Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SYNTHETIC TRITERPENOIDS AND METHODS FOR MODULATING STEM/PROGENITOR CELL GENE EXPRESSION
Document Type and Number:
WIPO Patent Application WO/2013/169740
Kind Code:
A2
Abstract:
Synthetic triterpenoids and methods of using the same to induce gene expression and differentiation of stem or progenitor cells are provided.

More Like This:
Inventors:
GRIBBLE GORDON W (US)
FU LIANGFENG (US)
SPORN MICHAEL B (US)
LIBY KAREN T (US)
Application Number:
PCT/US2013/039902
Publication Date:
November 14, 2013
Filing Date:
May 07, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DARTMOUTH COLLEGE (US)
International Classes:
C07C69/757; A61K31/19; A61K31/215; A61P39/00; C07C61/29
Foreign References:
US20090060873A12009-03-05
US20100056777A12010-03-04
Other References:
BENSASSON ET AL.: 'Efficacy ranking of triterpenoids as inducers of a cytoprotective enzyme and as inhibitors of a cellular inflammatory response via their electron affinity and their electrophilicity index.' CHEM BIOL INTERACT., [Online] vol. 186, no. 2, 2010, pages 118 - 126 Retrieved from the Internet: [retrieved on 2013-12-03]
HONDA ET AL.: 'Synthetic Oleanane and Ursane Triterpenoids with Modified Rings A and C: A Series of Highly Active Inhibitors of Nitric Oxide Production in Mouse Macrophages.' J. MED. CHEM. vol. 3, no. 22, 2000, pages 4233 - 4246
Attorney, Agent or Firm:
TYRRELL, Katheleen, A. et al. (66 E. Main StreetMarlton, NJ, US)
Download PDF:
Claims:
What is claimed is:

1. A triterpenoid compound having the structure Formula I, or a dimer thereof:

Formula I

wherein,

at least one of X1 and X2 is ORa, NRaRb, or SRa, and the other of X1 and X2 is hydrogen, 0Ra, NRaRb, or SRa, wherein

Ra is a hydrogen, cyano, -CF3, nitro, amino, or substituted or unsubstituted heteroaryl group;

Rb is hydrogen, hydroxyl , alkyl, aryl , aralkyl, acyl, alkoxy, aryloxy, acyloxy, alkylamino, arylamino, amido, or a substituted version of any of these groups;

or a substituent convertible in vivo to hydrogen; provided that Ra is absent when the atom to which it is bound is part of a double bond, further provided that when Ra is absent the atom to which it is bound is part of a double bond;

Y is CH2 or CH2-CH2;

Z is a covalent bond, -C(=0)-, alkanediyl, alkenediyl, alkynediyl, or a substituted version of any of these groups ;

the dashed bonds can be independently present or absen ; R1, R2, R3 and R4 are each independently a hydrogen, hydroxyl , alkyl, substituted alkyl, alkoxy or substituted alkoxy group;

at least one of R5, R6 , R7, R8, R9 or R10 is independently -OMs, -CH2OMs, -C(=0)C≡CRa, -C≡CC02Ra, C≡CS02Ra, -C≡CC (=0) Ra or -S02Ra, or

R5 and R6 , or R7 and R8, or R9 and R10 are together =CRcRd, wherein

Rc is hydrogen or alkylthiyl, and

Rd is hydrogen, halo, alkylthiyl, or substituted or unsubstituted alkylsulfonyl or alkylsulfonyl -O- ;

the remainder of R5, R6, R7, R8, R9 and R10 are independently hydrogen, hydroxyl, halo, cyano, =0, -C≡CRa, - C02Ra, -C0Ra, alkyl, alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl , alkoxy, aryloxy, acyloxy, alkylamino, arylamino, nitro, amino, amido, -C(0)Re or a substituted version of any of these groups, wherein

Re is hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or Ci-Ci5-alkyl , C2-Ci5-alkenyl , C2-C15- alkynyl, C6-C15-aryl, C7-Ci5-aralkyl , C;i-Ci5-heteroaryl , C2- C15-heteroaralkyl , Ci-C15-acyl, Ci-C15-alkoxy, C2-Ci5- alkenyloxy, C2-Ci5-alkynyloxy, C6-C15-aryloxy, C7-Ci5- aralkyloxy, Ci-Cis-heteroaryloxy, C2-Ci5-heteroaralkyloxy, Ci- C15-acyloxy, Ci-Ci5-alkylamino , C2-Ci5-dialkylamino, C1-C15- alkoxyamino, C2-Ci5-alkenylamino , C2 -C15-alkynylamino , Cg-C15- arylamino, C7-Ci5-aralkylamino, Cx-Cis-heteroarylamino, C2- Ci5-heteroaralkylamino, Ci-Ci5-alkylsulfonylamino, Cx-Cis- amido, Cx-Cis-alkylsilyloxy, or substituted versions of any of these groups;

R11 and R12 are each independently hydrogen, hydroxyl, halo, alkyl , alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, acyl, alkoxy, aryloxy, aralkoxy, heteroaryloxy, hetero-aralkoxy, acyloxy, alkylamino, dialkylamino, arylamino, aralkylamino, heteroarylamino , heteroaralkylamino , amido, or a substituted version of any of these groups, or

R11 and R12 are taken together and are alkanediyl, alkenediyl, arenediyl , alkoxydiyl, alkenyloxydiyl, alkylaminodiyl , alkenylaminodiyl , or alkenylaminooxydiyl ;

R13 is hydrogen, hydroxy or oxo;

R14 is hydrogen or hydroxyl ; and

R15 is

a hydrogen, hydroxyl, -NRfR9, cyano, halo, azido, phosphate, 1 , 3 -dioxoisoindolin-2 -yl , mercapto, silyl or - COOH group,

substituted or unsubstituted versions of C1-C15- alkyl, C2-CiS-alkenyl , C2-Ci5-alkynyl , C6-C15-aryl, C7-Ci5- aralkyl, Ci-Ci5-heteroaryl , C2~Ci5-heteroaralkyl , Ci-C15-acyl, Ci-Ci5-alkoxy, C2-Ci5-alkenyloxy, C2-Ci5-alkynyloxy, C6-Ci5- aryloxy, C7-Ci5-aralkyloxy, Ci-C15-heteroaryloxy, C2-Ci5- heteroaralkyloxy, Ci-Ci5-acyloxy, Ci-Ci5-alkylamino, C2-Ci5- alkenylamino, C2-Ci5-alkynylamino, C3-C15-arylamino, C7-Ci5- aralkylamino, Ci-C15-heteroarylamino, C2-Ci5- heteroaralkylamino, Cx-Cis-amido, Ci-C15-alkylthio, C2-Ci5- alkenylthio, C2-Ci5-alkynylthio, C6-Ci5-arylthio , C7-C15- aralkylthio, Ci-Ci5-heteroarylthio, C2-Ci5-heteroaralkylthio, Ci-Ci5-acylthio, C1-C12-thioacyl , Ci-C12-alkylsulfonyl , C2-C12- alkenylsulfonyl , C2-Ci2-alkynylsulfonyl , C6-C12-arylsulfonyl , C7-C12-aralkylsulfonyl , , C1-Ci2- heteroaralkylsulfonyl , Ci-Ci2-alkylsulfinyl , C2-Ci2- alkenylsulfinyl , C2-Ci2-alkynylsulfinyl , C6-Ci2-aryl sulfinyl, C7-Ci2-aralkylsulfinyl , Ci-CX2-heteroarylsulfinyl , Ci-Ci2-heteroaralkylsulfinyl , Cx-Ci2-alkylphosphonyl , Cx-Ci2- alkylphosphate, C2-C12-dialkylphosphate , Ci-C12- alkylammonium, Ci-Ci2-alkylsulfonium, Ci-C15-alkylsilyl , or a substituted version of any of these groups, a -C02Me, carbonyl imidazole, -CO-D-Glu (OAc) , CONH2, -CONHNH2, -CONHCH2CF3, or -C (=0) -heteroaryl group, or

Z and R15 form a three to seven-membered ring, such that Z and R15 are further connected to one another through one or more of -0- and alkanediyl, further wherein Z is - CH- and R15 is -CH2- or Z, R15 , and carbon numbers 13, 17 and 18 form a ring such that R15 is bound to carbon 13, wherein Y is methanediyl or substituted methanediyl and R15 is -0-, wherein

Rf and R9 are independently hydrogen, hydroxyl, alkyl, alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl , alkoxy, alkenyloxy, alkynyloxy, aryloxy, aralkoxy, heteroaryloxy, heteroaralkoxy, thioacyl, alkylsulfonyl , alkenylsulfonyl , alkynylsulfonyl , arylsulfonyl , aralkylsulfonyl , heteroarylsulfonyl , or heteroaralkylsulfonyl , or a substituted version of any of these groups.

2. The triterpenoid compound of claim 1, wherein the dimer has the structure of Formula II,

Formula II

wherein

X1, X2, Y, Z, R1, R2, R3, R4, R8, R9, R11, R12, R13 , R14 and R1S are as defined for Formula I, and L is -C≡C-R-C≡C-, -C(=0)-, -C≡C-, -C≡C-N(-R)-, -C(=0)- N(-R)-, -C≡C-C(=0)~, -Ar-C(=0)-, or -C≡C-C (=0) -Ar- , wherein

R is hydrogen, or an alkyl , aryl , alkenyl, or alkynyl group.

