Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
C17-ALKANEDIYL AND ALKENEDIYL DERIVATIVES OF OLEANOLIC ACID AND METHODS OF USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2014/040060
Kind Code:
A1
Abstract:
Disclosed herein are novel C17-alkanediyi and aikenediyl derivatives of oleanolic acid, including those of the formula (I), wherein the variables are defined herein. Also provided are pharmaceutical compositions, kits and articles of manufacture comprising such compounds. Methods and intermediates useful for making the compounds, and methods of using the compounds, for example, as antioxidant inflammation modulators, and compositions thereof are also provided.

Inventors:
BENDER CHRISTOPHER F (US)
JIANG XIN (US)
ANDERSON ERIC (US)
VISNICK MELEAN (US)
Application Number:
PCT/US2013/059027
Publication Date:
March 13, 2014
Filing Date:
September 10, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
REATA PHARMACEUTICALS INC (US)
International Classes:
C07J63/00; A61K31/56; A61P29/00
Domestic Patent References:
WO2009129548A12009-10-22
WO2009129545A12009-10-22
WO2012125488A12012-09-20
Foreign References:
US8129429B22012-03-06
US7915402B22011-03-29
US8124799B22012-02-28
US8071632B22011-12-06
US8338618B22012-12-25
US7943778B22011-05-17
Other References:
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SASAKI, KAZUE ET AL: "Preparation of triterpene derivatives for treatment of liver disease", XP002716627, retrieved from STN Database accession no. 2001:651412
P. H. STAHL & C. G. WERMUTH: "Handbook of Pharmaceutical Salts: Properties, and Use", 2002, VERLAG HELVETICA CHIMICA ACTA
"March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2007
"Handbook of Pharmaceutical Salts: Properties, and Use", 2002
ABRAHAM; KAPPAS, FREE RADICAL BIOL. MED., vol. 39, 2005, pages 1 - 25
AHMAD ET AL., CANCER RES., vol. 68, 2008, pages 2920 - 2926
AHMAD ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 35764 - 9
ARAUJO ET AL., J.IMMUNOL., vol. 171, no. 3, 2003, pages 1572 - 1580
BACH, HUM. IMMUNOL., vol. 67, no. 6, 2006, pages 430 - 432
CHAUHAN; CHAUHAN, PATHOPHYSIOLOGY, vol. 13, no. 3, 2006, pages 171 - 181
DICKERSON ET AL., PROG NEUROPSYCHOPHARMACOL BIOL. PSYCHIATRY, 6 March 2007 (2007-03-06)
DINKOVA-KOSTOVA ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, no. 12, 2005, pages 4584 - 4589
DUDHGAONKAR, EUR. J. PAIN, vol. 10, no. 7, 2006, pages 573 - 9
FORSTERMANN, BIOL. CHEM., vol. 387, 2006, pages 1521
"Handbook of Pharmaceutical Salts: Properties, and Use", 2002, VCRLAG HELVETICA CHIMICA ACTA
HANSON ET AL., BMC MEDICAL GENETICS, vol. 6, no. 7, 2005
HONDA ET AL., BIOORG. MED. CHEM. LETT., vol. 12, 2002, pages 1027 - 1030
HONDA ET AL., J. MED. CHEM., vol. 43, 2000, pages 4233 - 4246
HONDA ET AL., J. MED. CHEM., vol. 43, 2000, pages 1866 - 1877
HONDA ET AL., BIOORG. MED. CHEM. LETT., vol. 7, 1997, pages 1623 - 1628
HONDA ET AL., BIOORG. MED. CHEM. LETT., vol. 9, no. 24, 1999, pages 3429 - 3434
HONDA ET AL., BIOORG.MED. CHEM. LETT., vol. 8, no. 19, 1998, pages 2711 - 2714
HONDA ET AL., BIOORG. MED. CHEM. LETT., vol. 16, no. 24, 2006, pages 6306 - 6309
HONG ET AL., CLIN CANCER RES, vol. 18, no. 12, 2012, pages 3396 - 406
ISHIKAWA ET AL., CIRCULUTION, vol. 104, no. 15, 2001, pages 1831 - 1836
KAWAKAMI ET AL., BRAIN DEV., vol. 28, no. 4, 2006, pages 243 - 246
KENDALL-TACKETT, TRAUMA VIOLENCE ABUSE, vol. 8, no. 2, 2007, pages 117 - 126
KRUGER ET AL., J.PHARMACOL. EXP. THER., vol. 319, no. 3, 2006, pages 1144 - 1152
LEE ET AL., GLIA, vol. 55, no. 7, 2007, pages 712 - 22
LENCZ ET AL., MOL. PSYCHIATRY, vol. 12, no. 6, 2007, pages 572 - 80
LIBY ET AL., CANCER RES., vol. 65, no. 11, 2005, pages 4789 - 4798
LIBY ET AL., NAT. REV. CANCER, vol. 7, no. 5, 2007, pages 357 - 356
LIBY ET AL., MOL. CANCER THER., vol. 6, no. 7, 2007, pages 2113 - 9
LIU ET AL., FASEB J, vol. 20, no. 2, 2006, pages 207 - 216
LU ET AL., J.CLIN. INVEST., vol. 121, no. 10, 2011, pages 4015 - 29
MARCH'S ADVANCED ORGANIC CHEMISTRY: REACTIONS, MECHANISMS, AND STRUCTURE, 2007
MCLVER ET AL., PAIN, vol. 120, no. 1-2, 2005, pages 161 - 9
MORRIS ET AL., J. MOL. MED., vol. 80, no. 2, 2002, pages 96 - 104
MORSE; CHOI, ANI. J. RESPIR. CRIT. CARE MED., vol. 172, no. 6, 2005, pages 660 - 670
MORSE; CHOI, AM. J RESPIR. CRIT. CARE MED., vol. 27, no. 1, 2002, pages 8 - 16
PALL, MED. HYPOTH., vol. 69, 2007, pages 821 - 825
PERGOLA, N ENGL J MED, vol. 365, 2011, pages 327 - 336
PLACE ET AL., CLIN. CANCER RES., vol. 9, no. 7, 2003, pages 2798 - 806
RAJAKARIAR ET AL., PROC. NATL. ACAD. SCI. USA, vol. 104, no. 52, 2007, pages 20979 - 84
ROSS ET AL., AM. J. CLIN. PATHOL., vol. 120, 2003, pages 53 - 71
ROSS ET AL., EXPERT REV. MOL. DIAGN., vol. 3, no. 5, 2003, pages 573 - 585
RUSTER ET AL., SCAND. J. RHEUMATOL., vol. 34, no. 6, 2005, pages 460 - 3
SACERDOTI ET AL., CURR NEUROVASE RES., vol. 2, no. 2, 2005, pages 103 - 111
SALVEMINI ET AL., J. CLIN. INVEST., vol. 93, no. 5, 1994, pages 1940 - 1947
SARCHICLLI ET AL., CEPHALALGIA, vol. 26, no. 9, 2006, pages 1071 - 1079
SATOH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, no. 3, 2006, pages 768 - 773
SCHULZ ET AL., ANTIOXID. REDOX. SIG., vol. 10, 2008, pages 115
STREJAN ET AL., J. NEUROIMMUNOL., vol. 7, 1984, pages 27
SUH ET AL., CANCER RES., vol. 58, 1998, pages 717 - 723
SUH ET AL., CANCER RES., vol. 59, no. 2, 1999, pages 336 - 341
SZABO ET AL., NATURE REV. DRUG DISC., vol. 6, 2007, pages 662 - 680
TAKAHASHI ET AL., CANCER RES., vol. 57, 1997, pages 1233 - 1237
TAMIR; TANNEBAUM, BIOCHIM. BIOPHYS. ACTA, vol. 1288, 1996, pages F31 - F36
XIE ET AL., J.BIOL. CHEM., vol. 270, no. 12, 1995, pages 6894 - 6900
ZHOU ET AL., AM. J. PATHOL., vol. 166, no. 1, 2005, pages 27 - 37
Attorney, Agent or Firm:
VOGES, Mark, H. (1120 S. Capital of Texas HighwayBuilding One, Suite 20, Austin TX, US)
Download PDF:
Claims:
C AIM :;s

A

(l wherein:

Y is a covalent bond, -CH2- -C(0)-, -0-, or -NH-;

Ri and R2 are each independently -Ή, -OH, methyl, or as defined below; and

R¾ is:

hydrogen, hydroxy, halo, amino, N ! 10! i . or mercapto; aikyl(c<8), ail :enyl(c g), alkynyl(c 8), aiyl(c 8), aralkyi<c<si, heteroaryl(c s), heierocycloalkyl(c<s), acyl<c<s), alkoxy(c<8), alkenyioxyvcss), aryioxy(c<8), aralkoxy(c<8), heteroaryloxy(c<g), acyio y(c<8j, heterocycloalkoxy(c<s), alkylainmo(c<8), diaIkylamino(c<s), alkeny laminO(c 8), alkoxy mino, , arylaminO(c<8), aralkylaminO(c<s), heteroaiylamino(c≤8), heterocyclo- alkylaminO(c<s), alkylsulfonylamino(c<8), arnidO(c<8), -NH-amidO(c<8), or a substituted version of any of these groups;

R¾ and R], taken together, are -0-, -NR2- or a covalent bond between

Y and carbon atom 13, wherein Ra is hydrogen or alkyl(c<4); or R3 and R¾ taken together, are -0-, --NRa-- or a covalent bond between

Y and carbon atom 14, wherein Ra is hydrogen or alkyf(c≤4); or a pharmaceutically acceptable salt thereof. The compound of claim 3 further defined as:

wherein:

Y is Ci i : . -C(O)-, -O- or N i l ;

Ri and R2 are each independently -H, -OH, methyl, or as defined below; and R3 is:

hydrogen, hydroxy, halo, amino, -NHOH, or mercapto: aikyl(c 8), aikenyl(C<s), alkynyl(C<s)5 aryl(C<s), aralkyi{C<s), heteraaryl(C<s), heterocycloalkyl(c<8), acyi(C<8), aikoxy(e<8), alkenyioxy(C<8 ), aryloxy c<8), aralkoxy(c 8), heteroaryioxy(c<8 >, acyloxy<c≤8), heterocycloalkoxy(c≤8), alkylamino e<8), dialkylamino(c<8), alkenylamino(c<8), alkoxy amino, c<s,i, arylamino(c<s)» aralkylaminofc<8), heteroarylamino(c<8), heterocyc!o- alk}'1aminO(c<8), alkyisulfony!ammo(c<s), amido<c<s), -NH-amidO(c<8), o a substituted version of any of these groups;

R3 and Ri, taken together, are ~0~, -NRa- or a covalent bond between

Y and carbon atom 13, wherein Ra is hydrogen or alkyi,c<4>; or R? and R2, taken together, are -0·-. -NRa- or a covalent bond bet ween

Y and carbon atom 14, wherein Ra is hydrogen or alkyi(c<4); or a pharmaceutically acceptable salt thereof.

3. Th

wherein :

Y is a covalent bond, -CH2- ~C(0)~, -0-, or -NH-; and

¾ is:

hydrogen, hydroxy, halo, amino, -NHOH, or mercapto; or

aikyl(c<8), alkenyl(c<8), alkynyl(c 8), a.ryl(c<s), am i ky i, , . ,. heteroaryl(c<s), heierocycloalkyl(c<8), acyi<c<s), alkoxy(c<8), alkenyloxy(c<8 ), aryioxy'(c<8), aralkoxy(c<s>, heteroaryloxy(c<s >, acyloxy(c<s), heierocyck»aikoxy<c<s), aikylamino(c<8), diaikyla:mino(c<8), alkenylamino(c 8), alkoxyamino<c<8), arylamino(c<s), aralkyiaminoicss), heteroarylamino(c<8), heterocyclo- alkylaminO(c<S h alkyisulfonylaminO(c<8), amidols), -NH- amidO(c<8), or a substituted version of any of these groups;

or a pharmaceutically acceptable salt thereof.

4. The compound of claim 3, further defined as:

wherein:

Y is CU . . ( (() ! . -0-, or Ni l : and

R3 is:

hydrogen, hydroxy, halo, amino, -NHOH, or mereapio: or

aikyl(c<8), aikenyl<c<3;., alkynyl(c<s), aryl(c<g), aralkyi<c<S), heteroaryl(c<g), heierocycloalkyl, css.i, acyi<c<s), alkoxy(c<8), alkenyioxyvcss >, aryioxy'(c<8), aralkoxy(c<s>, heteroary loxytc s >, acyloxy(c<s), heierocycloalkoxy(c<S), aikylamino(c<8), dialkyla:mino(c<8), alkenylamino(c 8>, alkoxyamino<c<8), arylamino(c<8), aralkyiaminoicssi, heteroarylamino(c<8), heterocyclo- alkylamino,c<s,i, alkyisulfonylamino<c s,i, midols), -NH--amido: c<8), or a substituted version of any of these groups;

or a pharmaceutically acceptable salt thereof.

The compound according to any one of claims 1-4, wherein the bond between carbon atoms a and b is a single bond.

The compound according to any one of claims 1 -4, wherein the bond between carbon atoms a and b is a double bond.

{00086504} 99 The compound according to any one of claims 1 , 3 and 5-6, wherein Y is a covalent bond.

8. The compound according to any one of claims 1 -6, wherein Y is -CH2-.

9. The compound according to any one of claims 1 -6, wherein is -C(0)-~. 10. The compound according to any one of claims 1 -6, wherein Y is -0-.

1 1. The compound according to any one of claims 1 and 5- 10, wherein i is ~H.

12. The compound according to any one of claims 1 and 5- 1 0, wherein R2 is methyl.

13. The compound according to any one of claims 1 - 12, wherein R¾ is -H.

14. The compound according to any one of claims 1 - 12, wherein ¾ is -OH. 15. The compound according to any one of claims 1 - 12, wherein R¾ is amino.

16. The compound according to any one of claims 1 - 12, wherein R¾ is alkyl(c<¾).

1 7. The compound of claim 16, wherein R3 is methyl.

18. The compound according to any one of claims 1 - 12, wherein ¾ is heterocycloalkyl,'c<8)- 19. The compound of claim 18, wherein ¾ is morpholinyl, pyrrolidinyl, azetidirryl or piperazinyi.

20. The compound according to any one of claims 1 - 12, wherein ¾ is substituted heterocycioaikyl(c<8).

21. The compound of claim 20, wherem Rj is hydroxypyrrolidinyl, difluoropyrrolidinyl, hydroxypiperidinyl, or N-Boc-piperazinyl.

22. The compound according to any one of claims 1 - 12, wherein R3 is acyl(c<8).

23. The compound of claim 22, wherem R3 is acetyl.

{00086504} 100

24. The compound according to any one of claims 1-12, wherein R3 is substituted

25. The compound of claim 24, wherem R3 is ethylaminocarbonyl.

26. The compound according to any one of claims 1 -12, wherein R3 is alkoxy(c<s

27. The compound of claim 16, wherein R3 is methoxy, ethoxy, isopropoxy, feri-butoxy, or -O-cyclohexyl.

28. The compound according to any one of claims 1-12, wherein ¾ is aryloxy css)-

29. The compound of claim 28, wherein R j is -O-phenyl.

30. The compound according to any one of claims 1-12, wherein R¾ is aralkoxy(c<s)-

31. The compound of claim 30, wherein R3 is benzyloxy.

32. The compound according to any one of claims 1 - 12, wherem R¾ is substituted

33. The compound of claim 32, wherein R-, is ---OC(0)NHCH2C3¾.

34. The compound according to any one of claims 1 -12, wherein R3 is heterocyc ioaikoxy(c<8) .

35. The compound of claim 34, wherem R3 is -O-piperidinyl or N-Boe-piperidinyloxy.

36. The compound according to any one of claims 1-12, wherein R3 is alkylammO(c<s).

37. The compound of claim 36, wherein R3 is methylamino, ethylamino, isopropylamino, feri-butylamino or cyclohexylamino.

38. The compound according to any one of claims 1 - 12, wherem R¾ is substituted all y lamin (e<x).

39. The compound of claim 38, wherem 3 is 2,2,2 -trifluoroethylamino, - HCH2C(0)OCH3 or Ni i( i i >( ( {)){)! i.

40. The compound according io any one of claims 1-12, wherein R3 is dialky lamino(c<8).

41. The compound of claim 40, wherein ¾ is dimethylamino.

42. The compound according to any one of claims 1 -12, wherein R;, is alkoxyaminO(c<8) 43. The compound of claim 42, wherein R? is methoxyamino. 44. The compound according to any one of claims 1-12, wherein R/¾ is arylaminO(c<8). 45. The compound of claim 44, wherein R3 is phenylammo. 46. The compound according to any one of claims 1-12, wherein Rj is aralkylaminO(c<8j 47. The compound of claim 46, wherein R3 is benzylamino. 48. The compound according to any one of claims 1- 12, wherein ¾ heteroaryiaminO(c<8

49. The compound of claim 48, wherein ¾ is pyridinylamino. 50. The compound according to any one of claims 1 -12, wherein R3 is heterocyc alkylaminO(c<s i.

51. The compound of claim 50, wherein Rj is oxetanyj amino. 52. The compound according to any one of claims 1-12, wherein Rj is ~ H~amido(c<8). 53. The compound of claim 52, wherein R j is -NI-INHC(0)CH3. 54. The

{00086504} 102

or a pharmaceutically acceptable salt thereof.

wherein:

n is 1 to 6;

Ri and R2 are each independently -H, -OH, methyl, or as defined below; and ¾ is:

amino or -NHOH; or

N-heEeroaryl(c<s); N-heterocycloalkyl(c<8), alkylamino(c<8), dia1kylamino(c<8), alkenylamino(c g), alkoxyamino(c<8), aryiaminO(c 8), aralkylaminO(c<8j, heteroarylaminO(c<8j, heterocycloaikylamino(c<8j, alkylsulfonylamino(c<s), amido(c<s), -NH-amidO(c<8), or a substituted version of any of these groups;

R.3 and R|, taken together, are -NRa-, wherein Ra is hydrogen or alkyl(c<4); or R.3 and R¾ taken together, are --NRa-, wherein Ra is hydrogen or alkyl(c<4); or

or a pharmaceutically acceptable salt thereof.

The compound of claim 55, further defined as

wherein:

n is 1 to 6; and

amino or --NHOH; or

N-heterocycloalkyl<c<s), alky]ammO(c<s), dialkylammO(c 8), alkenylaminO(c 8), a1koxyaminO(c<8), ary{aniinO(c<8), aralkylamino<c<s), heteroaiylamino<c<s), heterocycloalkylarainO(c<8), alkyisulfonylaminO(c<s), amido(c<8>, -NH-amidO(c<8), or a substituted version of any of these groups:

or a pharmaceutically acceptable salt thereof.

57. The compound according to eiiher one of claim 55 and 56, wherein n is 1.

58. The compound according to either one of claim 55 and 56, wherein n is 2.

59. The compound according to either one of claim 55 and 56, wherein n is 3. 60. The compound according to any one of claims 55 and 57-59, wherem R-, is -H.

{00086504} 107

61. The compound according to any one of claims 55 and 57-59, wherein R2 is methyl.

62. The compound according to any one of claims 55-61, wherein R3 is N~heteroaryl(c<8

63. The compound of claim 62, wherein R? is imidazolyl.

64. The compound according to any one of claims 55-61, wherein R; is N-heterocycloalkyl<c<s)-

65. The compound of claim 64, wherein R j is morpholinyl or pyrroiidinyl.

66. The compound according to any one of claims 55-61 , wherein R3 is alk lamino(c<8).

67. The compound of claim 66, wherein R3 is ethylamino.

68. The compound of claim 55, further defined as:

or a pharmaceutically acceptable salt thereof.

A pharmaceutical composition comprising: a) the compound according to any one of claims 1 -68; and b) an excipient.

A method of treating and/or preventing a disease or a disorder in a patient in need thereof comprising admimstering to the paiient a compound according to any one of claims 1-68 in an amount sufficient to treat and/or prevent the disease or disorder.

Description:
DESCRIPTION

C17-ALKANEDIYL AND AL ENEDIYL DERIVATIVES OF OLEANOL1C ACID

AND METHODS OF USE THEREOF BACKGROUND OF THE INVENTION

This application claims the benefit of both United States Provisional Patent

Application No. 61/699, 122, filed on September 10, 2012 and United States Provisional Patent Application No. 61/780,540, filed on March 13, 2013, the entirety of both are incorporated herein by reference.

Pursuant to 37 C.F.R, 1.821(c), a sequence listing is submitted herewith as an ASCII compliant text file named "REATP0074US SequenceListing ST25.txt", created on September 5, 2013 and having a size of ~1 KB. The content of the aforementioned file is hereby incorporated by reference in its entirety.

I. Field of the In vention

The present invention relates generally to the fields of biology and medicine. More particularly, it concerns compounds, compositions and methods for the treatment and prevention of diseases such as those associated with oxidative stress and inflammation.

IL Description of Related Art

The anti-inflammatory and anti-proliferative activity of the naturally occurring triterpenoid, oieanolic acid, has been improved by chemical modifications. For example, 2- cyano-3,12-diooxooleana-l,9(l l)-dien-28-oic acid (CDDO) and related compounds have been developed (Honda et al, 1997; Honda et al, 1998; Honda et al, 1999; Honda et al, 2000a; Honda et al, 2000b; Honda, et al, 2002; Suh et al 1998; Suh et al, 1999; Place et al, 2003; Liby et al, 2005; and U.S. Patents 8,129,429; 7,915,402; 8,124,799; 8,071,632; 8,338,618; and 7,943,778). The methyl ester, bardoxolone methyl (CDDO-Me), has been evaluated clinically for the treatment of cancer and chronic kidney disease (Pergola et al, 201 1 ; Hong et al, 2012).

Synthetic triterpenoid analogs of oieanolic acid have also been shown to be inhibitors of cellular inflammatory processes, such as the induction by IFN-γ of inducible nitric oxide synthase (iNOS) and of COX-2 in mouse macrophages. See Honda et al. (2000a); Honda et al (2000b), and Honda et al. (2002). Synthetic derivatives of another triterpenoid, betulinic acid, have also been shown to inhibit cellular inflammatory processes, although these

{00086504} j compounds have been less extensively characterized (Honda et al, 2006). The pharmacology of these synthetic triterpenoid molecules is complex. Compounds derived from oleanolic acid have been shown to affect the function of multiple protein targets and thereby modulate the activity of several important cellular signaling pathways related to oxidative stress, cell cycle control, and inflammation (e.g., Dinkova-Kostova et al, 2005; Ahmad et al, 2006; Ahmad et al, 2008; Liby et al, 2007a). Derivatives of betulinic acid, though they have shown comparable anti-inflammatory properties, also appear to have significant differences in their pharmacology compared to OA-derived compounds (Liby et al, 2007b). Given that the biological activity profiles of known triterpenoid derivatives vary, and in view of the wide variety of diseases that may be treated or prevented with compounds having potent antioxidant and anti-inflammatory effects, and the high degree of unmet medical need represented within this variety of diseases, it is desirable to synthesize new compounds with diverse structures that may have improved biological activity profiles for the treatment of one or more indications.

