluoroacetate salt as a blue solid, MS (ES, m/s): 448 [M+H] + ; 1H-NMR (400 MHz, CD 3 OD) δ 7.71 (s, 1H), 7.49 (s, 2H), 7.27 (d, J = 4 Hz, 2H), 7.06 (d, J- 8 Hz, 1 11). 6.80 (m, IH), 5.44-5.30 (m, i f f). 5.07-5.03 ( , IH), 4.68-4.52 (m, IH), 3,95- 3.63 (m, 4H), 3.46-3.40 (m, 2H), 3.27-3.23 (m, IH), 2.48 (s, IH), 2.28-2.23 (m, 2H), 0.92-0.86 (m, 2H), 0.66-0.54 (ra, 2H).

Example 79

l-cvclopropyl-4-(f4-}(2 -dichlorophenvi)methoxy]oxaK^-yl1carbonyl)-1.2.3.4- tetrahydroquinoxaline

Example 79 1 - cycloprc. pyl-4-([4-[(2.5-dichlorophenyl)metlioxy]oxari-4- yl]carbonyl)-l ,23,4-tetrahydroquinoxaline: Example 79 was prepared as described for example 8, substituting oxan-4-one for cyclopentanone. This resulted in 4.8 mg (4%) of the title compound trifluoroacetate salt as a white solid. MS (ES, m/z) 461 [M+ H] + ; 1H-NMR (400 MHz, CD 3 OD) δ 7.30-7.40 (m, 4H), 7.02-7, 13 (m, 21 f). 6.66-6.70 (m, I E), 4.55 (s, 2H), 4.07 (s, 2H), 3.78-3.87 (m, 4H), 2.12-2.38 (m, 5H), 0.76-0.80 (m, 2H), 0.50 i s. 2H).

Example 80

3-f2,5-dic oro-4-( l-f(4-cyclopropvl-l,2 .4-tetjahydroquinoxa3in-l- yl)carbonyl]cyclopropoxy methyl)phenyl]-N-met yl-N-[(2

peniahYdroxyhexyljpropanamide

Scheme 80 : 1 , 1- [(4-cyclopropyl- 1 ,2,3 ,4-tetr ahydroquinoxalin- 1 - yl)carbonyl]cyclopropa.n-l -ol, K 2 C0 3 , KI, DMF; 2. TMSBr, DCM; 3. (2R 5 3R,4R,5S)-6- (metliylamino)hexane-l ,2,3,4,5-pentaol, HA ' fU, DIEA, DMF. Intermediate 80a 3-[2,5-dichloro-4-([l-[(4-cyclopropyl-i ,2,3,4- tetrahydro-quinoxalin- 1 -yl)carbonyl]cyclopropoxy]meth.yl)phenyl]propatioate: A stirred solution of tert-butyl 3-[4-(bromomethyl)-2,5-dichlorophenyl]propanoate- (200 mg. 0.54 mmol, 1.00 equiv), 1 -[(4-cyclopropyl-l ,2,3,4-i.etrahydroquinoxalin-l- yl)carbonyl]cyclopropan-l -ol, 9a (140 mg, 0.54 mmol, 1.00 equiv), potassium carbonate (1 50 mg, 1.09 mmol, 2.00 equiv), and KI (18 mg, 0.11 mmol, 0.20 equiv) dissolved in DMF (2 mL) in a sealed tube was stirred overnight at 30 °C in an oil bath. The resulting reaction mixture was concentrated under reduced pressure and purified, by preparative TLC, with a mobile phase of petroleum ether/ethyl acetate (5: 1) to provide 130 mg (44%) of 80a as a light-yellow oil.

Intermediate 80b 3-[2,5-dichloro-4-([1 ~[(4~cyclopropyl-l ,2,3,4- tetrahydro-quinoxaiin- l-yl)carbonyl]cyclopropoxy]meth.yl)phenyl]propanoic acid: To a stirred solution of tert- butyl 3-f2 s 5-dichloro-4-([l-((4-cyclopropyl-1 ,2,3,4- tetrahydroquinoxalin- 1 -yl)carbonyl]cyclopropoxy]methyl)phenyl]propanoate (130 mg, 0.24 mmol, 1 .00 equiv) in dichioromethane (2 mL) was added TMSBr (2 nil,). The resulting reaction mixture was stirred for 2 h at room temperature, then concentrated under reduced pressure, then diluted with ¾0 (50 mL). The resulting mixture was extracted with ethyl acetate (3x30 mL) and the organic layers combined, dried over sodium sulfate, and concentrated under reduced pressure to provide 100 mg (86%) of 80b as a light yellow solid.

Example 80 3 - [ ,5 -diebloro-4-ί [ 1 - [(4-cycl opropyl- 1 ,2,3 ,4-teirahydro- quinoxalin-1 -yl)carbonyl

2,3,4,5,6-pentahydroxyhexyljpropanamide: A solution of 3-[2,5-dichloro-4-([l -[(4- cyclopropyl-l,2,3,4 etrahydroquinoxalm-l-yl)carbonyi]cyclopropoxy]m

propanoic acid (100 mg, 0.20 mmol, 1.00 equiv), (2R,3R,4R,5S)-6-

(methyIammo)hexane-L23,4,5--pento1 (60 mg, 0.31 mmol, 1.50 equiv), HATU (1 17 mg, 0.31 mmol, 1.50 equiv), and DIEA (53 mg, 0. 1 mmol, 2.00 equiv) in DMF (2 mL) was stirred overnight at room temperature. The resulting reaction mixture was concentrated under reduced pressure and the crude product residue (1 50 mg) was purified by preparative HPLC with the following conditions: Column, SunFire preparative C18, 19*150mm 5μπα; mobile phase gradient, water containing 0.05% " FFA : CH 3 CN (38.0% CJ¾CN to 56.0% over 6 min); Detector, Waters 2545 UV detector at 254 and 220nm to provi de 95 mg (70%) of the title compound trifluoroacetate salt as an off-white solid. MS (ES, m/zy. 666 [M+Hf : 1H-NMR (300 MHz, CD 3 OD) 5 7.27 (dd, J - 4.8 Hz, 2H), 6,99-7.04 (rn, 2H), 6.50-6.68 (in, 2H), 4.32 (s, 2H), 3.87-3.94 (m, 3H), 3.55-3.75 (m ; 3H), 3.27-3.36 (m, 3H), 2.89-3.06 (m, 5H), 2.60-2,63 (m, 2H), 2.24 (s, 1H), 1.39 (s, 21-1), 1.14-1.17 (m, 2H), 0.64-0.66 (m, 2H), 0.17 (s, 2H). Example 81

l-cyclopropyl-4-rrf4RV3-r(2,5^

yi]carbonyl]-l ,2,3,4-tetrahydro uinoxaiine

Example 81 (S)-(4-cyelopropyl~3,4-di yd.roqiiinoxaline-- 3 (2H)-yl)(l-

(2,5-dicWorobenzyl)-2-meihylpyrrolidin-2-yl)methanone: Example 81 was prepared using the procedure described for the preparation of example 12, except that (4R)-2.2- ditnethyl-l ,3-tiiiazolidine-4-carboxylic acid was used in place of (S)-l-[(benzyloxy) carbonyljpynOiidine-2-carboxylie acid isolated as the bis TFA salt, a white solid, MS (ES, m/z): 476 [M+Hf . 'T T- IR (300 MHz, CD 3 OD) δ 7.39-7.20 (m, 211), 7.17-7.1 1 (m, 411), 6.66 (br s, Hi), 4.21 (s, IH), 3.51 (br s, I B), 3.28 (br s, 1H), 3.04 (br s, 2! ! }. 2.25 (s, I H), 2.00-1.26 (m, 6H), 0.71. (br s, 211), 0.34-0.07 (br s, 2H).

Exam le 82

(2 S)-N-(2 -cy clopropoxyphenyl) - 1 - [(2 , 5 -di chl orophenyDmethyl] -N-methy me- -carboxamide

Example 82 (2 S)-N-(2-cyciopropoxyphenyl)- 1 - [(2, 5 - dich]oropi'ienyl)niethyl]- -methylpyrrolidine-2-carboxaniide: Example 82 was prepared as described for example 12 substituting 2-cycIopropoxy-N-methylanilir.ie 73g for 1- cyclopropyl- 1 ,2,3,4-tetrahydro-quinoxalme. This resulted in 24 mg (33%) of the title compound trifluoroacetate salt as an off-white solid. LC--MS- (ES, m/z): 467 [M+H] T ; 2012/071251

' i !-NM (300 MHz, CD3OD) 6 7.31 (d, J - 7.8 Hz, IH), 7.09 (s, 1 1 1). 7.05 (d, J Hz, 1H), 6.71 -6.66 (m, 3H), 4.33 (s, 211), 4.39-4.86 (m, 2H) 5 3.36-3.34 (m, 2H), 2.24 (m, IH), 1.41-1.38 (m, 2H), 1.214.1.1 (m, 2H).

Example 83

3 -(2,5-dichloro-4-(Y( 1 -f 4-cyclopropyi- 1 ,2.3 ,4-tetrahydroquinoxaline- 1 -

Scheme 83 : 1. h/C, EtOAc; 2. NaBH 4 , MeOH; 3. 4 M HCl in dioxane.

Intermediate 83a: tert-butyl 3~{2,5-dicWoro-4-formyipheiiyi)propanoate: Hydrogen gas was introduced to a stirred solution of tert-butyl (2E)-3-(2,5-dichloro-4- formylpheny])prop-2-enoate (3 g, 9.96 mmol, LOO equiv) and 30% Rli/ ' C (l .Og) in ethyl acetate (30 mL). The resulting solution was stirred for 5 h at room temperature under a hydrogen atmosphere then the solids were removed by filtration and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (30:1 ) as the eluent to provide 2.5 g (83%) of 83a as a colorless solid. MS (ES, m/z): (400MHz, DMSO ¾): δ 10.21 (s, I H), 7.81 (s, IH), 7.63 (s, IH), 2.92-2.99 (m, 2H), 2.55-2.62 (m, 2H), 1.36 (s, 9H).

Intermediate 83b: t-Butyl 3-(2,5-dichloro-4-(((l-(4-cyclopTopyl-1. 5 2,3,4- tetrahydroquinoxaline-l-carb^^ A mixture of (l-aminocyclopropyl)(4-cyclopropyl-3,4-dihydroqiunoxalin-l-( 2H)- yl)methanone free base (10.8 mg, 0.04 mmol), prepared form 26a by treating it with 4 M hydrochloric acid in 1,4-dioxane and then washed with saturated aqueous NaHCth, and t-butyl 3-(2,5-dic loro-4-formylphenyl)propanoate (12.5 mg, 0.04 mmol) in methanol (0.16 mL) was stirred at rt for I h. The mixture was cooled to 0 °C and to the mixture was added NaBHj (3.2 mg, 0.08 mmol). The resulting mixture was stirred at 0 °C for 15 minutes and at room temperature for 5 minutes. The addition of NaB¾ was repeated three more times. The reaction mixture was quenched with 1 M aqueous NaOH, extracted with EtOAc (3x). The combined organic layers were washed with brine (Ix), dried over sodium sulfate, concentrated and purified by column to give 18 mg of 83b as yellow syrup.

Example 83 3-(2,5-Dichloro-4-(((l -(4-cyclopropyI-l ,2,3,4- tetrahydroqu oxaline- 1 -carbonyl)cyclopropy l)aniino)methyl)phenyl)propanoic acid: To t-butyl 3 -(2,5 -dicbl oro-4-((( 1 -(4-cycloprop I - 1 ,2,3 ,4-tetrahy droquinoxal ine- 1 - carbonyl)cyclopropyl)amino)meihyl)phenyi) propanoate (21 mg, 0.039 mmol) was added 4 M hydrochloric acid in dioxane (2 mL). The mixture was stirred at room temperature for 2 and concentrated to give 14 mg (crude) of 3-(2 s 5-dichlaro-4-(((l~(4- cycl opropyl- 1.2,3 ,4-tetrahydroquinoxaline- 1 - carbonyl)cyclopropyl)armn.o)methyl)phenyl)propanoic acid as a solid. Some of the solid (4,7 mg) was purified by pre-HPLC to give 2.5 mg of 3-(2,5-dichloro-4-(((l-(4- cyciopropyl-l,2,3,4 ettahydroquinoxaline-l-carbonyl)cyclopropyl)- amino)methyl)phenyl)propanoic acid TFA salt as a yellow solid. Ή-NMR (400MHz, CD 3 OD) 6 7.37 (s, IH), 7.29 (dd, ./ 7.9, 1.5 Hz, IH), 7.23 (dd, J = 8.3, 1.4 Hz, IH), 7.15 (ddd, J= 8.4, 7.3, 1.5 Hz, IH), 7.10 (s, IH), 6.78 - 6.72 (m, IH), 3.94 - 3.83 (m, 4H), 3.44 (t, J= 5.8 Hz, 2H), 2.97 (t, J = 7.6 Hz, 2H), 2.58 ft, J - 7.6 Hz, 2H), 2.45 - 2.37 fro, I H), 1.38 (q, J = 5.0 Hz, 2H), 1.11 (dd, J - 7.6, 5.0 Hz, 2H), 0.86 - 0.77 (m, 2H), 0.54 - 0.45 (m, 2H). MS (ES, m/z): 488 [M+H] + . Example 84

l-rff2Syi-r[2-cMoro-5-(trffluo^

cyclopropyl- 1 ,2 ,4-tetrahydroquinoxaline

Scheme 84: 1. 2-chloro-5-( rifluoromethy])benzaldehyde. NaB¾CN, eOH.

Example 84 1 - [ [(2 S)- 1 -[[2-chloro-5-(trifluorometfayl)phenyl]methyl]- pyrrolidin-2~yl]caihonyl]-4-cyclopfopy -l .2,3 ,4-tetrahydroquinoxalinc: To a stirred solution oi l-c clopropyl-4-[[(2S)-pyrrolidin~2~yI]carbori lj-l tetrahydroquinoxaline (50 mg, 0.1 8 mmol, 1.00 equiv), 2-chloro-5- (triiluorometliyl)benzaldehyde (50 mg, 0.24 mmol, 1.30 equiv) in diehloromethane (4 mL) was added NaB¾CN (50 mg, 0.80 mmol, 4.32 equiv). The resulting reaction mixture was stirred overnight at room temperatisre then quenched by the addition of 10 ml. of water. The resulting solution was extracted with ethyl acetate (3x10 mL) and the combined organic layers washed with brine (10 mL), dried, over anhydrous sodium sulfate and concentrated under reduced pressure. The crude residue (60 mg) was purified by preparative HPLC with the following conditions: Column, SmiFire preparative CI 8, 19* 150mm 5μπι; mobile phase gradient, water containing 0.05% TFA : C CN (30.0% CH 3 CN up to 47.0% in 0 min); Detector, Waters 2545 UV detector at

254 and 220nm to provide 11.1 mg (13%) of the title compound bis-trifiuoroacetate salt as a yellow solid. MS (ES, m/z) 463 [M+Hf ; 1H- MR (300 MHz, C¾OD) δ 8.03 (s, 1H), 7.68-7.80 (m, 2H), 7.05-7.23 (m, 3H), 6.56-6.81 (m, 1H), 4.57-4.92 (m, 3H), 3.96- 4.08 (m, 1H), 3.13-3.63 (m, 6H), 2.06-2.45 (m, 511), 0.51-0.87 (m, 4! !). Example 85

l-([l-[(2.5-cHchlorophenyto

naphthyridine

Scheme 85: 1. NaH, DMF; 2. LiOH, THF, H 2 0; 3. oxalyl dichloride, catalytic DMF, DCM; 4. l,2 ,4-tetraliydro-L8--iiaphth3' idine ; Et N, DCM.

Intermediate 85a Methyl 1 -[(2,5-dichlorophenyl)metlioxy]cyclopropane- 1-earboxylate: To a stirred 0 °C solution of methyl l -hydroxyeyclopropane-I- carboxylate (1 16 mg, 1.00 mmol, 1.00 equiv) in DMF (4 mL) was added sodium hydride (60 mg, 1.50 mmol, 1.50 equiv, 60% in mineral oil) in several batches. The resulting reaction mixture was stirred for 0.5 h at 0 °C, then 2-(bromomethyl)-l,4- dichlorobenzene (238 mg, 0.99 mmol, 0.99 equiv) was added. The resulting reaction mixture was stirred for 1 h at room temperature and quenched by the addition of water (20 mL). The resulting solution was extracted with ethyl acetate (3x20 mL) and the combined organic layers washed with, brine (20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to provide 270 mg (98%) of 85a as a yellow oil.

Intermediate 85b l-[(2.5-dichiorophenyl)methoxyjcyclopropane-l- carboxylic acid: To a stirred solution of methyl l-[(2,5- dichlorophenyl)metiioxy]cyclopropane-l-carboxylate (270 mg, 0.98 mmol, 1.00 equiv) in tetrahydrofuran (5mL) and ¾0 (2 mL) was added LiOH (240 mg, 10.02 mmol, 10,21 equiv), in portions. The resulting solution was stirred overnight at 30 °C in an oil bath. The pH value of the reaction mixture was adjusted to 5-6 with hydrogen chloride (2.0 M) then extracted with ethyl acetate (3x20 mL) and the combined organic layers dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 250 mg (98%) of 85b as a while solid.

Intermediate 85c 1 -[(2,5-dichlorophenyl)methoxyjcyclopropane-l- carbonyi chloride: To a stirred solution of l-[(2,5- dichlorophenyr)methoxy] cyclopropane- l-carboxylic acid (100 mg, 0.38 mmol, 1.00 equiv) in dichloromethane (4 mL) containing a catalytic amount of DMF was added oxalic dichioride (145 mg, 1 .14 mmol, 3.00 equiv) dropwise. The resulting reaction, mixture was stirred for 1 h at room temperature was concentrated under reduced pressure to provide 100 mg (93%) of 85c as a yellow solid.