3. The triterpenoid compound of claim 1, wherein the compound has the structure of :

Formula VI Formula VII

Formula VIII Formula IX wherein,

R11 and R12 are as defined for Formula I;

R18 is -OMs, -CH2OMs, -C(=0)C≡CRa, -C≡CC02Ra, -C≡CS02Ra, - C≡CC(=0)Ra, -S02Ra, =0 or =CRcRd, wherein

Rd is hydrogen, halo, alkylthiyl, or substituted or unsubstituted alkylsulfonyl or alkylsulfonyl -0- ;

R22 and R23 are independently a hydrogen, hydroxyl , halo, alkyl, nitro or amino group; R19, R20, R21 , R24 and R25 are independently a hydrogen, hydroxyl , -NRfR9, cyano, halo, azido, phosphate, 1,3- dioxoisoindolin-2 -yl , mercapto, silyl or -COOH group,

substituted or unsubstituted versions of Ci-C15-alkyl , C2-C15-alkenyl, C2-Ci5-alkynyl , Cg-C15-aryl, C7-Ci5-aralkyl , Cx- Ci5-heteroaryl , C2-CiS~heteroaralkyl , Ci-Ci5-acyl7 C1-C15- alkoxy, C2-Ci5-alkenyloxy, C2-Gi5-alkynyloxy, Ce-Ci5-aryloxy, C7-Ci5-aralkyloxy, Ci-CiS-heteroaryloxy, C2-Ci5- heteroaralkyloxy, Ci-Ci5-acyloxy, Ci-C15-alkylamino, C2-Ci5- alkenylamino, C2-C15-alkynylamino, C6-Ci5-arylamino, C7-Ci5- aralkylamino, Ci-C15-heteroarylamino, C2-Ci5- heteroaralkylamino, Ci-Ci5-amido, Ci-Ci5-alkylthio, C2-Ci5- alkenylthio, C2-C15-alkynylthio, C6-Ci5-arylthio, C7-Ci5- aralkylthio, Ci-Ci5-heteroarylthio, C2-Ci5-heteroaralkylthio, Ci-C15-acylthio, Ci-Ci2- thioacyl , Ci-C12-alkylsulfonyl , C2-C12- alkenylsulfonyl , C2-Ci2-alkynylsulfonyl , C6-Ci2-arylsulfonyl , C7-Ci2-aralkylsulfonyl , Ci-Ci2-heteroarylsulfonyl , Ci-C12- heteroaralkylsulfonyl , C1-Ci2-alkylsulfinyl , C2-C12- alkenylsulfinyl , C2-Ci2-alkynylsulfinyl , C6-Ci2-aryl sulfinyl, C7-Ci2-aralkylsulfinyl , Ci-C12-heteroarylsulfinyl , Ci-Ci2-heteroaralkylsulfinyl , Ci-Ci2-alkylphosphonyl , C!-Ci2- alkylphosphate, C2-Ci2-dialkylphosphate , Ci-Ci2- alkylammonium, C!-C12-alkylsulfonium, Ci-Ci5-alkylsilyl , or a substituted version of any of these groups, or

a -C02Me, carbonyl imidazole, -CO-D-Glu (OAc) 4 , -CONH2, -CONHNH2, -CONHCH2CF3, or -C ( =0) -heteroaryl group.

4. The triterpenoid compound of claim 1, wherein the compound has the structure of :

Formula X Formula XI

Formula XII Formula XIII wherein,

R11 and R12 are as defined for Formula I ;

X is =0 or -OMe;

dashed bonds are present or absent ;

R26 and R27 are independently a hydrogen, halo, alkylthiyl, or substituted or unsubstituted alkylsulfonyl or -O-alkylsulfonyl ; and

R28 and R29 are independently -H , or together are =0.

5. The triterpenoid compound of claim 1, wherein the compound has the structure of :

Formula XIV Formula XV

Formula XVI

wherein

R11 and R12 are as defined for Formula I;

R30, R31, R32 and R33 are each independently a hydrogen, cyano, -OMs, or -CH2OMs group; or

R30 and R31 or R32 and R33 together are =CH2; and

dashed bonds are either present or absent .

6. A pharmaceutical composition comprising the compound of claim 1, 2, 3, 4 or 5 in admixture with a pharmaceutically acceptable carrier.

7. A method for producing a stem or progenitor cell with induced gene expression comprising contacting a stem or progenitor cell with an effective amount of a synthetic triterpenoid to induce the expression of one or more of SOX9 (Sex determining region Y-box 9) , COL2A1 (Type II Collagen (alphal) ) , TGF (Transforming Growth Factor) -βΐ, TGF- 2, TGF- 3, BMP (Bone Morphogenic Protein) 2, BMP , BMPRII (Bone Morphogenic Protein Receptor II) , SMAD (Small Mothers Against Decapentaplegic) 3, SMAD4 , SMAD6, SMAD7, TIMP (Tissue Inhibitor of Metalloproteinase) -1 or TIMP-2 in the stem or progenitor cell, wherein the stem or progenitor cell is not a mesenchymal stem cell, periosteium cell or osteoprogenitor cell .

8. The method of claim 7, wherein the synthetic triterpenoid has the structure of Formula I, or a dimer thereof :

Formula I

wherein,

at least one of X1 and X2 is 0Ra, NRaRb, or SRa, and the other of X1 and X2 is hydrogen, ORa, NRaRb, or SR , wherein

Ra is a hydrogen, cyano, -CF3, nitro, amino, or substituted or unsubstituted heteroaryl group;

Rb is hydrogen, hydroxyl, alkyl, aryl , aralkyl, acyl , alkoxy, aryloxy, acyloxy, alkylamino, arylamino, amido, or a substituted version of any of these groups;

or a substituent convertible in vivo to hydrogen; provided that Ra is absent when the atom to which it is bound is part of a double bond, further provided that when Ra is absent the atom to which it is bound is part of a double bond;

Y is CH2 or CH2-CH2;

Z is a covalent bond, -C(=0)-, alkanediyl, alkenediyl, alkynediyl, or a substituted version of any of these grou s ;

the dashed bonds can be independently present or absent ;

R1, R2, R3 and R4 are each independently a hydrogen, hydroxyl, alkyl, substituted alkyl, alkoxy or substituted alkoxy group;

at least one of R5, R6, R7, R8, R9 or R10 is independently -OMs, -CH2OMs, -C(=0)C≡CRa, -C≡CC02Ra, C≡CS02Ra, -C≡CC(=0)Ra or -S02Ra, or

R5 and Rs, or R7 and R8, or R9 and R10 are together =CRcRd, wherein

Rc is hydrogen or alkylthiyl, and

Rd is hydrogen, halo, alkylthiyl, or substituted or unsubstituted alkylsulfonyl or alkylsulfonyl -O- ;

the remainder of R5, R6, R7, R8, R9 and R10 are independently hydrogen, hydroxyl, halo, cyano, =0, -C≡CRa, - C02Ra, -CORa, alkyl, alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl , alkoxy, aryloxy, acyloxy, alkylamino, arylamino, nitro, amino, amido, -C(0)Re or a substituted version of any of these groups, wherein

Re is hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or Ci-Ci5-alkyl , C2 -Ci5-alkenyl , C2-CiS- alkynyl, C6-Ci5-aryl, C7-Ci5-aralkyl , Ci-Ci5-heteroaryl , C2- C15-heteroaralkyl , Ci-Cis-acyl, Ci-C15-alkoxy, C2-C15- alkenyloxy, C2-Ci5-alkynyloxy, C6-Ci5-aryloxy, C7-Ci5- aralkyloxy, Ci-Ci5-heteroaryloxy, C2-Ci5-heteroaralkyloxy, Cx- Ci5-acyloxy, Ci-Cis-alkylamino , C2-Ci5-dialkylamino, Ci-C15- alkoxyamino, C2-C15-alkenylamino, C2-Ci5-alkynylamino, C3-Ci5- arylamino, C7-Ci5-aralkylamino, Ci-Ci5-heteroarylamino, C2- C15-heteroaralkylamino, Ci-Cis-alkylsulfonylamino, C1-C15- amido, Ci-C15-alkylsilyloxy, or substituted versions of any of these groups;

R11 and R12 are each independently hydrogen, hydroxyl , halo, alkyl , alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl , alkoxy, aryloxy, aralkoxy, heteroaryloxy, hetero-aralkoxy, acyloxy, alkylamino, dialkylamino, arylamino, aralkylamino, heteroarylamino, heteroaralkylamino, amido, or a substituted version of any of these groups, or

R11 and R12 are taken together and are alkanediyl, alkenediyl, arenediyl , alkoxydiyl , alkenyloxydiyl, alkylaminodiyl , alkenylaminodiyl , or alkenylaminooxydiyl ;

R13 is hydrogen, hydroxy or oxo ;

R14 is hydrogen or hydroxyl; and

R15 is

a hydrogen, hydroxyl, -NRfR3, cyano, halo, azido, phosphate, 1 , 3 -dioxoisoindolin-2 -yl , mercapto, silyl or - COOH group,

substituted or unsubstituted versions of Ci-Ci5- alkyl, C2-C15-alkenyl , C2-Ci5-alkynyl , C3-Ci5-aryl, C7-Ci5- aralkyl, Ci-Ci5-heteroaryl , C2-Ci5-heteroaralkyl , Ci-Ci5-acyl, C1-C15-alkoxy, C2-Ci5-alkenyloxy, C2-Ci5-alkynyloxy, C6-Ci5- aryloxy, C7-Ci5-aralkyloxy, Ci-C15-heteroaryloxy, C2-Ci5- heteroaralkyloxy, d-C15-acyloxy, Ci-Ci5-alkylamino, C2-Ci5- alkenylamino, C2-Ci5-alkynylamino, C6-Ci5-arylamino, C7-Ci5- aralkylamino, Ci-C15-heteroarylamino, C2-Ci5- heteroaralkylamino, Ci-Ci5-amido, Ci-CiS-alkylthio, C2-Ci5- alkenylthio, C2-C15-alkynylthio, C6-Ci5-arylthio, C7-Ci5- aralkylthio, Ci-C15-heteroarylthio, C2-C15-heteroaralkylthio, Ci-Cis-acylthio, Ci-Ci2-thioacyl , Ci-C12-alkylsulfonyl , C2-C12~ alkenylsulfonyl , C2-Ci2-alkynylsulfonyl , C6-Ci2-arylsulfonyl , C7-Ci2-aralkylsulfonyl , Ci-Ci2-heteroarylsulfonyl , C1-C12- heteroaralkylsulfonyl , C1-Ci2-alkylsulfinyl , C2-C12- alkenylsulfinyl , C2-Ci2-alkynylsulfinyl , C6-C12-aryl sulfinyl, C7-Ci2-aralkylsulfinyl , Ci-Ci2-heteroarylsulfinyl , Ci-Ci2-heteroaralkylsulfinyl , C1-Ci2-alkylphosphonyl , C1-C12- alkylphosphate , C2-C12-dialkylphosphate , Ci-Ci2- alkylammonium, C1-C12-alkylsulfonium, Cx-Cis-alkylsilyl , or a substituted version of any of these groups,

a -C02Me, carbonyl imidazole, -CO-D-Glu (OAc) 4 , CONH2, -CONHNH2 , -CONHCH2CF3, or -C (=0) -heteroaryl group, or

Z and R15 form a three to seven-membered ring, such that Z and Rls are further connected to one another through one or more of -0- and alkanediyl, further wherein Z is - CH- and R15 is -C¾- or Z, R15, and carbon numbers 13, 17 and 18 form a ring such that R15 is bound to carbon 13, wherein Y is methanediyl or substituted methanediyl and R15 is -0-, wherein

Rf and R9 are independently hydrogen, hydroxyl , alkyl, alkenyl, alkynyl , aryl , aralkyl, heteroaryl, heteroaralkyl , acyl, alkoxy, alkenyloxy, alkynyloxy, aryloxy, aralkoxy, heteroaryloxy, heteroaralkoxy, thioacyl, alkylsulfonyl , alkenylsulfonyl , alkynylsulfonyl , arylsulfonyl , aralkylsulfonyl , heteroarylsulfonyl , or heteroaralkylsulfonyl , or a substituted version of any of these groups .