SUMMARY OF THE INVENTION

The present disclosure provides novel synthetic triterpenoid derivatives, with antiinflammatory and/or antioxidant properties, pharmaceutical compositions, and methods for their manufacture, and methods for their use.

In one aspect, there are provided compounds of the formula:

wherein:

Y is a covalent bond, -CH 2 - -C(0}-, -Ό-, or Ni l .

{00086504} 2 Ri and R 2 are each independently -Ή, -OH, methyl, or as defined below; and R 3 is:

hydrogen, hydroxy, halo, amino, -NHOH, or mercapto;

alkyl(c s); alkenyl(c<8 alkynyl c<8), aryl(c<s), heteroaryl ( c<s), heterocycloalkyl(c<8 >, acyl(c<8 alkoxv(c<8 >, alkenyioxy(c<8 >, aiyloxy(c<s), aralkoxyross), heteroaryioxyvc<g), acyioxyvc<g), heterocycioalkoxy(c 8), alkylaminO(cv8), dialkylammO(c<8), alkenylamino ( c<8), alkoxyaminO(cv8), arylaminO(cv8) ? a alkylamino ( c<8), heteroaiylaminO(Ci8), heterocycl Q alkylammO(c<8), alkylsulfonylaminO(c<s), amidols), -NH-amidO(c<8), or a substituted version of any of these groups;

Rj and Rj, taken together, are -0-, -NR a - or a covalent bond between Y and carbon atom 13, wherein R a is hydrogen or a1kyl ( c<4); or

¾ and R 2j taken together, are -Q-, -NR a - or a covalent bond between Y and carbon atom 14, wherein R a is hydrogen or alkyl(c≤ ); harmaceutically acceptable salt thereof.

In some embodiments, the compounds are further defined as:

'herein:

Y is -Cl¾--, (·(() > . -0-, or -NH-;

Ri and R 2 are each independently -H, -OH, methyl, or as defined below

R; is: hydrogen, hydroxy, halo, amino, -NHOH, or mercapio;

alkyl(c<8), a]kenyl(c< 8 ), aik> nvi, , . ·. ,. aryl(c 8 ), aralkyl(c< 8 ), heteroaryi<c<s), heterocycloalkyl(c<8 ), acyl(c< 8 ), alkoxy(c< 8 >, alkenyioxy(c< 8 ), aiyloxv(c<8), araJkoxy(c< 8 ), heteroaryloxy ( c<s), acyloxy ( c≤s), heterocycloalkoxy(c≤8), alkylarnino<c<8), dialkylaminO(c<8), aikenyl amino- c<s), alkoxyaminO(c<s), arylarflino<c<s)> aralky1amino ( c<s), heteroarylaminO(c<8), heterocycloalkylamino(e<8 ), alkylsulfonylaminO(c<8), amidO(c<8>, _ NH-amidO(c< 8 ), or a substituted version of any of these groups;

¾ and R i , taken together, are ~ 0 ~ , -NR a - or a eovalent bond between Y and carbon atom 13, wherein R 3 is hydrogen or alkyl(c<4 ) 5 or

R3 and R.2, taken together, are -0-, -NR a - or a covaleiit bond between Y and carbon atom 14, wherein R a is hydrogen or alkyl(c<4);

or a pharmaceutically acceptable salt thereof.

In

wherein:

Y is a covalent bond, -CH?-, -C(O)-, -0-, or -NH-; and

R-3 is:

hydrogen, hydroxy, halo, amino, -NHOH, or mercapto; or

alkyl(c<8>, alkynyl(c<8), aryl(c<s>, aralkyi ( c<s), heteroaryl(c< 8 ), heterocycloalkyl(c s), acyl ( c 8), alkoxy(c 8 ), alkenyloxy ( c s),

{00086504} 4 aryioxv(c<8), a.raikoxy ( c<8), heteroafyloxy ( c<8), acyloxy(c<8), heteiOcycloalkoxy t c s >, alkylainino<c<s,>< dialkylamino(c<x), alkenylaminO(c s), alkoxyamino<c<8.,, arylamino,c<Sj, aralkylamino<e<S), heiefoarylaminO(c<s , i, hetei eycioaikyla:mmO(c<8), alkylsulfonylaniino(c<8), amidols), -NH---amido.;c<s), or a substituted version of any of these groups;

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compounds are further defined as:

wherein:

Y is CU . . ( ·(() > . -0-, or \ H : and

R 3 is:

hydrogen, hydroxy, halo, amino, -NHOH, or mercapto; or

alkyl (C S ), a1keny (c<8), alkynyl<c<s) > ar ' (c<8), ara Iky I c<8), heteroand-cssi, heterocycloalkyl(c 8 >, acyl c<s >, a lkoxy (C < 8 a ikenyloxy.;c<8 >, aryioxv(c<8), araikoxy(e 8), heteiOaryloxy<c<S), acyloxy ( c<s), heterocycloalkoxy(c≤8), alkylamino<c<s), dialkyla:mino(c<8), alkeny1aminO(c<8), alkoxyaminO(c<8), arylaraino<c<s)> aralkyJaraino<c<s)> heteroarylaminoicss), eterocycloalkylaniino(c<8 >, alkylsulfonylaniino(c<8), amido(c<8), -NH-amidO(C<8), or a substituted version of any of these groups;

or a pharmaceutically acceptable salt thereof.

{00086504} 5 In some embodiments, the bond between carbon atoms a and b is a single bond. In some embodiments, the bond between carbon atoms a and b is a double bond. In some embodiments, Y is a covalerit bond. In some embodiments, Y is -CH 2 -. In some embodiments, Y is -C(0)-. In some embodiments, Y is -0-. In some embodiments, Rj is -Ή. In some embodiments, R 2 is methyl.

In some embodiments, 3 is -H. In some embodiments, R3 is -OH. In some embodiments, R3 is amino. In some embodiments, R3 is alkyi ( c< 8) , for example methyl. In some embodiments, R3 is heterocycloalkyl(c<8), for example, morpholinyl, pyrrolidinyl, azetidinyl or piperazinyl. In some embodiments, R3 is substituted heterocycloaIkyl(c<8), for example, hydroxypyrrolidinyl, difluoropyrrolidinyl, hydroxypiperidinyl, or N-Boc- piperaziny]. In some embodiments, R3 is acyl ( c<s ) , for example, acetyl. In some embodiments, R3 is substituted acyl(c< ), for example, ethylaminocarbonyl. In some embodiments, R 3 is alkoxyvcss ) , for example, methoxy, ethoxy, isopropoxy, teri-butoxy, or -O-cyclohexyl. In some embodiments, R3 is aryloxy ( c<s), for example, -0-pheny3. In some embodiments, Rj is aralkoxy(c<8), for example, benzyloxy. In some embodiments, Rj is substituted aeyloxy ( c<s), for example, -OC(0)NHCH 2 CH3. In some embodiments, R3 is heterocycJoalkoxy(c<8), for example, -O-piperidinyl or N-Boc-piperidinyloxy. In some embodiments, R3 is alkylamino(c<g), for example, methylamino, ethylamino, isopropyJamino, ie/'/-buiylamino or cyclohexylamino. In some embodiments, R3 is substituted alkylamino(c 8), for example, 2,2,2-trifluoroethylamino, -NHCH 2 C(0)OCH3 or -NHCH 2 C(0)OH. In some embodiments, R3 is dialkylamino(c<s), for example, dimethylamino. In some embodiments, R3 is alkoxyamirtO(c<8), for example, methoxyamino. In some embodiments, R3 is arylamino(c 8), fo example, phenylam no. In some embodiments, R3 is aralkylamino(c<s), fo example, benzylamino. In some embodiments, R3 is heteroaiylamino,c<8), for example, pyrid nylamino. In some embodiments, R3 is h.eterocycIoalkylamino(c<8), for example, oxetanylamino. In some embodiments, R3 is -NH-amidO(c<8), for example, -NHNHC(0)C3¾.

{00086504}

{00086504} 7

{00086504} 9

or a pharmaceutically acceptable salts of any of these formulas.

In one aspect, there are pro vided compounds of the formula :

wherein:

Ώ is 1 to 6;

Ri and R2 are each independently -H, -OH, methyl, or as defined below; and R 3 is:

amino or -NHOH; or

N-heteroaryl(c 8), N-heterocycloalkyI c<s), alkylamino ( c<s), diaikylamino ( c<s), aikenylaminO(c<8), alkoxyaminO(c<s), arylaminO(c<Sj, aralkylaminO(c<8j, heteroarylamino(c<s), heterocycloalky1amino(c<8),

{00086504} 10 aikylsuifonylaminO(c<8), midols), -NH-amidO(c<s), or a substituted version of any of these groups;

R.3 and Rj, taken together, are -NR S -, wherein R a is hydrogen or or Rj and R?, taken together, are -NR a -, wherein R a is hydrogen or alkyl ( c<4); or or a pharmaceutically acceptable salt thereof.

In some embodiments, the compounds are further defined as:

wherein:

n is 1 to 6; and

¾ is:

amino or -NHOH; or

N-heteroaryl(c<s), N-heterocycloalkyl^a), alkylamin0(c<8j, diaUylamin0(c<8), afkeny]aminO(C<8), alkoxyamino(cv8), aiylamino(Ci8), aralky]amino ( c<8), heteroaiylamino(Ci8), lieterocycfoaikylam no(c<8), afkylsuffonylamin (c<8), amidO(c< 8 ), -NH-amidO(c< 8 ), o a substituted version of any of these groups;

or a pharmaceutically acceptable salt thereof.

In some embodiments, n is 1. In other embodiments, n is 2. In still other embodiments, n is 3.

In some embodiments, R; is -H. In some embodiments, R2 is methyl.

In some embodiments, R 3 is N-heteroaryi ( c 8), for example, imidazolyl. In some embodiments, R3 is N-heterocycloalkyl c<8), for example, morpholinyl or pyrrolidinyl. In some embodiments, R 3 is alkylaminO ( c 8), for example, eihyiamino.

{00086504} I I

or a pharmaceutically acceptable salt thereof.

In some aspects, there are provided pharmaceutical compositions comprising one or more of the above compounds and an excipient. In other aspects there are provided methods of treating and/or preventing a disease or a disorder in patients in need thereof, comprising administering to such patients one or more of the above compounds in an amount sufficient to treat and or prevent the disease or disorder.

Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in ihe art from this detailed description. Note that simply because a particular compound is ascribed to one particular generic formula doesn't mean that it cannot also belong to another generic formula.

{00086504} 12 ENTS

Disclosed herein are new compounds and compositions with antioxidant and'Or antiinflammatory properties, methods for their manufacture, and methods for their use, including for the treatment and/or prevention of disease. L Definitions

When used in the context of a chemical group: "hydrogen" means -H; "hydroxy" means -OH; "oxo" means =0; "carbonyl" means -C( ;; O) -; "carboxy" means -C(-0)OH (also written as -COOH or -C0 2 H); "halo" means independently -F, -CI, -Br or -I; "amino" means -NH 2 ; "hydroxyamino" means -NHOH; "nitro" means - O2; imino means =NH; "cyano" means -CN; "isocyanate" means -N=C=0; "azido" means -N3; a monovalent context "phosphate" means -OP(0)(OH) 2 or a deprotonated form thereof; in a divalent context "phosphate" means -QP(Q)(QH)Q- or a deprotonated form thereof; "mercapto" means -SH; and "thio" means =S; "sulfonyl" means -S(0) 2 -; and "sulflnyl" means -S(O)-.

In the context of chemical formulas, the symbol "-" means a single bond, "=" means a double bond, and ' =" means triple bond. The symbol " " represents an optional bond, which if present is either single or double. The symbol "==" represents a single bond or a double bond. Thus, for example, the formula

^). And it is understood that no one such ring atom forms part of more than one double bond. Furthermore, it is noted that the covafent bond symbol when connecting one or two stereogenic atoms, does not indicate any preferred stereochemistry. Instead, it cover ail stereoisomers as well as mixtures thereof. The symbol " <ΛΛΛ n t when drawn perpendicularly across a bond (e.g " .,— CH 3 for methyl) indicates a point of attachment of the group. It is noted that the point of attachment is typically only identified in this manner for larger groups in order to assist the reader in unambiguously identifying a point of attachment. The symbol " ·" ■■ ■ " means a single bond where the group attached to the thick end of the wedge is "out of the page." The symbol " "•Hi " means a single bond where the group attached to the thick end of the wedge is "into the page". The symbol "*ΛΛΛ " means a single bond where the geometry around a double bond (e.g., either E or Z) is undefined. Both options, as well as combinations thereof are therefore intended. The bond orders described above are not

{00086504} 13 limiting when one of the atoms connected by the bond is a metal atom (M). In such cases, it is understood that the actual bonding may comprise significant multiple bonding and/or ionic character. Therefore, unless indicated otherwise, the formulas M-C, M=C, M C, and M

= =(?,, each refers to a bond of any and type and order between a metal atom and a carbon atom. Any undefined valency on an atom of a structure shown in this application implicitly represents a hydrogen atom bonded to that atom. A bold dot on a carbon atom indicates ihai the hydrogen attached to that carbon is ori ented out of the plane of the paper.

When a group "R" is depicted as a "floating group" on a ring system, for example, in the formula: then R may replace any hydrogen atom attached to any of the ring atoms, including a depicted, implied, or expressly defined hydrogen, so long as a stable structure is formed. When a group "R" is depicted as a "floating group" on a fused ring system, as for example in the formula:

then R may replace any hydrogen attached to any of the ring atoms of either of the fused rings unless specified otherwise. Replaceable hydrogens include depicted hydrogens (e.g., the hydrogen attached to the nitrogen in the formula above), implied hydrogens (e.g. , a hydrogen of the formula above that is not shown but understood to be present), expressly defined hydrogens, and optional hydrogens whose presence depends on the identity of a ring atom (e.g., a hydrogen attached to group X, when X equals -CH-), so long as a stable structure is formed, in the example depicted, R may reside on either the 5-membered or the 6- membered ring of the fused ring system. In the formula above, the subscript letter "y" immediately following the group "R" enclosed in parentheses, represents a numeric variable. Unless specified otherwise, this variable can be 0, 1 , 2, or any integer greater than 2, only limited by the maximum number of replaceable hydrogen atoms of the ring or ring system.

For the groups and classes below, the following parenthetical subscripts further define the group/class as follows: "(Cn)" defines the exact number (n) of carbon atoms in the group/class. "(C<n)" defines the maximum number (n) of carbon atoms that can be in the

{00086504} 14 group/class, with the mmimum number as small as possible for the group in question, e.g. , it is understood that the minimum number of carbon atoms in the group "alkenyl<c<s)" or the class "alkene ( c<8)" s two. For example, "alkoxy(c≤io)" designates those alkoxy groups having from 1 to 10 carbon atoms. (Cn-n') defines both the minimum (n) and maximum number (η') of carbon atoms in the group. Similarly, "aikyl(C2 -iO)" designates those alky! groups having from 2 to 10 carbon atoms.

The term "saturated" as used herein means the compound or group so modified has no carbon-carbon double and no carbon-carbon triple bonds, except as noted below. In ihe ease of substituted versions of saturated groups, one or more carbon oxygen double bond or a carbon nitrogen double bond may be present. And when such a bond is present, then carbon- carbon double bonds that may occur as part of keto-enol tautomerisni or imine/enamine tautomerism are not precluded.

The term "aliphatic" when used without the "substituted" modifier signifies that the compound/group so modified is an acyclic or cyclic, but non-aromatic hydrocarbon compound or group. In aliphatic compounds/groups, ihe carbon aioms can be joined together in straight chains, branched chains, or non-aromatic rings (alicyclic). Aliphatic compounds/groups can be saturated, that is joined by single bonds (alkanes/alkyl), or unsaturated, with one or more double bonds (alkenes/alkenyl) or with one or more triple bonds (alkynes/alkynyl).

The term "alky!" when used without the "substituted" modifier refers to a monovalent saturated aliphatic group with a carbon atom as the point of attachment, a linear or branched, cycle, cyclic or acyclic structure, and no atoms other than carbon and hydrogen. Thus, as used herein cycloalkyl. is a subset of alkyl, with the carbon atom that forms the point of attachment also being a member of one or more non-aromatic ring structures wherein the cycloalkyl group consists of no atoms other than carbon and hydrogen. As used herein, the term does not preclude the presence of one or more alkyl groups (carbon number limitation permitting) attached to the ring or ring system. The groups -C¾ (Me), -CH2CH3 (Et), -CI-I2CH2CH3 (n-Pr or propyl), ( I li Cl h i . (ι-Pr, Ps or isopropyl), -CH(CH 2 )2 (cyclopropyl), C ' l i C ' i (>( 1 1 ·■( ! ! : (Λ-BU), C ! l( Ci O ! (sec-butyl), ( Ί i >( 1 li C ' i ! : !,· (isobutyl), -C(CHj) ¾ (teri- butyl, £- butyl, i-Bu or ¾u), -CH 2 C(CHj) ¾ (neo-pentyl), cyclobutyl, cyclopentyl, cyclohexyl, and cyclohexylmethyl are non-limiting examples of alkyl groups. The term "alkanediyl" when used without the "substituted" modifier refers to a divalent saturated aliphatic group, with one or two saturated carbon atom(s) as the point(s) of

{00086504} attachment, a linear or branched, cyclo, cyclic or acyclic structure, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen. s, --de ¬

(methylene), -CH 2 CH 2 - -CH 2 C(CH 3 ) 2 CH 2 - -CH 2 CH 2 CH 2 - and are non- limiting examples of alkanediyl groups. The term "alkylidene" when used without the "substituted" modifier refers to the divalent group ;;; CRR' in which R and R' are independently hydrogen, aikyl, or R and R' are taken together to represent an alkanediyl having at least two carbon atoms. Non-limiting examples of alkylidene groups include: =€H 2 , =CH(CH 2 CH3), and = Ο¾)?, An "alkane" refers to the compound H-R, wherein R is aiky l as this term is defined above. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2) -N0 2 , -C0 2 H, ( ( ) ·( ·! i ;. -CN, -SH, -OCH 3 , -OCH 2 CH 3 , -C(0)CH 3 , ~ NHCH 3 , -NHCH 2 CH 3 , -N(CH 3 ) ¾ -C(0)NH ¾ -OC(0)CH 3 , or - ~ 8(0) 2 NH 2 . The following groups are non-limiting examples of substituted aikyl groups: -CH 2 OH, -CH 2 C1, -CF 3 , -CH 2 CN, -CH 2 C(0)OH, -CH 2 C(0)OCH 3 , -CH 2 C(0)NH 2 , -CH 2 C(0)CH 3 , -CH 2 OCH 3 , -CH 2 OC(0)CH 3 , -CH 2 NH 2 , -CH 2 N(CH 3 ) 2 , and -CH 2 CH 2 C1. The term "haloalkyl" is a subset of substituted aikyl, in which one or more hydrogen atoms has been substituted with a halo group and no other atoms aside from carbon, hydrogen and halogen are present. The group, -CH 2 C1 is a non-limiting example of a haloalkyl. The term "fluoroalkyl" is a subset of substituted aikyl, in which one or more hydrogen has been substituted with a fluoro group and no other atoms aside from carbon, hydrogen and fluorine are present. The groups, -CH 2 F, -CF 3 , and -CH 2 CF 3 are non-limiting examples of fluoroalkyl groups.

The term "alkenyl" when used without the "substituted" modifier refers to an monovalent unsaturated aliphatic group with a carbon atom as the point of attachment, a linear or branched, cyclo, cyclic or acyclic structure, at least one nonaromatic carbon-carbon double bond, no carbon-carbon triple bonds, and no atoms other than carbon and hydrogen. Non-limiting examples of alkenyl groups include: -CH= H 2 (vinyl), -CH-CHCH 3 , -CH=CHCH 2 CH 3 , ί Ι ΚΉ < I l > fallyl), ( Π >< 1 ! Ci lCl . and CU Ci lCU C ! k The term "alkenediyl" when used without the "substituted" modifier refers to a divalent unsaturated aliphatic group, with two carbon atoms as points of attachment, a linear or branched, cyclo, cyclic or acyclic structure, at least one nonaromatic carbon-carbon double bond, no carbon-carbon triple bonds, and no atoms other than carbon and hydrogen. The

groups, i l l CU . , are non-limiting

{00086504} 16 examples of alkenediyl groups. It is noted that while the alkenediyl group is aliphatic, once connected at both ends, this group is not precluded from forming part of an aromatic structure. The terms "alkene" or "olefin" are synonymous and refer to a compound having the formula H-R, wherein R is aJkenyl as this term is defined above. A "terminal alkene" refers to an alkene having just one carbon-carbon double bond, wherein that bond forms a vinyl group at one end of the molecule. When any of these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, Ni l ,. -NO ? , -C0 2 H, ( () ,( 1 1 = . -CN, SH. -OCH 3 , OCU >( ' ! ! ;. ( ( { ))( ! ! : . -NHCH3, -NHCH2CH3, -N(CH 3 ) 2 , -C(0)NH 2 , -OC(0)CH 3 , or --S(0) 2 N1¾. The groups, -CH=CHF, -CH=CHC1 and -CH=CHBr, are non-limiting examples of substituted alkenyf groups.

The term "alkynyi" when used without the "substituted" modifier refers to an monovalent unsaturated aliphatic group with a carbon atom as the point of attachment, a linear or branched, cyclo, cyclic or acyclic structure, at least one carbon-carbon triple bond, and no atoms other than carbon and hydrogen. As used herein, the term alkynyi does not preclude ihe presence of one or more non-aromaiic carbon-carbon double bonds. The groups, -C=CH, -O CH 3 , and -CH 2 0≡CCH 3 , are non-limiting examples of alkynyi groups. An "alkyne" refers to the compound H-R, wherein R is alkynyi. When any of these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, - H 2 , -N0 2 , -C0 2 H, -C0 2 CH 3 , -CN, -SH, 0( 1 1 :. -OCH 2 C¾, -C(0)CH 3 , -NHCH.3, -NHCH 2 CH 3 , -N(CH 3 ) 2 , -C(0)NH 2 , -OC(0)CH 3 , or -S(0) 2 NH 2 .