Example 85 l-([l-[(2,5-dichlorophenyl)xnethoxy]cyclopropyi3carbonyl)- l,2,3,4-tetrahydro-l,8-naprithyridme: To a stirred solution of l-[(2 5 5- dichlorophenyl)methoxy]cyclopropane- 1 -carbonyi chloride (100 mg, 0.36 mmol, 1.00 equiv), 1 ,2,3 ,4-tetrahydro- 1 ,8-naphmyridine (51 mg, 0.38 mmol, - 1.06 equiv), and dichloroni ethane (4 mL) was added trietliyl amine (77 mg, 0.76 mmol, 2, 13 equiv) dropwise with stirring. The resulting reaction mixture was stirred for 3 h at room temperature, then quenched by the addition of water (10 mL), extracted with dichloromethane (3x20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude residue (100 mg) was purified by preparative HPLC with the tbllowing conditions: Column, SunFire preparative CI 8, 19* 150mm 5μχη; mobile phase gradient, water containing 0.05% TFA : CH 3 CN (38.0% CH 3 CN to 56.0% over 6 min); Detector, Waters 2545 UV detector at 254 and 220nm to provide 18.2 mg (13%) of the title compound trifluoroacetate salt as a yellow solid. MS (ES, m/z): 377 [M+H ; 1H-NMR (300 MHz, CD 3 OD) δ 8.26-8.24 (m, 1H), 7.95-7.92 (m, 1H), 7.43-7.38 (m, 1H), 7.28-7.25 (m, 1H), 7.18-7.14 (m, 1H), 7.07 (s, I H), 4.59 (s, 2H), 4.15 (t, J = 6.0 Hz, 2H), 2.82 (t, J = 6.3 Hz, 2H), 2.04-1.96 (ui, 2H), 1.504.39 (m, 2H), 1 .32-1.29 (m, 2H). Example 86

-C i (2.5-diciiloropheriyito

naphthyridine

Example 86 l-([l-[(2,5-dichlorophenyl)methoxy]cyclopropyl]carbonyl)- 1 ,2,3 ,4-tetrahydro- 1 ,5-naphthyridine: Example 86 was prepared as described for example 85 substituting l,2,3,4-tetrahydro-l,5-naphthyridine for l ,2,3,4-tetrahydro-l,8- naphthyridine to provide 47.2 mg (33%) of the title compound trifluoroacetate salt as a white solid, MS (ES, m/z): 377 [M+H] + ; 1H-NMR (300 MHz, CD3OD) δ 8.51-8,54 (m, 1 11 ). 8.35-8,38 (m, III), 7.65-7.70 (m, 1 H), 7.17-7.32 (m, H ). 4.60 (s, 2H), 4.07-4.11 (m, 2H), 3.05 (t, J = 6.9 Hz, 2H), 2.04-2, 12 (rn, 2H), 1.30-1.45 (m, 4H).

Example 87

3-(2,5 ¾cMoro-4 [(2S)-2 (4-eyclop^

vDcafbonyllpynT> ffl~l -yl]m^

pentahydroxyhexyljpropanamide

Example 87 3-(2,5-dicMoro-4-[[(2S)-2-[(4-cyclopropyl-l,2 5 3,4- tetrahy droquinoxalin- 1 - yl)carbonyi jpyrrolidin- 1 -y l]methyl]phenyl)-N-[(2S,3R,4R.5R.)- 2,3,4, 5,6-penta ydroxyhexyrjpropanamide: Example 87 was prepared as described for example 48, substituting (2R,3R.,4R,5S)-6-aminohexane-l ,2,3,4,5-pentol for (2R,3R,4R,5S)-6-(methylamino)hexane-i ,2,3,4,5-pentaol to provide 30 mg (23%) of the title compound bis-trifiuoroacetate salt as a white solid. MS (ES, m/z): 665 [M+H] ' ; Ή-NMR (300 MHz, CD3OD) δ 7.52 (s, 1H) 5 7.27 (s, 2H), 7.11 (d, J - 7.5 Hz, Hi), 5.50 (s, 1H), 4.58 (m, 211), 4.23 (m, ! ! 0. 3.65 (m, 4H), 3.42 (m, 2H), 3.1 5 (d, ,7 - 7.2 Hz, 2H), 2.60 (m, 3H), 2.07 (in, 3H), 0.88 (m, 2H), 0.67 (m. 2H).

Exani le 88

y . ear ony cyc opropoxy memy p em^

pentahydroxyhexyl]propanarnide

Example 88 3-[2,5-dichloro-4-([1 -[(4-cyclopropyl-l } 2,3,4- tetrahydroquinoxalin -yl)carb^^

2,3,4,5,6-pentahydroxyhexyljpropanamide: Example 88 was prepared as described for example 80, substituting (2R,3R,4R,5S)-6-aminohexane-l ,2,3,4,5-pentol for (2R 3 3R,4R,5S)-6~(methyIamino)hexaiie-L2 5 3,4,5-pentaol to provide 84.6 mg (40%) of the title compound irifluoroacetate salt as a pink solid. MS (ES, m/z): 652 [M+H] + ; Η- NMR (300 MHz, CD 3 OD) δ 7.27-7.32 (m, 2H), 7.01 -7.07 (m, 2H), 6.72 (t, J = 8.4 Hz, 2H), 4.36 (s, 2H), 3.92 (t, J - 5.4 Hz, 21 iL 3.32-3.81 (m, 9H), 2.96 (t, J - 7.5 Hz, 2H), 2.46-2.51 (m, 2H), 2.27 (t, J= 3.3 Hz, H I), 1.43 (s, 2H), 1.19 (del, J= 7.5 Hz, 2H), 0.68 (t, ./ ~ 8.1 Hz, 2\ \ ), 0,20 is. 21 1 ).

Example 89

(SV(4-CYclopropyi-3.4-dmydroqumoxalin-l(2ffl-yl)n-(2.5-dich1 oro- methoxybenzy )pyrrolidin-2-yl methanone

Example 89 (4~cyclopropyl-3 ,4-dihydroquinoxalin- 1 (2H)-yl)( 1 -( 1 -(2,5 - dichlorophenyl)ettioxy)cyclopropyl)methaiione: Example 89 was prepared as described for example 84 substituting 92a for 2-chloro-5-(trifluoromethy])benzaldehyde to provide 89 as the bis-trifluoroacetate salt. MS (ES, m/∑): 460 [M+H] + .

Example 90

(4-cyciopropyl-3^-dibydroqumoxalm^

y1)methanonc

Example 90 (4-cyclopropyl-3 } 4-dihydroquinoxalin-l(2H)-yl)(3-(2,5- dichlorobenzylamino)oxetan-3~yl)meihanone: Example 90 was prepared as described for example 26 substituting 3~(teri-bittoxyearboiiyla.mino)oxetarj.e-3-carboxyHc acid for l-(tert-butoxycarbonylamino)cyclopropanecarboxylic acid to provide 90. MS (ES, m/z): 432 [M+H ; 1H-NMR ( 00MIIz, CD 3 OD) δ 7.48 is, 2H), 7.36 (d, J = 8.5 Hz, 1H), 7.25 (dd, J ------ 8.5, 2.6 Hz, 1H), 7.18 (d, J= 8.0 Hz, 1H), 7.08 (s, 1H), 6.67 ( J= 8,2 Hz,

1H), 5.08 (s, 2H), 4.66 (s ( 2H), 3.95 - 3.59 (m, 4H), 3.39 (s, 2H), 2.48 ■■■■ 2.32 (m, iH), 0,87 - 0.74 (m, 2H), 0.50 is, 2H).

Exam le 91

3-(2.5-dichloro^-((l-(4-cyd^

carbonyl)cyc3opropylamm^

pcntaliydroxyhexyl)propanamide

Example 91 (4-cyelopropy]-3,4-dihydroqumoxalir:-l(2I-i)-yi)(3-(2,5- dichlorobenzyiamino)oxeian-3-yl)nietharione: Example 91 was prepared as described for example 80 substituting 83 for 80b to provide 91 as the TEA salt. ' i l-NMR. (400MHz, CD3OD) δ 7.40 (d, J = 2.7 Hz, 1H), 7.29 (dd, J - 7.9, 1.4 Hz, 1 H), 7.26 - 7.21 (m, 1H), 7.19 - 7.11 (m, 2H), 6.78 - 6.72 (m, 1H), 3.99 - 3.85 (m, 5H), 3.77 (dd, J - 11.0, 3.4 Hz, lH), 3,73 - 3.57 (m, 5H), 3.45 (t, J= 5.8 Hz, 2H), 3.42 - 3.32 (m, lH), 3.09 (s, 1.5H), 2.99 (t, J = 7.8 Hz, 2H), 2.96 (s, 1.5H), 2.91 - 2.72 (m, 3 H), 2.71 - 2.64 (m, 1H), 2.47 - 2.37 i rn. l i l t. 1.37 (q, . = 5.1 Hz, 2H), 1.18 - 1.09 (m, 2H), 0.88 - 0.79 (m, 2H), 0.56 - 0.47 (m, 2H). MS (ES, m zy. 665 [M+H] ' " .

Example 92

-(4-(3-aininopropylV2,5-dichk robenzylamino)cvclopropyi)(4

dihydroq uinoxai in- 1 (2R) -yl)methanone

Scheme 92: 1. Dichloro(methoxy)mei atie,TiCl4, DCM; 2. LiCl, DMF; 3. Tf 2 0, TEA, DCM; 4. N-(prop-2-yn-l-yl)carbamate, Pd(dppi)Cl 2 , Cui, 2 C0 3 , DMF; 5. Kh/C, H 2 , ethyl acetate. 6. NaBH 4 , MeOH. 7. 4 M HC1 in dioxane.

Intermediate 92a 2,5-dichloro-4-methoxybenzaldehyde: To a stirred 0 °C solution of 1 ,4-dichloro-2-methoxybenzene (25.0 g, 141.2 irimol, 1.00 equiv) and TiCL f (30.9 nil.) in dichloromethane (300 inL) was added dichloro(methoxy)methane (16.2 g, 140.9 mmol, 1.00 equiv) dropwise. The resulting reaction micture was stirred for 2 h at 60 °C then quenched by the addition of water/ice. The pH value of the solution was adjusted to 1.0 with concentrated HC1 extracted with ethyl acetate (4x500 ml,) and. the combined organic layers washed with brine (2x500 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide 31.0 g (crude) of 92a as a yellow solid.

Intermediate 92b 2 s 5-dichloro-4-hydroxybenzaldehyde: A solution of 2.5-dichioro-4-methoxybenzaldehyde (14.0 g, 68.3 mmol, 1.00 equiv), LiCl (11.6 g, 274 mrnol, 4.00 equiv) in DMF (150 mL) under an inert atmosphere of nitrogen was stirred overnight at 140 °C in an oil bath. The reaction mixture was then quenched by the addition of water/ice and the pH value of the solution was adjusted to 1 -2 with concentrated HQ. The resulting solution was extracted with ethylacetate (3x400 mL) and the combined organic layers were dried over anliydrous sodium sulfate and concentrated under reduced pressure. The resulting residue was purified using silica gel column chromatography with a ethyl acetate/petroleum ether (1 :10-1:5) gradient to provide 10.0 g (77%) of 92b as a light yellow solid. (300Hz, DMSO<¾): 8 1 1 .99(s, IH), 10.08(s, 111), 7.8 l is. I H ). 7.09(s, IH).

Intermediate 92c 2,5-dichloro-4-form.ylphenyl trilluoromethanesulfonate: To a stirred 0 °C solution of 2,5-dichIoro-4- hydroxybeiizaldehyde (3.0 g, 15.71 mmol, 1.00 equiv) and triethylamine (3.2 g. 31.62 mmol, 2.00 equiv) in dichloromethane (50 mL) was added a solution of trifluoromefhanesulfonic anhydride (6.8 g, 24.10 mmol, 1.50 equiv) in dichloromethane (10 mL) drop wise. The resulting reaction mixture was stirred for 30 min at. room temperature then washed with brine (2x30 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification by silica gel column chromatography with an e!uent gradient of ethyl acetate/petroleum ether (1 :50-1 : 10) provided 3.0 g (59%) of 92c as a white solid. 1H- NMR (300Hz, DMSC g): 10.22(s, IH), 8.14-8.15(m, 211).

Intermediate 92d tert-butyl N-[3-(2,5-dichloro-4-fonnylpheny!)prop-2- yn-i -yl] carbamate: A solution of 2,5-dichIoro-4-formylphenyi trifluoromethanesulfonate (5.0 g, 15.48 mmol, 1.00 equiv), tert-butyl N-(prop-2-yn-l - yl)carbamate (2.4 g, 15.46 mmol, 1.00 equiv), potassium carbonate (4.1 g, 29.7 mmol, 2.00 equiv), Pd(dppf)Cl 2 (1 .2 g, 1.64 mmol, 0.10 equiv) and Cul (290 mg, 1.52 mmol, 0.10 equiv) in DMF (45.0 mL) was stirred overnight at room temperature under an inert N2 atmosphere. The resulting reaction mixture was diluted with water (150 mL) extracted with ethyl acetate (3x150 mL) and the combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification of the residue by silica gel column chromatography with an eiuent gradient of ethyl acetate petroleum ether (1 :15-1 :10) provided 2.0 g (39%) of 92d as a light yellow solid.

Intermediate 92e tert-hutyl N-[3-(2 5 5-dichloro-4-formylphenyl)propy[]- carbamate: A solution of h/C (1.5 g), tert-hutyl N-[3-(2,5-dichloro-4- fomiylphenyl)prop-2-yn-l -yl]carbamate (3.0 g, 9.14 mmol, 1.00 equiv) in ethyl acetate (45 ml.) was stirred overnight under a hydrogen atmosphere at room temperature. Solids were removed from the reaction mixture and the filtrate was concentrated under reduced pressure. Purification of the resulting residue by silica gel column chromatography with an eiuent gradient of ethyl acetate/petroleum ether (1 :20-1 : 10) resulted in 2.4 g (79%) of 92e as a white solid. 1H-NMR (300Hz, DMSCWtf): 10.20(s 5 IH), 7.81 (s, IH), 7.68(s, IH), 6.90-6.94(m, IH), 2.93-2.99(m, 2H), 2.71 -2.76(m, 2J 1). 1.66-1.73(m, 2H), 1.37(s, 9H).

Example 92 3-(3-(2,5-dicWoro-4-((l-(4-cyclopropyl-l,2,3 ) 4-tetrahydro- quinoxaline- 1 -carbonyl)cyclopropylam.ino)methyl)phenyl)propyl)-l -methyl- 1 - ((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl)urea: Example 92 was prepared as described for example 83 substituting tert-butyl N-[3-(2,5-dichloro-4- formylphenyl.)propyl] carbamate 92e for 83a tert-butyl 3-(2,5-dichloro~4- formylphenyl)propanoate to provide 92 as the TFA salt. 1H-NMR (400MHz, CD 3 OD) 5 7.34 - 7.29 (m, 2H), 7.22 (dd, ,7 - 8.3, 1.4 Hz, IH), 7.16 - 7.10 (m, 1H), 6.99 (s, 1H), 6.74 (td, J = 7,7, 1.4 Hz, IH), 3.90 (t, J - 5.8 Hz, 2H), 3.70 (s, 2H), 3.45 (t J = 5.8 Hz, 2H), 2.95 (t, J— 8.0 Hz, 2H), 2.77 (t, J = 8.0 Hz, 2H), 2.47 - 2.36 (m, IH), 1.97 - 1.85 (m, 2H), 1.40 (q, J~ 4.5 Hz, 2H), 1.00 (q, J- 4.5 Hz, 2H), 0.83 - 0.75 (m, 21 1). 0.51 - 0.41 (m, 2H). MS (ES, m/z): 473 [M+H] + . Example 93

3-f3-(2,5-dicMoro-4-((l-(4-cy^

carbonyl)cyclopropylamino)metiiyl)phenyl)propyl)- 1 -methyl- 1 ~i(283R,4R,5 R)- -pentahydroxy hexy I)ur ea.

Ex mpl e 93 3 -(3 - (2 , 5 -dichloro-4 -((1 -(4-cyclopropyl- 1 ,2,3 ,4-tetrahydro- quinoxaline- 1 -carbonyl)cyc1opropylajacdno)meti .yl)pb.enyl)propyl)- 1 -methyl- 1 - ((2S > 3R,4R,5R)-2,3,4,5,6-penla ydroxyhexyl)i3rea: Example 93 was prepared as described for example 69 substituting 92 for 68 to provide 93 as the TFA salt. MS (ES, m/z): 694 [M+fff ; Ή-NMR (400MHz, CD 3 OD) δ 7.36 (s, 1H), 7.29 (dd, J = 7.9, 1 .4 Hz, 1 H), 7.23 (dd, J- 8.3, 1.4 Hz, HI), 7.19 - 7.12 (m, 1H), 7.10 (s, 1H), 6.75 (td, J= 7.6, 1.4 Hz, 1H), 3,98 - 3.86 (m, 5H), 3.77 (dd, J - 10.9, 3.2 Hz, 1H), 3.73 - 3.57 (m, 4H), 3.49 - 3.41 (m, 3H), 3.35 (d, J = 8.0 Hz, IH), 3.18 (t, J ~ 6.9 Hz, 2H), 2.94 (s, 311), 2.77 - 2.66 (in, 2H), 2.46 - 2.37 (m, 1H), 1.82 - 1.72 (m, 2H), 1 ,38 (q, J= 5.0 Hz, 2H), 1.13 (q, J= 5.1 Hz, 2Π). 0.86 - 0.78 (m, 2H) S 0.55 - 0.46 (m, 2H).

Ae 94

(4-cyclopropyl-3 ,4-diliydroquinoxalin- 1 (2H)-yl)(l-< -(2,5- dic oroph.enyl)etfaoxy)cyclopropyl)methanone

Example 94 (4-cyclopropyl-3 ,4-dihydroquinoxalin-l (2H)-yl)( 1 -( 1 -(2,5- dichlorophenyl)ethoxy)cycIopropyl)methanone was prepared as described for example 9 substituting 1 -(2,5~dieliloropheny3)ethyl metlianesulfonate for 2-(bromomeihy 1)- 1 ,4- dichlorobenzene to provide 94 as the TFA salt. MS (ES, m'z): 431 [M+H] 1 . N.jV'-r q'-fethaae-l ,2-diylbis( ' oxy))bis(e1faane-2, 1 Hvmbisf3-g5→iichloro-4-ft cyclopropyl-l,2,3,4-tetrahydroquinoxalinc-l- carbonyl)cyclopropyiamino)mcij yl)phenyl)propanamide)

Example 95 N,N' 2,2' ethane-l,2-diylbis(oxy))bis(ethme-2 -diyl^ dichloro-4-(( 1 -(4-cyclopropyl- 1 ,2, 3 ,4-tetrahydroq inoxaline- 1 -carbonyl)cyclopropyl- amino)methyl)-phen l)propanamide) : Example 95 was prepared as described for example 91 using example 83 as the starting material and one half of an equivalent of 2,2'-(ethane-l^-diylbis(oxy))-diet3ianamine in place of N-methyl-D-glucamiae to provide 95 as the TFA salt. MS (ES, m/z): 1089 [M+H] + .

Scheme 96: 1. NaBH 4 , MeOH; 2. NBS, PPh 3 , DCM, THF; 3. NaH, DMF; 4. HCl/Dioxane 5. Ν,Ν' -disuccinimidyl carbonate, THF.

Intermediate 96d 1 -(4-(3-amin.opropyl)-2,5-dichlorobenzyloxy)-N-(2- cycJopropoxyx>henyl)-N-methy!cyclopropaiiecarboxamid.e: Intermediate 96d was prepared as described for intermediate 80b substituting N-(2-cyclopropoxyphenyl)-l- hydroxy-N-metiiylcyclopropane-1. -carboxamide (73h) for 1 -[(4-cyclopropyl- 1.2,3, 4~ tetrahydroqiimoxalin-l-yl)carbonyl]cyclopropan-l-ol in step 1 to provide intermediate 96d.