9. The method of claim 7, wherein the stem or progenitor cell is multipotent .

10. A stem or progenitor cell produced by the method of claim 7.

11. A method for treating a degenerative disease or injury comprising transplanting into a patient suffering from a degenerative disease or injury a stem or progenitor cell of claim 10 thereby treating the patient's degenerative disease or injury, wherein the degenerative disease or injury is a skin wound or injury, muscle injury, Parkinson's disease, diabetes, nerve injury, amyotrophic lateral sclerosis, traumatic brain injury, cardiac injury or disease, baldness, myocardial infarction, muscular dystrophy, or liver disease or injury.

12. A method for treating a congenital disorder comprising transplanting into a patient suffering from a congenital disorder a stem or progenitor cell of claim 10 thereby treating the patient's congenital disorder, wherein the congenital disorder is familial dysautonomia, a congenital heart defect, a dysganglionic disorder in the gastrointestinal tract, or a pigment cell disorder.

13. A method for treating a degenerative disease or injury comprising administering to a patient suffering from a degenerative disease or injury an effective amount of a synthetic triterpenoid thereby treating the patient's degenerative disease or injury, wherein the degenerative disease or injury is a skin wound or injury, muscle injury, Parkinson's disease, diabetes, nerve injury, amyotrophic lateral sclerosis, traumatic brain injury, cardiac injury or disease, baldness, myocardial infarction, muscular dystrophy, or liver disease or injury.

14. A method for treating a congenital disorder comprising administering to a patient suffering from a congenital disorder an effective amount of a synthetic triterpenoid thereby treating the patient's congenital disorder, wherein the congenital disorder is familial dysautonomia, a congenital heart defect, a dysganglionic disorder in the gastrointestinal tract, or a pigment cell disorder .

15. A method for treating a chemoresistant cancer comprising administering to a patient with a chemoresistant cancer an effective amount of a synthetic triterpenoid, thereby treating the patient's chemoresistant cancer.

16. The method of claim 15, wherein the chemoresistant cancer is chemoresistant colon cancer or glioblastoma.

Description:
Synthetic Triterpenoids and Methods for Modulating Stem/Progenitor Cell Gene Expression

Introduction

[0001] This application claims the benefit of priority from U.S. Patent Application Serial Nos . 13/466,456, filed May 8, 2012, and 61/644,080, filed May 8, 2012, the contents of which are incorporated herein by reference in their entireties .

Background of the Invention

[0002] One of the major needs in cancer prevention is the development of effective and safe new agents for chemoprevention . In particular, there is a need for chemopreventative agents targeted at mechanisms known to be involved in the process of carcinogenesis. In recent years, there has been a resurgence of interest in the study of mechanisms of inflammation that relate to carcinogenesis and in the use of such mechanisms as the basis for development of new chemopreventative agents.

[0003] The concept that inflammation and carcinogenesis are related phenomena has been the subject of many studies that have attempted to link these two processes in a mechanistic fashion (Sporn & Roberts (1986) J. Clin. Invest. 78:329- 332; Ohshima & Bartsch (1994) Mutat. Res. 305:253-264) . The enzymes that mediate the constitutive synthesis of nitric oxide and prostaglandins from arginine and arachidonate , respectively, have relative little significance for either inflammation or carcinogenesis. In contrast, inducible nitric oxide synthase (iNOS) and inducible cycloxygenase

(COX- 2) both have critical roles in the response of tissues to injury or infectious agents (Moncada, et al . (1991) Pharmacol. Rev. 43:109-142; Nathan & Xie (1994) Cell 78:915-918; Siebert & Masferrer (1994) Receptor 4(l):17-23; Tamir & Tannebaum (1996) Biochim. Biophys . Acta 1288 :F31- F36) . These inducible enzymes are essential components of the inflammatory process, the ultimate repair of injury, and carcinogenesis. While physiological activity of iNOS and COX-2 may provide a definite benefit to the organism, aberrant or excessive expression of either iNOS or COX-2 has been implicated in the pathogenesis of many disease processes, particularly in chronic degeneration of the central nervous system, carcinogenesis, septic shock, cardiomyopathy, and rheumatoid arthritis.

[0004] Triterpenoids , biosynthesized in plants by the cyclization of squalene, are used for medicinal purposes in many Asian countries; and some, like ursolic and oleanolic acids, are known to exhibit anti-inflammatory and anti- carcinogenic activity (Huang, et al . (1994) Cancer Res. 54:701-708; Nishino, et al . (1988) Cancer Res. 48:5210- 5215) . However, the biological activity of these naturally- occurring molecules is relatively weak, and therefore the synthesis of new analogs to enhance their potency has been undertaken (Honda, et al . (1997) Bioorg. Med. Chem. Lett. 7:1623-1628; Honda, et al . (1998) Bioorg. Med. Chem. Lett. 8 (19) : 2711-2714) .

[0005] An ongoing effort for the improvement of antiinflammatory and antiproliferative activity of oleanolic and ursolic acid analogs led to the discovery of 2-cyano- 3, 12-dioxooleane-l, 9 (11) -dien-28-oic acid (CDDO) and related compounds (Honda, et al . (1997) supra; Honda, et al . (1998) supra; Honda, et al . (1999) Bioorg. Med. Chem. Lett. 9 (24) : 3429-3434 ; Honda, et al . (2000) J. Med. Chem. 43:4233-4246; Honda, et al . (2000) J. Med. Chem., 43:1866- 1877; Honda, et al . (2002) Bioorg. Med. Chem. Lett. 12:1027-1030; Suh, et al . (1998) Cancer Res. 58:717-723; Sun, et al. (1999) Cancer Res., 59 (2 ) : 336-341 ; Suh, et al . (2003) Cancer Res. 63:1371-1376; Place, et al . (2003) Clin. Cancer Res. 9:2798-2806; Liby, et al . (2005) Cancer Res. 65 :4789-4798) .

[ 0006 ] Furthermore, US Patent No. 6,326,507, US Patent No. 6,552,075, US Patent No. 7,288,568, US Patent No. 7,863,327, US Patent No. 8,034,955, US 2009/0060873, US 2009/0048204, WO 2008/136838 and WO 2009/023232 teach the use of 2-cyano-3 , 12 -dioxooleana- 1 , 9 (11) -dien-28-oic acid

(CDDO) , and derivatives thereof such as 2 -cyano-3 , 12 - dioxoolean-1, 9 (11) -dien-28-oic acid methyl ester (CDDO-Me ) and amide derivatives, for the treatment of diseases such as cancer, Alzheimer's disease, Parkinson's disease, inflammatory bowel diseases, and multiple sclerosis. Similarly, US Patent No. 6,974,801 and WO 2004/064723 teach the use of 2-cyano-3 , 12 -dioxooleana- 1 , 9 (11) -dien-28- onitrile (CNDDO) , 1 -( 2 -cyano-3 , 12 -dioxooleana- 1 , 9 ( 11 ) -dien- 28-oyl) imidazole (CDDO-Im) , 1- (2-cyano-3 , 12 -dioxooleana- 1, 9 (11) -dien-28-oyl) -2-methylimidazole, and 1- (2-cyano- 3, 12-dioxooleana-l, 9 (11) -dien-28-oyl) -4 -methylimidazole in the prevention or treatment of cancer, Alzheimer's disease, Parkinson's disease, multiple sclerosis, rheumatoid arthritis, and other inflammatory diseases. Furthermore, the use of triterpenoids such as CDDO, CDDO-Me , CDDO-Im, and CDDO-Ethylamide in stimulating the growth and repair of bone and cartilage (US 2008/0233195 and WO 2008/064132) as well as in inhibiting HIV-1 replication (WO 2005/046732) has been described. US 2009/0326063 further teaches the use of synthetic triterpenoids in the prevention and treatment of renal/kidney disease, insulin resistance/diabetes, fatty liver disease, and/or endothelial dysfunction/cardiovascular disease . [ 0007 ] Combination therapies of CDDO or CDDO-Me and a chemotherapeutic agent, immunosuppressive agent, or proteasome inhibitor are described in US Patent No. 7,435,755, US Patent No. 7,795,305, US 2009/0018146, US 2009/0048205, WO 2002/047611 and WO 2009/023845 for the treatment of cancer and graft versus host disease. Moreover, formulations for improved oral bioavailability of CDDO-Me are disclosed in WO 2010/093944.