The term "aryl" when used without the "substituted" modifier refers to a monovalent unsaturated aromatic group with an aromatic carbon atom as the point of attachment, said carbon atom forming part of a one or more six-membered aromatic ring structure, wherein the ring atoms are all carbon, and wherein the group consists of no atoms other than carbon and hydrogen. If more than one ring is present, the rings may be fused or unfused. As used herein, the term does not preclude the presence of one or more alky! or aralkyl groups (carbon number limitation permitting) attached to the first aromatic ring or any additional aromatic ring present. Non-limiting examples of aryl groups include phenyl (Ph), methylphenyi, (dimethyl)phenyl, -C 6 H4CH2CH 3 (ethylphenyi), naphthyi, and a monovalent group derived from biphenyl. The term "arenediyl" when used without the "substituted" modifier refers to a divalent aromatic group with two aromatic carbon atoms as points of attachment, said carbon atoms forming part of one or more six-membered aromatic ring

{00086504} | 7 structure(s) wherein the ring atoms are all carbon, and wherein the monovalent group consists of no atoms other than carbon and hydrogen. As used herein, the term does not preclude ihe presence of one or more alkyl, aryl or aralkyi groups (carbon number limitation permitting) attached to the first aromatic ring or any additional aromatic ring present. If more than one ring is present, the rings may be fused or unfused. Unfused rings may be connected via one or more of the following: a covaient bond, alkanediyl, or alkenediyl groups (carbon number limitation p

An "arene" refers to ihe compound H-R, wherein R is aryl as that term is defined above. Benzene and toluene are non-limiting examples of arenes. When any of these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -Ci, -Br, -I, N i b. -N0 2 , CO -H. ( ) >( ! h. -CN, SI I. (ΧΊ 1 .. -OCH2CH3, -C(0)CH 3 , N i iCi i .. -NHCH2CH3, -N(CH 3 ) 2 , -C(0)NH 2 , -OC(0)CH 3 , or -S(0) 2 NH 2 .

The term "aralkyi" when used without the "substituted" modifier refers to the monovalent group -alkanediyl-aryl, in which ihe terms alkanediyl and aryl are each used in a manner consistent with the definitions provided above. Non-limiting examples of aralkyls are: phenyimethyl (benzyl, Bn) and 2-phenyl-ethyl. When the term aralkyi is used with the "substituted" modifier one or more hydrogen atom from the alkanediyl and/or the aryl group has been independently replaced by -OH, -F, -CI, -Br, -1, Ni k -N0 2 , CO.-U . -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(0)CH 3 , -NHCH3, -NHCH2CH3, -N(CH 3 )¾ -C(0)N¾, - C(0)CHj, or -S(0) 2 NH ? , Non-limiting examples of substituted aralkyls are: (3-chloropheny l)-rneihyi, and 2-chloro-2-phenyl-eth- 1 -yl.

The term "heteroaryi" when used without the "substituted" modifier refers to a monovalent aromatic group with an aromatic carbon atom or nitrogen atom as the point of attachment, said carbon atom or nitrogen atom forming part of one or more aromatic ring structures wherein at least one of the ring atoms is nitrogen, oxygen or sulfur, and wherein the heteroaryi group consists of no atoms other than carbon, hydrogen, aromatic nitrogen, aromatic oxygen and aromatic sulfur. If more than one ring is present, the rings may be fused

{00086504} 18 or unfused. As used herein, the term does not preclude the presence of one or more alkyl, aryl, and/or aralkyl groups (carbon number limitation permitting) attached to the aromatic ring or aromatic ring system. Non-limiting examples of heteroaryl groups include furanyl, imidazoiyl, indofyl, indazofyl (Im), isoxazolyl, methylpyridinyl, oxazolyl, pheiiylpyiidinyl, pyridinyi, pyrrolyl, pyrimidinyl, pyrazinyl, qumolyl, quinazolyl, quinoxalinyl, triazinyl, tetrazolyl, thiazolyl, thienyi, and triazoiyi. The term "N-heteroaryl" refers to a heteroaryl group with a nitrogen atom as the point of attachment. The term "heteroarenediyl" when used without the "substituted" modifier refers to an divalent aromatic group, with two aromatic carbon atoms, two aromatic nitrogen atoms, or one aromatic carbon atom and one aromatic nitrogen atom as the two points of attachment, said atoms forming part of one or more aromatic ring structure(s) wherein at least one of the ring atoms is nitrogen, oxygen or sulfur, and wherein the divalent group consists of no atoms other than carbon, hydrogen, aromatic nitrogen, aromatic oxygen and aromatic sulfur. If more than one ring is present, the rings may be fused or unfused. Unfused rings may be connected via one or more of the following: a covalent bond, alkanediyi, or alkenediyi groups (carbon number limitation permitting). As used herein, the term does not preclude the presence of one or more alkyl, aryl, and/or aralkyl groups (carbon number {imitation permitting) attached to the aromatic ring or aromatic ring syste -limiting examples of heteroarenediyl groups include:

A "heteroarene" refers to the compound H-R, wherein R is heteroaryl. Pyridine and quinoline are non-limiting examples of heteroarenes. When these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH,

-F, -CI, -Br, -I, XH >. -N0 2 , -C0 2 H, ( >Π ! .. -CN, -SH, OCU .. -OCH 2 CH 3 -C(0)CH 3 , -NHCH3, -NHCH2CH3, -N(CH 3 ) 2 , -C(0)NH 2 , -OC(0)CH 3 , or -S(0) 2 NH 2 .

The term "heterocycloalkyl" when used without the "substituted" modifier refers to a monovalent non-aromatic group with a carbon atom or nitrogen atom as the point of attachment, said carbon atom or nitrogen atom forming part of one or more non-aromatic ring structures wherem at least one of the ring atoms is nitrogen, oxygen or sulfur, and wherein the heterocycloalkyl group consists of no atoms other than carbon, hydrogen, nitrogen, oxygen and sulfur. If more than one ring is present, the rings may be fused or unfused. As used herein, the term does not preclude the presence of one or more alkyl groups (carbon number limitation permitting) attached to the ring or ring system. Also, the term does not preclude the presence of one or more double bonds in the ring or ring system, provided that the resulting group remains non-aromatic. N on- limiting examples of heterocycloalkyl groups include aziridinyi, azetidinyl, pyrroHdinyl, piperidinyl, piperazinyl, morphoHnyl, thiomorpholinyl, tetrahydrofuranyl, tetrabydrotbiofuranyl, tetrahydropyranyl, pyranyl, oxiranyl, and oxetanyi. The term "N-heterocycloalkyl" refers to a heterocycloalkyl group with a nitrogen atom as the point of attachment. The term "heterocycloalkanediyl" when used without the "substituted" modifier refers to an divalent cyclic group, with two carbon atoms, two nitrogen atoms, or one carbon atom and one nitrogen atom as the two points of attachment, said atoms forming part of one or more ring siructure(s) wherein at least one of the ring atoms is nitrogen, oxygen or sulfur, and wherein the divalent group consists of no atoms other than carbon, hydrogen, nitrogen, oxygen and sulfur. If more than one ring is present, the rings may be fused or unfused. Unfused rings may be connected via one or more of the follo wing: a cov alent bond, alkanediyl, or alkenedivl groups (carbon number limitation permitting). As used herein, the ter does not preclude the presence of one or more alky] groups (carbon number limitation permitting) attached to the ring or ring system. Also, the term does not preclude the presence of one or more double bonds in the ring or ring system, provided that the resulting group remains non-aromatic. Non-limiting examples of heterocycloalkanediyl groups include:

When these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -N0 2 , -C0 2 H, -CO2CH 3 , -CN, -SH, 0( 1 1 : . ( X i l -C ! i :. -C(0)CH 3 , --NHCH3, -NHCH 2 CH 3 , " -N(CH 3 ) 2 , -C(0)NH 2) -OC(0)CH 3 , -S(0) 2 N¾, or -C(0)OC(CH 3 ) 3 (teri-butyloxycarbonyl, HOC }.

The term "acyl" when used without the "substituted" modifier refers to the group -"C(0)R, in which R is a hydrogen, alkyl, aryi, aralkyl or heteroaryl, as those terms are defined above. The groups, -CHO, -C(Q)CH (acetyl, Ac), -C(0)CH 2 CH 3 , -C(0)CH 2 CH 2 CH 3 , -C(0)CH(CH 3 ) 2 , -C(0)CH(CH 2 ) 2 , -C(0)C6H 5 , C ' (( ) }( ' ,,i I X ' l ! . -C(0)CH 2 C6H5, -C(0)(imidazolyl) are non-limiting examples of acyl groups. A "thioacyl" is defined in an analogous manner, except that the oxygen atom of the group - ~ C(0)R has been replaced with a sulfur atom, -C(S)R. The term "aldehyde" corresponds to an alkane, as defined above, wherein at least one of the hydrogen atoms has been replaced with a -CHO group. When any of these terms are used with the "substituted" modifier one or more

{00086504} 20 hydrogen atom (including a hydrogen atom directly attached the carbonyl or thiocarbonyl group, if any) has been independently replaced by -OH, -F, -CI, -Br, -1, -NH 2 , -N0 2 , -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(0)CH 3 , -NHCH3, -NHCH2CH3, -N(CH 3 ) 2 , -C(0)NH 2 , -OC(0)CH 3 , or -S(0) 2 NH 2 . The groups, -C(0)CH 2 CF 3 , -C0 2 H (carboxyi), -CO2CH3 (methylcarboxyl), -CO2CH2CH3, -C(0)NH 2 (carbamoyl), and -CO (CH3 2, are non- limiting examples of substituted acyl groups.

The term "alkoxy" when used without the "substituted" modifier refers to the group -OR, in which R is an alkyl, as that term is defined above. Non-limiting examples of alkoxy groups include: OCH ; (methoxy), ( )( ! ! ·.( i f (ethoxy), ( )( ! f( i CU ; . ΟΠ U Ci ! ·, ) > (isopropoxy), -0(CH 3 )3 (ie/ -butoxy), -OCH(CH 2 ) 2 , -O-cyclopentyl, and -O-cyclohexyl. The terms "alkenyloxy", "alkynyloxy", "aryloxy", "aralkoxy", "heteroaryloxy", "heterocycloalkoxy", and "acyloxy", when used without the "substituted" modifier, refers to groups, defined as -OR, in which R is alkenyl, alkynyl, aryl, aralkyi, heteroaryl, heterocycloalkyl, and acyl, respectively. The term "alkoxydiyl" refers to the divalent group -O-alkanediyl-, -O-alkanediyl-O-, or -alkanediyi-O-alkanediyl-. The term "alkylthio" and "acylthio" when used without the "substituted" modifier refers to the group -SR, in which R is an alkyl and acyl, respectively. The term "alcohol" corresponds to an alkane, as defined above, wherein at feast one of the hydrogen atoms has been replaced with a hydroxy group. The term "ether" corresponds to an alkane, as defined above, wherein at least one of the hydrogen atoms has been replaced with an alkoxy group. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, F. ( 1. -Br, -Ί, - ~ NH 2 , -NO2, -C0 2 H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(0)CH 3 , -NI-ICH3, -NHCH2CH3, -N(CH 3 ) ¾ -C(0)NH 2 , -OC(0)CH 3 , or -S(0) 2 NH 2 .

The term "afkylamino" when used without the "substituted" modifier refers to the group -NHR, in which R is an alkyl, as that term is defined above. Non-limiting examples of alkylamino groups include: -NHCH 3 and -NHCH2CH 3 . The term "dialkylamino" when used without the "substituted" modifier refers to the group -NRR', in which R and R' can be the same or different alkyl groups, or R and R' can be taken together to represent an alkanediyl. Non-limiting examples of dialkylamino groups include: -Ν(0¾) 2 , -N(CH3)(CH 2 CH3), and N-pyrrolidinyl. The terms "alkoxyamino", "alkenylamino", "alkynyl amino", "aiylamino", "aralkylamino", "heteroarylamino", "heterocycloalkylamino" and "alk lsulfonylamino" when used without the "substituted" modifier, refers to groups, defined as -NHR, in which R is alkoxy, alkenyl, alkynyl, aryl, aralkyi, heteroaryl,

{00086504} T j heterocycloalkyl, and alkylsulfonyl, respectively. A non-limiting example of an arylamino group is --NHC 0 H 5 . The term "amido" (acylamino), when used without the "substituted" modifier, refers to the group -NHR, in which R is acyl, as that term is defined above. A non- limiting example of an amido group is -NHC(0)CH3. The term "alkylimino" when used without the "substituted" modifier refers to the divalent group -NR, in which R is an alkyi, as that term is defined above. The term "alkylaminodiyl" refers to the divalent group -NH-alkanediyl-, -NH-alkanediyl-NH-, or -alkanediyl-NH-alkanediyl-. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, (1. -Br, -I, -N¾, -N0 2 , -C0 2 H, ( ( ) .( ! ! :. -CN, -SH, -OCH3, -OCH2CH3, -C(0)CH 3 , -NHCH3, -NHCH2CH3, -N(CH 3 ) 2 , -C(0)NH 2 , -OC(0)CH 3 , or -S(0) 2 NH 2 . The groups -NHC(0)OCH3 and -NHC(0)NHCH 3 are non- limiting examples of substituted amido groups.

The terms "alkylsulfonyl" and "alkylsulfinyl" when used without the "substituted" modifier refers to the groups -S(0) 2 R and -S(0)R, respectively, in which R is an alkyi, as that term is defined above. The terms "alkenylsuifonyl", "alkynylsulfonyl", "arylsulfonyl", "araikylsuifonyl", "heteroarylsulfonyl", and "heterocycloalkylsulfonyl" are defined in an analogous manner. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO2, -C0 2 H, -CO CH 3 , -CN, -SH, -OCH 3 , -OCH 2 CH 3 , ( i OK ' i . -NHCH 3 , -NHCH 2 CH 3 , -N(CH 3 ) ¾ -C(0)NH 2 , OO O K ' M ;. or -S(0) 2 N¾.

The term "alkylphosphate" when used without the "substituted" modifier refers to the group -OP(0)(OH)(OR), in which R is an alkyi, as that term is defined above. Non-limiting examples of alkylphosphate groups include: -OP(0)(OH)(OMe) and -OP(0)(OH)(OEt). The term "dialkylphosphate" when used without the "substituted" modifier refers to the group -OP(0)(OR)(OR'), in which R and R' can be the same or different alkyi groups, or R and R' can be taken together to represent an alkanediyl. Non-limiting examples of dialkylphosphate groups include: -OP(0)(OMe) 2 , -OP(0)(OEt)(OMe) and -QP(Q)(QEt) 2 . When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -N0 2 , -C0 2 H, -C0 2 CH 3 , -CN, -SH, -OCH3, -OCH 2 CH 3 , O OK i h. -NPICH3, -NHCH2.CH3, N i ! : ! >. -C(0)NH 2 , )C ' i ( ))( ! l :. or -S(0) 2 NH 2 .

The use of the word "a" or "an," when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one,"

{00086504} 22 Throughout this application, the tenn "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.

As used herein, a "chiral auxiliary" refers to a removable chiral group that is capable of influencing the stereoselectivity of a reaction. Persons of skill in the art are familiar with such compounds, and many are commercially available.

The terms "comprise," "have" and "include" are open-ended linking verbs. Any forms or tenses of one or more of these verbs, such as "comprises," "comprising," "has," "having," "includes" and "including," are also open-ended. For example, any method that "comprises," "has" or "includes" one or more steps is not limited to possessing only those one or more steps and also covers other unlisted steps.

The term "effective," as that term is used in the specification and/or claims, means adequate to accomplish a desired, expected, or intended result. "Effective amount," "Therapeutically effective amount" or "pharmaceutically effective amount" when used in the context of treating a patient or subject with a compound means that amount of the compound which, when administered to a subject or patient for treating a disease, is sufficient to effect such treatment for the disease.

As used herein, the term "IC50" refers to an inhibitory dose which is 50% of the maximum response obtained. This quantitative measure indicates how much of a particular drug or other substance (inhibitor) is needed to inhibit a given biological, biochemical or chemical process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half.

An "isomer" of a first compound is a separate compound in which each molecule contains the same constituent atoms as the first compound, but where the configuration of those atoms in three dimensions differs.

As used herein, the term "patient" or "subject" refers to a living mammalian organism, such as a human, monkey, cow, sheep, goat, dog, eat, mouse, rat, guinea pig, or transgenic species thereof. n certain embodiments, the patient or subject is a primate. Non- limiting examples of human subjects are adults, juveniles, infants and fetuses.

As generally used herein "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.

{00086504} "Pharmaceutically acceptable salts" means salts of compounds of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as 1 ,2-emanedisulfonic acid, 2-liydroxyethanesulfonic acid, 2-naphthalenesuifonie acid, 3 -phenylpropionic acid, 4,4'-methylenebis(3-hydroxy-2-ene- 1 -carboxylic acid), 4-methylbicyclo[2.2.2]oct-2-ene- 1-carboxylic acid, acetic acid, aliphatic mono- and dicarboxylic acids, aliphatic sulfuric acids, aromatic sulfuric acids, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, carbonic acid, cinnamic acid, citric acid, cycJopentanepropionic acid, ethanesulfonic acid, fumarie acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, heptanoic acid, hexanoic acid, hydroxynaphthoic acid, lactic acid, laurylsulfuric acid, maleic acid, malic acid, maionic acid, mandelic acid, methanesulfonic acid, muconie acid, o-(4-hydroxybenzoyl)benzoic acid, oxalic acid, p-chJorobenzenesulfonic acid, phenyl-substituted alkanoic acids, propionic acid, / oluenesuifonic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, tartaric acid, tertiarybutylacetic acid, trimethylacetic acid, and the like. Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include ethanolamine, dieihanolamine, triethanolamine, tromethamine, N-methylglucamine and the like. It should be recognized that the particular anion or cation forming a part of any salt of this invention is not critical, so long as the salt, as a whole, is pharmacologically acceptable. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are presented in Handbook of Pharmaceutical Salts: Properties, and Use (P. H. Sta ! & C. G. Wermuth eds., Verlag Helvetica Chimica Acta, 2002).

The term "pharmaceutically acceptable carrier," as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent.

"Prevention" or "preventing" includes: (1) inhibiting the onset of a disease in a subject or patient which may be at risk and/or predisposed to the disease but does not yet experience or display any or all of the pathology or symptomatology of the disease, and/or (2) slowing the onset of the pathology or symptomatology of a disease in a subject or patient which may be at risk and/or predisposed to the disease but does not yet experience or display any or all of the pathology or symptomatology of the disease.

"Prodrug" means a compound that is convertible in vivo metabolically into an inhibitor according to the present invention. The prodrug itself may or may not also have activity with respect to a given target protein. For example, a compound comprising a hydroxy group may be administered as an ester (hat is converted by hydrolysis in vivo to the hydroxy compound. Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, phosphates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-P-hydroxynaphthoate, gentisates, isethionates, di-/ !oluoyltartrates, methanesulfonates, eihanesulfonates, benzenesulfonates, j P-toiuenesulfonates, eyciohexylsulfamates, quinates, esters of amino acids, and the like. Similarly, a compound comprising an amine group may be administered as an amide that is converted by hydrolysis in vivo to the amine compound.

A "stereoisomer" or "optical isomer" is an isomer of a given compound in which the same atoms are bonded to the same other atoms, but where the configuration of those atoms in three dimensions differs. "Enantiomers" are stereoisomers of a given compound that are mirror images of each other, like left and right hands. "Diastereomers" are stereoisomers of a given compound that are not enantiomers. Chiral molecules contain a chiral center, also referred to as a stereocenter or stereogenic center, which is any point, though not necessarily an atom, in a molecule bearing groups such that an interchanging of any two groups leads to a stereoisomer. In organic compounds, the chiral center is typically a carbon, phosphorus or sulfur atom, though it is also possible for other atoms to be stereocenters in organic and inorganic compounds. A molecule can have multiple stereocenters, giving it many stereoisomers. In compounds whose stereoisomerism is due to tetrahedral stereogenic centers (e.g., tetrahedral carbon), the total number of hypothetical!}' possible stereoisomers will not exceed 2n, where n is the number of tetrahedral stereocenters. Molecules with symmetry frequently have fewer than the maximum possible number of stereoisomers. A 50:50 mixture of enantiomers is referred to as a racemic mixture. Alternatively, a mixture of enantiomers can be enanttomerically enriched so that one enantiomer is present in an amount greater than 50%. Typically, enantiomers and/or diastereomers can be resolved or separated using techniques known in the art. It is contemplated that that for any stereocenter or axis of chiraiity for which stereochemistry has not been defined, that stereocenter or axis of chiraiiiy can be present in its R form, S form, or as a mixture of the R and S forms, including racemic

{00086504} and non-racemic mixtures. As used herein, the phrase "substantially free from other stereoisomers" means that the composition contains < 15%, more preferably < 10%, even more preferably < 5%, or most preferably < 1% of another stereoisomer(s).

"Treatment" or "treating" includes (1 ) inhibiting a disease in a subject or patient experiencing or displaying the pathology or symptomatology of the disease (e.g., arresting further development of the pathology and/or symptomatology), (2) ameliorating a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease (e.g., reversing the pathology and/or symptomatology), and/or (3) effecting any measurable decrease in a disease in a subject or patient that is experiencing or displaying the pathology or symptom atology of the disease.

Other abbreviations used herein are as follows: DMSO, dimethyl sulfoxide; LiAlPLs, lithium aluminum hydride; DMF, dimethylformamide; MeCN, acetonitrile; MeOH, methanol: EtOH, ethanoi; EtOAc, ethyl acetate " 'BuOH, tert-buianol; 'PrOH, isopropanoi; c HexQH, cyclohexanol; Pd/C, palladium on carbon; A.C2O, acetic anhydride; AcOOH, peracetic acid; tiCO^Et, ethyl formate; MeOTf, methyl trifluoromethatisulfonate; EtNCO, ethyl isocyanate; THF, tetrahydrofuran; KOTiu, potassium tert-butoxide; NaOMe, sodium methoxide; MTBE, methyl tert~bu†yl ether; DME, dimethoxy ethane; NBS, N- bromosuccinimide; DIBAL-H, diisohutylaluminium hydride; GDI, carbonyldiimidazoie; DIEA, diisopropylethylamine; HOBt-xH 2 0, hydroxybenzotriazole hydrate; TEA, triethylamine; DMAP, dimethylaminopyridine; EDC1. 1 -ethyl- 3- (3 - dimethykminopropyl)carbodiimide; 4 A MS, 4 angstrom molecular sieves; NMO, N- methylmorphoime N-oxide; TPAP, tetrapropylammonium perruthenate; DBDMH, 1 ,3- d " ibromo-5,5-dimethylhydantoin; NO, nitric oxide; iNOS, inducible nitric oxide synthase; COX-2, eyclooxygenase-2; FBS, fetal bovine serum; IFNy or TFN-y, interferon-γ; TNFa or TNF-a, tumor necrosis factor- ; IL-l β, interleukin- 1 β; HO-1 , inducible heme oxygenase.