Example 96 iV-(2-cyclopropoxyphcnyl)- 1 -(2 5 5-dichloro-4-(3-(3- ((2S R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl) ireido)propyl)benzyloxy)-N- methykyclopropanecarboxamide: Example 96 was prepared as described for example 69 substituting (2R,3R,4R,5S)-6-aminohexane-l ! 2,3,4,5-pentol for (2R,3R,4R,5S)-6- (methylamino)hexane-l ,2,3,4 5 5-pentaol to provide the title compound. MS (ES, m/z); 670 [M+H] + ,

Example 97

3 -(2,5-dk lorp-4-(((R)-4-(4-cyclopropyl- 1 ,2,3 ,4-te rahydroquLnoxaline- 1 - earbonv])thiazolidio-3-yl)metind)phenyl)~N-i S3R,4R,5R -23.4,5 j--

Example 97 3-(2,5-dichloro-4-(((R)-4-(4-cyclopropyl-l ,2,3,4- tetrahydroquinoxaHne-1 -carbon^^ ^

2^A5,6-pentahydroxyhexyl)propanamide: Example 97 was prepared as described for example 48 substituting (R)-(4-cyciopropy !-3,4-dihydroquinoxalin-l (2H)- y].)(thiazolidin » 4-yl)methanone tor (S)-(4-cyclopropyl-3,4-<iihydroqirbioxalin-l(2H)- yl)(pyrroiidin-2-yl)metba >ne in step 6 and (2R,3R,4R,5 S)-6-aminohexane- 1 ,2,3,4,5- pentol for (2R^R,4R,5S)-6-(methyiainino)hexane-i,2,3,4,5-pentoi in step 8 to provide 97 as the bis TEA salt. MS (ES, /z) 683 [M+H] + .

Example 98

3-(2,5-dic oro-4-(((R)-4-(4-cyclopropyl- 1 ,23,4-tctrab.ydroquinoxalirie- 1- earbonyl)thiazoUd^

pentahydroxyhexyl)propanarnide

Example 98 3-(2,5-dichloro-4-((( )-4-(4-cyclopropyl-l ,2,3,4- tetrahydroquiiioxaline- 1 -carbonyI)thiazolidm-3-y l)methyl)pheayl)-N-methyl-N- ((2S,3R,4S,5R)"2,3,4,5,6-pentahydroxyhexyl)propanamide: Example 98 was prepared as described for example 48 substituting (R)-(4-cyclopropyl-3,4-dihydroquinoxalin- 1 (2H)-yl)(ihiazolidin-4-yl)methanone for (S)-(4-cyclopropyl-3.4-dihydroqmnoxalm- l(2H)-yI)(pyiTolidin-2-}i)rriethanorie in step 6 and (2R,3S,4R.,5S)-6- (niethylamino)hexane-l .,2,3,4,5-pentaol for (2R,3R,4R,5S)-6-(m.ethy]a.mino)liexane- 1,2,3,4,5-pentol in step 8 to provide 98 as the bis TFA salt. MS (ES, m ): 697 [M+H ,

Examples 99-158

Compounds 99-1.58 are prepared from commercial or known starting materials according to the general methods described in Examples 1-98 and methods known to those skilled in the art.

- 5 - - 3-{3-[2,5~dichloro-4- 1

({ l -[(4-cycIopropyl- ! 1.23,4·· i tetrahydroquinoxalin- 1 - y].)carbonyl]cyclopropo xy } methyl)phenyi] prop vl}-W3-(4-{3-[({3- '

125 [2,5-dichloro-4-({l-[(4- cyciopropyl- 1 ,2, ,4- tetrahydroquin oxalin- 1 - yl)carbonyl]cyclopropo j xy } methyl)pheny 1 ] prop yl}carbaraoyl)amino]pr opyl}piperazin-l - yl)propyl]urea

3-[3-(2,5-dich!oro-4-

{[(2S)-2-[(4- cycloprop l- 1 ,2,3,4- tetrahydroquinoxaliii- 1 - i yl)carbonyl]pYrrolidin- 1- " vl]met yl}p eny] prop ' l j.. i -(3- i4-[3-i ; |3- (2,5-dichloro-4-{[(2S)-

126

2-[(4-cyelopropyi-

1 ? 4- tetraliydroquinoxailn- 1 -

. o yl)carbonyl]pyrrolidin- " 1 - ' yl] methyl } phenyl)prop yijcarbamoyl} amino)pr opyl]piperazm-l- yl}pro yl) rea

3-{3-[2,5-dichloro-4-

({ 1 -[(4-cyclopropyl- 1,2,3,4- tetrahydroquinoxalin- 1 - yl)carbonyl]cyclopropo xy}methyl)phenyl]prop yl}-l-f2-(2-{2-[(f3-

127

[2,5-dichloro-4-({l -[(4-

» o,-"¾ cyclopropyl-1 ,2,3,4- y y ' tetrahydroquinoxalin- 1 - yl)carbon.yi]cyclopropo x } methy3)phenyl]prop

V-i yl}carbamoyl)amino]et hox } eiiioxy)ethyi jurea

y)ethyljpropanamide 1251

Example 159

hydroxymethyl)benzyi)thiazolidin-4-yl)methanone

Scheme 159: 1. (Boc) 2 0, NaOH, dioxane, H 2 0; 2. 1 -cyclopropyl- 1 ,2,3,4- tetrahydroquinoxaline, HATU, DIE A, DMF; 3. Cone. HC1; 4. NBS, benzoyl peroxide, CCI 4 ; * 5. AgN0 3 , acetone, H 2 0; 6. 159c, Na(OAc) 3 BH, DCE.

Intermediate 159a (4 )-3-[(tert-butoxy)carbonyl]-13-iMazolidine-4- carboxylic acid. To a solution of (4R)-l,3-thiazolidine-4-carboxylic acid (9 g, 67.58 mmol, 1.00 equiv) in. dioxane (100 mL) was added sodium hydroxide (8.1 g, 202,5 mmol, 3.00 equiv) in water (350 mL) and then (Boc) 2 0 (22 g, 100.8 mmol, 1,49 equiv). The resulting solution was stirred overnight at room temperature. The pH value of the solution was adjusted to 4 with hydrogen chloride (1 mof/L) and was then extracted with ethyl acetate (3 x 250 mL). The combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous sodium sulfate and concentrated under vacuum to afford 15 g (95%) of 159a as a white solid with was used without further . purification.

Intermediate IS9b (tert-buiyl (4R)~4 i4-cyclopropyl-L2,3,4- tctrahydroquinoxalin-l -yI)carbonyl]-l ,3-thiazoiidine-3-carboxylate, A solution of (4R)-3-[(tert-butoxy)carbonyl]-l > 3-thiazolidine-4-carboxylic acid (8.0 g, 34.29 mmol, 1.00 equiv), l-cyclopropyl-l,23,4-tetxahydfoqiiinoxalirie (6 g, 34.43 mmol, 1.00 equiv), HATU (17 g, 44.71 mmol, 1.30 equiv), and DIEA (6.7 g, 51.84 mmol, 1.51 equiv) was stirred in DMF (80 mL) overnight. The resulting solution was diluted with 1¾0 (500 mL), extracted with ethyl acetate (2 x 250 mL) and the combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous sodium sulfate, and concentrated under vacuum. The residue was purified by silica gel column chromatography with an eluent gradient of petroleum ether/ethyl acetate (20:1 to 10:1) to furnish 159b (12 g, 90%) as a yellow oil.

Intermediate 159c l-cyclopropyl-4-[[(4R)-l,3-thiazolidin-4-yl]carbonyi]- 1 ,2,3,4-tetrahydroquinoxaline. To a solution of tert-butyl (4R)-4-[(4-cyciopropyl- l,2,3,4-tetrahydroquinoxalin-l-yl)carbon^ (10 g,

25.67 mmoi, 1.00 equiv) in 1.4-dioxane (150 mL) was added concentrated HCl (50 ml , ). The resulting solution was stirred for 1 h at room temperature, then the pH value of the solution was adjusted to 6-7 with aqueous sodium hydroxide and the resulting solution, was extracted with ethyl acetate (2 x 300 mL). The organic layers were combined, washed with brine (3 x 500 mL), dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by silica gel column chromatography with an eluent gradient of ethyl acetate/petroleum ether (1 : 10 to 1 :4) to furnish 159c (4.98 g, 67%) as light yellow oil. MS (ES, m/z); 290 [M + H] + . 1H- NMR (400MHz, CDCI 3 ): 7.28-7.11 (m, 3H), 6.74-6.70 (m, 1H), 4.45-4.43 (d, J==9.6Hz, 1H), 4.14-4.00 (m, 3H), 3.80-3,77 (m, IH), 3.44-3.41 (t, J-5,6, 6Hz, 2H), 2.97-2.93 (t, J==9.6, 6.8Hz, IH), 2,74-2.69 (t, J=9.6,9.2Hz, 2H), 2.48-2.44 (m, IH), 0.88-0.84(m. 2H), 0.69-0.60(m, 211).

Intermediate 159d: l,4-dichLoro-2,5-bi.s(dibromomethyl)benzene. To a solution of L4-dichloro-2,5-dimethylbenzene (5 g, 28.56 mmol, 1.00 equiv) in CC (50 mL) was added NBS (25.4 g, 142.71. mmol, 5.00 equiv) and benzoyl peroxide (490 mg, 2.02 mmol, 0.07 equiv) and the resulting solution was stirred overnight at 80 °C in an oil bath, The solids were filtered out, the filter cake was washed with 4 x 100 mL of ethyl acetate, and the organic layers were combined, washed with 2 x 100 mL of water, 1 x 150 ml., of saturated a 2 S 2 0 3 and 1 x 150 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum to give 15 g of intermediate 159d as a light yellow solid, which was used without further purification.

Intermediate 159e: 2,5-dichlorobenzene-l,4-dicarbaIdehyde. To a solution of intermediate 159d (15 g, 30.57 mmoi, 1.00 equiv) in acetone (100 mL) was added a solution of AgN0 3 (21.8 g, 128.31 mmol, 4.20 equiv) in water (30 mL) dropwise with stirring at 65 °C. The resulting solution was stirred for 2 h at 65 °C in an oil bath. The resulting solution was diluted with 500 mL of ethyl acetate. The resulting mixture was washed with 1 x 100 mL of water, 1 x 120 mL of hydrogen chloride ( IN), 1 x 100 mL of NaHC0 3 (sat.) and 1 x 100 mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :20~EA) to afford 2.5 g (40%) of intermediate 159e as a light yellow solid.

Example 159: (R (4~eye]opropyS-3,4~dihyd

dichloro~4-(hydroxwnethyl)ben^ To a solution of mtermediate 159e (2.4 g, 11.82 mmol, 1.20 equiv) in 1,2-dichloroethane (60.0 mL) was added. 159c (3.0 g, 10.37 mmol, 1.00 equiv) and the mixture was stirred for 1 h. To this was added NaBH(OAc)3 (8.8 g, 41.53 mmol, 4.00 equiv) in several batches and the resulting solution was stirred overnight. The reaction was then quenched by the addition of 100 mL of water and extracted with 3 x 100 mL of dichloromethane. The combined organic layers were washed with 1 x 100 mL of brine, dried over anhydrous sodium sulfate, and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :20-1 :8) to afford 2.1 g (42%) of Example 159 as a while solid.

Example 160

(¾)-(3-(4-(5-aimnopentyl)-2,5-di^

dihydroqmnoxaim-l(2H)-yl)methanone

,O s -— - ? . . . << <:

160b 160c

MeOH; 7. NBS, PPh 3 , THF/DCM; . 159c K 2 C 3 , ; . oxane.

Intermediate 160a: pent-4-ynyl methanesulfonate. To pent-4-yn-l-ol (.10 g, 118.88 mmol, 1.00 equiv) in DCM (150 mL) at 0 °C was added TEA ( 18.04 g, 178.28 mmol, 1.50 equiv) followed by the drop-wise addition of a solution of methaiiesulronyl chloride (16.36 g, 142.82 mmol, 1.20 equiv) in DCM (50 mL) and the resulting solution was stirred for 1.5 h. The reaction was quenched by the addition of water (100 mL) and then extracted with 2 x 100 mL of dichloromethane. The organic layers were combined, washed with 1 x 200 irsL of sodium biearbonate(sat). 1 x 250 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 18.05 g (94%) of intermediate 1 0a as a brown oil.

Intermediate 160b: pent-4-yn-l -amine. To intermediate 160a (1 1.4 g, 70.28 rnmol, 1.00 equiv) in a 250 mL high-pressure seal able tube was added liquid ammonia (60 mL), the tube was sealed, and the mixture was stirred overnight at 80 °C. The reaction mixture was then cooled to 0 °C and the tube opened, the contents diluted with 150 mL of ether, the mixture was filtered and then the filtrate was concentrated under vacuu to afford 4.91 g (84%) of intermediate 160b as brown oil.

intermediate 160c: tert-butyl pent-4-ynylcarbamate. To intermediate 160b (4.91 g, 59,06 rnmol, LOO equiv) in DCM (40 mL) at 0 °C was added TEA (8.95 g, 88.45 mmol 5 1.50 equiv) followed by the drop-wise addition of a solution of di-tert-butyl dicarbonate (12.88 g, 59.02 rnmol, 1.00 equiv) in DCM (20 mL). The resulting solution was allowed to warm to RT and then stirred overnight at room temperature. The mixture was concentrated under vacuum and then purified via silica gel chromatography (ethyl acetate/petroleum, ether 1 :50-1 :40) to afford 4.59 g (43%) of intermediate 160c as a light yellow oil.

intermediate 160d: tert-butyl 5-(2,5-dichioro-4-formyiphei l)pent-4- ynyicarbamate. To intermediate 92c (7.71 g, 23.86 rnmol, LOO equiv) in DMF (100 mL) was added intermediate 160c (4.59 g, 25.05 rnmol, 1.05 equiv), Pd(PPh 3 ) 2 Ci 2 (1.67 g, 2.38 rnmol, 0.10 equiv), Cul (450 rng, 2.36 rnmol, 0,10 equiv) and DIEA (6.61 g, 51.15 rnmol, 2.00 equiv) and the resulting solution was stirred overnight. The mixture was diluted with 500 mL of ethyl acetate, washed with 3 x 200 ml. of brine and the organic layer was dried over sodium sulfate and then concentrated under vacuum. The residue was purified via silica gel chromatography (petroleum ether/ethyl acetate. 50:1- 10:1) to afford 3.7 g (44%) of intermediate 160d as a brown syrup.

Intermediate 160e: tert-butyl 5-(2,5-dic oro-4-formylphenyl)pent.ylcarbamate.

To intermediate 160d (3.21 g, 9.01 mraol, 1.00 equiv) in ethyl acetate (90 mL) was added Rh/C (3.60 g) and the suspension stirred under a hydrogen atmosphere overnight The solids were filtered out and the filtrate was concentrated under vacuum to afford 3.1 g (95%) of intermediate 160e as a brown oil.

Intermediate 160f: tert-buiyl 5-(2 J 5-dichloro-4-(hydroxymetliyl)phenyl)pentyl- carhamate. To intermediate 160e (3.1 g, 8.60 mrnol, 1.00 equiv) in methanol (100 mL) at 0 °C was added portion-wise NaB¾ (810 mg, 21.41 rnmol, 2.49 equiv) over 30 min. The resulting mixture was stirred for 1 h at 0 °C, then quenched by the addition of 50 mL of water. The mixture was concentrated under vacuum to remove the organic solvents, then extracted with. 3 x ! 00 mL of dichloromethane. The organic layers were combined, washed with 3 x 100 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 2.70 g (87%) of intermediate 160f as light yellow oil.

Intermediate 160g: tert-butyl 5-(4-(bromomethyl)-2,5-dichlorophenyl)pentyi- carbamate. To intermediate 160f (250 mg, 0.69 mrnol, 1.00 equiv) in DCM/THF (2/2 ml.) at 0 °C was added NBS (235 mg, 1.32 mrnol, 1.90 equiv) followed by the batch- wise addition of triphenyiphosphine (373 mg, 1.42 mrnol, 1.50 equiv). The reaction was allowed to warm to RT and then stirred for 1 h. The resulting mixture was concentrated under vacuum and the residue purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 :50) to afford. 173 mg (59%) of intermediate 160g as light yellow oil.

Intermediate 160h: (R)-tert-b tyl 5-(2,5-dichloro-4-((4-(4-cyclopropyl-l > 2,3 > 4- tetrahydroquinoxdine-l-carbonyl)^ To intermediate 160g (3 g, 7.06 mrnol, 1 .00 equiv) in DMF (10 mL) was added intermediate 159c (2 g, 6.91 mrnol, 1.00 equiv) and potassium carbonate (2 g, 14.47 mrnol, 2.00 equiv) and the reaction was stirred overnight. The mixture was diluted with 20 mL of water, extracted with 3 x 30 ml, of ethyl acetate, the organic layers combined, washed with 1 50 ml, of brine and then dried. The solution was concentrated under vacuum and. the residue was purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 :20) to afford 1.7 g (38%) of intermediate 160h as a brown solid.

Example 160: (R)-(3-(4-(5-aminopentyl)-2,5-dicUorobenzyl)thiazolidm-4-yl) (4- cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)methanone. To intermediate 160h (1.7 g, 2.68 mrnol, 1.00 equiv) was added 1 M IIC1 in dioxne (5 mL) and the resulting solution was stirred for 1 h. The resulting mixture was concentrated under vacuum, diluted with 30 niL of ethyl acetate, and washed with 3 x 10 mL. of aqueous sodium carbonate, The organic layer was dried over sodium sulfate and concentrated under vacuum to afford 1.4 g (98%) of example 160 as a yellow solid. LCMS (ES, m/z): 533 [M+l] + . 1H-NMR (300 MHz, CDCi ppm): 7.49 (s, 1H), 7.21 (s, 1H), 4,74 (s, 211), 4.46 -4.40 (m, 1 H), 3.13 (s, 2H), 2.72 -2.0? (m, 2H), 1.87 (s, H i). 1.68 -1.53 (m, 4H), 1.51 (s, 9 1 ,46-1.36 (m, 21-1).