[ 0008 ] Furthermore, CDDO-Me suppresses the induction of several important inflammatory mediators, such as iNOS, COX-2, TNF , and IFNy, in activated macrophages. CDDO-Me has also been reported to activate the Keapl/Nrf2 /ARE signaling pathway resulting in the production of several anti - inflammatory and antioxidant proteins, such as heme oxygenase-1 (HO-1) . These properties have made CDDO-Me a candidate for the treatment of neoplastic and proliferative diseases, such as cancer. Moreover, synthetic triterpenoids have been found to induce apoptosis and differentiation and inhibit proliferation in human leukemia cells (Ikeda, et al . (2003) Cancer Res. 63:5551-5558; onopleva, et al . (2002) Blood 99 (1) :326-335; Suh, et al . (1999) supra; Ito, et al . (2000) Mech. Dev. 97:35-45), induce osteoblastic differentiation in osteosarcoma cells (Ito, et al . (2001) Antimicrob . Agents Chemother. 45:1323-1336), enhance neuronal growth factor-induced neuronal differentiation of rat PC12 pheochromocytoma cells, and induce adipogenic differentiation of fibroblasts into adipocytes (Suh, et al . (1999) supra) . CDDO-Me has also been found an effective drug for improving kidney function in patients suffering for renal/kidney disease using CDDO-Me (U.S. Patent No. 8, 129, 429) .

[ 0009 ] Given the activity of CDDO and CDDO-Me, additional oleanolic acid derivatives have been developed for use in treating cancer, cardiovascular disease, neurodegenerative disease, renal/kidney disease, diabetes, arthritis and inflammatory conditions such as obesity, hypertension, atherosclerosis, coronary heart disease, stroke, peripheral vascular disease, hypertension, nephropathy, neuropathy, myonecrosis, ulcerative colitis, Crohn's disease, irritable bowel syndrome, retinopathy and metabolic syndrome. See US Patent No. 7,915,402, US Patent No. 7,943,778, US 2010/0048887, US 2010/0048892, US 2010/0048911, US 2011/0245206 and US 2011/0245233.

[0010] In view of the therapeutic activities of this class of triterpenoids , it would be advantageous to have compounds with improved activity.

Summary of the Invention

[0011] The present invention encompasses new triterpenoid compounds and the specific finding that such triterpenoid compounds alter gene expression in cells. The present invention therefore provides compositions and methods for inducing cell differentiation, in a variety of important contexts and applications.

[0012] Among other things, this invention provides a triterpenoid compound of Formula I, II, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, or XVI, as defined herein and a pharmaceutical composition containing the same.

[0013] Furthermore, the present invention provides methods for producing a cell, such as a stem or progenitor cell, with induced gene expression by contacting a stem or progenitor cell with an effective amount of a synthetic triterpenoid to induce the expression of one or more of SOX9 (Sex determining region Y-box 9) , COL2A1 (Type II Collagen (alphal) ) , TGF-βΙ, TGF-p2, TGF-p3, BMP2 , BMP4, BMPRII (Bone Morphogenic Protein Receptor II) , SMAD (Small Mothers Against Decapentaplegic) 3, SMAD4 , SMAD6, SMAD7, TIMP (Tissue Inhibitor of Metalloproteinase) -1 or TIMP-2 in the stem or progenitor cell, wherein the stem or progenitor cell is not a mesenchymal stem cell, periosteium cell or osteoprogenitor cell. In one embodiment, the stem or progenitor cell is multipotent .

[0014] A stem or progenitor cell produced by the method is also provided, as are methods of using such cells in the treatment of a degenerative disease, disorder, condition, or injury such as skin wounds or injury, muscle injury, Parkinson's disease, diabetes, nerve injury, amyotrophic lateral sclerosis, traumatic brain injury, cardiac injury or disease, baldness, myocardial infarction, muscular dystrophy, or liver disease or injury. In addition, treatment of a congenital disorder such as familial dysautonomia, a congenital heart defect, a dysgangl ionic disorder in the gastrointestinal tract, or a pigment cell disorder is also provided.

[0015] Methods for treating a degenerative disease or injury, a congenital disorder, or a chemoresistant cancer such as chemoresistant colon cancer or glioblastoma by administering to a patient an effective amount of a synthetic triterpenoid are also provided.

Detailed Description of the Invention

[0016] Triterpenoids, including CDDO-Me derivatives, have now been developed and shown to induce the expression of SOX9, COL2A1, TGF-βΙ, TGF^2, TGF^3, BMP2, BMP4 , BMPRII, SMAD3, SMAD4, SMAD6 , SMAD7 , TIMP-1 and/or TIMP-2 in stem cells. For example, the synthetic triterpenoids CDDO-Im and CDDO-EA induce the expression of each of the above- referenced genes in mesenchymal stem cells and induce chondrogenesis in newborn mouse calvaria. In this respect, the present invention provides synthetic triterpenoid compounds and methods of using to same to produce stem/progenitor cells with altered expression of one or more of SOX9 , C0L2A1, TGF-βΙ, TGF- 2, TGF-p3, BMP2 , BMP4, BMPRII, SMAD3, SMAD4 , SMAD6 , SMAD7 , TIMP-1 or TIMP-2, as compared to a control (e.g., a cell not contacted with the synthetic triterpenoid) , and application of such cells in the prevention or treatment of disease, where the cells can contacted with the triterpenoid in situ, in vivo, ex vivo or in vitro. In some embodiments, expression of one or more of said genes is indicative of differentiation of the stem/progenitor cell.

[0017] Compounds particularly embraced by this invention have the structure of Formula I, which includes hydrates, isomers, prodrugs or pharmaceutically acceptable salts of Formula I :

Formula I

wherein,

at least one of X 1 and X 2 is OR a , NR a R b , or SR , and the other of X 1 and X 2 is hydrogen, OR a , NR a R b , or SR a wherein

R a is a hydrogen, cyano, -CF 3 , nitro, amino, or substituted or unsubstituted heteroaryl group;

R b is hydrogen, hydroxyl , alkyl, aryl , aralkyl, acyl , alkoxy, aryloxy, acyloxy, alkylamino, arylamino, amido, or a substituted version of any of these groups; or a substituent convertible in vivo to hydrogen;

provided that R a is absent when the atom to which it is bound is part of a double bond, further provided that when R a is absent the atom to which it is bound is part of a double bond;

Y is CH 2 or CH 2 -CH 2 ;

Z is a covalent bond, -C(=0)-, alkanediyl, alkenediyl, alkynediyl, . or a substituted version of any of these groups ;

the dashed bonds can be independently present or absent ;

R 1 , R 2 , R 3 and R 4 are each independently a hydrogen, hydroxyl, alkyl, substituted alkyl, alkoxy or substituted alkoxy group;

at least one of R 5 , R 6 , R 7 , R 8 , R 9 or R 10 is independently -OMs, -CH 2 OMs, -C(=0)C≡CR a , -C≡CC0 2 R a , C≡CS0 2 R a , -C≡CC (=0) R a or -S0 2 R a , or

R 5 and R 6 , or R 7 and R 8 , or R 9 and R 10 are together or =CRR d , wherein

R c is hydrogen or alkylthiyl, and

R d is hydrogen, halo, alkylthiyl, or substituted or unsubstituted alkylsulfonyl or alkylsulfonyl-0- ;

the remainder of R 5 , R 6 , R 7 , R 8 , R 9 and R 10 are independently hydrogen, hydroxyl, halo, cyano, =0, -C≡CR a , - C0 2 R a , -C0R a , alkyl, alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl, alkoxy, aryloxy, acyloxy, alkylamino, arylamino, nitro, amino, amido, -C(0)R e or a substituted version of any of these groups, wherein

R e is hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or Ci-Ci 5 -alkyl , C 2 -C 15 -alkenyl , C 2 -Ci 5 - alkynyl, C 6 -C 15 -aryl, C 7 -Ci 5 -aralkyl , Ci-C 15 -heteroaryl , C 2 - C 15 -heteroaralkyl , Ci-Ci 5 -acyl, Ci-C 15 -alkoxy, C 2 -Ci 5 - alkenyloxy, C 2 -Ci 5 -alkynyloxy, C 3 -Ci 5 -aryloxy, C 7 -Ci 5 - aralkyloxy, Cx-Cis-heteroaryloxy, C 2 -C 15 -heteroaralkyloxy, C - Ci 5 -acyloxy, Ci-C 15 -alkylamino, C 2 -Ci 5 -dialkylamino, Ci-Ci 5 - alkoxyamino, C 2 -Ci 5 -alkenylamino, C 2 -C 15 -alkynylamino, C 6 -Ci 5 - arylamino, C 7 -Ci 5 -aralkylamino, Cx-Cxs-heteroarylamino , C 2 - C ls -heteroaralkylamino, Ci-C 15 -alkylsulfonylamino, C 1 -C15- amido, Ci-Ci 5 -alkylsilyloxy, or substituted versions of any of these groups;

R 11 and R 12 are each independently hydrogen, hydroxyl , halo, alkyl , alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl , alkoxy, aryloxy, aralkoxy, heteroaryloxy, hetero-aralkoxy, acyloxy, alkylamino, dialkylamino, arylamino, aralkylamino, heteroarylamino, heteroaralkylamino, amido, or a substituted version of any of these groups, or

R 11 and R 12 are taken together and are alkanediyl, alkenediyl, arenediyl , alkoxydiyl, alkenyloxydiyl , alkylaminodiyl , alkenylaminodiyl , or alkenylaminooxydiyl ;

R 13 is hydrogen, hydroxy or oxo;