The above definitions supersede any conflicting definition in any of the reference that is incorporated by reference herein. The fact that certain terms are defined, however, should not be considered as indicative that any term that is undefined is indefinite. Rather, all terms used are believed to describe the invention in terms such that one of ordinary skill can appreciate the scope and practice the present invention.

II. Compounds and Synthetic Methods

The compounds provided by the present disclosure are shown above in the summary of the invention, in the claims, and in the sections below. They may be made using the

{00086504} % methods outlined in the Examples section. These methods can be further modified and optimized using the principles and techniques of organic chemistry as applied by a person skilled in the art. Such principles and techniques are taught for example, in March 's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure (2007), which is incorporated by reference herein.

Compounds of the invention may contain one or more asymmetrically-substituted carbon or nitrogen atoms, and may be isolated in optically active or racemic form. Thus, all chirai, diastereomeric, racemic form, epimeric form, and all geometric isomeric forms of a chemical formula are intended, unless the specific stereochemistry or isomeric form is specifically indicated. Compounds may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. in some embodiments, a single diastereomer is obtained. The chirai centers of the compounds of the present invention can have the S or the R configuration.

Chemical formulas used to represent compounds of the invention will typically only show one of possibly several different tautomers. For example, many types of ketone groups are known to exist in equilibrium with corresponding enoi groups. Similarly, many types of imine groups exist in equilibrium with enamine groups. Regardless of which tautomer is depicted for a given compound, and regardless of which one is most prevalent, all tautomers of a given chemical formula are intended.

Atoms making up the compounds of the present invention are intended to include all isotopic forms of such atoms. Compounds of the present invention include those with one or more atoms that have been isotopicaily modified or enriched, in particular those with pharmaceutically acceptabie isotopes or those usefui for pharmaceutically research. Isotopes, as used herein, include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include deuterium and tritium, and isotopes of carbon include :J C and 14 C. Similarly, it is contemplated that one or more carbon atom(s) of a compound of the present invention may ¬ be replaced by a silicon atom(s). Furthermore, it is contemplated that one or more oxygen atom(s) of a compound of the present invention may be replaced by a sulfur or selenium atom(s).

Compounds of the present invention may also exist in prodrug form. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability,, manufacturing, etc.), the compounds employed in some methods of the invention may, if desired, be delivered in prodrug form. Thus, the invention contemplates

{00086504} 27 prodrugs of compounds of ike present nvers on as well as metho s of delivering prodrugs. Prodrugs of the compounds employed it! the invention may be prepared by modifying functional groups present in tire compound in such a way that tire modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Accordingly, prodrugs include, for example, compounds described herein in which a hydroxy, amino, or carboxy group is bonded to any group that, when the prodrug is administered to a subject, cleaves to form a hydroxy, amino, or carboxylic acid, respectively.

It should be recognized thai the particular anion or caiion formi g a part of any salt of this invention is not critical, so long as the sals, as a whole, is pharmacologically acceptable. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are presented in Handbook of Pharmaceutical Salts: Properties, and Use (2002), which is incorporated herein by reference.

It should be further recognized that the compounds of the present invention include those that have been further modified to comprise substituents that are convertible to hydrogen in vivo. This includes those groups that may be convertible to a hydrogen atom by enzymological or chemical means including, but not limited to, hydrolysis and hydrogenolysis. Examples include hydrolyzable groups, such as acyl groups, groups having an oxycarbonyl group, amino acid residues, peptide residues, o-nitrophenylsulfenyl, trimethy isilyl, tetrahydropyranyl, dipheny lphosphinyl, and the like. Examples of acyl groups include forniyi, acetyl, rrifluoroaeetyl, and the like. Examples of groups having an oxycarbonyl group include ethoxycarbonyl, tert-butoxycarbonyi (-C(0)OC(C¾)3, Boc), benzyl oxycarbonyl, j-methoxybenzyloxycarbonyl, vinyioxycarbonyl, β-(ρ- toluenesuifonyl)ethoxycarbonyl, and the like. Suitable amino acid residues include, but are not limited to, residues of Giy (glycine), Ala (alanine), Arg (arginine), Asn (asparagine), Asp (aspartic acid), Cys (cysteine), Glu (glutamic acid), His (histidine), He (isoleucine), Leu (leucine), Lys (lysine), Met (methionine), Phe (phenylalanine), Pro (proline), Ser (serine), Thr (threonine), Trp (tryptophan), Tyr (tyrosine), Val (valine), Nva (norvaline), Hse (homoserine), 4-Hyp (4-hydroxyproline), 5-Hyl (5-hydroxylysine), Orn (ornithine) and β- Ala, Examples of suitable amino acid residues also include amino acid residues that are protected with a protecting group. Examples of suitable protecting groups include those typically employed in peptide synthesis, including acyl groups (such as formyl and acetyl), arylmethoxycarbonyl groups (such as benzyloxycarbonyl and p-nitrobenzyloxycarbonyl), tert-butoxycarbonyl groups (-C(0)OC(CH3)3, Boc), and the like. Suitable peptide residues include peptide residues comprising two to five amino acid residues. The residues of these

{00086504} amino acids or peptides can be present in stereochemical configurations of the D-form, the L- forni or mixtures thereof. In addition, the amino acid or peptide residue may have an asymmetric carbon atom. Examples of suitable amino acid residues having an asymmetric carbon atom include residues of Ala, Leu, Phe, Trp, Nva, Val, Met, Ser, Lys, Thr and Tyr. Peptide residues having an asymmetric carbon atom include peptide residues having one or more constituent amino acid residues having an asymmetric carbon atom. Examples of suitable amino acid protecting groups include those typically employed in peptide synthesis, including acyl groups (such as formyl and acetyl), arylmethoxycarbonyl groups (such as benzyloxycarbonyl and >-nitrobenzyloxycarbonyi), tert-butoxycarbonyl groups (-C(0)OC(C¾)3), and the like. Other examples of substituents "convertible to hydrogen in vivo" include reductively eliminable hydrogenolyzable groups. Examples of suitable reductively eliminable hydrogenolyzable groups include, but are not limited to, arylsuifonyl groups (such as o-toluenesulfonyi); methyl groups substituted with phenyl or benzyioxy (such as benzyl, trityi and benzyloxymethyi); arylmethoxycarbonyl groups (such as benzyloxycarbonyl and o-methoxy-benzyloxycarbonyf); and haloethoxycarbonyl groups (such as β,β,β-trichloroethoxycarbonyl and β-iodoethoxycarbonyl).

Compounds of the invention may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g., higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the indications stated herein or otherwise.

ΓΠ. Biological Activity

Assay results for the suppression of IF y-induced NO production are shown for several of the compounds of the present invention in ' T able 1 below. In the right-hand column of this table under the RAW264.7 heading, the results are compared to those of bardoxolone methyl (RTA 402, CDDO-Me). Details regarding this assay are provided in the Examples section below.

{00086504} 29 Table ί. Suppression

IV, Diseases Associated with Inflammation and/or Oxidative Stress

Inflammation is a biological process thai provides resistance to infectious or parasitic organisms and the repair of damaged tissue. Inflammation is commonly characterized by localized vasodilation, redness, swelling, and pain, the recruitment of leukocytes to the site of infection or injury, production of inflammatory cytokines such as TNF-a and IL-I , and production of reactive oxygen or nitrogen species such as hydrogen peroxide, superoxide and peroxynitrite. In later stages of inflammation, tissue remodeling, angiogenesis, and scar formation (fibrosis) may occur as part of the wound healing process. Under normal circumstances, the inflammatory response is regulated and temporary and is resolved in an orchestrated fashion once the infection or injury has been dealt with adequately. However, acute inflammaiion can become excessive and life-threatening if regulatory mechanisms fail. Alternatively, inflammation can become chronic and cause cumulative tissue damage or systemic complications. Based at least on the evidence presented above, the compounds of this invention may be used in the treatment or prevention of inflammation or diseases associated with inflammation.

Many serious and intractable human diseases involve dys egulation of inflammatory processes, including diseases such as cancer, atherosclerosis, and diabetes, which were not traditionally viewed as inflammatory conditions. In the case of cancer, the inflammatory processes are associated with tumor formation, progression, metastasis, and resistance to therapy. Atherosclerosis, long viewed as a disorder of lipid metabolism, is now understood to be primarily an inflammatory condition, with activated macrophages playing an important role in the formation and eventual rupture of atherosclerotic plaques. Activation of inflammatory signaling pathways has also been shown to play a role in the development of insulin resistance, as well as in the peripheral tissue damage associated with diabetic hyperglycemia. Excessive production of reactive oxygen species and reactive nitrogen species such as superoxide, hydrogen peroxide, nitric oxide, and peroxynitrite is a hallmark of inflammatory conditions. Evidence of dysregulated peroxynitrite production has been reported in a wide variety of diseases (Szabo et at, 2007; Schulz et at, 2008; Forstermami, 2006; Pall, 2007).

Autoimmune diseases such as rheumatoid arthritis, lupus, psoriasis, and multiple sclerosis involve inappropriate and chronic activation of inflammatory processes in affected tissues, arising from dysfunction of self vs. non-self recognition and response mechanisms in the immune system. In neurodegenerative diseases such as Alzheimer's and Parkinson's

{00086504} diseases, neural damage is correlated with activation of microglia and elevated levels of proinflammatory proteins such as inducible nitric oxide synthase (iNOS). Chrome organ failure such as renal failure, heart failure, liver failure, and chronic obstructive pulmonary disease is closely associated with the presence of chronic oxidative stress and inflammation, leading to the development of fibrosis and eventual loss of organ function. Oxidative stress in vascular endothelial cells, which line major and minor blood vessels, can lead to endothelial dysfunction and is believed to be an important contributing factor in the development of systemic cardiovascular disease, complications of diabetes, chronic kidney disease and other forms of organ failure, and a number of other aging-related diseases including degenerative diseases of the central nervous system and the retina.

Many other disorders involve oxidative stress and inflammation in affected tissues, including inflammatory bowel disease; inflammatory skin diseases; mucositis related to radiation therapy and chemotherapy; eye diseases such as uveitis, glaucoma, macular degeneration, and various forms of retinopathy; transplant failure and rejection; ischemia- reperfusion i jur ''; chronic pain; degenerative conditions of the bones and joints including osteoarthritis and osteoporosis; asthma and cystic fibrosis; seizure disorders; and neuropsychiatric conditions including schizophrenia, depression, bipolar disorder, posttraumatic stress disorder, attention deficit disorders, autism-spectrum disorders, and eating disorders such as anorexia nervosa. Dysregulation of inflammatory signaling pathways is believed to be a major factor in the pathology of muscle wasting diseases including muscular dystrophy and various forms of cachexia.

A variety of life-threatening acute disorders also involve dysregulated inflammatory signaling, including acute organ failure involving the pancreas, kidneys, liver, or lungs, myocardial infarction or acute coronary syndrome, stroke, septic shock, trauma, severe burns, and anaphylaxis.

Many complications of infectious diseases also involve dysregulation of inflammatory responses. Although an inflammatory response can kill invading pathogens, an excessive inflammatory response can also be quite destructive and in some cases can be a primary- source of damage in infected tissues. Furthermore, an excessive mflammatory response can also lead to systemic complications due to overproduction of inflammatory cytokines such as T F-a and IL-1. This is believed to be a factor in mortality arising from severe influenza, severe acute respiratory syndrome, and sepsis.

{00086504} 46 The aberrant or excessive expression of either iNOS or cyclooxygenase-2 (COX-2) has been implicated in the pathogenesis of many disease processes. For example, it is clear that NO is a potent mutagen (Tamir and ' T ansiebaum, 1996), and that nitric oxide can also activate COX-2 (Salvemini et at, 1994). Furthermore, there is a marked increase in iNOS in rat colon tumors induced by the carcinogen, azoxymethane (Takahashi el at, 1997). A series of synthetic triterpenoid analogs of oleanolic acid have been shown to be powerful inhibitors of cellular inflammatory processes, such as the induction by IFN-y of inducible nitric oxide synthase (iNOS) and of COX-2 in mouse macrophages. See Honda el at (2000a); Honda el at (2000b), and Honda ei at (2002), which are ail incorporated herein by reference.

In one aspect, compounds disclosed herein are characterized by their ability to inhibit the production of nitric oxide in macrophage-derived RAW 264.7 cells induced by exposure to γ-interferon. They are further characterized by their ability to induce the expression of antioxidant proteins such as NQOl and reduce the expression of pro-inflammatory proteins such as COX-2 and inducible nitric oxide synthase (iNOS). These properties are relevant to the treatment of a wide array of diseases and disorders involving oxidative stress and dysregulation of inflammatory processes including cancer, complications from localized or total-body exposure to ionizing radiation, mucositis resulting from radiation therapy or chemotherapy, autoimmune diseases, cardiovascular diseases including atherosclerosis, isehemia-reperfusion injury, acute and chronic organ failure including renal failure and heart failure, respiratory diseases, diabetes and complications of diabetes, severe allergies, transplant rejection, graft-versus-host disease, neurodegenerative diseases, diseases of the eye and retina, acute and chronic pain, degenerative bone diseases including osteoarthritis and osteoporosis, inflammatory bowel diseases, dermatitis and other skin diseases, sepsis, burns, seizure disorders, and neuropsychiatric disorders.

Without being bound by theory, the activation of the antioxidant/anti-inflammatory

Keap l/Nrfi/ARE pathway is believed to be implicated in both the anti-inflammatory and anti-carcinogenic properties of the compounds disclosed herein.

In another aspect, compounds disclosed herein may be used for treating a subject having a condition caused by elevated levels of oxidative stress in one or more tissues. Oxidative stress results from abnormally high or prolonged levels of reactive oxygen species such as superoxide, hydrogen peroxide, nitric oxide, and peroxynitrite (formed by the reaction of nitric oxide and superoxide). The oxidative stress may be accompanied by either acute or chronic inflammation. The oxidative stress may be caused by mitochondrial

{00086504} 47 dysfunction, by activation of immune cells such as macrophages and neutrophils, by acute exposure to an external agent such as ionizing radiation or a cytotoxic chemotherapy agent (e.g. , doxorubicin), by trauma or other acute tissue injury, by ischemia/reperfusion, by poor circulation or anemia, by localized or systemic hypoxia or hyperoxia, by elevated levels of inflammatory cytokines and other inflammation-related proteins, and/or by other abnormal physiological staies such as hyperglycemia or hypoglycemia.

In animal models of many such conditions, stimulating expression of inducible heme oxygenase (HO-1), a target gene of the Nrf2 pathway, has been shown to have a significant therapeutic effect including models of myocardial infarction, renal failure, transplant failure and rejection, stroke, cardiovascular disease, and autoimmune disease (e.g. , Sacerdoti et al, 2005; Abraham & Kappas, 2005; Bach, 2006; Araujo et al, 2003; Liu et al, 2006; Ishikawa et al, 2001 ; Kruger et al, 2006; Satoh et al, 2006; Zhou et al, 2005; Morse and Choi, 2005; Morse and Choi, 2002). This enzyme breaks free heme down into iron, carbon monoxide (CO), and biliverdin (which is subsequently converted to the potent antioxidant molecule, bilirubin).

In another aspect, compounds of this invention may be used in preventing or treating tissue damage or organ failure, acute and chronic, resulting from oxidative stress exacerbated by inflammation. Examples of diseases that fall in this category include: heart failure, liver failure, transplant failure and rejection, renal failure, pancreatitis, fibrotic lung diseases (cystic fibrosis, COPD, and idiopathic pulmonary fibrosis, among others), diabetes (including complications), atherosclerosis, isehemia-reperfusion injury, glaucoma, stroke, autoimmune disease, autism, macular degeneration, and muscular dystrophy. For example, in the case of autism, studies suggest that increased oxidative stress in the central nervous sysiem may contribute to the development of the disease (Chauhan and Chauhan, 2006).

Evidence also finks oxidative stress and inflammation to the development and pathology of many other disorders of the central nervous system, including psychiatric disorders such as psychosis, major depression, and bipolar disorder; seizure disorders such as epilepsy; pain and sensory syndromes such as migraine, neuropathic pain or tinnitus; and behavioral syndromes such as the attention deficit disorders. See, e.g., Dickerson et al, 2007; Hanson et al, 2005; Kendall-Tackett, 2007; Lencz et al, 2007; Dudhgaonkar el al, 2006; Lee et al, 2007; Morris et al, 2002; Ruster et al, 2005; Mclver et al, 2005; Sarchielli et al, 2006; Kawakami et al, 2006; Ross et al, 2003, which are all incorporated by reference herein. For example, elevated levels of inflammatory cytokines, including TNF, interferon-γ,

{00086504} 48 and IL-6, are associated with major mental illness (Dickerson et al., 2007). Microglial activation has also been linked to major mental illness. Therefore, downregulating inflammatory cytokines and inhibiting excessive activation of microglia could be beneficial in patients with schizophrenia, major depression, bipolar disorder, autism-spectrum disorders, and other neuropsychiatric disorders.

Accordingly, in pathologies involving oxidative stress alone or oxidative stress exacerbated by inflammation, treatment may comprise administering to a subject a therapeutically effective amount of a compound of this invention, such as those described above or throughout this specification. Treatment ma be administered preventively, in advance of a predictable state of oxidative stress (e.g., organ transplantation or the administration of radiation therapy to a cancer patient), or it may be administered therapeutically in settings involving established oxidative stress and inflammation.

The compounds disclosed herein may be generally applied to the treatment of inflammatory conditions, such as sepsis, dermatitis, autoimmune disease and osteoarthritis. In one aspect, the compounds of this invention may be used to treat inflammatory pain and/or neuropathic pain, for example, by inducing Nrf2 and/or inhibiting NF-KB.

In some embodiments, the compounds disclosed herein may be used in the treatment and prevention of diseases such as cancer, inflammation, Alzheimer's disease, Parkinson's disease, multiple sclerosis, autism, amyotrophic lateral sclerosis, Huntington's disease, autoimmune diseases such as rheumatoid arthritis, lupus, Crohn's disease and psoriasis, inflammatory bowel disease, all other diseases whose pathogenesis is believed to involve excessive production of either nitric oxide or prostaglandins, and pathologies involving oxidative stress alone or oxidative stress exacerbated by inflammation.

Another aspect of inflammation is the production of inflammatory prostaglandins such as prostaglandin E. These molecules promote vasodilation, plasma extravasation, localized pain, elevated temperature, and other symptoms of inflammation. The inducible form of the enzyme COX-2 is associated with their produc tion, and high levels of COX-2 are found in inflamed tissues. Consequently, inhibition of COX-2 may relieve many symptoms of inflammation and a number of important anti- inflammatory drugs (e.g. , ibuprofen and celecoxib) act by inhibiting COX-2 activity. Recent research, however, has demonstrated that a class of cyclopentenone prostaglandins (cyPGs) (e.g., 15-deoxy prostaglandin J2, a.k.a. PGJ2) plays a roie in stimulating the orchestrated resoiution of inflammation (e.g., Rajakariar et al, 2.007). COX-2 is also associated with the production of cyclopentenone prostaglandins.

{00086504} 49 Consequently, inhibition of COX-2 may interfere with the full resolution of inflammation, potentially promoting the persistence of activated immune cells in tissues and leading to chronic, "smoldering" inflammation. This effect may be responsible for the increased incidence of cardiovascular disease in patients using selective COX-2 inhibitors for long periods of time.

In one aspect, the compounds disclosed herein may be used to control the production of pro-inflammatory cytokines within the cell by selectively activating regulatory cysteine residues (RCRs) on proteins that regulate the activity of redox-sensitive transcription factors. Activation of RCRs by cyPGs has been shown to initiate a pro-resolution program in which the activity of the antioxidant and cytoprotective transcription factor Mrf2 is potently induced and the activities of the pro-oxidant and pro-inflammatory transcription factors NF- Β and the STATs are suppressed. Tn some embodiments, this increases the production of antioxidant and reductive molecules (NQOL HQ-1, SODL y-GCS) and decreases oxidative stress and the production of pro-oxidant and pro-inflammatory molecules (i OS, COX-2, TNF-cc). In some embodiments, the compounds of this invention may cause the cells that host the inflammatory event to reveri to a non-inflammatory state by promoiing the resolution of inflammation and limiting excessive tissue damage to the host.

V. Pharmaceutical Formulations and Routes of Administration

The compounds of the present disclosure may be administered by a variety of methods, e.g., orally or by injection (e.g. subcutaneous, intravenous, intraperitoneal, etc.). Depending on the route of administration, the active compounds may be coated in a material to protect the compound from the action of acids and other natural conditions which may inactivate the compound. They may also be administered by continuous perfusion/infusion of a disease or wound site.

'TO administer the therapeutic compound by other than parenteral administration, it may be necessar '- to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the therapeutic compound may be administered to a patient in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Liposomes include water-in- oil-in-water CGF emulsions as well as conventional liposomes (Strejan el ah, 1984).

The therapeutic compound may also be administered parenterally, intraperitoneally, intraspinally, or intracerebrally. Dispersions can be prepared in glycerol, liquid polyethylene

{00086504} srrj glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.

Pharmaceutical compositions suitable for injectable use include: sterile aqueous solutions (where water soluble), dispersions, and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. See for example U.S. Patent Application by J. Zhang, entitled "Amorphous Solid Dispersions of CDDO-Me for Delayed Release Oral Dosage Compositions," filed February 13, 2009, which is incorporated herein by reference. In all cases, the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (such as, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, sodium chloride, or polyalcohois such as mannitol and sorbitol, in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.

Sterile injectable solutions can be prepared by incorporating the therapeutic compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the therapeutic compound into a sterile carrier which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the ease of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., the therapeutic compound) plus any additional desired ingredient from a previously sterile-filtered solution thereof.

The therapeutic compound can be orally administered, for example, with an inert diluent or an assimilable edible earner. The therapeutic compound and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the

{00086504} sr i therapeutic compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. The percentage of the therapeutic compound in the compositions and preparations may, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.

It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic compound for the treatment of a selected condition in a patient.

The therapeutic compound may also be administered topically to the skin, eye, or mucosa. Alternatively, if local delivery to the lungs is desired the therapeutic compound may be administered by inhalation in a dry-powder or aerosol formulation.

Active compounds are administered at a therapeutically effective dosage sufficient to treat a condition associated with a condition in a patient. For example, the efficacy of a compound can be evaluated in an animal model system that may be predictive of efficacy in treating the disease in humans, such as the model systems shown in the examples and drawings.