Example 161

H ' (2,5-dielilorQ-4-((4-(4-cvclopropyl-1 ,2,3,4-tetrahydroquinoxaline-l- / l)thiazolidin-3-yl)methyl)ben2yr)tfaio)- 1 -methyl-1 H-imidazole-S-carboxylic acid

Scheme 161 : 1. Methyl 2-mercapto-l-methyl-lH-imidazole-5-carboxylate, DEAD, PPh 3 , toluene; 2. LiOH, THF, ¾0,

Intermediate 161a: (R)-methyl 2-((2 s 5-dichloro-4-((4-(4-cyciopropyl-l, 2,3,4- tetrahydroquinoxaline- 1 -carboTiyI)thiazolidin-3-yI)methyI) be.nzyl)tbio)- 1 -methyl- 1 H- imidazole-5-carboxylate, To a mixture of example 159 (60 mg, 0.125 mmol, 1 equiv), methyl 2-mercapto-l-methyl-lH-imidazole-5-carboxylate (30.2 mg, 0,176 mmol, 1.4 equiv) and PPh 3 (46.2 mg, 0.176 mmol. 1.4 equiv) in toluene (0.35 mL) at 0 °C was added dropwise diethyl azodicarboxylate (40 % wt in toluene, 80 μΕ, 0, 176 mmol. 1.4 equiv). The mixture was stirred at rt for 3 h, concentrated, and then purified by column to give 79 mg (100%) of intermediate 161a as a white solid.

Example 161: (R)-2-((2 ) 5-dichloro-4-((4-(4-cyclopropyl-l > 2 > 3 > 4- tetrahydroquinoxaline- 1 -carbonylithi azolidin-3 -yl)methyl)benzy!)tbio)- 1 -metbyl-l H- imidazole-5-earboxylic acid. To a mixture of intermediate 161a (79 mg, 0,125 mmol, 1 equiv) in THF (0.4 mL) and water (0.2 mL) was added LiOH « H 2 0 (26.2 mg, 0.625 mmol, 5 equiv) and the reaction was stirred overnight. The mixture was concentrated, diluted with ¾0 (0.3 mL), acidified by 1M HQ to pH 3, and then extracted with EtOAc. The organic layer was washed with brine (1 X), dried and concentrated to give 51 mg (66 %) of example 161 as a white solid. LCMS (ES, m/z): 618.10 [M + Hj + .

(R)-(4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yI)(3-(^

hydroxybenzyl)thiazolidin-4-yl)methanone

Scheme 162: 1. 92b. NaBHfOAc , AcOH, DCE Example 162: ( )-(4-cyclopropyl~3,4-dihydroquinoxdin-l(2H)-yl)(3-(2,5-dichl oro-4- hydroxybenzyl)tW.azoUdin-4-yl)methanone. To a solution of 159c (200 mg, 0.691 mmol), 92b (132 mg, 0.691 mmol) and AcOH (40 μΕ, 0.69 mmol) in DCE (3 mL) was added NaBH(OAc) 3 (234 mg, 1.11 mmol) and the resulting mixture stirred for 16 h. The excess NaBH(OAc) 3 was quenched with IM aqueous HQ, and the mixture then extracted with DCM. The organic layer was dried over Na 2 S0 . filtered, and then the solvent removed under reduced pressure. The resulting residue was purified by flash column chromatography, using 0 to 50% EtOAc in hexanes as eluent to give Example 162 as a white powder (140 mg, 44%). MS (ES, m/z): 464.16 [M + H] + . Example 163

(R)-5-(2,5<UeMoro^-((4-(4-ey^

carbon l)thiazolidm-3 -vl methyl phenyl)pentanoic acid

Scheme 163: 1 , SOC! 2 , MeOH: 2. 92c, Pd/PP s^CL, CuL BIPEA, DMF; 3. Rli/C, ¾, EtOAc; 4. NaBHU, MeOH; 5. NBS, PPh 3 , ! ' )(. ' Nl Ί ? !{· ' ; 6. 159c, K 2 C() 3 , DMF; 7. LiOH, THF, H 2 0: intermediate 163a: methyl pent-4-ynoate. To a mixture of pent-4-ynoic acid (5 g, 50.97 mmol, 1 ,00 equiv) in methanol (250 mL) at 0 °C was added thionyl chloride (4.45 mL) dropwise and the resulting solution was stirred overnight. The mixture was diluted with 800 mL of dicliloromethane, washed with 2 x 500 mL of water, dried over anhydrous sodium sulfate and then concentrated to give 5.9 g (crude) of intermediate 163a as light yellow oil

intermediate 163b: methyl 5-(2,5-dichloro-4-formylpheiiyl)pent-4-ynoate. To a mixture of 92c (6.0 g, 18.57 mmol. 1.00 equiv), intermediate 163a (2.50 g, 22,30 mmol, 1 .20 equiv) and DIE A (4.79 g, 37.06 mmol, 2.00 equiv) in DMF (45 mL) was added Pd(PPh 3 ) 2 Cl 2 (1.30 g, 1.85 mmol, 0.10 equiv) and Cui (354 mg, 1.86 mmol, 0.10 equiv) and the resulting solution was stirred overnight. The mixture was diluted with 300 mL of ethyl acetate, washed with 2 x 200 mL of water and 2 x 200 mL of brine, the organic- layer dried over anhydrous sodium sulfate and then concentrated under vacuum. The residue was applied onto a silica gel column with petroleum ether/ethyl acetate (40:1) to afford 3,05 g (58%) of intermediate 163b as a light yellow solid

Intermediate 163c: methyl 5-(2.5-dichloro-4-formylphenyl.)pentanoate: To a mixture of intermediate 163b (3.05 g, 10.70 mmol, 1.00 equiv) in ethyl acetate (100 mL) was added Rh/C (3.23 g) and the suspension was stirred under a H 2 atmosphere overnight. The solids were filtered out and the filtrate concentrated to provide 2.52 g (81%) of intermediate 163c as brown oil. Intermediate 163d: methyl 5-[2,5-dichloro-4-(hydroxymethyl)phenyl]pentanoaie. To a solution of intermediate 163c (2.52 g, 8.72 mmol, 1.00 equiv) in methanol {40 mL) at 0 °C was added NaBH (660 mg, 17.45 mmol, 2.00 equiv) in several batches over 1 h. The reaction was stirred for 1 h at 0-5 °C and then quenched by the addition of 50 ml, of waier/ice. The mixture was concentrated under vacuum to remove the organic solvents and fhenextracted with 3 50 mL of DCM. The organic layers were combined, washed with 1 x 100 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 2.42 g (95%) of intermediate 163d as an off-white solid.

Intermediate 163e: methyl 5-(4-(bromomethyl)-2,5-dichlorophenyl)pentanoate. To a mixture of intermediate 163d (200 mg, 0.686 mmol, 1 equiv) in DCM (1.3 mL) and THF (1 .3 mL) at 0 °C was added NBS (269 mg, 1.51 mmol 2.2 equiv) and P.Ph 3 (234 tag, 0.892 mmol, 1.3 equiv) and the mixture was stirred for 1 h. The reaction was quenched with brine, extracted with EtOAc, the organic layer was dried, concentrated, and purified by column to give 227 mg (93 %) of intermediate 163e as clear oil. Intermediate 163f: (R)-methyl 5-(2,5-dic oro-4-((4-(4-eyclopropy tetrahydroquinoxaime-l-carbonyI)tM^ To a mixture of 159c (20.3 mg. 0.07 inmoi, i equiv) in DMF (0.3 mL) was added intermediate 163e (27.3 mg, 0.077 mmol, 1.1 equiv) and K 2 CG 3 (19.4 mg, 0.1.4 mmol, 2 equiv) and the mixture stirred for 3 h and then, heated to 60 °C and stirred overnight. The mixture was diluted with EtOAc, washed with ¾0 (2 x) and brine (1 x), the organic layer was dried, concentrated, and purified by column to give 12.6 mg (32 %) of intermediate 1 3f as clear syrup.

Example 163: ( 5-(2,5-dichloro-4-((4-(4-cyclopropyl- 1 ,2,3,4- tetrahydroqumoxaline-l-carbony acid. To a mixture of intermediate 163f (10.8 mg, 0.0193 mmol, 1 equiv) in THF (0.12 ml.) and water (0.06 mL) was added LiOH » H20 (1.6 mg, 0.0385 mmol, 2 equiv) and the reaction was stirred, for 6 h. The mixture was acidified with 1 M HC1 (42 μΐ,), concentratedand then lyophilized to give 1 1.4 mg of Example 163 as a white solid. LCMS (ES, m/z): 548.09 [M + Hf Exam le 164

(4-cyclopropyl-3,4-dihydroquinoxa1m^

(trifluoromethyl)berj2^damino cvclopropyl)methanone .

Scheme 164; 1. H 2 80 4 , NaN0 2 , I; 2. F8C) 2 CF 2 COOMe, CuBr, NMP; 3, Fe, NH 4 C-L MeOH; 4. ¾S0 4 , Na 0 2 , KI; 5, NBS, benzoyl peroxide, CC1 4 ; 6. FT: 7. 164e, K 2 C0 3 , DMF, KL

Intermediate 164a: 1 -iodo-2-methyl-4-nitrobenzene. To 2-methyi-4-nitroaniline (20.0 g, 131 .45 mmol, 1.00 equiv) in H 2 0/acetone (80/50 mL) al 0-5 °C was added cone. H.2SO4 (27.1 g, 276,53 mmol, 2.10 equiv) followed by the drop-wise addition of a solution of NaN(>> (10.0 g, 144.93 mmol, 1 .10 equiv) in water (20 ml.) and the resulting solution was stirred for 1 h. To this was added drop- wise a solution of KI (30.6 g, 184.34 mmol, 1.40 equiv) in water (20 mL) and the reaction allowed to warm to RT and then stirred for an additional 2 h. The mixture was diluted with 500 mL of ethyl acetate, washed with 2 x 200 mL of water, 3 x 200 mL of aqueous Na 2 S () 3, dried over anhydrous sodium sulfate and concentrated. The residue was purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 :1000) to afford 21.7 g (63%) of intermediate 164a as a white solid.

Intermediate 164b: 2-memyl-4-nitTo-l -(trifluoromethyl)benzene. To intermediate 164a (21.9 g, 83.26 mmol, 1.00 equiv) in NMP (150 mL) was added methyl 2,2-difluoro-2-(fluorosulfonyl)aeetate (23,73 g, 123.52 mmol, 1.50 equiv) and CuBr (1.45 g, 10.11 mmol, 0.12 equiv) and the mixture was stirred ai 120 °C overnight. The mixture was diluted with 500 mL of ethyl acetate, washed with 3 x 200 mL of brine, dried over anhydrous sodium sulfate and concentrated. The residue was purified via silica gel chromatography (ethyl acetate/petroleum ether, 1:100) to afford 15.0 g (88%) of intermediate 164b as a yellow oil. Intermediate 164c: 3-methyl-4-(triiluoromethyl)aniline. To intermediate 164b (15.0 g, 73.12 mmol, 1.00 equiv) in methanol/HiQ (100/25 mL) was added elemental Fe (15.0 g, 267.86 mmol, 3.66 equiv) and NH 4 C1 (1.5.0 g, 280.43 rnmol, 3.83 equiv) and the reaction was stirred at 60 °C for 3 h. The mixture was filtered and the filtrate concentrated, diluted with 200 mL of ethyl acetate, washed with 2 x 100 mL of brine, dried over sodium sulfate and then concentrated to afford 8.0 g (62%) of intermediate 164c as a yellow oil.

Intermediate 164d: 4-iodo-2-methyl- 1 -(trifluoromethyl)benzene. To intennediate 164c (6.0 g, 34,26 mmol, 1 .00 equiv) in water (50 mL) at 0 °C was added sulfuric acid (7.06 g, 71.98 mmol, 2.10 equiv) followed by the drop-wise addition of a solution of NaN0 2 (2.60 g, 37.68 mmol, 1.10 equiv) in water (40 mL) and the mixture was stirred for 1 h. To this was added drop-wise a solution of KI (7.97 g, 48.01 mmol, 1.40 equiv) in water (40 mL) and the reaction was allowed to warm to RT and stirred for 1 h. The mixture was diluted with 200 ml, of ethyl acetate, washed with 2 x 200 ml. of Brine, 1 x 200 mL of aqueous Na^SOa , dried over anhydrous sodium sulfate and concentrated. The residue was purified via silica, gel chromatography (petroleum ether/ethyl acetate, 100:1) to afford 8.2 g (85%) of intermediate 164d as a yellow oil.

Intermediate 164e: 2-(bromoniethyl)~4-iodo-l -(trifluoromethyl)benzene. To intermediate 164d (3.5 g, 12.24 mmol, 1.00 equiv) in CC1 4 (40 mL) at 60 °C was added benzoyl peroxide (1.7 g, 7.02 mmol, 0.57 equiv) followed by the batch-wise addition of NBS (2.37 g, 13.32 mmol, 1.09 equiv) and the reaction stirred at reflux overnight. The solids were filtered out and the filtrate concentrated to afford 1.6 g (36%) of intermediate 164e as a red oil.

Intermediate 164f: (l-aminocyclopropyl)(4-cyclopropyl-3,4-dihydroquinoxaiin- l(2H)-yl)methanone. Intermediate 164f was prepared from intermediate 26a using the procedures described in Example 59.

Example 164: (4rcyclopropyl-3,4-dihydroquinoxaim-l(2H)-yi)(l~(5-iodo-2- (triilu.oromethyl)benz}damino)cyclopropyl)metha.none. To intermediate 164e (364 mg, 1.00 mmol, 1.00 equiv) in DMF (5 mL) was added intermediate 164f (257 mg, 1.00 mmol, 1.00 equiv), potassium carbonate (208 mg, 1 .50 mmol. 1.50 equiv) and KJ. (166 mg. 1.00 mmol, 1.00 equiv) and the reaction was stirred overnight. The mixture was diluted with 20 mL of ethyl acetate, washed with 2 x 20 mL of brine, dried over anhydrous sodium, sulfate, concentrated and then purified via silica gel chromatography (petroleum ether/ethyl acetate, 5:1) to afford Example 164 of a purity suitable for use in the next step, A 300 mg aliquot was further purified via reverse phase (CI 8) Prep- HPLC to afford 150 mg (28%) of the title compound as an off-white solid. LCMS (ES, m/z): 542 [M+lf. l H NMR (400 MHz, CD 3 OD, ppm): 8.33-7.78 (m, 1H), 7.70-7.22 (m, 5H), 7.07-6.78 (m, IH), 4.11-3.82 (m, 4H), 3.45-3.09 (m, 2H), 2.44 (s, 1 H),L41 (s, 2H), 1.04 (s, 2H), 0.83 (d, J = ό.9Πζ, 2H), 048 (s, 2H).

Example 65

(4-Cyclopropyl-3 ,4-dihydroquinoxalia- 1 (2H)-yl)(l -((2,5-dichloro-4- hydroxybenzyl amino cyclopropyl me1iianorte

Scheme 165: 1. 92b, NaBH 4 , MeOH.

Example 165 : (4-cyciopropyI-3 ,4-dihydroquinoxalin- 1 (2H)~yl)( 1 -((2 ,5 -d ί eh ] oro- 4-hydroxyberrzyl)amino)cyclopropyl)methanone bis-TFA salt. To a mixture of intermediate 164f (50.6 mg, 0.197 mmol, 1.0 equiv) in methanol (0.8 mL) was added 92b (37.6 mg, 0.197 mmol, 1.0 equiv). The mixture was stirred at room temperature for 1.5 h and cooled to 0 °C. To the mixture was added acetic acid (1 1.3 pJL, 0.197 mmol, 1.0 equiv), followed by addition of sodium borohydride (11.9 mg, 0.315 mmol. 1.6 equiv). The mixture was stirred at 0 °C for 20 minutes and purified by preparative. IIPLC to give the title compound. (74.7 mg, 57%) bis-TFA salt as a pale yellow solid. MS (ES, m/z): 432.03 [M + H] + , l H NMR. (400 M! f . CD3OD) δ 7.27 (td, J = 8.4, 1 .4 Hz, 2H), 7.20 (s, IH), 7.19 - 7.13 (m, I H), 6.96 (s, 1H), 6.75 (td, J - 7.6, 1.3 Hz, IH), 4.04 (s, 2H), 3.89 (t, J = 5.8 Hz, 2H), 3.44 (t, J= 5.8 Hz, 2H), 2.50 - 2.36 (m, IH), 1 .35 (dd, J = 7.8, 5.6 Hz, 2H), 1.24 (dd, j = 7.6, 5.7 Hz, 2H), 0.91 - 0.79 (m, 2K), 0.61 - 0.53 Urn 2H).

Example 166 l~(4-cycl 0 propY 2 , 5- dichlorobenzoate

Scheme 166: 1. 2,5-cUch!orobenzoic acid, EDOHC1, DMAP, DCM.

Example 16 : 1 -(4-cyclopropyl- 1 ,2,3 ,4-tetrahydroquinoxaline- 1 - carbanyl)cyclopropyi 2,5-dichlorobenzoate TFA salt. To a mixture of intermediate 9a (28.3 mg, 0.1 1 mmol, 1.0 equiv) in DCM (0.4 m.L) were added 2,5-dichlorobenzoic acid (42 mg, 0.22 mmol, 2.0 equiv) and DMAP (27 nig, 0.22 mmol, 2.0 equiv). The mixture was cooled to 0 °C and then EDC.HCl (42 mg, 0.22 mmol, 2.0 equiv)was added. The mixture was stirred at room temperature over weekend, concentrated, and purified by preparative. HPLC to give the title compound (36.4 mg, 61%) TFA salt as a yellow solid. MS (ES, m/z): 431.05 [M + H] + , ! H NMR (400 MHz, CD 3 OD) δ 7.44 (dd, J = 8.6, 2.5 Hz, 1H), 7.39 (d, J = 8.6 Hz, 1 H), 7.27 (dd, J = 7.9, 1.4 Hz, 1H), 7.13 - 7.07 (m, 1H), 6.90 (dd, J - 8.3, 1.2 Hz, 3H), 6.77 (td, J= 7.6, 1.3 Hz, 1H), 6.54 (s, 1H), 3.77 (s, 2H), 3.34 (t, J - 5.7 Hz, 2H), 2.31 - 2.20 (m, 1H), 1.82 - 1.73 (m, 2H), 1.31 - 1.23 (m, 2H), 0.61 (dd, ./ ' 6.5, 1.8 Hz, 2H), 0.05 (s, 2H).

Example 167

hydroxybenzyl)ammo)cyclopropyl)metfaanorie

Scheme 167: 1. BH 3 THF, THF; 2. pyridinium cMorochromate, DCM; 3. NaBH 4 , MeOH.

Intermediate 167a: 2,5-dichloro-3-(hydroxyraet yl)phenol. To a mixture of 2,5- dichloro-3-hydroxybenzoic acid (1.02 g, 4.93 mmol, 1.00 equiv) in THF (6.7 nxL) at 0 °C was added borane-tetrahydrofuran complex solution (1 M, 14.8 mL, 14.8 mmol, 3.00 equiv) dropwise. The mixture was stirred at 80 °C overnight. The mixture was cooled to room temperature, quenched with 2N HQ, and extracted with ethyl acetate. The organic layer was washed with 2N HQ (Ix), ¾0 (Ix), and brine (Ix), dried, concentrated, and purified by column to give 0.435 g (46%) of 2,5-dichloro-3- (hydroxymethyl)phenol as a yellow solid.