R 14 is hydrogen or hydroxyl; and

R 15 is

a hydrogen, hydroxyl, -NR f R 9 , cyano, halo, azido, phosphate, 1 , 3 -dioxoisoindolin-2 -yl , mercapto, silyl or - COOH group,

substituted or unsubstituted versions of C 1 -C15- alkyl, C 2 -Ci 5 -alkenyl , C 2 -C 15 -alkynyl , C e -C 15 -aryl, C 7 -Ci 5 - aralkyl, Ci-Ci 5 -heteroaryl , C 2 -Ci 5 -heteroaralkyl , Ci-Ci 5 -acyl, Ci-Ci 5 -alkoxy, C 2 -Ci 5 -alkenyloxy, C 2 -Ci 5 -alkynyloxy, C 3 -Ci 5 - aryloxy, C 7 -Ci 5 -aralkyloxy, Ci-Ci S -heteroaryloxy, C 2 -Ci 5 - heteroaralkyloxy, Cx-C 15 -acyloxy, Ci-Ci 5 -alkylamino, C 2 -Ci 5 - alkenylamino, C 2 -Ci 5 -alkynylamino, C 6 -C 15 -arylamino, C 7 -Ci 5 - aralkylamino, Ci-Ci 5 -heteroarylamino, C 2 -C 15 - heteroaralkylamino, Ci-Ci 5 -amido, Ci-Ci 5 -alkylthio, C 2 -C 15 - alkenylthio, C 2 -Ci 5 -alkynylthio, C 6 -Ci 5 -arylthio, C7-C15- aralkylthio, Ci-Ci 5 -heteroarylthio, C 2 -Ci 5 -heteroaralkylthio, Ci-Ci 5 -acylthio, Cx-Ci 2 -thioacyl , Ci-Ci 2 -alkylsulfonyl , C 2 -Ci 2 - alkenylsulfonyl , C 2 -Ci 2 -alkynylsulfonyl , C 6 ~Ci 2 -arylsulfonyl , C 7 -C 12 -aralkylsulfonyl , Ci-Ci 2 -heteroarylsulfonyl , Ci-Ci 2 - heteroaralkylsulfonyl , Ci-C 12 -alkylsulfinyl , C 2 -C 12 - alkenylsulfinyl , C 2 -C 12 -alkynylsulfinyl , C 3 -Ci 2 -aryl sulfinyl, C 7 -Ci 2 -aralkylsulfinyl , Ci-Ci 2 -heteroarylsulfinyl , Ci-Ci 2 -heteroaralkylsulfinyl , Ci-Ci 2 -alkylphosphonyl , Ci-Ci 2 ~ alkylphosphate , C 2 -Ci 2 -dialkylphosphate , Ci-Ci 2 - alkylammonium, Ci-C 12 -alkylsulfonium, Ci-Ci 5 -alkylsilyl , or a substituted version of any of these groups,

a -C0 2 Me, carbonyl imidazole, -CO-D-Glu (OAc) 4 , CONH 2 , -CONHNH 2 , -CONHCH 2 CF 3 , or -C (=0) -heteroaryl group, or

Z and R 15 form a three to seven-membered ring, such that Z and R 15 are further connected to one another through one or more of -0- and alkanediyl, further wherein Z is - CH- and R 15 is -CH 2 - or Z, R 15 , and carbon numbers 13, 17 and 18 form a ring such that R 15 is bound to carbon 13, wherein Y is methanediyl or substituted methanediyl and R 15 is -0-, wherein

R f and R 3 are independently hydrogen, hydroxy1 , alkyl, alkenyl , alkynyl, aryl , aralkyl , heteroaryl, heteroaralkyl , acyl , alkoxy, alkenyloxy, alkynyloxy, aryloxy, aralkoxy, heteroaryloxy, heteroaralkoxy, thioacyl, alkylsulfonyl , alkenylsulfonyl , alkynylsulfonyl , arylsulfonyl , aralkylsulfonyl , heteroarylsulfonyl , or heteroaralkylsulfonyl , or a substituted version of any of these groups .

[0018] In certain embodiments, the bond between C 2 and C 3 in the A-ring is a double bond. In other embodiments, the bond between C 2 and C 3 in the A-ring is a single bond. [ 0019 ] In some embodiments, the compound of the invention is a di

Formula II

wherein X 1 , X 2 , Y, Z, R 1 , R 2 , R 3 , R 4 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 and R 15 are as defined for Formula I and L is -C≡C-R-C≡C- , - C(=0)-, -C≡C-, -C≡C-N(-R)-, -C(=0) -N(-R) -, -C≡C-C(=0)-, - Ar-C(=0)-, or -C≡C-C (=0) -Ar- , wherein R is hydrogen, or an alkyl, aryl , alkenyl, or alkynyl group. Exemplary dimers include compounds 10 - 17 .

[ 0020 ] In yet other embodiments, the compound of the invention has the structure as set forth in Formulae VI- XVI. In particular embodiments, the triterpenoid compound of the invention is a compound selected from compound 18 - 75 .

[ 0021] As used herein, "hydrogen" means -H; "hydroxyl" means -OH; "oxo" means =0; "halo" or "halogen" means independently -F, -CI, -Br or -I; "hydroxyamino" means - NHOH; "nitro" means -N0 2 ; "cyano" means -CN; "azido" means - N 3 ; "mercapto" means -SH; "thio" means =S; "sulfonyl" means -S(0) 2 - (see additional definitions of groups containing the term sulfonyl, e.g., alkylsulfonyl ) ; and "silyl" means -SiH 3

(see additional definitions of group (s) containing the term silyl, e.g., alkylsilyl) .

[ 0022 ] For the groups below, the following parenthetical subscripts further define the groups as follows: " (Cn) " defines the exact number (n) of carbon atoms in the group. For example, "Ci-Ci 5 -alkoxy" designates those alkoxy groups having from 1 to 15 carbon atoms {e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, etc. or any range derivable therein {e.g., 3- 10 carbon atoms) ) .

[ 0023 ] The term "alkyl" refers to a non-aromatic monovalent group with a saturated carbon atom as the point of 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. The groups, -CH 3 , CH 2 CH 3 , -C¾CH 2 CH 3 , -CH(CH 3 ) 2 , -CH(CH 2 ) 2 , -CH 2 CH 2 CH 2 CH 3 , CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH 2 C(CH 3 ) 3 , cyclobutyl, cyclopentyl, cyclohexyl, and cyclohexylmethyl are non- limiting examples of alkyl groups.

[ 0024 ] The term "alkanediyl" refers to a non-aromatic divalent group, wherein the alkanediyl group is attached with two σ-bonds, with one or two saturated carbon atom(s) as the point (s) of 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. The groups, -CH 2 - (methylene) , -CH 2 CH 2 -, -CH 2 C (CH 3 ) 2 CH 2 - , and - CH 2 CH 2 CH 2 - are non-limiting examples of alkanediyl groups.

[ 0025 ] The term "alkenyl" refers to a monovalent group with a nonaromatic 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=CH 2 , -CH=CHCH 3 , -CH=CHCH 2 CH 3 , -CH 2 CH=CH 2/ -CH 2 CH=CHCH 3 , and -CH=CH-C 6 H 5 .

[ 0026 ] The term "alkenediyl" refers to a nonaromatic divalent group, wherein the alkenediyl group is attached with two σ-bonds, 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, -CH=CH-, CH=C (CH 3 ) CH 2 - , and -CH=CHCH 2 - are non-limiting examples of alkenediyl groups .

[ 0027 ] The term "alkynyl" refers to a monovalent group with a nonaromatic 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. The groups, -C≡CH, -C≡CCH 3 , C≡CC 6 H 5 and -CH 2 C≡CCH 3 , are non- limiting examples of alkynyl groups .

[ 0028 ] The term "alkynediyl" refers to a nonaromatic divalent group, wherein the alkynediyl group is attached with two σ-bonds, with two carbon atoms as points 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. The groups, -C≡C-, - C≡CCH 2 -, and -C≡CCH(CH 3 )- are non-limiting examples of alkynediyl groups .

[ 002 9 ] The term "aryl" refers to a monovalent group with an aromatic carbon atom as the point of attachment, said carbon atom forming part of a six-membered aromatic ring structure wherein the ring atoms are all carbon, and wherein the monovalent group is composed of carbon and hydrogen. Non-limiting examples of aryl groups include phenyl , methylphenyl , (dimethyl ) phenyl , - ethylphenyl , propylphenyl , -C e H 4 CH (CH 3 ) 2 , -C 6 H 4 CH (CH 2 ) 2 , methylethylphenyl , vinylphenyl , naphthyl , and the monovalent group derived from biphenyl .

[ 003 0 ] The term "arenediyl" refers to a divalent group, wherein the arenediyl group is attached with two σ-bonds, with two aromatic carbon atoms as points of attachment, said carbon atoms forming part of one or more six-membered aromatic ring structure (s) wherein the ring atoms are all carbon, and wherein the monovalent group is composed of carbon and hydrogen. Non-limiting examples of arenediyl

[ 0031] The term "aralkyl" refers to the monovalent group - alkanediyl -aryl , in which the terms alkanediyl and aryl are each used in a manner consistent with the definitions provided above. Non-limiting examples of aralkyls include 1 -phenyl -ethyl , 2 -phenyl -ethyl , indenyl and 2,3-dihydro- indenyl, provided that indenyl and 2 , 3-dihydro- indenyl are only examples of aralkyl in so far as the point of attachment in each case is one of the saturated carbon atoms .

[ 0032 ] The term "heteroaryl" refers to a monovalent group with an aromatic carbon atom or nitrogen atom as the point of attachment, said carbon atom or nitrogen atom forming part of an aromatic ring structure wherein at least one of the ring atoms is nitrogen, oxygen or sulfur, and wherein the monovalent group is composed of carbon, hydrogen, aromatic nitrogen, aromatic oxygen or aromatic sulfur. Non- limiting examples of aryl groups include acridinyl, furanyl , imidazoimidazolyl , imidazopyrazolyl , imidazopyridinyl , imidazopyrimidinyl , indolyl , indazolinyl, methylpyridyl , oxazolyl, phenylimidazolyl , pyridyl, pyrrolyl, pyrimidyl, pyrazinyl, quinolyl , quinazolyl, quinoxalinyl , tetrahydroquinolinyl, thienyl, triazinyl, pyrrolopyridinyl , pyrrolopyrimidinyl , pyrrolopyrazinyl , pyrrolotriazinyl , pyrroloimidazolyl , chromenyl (where the point of attachment is one of the aromatic atoms) , and chromanyl (where the point of attachment is one of the aromatic atoms) .

[0033] The term "heteroaralkyl" refers to the monovalent group -alkanediyl-heteroaryl, in which the terms alkanediyl and heteroaryl are each used in a manner consistent with the definitions provided above. Non-limiting examples of aralkyls include pyridylmethyl , and thienylmethyl .