The actual dosage amount of a compound of the present disclosure or composition comprising a compound of the present disclosure administered to a subject may be determined by physical and physiological factors such as age, sex, body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the subject and on the route of administration. These factors may be determined by a skilled artisan. The practitioner responsible for administration will typically determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject. The dosage may be adjusted by the individual physician in the event of any complication.

An effective amount typically will vary from about 0.001 mg/kg to about 1000 mg/kg, from about 0.01 mg kg to about 750 mg/kg, from about 100 mg/kg to about 500 mg/kg, from about 1.0 mg/kg to about 250 mg kg, from about 10.0 mg/kg to about 150 mg/kg in one or more dose admmistrations daily, for one or several days (depending of course of the mode of administration and ihe factors discussed above). Other suitable dose ranges include 1 nig to 10000 mg per day, 100 mg to 10000 mg per day, 500 mg to 10000 mg per day, and 500 mg to 1000 mg per day. in some particular embodiments, the amount is less than 10,000 mg per day with a range of 750 mg to 9000 mg per day.

The effective amount may be less than 1 mg/kg/day, less than 500 mg/kg/day, less than 250 mg/kg/day, less than 100 mg/kg/day, less than 50 mg/kg day, less than 25 mg/kg/day or less than 10 mg/kg/day. It may alternatively be in the range of 1 mg/kg/day to 200 mg/kg day. For example, regarding treatment of diabetic patients, the unit dosage may be an amount that reduces blood glucose by at least 40% as compared to an untreated subject. In another embodiment, the unit dosage is an amount that reduces blood glucose to a level that is ± 10% of the blood glucose level of a non-diabetic subject.

In other non-limiting examples, a dose may also comprise from about 1 micro- gram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg body weight, about 50 microgram/kg body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligranV ' kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg/body weight to about 100 mg/kg body weight, about 5 microgram kg/body weight to about 500 miliigram/kg/body weight etc., can be administered, based on the numbers described above.

In certain embodiments, a pharmaceutical composition of the present disclosure may- comprise, for example, at least about 0.1% of a compound of the present disclosure. In other embodiments, the compound of the present disclosure may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein.

Single or multiple doses of the agents are contemplated. Desired time intervals for delivery of multiple doses can be determined by one of ordinary skill in the art employing no more than routine experimentation. As an example, subjects may be administered two doses daily at approximately 12 hour intervals. In some embodiments, the agent is administered once a day.

{00086504} The agent(s) may be administered on a routine schedule. As used herein a routine schedule refers to a predetermined designated period of time. The routine schedule may encompass periods of time which are identical or which differ in length, as long as the schedule is predetermined. For instance, the routine schedule may involve administration twice a day, every day, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks therebetween. Alternatively, the predetermined routine schedule may involve administration on a twice daily basis for the first week, followed by a daily basis for several months, etc. In other embodiments, the invention provides that the agent(s) may be taken orally and that the timing of which is or is not dependent upon food intake. Thus, for example, the agent can be taken every morning and/or every evening, regardless of when the subject has eaten or will eat.

VI. Combination Therapy

In addition to being used as a monotherapy, the compounds of the present invention may also find use in combination therapies. Effective combination therapy may be achieved with a single composition or pharmacological formulation that includes both agents, or with two distinct compositions or formulations, administered at the same time, wherein one composition includes a compound of this invention, and the other includes the second agent(s). Alternatively, the therapy may precede or follow the other agent treatment by intervals ranging from minutes to months.

Non-limiting examples of such combination therapy include combination of one or more compounds of the invention with another anti-inflammatory agent, a chemotherapeutic agent, radiation therapy, an antidepressant, an antipsychotic agent, an anticonvulsant, a mood stabilizer, an ants -infective agent an antihypertensive agent, a cholesterol-lowering agent or other modulator of blood lipids, an agent for promoting weight loss, an antithrombotic agent, an agent for treating or preventing cardiovascular events such as myocardial infarction or stroke, an antidiabetic agent, an agent for reducing transplant rejection or graft-versus-host disease, an anti-arthritic agent, an analgesic agent, an anti-asthmatic agent or other treatment for respiratory diseases, or an agent for treatment or prevention of skin disorders. Compounds of the invention may be combined with agents designed to improve a patient's immune response to cancer, including (but not limited to) cancer vaccines. See Lu et at (201 1), which is incorporated herein by reference.

{00086504} VIL Examples

The following examples are included to demonstrate preferred embodiments of the invention, t should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of ihe invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention. Methods and Materials

Nitric Oxide Production and Cell Viability Assay. RAW264.7 mouse macrophages were plated in 96-well plates at 30,000 cells/well in triplicate in RPMJ1640 + 0.5% FBS and incubated at 37°C with 5% CO ? .. On the next day, cells were pre-treated with DMSO or drug (0-200nM dose range) for 2 hours, and then treated with recombinant mouse IFNy (R&D Systems) for 24 hours. Nitric Oxide concentration in media was determined using the Griess reagent system (Promega). Cell viability was determined using WST-1 reagent (Roche). IC 50 values were determined based on the suppression of IFNy induced Nitric Oxide production normalized to cell viability.

NQOl-ARE Luciferase Reporter Assay. This assay allows for quantitative assessment of the endogenous acti vity of the Nrf2 transcription factor in cultured mammalian cells. Expression of Firefly luciferase from NQOl -ARE luciferase reporter plasmid is controlled by binding of Nr£2 to a specific enhancer sequence corresponding to the antioxidant response element (ARE) that was identified in the promoter region of the human NADPH:quinone oxidoreductase 1 (ΝΟΟΓ) gene (Xie el a!., 1995). The plasmid was constructed by inserting a sequence:

5'- CAGTCACAGTGACTCAGCAGAATCTG-3' (SEQ ID NO: l)

encompassing the human NQOl-ARE into the pLuc-MCS vector using Hindlll/Xhol cloning sites (GenScript Corp., Piscaiaway, NJ), The assay is performed in Hull? cells maintained in DMEM (Tnvitrogen) supplemented with 10% FBS and 100 Li/ml (each) of penicillin and streptomycin. For the assay, cells are plated in 96-well plates at 17,000 cells per well. Twenty four hours later, the cells are co-transfected with 50 ng each of NQOl-ARE reporter plasmid and pRL-TK plasmid using Lipofectamine 2.000 transfection reagent (Invitrogen).

{00086504} ir e pRL-TK plasmid constitutively expresses Renilia luciferase and is used as an internal control for normalization of transfection levels. Thirty hours after transfeetion, the ceils are treated with compounds (at concentrations ranging from 0 to 1 μΜ) for eighteen hours. Firefly and Renilia luciferase activity is assayed by Dual-Glo Luciferase Assay (Promega Corp., Madison, WI), the luminescence signal is measured on an L-Max II lummometer (Molecular Devices), Firefly luciferase activity is normalized to the Renilia activity, and fold induction over a vehicle control (DMSO) of normalized Firefly activity is calculated. The fold induction at 62.5 nM concentration is used for comparing relative potencies of compounds to induce Nrf2 transcriptional activity. See Xie et al, 1995, which is incorporated herein by reference.

{00086504} 56 Yields

{00086504} 57

Reagents and conditions: (a) LiAlH 4 , THF, 0 °C to rt, 72%; (b) Phl(OAc) 2 , TEMPO, H 2 0, CH 2 C1 ¾ rt, 72%; (c) triethyl phosphonoacetate, NaH, 0 °C to rt,, 67%; (d)TPAP, NMO, 4 A MS, CH 2 <¾ rt, 88%; (e) ¾ Pd/C, THF, rt, 99%; (t) EtOCHO, NaOMe, MeOH, rt; (g) i) NH 2 OH « HCL EtOH, H 2 0, 55 °C, ii) MeOH, HCl, rt, 80% from 5; (h) NaOMe, MeOH, 55 °C, 99%; (i) i) DBDMH, DMF, 0 °C. ii) pyridine, 55 °C, TX63762: 17%, TX63742: 75%; (j) HCl, H 2 0, MeCN, 65 °C, 98%.

Reagents and conditions: (a) HC0 2 Et, NaOMe, MeOH, rt; (b) \i !■()!!·! !C!. EtOH, H 2 0, 55 °C, 81% from S; (c) NaOMe, MeOH, 55 °C, quantitative; (d) i) DBDMH, DMF, 0 °C. ii) pyridine, 55 °C, 63% from 11a, lib.

{00086504} 59

Reagents and conditions: (a) ROH, HQ, ft to 55 °C, TX 64042: 65%, TX64843: 79%, TX64058

TX64046 R = Ph

Reagents and conditions: (a) ROH, TEA, DMAP, EDCi. CH 2 C1 2 , rt, TX64046: 64%, TX64066: 64%, TX64067: 15%, TX64053: 70%; (b) HC1 CH 2 C¾, l ,4-dioxane, rt, 83%.

i00086504i 60

Scheme 5

TX64045: R

TX84052: R = NHCH 2 C0 2 Me -

TX64054: R = NHCH 3 CO;. : H *-

TX64124: R = 4-Bocpjperazine

TX8413S: R = piperazirte *

Reagents and conditions: (a) amine, TEA, DMAP, EDCI, CH 2 Ci 2 , rt, TX64064: 22%, ΤΧ64Θ65: 29%, TX64057: 32%, TX64059: 71%, TX64075: 25%, TX63949: 47%, TX63763: 82%, TX64045: 45%, TX64048: 51 %, TX64068: 12%, TX64124: 88%; (b) amine-HCl, TEA, DMAP, EDCI, CH2CI2, rt, TX64056: 57%, TX63770: 81%, TX63768: 70%, TX63764: 76%, ΤΧ6406Θ: 77%, TX63950: 71%, TX64049: 59%, TX64047: 46%, TX64044: 65%, TX64052: 64%; (c) HCl, ¾0, MeCN, 55 °C, 71%; (d) HCi, 1 ,4-dioxane, CH 2 Ci 2 , rt, 35%.

Reagents and conditions: (a) 4-ra rphoHnecar onyl chloride, DMAP, pyridine, r o 90 °C, 8 h, 49%.

{00086504} 64

{00086504}

Sche

Reagents and conditions: (a) LiAlELj, THF, 0 °C to reflux; (b) NaOCL NaHC0 3 , NaBr, TEMPO, ¾0, C¾C1 ¾ 0 °C to rt, 79% from oleanolic acid: (c) i) KO'Bu, (methoxymethyi)tripheiiylphosphonium chloride, THF, rt, ii) H 2 SO4, rt, 96%; (d) NaBFi 4 , MeOFl, THF, rt, quantitative; (e) Ac 2 0, pyridine, DMAP, CH 2 C1 2 , rt, 75% from 19; (f) AcOOH, Na 2 C0 3 , AcOH, 50 °C, 96%; (g) Br,, HCI, 1 ,4-dioxane, MeCN, 35 °C; (h)

;0008650 ; Qi

H 2 S0 4 , MeOH, THF, 35 °C to reflux, 74% from 22; (i) NaOCL AcOH, H 2 0, rt, 80%; (j) NaOMe, MeOH, EtOCHO, 0 °C to rt; (k) NH 2 0H-HC1, EtOH, H 2 0, 55 °C; (1) i) NaOMe, MeOH, 55 °C, ii) HCL 1,4-dioxane, 55 °C, 71% from 25; (m) i) DBDMH, DMF, 0 °C, ii) pyridine, 55 °C, 80%; (n) MeOTf, 2,6-di-feri-butyl-4-methylpyridine, CH 2 Ci 2 , rt, 76%.

Scheme 13

Reagents and conditions: (a) Ac 2 0, pyridine, DM A P. CH 2 <¾, rt, 76%. Scheme 14

Reagents and conditions: (a) EtNCO, toluene, rt to 70 °C, 73%.

Synthesis and Characterization of Compounds and intermediates

Compound 2: l .iA ll L (2.0 M in THF, 50 mL, 100 rnmol) was carefully added to a 0 °C solution of compound 1 (9.65 g, 20.0 rnmol) in THF (350 mL) over -10 min. The ice bath was removed after 30 min and the reaction stirred an additional 2.5 h. The solution was diluted with MTBE (200 mL); cooled to 0 °C; and quenched by sequential addition of 5.3 mL each water, 4 M NaOH, and water. The mixture was stirred at room temperature for 30 min, filtered through a plug of eeliie, eluted with MTBE, and concentrated to give a white solid. The resultant solid was suspended in CH 2 CI 2 , stirred at room temperature for 2 h, and the solid isolated by filtration [washed with CH 2 CI 2 , then dried] to give compound 1 (6.56 g, 72%) which was used without further purification: m/z 441.3 (M-H 2 0+1).

Compound 3: Phl(OAc) 2 (10.40 g, 32,3 nmio!), TEMPO (2.27 g, 14.5 rnmol), and water (15 ml.) were added to a room temperature suspension of compound 2 (6.56 g, 14,3 mmol) in CH 2 C1 2 ( 1.5 L) and stirred vigorously for I S h. The resultant biphasic, homogeneous solution was treated with anhydrous Na2S04, stirred an additional 15 min, filtered and concentrated to a viscous oil. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound 4 (4,72 g, 72%) as an off-white solid: m/z 439.3 ( M - I I .O - h.

Compound 4: Triethyl phosphonoacetate (18,8 mL, 94.8 rnmol) was added to a 0 °C suspension of NaH (3,81 g, 60%, 95.3 mmol) in THF (285 mL), and the resultant mixture stirred at 0 °C for 15 min, then warmed to room temperature over 30 min. The mixture was cooled to 0 °C, a solution of compound 3 (8.67 g, 19.0 mmol) in THF (75 mL) was added, and the transfer completed with THF (20 mL) wash. The reaction was maintained at 0 °C for approximately 4 h and then warmed to room temperature overnight. The reaction was again cooled to 0 °C and quenched by the carefid addition of 1 N HCl, stirred vigorously at room temperature for 15 min, and extracted with MTBE. The combined organic fractions were washed with brine, then dried over Na 2 S04, and concentrated. The residue was purified by- flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound 4 (6.68 g, 67%) as a white solid: m/z 509.3 (M-H2O+I).

Compound 5: A mixture of compound 4 (6.68 g, 12.7 mmol), 4 A MS (12,7 g), NMO (4.47 g, 38.2 mmol), and CH 2 CI 2 (250 mL) was stirred at room temperature for 15 min. TPAP (458 mg, 1.30 mmol) was then added, and the reaction was stirred for 1 h. The reaction was concentrated to -Λ 0 mL and purified by flash chromatography (silica gel, 0% to

{00086504} 71 100% EtOAc in hexanes) to give compound 5 (5.89 g, 88%) as a white foam solid: m/z 523.3 (M+l).

Compound 6: A flask containing a suspension of compound S (5.89 g, 1 1.3 mmol), Pd/C (10%, 1 .45 g), and THF (250 mL) was purged with N? and then H 2 , and the reaction was vigorously stirred under H 2 (balloon pressure) for 5 h. The resultant suspension was sparged with N? for 30 min, filtered through a short plug of celite, eiuted with CH 2 Ci 2 , and concentrated to give compound 6 (5.84 g, 99%) as a white solid: m/z 525.4 (M+l ).

Compounds 7a,7b: A solution of compound 6 (5.84 g, 1 1.1 mmol), NaOMe (25 % in MeOH, 35 mL) and HCQ 2 Et (70 mL) was siirred at room temperature for 3 h, diluted with 1 N HQ, and extracted with EtOAc. The combined organic fractions were washed with brine, dried with Na2SC>4, and concentrated to give compounds 7a,b (Me-ester:Et-ester = 9:91 ) as an off-white foam solid that was used without further purification: 7a m/z 539.3 (M+l ), 7b m/z 553.4 (M+l ).

Compound 8a, 8b: A mixture of compounds 7a, 7b (all above obtained, -1 1.3 mmol), NH 2 OH » HCi (1.06 g, 15.3 mmol), EtOH (100 mL) and water ( 1 7 mL) was heated to 55 °C for 16 h. The resultant solution was cooled to room temperatm'e, diluted with 1 N HQ and extracted with EtOAc. The combined organic fractions were washed with 1 N HQ and brine, dried with Na 2 S0 4 , and concentrated. The residue was dissolved in MeOH (700 mL), treated with 12 N HQ ( 1.0 mL), and stirred at room iemperature for 5 h. The mixture was concentrated to -30 mL, diluted with CH 2 C1 2 , washed with brine, dried with Na 2 S0 4 and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 60% EtOAc in hexanes) to give compounds 8a, 8b (4.84 g, Me-ester:Et-ester - 25:75, 80% from 6) as a white solid: 8a m/z 536.4 (M+ l), 8b m/z 550.3 (M+ l).

Compounds 9a, 9b: A solution of 8a, 8b (4.84 g, 8.84 mmol), NaOMe (25% in MeOH, 5.3 mL), and MeOH (1 10 mL) was heated to 55 °C for 5 h. The resultant mixture was diluted with 1 N HQ and extracted with CH 2 C1 2 . The combined organic fractions were washed with brine, dried with r>!a 2 S0 4 , and concentrated to give compounds 9a, 9b (4.69 g, carboxylic acid:Me-ester = 17:83, 99%) as a yellow solid that was used without further purification: 9a m/z 522.3 (M+l ), 9b m/z 536.3 (M+ l ).

Compounds TX63762 and TX63742: l ,3-dibromo-5,5-dimethylhydantoin ( 1.25 g,

4.37 mmol) was added to a 0 °C solution of 9a, 9b (4.69 g, 8.79 mmol) in DMF (106 mL). The mixture was stirred at 0 °C for 4 h. Pyridine (2.83 mL, 35.0 mmol) was then added, and the reaction was heated to 55 °C. The reaction was cooled to room temperature after 4 h and was stirred an additional 3 d. The resultant solution was diluted with 1 N HQ and extracted

{00086504} 72 with EtOAc. The combined organic fractions were washed with water and brine, dried with Na. 2 S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63762 (780 mg, 17%) as a white solid and compound TX63742 (3.53 g, 75%) as an off-white solid: TX63762 ¾ NMR (400 MHz, CDC1 3 ) 5 8.04 (s, IH), 5.98 (s, 1H), 3.06 (d, IH, J - 4.7 Hz), 2.34 (m, 2H), 2.24 (td, 1H, J - 4.3, 13.4 Hz), 1.69 (m, M l ! }. 1.50 (s, 3H), 1.49 (s, 3H), 1.26 (s, 3H), 1.26 (m, 4H), 1.18 (s, 3H), 1.04 (m, 2H), 1.02 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 520.3 (M+l ); TX63742 ; H NMR (400 MHz, CDCI 3 ) δ 8.04 (s, 1 H), 5.97 (s, I H), 3.67 (s, 3H), 3.06 (d, 1H, J - 4.7 Hz), 2.30 (m, 2H), 2.21 (td, IH, ,/ - 4.6, 13.0 Hz), 1 .74 (m, 9H), 1 .50 (s, 6H), 1 .46 (m, 2H), 1 .26 (s, 3H), 1.25 (m, 4H), 1.18 is, 3H), 1.02 (m, 2H), 1.01 (s, 3H), 0.92 fs, 3H), 0.87 (s, 3H); m/z 534.3 (M+l).

Compound TX63762: A. suspension of TX63742 (3.53 g, 6.61 mmol) in MeCN (200 ml.) and 1 N HQ (66 mL) was heated to 65 °C for 17 h. The resultant solution was cooled to room temperature and extracted with EtOAc. The combined organic fractions were washed with brine, dried with Na 2 S04, and concentrated to give compound TX63762 (3.354 g, 98%) as a pale yellow solid.

Compounds 10a, 10b: A solution of compound 5 (250 mg, 0.478 mmol), NaOMe (25 % in MeOH, 1.25 mL) and HC0 2 Et (3.75 mL) was stirred at room temperature for 3 h, diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S04, and concentrated to give compounds 10a, 10b (Me~ester:Et-ester = 23:77) as an off-white foam solid that were used without further purification: l(5a m/z 537.3 (M+l ), 10b m/z 551 .4 (M+l ).

Compounds 11a, lib: A mixture of compounds 10a, 10b (all above obtained, 0.48 mmol), NH 2 OH » HCl (45.1 g, 0.649 mmol), EtOH (4.25 mL) and water (0.75 mL) was heated to 55 °C for 20 h. The resultant solution was cooled to room temperature, diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated. The crude residue was purified by flash chromatography (silica gel, 0% to 60% EtOAc in hexanes) to give compounds H a, l ib (210 mg, Me-ester:Et-ester = 21 :79, 81% from 5) as a white solid: 11a m/z 534.3 (M+l), 111) m/z 548.3 (M+ l).

Compound 12: A mixture of compounds 11a, lib (210 mg, 0.385 mmol), NaOMe (25% in MeOH, 0.2 mL) and MeOH (4.8 mL) was heated to 55 °C for 3.5 h. The resultant mixture was diluted with 1 N HCl and extracted with EtOAc. The combined organic fractions were dried with Na 2 S04 and concentrated to give compound 12 (206 mg,

{00086504} 73 quantitative) as a glassy, white solid that was used without further purification: m/z 534.3 (M+l).

Compounds TX63743: 1 ,3-dibromo-5,5-dimethylhydantoin (57.6 mg, 0.201 mmol) was added to a 0 °C solution of compound 12 (206 mg, -0.385 mmol) in DMF (3.9 mL). The mixture was stirred at 0 °C for 2 h, pyridine (0.13 mL, 1.6 mmol) was added, and the reaction heated to 55 °C. The reaction was cooled to room temperature after 4 h and stirred overnight. The resultant solution was diluted with EtOAc, washed with 1 N HC1, 10% Na 2 S0 3 , and brine, then dried with Na?S0 and concentrated. The cmde residue was purified by flash chromatography (silica gel, 0% to 45% EtOAc in hexanes) to give compound TX63743 (129 mg, 63% from 11a, lib) as a white solid: 'H MR (400 MHz, CDC1 3 ) δ 8.02 (s, IH), 6.95 (d, 1H, J = 16.3 Hz), 5.97 (s, 1H), 5.87 (d, 1H, J = 16.4 Hz), 3.74 (s, 3H), 2.85 (d, I H, J - 4.6 Hz), 2.57 (td, IH, J - 4.1, 13.2 Hz), 2.06 (dt, IH, J - 4.0, 13.8 Hz), 1.65 (m, 8H), 1.47 (s, 3H), 1.32 (s, 3H), 1.25 (s, 3H), 1.20 (m, 6H), 1.17 (s, 3H), 1.01 (s, 3H), 0.98 (s, 3H), 0.91 (s, 3H); m/z 532.3 (M+l ).