Intermediate 167b: 2,5-dicbloro-3-hydroxybenzaldehyde. To a mixture of intermediate 2a (350 mg, 1.81 mmol, 1.00 equiv) in DCM (4 mL) at room temperature was added pyridinium chlorochromate (437 mg, 2.03 mmol, 1.12 equiv). The mixture was stirred at room temperature for 5h, concentrated, and purified by column to give 179 mg (52%) of 2,5 -dichloro-3 -hydrox benzaldehyde as a white solid. Ή NMR. (400 MHz, COCK) δ 10.34 (s, 1H), 7.49 (s, 1 H), 7.28 (s, 1H), 6.07 (s, IH).

Example 167 : (4-cyclopropyl-3 ,4-dihydroquinoxalin- 1 (2H)-yl)(l -((2,5-dichloro- 3-hydroxyber.zyl)amino)cyc3opropyl)metb.anone bis TFA salt. Example 167 was prepared using the procedures described in Example 165. MS (ES, m/z): 432.1 1 [M+H] + . S M NMR (400 MHz, CD : ( .)] )) δ 7.30 (dd, J = 7.9, 1.3 Hz, I H), 7.24 (dd, J == 8.3, 1.2 Hz, IH), 7.19-7.13 (m, IH), 6.89 (d, J - 2.4 Hz, IH), 6.79-6.73 (m, I H), 6.69 (d, J= 2.3 Hz, IH), 4.04 (s, 2H) 5 3.89 (t, J = 5.7 Hz, 2H), 3.44 (t, J - 5.8 Hz, 2H), 2.46- 2.39 (m, III), 1.37 (dd, J = 7.8, 5.4 Hz, 2H), 1.21 (dd, J= 7.8, 5.4 Hz, 2H), 0.86-0.79 (m, 2H), 0.58-0.49 (m, 2H).

Example 168 (4-c vclo ropvi-3 ,4-dihydroquinoxaiin- 1 (21 0-νΤ)Π -(( 3 ,6-diciiloro-2- hyciroxybenzyl)ainino)cyclopropyi)methanone

i68a 168b 1S8c

Scheme 168: 1 , BH 3 THF, THF; 2. pyridinium clilorochromate. DCM, 3, BB.r 3j DCM; 4. NaBH 4 , MeOH,

Intermediate 168a: (3,6-dichloro-2-met oxyphenyl)meth.anol, To a mixture of 3,6-dicliloro-2-raethoxybenzoic acid (1.0 g, 4.52 mmol. 1.00 equiv) in THF (6 mL) at 0 °C was added bora e-ietraliydrofuraii complex solution (1M, 9mL, 9.0 mrnol, 2.00 equiv) dropwise. The mixture was stirred at 80 °C overnight. The mixture was cooled to room temperature, quenched with 2N HCI, and extracted with ethyl acetate. The organic layer was washed with 2N HCI (Ix). ¾0 (Ix), and brine (ix), dried, and concentrated to give 0.866 g (92%) of (3,6-dk oro-2-methoxyphenyl)methanol as a white solid.

Intermediate 168b: 3,6-dicMoro-2-methoxybenzaidehyde. To a mixture of intermediate 168a (94.3 mg, 0.456 mmol, 1.0 equiv) in DCM (1 mL) at room temperature was added pyridinium chlorochromate (118 mg, 0.547 mmol. 1.2 equiv). The mixture was stirred at room temperature overnight, concentrated, and purified by column to give 85 mg (91%) of 3.6~dich1oro-2-methoxybenzaldehyde as a white solid. 1H NMR (400 MHz, CDC1 3 ) δ 10.42 (s, 1H), 7.51 (dd, J = 8.7, 0.5 Hz, 1H), 7.19 (d, J - 8.7 Hz, lH), 3.96 (s, 3H).

Intermediate 168c: 3,6-dichloro-2-hydroxybeazaldehyde. To a mixture of intermediate lb (52.6 mg, 0.255 mrnol, 1.0 equiv) in DCM (3 mL) at 0 °C was added boron tribromide solution (1M, 0.77mL, 0.77mmoi, 3 equiv). The mixture was stirred at room temperature overnight and at 45 °C for 3 h. The resulting mixture was cooled to room temperature, quenched with sat. aqu. NaHC0 3 , and extracted with ethyl acetate. The organic layer was dried and concentrated to give 47 mg (96%) of 3,6-dichloro-2- ydroxybenzaldehyde as a yellow solid. ¾ NMR (400 MHz, CDC1 3 ) δ 12.44 (s, 1H), 10.40 (s, 1 H), 7.61 - 7.43 (m, Ιίϊ), 7.01 - 6.85 (m, lH).

Example 168: (4-cyclopropy{-3,4-dih.ydroquinoxaiin- 1 (2H)-yl)(l -((3,6-dichloro- 2-hydroxybenzyl)amino)cycIopropyl)methanone bis TFA salt Example 168 was prepared using the procedures described in Example 165 substituting 167b in place of 92b. MS (ES, m/z): 432.15 [M+H] + . l H NMR (400 MHz, CD 3 OD) δ 7.36 (d, J = 8.7 Hz, 1H), 7.30 - 7.24 (m, 2H), 7.19 - 7.12 (m, 1H), 6.98 (d, J = 8.7 Hz, IF), 6.77 - 6.70 (m, 1H), 4.46 (s, 2Ii), 3.94 (t, J= 5.7 Hz, 2H), 3.47 (t, J ------ 5.7 Hz, 2H), 2.51 - 2.40 (m, 1H), 1.45 - 1.26 (m, 4H), 0.91 ■■■■ 0.83 (m, 2H), 0.67 - 0.58 (ra, 2H).

hydroxybenzyl)thiazolidm-4-yl)methanone

Example . 169: (R)-(4-cyclopropyl-3,4-dihydroquiaoxdin-l(2H)-yl)(3-(2 5 5- dicMoro-3-hydroxyber^l)lhiazolidhi-4-yl)meihaiione bis TFA salt. Example 169 was prepared using the procedures described in Example 162 substituting 167b in place of 92b., MS (ES, m/z): 464.10 [M+H] + . ¾ NMR (400 MHz, CD 3 OD) δ 7.20 (dd, J= 8.3, 1.3 Hz, 1H), 7.18 - 6.98 (m, 2H) S 6.87 (s, 1 11 ). 6.76 (s, 1H), 6.67 (t J = 7.1 Hz, 1H), 4.77 (s, 1H), 4.27 (d, J- 9.6 Hz, 1 H), 4.15 - 4,03 (m, 1 H), 4.02 - 3.87 (m, 2H), 3.87 - 3.73 (m, 1H), 3.73 - 3.57 (m, 1H), 3,47 - 3.33 (m, 2H), 3.14 (s, 2H), 2.44 (s, 1H), 0.90 - 0.73 (m, 2H), 0.67 - 0.46 (m, 2H).

Example 170 (R " )-(4-cyclopropyl-3 ,4-dih ydroquinoxalin- 1 (2H)-yl)(3-(3 ,6-dichloro-2- hydroxvbeii2A ¾hiazolidm-4-yl)methanone

Example 170: (R)-(4-cyclopropyl-3,4-dihy<iroqumoxalin-l(2H)-yl)(3-(3,6 - dicMoro-2-hydroxybenzyl)thiazoUdin-4-yl)me1hanone bis TFA salt. Example 170 was prepared using the procedures described in Example 162 substituting 168c in place of 92b.. MS (ES, m/z): 464.06 [M+Hf. ! H NMR (400 MHz, CD 3 OD) δ 7.34 (d, J ------ 8.2

Hz, 1 H), 7.25 - 7.12 (m, 2H), 7.08 - 6.97 (ra, .1 H), 6.96 - 6.86 (m, 1H), 6.77 - 6.62 (m, I I I ). 4.84 - 4.75 (m, I ! I). 4.49 (d, J = 10.0 Hz, 1H), 4.42 - 4.27 (m, 2H), 4.26 - 4.15 (m, 1H), 3.94 - 3.80 (m, HI), 3.77 - 3.68 (m, 1H), 3.48 - 3.39 (m, 1 1 1). 3.38 - 3.33 (m, 1H), 3.23 - 3.09 (m, H I). 3.03 - 2.89 (m, 1H), 2.46 (s, 1H), 0.92 - 0.78 (m, 2H), 0.72 - 0.47 (m, 2IT).

Example 171

f(4-eyclopropyl-3 ,4-dihydf oquinoxalin- 1 (21 Γ)-νί)( 1 -((2,5 -di chloro-4 -(4-

(metfayl((2S,3 .4R,5R)-2.3.4.5,6- per ! tahydroxy1iexyI)aj¾uno)butvl)phenoxy)methyl)

Scheme 171 : I . KOH, MeOH; 2. Oxalyl chloride, DCM, DMF (cat.); 3. Intermediate le, TEA, DCM; 4, NaOH, THF, H 2 0; 5. isobutylchloroformate, TEA, DCM; 6. NaBH 4? DEM/H2O; 7. MsCl, TEA, DCM; 8. 2,5-dichloro-4-iodophenol, K 2 C(X acetone; 9. but- 3-yn-l-oL Pd(PPh 3 ) 2 Cl 2 , Cul, DIEA, DMF; 10, Rh C, H 2 , EtOAc; 11. MsCl, TEA, DCM; 12. (IR.SR^SS -Cmethyiajminoiliexane-l^^^^- entaol, TEA, KI, DMF

Intermediate 171a: l-(methoxycarbonyl)cyclopropanecarboxylic acid. To 1,1- diethyl cyclopropane- 1,1 -dicarboxylate (15.0 g, 80,56 mmol, 1.00 equiv) in methanol (90 mL) at 0 °C was added batch- wise potassium hydroxide (6.3 g, 112.28 mmol, 1.40 equiv) and the resulting solution allowed to warm to RT and stirred for 2 b. The mixture was concentrated under vacuum., diluted with 100 mL of water and then washed with 1 x 50 mL of ethyl acetate. The pH value of the aqueous solution was adjusted to 3-4 with cone. HCl, extracted with 3 x 50 mL of ethyl acetate, the organic layers combined and then washed with brine. The organic layer was dried over anhydrous, sodium sulfate and concentrated to afford 9.2 g (79%) of intermediate 171a as a colorless liquid.

Intermediate 171b: methyl l-(chlorocarbonyl)cyclopropanecarboxylate. To intermediate 171a (3.6 g, 24.98 mmol, 1.00 equiv) in DCM (15.0 mL) at 0 °C was added DMF (150 nig, 2.05 mmol, 0, 10 equiv) followed by the drop-wise addition of oxaiyl dichloride (4.3 g. 33.88 mmol, 1.50 equiv) and the resulting solution was allowed to warm to RT and then stirred for 1.5 h. The mixture was concentrated, under vacuum to afford 3.7 g (91%) of intermediate 171b as a yellow oil, which was used without further purification.

intermediate 171c: methyl 1 -(4-cyclopropyl-l,2.3 5 4-tetrahydroquinoxaHne-l- carbony])cyclopropanecarboxylate.. To intermediate le (4.0 g, 22.96 mmol, 1.00 equiv) in dichloromethane (40.0 rnL) at 0 °C was added TEA (3.5 g, 34.59 mmol, 1.50 equiv) followed by the drop-wise addition of a solution of intermediate 171b (3.7 g, 22.76 mmol, 1.00 equiv) in DCM (5.0 mL), and the resulting solution was allowed to warm, to RT and. then stirred for 0,5 h. The mixture was diluted with 45 mL of DCM, washed with 1 x 50 mL of brine, dried over anhydrous sodium sulfate and. then concentrated under vacuum. The residue was purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 :20-1 :5) to afford 6.5 g (94%) of intermediate 171c as a yellow oil.

Intermediate 171d: 1 -(4-cyclopropyl- 1 ,2,3 ,4-tetrahydroquinoxaline- 1 - carbonyl)cycJopropaiiecarboxylic acid. To interniediaie 171c (6.5 g, 21.64 inmol, 1.00 equiv) in 1 : 1 THF/H 2 0 (50 mL) at 0 °C was added sodium hydroxide (1.7 g, 42.50 mmol, 2.00 equiv) and the resulting solution was allowed to warm to RT and then stirred overnight. The solution was adjusted to pH 2-3 with aqueous 1 M HC1 and then extracted with 3 x 25 ml, of ethyl acetate. The organic layers were combined, washed with 1 x 30 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 6.0 g (97%) of intermediate 171d as a light yellow solid.

intermediate 171 e: 1 -(4-cyclopropyl- 1 ,2,3 ,4-teirahydroquinoxaline- 1 - carbonyl)cyciopropanecarboxylic (isobuty! carbonic) anhydride. To intermediate 17 (7.0 g, 24.45 mmol, 1.00 equiv) in DCM (80.0 mL) at 0 °C was added TEA (3.6 g, 35.58 mmol, 1.50 equiv) followed b the drop-wise addition of 2-meth.ylpropyl chloroformate (3.9 g, 28.56 mmol, 1.05 equiv) and the resulting solution was allowed to warm to RT and then stirred for 0.5 h. The mixture was diluted with 80 mL of DCM, washed with 1 x 50 mL of brine, dried over anhydrous sodium sulfate and then concentrated under vacuum. The residue was purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 :10-1 :5) to afford 10.0 g (crude) of intermediate 171e as a light yellow oil.

Intermediate 171f: (4-cyc!opropyl-3 > 4-dihydroquinoxalin-l (2H)-yl)(l - (hydroxymethyl)cyclopropyl)methanone. To intermediate 171e (10 g, 25.88 mmol, 1.00 equiv) in ethylene glycol dimetliyl ether (250 mL) at -30 °C was added drop-wise a solution of NaBH 4 (2.0 g, 52.87 mmol, 2,00 equiv) in water (12.0 mL) and the resulting solution was allowed to warm to 0 °C and then stirred for 2 h. The solution was adjusted, to pH 2-4 with aqueous 1 M HCL diluted with 100 mL of water and then extracted with 3 x 200 mL of ethyl acetate. The organic layers were combined, dried (anhydrous sodium, sulfate), concentrated and then purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 : 10-1 : 1) to afford 2.5 g (35%) of intermediate I71f as a purple solid.

Intermediate 171g: (l-(4-eyclopropyl-l ,2,3,4-teirah.ydroqumoxaline~l- carbonyl)cyclopropyl)methyl metlianesulfonate. To intermediate 171f (2.3 g, 8.45 mmoL 1.00 equiv) in DCM (45.0 mL) at 0 °C was added TEA (1.3 g, 12.85 mmol, 1.50 equiv) followed by the drop-wise addition of MsCl (1.1 g, 9.60 mmol, 1.10 equiv) and the resulting solution, was allowed to warm to RT and stinred for 0.5 h. The mixture was washed with 1 x 30 mL of brine, dried over anhydrous sodium sulfate, concentrated and then purified vi silica gel chromatography (ethyl acetate/petroleum ether, 1 :20-1 :1) to afford 2.7 g (91%) of intermediate 171g as a light yellow solid.

Intermediate 171 h. : (4-ey clo propyl-3 ,4-dihydroquinoxalin- 1 (2H)-yl)(l-((2,5- dichk ro-4-ioclophenoxy)methyl)eyclopropy])methanoHe. To intermediate 171g (1.5 g, 4.28 mmol, LOO equiv) in acetone (70.0 mL) was added 2,5-dic oro-4-iodophenol (1.86 g, 6.44 mmol, 1.50 equiv) and potassium carbonate (1.18 g, 8.54 mnioi, 2.00 equiv) and the reaction was stirred at 55 °C overnight. The mixture was filtered, the filtrate concentrated and the residue purified via silica gel chromatography (ethyl acetate/petroleum ether, 1:10-1:2) to afford 1.7 g (73%) of intermediate 171h as a white solid.

Intermediate 1711: (4-cyclopropyl-3,4-dihydioquinoxalin-l(2H)-yl)(l -((2,5- dicMoro-4~(4~hydroxybut^ To intermediate 171h (160 mg, 0.29 mmol, 1.00 equiv) in DMF (5.0 mL) was added but-3- yn-l-ol (20.1 mg, 0.29 mmol, 1.00 equiv), DIEA (77.5 mg, 0.60 mmol, 2.00 equiv), Pd(PPli 3 )7.Ci 2 (21.1 mg, 0.03 mmol, 0.10 equiv) and Cul (5.7 mg, 0.03 mmol, 0.10 equiv) and the mixture was stirred for 2 h at RT. The reaction was diluted with 20 mL of water, extracted with 3 x 30 mL of ethyl acetate and the organic layers combined, washed with 1 x 50 ml. of brine and then dried over anhydrous sodium sulfate. The solution was concentrated and the residue purified via silica gel chromatography (ethyl acetate/petroleum ether, 1 : 10-1 :2) to afford 0.18 g of intermediate 1711 as a red oil. intermediate 171j : (4-cyclopropyl-3,4-dihydroquinoxalin-l(2H)-yl)(l-((2,5- dicWora-4-(4-hydroxybu1yl)phenoxy)methyi)cyclopropyl)metbano ne. To intermediate 1711 (300 mg, 0.62 mmol) in ethyl acetate (15.0 mL) was added h/ ' C (350 mg) and the resulting suspension was stirred under a hydrogen atmosphere overnight. The mixture was diluted with 20.0 ml, of methanol, filtered and the filtrate concentrated to afford 0.30 g (99%) of intemiediate 171 j as a red oil.

Intermediate 171k: 4-(2,5-dichloro-4-((l -(4-cyclopropyl- 1,2,3,4- tetrahydroquinoxaline- 1 -carbonyl)cyclopropyl)methoxy)phenyl)butyl

raeihanesuifonate. To intemiediate 171] (250 mg, 0.51 mmol, 1.00 equiv) in DCM (10.0 mL) at 0 °C was added TEA (77.4 mg, 0.76 mmol, 1.50 equiv) followed by the drop-wsie addition of MsCl (70.2 mg, 0.61 mmol, 1.20 equiv) and the resulting solution allowed to warm to RT and then stirred for 0.5 . The mixture was diluted with. 20.0 mL of DCM, washed with 1 x 20 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 260 mg (90%) of intermediate 171k as a. purple oil.

Example 171: (4-cyclopropyl-3,4-dih.ydroquinoxalin- 1 (2H)-yl)(l -((2,5-dichloro- 4-(4-(memyl((2S,3R,4R,5R)-2,3,4,5,6- peiitahyclroxyhexyl)am.mo)butyl)phen^^ To intermediate 171k (320 mg, 0.56 mmol, LOO equiv) in DMF (7.0 mL) was added (2R3R,4R,5S)-6-(methylamino)hexane-l,2.3,4,5"pentaol (160 mg, 0.82 mmol, 1.50 equiv), TEA (85.0 mg, 0.84 mmol, 1.50 equiv) and KI (93.6 mg, 0.56 mmol, 1.00 equiv) and the reaction was stirred overnight at 75 °C. The mixture was then filtered, concentrated and the residue purified by reverse-phase (CI S) prep-HPLC to afford 24.6 mg (7%) of Example 171 trifluoro acetic acid salt as an off-white solid. LCMS (ES, m/z): 666 [M + H] + . Ή NMR (300 MHz, CO 3 OlX ppm): 7.41-7.43 (d, J-7.5 Hz, 1H), 7.35 (s, 1 H), 7.10-7.12 (ni, 2Ή), 6,54-6.72 (m, 2H), 4.10-4.18 (m, 1H), 3.88-3.92 (m, 2H), 3.81-3.83 (m, 2H), 3.66-3.79 (m, 511), 3.41.-3.433 (m, 2H), 3.26-3.29 (m, 4H), 2.92-2.94 (m, 2H), 2.72-2.77 (m, 2H), 2.28 (m, Hi), . 1.68-1.70 (m, 4H), 1.35-1.38 (m, 2H), 0.96-1.00 (m, 2H), 0.67-0.69 (m, 2H), 0.19-0.21. (m, 2H).