[0034] The term "acyl" refers to a monovalent group with a carbon atom of a carbonyl group as the point of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure. The groups, -CHO, -C(=0)CH 3 , C(=0)CH 2 C¾, -C(=0)CH 2 CH 2 CH 3 , -C (=0) CH (CH 3 ) 2 , -C ( =0) CH (CH 2 ) 2 , -C(=0)C s H 5 , -C (=0) C 6 H 4 CH 3 , and -C (=0) C e H 4 CH 2 CH 3 are non- limiting examples of acyl groups. The term "acyl" therefore encompasses, but is not limited to groups sometimes referred to as "alkyl carbonyl" and "aryl carbonyl" groups.

[0035] The term "alkoxy" refers to the group -OR, in which R is an alkyl, as that term is defined herein. Non-limiting examples of alkoxy groups include -OCH 3 , -OCH 2 CH 3 , OCH 2 CH 2 CH 3 , -OCH(CH 3 ) 2 , -OCH(CH 2 ) 2 , -O-cyclopentyl , and -O- cyclohexyl .

[0036] Similarly, the terms "alkenyloxy , " "alkynyloxy, " "aryloxy, " "aralkoxy, " "heteroaryloxy, " "heteroaralkoxy" and "acyloxy, " refer to groups, defined as -OR, in which R is alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl and acyl, respectively, as those terms are defined above.

[0037] The term "alkoxydiyl" refers to a non-aromatic divalent group, wherein the alkoxydiyl group is attached with two σ-bonds, with (a) two saturated carbon atoms as points of attachment, (b) one saturated carbon atom and one oxygen atom as points of attachment, or (c) two oxygen atoms as points of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure, no carbon- carbon double or triple bonds in the group's backbone, further having no backbone atoms other than carbon or oxygen and having at least one of each of these atoms in the group's backbone. The groups, -0-CH 2 CH 2 -, -CH 2 -0-CH 2 CH 2 - , -0-CH 2 CH 2 -0- and -0-CH 2 -0- are non-limiting examples of alkoxydiyl groups .

[ 0038 ] The term "alkenyloxydiyl" refers to a divalent group that is nonaromatic prior to attachment, wherein the alkenyloxydiyl group is attached with two σ-bonds, which may become aromatic upon attachment, with (a) two carbon atoms as points of attachment, (b) one carbon atom and one oxygen atom as points of attachment, or (c) two oxygen atoms as points of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure, at least one carbon-carbon double bond that is non-aromatic at least prior to attachment, further having no backbone atoms other than carbon or oxygen and having at least one of each of these atoms in the group's backbone. The groups, -0-CH=CH-, -0-CH=CHO- and -0-CH=CHCH 2 - are non- limiting examples of alkenyloxydiyl groups.

[ 0039 ] The term "amino" refers to a moiety of the formula - NRR' , wherein R and R' are independently hydrogen, alkyl, cycloalkyl, heterocycloalkyl , aryl or heteroaryl .

[ 0040 ] The term "alkylamino" 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 , NHC¾CH 3 , -NHCH 2 CH 2 CH 3 , -NHCH(CH 3 ) 2 , -NHCH(CH 2 ) 2 ,

NHCH 2 CH 2 CH 2 CH 3 , -NHCH (CH 3 ) CH 2 CH 3 , -NHCH 2 CH (CH 3 ) 2 , -NHC(CH 3 ) 3 , - NH-cyclopentyl , and -NH-cyclohexyl .

[ 0041] Similarly, the terms "alkoxyamino, " "alkenylamino, " "alkynylamino, " "arylamino," "aralkylamino, " "heteroarylamino, " "heteroaralkylamino, " and

"alkylsulfonylamino" refer to groups, defined as -NHR, in which R is alkoxy, alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl and alkylsulfonyl , respectively, as those terms are defined above. A non-limiting example of an arylamino group is -NHC S H 5 .

[ 0042 ] The term "dialkylamino" refers to the group -NRR', in which R and R' can be the same or different alkyl groups, or R and R 1 can be taken together to represent an alkanediyl having two or more saturated carbon atoms, at least two of which are attached to the nitrogen atom. Non- limiting examples of dialkylamino groups include -NHC(CH 3 ) 3 , -N(CH 3 ) CH 2 CH 3 , -N(CH 2 CH 3 ) 2 , N-pyrrolidinyl , and N- piperidinyl .

[ 0043 ] The term "alkylaminodiyl" refers to a non-aromatic divalent group, wherein the alkylaminodiyl group is attached with two σ-bonds, with (a) two saturated carbon atoms as points of attachment, (b) one saturated carbon atom and one nitrogen atom as points of attachment, or (c) two nitrogen atoms as points of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure, no double or triple bonds in the group's backbone, further having no backbone atoms other than carbon or nitrogen and having at least one of each of these atoms in the group's backbone. The groups, -NH-CH 2 CH 2 -, -CH 2 -NH-CH 2 CH 2 - , -NH- CH 2 CH 2 -NH- and -NH-CH 2 -NH- are non-limiting examples of alkylaminodiyl groups.

[ 0044 ] The term "alkenylaminodiyl" refers to a divalent group that is nonaromatic prior to attachment, wherein the alkenylaminodiyl group is attached with two σ-bonds, which may become aromatic upon attachment, with (a) two carbon atoms as points of attachment, (b) one carbon atom and one nitrogen atom as points of attachment, or (c) two nitrogen atoms as points of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure, at least one carbon-carbon double bond or carbon-nitrogen double that is non-aromatic at least prior to attachment, further having no backbone atoms other than carbon or nitrogen. The groups -NH-CH=CH-, -NH-CH=N- and -NH-CH=CH-NH- are non-limiting examples of alkenylaminodiyl groups .

[ 0045 ] The term "alkenylaminooxydiyl" refers to a divalent group, wherein the alkenylaminooxydiyl group is attached with two σ-bonds, which may become aromatic upon attachment, with two atoms selected from the group consisting of carbon, oxygen and nitrogen as points of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure, at least one carbon-carbon double bond, carbon-nitrogen double, or nitrogen-nitrogen double bond that is non-aromatic at least prior to attachment, further having no backbone atoms other than carbon nitrogen or oxygen and having at least one of each of these three atoms in the backbone. The group -0-CH=N-, is a non-limiting example of an alkenylaminooxydiyl group.

[ 0046 ] The term "amido" (acylamino) refers to the group - NHR, in which R is acyl , as that term is defined herein. A non-limiting example of an acylamino group is -NHC(=0)CH 3 .

[ 0047 ] The term "alkylthio" refers to the group -SR, in which R is an alkyl, as that term is defined above. Non- limiting examples of alkylthio groups include -SCH 3 , SCH 2 CH 3 , -SCH 2 CH 2 CH 3 , -SCH(CH 3 ) 2 , -SCH(CH 2 ) 2 , -S-cyclopentyl , and - S-eye1ohexy1.

[ 0048 ] Similarly, the terms "alkenylthio, " "alkynylthio, " "arylthio," "aralkylthio , " "heteroarylthio, "

"heteroaralkylthio" and "acylthio" refer to groups, defined as -SR, in which R is alkenyl, alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl and acyl , respectively, as those terms are defined above.

[ 0049 ] The term "thioacyl" refers to a monovalent group with a carbon atom of a thiocarbonyl group as the point of attachment, further having a linear or branched, cyclo, cyclic or acyclic structure. The groups -CHS, -C(=S)CH 3 , - C(=S)CH 2 CH 3 , -C(=S)CH 2 CH 2 CH 3 , -C (=S) CH (CH 3 ) 2 , -C (=S) CH (CH 2 ) 2 , -C(=S)C 6 Hs, -C(=S)C 6 H 4 CH 3 , -C (=S) C 6 H 4 CH 2 CH 3 , -C (=S) C 6 H 3 (CH 3 ) 2 , and -C (=S) CH 2 C e Hs , are non-limiting examples of thioacyl groups. The term "thioacyl" therefore encompasses, but is not limited to, groups sometimes referred to as "alkyl thiocarbonyl" and "aryl thiocarbonyl" groups.

[ 0050 ] The term "alkylsulfonyl" refers to the group S(=0) 2 R, in which R is an alkyl, as that term is defined above. Non-limiting examples of alkylsulfonyl groups include: -S(=0) 2 CH 3 , -S (=0) 2 CH 2 CH 3 , -S (=0) 2 CH 2 CH 2 CH 3 , S(=0) 2 CH(CH 3 ) 2 , -S (=0) 2 CH(CH 2 ) 2 , -S (=0) 2 -cyclopentyl , and - S (=0) 2 -cyclohexyl .

[ 0051 ] Similarly, the terms "alkenylsulfonyl , "

"alkynylsulfonyl , " "arylsulfonyl , " "aralkylsulfonyl , "

"heteroarylsulfonyl , " and "heteroaralkylsulfonyl" refer to groups, defined as -S(0) 2 R, in which R is alkenyl, alkynyl, aryl, aralkyl, heteroaryl, and heteroaralkyl, respectively, as those terms are defined above.

[ 0052 ] The term "alkylsulfinyl" refers to the group S(=0)R, in which R is an alkyl, as that term is defined above. Non-limiting examples of alkylsulfinyl groups include -S(=0)CH 3 , -S (=0) CH 2 CH 3 , -S (=0) CH 2 CH 2 CH 3 , S (=0) CH (CH 3 ) 2 , -S(=0)CH(CH 2 ) 2 , -S (=0) -cyclopentyl , and S (=0) -cyclohexyl .

[ 0053 ] Similarly, the terms "alkenylsulfinyl , "

"alkynylsulfinyl , " "arylsulfinyl , " "aralkylsulfinyl , "

"heteroarylsulfinyl" and "heteroaralkylsulfinyl" refer to groups, defined as -S(=0)R, in which R is alkenyl, alkynyl , aryl, aralkyl, heteroaryl, and heteroaralkyl, respectively, as those terms are defined above.

[ 0054 ] The term "alkylammonium" refers to a group, defined as -NH 2 R + , -NHRR 1 + , or -NRR 1 R" + , in which R, R ' and R" are the same or different alkyl groups, or any combination of two of R, R' and R" can be taken together to represent an alkanediyl . Non-limiting examples of alkylammonium cation groups include -NH 2 (CH 3 ) + , -NH 2 (CH 2 CH 3 ) + , -NH 2 (CH 2 CH 2 CH 3 ) + , - NH(CH 3 ) 2 \ -NH(CH 2 CH 3 ) 2 + , -NH (CH 2 CH 2 CH 3 ) 2 + , -N(CH 3 ) 3 + , N(CH 3 ) (CH 2 CH 3 ) 2 + , -N(CH 3 ) 2 (CH 2 CH 3 ) + , -NH 2 C (CH 3 ) 3 + ,

NH (cyclopentyl) 2 + , and -NH 2 (cyclohexyl ) + .