Compound TX64042: A mixture of TX63762 (53.0 mg, 0.102 mmol), EtOH (2 mL) and 12. N HCi (2. drops) was stirred at room temperature for 21 li, then at 50 °C for 5 h. The resultant suspension was diluted with EtOAc to give a homogeneous solution that was washed with brine, dried with Na^SC , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64042 (36.5 mg, 65%) as a white solid: ! H NMR (400 MHz, CDCh) δ 8.04 (s, 1 H), 5.97 (s, IH), 4.14 (d, H, J - 7.1 Hz), 4.10 (d, H, J - 7.1 Hz), 3.07 (d, I H, J - 4.7 Hz), 2.27 (m, 3H), 1.68 ( in. 1 IH), 1.50 (s, 6H), 1.26 fs, 3H), 1.26 (t, 3H, ./ 7.1 Hz), 1.24 (m, 4H), 1.18 (s, 3H), 1 .01 (m, 2H), 1.01 (s, 3H), 0.93 (s, 31 ! :·. 0.88 (s, 3H); m/z 548.3 (M+l ).

Compound TX04043: A mixture of TX63762 (52.4 mg, 0.101 mmol), /-PrOH (2 mL) and 12 N HCi (2 drops) was stirred at room temperature for 21 h, then at 50 °C overnight. The resultant suspension was diluted with EtOAc to give a homogeneous solution that was washed with 1 N HCI and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64043 (44.8 mg, 79%) as a white sohd: Tl NMR (400 MHz, CDCI3) 8 8.04 (s, I H), 5.97 (s, IH), 4.99 (sept IH, J = 6.3 Hz), 3.08 (d, IH, J = 4.7 Hz), 2.24 (m, 3H), 1 .73 (m, 10 H), 1.50 (s, 6H), 1.43 (m, IH), 1.26 (m, 3H), 1.22 (m, iOH), 1.18 (s, 3H), 1.03 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 562.3 (M+l).

{00086504} 74 Compound TX64058: A mixture of TX63762 (40.0 mg, 0.0770 mmoi), e-HexQH (2 mL) and 37% HC1 (2 drops) was stirred at 55 °C for 21 h. The resultant suspension was diluted with EtOAc to give a homogeneous solution that was washed with water and brine, dried with a2S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64058 (34.8 mg, 75%) as a white solid: 1 ! NMR. (400 MHz, CDC1 3 ) δ 8.04 (s, IH), 5.97 (s, IH), 4.73 (m. I I I ). 3.08 (d, IH, J = 4.7 Hz), 2.26 (m, 3H), 1 .77 (m, 12H), 1.55 (s, 3H), 1.50 (s, 3H), 1 .32 (m, 13H), 1.26 (s, 3H), 1 .1 8 (s, 3H), 1 .03 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 602.4 (M+l ).

Compound TX64046: A mixture of compound TX63762 (39.0 mg, 0.0750 mmoi), TEA (0.03 mL, 0.2 mmol), DMAP (18.1 mg, 0.148 mmol), phenol (22.5 mg, 0.239 mmol) and CH2CI2 (2 mL) was stirred at room temperature for 30 min. EDC1 (31.6 mg, 0.165 mmol) was added, and the reaction was stirred at room temperature for 18 h. The resultant mixture was diluted with EtOAc, washed with 1 N HC1 and brine, dried with Na2SC>4, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64046 (28.5 mg, 64%>) as a white solid: ] H NMR (400 MHz, CDCI 3 ) δ 8.04 (s, I H), 7.38 (t, 2H, J - 7.8 Hz), 7.23 (t, 1H, J - 7.4 Hz), 7.07 (m, 2H), 5.98 (s, IH), 3.1 1 (d, 1H, J = 4.7 Hz), 2.56 (m, 2H), 2.29 (td, 1H, J = 4.7, 13.0 Hz), 1.77 (m, I I H), 1 .49 (s, 6H), 1.28 (m, 4H), 1 .26 (s, 3H), 1.17 (s, 3H), 1.08 (m, 2H), 1 .03 (s, 3H), 0.96 (s, 3H), 0.90 (s, 3H); m/z 596.3 (M+l).

Compound TX64066: A mixture of compound TX63762 (53.8 mg, 0. 104 mmol),

TEA (0.05 mL, 0.4 mmol), DMAP (37.2 mg, 0.304 mmol), benzyl alcohol (0.06 mL, 0.7 mmol) and CiLCl? (2 mL) was stirred at room temperature for 15 min. EDCI (59.8 mg, 0.312 mmoi) was added, and the reaction was stirred at room temperature for 17 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCJ and brine, dried with N 280/j, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64066 (40.5 mg, 64%) as a white solid: Ή NMR (400 MHz, CDCI 3 ) δ 8.03 (s, IH), 7.35 (m, 5H), 5.97 (s, IH), 5.12 (d, IH, J = 12.3 Hz), 5.09 (d, IH, J - 12.3 Hz), 3.05 (d, IH, J - 4.7 Hz), 2.35 (m, 2H), 2.23 (td, IH, J - 4.7, 13.1 Hz), 1 .80 (m, 7H), 1.50 (m, 4H), 1.49 (s, M l ). 1.44 (s, 3H), 1 .26 (s, i n. 1.21 (m, 4H), 1.18 (s, 3H), 1.01 (m, 2H), 1.00 (s, 3H), 0.91 (s, 3H), 0.87 (s, 3H); m/z 610.3 (M+l).

Compound TX64067: A mixture of compound TX63762 (51.6 mg, 0.0993 mmol), TEA (0.05 mL, 0.4 mmol), DMAP (38.8 mg, 0.318 mmol), i-BuOH (0.10 mL, 1.0 mmol) and CH2CI2 (2 mL) was stirred at room temperature for 1 5 min. EDCT (60.7 mg, 0.31 7 mmol)

{00086504} 75 was added, and the reaction was stirred at room temperature for 17 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with Na. 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes), then purified by additional flash chromatography (Ci g silica gel, 0% to 100% MeCN in water) to give compound TX64067 (8.3 mg, 15%) as a white solid: Ή NMR (400 MHz, CDCI 3 ) δ 8.04 (s, IH), 5.97 (s, IH), 3.07 (d, I H, J = 4.7 Hz), 2.18 (m, 31 Π. 1.79 (m, 6H), 1.51 (s, 3H), 1.51 (m, 6H), 1.50 (s, 3H), 1 .44 (s, 9H), 1.26 (s, 3H), 1.24 (m, 3H), 1.1 8 (s, 3H), 1.02 (m, 2H), 1 .01 (s, 3H), 0.93 (s, 3H), 0.87 (s, M l ) : m/z 520.3 ( -r-Bu+H+1 ).

Compound TX64053: A mixture of compound T 3762 (70 mg, 0.14 mmol), TEA (0.06 mL, 0.4 mmol), DMAP (52.9 mg, 0.433 mmol), l -Boc-4-hydroxypiperidine (53 ,8 mg, 0.267 mmol) and CH 2 C1 2 (3 mL) was stirred at room temperature for 15 min. EDCI (78.1 mg, 0.407 mmol) was added, and the reaction w r as stirred at room temperature for 17 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gei, 0% to 100% EtOAc in hexanes) to give compound TX64053 (68.7 mg, 70%) as a white solid: Ή. NMR (400 MHz, ( IX h i δ 8.04 (s, IH), 5.98 (s, i l l ). 4.90 (m, IH), 3.72 (m, 21 ! }. 3.21 (m, 2H), 3.06 (d, 1 H, J = 4.6 Hz), 2.27 (m, 3H), 1.68 (m, 15H), 1.50 (s, 6H), 1 .46 (s, 9H), 1 .26 (s, 3H), 1.25 (m, 4H), 1.18 (s, 3H), 1.04 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 603.4 (M-Boc+H+1).

Compound TX64055: A mixture of TX64053 (21 .5 mg, 0.0306 mmol), 4 N HC1 in

1 ,4-dioxane (0.1 ml, 0.4 mmol) and CH 2 Cl 2 (0.9 mL) was stirred at room temperature for 5 h and cooled to -20 °C overnight. The resultant mixture was diluted with EtOAc, washed with brine, dried with Na 2 S0 4 , and concentrated to give compound TX640S5 ( 15.4 mg, 83%) as a white solid: J H NMR (400 MHz, CDCI 3 ) δ 8.04 (s, IH), 5.98 (s, I H), 4.83 (tt, I H, ./ - 4.2, 9.0 Hz), 3.07 (d, IH, J = 4.9 Hz), 3.05 (m, I H), 2.71 (m, 2H), 2.27 (m, 3H), 1.69 (m, 17H), 1 .50 (s, 6H), 1.26 (s, 3H), 1.25 (m, 4H), 1 .18 (s, 3H), 1.03 (m, 2H), 1 .01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 31 I K m/z 603.4 (M+l).

Compound TX640S6: A mixture of compound TX63762 (40.3 mg, 0.0775 mmol), TEA (0.04 mL, 0.3 mmol), DMAP (18.4 mg, 0.151 mmol), methoxyamme hydrochloride (26.6 mg, 0.318 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCI (30.8 mg, 0.161 mmol) was added, and the reaction was stirred at room temperature for 20 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica

{00086504} 76 gel 0% to 100% EtOAc in hexanes) to give compound TX64056 (24.1 mg, 57%) as a white solid: ] H NMR (400 MHz, CDCi 3 ) δ 8.04 (s, 1H), 5.97 (s, 1H), 3.76 (s, 3H), 3.09 (d, 1 H, J = 4.5 Hz), 2.23 (m, 2H), 2.04 (m, 2H), 1.67 (m, lOH), 1.52 (s. M l ). 1.50 (s, 3H), 1.26 (s, 3H), 1.24 (m, 4H), 1 .18 (s, 3H), 1.03 (m, 2H), 1 .00 (s, 3H), 0.92 (s, 3H), 0.88 (s, 3H); m/z 549.3 (M+l).

Compound TX63770: A mixture of compound TX63762 ( 108.4 mg, 0.209 mmol), TEA (0.06 mL, 0.4 mmol), DMAP (49.8 mg, 0.408 mmol), nethyiamine hydrochloride (29.3 mg, 0.434 mmol) and CH2CI2 (3 mL) was stirred at room temperature. EDCI (78,4 mg, 0.409 mmol) was added, and the reaction was stirred at room temperature for 19 li. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na?S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63770 (90.1 mg, 81%) as a white solid: Ή NMR (400 MHz, CDCI 3 ) δ 8.04 (s, 1H), 5.96 (s, 1H), 5.44 (s, 1H), 3.09 i d. i l l. .1 4.7 Hz), 2,80 (d, 3H, J - 4.8 Hz), 2.22 (td, 1H, J - 4.3, 13.5 Hz), 2.13 (t, 2H, J - 8.2 Hz), 1.67 (m, 1 1H), 1.53 (s, 3H), 1.50 (s, 3H), 1.26 (s, 3H), 1.24 (m, 4H), 1.18 (s, 3H), 1.02 (m, 2H), 1.00 (s, 3H), 0.92 (s, 3H), 0.87 (s, 3H); m z 533.3 (M+l).

Compound TX63768: A mixture of compound TX63762 (77.5 mg, 0.149 mmol), TEA (0.04 mL, 0.3 mmol), DMAP (35.5 mg, 0.291 mmol), ethyiamme hydrochloride (25 mg, 0.31 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 30 min. EDCI (55.5 mg, 0.290 mmol) was added, and the reaction was stirred at room temperature for 17 h. The resultant mixture was diluted with CH 2 C1 2 , washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63768 (56.6 mg, 70%) as a white solid: Ή NMR (400 MHz, CDCI 3 ) δ 8.02 (s, 1H), 5.96 (s, ! H), 5.34 (s, 1H), 3.27 (dq, 2H, J = 5.1 , 7.1 Hz), 3.10 (d, 1H, ,/ = 4.6 Hz), 2.24 (td, M i . J = 4.0, 13.1 Hz), 2.12 (t, 2H, J = 8.1 Hz), 1.68 (m, ΠΗ), 1.50 (s, 3H), 1 .48 (s, 3H), 1.26 (s, 3H), 1.24 (m, 4H), 1 .18 (s, 3H), 1.14 (t, 3H, J = 7.3 Hz), 1.03 (m, 2H), 1.02 (s, 3H), 0.94 (s, 3H), 0.89 (s, 3H); m/z 547.4 (M+l ).

Compound TX63764: A mixture of compound TX63762 (75.9 mg, 0.147 mmol), TEA (0.04 mL, 0.3 mmol), DMAP (35.9 mg, 0.294 mmol), 2,2,2-trifluoroethylamine hydrochloride (39.1 mg, 0.289 mmol) and C1¾C1? (2 mL) was stirred at room temperature for 10 min. EDCI (56.8 mg, 0.296 mmol) was added, and the reaction was stirred at room temperature for 17 h. The resultant mixture was diluted with CH 2 C1 2 , washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash

{00086504} chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63764 (66.4 mg, 76%) as a white solid: Ή NMR (400 MHz, CDCI 3 ) δ 8.03 (s, I H), 5.98 (s, 1H), 5.65 is, i 1 i ·. 3.91 (m, 2H), 3.07 (d, H i. J = 4.7 Hz), 2.25 (m, 1H), 2.23 (t, 2H, J - 8.1 Hz), 1 .71 (m, l iH), 1.52 (s, 3H), 1.51 (s, 3H), 1.26 (s, 3H), 1.26 (m, 4H), 1 .19 (s, 3H), 1.04 (m, 2H), 1.03 (s, 3H), 0.94 (s, 3H), 0.89 (s, 3H); m/z 601.3 (M+ 1 ).

Compound TX64064: A mixture of compound TX63762 (42.1 mg, 0.0810 mmol), TEA (0.04 mL, 0.3 mmoi), DMAP (21.2 mg, 0.165 mmoi), isopropylamine (0.02 mL, 0.2 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 20 min. EDCI (30.1 mg, 0.157 mmol) % r as added, and the reaction was stirred at room temperature for 16 h. Additional isopropylamine (0.05 mL, 0.6 mmol) and EDCI (50 mg, 0.26 mmoi) were added, and the reaction stirred overnight at room temperature. The resultant mixture was diluied with EtOAc, washed with 1 N HCl and brine, dried with >la 2 S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) and was then purified by additional flash chromatography (silica gel, 0% to 100% EtOAc in hexanes, each containing 0.5% TEA) to give compound ΤΧ64Θ64 (10.2 mg, 22%) as a white solid: ¾ NMR (400 MHz, CDCI 3 ) δ 8.04 (s, M Y). 5.96 (s, IH), 5.23 (d, 1 H, J = 8.0 Hz), 4.06 (m, I H), 3.10 (d, I H, J = 4.6 Hz), 2.22 (td, I H, J = 4.5, 13.1 Hz), 2.09 (t 2H, J = 8.1 Hz), 1.68 (m, 1 1 H), 1 .53 (s, 3H), 1.50 (s, 3H), 1.26 (s, 3H), 1 .24 (m, 4H), 1.1 8 (s, 3H), 1 .14 (m, 6H), 1.02 (m, 2H), 1.00 (s, 3H), 0.92 (s, 3H), 0.88 (s, 3H); m/z 561.3 (M+l ).

Compound ΤΧ64Θ65: A mixture of compound TX63762 (50.8 mg, 0.0977 mmol),

TEA (0.05 mL, 0.4 mmoi), DMAP (36.7 mg, 0.300 mmol), fer/--buiylamine (0.06 mL, 0.6 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCI (59,2 mg, 0.309 mmol) was added, and the reaction was stirred at room temperature for 17 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with Na 2 S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64065 (16.1 mg, 29%) as a white solid: Ή NMR (400 MHz, CDCL) 6 8.04 (s, IH), 5.96 (s, IH), 5.24 (s, IH), 3.08 i d. IH, J - 4.6 Hz), 2.22 (td, IH, ./ - 4.6, 13.2 Hz), 2.05 (t, 2 \ l. J 8.1 Hz), 1.67 (m, 1 1 H), 1.53 (s, 3H), 1.50 (s, 3H), 1.34 (s, 9H), 1.26 (s, 3H), 1.24 (m, 4H), 1.18 (s, 3H), 1.01 (m, 2H), 1.00 (s, 3H), 0.92 (s, 3H), 0.87 (s, 3H); m/z 575.4 (M+l).

Compound TX64057: A mixture of compound TX63762 (40.4 mg, 0.0777 mmol), TEA (0.04 mL, 0.3 mmol), DMAP (19.3 mg, 0.158 mmol), cyclohexylamine (0.04 mL, 0.4 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCI (29.8 mg,

{00086504} 78 0.155 mmoi) was added, and the reaction was stirred at room temperature for 21 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with a 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in liexanes) to give compound TX64057 (14.7 mg, 32%) as a white solid: : H NMR (400 MHz, CDCL) 5 8.04 (s, IH), 5.96 (s, 1H), 5.27 (d, 1H, J - 8.2 Hz), 3.74 (m, IH), 3.10 (d, IH, J = 4.6 Hz), 2.22 (td, I H, J = 4.5, 13.1 Hz), 2, 10 (t, 2H, J = 8.1 Hz), 1.45 (m, 271 ! }. L52 (s, 31 1 :·. 1.50 (s, 3H), 1.26 (s, 31 1 :·. 1.1 8 (s, 3H), 1.00 (s, 31 1 :·. 0.92 (s, 3H), 0.88 (s, 3H); m/z 601.4 (M+l ).

Compound TX64059: A mixture of compound TX63762 (40.3 mg, 0.0775 mmoi), TEA (0.04 mL, 0.3 mmoi), DMAP (19.3 mg, 0.158 mmoi), aniline (0.02 mL, 0.2 mmoi) and CH 2 CI? (2 mL) was stirred at room temperature for 20 min. EDC1 (30.5 mg, 0, 159 mmoi) was added, and the reaction was stirred at room temperature for 16 h. The resultant mixture was diluted with EtOAc, washed with 1 N HC1 and brine, dried with Na2SC> , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64059 (32.6 mg, 71%) as a white solid: ] H NMR (400 MHz, CDCI 3 ) δ 8.05 (s, IH), 7.49 (d, 2H, J - 8.0 Hz), 7.32 (t, 2H, J - 7.8 Hz), 7.16 (s, IH), 7.11 (t, LH, J = 7.4 Hz), 5.98 (s, IH), 3.09 (d, IH, J = 4.6 Hz), 2.34 (t, 2H, J = 8.2 Hz), 2.28 (td, I i 1. ./ 4.6. 13.1 Hz), 1.73 (m, 1 1H), 1.52 (s, i n. 1.49 s, 3H), 1.27 (m, 4H), 1.26 (s, 3H), 1.18 (s, 3M ). 1.06 (m, 2H), 1.02 (s, 3H), 0.94 (s, 3H), 0.89 (s, 31 1 ): m z 595.4 (M+l).

Compound TX64075: A mixture of compound TX63762 (49.2 mg, 0.0947 mmoi),

TEA (0.04 mL, 0.3 mmoi), DMAP (23.5 mg, 0.192 mmoi), benzylamine (0.02 mL, 0.2 mmoi) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 mm. EDCI (37,2 mg, 0.194 mmoi) was added, and the reaction was stirred overnight at room temperature. The resultant mixture was diluted with EtOAc, washed with 1 N HC1 and brine, dried with Na 2 SC>4, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64075 (14,4 mg, 25%) as a white solid: Ή NMR (400 MHz, CDCL) δ 8.04 (s, IH), 7.31 (m, 5H), 5.97 (s, IH), 5.69 (s, IH), 4.43 (m, 2H), 3.10 (d, IH, J - ---- 4.6 Hz), 2.24 (id, IH, J - ----- 4.1, 13.4 Hz), 2.18 (t, 2H, J = 8.2 Hz), 1.69

(m, 1 1H), 1.52 (s, 3H), 1.50 (s, 3H), 1.26 (s, 3H), 1.25 (m, 4H), 1.18 (s, 3H), 1.02 (m, 2H), 1.01 (s, 3H), 0.91 (s, 3H), 0.87 (s, 3H); m/z 609.4 (M+l ).

Compound ΤΧ64Θ60: A mixture of compound TX63762 (43.9 mg, 0.0845 mmoi), TEA (0.04 mL, 0.3 mmoi), DMAP ( 18.8 mg, 0.154 mmoi), dimethylamine hydrochloride (13.4 mg, 0.164 mmoi) and CH 2 Ci 2 (2. mL) was stirred at room temperature for 20 min.

{00086504} 79 EDCI (30.9 mg, 0.161 mmol) was added, and the reaction was stirred at room temperature for 16 h. The resultant mixture was diluted with EtOAc, washed with 1 N HC1 and brine, dried with Na 2 S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 1 00% EtOAc in hexanes) to give compound ΤΧ64Θ6Θ (35.6 mg, 77%) as a white solid: Ή NMR (400 MHz, CDC1 3 ) 8 8.04 (s, 1H), 5.96 (s, 1H), 3.10 (d, 1H, J - 4.6 Hz), 3.03 (s, 3H), 2.94 (s, 3H), 2.34 (ddd, i l l. J = 5.4, 1 1.3, 15.7 Hz), 2,22 (m, 2H), 1.67 (m, 1 IH), 1.51 (s, 3H), 1.49 (s, 3H), 1 .26 (s, M l ). 1.26 (m, 4H), 1.18 (s, 3H), 1 .02 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 547.3 (M+l).

Compound TX63950: A mixture of compound TX 3762 (521 mg, 1 .00 mmol), TEA (0.42 mL, 3.0 mmol), DMAP (250 mg, 2.05 mmol), azetidine hydrochloride ( 188 mg, 2,01 mmol) and CH2CI2 (20 mL) was stirred at room temperature for 15 min. EDCI (391 mg, 2.04 mmol) was added, and the reaction was stirred at room temperature for 25 h. The resultant mixture was diluted with EtOAc, w r ashed with 1 N HQ and brine, dried with and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX639S0 (394.6 mg, 71%) as an off-white solid: Ή NMR (400 MHz, CDCI3) δ 8.03 (s, I H), 5.96 (s, IH), 4.15 (m, 2H), 3.99 (t, 2H, J = 7.7 Hz), 3.10 (d, IH, J = 4.6 Hz), 2.26 (m, 3H), 1.77 (m, 13H), 1 .53 (s, 3H), 1.49 (s, 3H), 1.26 (s, 3H), 1.24 (m, 4H), 1 .17 (s, 3H), 1.01 (m, 2H), 1.00 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 559.3 (M+l ).