Example 172

5-(3-((l "(4-cyclopropyl- 1 ,2,3 ,4-tetrahydroq urn oxaline- 1

((2S,3R,4R R)-23,4,5.6-pental Ax xYhex>4)pentanamide

P T/US2012/071251

Intermediate 172a: methyl 5-(3-((l -(4-cyciopropyl- 1 ,2,3,4- tetoahy(koquinoxaline-l-carbonyl)cyclopropylamino)methyl)-4- (trifluoromethyl)phejtxyi)pent-4-ynoate. To Example 164 (500 mg, 0.92 mmol, 1.00 equiv) in DMF (6 mL) was added methyl pent-4-ynoate (155 nig, 1.38 mmol, 1.50 equiv), Pd(Pl¾ )2 Cl 2 (324 mg, 0.46 mmol, 0.50 equiv), Cul (175 mg, 0.92 mmol, 0.99 equiv) and triemylamme (186 mg, 1.84 mmol, 1.99 equiv) and the reaction was stirred for 2 h. The mixture was diluted with 50 mL of ethyl acetate, washed with 2 x 50 mL of brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified via silica gel cinematography (ethyl acetate/petroleum ether, 1 :5) to afford 489 rng (100%) of intermediate 172a as a yellow oil.

intermediate 172b: methyl 5 3-((l -(4-cyclopropyl- 1.2,3,4- ietrahydroquittoxaline- 1 -carbonyl)eyclopropylamiiio)methyl)-4-

(trifliiOromethyi)phenyI)pentaiioate. To intermediate 172a (489 mg, 0.93 mmol, 1.00 equiv) in ethyl acetate (40 mL) was added Rli'C (734 mg) and the suspension was stirred under a hydrogen atmosphere at 30 °C for 2 days. The solids were filtered out and the filtrate concentrated to afford 1 1 mg of intermediate 172b as a yellow oil.

Intermediate 172c: 5-(3-((l -(4-cyclopropyl- 1 ,2,3,4-tctrahydroquinoxaIine- 1 - cmbonyl)cyclopropylan¾no)me acid. To intermediate 172b (511 mg, 0.96 mmol, 1.00 equiv) in THF/H 2 0 (20/10 mL) was added LiOH*H20 (406 mg, 9.68 mmol, 10.02 equiv) and the reaction was stirred for 3 h. The solution was adjusted to pH 3-4 with aqueous 6 M HCl and then extracted with 2 x 50 nil, of ethyl acetate. The organic layers were combined, washed with 2 x 50 mL of brine, dried over anhydrous sodium sulfate and then concentrated to afford 420 ng (84%) of intermediate 172eas a brown oil.

Example 172 : 5-(3 -((1 -(4-cyclopropyl- 1 ,2,3 ,4-letrahydroqmnoxalirie-l - carbonyl)cy opropylarnmo)m^

((2S,3R,4R,5R)-2,3,4,5,6-pentahydroxyhexyl)pentananiide. To intermediate 172c (220 mg, 0.43 mmol, 1.00 equiv) in DMF (6 mL) was added (2R,3R,4R,5S)-6- (methylamino)hexane-l,2,3,4,5~pentol (84 mg, 0.43 mmol, 1.00 equiv), HATU (195 mg, 0.51 mmol, 1.20 equiv) and DIEA (66 mg, 0.51 mol, 1.20 equiv) and the reaction stirred for 1 h. The mixture was diluted with 50 mL of ethyl acetate, washed with 2 x 50 mL of brine, dried over anhydrous sodium sulfate, concentrated and then purified by reverse-phase (CI 8) Prep-HPLC to afford 67.2 mg (23%) of Example 172 as a green solid. LCMS (ES, /z): 693 [M + H] + . ! H NMR (300 MHz. CD 3 OD, ppm): 7.58 (d, J=8.1Hz, 1H), 7.34-7.21 (m, 4H), 7.96 (d, = 6.3Hz, 1 H), 6.80 (d, J-7.2, 1H), 4.03-3.92 (trt, 3H), 3.90 (d, J=5.7Hz, 2H), 3.77-3.65 (m, 73Ε-Γ), 3.43 (d, J ------ 6.0Hz, 3H), 3.18-3.15

(m, 21 1). 3.01-2.98 (m, 2H), 2.67- 2.56 (m, 3H), 2.46-2.42 (m, 2H), 1.65 (s, 4H). 1.43- 1.39 (m, 2H), 1.18-1.14 (m, 2H), 0.82-0.80 (m, 2H), 0.46 (s, 2H).

Example 173

2-({4-[bis(2-hydroxyemyl)am

1 ,2,3 ,4-tetrahydroqtrinoxalin- 1 -yf jcarbonyll -1 ,3 -thiazolidin-3 - yl metiwUphenyl)pent l]a.mino}-l , 3, 5-triazin-2-yl)(2 -hvdroxyeth.yl)amino)ethan-l-ol

Scheme 173:

Example 173: 2-({4-[bis(2-hydroxye l)mmno]-6-{f5-(2,5-clichloro-4- 4-[(4-eyclopropyl-l,2,3,4-tetrahydroquin^

y1]mel l}phenyl)pentyl]amino}-^

To 2,4,6-trichloro-l ,3,5-triazine (17 mg, 0.094 mmol) in THF (0.5 mL) at 0 °C was added a 0 °C solution of 160 (50 mg, 0.094 mmol) in THF (0.5 mL) followed by DIEA (48 μΐ, 0.28 mmol) and the solution stirred for 30 min at 0 °C. The mixture was allowed to warm to room temperature and then stirred and additional 30 min. The solvent was then removed and the resulting residue dissolved in DMF (1 mL), then DIEA (48 ul, 0.28 mmol) and diethanolamine (39 mg. 0.28 mmol ) were added, and the resulting mixture was stirred at 60 °C for 6 h. The mixture was then diluted with ¾0 5 acidified with TFA, and then purified by preparative H LC with a CI 8 silica gei stationary phase using a gradient of H 2 0 0,05% TFA : CH 3 CN 0.05% TFA (70 : 30 to 5 : 95) and detection by UV at 254 nm to give the title compound (23 mg, 21%) tri-TFA salt. MS (ES, m/z): 818.26 [M + H] + ; 1H NMR (400 MHz, CD 3 OD) 8 7,30 - 6.98 (m, 5H), 6.64 (t, J = 7.6 Hz, 1H), 4.76 - 4.62 (m, 1H), 4.10 (d, J = 9.3 Hz, 2H), 3.98 - 3.61 (m, 12H), 3.60 - 3.33 (m, 6H), 3.27 - 3.05 (m, 8H), 2.72 (t J = 7,5 Hz, 2H), 2.46 - 2.31 (m, I H), 1.72 - 1,59 (m, 4H), 1 .49 - 1.39 (m, 2H), 0.81 (d, J = 6.5 Hz, 2H), 0.61 - 0.41 (m, 2H).

2-(3-(2,5-dicMoro- -(((R)-4-(4-cyclopropyl- 1 ,2,3 ,4-tetrahvdroq¾inoxaline- 1 - carbonyl)tbiazolidin-3-vl)met vl)phenvl)propvl)-Nl.N3-bis((2R,3S,4S.5SV

xybex yl )malonami de

Scheme 174: 1. 92c, NaBH(QAc) 3 , DCM; 2. Diethyl 2-aUylmalonate, 9-BBn, THF, 0 °C then K 3 PO 4 , H 2 0, Pd{dppi) 2 Cl 2j 70 °C; 3. NaOH, THF, 50 °C; 4. D-Glucamine. HAITI. DIE A, DMF. intermediate 174a (Jt)-2,5-dichloro-4-((4-(4-cyd

tetrahydroquinoxdme-l-carbonyl^

trifluoromethanesulfonate: To a solution of 159c (335 mg, 1.03 mrnol, 1.2 equiv) and 92c, (250 mg, 0.87mmol, 1.0 equiv) in dichloromethane (1.7 mL) was added NaBH(OAc)3 (275 mg, 1.29 ' mrnol, 1.5 equiv), The resulting solution was stirred at room temperature for 18 hours and then quenched with aqueous NaHCOs, diluted with dichloromethane (50 mL) and washed with water (4 x 50 mL) and brine (50 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by silica gel column chromatography with an ehient gradient of hexane: ethyl acetate (100: 1 to 5: 1) to furnish 174a (324 rag, 64%). MS (ES, m/∑): 596.0 [M + H ~ f.

Intermediate 174b (R)-diethyf 3-(3-(2,5-dichloro-4-((4-(4-cyclopropyl-l ,2,3,4- tetrahydroquinoxaiine-l -c^

dioxopentanedioate: To diethyl 2-allylmal.onate (200 mg, Immol, 1 ,0 equiv) in dry tetrahydrofuran (0.6 mL) at 0 °C was added dropwise a solution of 9-BBn (0.5M in THE, 2 mL, 1.0 equiv) over 3 minutes. The ice-bath was removed and the reaction mixture stirred overnight Aqueous K 3 PO 4 (636 mg in 0.7 mL H>Q, 3 ,0 equiv) was added dropwise. Half of the resulting solution (1.3 mL, 2.8 equiv) was added to (R)- 2,5-dichloro-4-((4-(4-cyclopropyi-l ,2,3 ,4-tetrahydroquinoxaline- 1 - '

carbonyl)thiazolidin-3-yl)meth.yl)phenyl trifluoromefhanesulfonate (1 08 mg, O. l Smmol, 1.0 equiv), the mixture was purged with N 2 (3 x N 2 /vacuuni cycles), and then Pd(dppf)Cb ( 13.2 mg, 0.09 equiv) was added, The mixture was again purged with N? (3 x ^/vacuum cycles) and then heated to 70 °C under an inert atmosphere. After 2 hours additional alkyl borate solution (0.5 mL, 0.15 mmol, 1.0 equiv) and Pd(dppi)Cl 2 (2.0 mg, 0.013 mmol) were added. After an additional hour the reaction mixture was cooled, diluted with ethyl acetate (50 mL) and washed with w r ater (3 x 50 mL) and brine (50 mL), The combined organic layers were dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by silica gel column chromatography with an eluent gradient of hexane:ethyl acetate (100: 1 to 5:1) to furnish 174b (50 mg, 43%). MS (ES, m/z): 704.3 [M + 11 ! ' .

Intermediate 174c (R)-3-(3~(2,5~dichloro-4-((4-(4-cyciopropyl-l , 2,3,4- tetrahydroquinoxal ii ^ ! -carbo

dioxopentanedioic acid: To a solution of (R)-diethyl 3-(3-(2,5-dichloro-4-((4-(4- cyclopropyl-1. ,2,3,4~tetrahydroquinoxaline- 1 -carbotiyl)thiazolidin-3- yl)methyl)phenyl)propyl)-2,4-dioxopentanedioate (50 mg, 0.077 mmol, 1.0 equiv) in tetrahydrofuran (0.5 mL) was added aqueous NaOII (3M, 0,39 mmol, 0.128 mL, 5.0 equiv) and the resulting mixture was stirred vigorously overnight at 50 °C, The reaction was diluted with ethyl acetate (5 mL) and water (5 mL) and the pH was adjusted to 3 with, aqueous HC1 (1M). The aqueous layer was extracted with ethyl acetate (3 x 5 mL) and the combined organic layers washed with brine (10 mL). The resulting mixture was dried over anhydrous sodium sulfate and concentrated over vacuum to afford 174c (30 nig, 60%) which was used without further purification. MS (ES, m/z): 648.2 [M + fff .

Example 174 (2-(3-(2,5-dichloro-4-(((R)-4-(4-cyclopiOpyl-l 5 2,3,4- tetrahydroquinoxaline- 1 -carbony1)Jliiazolidin.-3-yl)methyl)phenyl)propyl)-N 1 ,N3 - bis((2R,3S 7 4S,5S)-2 ! 3,4 J 5,6-pentahydroxyhexyl)ma!onamide TEA salt. To a solution of 174c (30 mg, 0.05 mmol, 1,0 equiv), D-Gmcamine (20 mg, 0.1 1 mmol, 2.2 equiv) and DIEA (12.9 mg, 0.1 mmol, 2.0 equiv) in DMF (0.5 ml.) was added dropwise HATU (38.2 mg, 0.1 mmol, 2.0 equiv) in DMF (0.5 niL) and the reaction mixture was stirred at room temperature for 5 minutes. The crude solution was diluted with DMF:H 2 0 (1 :T) to 4 ml, acidified with TF , and purified by preparative HPLC with a C18 silica gel stationary phase using a gradient of H 2 0 0.05% TFA : CiI 3 C 0.05% TFA (90 : 10 to 10 : 90) over 30 min and detection by UV at 254 run to give 7.7 mg (13%) of the title compound as a white solid. MS (ES, m/z): 918.5 [M + Ff.

Example 175

pentahydroxyhexy¾)-lH-imidazole-5-carboxamide

Scheme 175: 1. D-glucamine, HATU, DIPEA, DMF.

Example 175: 2-((2.5-dichloro-4-(((R)-4-(4-cyclopropyl-l ,2,3 ,4-tetrali droquinoxaiine- l-carbonyl)tMazolidm-3-yl)^

pentahydroxyhexyl)-lH-imidazole-5-carboxamide. To a mixture of example 161 (23.4 mg, 0.038mmoL 1 equiv) and D-glucamine (8.2 mg, 0.0454 mmol, 1.2 equiv) in DMF (0.2 mL) were added HATU (17.3 mg, 0.045 mmol, 1.2 equiv) and DIPEA (33 uL, 0.19 mmol, 5 equiv). The mixture was stirred at rt for 30 minutes and then purified by prep- HPLC to give 13.3 mg (31 %) of example 175 bis TFA salt as a white solid. Ή NMR (400 MHz. CD 3 OD) δ 7.95 (s, 1H), 7.20 (dd, J - 8.3, 1.3 Hz, 2H), 7.17 - 7.13 (m, 1H), 7.13 - 6.99 (m, 2H), 6.64 (td, ,/ - 7.7, 1.2 Hz, 1H), 4.81 - 4.69 (m, lH), 4.35 (s, 2H), 4.20 - 4.08 (m, 1 H), 4.05 (d, J = 9.8 Hz, IE), 3.98 - 3.89 (m, 1H), 3.88 - 3.81 (m, 1H), 3.82 - 3.75 (m, 6H) 5 3.74 - 3.69 (m, 1H), 3.68 (d, J = 1.9 Hz, 1H), 3.67 - 3.62 (m, ! H), 3.62 - 3.59 (m, 1H), 3.59 - 3.55 (m, 1H), 3.52 - 3.33 (m, 5H), 3.18 - 3.01 (m, HI), 2.48 - 2.38 (m, ill), 0.90 - 0.76 (m, 2H), 0.65 - 0.38 (m, 211). LCMS (ES, m/z): 781.24

Example 176

4-(2,5-dichloro-4-{[(4R -4-[ " (4-cyclopropyl-l ,23,4-letrahy

pentah ydroxyfaexyi benzami dc

Scheme 176

LiOH»H 2 0, 1 ,4-dioxane, ¾0; c. D-glucamine, HATU, DIEA, DMF.

Example 176: 4-(2,5-dichloro-4- { [(4R)-4-[(4-cyclopropyl- 1 ,2,3,4- tetrahydroquinoxalin- 1 -yi)carbonyl]~ l. ,3-thi.azolidtn-3-yl]methyl}phenoxymethyi)-N- [(2S,3R,4 ,5R)-2,3.4,5.6-pentahydroxyhexyl]benzamide. To Example 162 (172 mg, 0.370 mmol), methyl 4-(hydroxymethyl)benzoie acid (77 mg, 0.46 mmol), and tripheayphosphine (121 mg, 0.46 mmol) in DCM (3 mL) at 0 °C was added diisopropyl azodicarboxylate (91 ,uL, 0.46 mmol) and the mixture was allowed to warm to room temperature and then stirred for 16 h. The solvent was removed and the residue dissolved in a. mixture of H 2 0 (5 mL) and 1 ,4-dioxane (25 mL). To this was added LiOH*H 2 0 (62 mg, 1.5 mmol) and the mixture stirred at room temperature for 2 h. The solvent was removed, the residue dissolved in DCM and then washed with 1M aqueous HC1. dried over Na 2 S0 4 , then filtered and concentrated. To a portion of the crude residue (-0.123 mmol), D-grucamine (42 mg, 0.23 mmol), and DiEA (128 jiL, 0.740 mmol) in DMF (2 mL) was added HATU (70 mg, 0.19 mmol) and the reaction stirred for 1 h. The mixture was diluted with j¾0, acidified with TFA, and. then purified by preparative HPLC with a C18 silica gel stationary phase using a gradient of ¾0 0.05% TFA : CH 3 CN 0.05% TFA (70 : 30 to 5 : 95) and detection by UV at 254 ran to give the title compound (21 mg, 17%) as a bis-TFA salt. MS (ES, m/z): 761.34 [M + Hf; Ή NMR (400 MHz, CD 3 OD) δ 7.88 (d, J = 8.0 Hz, 2H), 7.58 (cl, J = 7.1 Hz, 2H), 7.25 is, 1 H), 7.20 (dd, J = 8.3, 1.3 Hz, 1.H), 7.17 - 7.08 (m, 2H), 7.08 - 6.99 (m, 1H), 6.67 (t, J - 7.2 Hz, 1 H), 5.25 (s, 2H), 4.80 - 4.72 (m, IH), 4.29 (d, J - 9.9 Hz, 1H), 4.14 - 3.86 (in, 4H), 3.86 -3.76 (m, 3H), 3.76 - 3.61 (m, 5H), 3.61 - 3.51 (m, IH), 3.47 (dd, J - 13.7, 7.3 Hz, i l l). 3.43 - 3.35 (m, 1 H), 3.21 - 3.09 (m, 2H), 2.44 (s, IH), 0.80 (s, 2H), 0.56 (s, 2H).