[ 0055 ] The term "alkylthiyl" refers to the group -SR. Non- limiting examples of alkylthiyl groups include -S(CH 3 ), S(CH 2 CH 3 ), -S (CH 2 CH 2 CH 3 ) , -S (cyclopentyl ) , and

S (cyclohexyl ) .

[ 0056 ] The term "alkylsilyl" refers to a monovalent group, defined as -SiH 2 R, -SiHRR ' , or -SiRR'R", in which R, R ' and R" can be the same or different alkyl groups, or any combination of two of R, R' and R" can be taken together to represent an alkanediyl. The groups -SiH 2 CH 3 , -SiH(CH 3 ) 2 , - Si(CH 3 ) 3 and -Si (CH 3 ) 2 C (CH 3 ) 3 , are non-limiting examples of unsubstituted alkylsilyl groups.

[ 0057 ] The term "alkylphosphonyl" refers to the group - OPO(OR) 2 , where R is alkyl, as defined herein.

[ 0058 ] The term "alkylphosphate" refers to the group 0P(=0) (OH) (OR), in which R is an alkyl, as that term is defined above. Non-limiting examples of alkylphosphate groups include -OP(=0) (OH) (OMe) and -OP(=0) (OH) (OEt) .

[ 0059 ] The term "dialkylphosphate" refers to the group - OP(=0) (OR) (OR'), 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 having two or more saturated carbon atoms, at least two of which are attached via the oxygen atoms to the phosphorus atom. Non-limiting examples of dialkylphosphate groups include -OP (=0) (0Me) 2 , 0P(=0) (OEt) (OMe) and -0P(=0) (0Et) 2 -

[ 0060 ] "Cycloalkyl" means a non-aromatic mono- or multicyclic ring system including about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms. Non-limiting examples of suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. Non-limiting examples of suitable multicyclic cycloalkyls include 1-decalinyl, norbornyl , adamantyl and the like.

[ 0061] "Heterocyclyl" or "heterocycloalkyl" means a non- aromatic saturated monocyclic or multicyclic ring system including about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Preferred heterocyclyls contain about 5 to about 6 ring atoms. The prefix aza, oxa or thia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom. The nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S- oxide or S,S-dioxide. Non-limiting examples of suitable monocyclic heterocyclyl rings include piperidyl, pyrrolidinyl , piperazinyl, morpholinyl, thiomorpholinyl , thiazolidinyl , 1 , 4 -dioxanyl , tetrahydrofuranyl , tetrahydrothiophenyl, lactam, lactone, and the like. Non- limiting examples of suitable bicyclic heterocyclyl rings include decahydro- isoquinoline , decahydro-

[2 , 6] naphthyridine , and the like. [0062] Any of the groups described herein may be unsubst ituted or optionally substituted. When modifying a particular group, "substituted" means that the group the term modifies may, but does not have to, be substituted. Substitutions typically replace an available hydrogen with an alkyl, alkenyl, alkynyl, aryl , heteroaryl, aralkyl, alkylaryl, heteroaralkyl , heteroarylalkenyl , heteroarylalkynyl , alkylheteroaryl , hydroxy, hydroxyalkyl , alkoxy, aryloxy, aralkoxy, alkoxyalkoxy , acyl , halo, nitro, cyano, carboxy, alkoxycarbonyl , aryloxycarbonyl , aralkoxycarbonyl , alkylsulfonyl , arylsulfonyl , heteroarylsulfonyl , alkylthio, arylthio, heteroarylthio, aralkylthio, heteroaralkylthio, cycloalkyl, or heterocyclyl .

[0063] Any undefined valency on an atom of a structure shown in this application implicitly represents a hydrogen atom bonded to the atom.

[0064] The term "hydrate" when used as a modifier to a compound means that the compound has less than one (e.g., hemihydrate) , one (e.g., monohydrate) , or more than one

(e.g., dihydrate) water molecules associated with each compound molecule, such as in solid forms of the compound.

[0065] 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.

[0066] "Pharmaceutically acceptable salts" means salts of compounds of the present invention which are pharmaceutically acceptable, 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 -ethanedisulfonic acid, 2 -hydroxyethanesulfonic acid, 2 -naphthalenesulfonic acid, 3 -phenylpropionic acid, 4 , 4 ' -methylenebis (3 -hydroxy-2 -ene- 1-carboxylic acid) ,

4-methylbicyclo [2.2.2] oct-2-ene-l-carboxylic acid, acetic acid, aliphatic mono- and di -carboxylic acids, aliphatic sulfuric acids, aromatic sulfuric acids, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, carbonic acid, cinnamic acid, citric acid, cyclopentanepropionic acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, heptanoic acid, hexanoic acid, hydroxynaphthoic acid, lactic acid, laurylsulfuric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, muconic acid, o- (4 -hydroxybenzoyl ) benzoic acid, oxalic acid, p- chlorobenzenesulfonic acid, phenyl-substituted alkanoic acids, propionic acid, p-toluenesulfonic 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 > diethanolamine , 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. Stahl & C. G. Wermuth eds . , Verlag Helvetica Chimica Acta, 2002) .

[ 0067 ] Compounds of the 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 prodrugs of compounds of the present invention as well as methods of delivering prodrugs. Prodrugs of the compounds employed in the invention may be prepared by modifying functional groups present in the compound in such a way that the 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 patient, cleaves to form a hydroxy, amino, or carboxylic acid, respectively. For example, a compound comprising a hydroxy group may be administered as an ester that 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- β -hydroxynaphthoate , gentisates , isethionates , di-p-toluoyltartrates, methanesulfonates , ethanesulfonates , benzenesulfonates , p-toluenesulfonates , cyclohexylsulfamates , 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. [0068] The novel compounds of this invention find use in modulating IFN-γ- induced NO production in macrophages, said composition having an IC 50 value of at least less than 0.6 μΜ, more preferably less than 0.001 μΜ.

[0069] In one embodiment, the novel triterpenoid compounds are of use in a method of modulating excessive nitric oxide or prostaglandin formation in a subject by administering to a subject a pharmaceutically effective amount of one or more triterpenoid compounds, such that the nitric oxide or prostaglandin formation is modulated.

[0070] In a further embodiment, the triterpenoid compounds of the invention are of use in a method of preventing or treating a disorder characterized by overexpression of iNOS or COX-2 genes, wherein the method includes administering to a subject a pharmaceutically effective amount of one or more triterpenoid compounds, such that the disorder is prevented or treated. In a preferred embodiment, the disorder includes cancer, diabetic nephropathy, neurodegenerative disease, rheumatoid arthritis, inflammatory bowel disease, and other diseases whose pathogenesis is believed to involve excessive production of either nitric oxide or prostaglandins. In a particular embodiment, the neurodegenerative disease includes Parkinson's disease, Alzheimer's disease, multiple sclerosis, and amyotrophic lateral sclerosis. The cancer may include, e.g., a leukemic cancer or a solid cancer. A leukemic cancer is a cancer of a blood cell, a myeloid cell, a monocytic cell, a myelocytic cell, a promyelocytic cell, a myeloblastic cell, a lymphocytic cell, or a lymphoblastic cell. A solid cancer is a cancer of a bladder cell, a breast cell, a lung cell, a colon cell, a prostate cell, a liver cell, a pancreatic cell, a stomach cell, a testicular cell, a brain cell, an ovarian cell, a skin cell, a brain cell, a bone cell, or a soft tissue cell.

[ 0071] Moreover, the invention provides methods for the treatment and prevention of graft versus host disease (GVHD) by providing a triterpenoid compound of the invention either alone or in conjunction with another agent, such as an immunosuppressive agent such as a corticosteroid or tacrolimus, or a chemotherapeutic agent for the treatment of GVHD. In graft versus host disease the donor immune system mounts a response against the host 1 s organs or tissue. As CDDO compounds, either alone or in conjunction with other agents, can induce apoptosis by inhibiting Bcl-2 and have activity in lymphoid tissue, it is contemplated that the instant triterpenoid compounds can be used to provide therapy for graft versus host diseases.

[ 0072 ] As a further embodiment of this invention, known and novel triterpenoid compounds described herein can be used to produce stem/progenitor cells with altered expression of one or more of SOX9, COL2A1, TGF-βΙ, TGF~ 2, TGF- β3 , BMP2, BMP4, BMPRII, SMAD3 , SMAD4 , SMAD6 , SMAD7 , TIMP-1 or TIMP-2, as compared to a control. As is known in the art, stem cells are primal cells found in all multi-cellular organisms. They retain the ability to renew themselves through mitotic cell division and can differentiate into a diverse range of specialized cell types. Mammalian stem cells include embryonic stem cells, derived from blastocysts; adult stem cells, which are found in adult tissues; and cord blood stem cells, which are found in the umbilical cord. In a developing embryo, stem cells can differentiate into all of the specialized embryonic tissues. In adult organisms, stem cells and progenitor cells act as a repair system for the body, replenishing specialized cells. [0073] As stem cells can be grown and transformed into specialized cells with characteristics consistent with cells of various tissues such as muscles or nerves through cell culture, stem cells are of use in medical therapies. In particular, embryonic cell lines, autologous embryonic stem cells generated through therapeutic cloning, and highly plastic adult stem cells from the umbilical cord blood or bone marrow are promising candidates.

[0074] "Potency" specifies the differentiation potential (the potential to differentiate into different cell types) of the stem cell. Totipotent stem cells are produced from the fusion of an egg and sperm cell. Cells produced by the first few divisions of the fertilized egg are also totipotent. These cells can differentiate into embryonic and extraembryonic cell types. Pluripotent stem cells are the descendants of totipotent cells and can differentiate into cells derived from any of the three germ layers. Multipotent stem/progenitor cells can produce only cells of a closely related family of cells (e.g., hematopoietic stem cells differentiate into red blood cells, white blood cells, platelets, etc.) .