Compound TX63949: A mixture of compound TX63762 (520 mg, 1.00 mmol),

TEA (0.42 mL, 3.0 mmol), DMAP (241 mg, 1.97 mmol), pyrrolidine (0.17 mL, 2.06 mmol) and CH2CI2 (20 mL) was stirred at room temperature for 15 min. EDCT (381 mg, 1 .99 mmol) was added, and the reaction was stirred at room temperature for 25 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCi and brine, dried with a?S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give TX63949 (266.9 mg, 47%) as an off-white solid: Ή NMR (400 MHz, CDCI 3 ) δ 8.04 (s, IH), 5.96 (s, IH), 3.44 (t, 4H, J = 6.9 Hz), 3.12 (d, IH, 4.7 Hz), 2.22 (m, 3H), 1.71 (m, 15H), 1.53 (s, 3H), 1 .49 (s, 3H), 1.26 (s, 3H), 1.26 (m, 4H), 1.18 (s, M l ). 1.03 (m, il l s. 1.01 (s, 3H), 0.96 (s, 3H), 0.88 (s, 3H); m/z 573.4 (M+l).

Compound TX63763: A mixture of compound TX63762 (77.2. mg, 0.149 mmol),

TEA (0.04 mL, 0.3 mmol), DMAP (35.6 mg, 0.291 mmol), morpholine (25 μΐ,, 0.29 mmol) and CH2CI2 (2 mL) was stirred at room temperature for 10 min. EDCI (57.8 mg, 0.301 mmol) was added, and the reaction was stirred at room temperature for 1 6 h. The resultant

{00086504} 80 mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na?S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63763 (66. 1 mg, 82%) as a white solid: Ή NMR (400 MHz, CDC1 3 ) δ 8.04 (s, 1H), 5.97 (s, 1H), 3.68 (m, 4H), 3.60 (m, 2H), 3.48 (m, 2l h. 3.08 (d, 1H, J - 4.6 Hz), 2.33 (ddd, 1H, ./ - 5.4, 1 1.3, 16.4 Hz), 2.21 (m, 2H), 1.69 (m, 1 1 H), 1.50 (s, 3H), 1.49 (s, 3H), 1.26 (s, 3H), 1.26 fm, 4H), 1.18 (s, 3H), 1.04 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, M l ): m/z 589.4 (M+l).

Compound ΤΧ64Θ45: A mixture of compound TX63762 (39.2 mg, 0.0754 mmol), TEA (0.03 mL, 0.2 mmol), DMAP (19.5 mg, 0.160 mmol), acetic hydrazide (14.5 mg, 0.196 mmol) and CiLCl? (2 mL) was stirred at room temperature for 30 min. EDCI (31 .8 mg, 0.166 mmol) was added, and the reaction was stirred at room temperature for 18 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64045 ( 19.7 mg, 45%) as a white solid: ! H NMR (400 MHz, CDCI 3 ) δ 8.04 (s, 1 H), 8.00 (s, I I I ). 7.96 (s, 1 H), 5.98 (s, 1H), 3.07 (d, H i. J - 4.6 Hz), 2.24 (m, 3H), 2.06 (s, 3H), 1.68 (s, 1 1H), 1.51 (s, 3H), 1.50 (s, 3H), 1.25 (m, 4H), 1.26 (s, 3H), 1.1 8 (s, 3H), 1.03 (m, 2H), 1 .01 (s, 3H), 0.92 (s, 3H), 0.88 (s, 3W); m/z 576.3 (M+l).

Compound TX64049: A mixture of compound TX63762 (40.3 mg, 0.0775 mmol), TEA (0.03 mL, 0.2 mmol), DMAP ( 17.9 mg, 0.147 mmol), 3,3-difluoropyrrolidine hydrochloride (23.2 mg, 0.162. mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature foi ¬ l s min. EDCT (29.3 mg, 0. 153 mmol) was added, and the reaction was stirred at room temperature for 18 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64049 (27.9 mg, 59%) as a white solid: l B NMR (400 MHz, CDCI 3 ) δ 8.04 (s, 1H), 5.97 (s, 1H), 3.75 (m, 4H), 3.09 (t, 1H, J - 4.4 Hz), 2.32 (m, 5H), 1.70 (m, 1 1 H), 1.52 (s, 3H), 1 .50 (s, 3H), 1.26 (s, 3H), 1.26 (m, 4H), 1.18 (s, 3H), 1.03 (m, 2H), 1 .01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 609.3 (M+l).

Compound ΤΧ64Θ47: A mixture of compound TX63762 (40.2 mg, 0.0774 mmol),

TEA (0.03 mL, 0.2 mmol), DMAP (17.9 mg, 0.147 mmol), (S)-3-hydroxypyrrolidine hydrochloride (34.1 mg, 0.276 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCI (28.5 mg, 0.149 mmol) was added, and the reaction was stirred at room

{00086504} 81 temperature for 18 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na. 2 S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64047 (20.8 mg, 46%) as a white solid: H NMR (400 MHz, CDClj) 8 [8.04 (s), 8.03 (s) (1 : 1 , l H)j, [5.97 (s), 5.96 (s) (1 : 1 , IH)], 4.52 (s, IH), 3.58 (m, 4H), [3.12 (d, J - 4.5 Hz), 3.09 (d, J - 4.5 Hz) (1 : 1 , IH)], 1.87 (m, 17H), [1.53 (s), 1.52 (s) (1 : 1 , 3H)], [1.49 (s), 1.50 (s) (1 : 1 , 3H)j, 1.26 (s, 3H), 1 .18 (s, 3H), 1.07 (m, 6H), 1.00 (s, 3H), [0.95 (s), 0.93 (s) (1 : 1 , 31 ! :· ]. 0.88 (s, 3H); .wz 589.3 (M+l ).

Compound TX64048: A mixture of TX63762 (40.1 mg, 0.0772 mmol), TEA (0.03 mL, 0.2 mmol), DMAP (19.2 mg, 0.157 mmol), 4-hydroxypiperidine ( 16.5 mg, 0.163 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCi (28.4 mg, 0.148 mmol) was added, and the reaction was stirred at room temperature for 18 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na2SC>4, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64048 (23.9 mg, 51%) as a white solid: ] H NMR (400 MHz, C DC! . } δ 8.04 (s, IH), 5.97 (s, I H), 4.10 (m, I H), 3.95 (m, I H), 3.72 (m, 21 ! }. 3.22 (m, 2H), 3.09 (s, IH), 2.29 (m, 5H), 1 .69 (m, 13H), 1 .51 (s, 3H), 1 .50 (s, 3H), 1 .26 (m, 4H), 1.26 (s, 3H), 1.18 (s, 3H), 1.03 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 603.4 (M+l).

Compound TX64068: A mixture of compound TX63762 (51.3 mg, 0.0987 mmol),

TEA (0.05 mL, 0.4 mmol), DMAP (37.2 mg, 0.304 mmol), 2-aminopyridine (52.5 mg, 0.558 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCi (59,2 mg, 0.309 mmol) was added, and the reaction was stirred at room temperature for 17 h. The resultant mixture was directly purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes, each containing 0.5% TEA), then was purified by additional flash chromatography (Qg silica gel, 0% to 100% MeCN in water) to give compound TX64068 (7.0 mg, 12%) as a white solid: ! H NMR (400 MHz, CDC1 3 ) δ 8.26 (d, IH, J = 4.8 Hz), 8.16 (d, IH, ./ = 8.5 Hz), 8.04 (s, I H), 7.83 (s, I H), 7.69 (m, IH), 7.04 (dd, H i. J = 4.8, 7.3 Hz), 5.98 (s, IH), 3.10 (d, IH, .7 - 4.7 Hz), 2.36 (m, 2H), 2.26 (id, IH, J = 4,7, 13,4 Hz), 1.75 (m, 1 I H), 1 .52 (s, 3H), 1 .50 (s, 3H), 1.26 (m, 4H), 1 .26 (s, 3H), 1 .18 (s, 3H), 1.05 (m, il l s. 1 .02 (s, 3H), 0.94 (s, 3H), 0.89 (s, 3H); m/z 596.4 (M+l).

Compound ΤΧ64Θ44: A mixture of compound TX63762 (39.7 mg, 0.0764 mmol), TEA (0,03 mL, 0,2 mmol), DMAP (20.3 mg, 0.166 mmol), 3 -oxeiananiine hydrochloride

{00086504} 82 (17.0 mg, 0.155 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 30 min. EDCl (30.6 mg, 0.160 mmol) was added, and the reaction was stirred at room temperature for 8 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S0 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound ΤΧ64Θ44 (28.5 mg, 65%) as a white solid: Ή NMR (400 MHz, CDCL) δ 8.04 (s, 1H), 5.97 (s, 1 H), 5.94 (s, 1H), 6.74 (m, 1H), 4.92 (dt, 2H, J = 2.3, 7.1 Hz), 4.48 (dt, 2H, J = 2.9, 6.4 Hz), 3.06 (d, 1 H, J = 4.6 Hz), 2.20 (m, 3H), 1.69 (m, 1 1H), 1.51 (s, M l ) . 1 .50 (s, 3H), 1.26 (s, 3H), 1.25 (m, 4H), 1 .18 (s, 3H), 1.02 (m, 2H), 1.00 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 575.3 (M+l ).

Compound TX64052: A mixture of compound TX63762 (71 mg, 0.137 mmol),

TEA (0.06 mL, 0.4 mmol), DMAP (46.2 mg, 0.378 mmol), glycine methyl ester hydrochloride (33.9 mg, 0.270 mmol) and CH 2 C1 2 (3 mL) was stirred at room temperature for 15 min. EDCT (78.1 mg, 0.407 mmol) was added, and the reaction was stirred at room temperature for 16 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64052 (52.0 mg, 64%) as a white solid: [ ll NMR (400 MHz, CDCL,) δ 8.04 (s, 1H), 5.97 (s, 1H), 5.95 (s, 1H), 4.04 (dd, 2H, J - 5.1 , 9.2 Hz), 3.77 (s, 3 i n. 3.08 (d, 1H, J - 4.7 Hz), 2.22 (m, 3H), 1.67 fm, I IH), 1.52 fs, 3H), 1.50 (s, 3H), 1.26 (s, 3H), 1.25 (m, 4H), 1.18 (s, 3H), 1.03 (m, 2H), 1.00 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 591.3 (M+l ).

Compound ΤΧ64Θ54: A mixture of compound TX64052 ( 20.9 mg, 0.0354 mmol), 1 N HCl (0.6 mL), and MeCN (1.2 mL) was heated to 55 °C for 22 h. The resultant mixture was diluted with EtOAc, washed with brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes, each with 0.5% HOAc) to give compound ΤΧ64Θ54 (14.6 mg, 71%) as a white solid: Ή NMR (400 MHz, CDClj) 8 8.04 (s, lH), 6.17 (s, i l l ). 5.99 (s, IH), 4.09 (dd, 2H, J - ----- 5.0, 9.7

Hz), 3.07 (d, H i. ./ 4.6 Hz), 2.24 (m, 3H), 1.68 (m, I IH), 1 .51 (s, 3H), i .50 (s, 3H), 1.26 (s, 3H), 1.23 (m, 4H), 1.18 (s, 3H), 1.03 (m, 2H), 1.01 (s, 3H), 0.91 (s, 3H), 0.88 (s, 3H); m/z 577.3 (M+l).

Compound TX64124: A mixture of compound TX63762 (47.8 mg, 0.0920 mmol),

TEA (0.04 mL, 0.29 mmol), DMAP (23.5 mg, 0.192 mmol), 4-Boc-piperazine (36.3 mg, 0.195 mmol) and CH 2 C1 2 (2 mL) was stirred at room temperature for 15 min. EDCl (36.8 mg, 0, 192 mmol) was added, and the reaction was stirred at room temperature for 16 h. The

{00086504} 83 resultant mixture was diluted with EtOAc, washed with 1 N HCT and brine, dried with Na. 2 S04, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX64124 (55.9 mg, 88%) as a white solid: ¾ NMR (400 MHz, CDCLj 8 8.04 (s, 1H), 5.97 (s, i l l ). 3.57 (m, 2H), 3.42 (m, 61 1 :·. 3.08 (d, 1 H, J - 4.6 Hz), 2.35 (m, 1 H), 2.23 (m, 2H), 1 .68 (m, 1 1 H), 1 .50 (s, 3H), 1 .49 (s, 3H), 1 .47 (s, 9H), 1.27 (m, 4H), 1.26 (s, 3H), 1.18 (s, 3H), 1.01 is, 3H), 1.00 (m, 2H), 0.93 (s, 31 ! }. 0.88 (s, 3H); m/z 588.4 (M-Boc+H+1).

Compound TX64135: HQ (4.0 M in 1 ,4-dioxane, 0.2 mL) was added to a room temperature solution of compound TX64124 (49 mg, 0.071 mmol) in CH 2 C1 2 (5 mL), and the mixture was stirred at room temperature. Additional HC1 solution (4.0 M in 1 ,4-dioxane) was added after 17 h (0.5 mL), 20 h (0.5 mL) and 2 d (2.0 mL). The resultant mixture was diluted with EtOAc, washed with 1 N NaOH and brine, dried with Ma 2 S04, and concentrated to give compound TX64135 (14.4 mg, 35%) as an off-white solid: 'H NMR (400 MHz, CDCL) δ 8,04 (s, 1H), 5.97 (s, IH), 3.62 (m, 5H), 3,09 (d, 1H, J = 4.7 Hz), 2.86 (app. td, 4H, ./ 5.1 , 17.7 Hz), 2.34 (m, lH), 2.22 (m, 2H), 1.68 (m, 1 1H), 1 .51 (s, 3H), 1.49 (s, 3H), 1 .26 (s, 3H), 1 .24 (m, 4H), 1.1 8 (s, M l ). 1.03 (m, 21 ! ) 1.01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 588.4 (M +1).

Compound TX64074: A mixture of compound TX63762 (51.9 mg, 0.0999 mmol), NH4CI (18.0 mg, 0.337 mmol), EDCI (28.7 mmol, 0.150 mmol), HOBt-xH 2 0 (23 mg, 0.17 mmol), DIEA (0.03 mL, 0.2 mmol) and DMF (1 mL) was stirred at room temperature for 17 h. The resultant mixture was diluted with EtOAc, washed with 1 N HCT and brine, dried with N 2SO/|, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes, each containing 0.5% TEA) to give compound TX64074 (25.3 mg, 49%) as a white solid: 1 ! NMR (400 MHz, CDCI 3 ) δ 8.04 (s, IH), 5.97 (s, 1 1 ! ) 5.39 (s, 1H), 5.24 (s, IH), 3.08 (d, IH, J = 4.7 Hz), 2.21 (m, 3H), 1.70 (m, 1 1H), 1.51 is, 3H), 1.50 (s, 3H), 1.26 (s, 3H), 1.24 (m, 4H), 1 . 18 (s, 3H), 1.03 (m, 2H), 1.01 (s, 3H), 0.93 (s„ 3H), 0.88 (s, 3H); w/z 519.3 (M+i).

Compound 13: Compound 13 was synthesized as reported in U.S. Patent No. 7,943,778, which issued on May 17, 201 1, the entirety of which is incorporated herein by reference.

Compound 14: The flask containing a suspension of compound 13 (4,81 g, 10.1 mmol), Pd/C (10% w/w, 642 mg), EtOAc (100 mL), and C¾C1 2 (100 mL) was thoroughly purged with N 2 , followed by H 2 . The resultant mixture was stirred vigorously for

{00086504} 84 2 h, sparged with N? for ~2 h, stirred overnight at room temperature, filtered through a plug of ceiite (3 cm) eluting with CH 2 G 2 (200 mL), and concentrated to give compound 14 (5.0 g) as a pale yellow foam that was used without further purification: m/z 480.3 (M+l).

Compound 15: A suspension of compound 14 (all above obtained, < 10.1 mmol), HBr (48% w/w aq., 0.49 mL, 4.4 mmol), and MeCN was heated to 35 °C for 30 min. Br 2 (0.62 mmol, 12.1 mmol) was added, and the resultant mixture was heated to 35 °C for an additional 17 h. The solution was cooled to room temperature, 10% Na 2 S03 (50 mL) added, and the biphasic mixture was stirred for 15 min at room temperature. The mixture was diluted with EtOAc. The organic fraction was separated, washed with a 1 : 1 mixture of 10% and sat. NaHC(¼ and brine, dried with Na 2 S0 4 , and concentrated to give compound IS (5.1 g) as a yellow foam that was used without further purification: m/z 478.3 (M+l).

Compound 16: NaOMe (25% w/w in MeOH, 3.00 mL, 13.1 mmol) was added to a suspension of compound 15 (ail above obtained, < 10.1 mmol) in MeOH ( 100 mL), and the resultant mixture heated to 50 °C for 2. li. The yellow solution was cooled to room temperature, diluted with MTBE, washed with 1 M HCf and brine, dried with Na 2 S0 4 , and concentrated to give compound 16 (5.15 g) as a yellow foam that was used without further purification: m/z 478.3 (M+l ).

Compound TX63403: A solution of l,3-dibromo-5,5-dimethylhydantoin (1.445 g, 5.05 mmol) in DMF (12 mL) was added over 5 min to a 0 °C solution of 30 (all above obtained, < 10.1 mmol) in DMF (40 mL). The vial containing the DBDMH solution was washed with an addition 8 mL of DMF, and that solution was added to the reaction. A fter 1 h at 0 °C, pyridine (2.45 mL, 30.3 mmol) was added, and the reaction was heated to 55 °C for 4 h. The resultant solution was cooled to room temperature, diluted with EtOAc, washed with 10% Na 2 S03, water, 1 N HC1, and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 42% EtOAc in hexanes), and the resultant product was triturated with EtOH (50 mL) at 50 °C for 30 min to give compound TX63403 (3.01 g, 63% from 13) as a white solid: 1 H N MR (500 MHz, CDCI 3 ) δ 8.05 (s, 1H), 5.97 (s, 1 1 1 !. 3.02 (d, M i . J = 4.7 Hz), 2,23 (id, 1H, ./ - 4.3, 13.5 Hz), 1.78 (m, 6H), 1.54 (m, 3H), 1.50 (s, 31 1 :·. 1 .46 (s, 3H), 1.40 (m, I l l s. 1 .26 (s, 3H), 1.22 (m, 5H), 1 .18 (s, 3H), 1.03 (m, 2H), 1.00 (s, 3H), 0.92 (s, 3H), 0.87 (s, 3H), 0.83 (t, 3H, J - 7.4 Hz); m/z 476.3 (M+l ).

Compound TX63868: Compound TX63355 (500 mg, 1.05 mmol) and 1 , 1 '-carbonyldiimidazole (221 mg, 1.36 mmol) were dissolved in MeCN (10 mL). The reaction was heated at reflux for 5 li and was then cooled to room temperature. The

{00086504} 85 precipitate was collected by filtration and was then washed with MeCN and dried under vacuum to give compound TX63868 (446 mg, 75%) as a white solid: ¾ NMR (400 mHz, (ΊΧΊ·.) δ 8.15 (s, 1H), 8.03 (s, iH).7.42 (s, III).7.10 (s, ill}.6.00 (s, 1H), 4.43 (d, 1H, J =

10.8 Hz), 4.36 (d, IH, ./ 10.8 Hz), 3.00 id. IH, J - 4.7 Hz), 2.44 (m, ill!.2.00 (ddd, 111. J = 4.3, 13.9, 13.9 Hz), 1.72-1.90 (m, 7H), 1.58 (m, IH), 1.52 (s, 3H), 1.51 (s, 3H), 1.26 (s,

3H), 1.18 (s, 3H), 1.15-1.44 (m, 6H), 1.05 (s, 3H), 0.97 (s, 3H), 0.91 (s, 3H); m/z 572.3 (M+l).

Com o nd TX63926: A mix Sure of compound TX6335S (200 mg, 0.42 mmol), 4- morrdroiinecarbonyi chloride (0.15 raL, 1.28 mmol), DMAP (5 mg, 0,041 rxmiol), and pyridine (1 mL was stirred at ri for 30 mm, then at 90 °C for 18 h. The reaction was cookd to rt and Ac ; ?0 (0.2 mL) was added. After the reaction was stirred for 30 mis, aq. NaHCCA solution was added. The reaction was stirred for another 30 min and was then extracted wish EiOAc. The combined organic extracts were washed with 1 HC1 and then NaHCCb solution, dried with Na?S0 , and concentrated. The residue was purified by column chromatography (silica gel, 0% to 45% EtOAc in hexanes) to give compound TX63 26 (122 mg, 49%) as a white foam: Tl NMR δ 8.06 (s, IH), 6.00 (s, IH), 4.20 (d, IH, ./ 11.0 Hz), 4.04 (d, 1H,J= 11.0 Hz), 3.69 (hs, 4H), 3.51 (hs, 4H), 3.07 (d, IH, J = 4.6 Hz), 2.42 (m, IH), 1.72-1.98 (m, 7H), 1.61 (m, if 11 1.53 (s, 3H), 1.52 (s, 3H), 1.49 (in, IH), 1.28 (s, 3H), 1.20 (s, 3H), 1.08-1.36 (m, 6H), 1.04 (s, 3H), 0.96 (s, 3H), 0.91 fs, 3H); m/z 591.4 (M+i).

Com ound TX63927: A mixture of compound TX63355 (200 mg, 0.42 mmol), 1- pyrrolidinecarbonyi chloride (0.14 mL, 1.27 mmol), DMAP (5 mg, 0.0 1 mmol} and pyridine (1 mL) was stirred at rt for 30 min, and then at 90 °C for 18 h. The reaction was cooled to rs, and Ac 2 0 (0.2 mL) was added. After the reaction was stirred for 30 min, aq. aHCCh solution was added. The reaction was stirred for another 30 min and was then extracted with EtOAc. ' The combined organic extracts were washed with i N HQ and NaHC A solution, dried with a2S0 , and concentrated. The residue was purified by column chromatography (silica gel, 0% to 40% EtOAc in hexanes) to give compound TX63927 (85 mg, 35%) as a white foam: 'HNMR δ 8.07 (s, IH), 5.99 (s, IH), 4.20 (d, IH, J= 10.9 Hz), 3.94 (d, 1H,J =

10.9 Hz), 3.40 (in, 4! if 3.09 (d, !ii. J= 4.6 Hz), 2.48 (m, if If 1.72-2.00 (m, 1 IH), 1.60 (m, ill). 1.53 (s, 311;·.1.52 (s, Ml).1.49 (rn, IH), 1.28 (s, 3H), 1.20 (s, 3H), 1.08-1.36 (m, 6H),

1.04 (s, 3H), 0.97 (s, 3H), 0.91 (s, Us; m/z 575.4 (M+l).