Example 177

l-(4-(2, 5 -di chloro -4 - ((( R)-4-( 4-cvc 1 opropyl- 1.2,3 ,4-tetrahydroquinoxaline- 1 - eaxbonvI)miazoiidm-3~yi)^^^^^

pentahydroxyhexypurea

Scheme 177: 1. NaB¾, MeOH. 2. a. MsCl, TEA, DCM: b. 159c, K 2 C0 3 . Nal, DMF; 3. TFA, TES, FLO; 4. DSC, ACN, D-Glucamine. (0.5 mL).

Intermediate 177a: tert-butyl 4-(2,5-dichloro-4-formylph.en.yl)butylcarbamate. Teri-butyl 4-(2,5-dichloro-4-formylphenyl)butylcarbamate was prepared using the procedures described in the synthesis of intermediate 160d, substituting but-3-yn-l- amine for pent-4-yn- 1 -amine, to afford 177a (2.1 Ig) as a brown oil.

Intermediate 177b: tert-butyl N-[4-[2,5-dichloro-4- (hydiOxymethyl)phenyl] butyl] carbamate. To tert-butyl N-[4-(2,5-dichloro-4- formyJphenyl)butyl]carbamate (2.1 1 g, 6.09 mmol, 1.00 equiv) in methanol (30 mL) at 0-5 °C was added NaBH 4 (460 mg, 12.16 mmoi. 2.00 equiv) in several batches over a period of I h. The reaction was stirred for 1 h at 0-5 °C and then quenched by the addition of 50 mL of water. The resulting mixture was concentrated under vacuum and extracted with dichlorom ethane (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous sodium sulfate and concentrated under vacuum to afford 1.70 g (80%) of 177b as brown oil, ^.-NMR (300MHz, CDC1 3 ): 7.45(s, 1H), 7.17(s, 1H), 4.69(s, 2H), 4.69(s, 0.64H), 3.13~3.1 1(m, 2H), 2.70~2.65(m, 2H), 1.93(s, 0.61 i 1.62 ~1.45(m, 4H), 1 .4 l i s. 8.9H). intermediate 177c: (R)-tert-butyl 4-(2,5-dicbloro-4-((4-(4-cyclopropyl-l ,2,3,4- tetrahydroqumoxahhu l -carbony To a solution of tert-butyl N-[4-[2,5-dichloro^~(hydioxyineihyl)phenyl3butyl3carbam^ (150 mg, 0.43 mmol, 1 ,0 equiv) and triethyiamine (87 mg, 0.86 mmoi, 2.0 equiv) in dicMoromethane (0.8 mL) at 0 °C was added niethanesulfonyi chloride (49 mg, 0.43 mmol, 1.0 equiv) and the reaction stirred for 20 minutes. The solvent was removed, the residue dissolved in DMF (0.5 mL), and the mixture was then added to a solution of 159c (1.24 mg, 0.43 mmol, 1.0 equiv) and K 2 C0 3 (65 mg, 0.47 mmol, 1.1 equiv) in DMF (0.5 ml,). Nal (6.4 mg, 0, 1 equiv) was then added and the solution heated to 70 °C overnight. The reaction was diluted with ethyl acetate (40 mL), washed with water (3 x 30 mL) and brine (30 mL), and dried over anhydrous sodium sulfate, The solution was concentrated by vacuum and the residue purified by silica gel column chromatography with an eluent gradient of hexane: ethyl acetate (100: 1 to 4: 1) to furnish 177c (32.9 mg, 12%) as a yellow oil. MS (ES, m/z) 619.1 [M + H] + .

Intermediate 177d: (R)~(3-(4-(4-aminobutyl)-2,5-dich3oroberi2yl)tMazolidm-4- yl)(4-cyclopropyl-3,4-dihydroqxrinoxa]in-l(2H)-yl)methanone. To (R)-tert-butyl 4- (2,5-dichloro-4-((4-(4-cyclopropyl-l ,2,3,4-tetrahyaroqumoxaline-l- carbonyl.)tbiazolidin-3-yi)methyl)pherjiy].)butylcarbamate (33 mg, 0.05 mmol, 1 .0 equiv) was added a solution of irifluoroacetic acid:triethyisilane:water (95:5:5) and the reaction stirred for 5 minutes. The mixture was quenched with aqueous NaHC0 3 (-20 mL), extracted with dichloromethane (2 x 20 mL) and the organic layer was concentratedto afford 177d (30.5 mg, 100%) which was used without further purification. MS (ES, m z): 519.1 [ + H] + .

Example 177: l"(4-(2 . ,5-dichloro-4-(((R)-4-(4-cyclopropyl-l,2,3 5 4- tetrahydroq ' uinoxa]ine-l-carbonyl)thiazolidin-3-yl)

((2R,3S,4S,5S)-2,3,4,5,6-pentahydToxyhexyl)urea. To a solution of 177d (30.5 mg, 0.6 mmol, 1.0 equiv) in acetonitrile (0.5 mL) was added N-N'-disuccmimidyl carbonate (16.5 nig, 0,65 mmol, 1.1 equiv). After 30 minutes, D-glucamine (16 mg, 0.09 mmol, 1.5 equiv) and DMF (0.3 mL) were added and the reaction mixture was stirred at 80 °C for 90 minutes. The resulting mixture was diluted to 4 mL with acetomtrile:waler (1 :1), acidified with TFA, and then purified by preparative HPLC with a C18 silica gel stationary phase using a gradient of ¾0 0.05% TFA : CH 3 C 0.05% TFA (90 : 10 to 5 : 95) and detection by UV at 254 nm to give the title compound (16.7 mg, 29%) as the TFA salt MS (ES, m/z) 726.3 [M + Hf .

Example 178

5 -(2, -dichloro-4-(((R)-4-(4-cyciopropyl- 1,2,3 ,4-tetrah ydroqumoxaline- 1 - carbonvDtMazoUd^

pentahydroxyhexyDpentanamide Scheme 178: 1. D-glucainine, IIATU, DiPEA, DMF.

Example 178: 5-(2,5-dichloro-4-(((R)-4-(4-cyclopropyl- 1 ^ t ~ te†rah droquinoxaline-i -earbony^

2,3,4,5,6-pentahydroxyhexyl)pentanamide. To a mixture of intermediate 163 (1 1 .4 mg, 0.0193 mmol, 1 equiv) and D-glucamine (4.2 mg, 0.0232 mmol, 1.2 equiv) in DMF (0.14 mL) was added HA.TU (8.8 mg, 0.232 mmol, 1.2 equiv) and DiPEA (13.4 μΕ, 0.77 mmol, 4 equiv). The mixture was stirred at rt for 1 h and then purified, by prep- HPLC to give 9,3 mg (52 %) of Example 178 as a white solid. 1H-NMR (400MHz, CD3OD , ppm)\ δ 7.63 (s, 0.4H), 7.41 (s, 0.6H), 7.32 (s, 0.7H), 7.20 (dd, J = 8.3, 1.4 Hz, 1.3H), 7.17 - 7.01 (m, 2H), 6.73 - 6.59 (m, 1 H), 4.86 - 4.65 (m, 1H), 4.23 (d, J - 10.3 f i .. 1 H), 4.17 - 3.86 (m, 3H), 3.84 - 3.80 (m, IH), 3.79 (d, J - 3.3 Hz, IH), 3.76 (d, J 3.5 Hz, IH), 3.72 (dd, J = 4.5, 2.1 Hz, 1H), 3.71 - 3.66 (m, 1H), 3.66 - 3.63 (m, 1H), 3.63 - 3.58 (m, 1 H), 3.47 (d, J - 4.7 Hz, 0.4H), 3.43 (d, J -- 4.7 Hz, 0.6H), 3.39 (dd, J = 1.1.3, 5.8 Hz, I H), 3.36 3.32 (m, 2H), 3.24 (dd, J - 13.8, 7.4 Hz, I H), 3.19 - 3.04 (m, 2H), 2.73 (t, J = 7.2 Hz, 2H), 2.49 - 2.34 (m, IH), 2.33 - 2.15 (m, 211), 1.75 - 1.51 (m, 4H), 0.89 - 0.74 (m, 2H), 0.67 - 0.40 (m, 2H). LCMS (ES, m/z): 711.24 [M + HI .

Example 179

i4-cyclopropy -3,4-di iydroqutnoxalin-l(2HVyl i3-(2,5-di

yDmethanone

Scheme 179: 1. methyl 2-hydroxyacetate, NaH, DMF: 2. DIBAL-H, THF; 3.

formaldehyde, KOH, EtOH/¾0; 4. dieihylcarbonate, EtONa; 5. TEMPO, NaC10 2 , NaCIO, ACN/Phosphate buffer, ¾0; 6. HOAt, EDC1, DMF.

Intermediate 179a: methyl 2-(2,5-dichiorobenzyloxy)acetate. To 2- (bromomethyl)-l ,4-dichlorobenzen.e (5 g, 20.84 mmol, LOO equiv) in DMF (30 ml.) was added methyl 2-hydroxyacetate (1.89 g, 20.98 mmol, 1.01 equiv) and the solution cooled to 0 °C. To this was added in portions sodium hydride (1.0 g, 41.67 mmol, 2.00 equiv). and the resulting solution was allowed to warm to T and then stirred, overnight. The reaction was quenched by the addition of 50 mL of water, the resulting solution was extracted with 3 x 50 mL of ethyl acetate and the organic layers were combined and then w¾shed with 1 x 50 mL of brine. The organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum to afford 5.2 g (80%) of intermediate 179a as yellow oil.

Intermediate 179b: 2-(2,5-dichlorobenzyloxy)acetaldehyde. To intermediate 179a (2 g, 8.03 mmol, 1.00 equiv) in THF (10 niL) at -60 °C was added drop-wise DIBAL-H (25% w/w in Hexane; 6.87 g, 12.10 mmol, 1,50 equiv) and the resulting solution was stirred for 3 li. The reaction was then quenched by the addition of water (20 mL), the pH value of the solution was adjusted to 5 with 1 M aqueous HC1 and the resulting solution was extracted with 3 x 20 mL of ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under vacuum to afford 1.72 g (98%) of intermediate 179b as a yellow oil,

. Intermediate 179c: 2-(2,5-dichiorobenzyIoxy)-2-(hydroxymethyl)propane-L3- diol. To intermediate 179b (1.72 g, 7.85 mmol. 1.00 equiv) in 1 :1 ethano]/H 2 0 (10 mL) was added formaldehyde (40%) in water; 5.91 g, 78.73 mmol, 10.00 equiv) followed by the drop-wise addition of a solution of potassium hydroxide (442 mg, 7.88 mmol, 1.00 equiv) in 1 :1 e hane I/H 2 O (5 mL) and the resulting solution was stirred for 3 h. The mixture was concentrated under vacuum, diluted with water (10 mL) and then extracted with 3 x 20 mL of ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate, concentrated and then purified by silica gel chromatography (dichloromethane/methanol 50/1) to afford 700 mg (32%) of intermediate 179c as a white solid.

Intermediate 179d: (3-(2 J 5-dichlorobeiizyloxy)oxetan-3-yl)methanoI. To intermediate 179c (340 mg, 1.21 mmol, 1.00 equiv) was added diethyl carbonate (215 mg, 1.82 mmol, 1.50 equiv) and sodium ethoxide (160 mg. 2.35 mmol, 0.20 equiv) and the resulting solution was stirred for 1 h at 140 °C and then for an additional 1 h at 190 °C. The reaction mixture was cooled to RT, quenched by the addition of wa ter (10 mL) and the resulting solution was extracted with 3 x 20 mL of ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate, concentrated and then purified by preparative TLC (dichloromethane/methanol 25/1) to afford 100 mg (3 %) of intermediate 179d as yellow oil.

Intermediate 179e: 3-(2 ; 5-dichlorobenzyloxy)oxetane-3-earboxylic acid. To intermediate 179d (150 mg, 0.57 mmol. 1.00 equiv) in ACN/phosphate buffer (7/3.5 mL) was added TEMPO (8.9 mg, 0.06 mmol, 0.10 equiv), aC10 2 (129 mg, 1.43 mmol, 2.51 equiv), NaCIO (11% in water; 19 mg, 0.03 mmol, 0.05 equiv) and the resulting solution was stirred for 20 h at 77 °C. The pH value of the solution was adjusted to 5-6 with aqueous 1 M HC1 and the resulting solution was extracted with 3 x 20 mL of ethyl aeetate. The organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under vacuum to afford 130 mg (82%) of intermediate 179e as a. yellow oil.

Example 179: (4-cyclopropy3.-3,4-dihydroquinoxalin-l(2H)-yl)(3-(2,5- dichlorobenzyloxy)oxetan~3-yl)mefbanone. To intermediate lc (130 mg. 0.47 mmol, 1.00 equiv) in DMF (3 mL) was added 1 -eyciopropyl- 1 ,2,3 ,4-tetrahydroqmnoxaline (82 mg, 0.47 mmol, 1.00 equiv), HOAt (128 mg, 0.94 mmol, 2.00 equiv) and EDC-HCi (1 80 mg, 0.94 mmol, 2,00 equiv) and the resulting solution was stirred for 2 h. The reaction was quenched by the addition of water (10 ml,) and then extracted with 3 x 20 mL of ethyl acetate. The organic layers were combined, washed with 1 x 10 mL of brine, dried over anhydrous sodium sulfate, concentrated and the crude product (100 mg) was purified by reverse-phase (C I S) Prep-HPLC to afford 30 mg (15%) of Example 179 trifiuoroacetic acid salt as an off-white solid. LCMS (ES, m/z): 433 [M +

Hf , l H- MR (300MHz, CDC13, ppm): 7.52-7.27 (m, 1H), 7.26-7.05 (m, 5H), 6.87- 6.50 (m, 1H), 5.20-5.17 (m, 1H), 4.83-4.77 (m, 2H), 4.52 (s, 3H), 3.95-3.61 (m, 2H), 3.47-3.37 (m, 2H), 2.37 (s, 1 H), 0.88-0.79 (m, 2H), 0.68-0.60 (m, 1H), 0.42 (s, 1H).

Example 180

(4-cyciopropvl-3,4-dihydroquinoxalin- 1 (2H)-yl)( 1 -((2,5-

Scheme 180: 1

Example 180 : (4-cy cl opropy 1-3 ,4-dihydroquinoxalm- 1 (2H)-yl)(l -((2,5 -dichloro- phenylthio)methyl)cyclopropyl)methanone. To intermediate 32c (30 mg, 0.10 mmol, 1.00 equiv) in DMF (2.0 mL.) was added 2,5-dichlorobenzenethiol (27.7 mg, 0.15 mmol, 1 ,00 equiv), potassium carbonate (27.6 mg, 0.20 mmol, 2.00 equiv), and KI (1.7 mg. 0.01 rnmol, 0.10 equiv) and the resulting solution was stirred overnight. The reaction was quenched by the addition of water (5 mL) and the resulting solution extracted with 3 5.0 mL of ethyl acetate, the organic layers were combined, dried over anhydrous sodium sulfate and then concentrated under vacuum. The crude product was purified by reverse-phase (C 18) Prep-HPLC to afford 7.8 mg (17%) of Example 180 as a light yellow solid. LCMS (E8, m/z): 433 [M + Hf ; 1H-NMR (300Hz, CD30D, ppm): 7.30-7.34 (m, 21 1 ). 7.05-7.16 (m, 3H), 6.74-6.80 (m, 1H), 6.69-6.74 (m, 1H), 3.83-3.87 (m, 21T), 3.43-3.46 (m, 2H), 2.70 (s, 2H), 2.20-2.27 (m, I II), 1 .43-1.45 (m, 2H), 0.70- 0.90 (m, 2H), 0.67-0.69 (m, 2H), 0.20-0.30 (m, 2H).

Example 181

(R)-(3-(4-(5-guaindipylpentv^^

dihydroqriinoxalin-l 2H - i methanone

Scheme 181 : 1. 1 H-pyrazole- 1 -carboximidarnide HC1, D1EA, DMF.

Example

yl)(4-cyc propyi-3 ,4-dihydroquinoxalin- 1 (2H)-yl)methanone. To intermediate 160 (70 mg, 0.13 rnmol, 1.00 equiv) in DMF (3 mL) at 5 °C was added lH-pyrazole-l- carboximidamide hydrochloride (38.7 mg, 0.26 rnmol, 2.00 equiv) and DIEA (101 ,8 mg, 0.79 rnmol, 6.00 equiv) and the reaction was allowed to warm to RT and then stirred overnight. The reaction was diluted with 10 mL of H 2 0, extracted with 2 x 1 5 mL of ethyl acetate and then the organic layers were combined, washed with. 2 x 30 mL of sodium, chloride and dried over anhydrous sodium sulfate. The solution was concentrated and the residue was purified via reverse-phase (CI 8) Prep-HPLC to afford 32.7 mg (43%) of Example 181 tri-trifluoroacetate as a white solid. LCMS (ES, m/z); 575 [M + Hf . M i-N'M R (400MHz, CD 3 OD, ppm): 7.36-7.20 (m, 2H), 7.17 (m, 1 1 I k 7.12-7.07 (m, 2H), 6.68-6.65 (m, 1H), 4.93-4.56 (s, I Hf 4.1 1 -4.09 (m, 2H), 3.88-3.71 (m, 3H), 3.50-3.42 (m, 111), 3.40-3.46 (m, 3H), 3.33-3.32 (m, 1H), 3.24-3.21 (m, 2H), 3.19-3.12 (m, 1H), 2.76-2.68 (m, 2H), 2,44 (m, 1H), 1.71-1.62 (ra, 4H), 1.50-1.42 (m, 21 1). 0.84-0.82 (m, 2H), 0.60-0.50 (m, 2H).

Example 182

4-{2.5~dicMQro-4~({(R)~4~( ~c^

earbonyl¼hiazolidm-3-yDme ^

entahydroxyhexyDbeiizamide

Scheme 182: 1. a. Methyl 4-hydroxybenzoate, DEAD, PPh 3 , toluene; b. LiOH, THF, l¾ ; 2. D-glucamine, HATU, DIPEA, DMF.

Intermediate 182a: (R)-4-(2,5-dichloro-4-((4-(4-cyclopropyl-l ,2,3 5 4- tetxahydroquinoxaline-l-carbonyl)m^ acid. Intermediate 182a was prepared using the procedures described for the preparation of example 161 except that methyl 4-bydroxybenzoate .was used in place of methyl 2- mercapto- 1. -methyl- 1 H-imidazol e-5-carboxylate.

Example 182: 4-(2,5-dichloro-4-(((R)-4-(4-cyclopropyl-l,2,3,4- tetrahydroqiMnoxal e- i-carbo^

((2S,3R.,4R,5R)-2 ,4,5 5-pentahydroxyhexy.i)benzamide. Example 182 was prepared using the procedures described for the preparation of example 175 to provide the title compound bis TFA salt as a white solid. (ES, rn/z): 761 [M + H] + , (400MHz, CD 3 OD, ppm): 7.8-5-7.87 (m, 2H), 7.65-7.67 (m, 1 H), 7.59 (s, 1H), 7.12-7,34 (m, 5H), 6.69 (s, 1 H), 5.23(s, 2H), 3.63-4.31 (m, 18H), 3.19-3.33 (m, 211), 2.45 (s, 1H), 0.83-0.84 (m, 2H), 0,56 (m, 2H).

Example 183 (l -(2-cMoro-5-cyclopropvlbenzylamino)cvclopropyl)(4-cvc1opropy l-3,4- dihydroqumoxalm- 1 (2K)-yl)methanone

Scheme 183: 1. 1641 2 C0 3 , DMF; 2, Cyclopropylboronic acid, Pd( ' dppf)Cl 2 , K 2 C0 3 , Toluene, H 2 0.

Ϊ niermediate 183 a : (l-(5 -brom o-2-chlorobenzylamino)cyc 1 opropy 1)(4~ cyciopropyl-3,4-dihydroquirtoxalin- 1 (2H)-yl)methanone. To a solution, of intermediate 4f (100 mg, 0.39 mmol, 1.00 equiv) in DMF (3 mL) was added 4-bromo-2- (bromomethyl)- 1 -chlorobenzetie (128.4 mg, 0.45 mmol, 1.16 equiv) and potassium carbonate (107.4 mg, 0.78 mmol, 2.00 equiv) and the reaction was stirred overnight. The mixture was diluted with 10 mL of 1¾0, extracted with 25 mL of ethyl acetate, and the organic layer was washed with brine (2 x 25 mL), dried over anhydrous sodium sulfate and concentrated to afford 200 mg crude 183a as brown oil.

Example 183 (l-(2-cUoro-5-cyclopropylbenzylamirio)cyclopropyl)(4- cyclopropyl-3,4-dmyckoquinoxdin-l(2H)-yl)methanone. To 183a (160 mg, 0.35 mmol, 1.00 equiv) in tomene/¾0 (2/0.2 mL) was added cyclopropylboronic acid (90 mg, 1.05 mmol, 3.00 equiv), Pd(dppf)Cl 2 (25 mg, 0.03 mmol, 0.10 equiv) and potassium carbonate (145 mg, 1 .05 mmol. 3.00 equiv) and the reaction was stirred at 80 °C overnight. The mixture was then concentrated under vacuum and the crude product (200 mg) purified by reverse-phase (CI S) prep-HPLC to afford 31.9 mg (22%) of Example 183 as a brown solid. (ES, 422 [M + H] + (300MHz, CD 3 OD , ppmy, 7.28 (d, J - 6.6Hz, 1H), 7.26-7.12 (m, 3H), 6.94 (d, J = 2.4Hz, IH), 6.72-6.62 (m, ill), 6.62 (s, IH), 3.86-3,79 (m, 411), 3.40-3.36 (m, 2H), 2.37 (m, I E), 1.79 (m, IH), 1.37-1.35 (m, 2H), 1.07-1.05 (m, 2H), 0.94-0.91 (m, 2H),0.76-0.74 (m, 2H), 0.60-0.56 f.m. 21 ! ). 0.43-0.41 (m.2! f t. Examples 184-291

Table 1 illustrates the method of preparation for examples 184-291, which were prepared using commercial or known starting materials according to the general methods described in examples 1-98 and 159-183 and methods generally known to those skilled in the art.

General synthetic method A: addition of an amine nucleophile inteanediate, for example 160 or the like, with a reactive di or trihaloaromatic or heteroaromatic ring, such as 2,4,6-tri.chloro-l,3,5-triazine, followed by sequential reactio with one or more amine nucieophil.es to provide the example compound. The synthesis of Example 173 is a typical procedure.

General synthetic method B: hydroboration of an aikene followed by Suzuki coupling to a halide or inflate intermediate, tor example intermediate 174d or the like. This method allows incorporation of one or more reactive functional groups and permits further functionality to be appended. The synthesis of Example 174 is a typical procedure.

General synthetic method C: reductive amination of a diaidehvde to provide an alcohol, intermediate such as 159. Mitsunobu alkylation allows additional functionality to be appended to the free alcohol. The synthesis of Example 182 is a typical procedure,

General, synthetic method D: reductive amination with a hydroxyaldehyde to provide a phenol intermediate such as example 162. Mitsunobu alkylation allows additional functionality to be appended. The synthesis of Example 176 is a typical procedure.

. General synthetic method E: the synthesis of alkyamine, aJkykarboxylate, and alcohol intermediates such as 160, 178g and 171 j by Sonogashira coupling to an. arylhalide or inflate followed by reduction of the alkyne is demonstrated in several examples. Typical functional, group transformations allow functionality to be appended to these Intermediates through ureas, amides amines, guanidines and sulfonamides. The synthesis of examples 171, 172, 177, .1.78, and 181, are typical procedures. Table!

IJ54

Ϊ60

837.34

536.22

[Mi H]

862.33

223 A 861.31

[M+H]÷

905.29

224 A 904.35

ħĮ73

IJ74

no

.80

>83

Example 448.1

447,09

291 [M+Hj 4

Example 292 Cell-Based TGR5 Assays

Two primar cell based screens were performed. The first utilized. HEK293 cells stably transfected to heterologously express human TGR5. The second screen used the human caecum carcinoma, ceil line NCI- H71.6 which endogenously expresses human TGR5. In both assays, ceils are treated with candidate TGR.5 activators and assessed, for the increased intercellular levels of c AMP.

HEK293 cells were transfected with a vector that expresses a gene encoding human TGR5, and a stable cell line was isolated using drug selection following standard techniques. Cells were grown overnight at 37 °C 15% C0 2 prior to assay. NCJ-H716 were grown in culture dishes coated with Ma rigel (Becton Dickinson) per the supplier's instructions and grown at 37 °C 15% C0 2 for 48h prior to assay.

TGR5-mediated cAMP generation was measured using a homogeneous time resolved fluorescence (HTRF) detection method (Cisbio). Test compounds were dissolved in DMSO to a final concentration of 10 rnM. Serial 3 -fold dilutions of the stock solution were made in DMSO, and these solutions were diluted 100-fold into Hanks Balanced Salt Solution supplemented with 10 mM HEPES pH 7.4 and 0.5 raM isobutyl methylxanthine (IBMX). Prior to assay, culture medium was replaced with. fresh medium, and test compounds diluted in HBSS/HEPES/IBMX were added to the cells and incubated at 37 °C for 30 minutes. Each compound was tested in duplicate at 1 2 concentrations ranging from 0.05 nM to 10 μΜ HEK293/hTGR5) or 22 nM to 50 μΜ (NCI H716).

Following incubation with test compounds, cAMP was detected through the successive addition of cAMP labeled with the modified allophyocyanin dye d2 (cAMP-d2) and cryptate-labeled anti-cAMP in lysis buffer, and reading HTRF per the manufacturer's instructions. A standard curve was used to convert the raw HTRF data into [cAMP]. The concentration of cAMP was plotted against log [test compound] and the resulting curves were fit to a 3 -parameter logistical equation using GraphPad Prism to determine pECso (the negative log of the ECso) and the magnitude of the response. ECso values are reported in the table below. The magnitude of the maximum response was typically between 50 and 200% of the maximum response elicited by a benchmark compound that had a maximum response similar to that elicited by lithocholic acid.

The results of this assay are set forth in Table 2.

Table 2

pECso Values of Representative Compounds

2012/071251

73 A A

74 B

75 B B

76 A B

77 A C

78 A B

79 B C

80 A B

S i B C

82 B C

83 B c

A B

85 B C

86 D K

87 A B

88 A A

89 B C

90 A B

91 B B

92 C C

93 B B

94 C C

95 A c

96 A A

97 A A

98 A B

159 A

160 B

161 B

162 B

163 A

164 B

165 A

166 A

167 B

168 B

169 A

170 B

171 B

172 C

173 B

174 C 175 A

176 B

177 A

178 A

179 A B

1 80 A B

181 A

182 B

1 83 A

184 B B

185 B C

186 B C

1S7 B C

188 B B

189 A B

190 C

191 A B

192 A

193 A

194 A

195 B

196 B

197 c

198 A

199 B

200 B

20 i B

202 B

203 B

204 B

205 B

206 A

207 B

208 A

209 C

210 A

21 1 A

212 C C

213 B c

214 B B

215 B C

216 B 1 c 259 A

260 B

261 B

262 C

263 C

264 B

265 B

266 C

267 c

268 A B

269 C

270 B

271 C

272 c

273 B

274 B

275 B

276 A

")ΊΊ B

278 B

279 A

280 B

281 A

282 A

28.3 B

284 C

285 A

286 B

287 B

288 B

289 B

290 B

291 C

*pEC 50 values are expressed as the follo wing ranges: A is a pEC 5 o of 7+, B is a pECso of 6-6.9, C is a pEC 50 of 43-5.9, D is a pEC 50 of < 5, E is a EC 50 of < 4,3

Example 293

In Vivo GLP-1 Secretion and Gallbladder Measurement T/US2012/071251

C57BL/6 male mice on regular chow had food removed in the morning and were dosed with, vehicle (10% hydroxypropyl-p-cyclodextrin or 2% DMSO in 0.4% hydroxypropy] methylcellulose) or the test conipoimd in vehicle to achieve a dose of 30 mg/kg. Eight hours later, each mouse was heavily anesthetized with isofiurane, the peritoneal cavity was opened and the gallbladder, with its entire contents, was carefully excised and weighed. Blood was collected from the left ventricle of the heart and processed to plasma in EDTA-coated tubes containing aprotinin and DPPIV inhibitor for measurement of total GLP-1 (K.150FCC; Meso Scales Discovery, Gaithersburg MD).

As shown in Tables 2 and 3, of the compounds tested, 18 significantly increased GLP-1 levels, and of these 18, 8 had no significant effects on gallbladder weight/body weight.

GLP-1 Levels

n.s. = non significant.

Table 4 Gallbladder Weight

n.s. = non significant

Example 294

Determination of Compound Concentration in Gall Bladder Compound concentrations in the gall bladder were determined as follows: each mouse was heavily anesthetized with isoflurane, the peritoneal cavity was opened and the gallbladder, with its entire contents, was carefully excised. After harvest, gall bladders were homogenized in 100 μΐ, water using a micro-homogeriizer. Samples of homogenate were diluted 1 :5 in water and precipitated with three volumes of neat acetonitrile. After centrifugation, supernatants were analyzed by LC-MS MS. The level of test compound present in gall bladder samples was interpolated from a standard curve for each individual compound prepared in a matrix of gallbladder homogenate from vehicle treated animals. Table 5 summarizes data collected for selected example compounds and shows compound concentration in gallbladders collected from the mice in example 293. Table 5

Example Compound Concentration in Gallbladder at 8 Hours Post Dose

Number of Mice

Mean Example with

Gallbladder Gallbladder Concentration Concentrat on <

Example LLOQ (μΜ) (μΜ) LLOQ

98 0.036 25.76 0/8

165 0.012 0.6 0/8

166 0.012 - 8/8

167 0.012 3.3 0/8

169 0.012 ] ? 0/8

175 0.003 0.06 0/8

176 0.013 0.1 0/8

177 0.014 0.1 0/7

178 0.007 0.3 0/8

191 0.003 0.1 0/8

193 0.015 0.9 0/8

198 0.037 1.3 0/8

208 0.003 0.2 0/7

218 0.004 0.1 0/8

0.007 1.73 0/7

228 0.003 0.60 0/8

237 0.004 1.6 1/8

238 0.034 4.4 0/8

239 0,003 _ 8/8

240 0.003 3.92 0/8

241 0.060 - 8/8

258 0.004 0.7 0/8

259 0.004 0.1 0/8

276 0.077 4.7 0/8

278 0.012 3.1 0/8

279 0.738 30.8 0/7

282 0.007 0.08 0/7

283 0.037 3.06 0/8

285 0.003 0.0.4 0/8

LLOQ^ Lower Limit of Quantification

Example 295

Determination, of Compound Plasma Concentration Blood samples collected as described in Example 293 were processed to plasma by cenirifugation. Plasma samples were treated with acetonitrile containing an internal standard, precipitated proteins removed by centriiiigation. Supernatants were analyzed by LC-MS/MS and compound concentrations were determined by interpolated from, a standard curve prepared in plasma. Table 6 summarizes data collected for selected example compounds for compound concentration in plasma collected in example 293.

Table 6

Example Compound Concentration in Plasma at 8 Hours Post Dose

Number of

Mean Example Mice with

Plasma Plasma

LLOQ Concentration Concentration

Example (ng/ ' mL) (ng/mL) < LLOQ

239 2 2.0 678

240 0.5 28.1 0/8

241 10 - 8/8

258 1 9.4 7/8

259 1 L I 6/8

276 , 7.6 0/8

278 2.3 5/8

279 1 2.0 0/8

282 0.5 2.8 0/8

283 10 24.8 7/8

285 0.5 0.7 2/8

LLOQ - Lower Limit of Quantification

Example 296

Counter screens vs. Ileal bile acid transporter (IB AT) and Farnesoid X Receptor (FXR)

IBAT: HEK293 cells were transfected with a vector that expresses a. gene encoding human IBAT and inhibition of the uptake of [Taurine "3 H]iauroeholic acid was measured in a manner similar to that described by Craddock (Craddock_1998). IBAT-transfected cells were overiayed with uptake buffer (10 mM HEPES, 116 mM sodium chloride, 5.3 mM KC1, 3.8 mM CaCl 2 , 1 1 mM glucose, 1.1 mM H 2 PO^ pFI 7.4) containing 10 μΜ f 3 H]taurocholic acid (American Radiolabeled Chemicals, St. Louis, MO) and 0 to 26 uM test compound. Following a 40-min incubation, the solution was removed, and the cells were washed twice with uptake buffer. Cells were lysed by addition of 20 μί- 0.1% Tween 80 followed by 100 μΐ , scintillation fluid and counted using a TopCoimt (Perkin Elmer).

Taurocholate, deoxychoiate, and chenodeoxycholic acid each inhibited uptake of [ 3 H]taurocholic acid with potency similar to that reported by Craddock (Craddock_1998) ; none of the test compounds inhibited [ 3 H]taurochoiic acid uptake (pi:C50 < 4.6; Table 7). FXR: The ability of test compounds to activate FXR (NR1H4) was measured using a cell-based assay kit obtained from Indigo Biosciences (State College, PA). Cells expressing human FXR and a FXR-responsive luciferase reporter gene were grown in duplicate in the presence of 0.4 to 50 μΜ test compound in buffer according to the manufacturer's instructions. The assay was benchniarked. using GW 4064 and chenodeoxycholic acid, which showed ECso values of 6.8 and ~ 4-4.5, respectively, similar to literature reports (Maloney, 2000). None of the compounds tested showed any inhibition of human FXR (pEC50 < 4.3; Table 7), References:

Craddock, A.L., Love, M.W., Daniel, R.W., Kirby, L.C., Waiters, H.C., Wong, M,H. and Dawson, P. A., .1998, Expression and transport properties of the human ileal and renal sodium-dependent bile acid transporter: Am. J. Gastrointest. Liver. Physiol, v.274, p. Gl 57-69.

Maloney, P.R. et al., 2000, Identification of a chemical tools for the orphan nuclear receptor FXR: J . Med. Chem, v. 43, no. 16, p. 2971 -4.

Table 7

ECso Values of Representative Compounds**

15 . G

16 G

ί 7 G

18 G

19 G

20 G

21 G

ΊΊ G

23 G

24 G

25 G

26 G E ι, 1 G

28 G

29 G

31 G

32 E

34 E

36 G

37 G

40 G

56 G

G

61 G

62 G

63 G

64 G

65 G

66 G

73 E

75 E

80 E

88 E

175 G 176 G E

1 77 G E

1 78 G E

193 G

198 G

218 G

228 G

238 G

240 G

24 G

** EC.5o values are expressed as the following ranges: A is a pEC 5 ,j of 7+, B is a pECso of 6-6.9, C is a pEC 50 of 4.3-5.9, D is a pEC 50 of < 5, E is a pEC S o of < 4.3, F is a pEC 50 of < 5.5, G is a EC'sc. of < 4.6.

Example 297

In Vivo Gallbladder Measurement 16 Hours Post-Dose C57BL/6 male mice on regular chow had food removed (to prevent gallbladder emptying) and were dosed in the early evening with vehicle (2% DMSO in 0.4% hydrox propyl methylcellulose) or 30 mg/kg of examples 176, 177, or 178 formulated in vehicle. The next morning (~16 h post-dose), mice were heavily anesthetized with isoflurane, the peritoneal cavity was opened and. gallbladders (with contents) were carefully excised and weighed. Compound levels in gallbladder were analyzed as described in Example 293.

Example compounds 1.76 and 178 had no significant effect on gallbladder weight/body weight, whereas example 177 did (Table 8).

Table 8

Gallbladder Weight 16 j lours Post Dose

*p < 0.05 vs. vehicle; One-way AN OVA followed by

Dunnett's test

n.s. ~: non significant

Table 9

Example Compoimd Concentration in Gallbladder at 16 Honrs Post Dose

l .i Oij Lower Limit of Quantification

Example 298

In Vivo Measurement of Gallbladder . Emptying CD-I female mice on regular chow had food removed (to prevent gallbladder emptying) in the late evening. The next morning (~ 16 h later), groups of mice were dosed orally with vehicle (10% hydroxypropyl-3-cyclodextrin; 2 groups), devazepicle in water (1 mg/kg; a CC receptor antagonist), or examples 176 and 1 78 (30 mg kg in vehicle). One hour later, 1 group of vehicle-treated mice was dosed orally with saline, and all other groups were dosed orally with lyophilized egg yolk [0.75 mL; 30% (wt/vol) reconstituted in saline for induction of CCK-mediated gallbladder emptying]. Fifteen minutes later, mice were heavily anesthetized with isoflurane, the peritoneal cavity was opened and gallbladders (with contents) were carefully excised and weighed. Examples 176 and 178 did not inhibit CC -mediated gallbladder emptying, whereas devazepide, as expected, did (Figure 1).

Example 299

Time Course of In Vivo Induction of GLP-1 and PY Y

C57BL/6 male mice on regular chow received an oral close of vehicle (2% DMSO in 0,4% hydroxypropyl methylcellulose) or examples 176 or 178 (at 30 mg/kg). Food was removed prior to/or at the time of dosing: 4, 8, 12, and 16 h groups fasted for 7, 8, 12 and 16 h, respectively. At the appropriate time after dosing, mice were heavily anesthetized with isoflurane, and blood was collected from the left ventricle of the heart and processed to plasma as described in Example 293 for measurement of total (t) GLP-1 and, additionally, total ( t)PYY ( 45ZA-1 ; Meso Scales Discovery, Gaithersburg MD).

Examples 176 and 178 caused a sustained induction of tGLP-l and tPYY levels following a single oral dose. tGLP-l was significantly elevated at 4, 8, 12, and 16 h and 4 and 8 for examples 176 and 178, respectively (Figure 2, panel A), and tPYY levels were significantly elevated at 8, 12, and 16 h and 8 h for Examples 176 and 178, respectively (Figure 2, panel B).

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.