Comp und ΪΧ63.93Θ: A mixture of compound TX63355 (200 mg, 0.42 mmol), ethyl isocyanate ( 0,33 rnL, 4.17 mmol), and toluene (1 mL; was stirred at rt for 30 min, and (hen at 90 °C for 16 h. The reaction was cooled to t and was purified directly by column

100086504} 86 rng, 85% yield? as a white foam: U NMR δ 8.07 (s, lH), 6.00 (s, 1H), 4.70 (m, 1H), 4.12 (d,

IH, J = 11.0 Hz), 4.00 (d, 1H, J = 11.0 Hz), 3.24 (m, 2H), 3.08 (d, 1H, J = 4,4 Hz), 2.42 (m, 1H), 1.53 (s, 3H), 1.52 (s, 3H), 1 .28 (s, 3H), 1.20 (s, 3H), 1.17 (t, 3H, J= 7.3 Hz), 1.08-1.97 (m, 15H), 1.04 (s, 3H), 0.97 (s, 3H), 0.90 (s, 3H); /n/ir 549.3 (M-H ).

Compound 17: LiAlH 4 (2.0 M in THF, 100 mL, 200 mmol) was added to a 0 °C solution of oleanolic acid (25.1 g, 55.0 mmol) in THF (1.25 L). The mixture was heated to reflux for 3 h, cooled to room temperature overnight, cooled to 0 °C, quenched by the successive addition of water (10.6 mL), 4 M aOH (10.6 mL) and water (10.6 mL), warmed to room temperature over 15 min, filtered through celite, eluted with MTBE, and concentrated to give compound 17 as a white solid that was used without further purification: m /z 443.4 (M+l).

Compound 18: A solution of NaOCl (6.0%, 80 mL, 65 mmol) and water (180 mL) was added to a 0 °C triphasic solution of compound 17 (all above obtained, < 55.0 mmol), NaHC0 3 (4.65 g, 55.4 mmol), NaBr (5.68 g, 55.2 mmol), TEMPO (4.30 g, 27.5 mmol), water (360 mL) and CH2CI2 (1 L) over 1 h. The mixture was warmed to room temperature over 16 h, quenched by the addition of 10% I^SOs and stirred for 15 min. The organic fraction was separated, and the aqueous fraction was extracted with CH 2 C] 2 . The combined organic fraction was washed with brine, dried with Na 2 SC>4, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 15% EtOAc in hexanes, then 10% EtOAc in CH 2 C1 2 ) to give compound 18 (19.22 g, 79% from oleanolic acid) as an off-white solid: m/z 441.4 (M÷l ).

Compound 19: KO¾u (6.36 g, 56.8 mmol) was added to a room temperature suspension of (niethoxyniethyl)triphenylphosphonium chloride (23.36 g, 68.14 mmol) in THF (1 15 mL) and stirred at room temperature for 45 min. A solution of 19 (5.00 g,

I I.4 mmol) in THF (85 mL) was added over 10 min to the reaction mixture, and the transfer was completed with THF (30 mL). The reaction was stirred at room temperature for 21 h, then quenched by the addition of H 2 S0 4 (30%, 23 mL), stirred 2 h, diluted with EtOAc and water, and made basic with 4 N NaOH. The organic fraction was separated, washed with brine, dried with Na 2 SC>4, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 30% EtOAc in hexanes) to give compound 19 (4.93 g, 96%) as a pale yellow solid: m/z 455.4 (M+l).

Compound 20: NaBH 4 (856 mmol, 22.6 mmol) was added to a 0 °C solution of compound 19 (4.93 g, 10.8 mmol) in MeOH/THF (2: 1 mixture, 300 mL), and the reaction

{00086504} 7 was allowed to warm to room temperature. After 2 h at room temperature, the mixture was concentrated to reduced volume, diluted with EtOAc, washed with 1 N HCl and brine, dried with Na 2 S0 4 , and concentrated to give compound 20 (6 g, quantitative) as a white solid: m/z 457.4 (M+1 ).

Compound 21 : A solution of 28 (all above obtained, < 10.8 mmol), Ac 2 0 (8.2 mL,

87 mmol), pyridine (10.5 mL, 130 mmol), and DMAP (133 mg, 1.09 mmol) in CH 2 C1 2 (225 mL) was stirred at room temperature. Additional Ac 2 0 (4.1 mL, 43 mmol), pyridine (5.25 mL. 64.9 mmol), and DMAP ( 133 mg, 1.09 mmol) were added after 7 h and again after an additional 16 h. The reaction was stirred an additional 3 d, then diluted with CH 2 CI 2 , washed with 1 HCl, saturated NaHCOj and brine, dried with a 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 30% EtOAc in hexanes) to give compound 21 (4.40 g, 75% from 19) as a white foam solid.

Compound 22: AcOOH (39% in AcOH, 2.1 mL, 12.4 mmol) was added to a 50 °C suspension of 21 (4.40 g, 8.14 mmol) and Na 2 CC>3 (1.13 g, 10.7 mmol) in AcOH (81 mL). Additional AcOOH (0.84 mL, 5.0 mmol) was added after 17 h and again (0.2.1 mL, 1.2 mmol) after an additional 8 h. The reaction was stirred at 50 °C for an additional 18 h, quenched by the addition of excess Na 2 S0 3 , stirred 15 min, diluted with toluene, and concentrated to dryness. The residue was dissolved in CH 2 C1 2 , washed with water, saturated aHCOs and brine, dried with Na 2 S0 4 , and concentrated to give compound 22 (4.36 g, 96 %) as a white solid: m/z 557.4 (M+1).

Compound 23: Br 2 (0.60 mL, 12 mmol) was added to a. 35 °C suspension of compound 22 (5.41 g, 9.72 mmol) and HCl (4.0 M. in 1 ,4-dioxane, 0.97 mL, 3.9 mmol) in MeCN (96 mL). The mixture was stirred overnight between room temperature and 35 °C. The mixture was heated to 35 °C, and additional Br 2 (0, 15 mL, 2.9 mmol) was added and again added (0.30 mL, 5.8 mmol) after an additional 1 h. The reaction was stirred at 35 °C for an additional 1 h, then quenched by the addition of 4% Na 2 SO¾ (100 mL) and stirred at room temperature for 30 min. The organic fraction was washed with saturated NaHCOr, and brine, dried with Ma 2 S0 4 , and concentrated to give compound 23 as a yellow solid: m/z 555.4 (M+i).

Compound 24: A solution of compound 23 (all above obtained, < 9.72 mmol) and concentrated H 2 S0 4 (0.1 mL) in MeOH/THF ( 1 : 1 mixture, 200 mL) was heated to 35 °C for 1 h. Additional H 2 S0 4 (0.9 ml) was added, and the reaction was then brought to reflux for 19 h. The mixture was concentrated to reduced volume, then diluted with EtOAc, washed with 1 N NaOH and brine, dried with Na. 2 S0 4 , and concentrated. The residue was purified by flash

{00086504} o chromatography (silica gel, 0% to 65% EtOAc in CH 2 C1 2 ) to give compound 24 (3.41 g, 74% from 22) as a pale-yellow solid: m/z 471.4 (M+ l).

Compound 25: A solution of NaOCI (6%, 6.5 mL, 5.2 mmol) was added to a room temperature solution of compound 24 (1.70 g, 3.61 mmol) in AcOH (27.5 mL), and the reaction mixture was stirred at room temperature for 1.5 h. The reaction was diluted with EiOAe, washed with 10% Na 2 S(½ and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 55% EtOAc in CH 2 C1 2 ) to give compound 25 (1.36 g, 80%) as a white solid: m/z 469.3 (M+ l).

Compound 26: NaOMe (25% in MeOH, 6.5 mL) was added to a 0 °C solution of compound 25 (1.36 g, 2.90 mmol) in ethyl formate (16.5 mL). The mixture was stirred at 0 °C for 15 m n, then warmed to room temperature. After 7 the mixture was diluted with EtOAc, washed with 1 N HC1 and brine, dried with Na 2 S0 4 , and concentrated to a give compound 26 (plus mixture of formate esters) as a yello foam: m/z 497.3 (M+l).

Compound 27: A mixture of compound 26 (all above obtained, < 2.90 mmol) and NH?OH-HQ in EtOH water was heated to 55 °C for 17 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with a2,S04, and concentrated to give compound 27 as a yellow foam: m/z 494.4 (M+l ).

Compound 28: A solution of 27 (all above obtained, < 2.90 mmol) and NaOMe (25% in MeOH, 1.7 mL) in MeOH (39 mL) was heated to 55 °C for 6 h. The mixture was cooled to room temperature overnight, acidified with HC1 (4 M in 1,4-dioxane, 10 mL), and heated to 55 °C for 7 h. The resultant mixture was diluted with EtOAc, washed with 1 N HC1 and brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound 28 (1.01 g, 71% from 25) as a pale-yello foam solid: m/z 494.3 (M+l).

Compound TX63608: A solution of l ,3-dibromo-5,5-dimethylliydantoin (309 mg,

1.08 mmol) in DMF (3 mL) was added to a 0 C C solution of compound 28 (1.01 g, 2.05 mmol) in DMF (15 mL), with additional DMF (2 mL) used to complete the transfer. The mixture was stirred at 0 °C for 3.5 h, then pyridine (0.66 mL, 8.2 mmol) was added, and the reaction was heated to 55 °C for 18 h. The resultant solution was diluted with EtOAc, washed with 1 N HQ, 5% Na 2 S03, and brine, dried with Na?804, and concentrated. The residue was purified by column chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63608 (805 mg, 80 %) as an off-white solid: Ή NMR (400 MHz, CDC1 3 ) 8 8.04 (s, 1H), 5.98 (s, 1H), 3.78 (m, 2H), 3.1 (d, 1H, J = 4.7 Hz), 2.25 (td, i l l. ./ =

{00086504} 4.0, 13.6 Hz), 1.65 (m, 121 ! }. 1.50 (s, 3H), 1.50 (s, 3l h. 1.26 (s, 3H), 1.25 (m, 4H), 1.1 8 (s, M l ). 1.06 (m, 11 1 ). 1.01 (s, 3H), 0.93 (s, 3H), 0.87 (s, 3H); m/z 492.3 (M+l ).

Compound TX63609: A solution of compound T 63608 (50.5 mg, 0.103 mmol), MeOTf (57 μΕ, 0.50 mmol) and 2,6-di-ieri-butyl-4-methylpyridine (128 mg, 0.623 mmol) in CH2CI2 (2 mL) was stirred at room temperature for 19 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ and brine, dried with Na 2 S04, and concentrated. The residue was purified by column chromatography (silica gel, 0% to 1 00 % EtOAc in hexanes) to give compound TX63609 (39.5 mg, 76 %) as a white solid: ¾ NMR (400 MHz, CDCI3) 8 8.04 fs, lH), 5.97 (s, I i n. 3.45 (m, 2H), 3.32 (s, 3H), 3.09 (d, 1H, ./ 4.7 Hz), 2,25 (id, H i. J - ---- 4.1 , 13.4 Hz), 1.66 (m, 1 1H), 1 .50 (s, 3H), 1.48 fs, 3H), 1 .26 (s, 3H), 1.25 (m, •U n. 1.18 is, 3H), 1.05 (m, 2H), 1.01 (s, 3H), 0.94 (s, 3H), 0.87 (s, 3H); m/z 506.3 (M+l ).

Compound TX63610: A solution of compound TX 63608 (49.6 mg, 0.101 mmol), Ac 2 0 (48 μΕ, 0.51 mmol), pyridine (81 uL, 1.0 mmol) and DMAP (3.4 mg, 0.028 mmol) in CH2CI2 (2 mL) was stirred at room temperature for 20 h. The resultant mixture was diluted with EtOAc, washed with 1 N HQ, saturated NaHCO;,, and brine, dried with Na?S0 4 , and concentrated. The residue was purified by column chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63610 (41 . 1 mg, 76%) as a white solid: Ή NMR (400 MHz, CDCI 3 ) 8 8.04 (s, 1 H), 5.98 (s, 1 H), 4.16 (m, 2H), 3.08 (d, H i. J - ----- 4.7 Hz), 2,25

(td, H i. J - 4.0, 1 3.5 Hz), 2.03 (s, 3H), 1 .66 (m, l lH), 1 .50 (s, 3H), 1.49 (s, 3H), 1.26 (s, 3H), 1.25 (m, 4H), 1.18 (s, 3H), 1 .07 (m, 2H), 1.01 (s, 3H), 0.93 (s, 3H), 0.87 (s, 3H); m/z 534.3 (M+l).

Compound TX63981 : A solution of compound TX 3608 (41.4 mg, 0.0842 mmol) and EtNCO (64 uL, 0.81 mmol) in toluene (0.5 mL) was stirred at room temperature for 1 h, then heated to 70 °C for >4 h. The reaction mixture was cooled to room temperature and was purified directly by column chromatography (silica gel, 0% to 100 % EtOAc in hexanes) to give compound TX63981 (34.5 mg, 73%) as a white solid: ! H NMR (400 MHz, CDCI3) δ 8.04 (s, 1H), 5.97 (s, IH), 4.58 (s, 1 H), 4.23 (m, 1H), 4.12 (m, 1H), 3.19 (m, 2H), 3.09 (d, 1 H, J - 4.6 Hz), 2.30 (td, 1 H, J - 4.1 , 13.2 Hz), 1.67 (m, 1 1 H), 1 .50 (s, 3H), 1.49 i s, 3H), 1.26 (s, 3H), 1.19 (m, 6 i n. 1.1 8 (s, 3H), 1.12 (t, 3H, J ------ 12 Hz), 1 .01 (s, 3H), 0.94 (s, 3H), 0.88 (s, 3H); m/z 563.4 (M+l).

Compound 29: D1BAL-H (1.0 M in THF, 5 mL, 5.0 mmol) was added to a 0 °C solution of 8a, 8b (8a:8b = 2:3, 0.50 g, 0.92 mmol) in THF (10 mL). The reaction mixture was stirred at 0 °C for 30 min, then warmed to room temperature for 2.5 h. The mixture was

{00086504} 90 cooled to 0 °C, quenched with saturated NaK tartrate (10 mL), diluted with MTBE (25 mL), warmed to room temperature over 1 h, diluted with additional saturated NaK tartrate (40 mL), then and extracted with MTBE, The combined organic fractions were washed with brine, dried with Na?S04, filtered through a short plug of celite, eluted with MTBE, and concentrated to give compound 29 (491 mg, mixture of C12-epimers, quantitative) as a white solid: m/z 492.3 (M+l).

Compound 30: NBS (244 mg, 1.37 mmol) was added to a room temperature solution of compound 29 (all above obtained, < 0.92 mmol) in DME/water (9: 1 mixture, 10 mL). The reaction was stirred at room temperature for 2 h, quenched by the addition of 5% Na 2 S0 3 , stirred 15 rain at room temperature, and then extracted with EtOAc. The organic fraction was washed with brine, dried with Na 2 SC>4, and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound 30 (491 mg, quantitative) as a white solid: m/z 508.3 (M+ l).

Compound 31: A solution of compound 30 (all above obtained, < 0.92. mmol) and NaOMe (25% in MeOH, 1.3 mL) in MeOH was stirred at 55 °C for 16 h. The resultant mixture was diluted with 1 N HC1 and extracted with EtOAc. The combined organic fractions were washed with brine, dried with Na 2 S0 4 , and concentrated. The residue was purified by flash chromatography (silica gel, 0% to 80% EtOAc in hexanes) to give compound 31 (151 mg, 32%) as a white solid: m/z 508.3 (M+l).

Compound TX63744: l,3-dibromo-5,5-dimethy3hydantoin (45 mg, 0, 16 mmol) was added to a 0 °C solution of compound 31 (151 mg, 0.278 mmol) in DMF (10 mL). The mixture was stirred at 0 °C for 2.5 h. Pyridine (0.10 mL, 1.2 mmol) was then added, and the reaction heated to 55 °C overnight. The resultant solution was diluted with EtOAc, washed with 1 N HQ, 10% Na 2 S<¼, and brine, dried with Na 2 S04, and concentrated. The residue was purified by column chromatography (silica gel, 0% to 80% EtOAc in hexanes) to give compound TX63744 (82 mg, 58%) as a white solid: ' Ή NMR (400 MHz, CDCL) δ 8.04 (s, IB), 5.97 (s, 1H), 3.66 (m, 2H), 3.05 (d, 1H, J ----- 4.7 Hz), 2,25 i id. 1H, J - ----- 4.1 , 13.6 Hz),

1.66 (m, 14H), 1.49 (s, 3H), 1.47 (s, 3H), 1.26 (s, 3H), 1.23 (m, 4H), 1.18 (s, 3H), 1.04 (m, 2H), 1.01 (s, 3H), 0,93 (s, 3H), 0.87 (s, 3H); m/z 506.3 (M+l).

Compound TX63983: A solution of compound TX63744 (24.1 mg, 0.0477 mmol) and EiNCO (39 iL, 0.49 mmol) in toluene (0.5 mL) was heated to 70 °C for 20 h, then purified directly by column chromatography (silica gel, 0% to 100% EtOAc in hexanes) to give compound TX63983 (20.7 mg, 75%) as a white solid: ] H NMR (400 MHz, CDC1 3 ) δ

{00086504} 91 8.04 (s, Π π. 5.98 (s, i l l ). 4.55 (s, H i s. 4.04 ·: m. 21 h. 3.21 (m, 2H), 3.02 i d. I ll, J - ----- 4.6 Hz),

2.24 (td, H i. ,/ 4.0. 12.8 Hz), 1.66 (m, 13H), 1.50 (s, 3H), 1.46 (s, 3H), 1.26 (s, 3H), 1.25 (m, 4H), 1.18 (s, 3H), 1.14 (t, 3H, J = 7,2 Hz), 1 .04 (m, 2H), 1 .01 (s, 3H), 0.93 (s, 3H), 0.88 (s, 3H); m/z 577.4 (M+l).

All of the compounds, compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the disclosure may have only been described in terms of certain embodiments, it will be apparent to those of skill in the art that variations may be applied to the compounds, compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skiiled in the art are deemed to be within the spirii, scope and concept of the invention as defined by the appended claims.

{00086504} 92 The following references to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.

U.S. Patent No. 7,915,402

U.S. Patent No. 7,943,778

U.S. Patent No. 8,071,632

U.S. Patent No, 8,124,799

U.S. Patent No, 8,129,429

U.S. Patent No. 8,338,618

Abraham and Kappas, Free Radical Biol. Med., 39: 1-25, 2.005.

Ahmad el al, Cancer Res., 68:2920-2926, 2008.

Ahmad et al. , J. Biol. Chem. , 281 :35764-9, 2006.

Araujo et al., J. Immunol , 171 (3): 1572- 1580, 2003.

Bach, Hum. Immunol., 67(6):430-432, 2.006.

Chauha and Chauhan, Pathophysiology, 13(3): 171- 181 2006.

Dickerson et al, Prog Neuropsychopharmacol Biol Psychiatry, March 6, 2007.

Dinkova-Kostova et al, Proc. Natl. Acad. Set USA, 102(12):4584-4589, 2005.

Dudhgaonkar et al, Eur. J. Pain, 10(7):573-9, 2006.

Forstermann, Biol. Chem., 387: 152 , 2006.

Handbook of Pharmaceutical Salts: Properties, and Use, Siahl and Wermuth Eds.), Verlag Helvetica Chirnica Acta, 2002.

Hanson et al, BMC Medical Genetics, 6(7), 2005.

Honda et al. Bioorg. Med. Chem. Lett., 12: 1027-1030, 2002.

Honda et al. , J. Med. Chem., 43 :4233-4246, 2000a.

Honda, et al, J. Med. Chem., 43: 1866-1877, 2000b.

Honda et al, Bioorg. Med. Chem. Lett., 7: 1623- 1628, 1997.

Honda et al, Bioorg. Med. Chem. Lett., 9(24):3429-3434, 1999.

Honda et al, Bioorg. Med. Chem. Lett, 8(19):271 1-2714, 1998.

Honda et al. Bioorg. Med. Chem. Lett, 16(24):6306-6309, 2006.

Hong et al, Clin Cancer Res, 18( 12):3396-406, 2012.

{00086504} Ishikawa et al.. Circulation, 104( 15):! 831-1836, 2001.

Ka.waka.mi et al. Brain Dev., 28(4):243-246, 2006,

Kendall- Tackett, Trauma Violence Abuse, 8(2): 1 17- 126, 2007.

Kroger et al. , J. Pharmacol. Exp. " Cher., 319(3): 1 144-1 152, 2006.

Lee et al, Glia., 55(7):712-22, 2007.

Lencz et al, Mol Psychiatry, 12(6):572-80, 2007.

Liby et al, Cancer Res., 65(1 l ):4789-4798, 2005.

Liby et al, Nat. Rev. Cancer, 7(5):357-356, 2007a.

Liby et al, Mol Cancer Then, 6(7):21 13-9, 2007b.

Liby et al., 2007b

Liu et al, FASEB J., 20(2):207-216, 2006.

La el al, ./. Clin. Invest., 12 i( i0):4015-29. 201 1.

March 's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 2007, vie h er et al, Pain, 120(1-2): 161-9, 2005.

Morris et al. J. Mol Med., 80(2):96-104, 2002.

Morse and Choi, Am. J. Respir. Oil. Care Med., 172(6):660-670, 2005.

Morse and Choi, Am. J. Respir. Crit. Care Med., 27(1):8-16, 2002.

Pall, et/. Hypotk , 69:821-825, 2007.

Pergola el. al, N Engl. J Med, 365:327-336, 201 1.

Place et al, Clin. Cancer Res., 9(7):2798-8G6, 2003.

Rajakariar ei al, Proa Natl. Acad Sci. USA, 104(52):20979-84, 2007.

Ross el al, Am. J. Clin. Pathol, 120(Suppl):S53-71 , 2003.

Ross et a Expert Rev. Mol. Diagri., 3(5):573-585, 2003.

Ruster et al, Scand. J. Rheumatol , 34(6):460-3, 2005.

Sacerdoti et al , Curr Neurovasc Res. 2(2) : 103 - 1 1 1 , 2005.

Salvemini el al, J. Clin. Invest.. 93(5): 1940- 1947, 1994.

Sarchielli έ'ί al, Cephalalgia. 26(9): 1071 -1079 , 2006.

Satoh et al, Proc. Natl Acad. Sci. USA, 103(3):768-773, 2006.

Scliulz et al, Antioxid. Redox. Sig., 10: 1 15, 2008.

S!rejan et al, J. Neuroimmunol., 7:27, 1984.

Sa et aL, Ca cer Res., 58:717-723, 1998.

Suh et a Cancer Res., 59(2):336-341 , 1999.

Szabo et al, Nature Rev. Drug Disc, 6:662-680, 2007.

Takahashi et al.. Cancer Res., 57: 1233-1237, 3997,

{00086504} Q 4 Tamil and Tannebaum, Biochim. Biophys. Acta, 1288:F31-F36, 1996. Xie et al., J. Biol Chem., 270(12):6894--6900, 1995.

Zhou et αί. , Ληι. J. Pathol., 166(l):27-37, 2005.

{00086504} 95