[0075] The term "differentiation" or "differentiated," as used herein, refers to the developmental process wherein an unspecialized or less specialized cell becomes more specialized for a specific function, such as, for example, the process by which a mesenchymal stem cell becomes a more specialized cell such as a cartilage cell, a bone cell, a muscle cell, or a fat cell. Differentiation can be assessed by identifying lineage-specific markers, such as, for example, aggrecan, a proteoglycan specific for cartilage.

[0076] Embryonic stem (ES) cell lines are cultures of cells derived from the epiblast tissue of the inner cell mass (ICM) of a blastocyst. A blastocyst is an early stage embryo, approximately 4 to 5 days old in humans and composed of 50-150 cells. ES cells are pluripotent, and give rise during development to all derivatives of the three primary germ layers: ectoderm, endoderm and mesoderm. In other words, they can develop into each of the more than 200 cell types of the adult body when given sufficient and necessary stimulation for a specific cell type. They do not contribute to the extra-embryonic membranes or the placenta .

[ 0077 ] A human embryonic stem cell is defined by the presence of several transcription factors and cell surface proteins. The transcription factors Oct -4, Nanog, and Sox2 form the core regulatory network, which ensures the suppression of genes that lead to differentiation and the maintenance of pluripotency. The cell surface proteins most commonly used to identify hES cells are the glycolipids SSEA3 and SSEA4 and the keratan sulfate antigens Tra-1-60 and Tra-1-81. Because of the combined ability of unlimited expansion and pluripotency, embryonic stem cells remain a potential source for regenerative medicine and tissue replacement after injury or disease. Therefore, the present invention contemplates the use of embryonic stem cells. Cells derived from embryonic sources may include embryonic stem cells or stem cell lines obtained from a stem cell bank or other recognized depository institution.

[ 0078 ] An adult or somatic stem cell, a cell which is found in a developed organism, has two properties: the ability to divide and create another cell like itself, and also divide and create a cell more differentiated than itself. Adult stem cell treatments have been used for many years to successfully treat leukemia and related bone/blood cancers through bone marrow transplants. Therefore, in particular embodiments of the present invention, adult stem cells are used in the methods of the disclosed herein. Pluripotent adult stem cells are rare and generally small in number but can be found in a number of tissues including umbilical cord blood. Most adult stem cells are lineage restricted (multipotent) . Therefore, in certain embodiments, the adult stem cells of the invention are multipotent.

[ 0079 ] Adult stem cells have been identified in many organs and tissues, including brain, bone marrow, peripheral blood, blood vessels, skeletal muscle, skin, teeth, heart, gut, liver, ovarian epithelium, and testis, and are thought to reside in a specific area of each tissue (called a "stem cell niche") . In this respect, stem cells are generally referred by the tissue from which they are derived, e.g., cardiac stem cells are stems that naturally reside within the heart, adipose-derived stem cells are stem cells found in fat tissue, myoblasts are muscle stem cells, dental pulp stem cells are found in the teeth, etc.

[ 0080 ] Stem cells give rise to a number of different cell types. For example, mesenchymal stem cells (CD105 + , CD90 + , CDllb + , CD34 + and CD45 + ) give rise to bone cells (osteocytes) , cartilage cells (chondrocytes) , fat cells (adipocytes) , and other kinds of connective tissue cells such as those in tendons. Hematopoietic stem cells (Lin " , CD34 + ' CD90 + , CD34 " and CD45 " ) give rise to all the types of blood cells including red blood cells, B lymphocytes, T lymphocytes, natural killer cells, neutrophils, basophils, eosinophils, monocytes, and macrophages. Neural stem cells in the brain (CD133 + , FA-1 + , CD34 " and CD45 " ) give rise to nerve cells (neurons), astrocytes and oligodendrocytes. Epithelial stem cells in the lining of the digestive tract occur in deep crypts and give rise to several cell types including absorptive cells, goblet cells, paneth cells, and enteroendocrine cells. Skin stem cells occur in the basal layer of the epidermis and at the base of hair follicles, and give rise to keratinocytes , which migrate to the surface of the skin and form a protective layer. The follicular stem cells can give rise to both the hair follicle and to the epidermis. Neural crest stem cells (p75 receptor "1" , a 4 integrin receptor 4" , CD29 + and CD9 + ) differentiate into the cells of the peripheral nervous system. In some embodiments, the invention includes the use of mesenchymal stem cells, hematopoietic stem cells, neural stem cells, epithelial stem cells, skin stem cells, follicular stem cells and/or neural crest stem cells. In other embodiments, the invention includes the use of hematopoietic stem cells, neural stem cells, epithelial stem cells, skin stem cells, follicular stem cells and/or neural crest stem cells. In certain embodiments, stem cells in accordance with the present invention are not mesenchymal stem cells.

[ 0081] Progenitor cells are cells that are direct descendants of stem cells, are typically less potent than stem cells, and have diminished capacity for self -renewal relative to stem cells, but retain the ability to become at least one, if not multiple, cell types. In this respect, multipotent progenitor cells are also encompassed within the scope of the present invention. Multipotent progenitor cells are similar to mesenchymal stem cells as they have the ability to differentiate, in vitro, into cells with phenotypic characteristics of cells from all three germ cell layers (mesoderm, ectoderm and endoderm) . Multipotent progenitor cells can be isolated in a fashion similar to mesenchymal stem cells, with adherence to plastic as an initial staple property; however additional separation techniques including magnetic activated cell sorting for CD45 " /TER119 ~ and 96-well single cell isolation/expansion on fibronectin in enriched media are required (Breyer, et al .

(2006) Exp. Hematol . 34:1596-601) . Multipotent progenitor cells possess certain cell surface markers (distinct from mesenchymal stem cells) including CD13, CD31 and SSEA-1 and lack markers including CD3 , CDllb, CD19, CD34, CD44, CD45, MHC I and MHC II (Breyer et al . (2006) supra; Jiang, et al .

(2002) Exp. Hematol. 34:809) . Progenitor cells include satellite cells found in muscles; intermediate progenitor cells formed in the subventricular zone; bone marrow stromal cells; periosteum cells, which includes progenitor cells that develop into osteoblasts or chondroblasts ; pancreatic progenitor cells; angioblasts or endothelial progenitor cells; and blast cells involved in the generation of B- and T-lymphocytes . In some embodiments, the invention includes the use of satellite cells, intermediate progenitor cells, bone marrow stromal cells, pancreatic progenitor cells, endothelial progenitor cells, and/or blast cells. In other embodiments, the invention includes the use of satellite cells, intermediate progenitor cells, bone marrow stromal cells, pancreatic progenitor cells, endothelial progenitor cells, and/or blast cells. In certain embodiments, a progenitor cell is not a periosteium cell or osteoprogenitor cell.

[ 0082 ] The invention applies to cells of an embryonic or adult origin in mammals, both human and non-human mammals, including but not limited to human and non-human primates, ungulates, ruminants and rodents. Ungulate species include, but are not limited to, cattle, sheep, goats, pigs, horses. Rodent species include, but are not limited to, rats and mice. The invention may also find application in other mammalian species such as rabbits, cats and dogs. Examples of preferred stem/progenitor cell populations which can be used in accordance with the methods of the present invention include primate stem/progenitor cells, such as human stem/progenitor cells. Such cells include adult human mesenchymal stem cells. As will be appreciated by one of skill in the art, a stem or progenitor cell referred to herein generally refers to a population of stem or progenitor cells.

[0083] Stem cells of the present invention can be isolated by conventional methods including Fluorescence Activated Cell Sorting (FACS) , microbead separation, or affinity chromatograph using antibodies specific to cell surface antigens. Alternatively, stem cells can be isolated by enzymatic digestion of source tissue and gradient separation with, e.g., PERCOLL or HISTOPAQUE. Once isolated, the stem cells can be maintained and propagated in vitro under controlled conditions. Cells may be cultured in a variety of types of vessels constructed of, for example, glass or plastic. The surfaces of culture vessels may be pre-treated or coated with, for example, collagen / polylysine, or components of the extracellular matrix, to facilitate the cellular adherence. In addition, layers of adherent cells or feeder cells, which are used to support the growth of cells with more demanding growth requirements, may be used.

[0084] Cells are normally cultured under conditions designed to closely mimic those observed in vivo. In order to mimic the normal physiological environment, cells are generally incubated in a C0 2 atmosphere with semi-synthetic growth media. Culture media is buffered and contains, among other things, amino acids, nucleotides, salts, vitamins, and also a supplement of serum such as fetal calf serum

(FCS) , horse serum or even human serum. Culture media may be further supplemented with growth factors and inhibitors such as hormones, transferrin, insulin, selenium, and attachment factors .

[0085] In certain aspects of the instant invention, cells are cultured prior to contact with a synthetic triterpenoid . They may also be cultured after contact, i.e., after they have been induced to express one or more of the genes disclosed herein or differentiate toward a given or specific phenotype . Cells will be cultured under specified conditions to achieve particular types of differentiation, and provided with various factors necessary to facilitate the desired differentiation.

[0086] In some embodiments, cells contacted with a synthetic triterpenoid are also contacted with one or more cell growth and differentiation factors. Cell growth and differentiation factors are molecules that stimulate cells to proliferate and/or promote differentiation of cell types into functionally mature forms. In some embodiments of the invention, cell growth and differentiation factors may be administered in combination with synthetic triterpenoids of the invention in order to direct the administered cells to proliferate and differentiate in a specific manner. One of ordinary skill would recognize that the various factors may be administered prior to, concurrently with, or subsequent to the administration of one or more synthetic triterpenoids of the present invention. In addition, administration of the growth and/or differentiation factors may be repeated as needed.

[0087] It is envisioned that a growth and/or differentiation factor may constitute a hormone, cytokine, hematapoietin, colony stimulating factor, interleukin, interferon, growth factor, other endocrine factor or combination thereof that act as intercellular mediators. Examples of such intercellular mediators are lymphokines, monokines, growth factors and traditional polypeptide hormones . Included among the growth factors are growth hormones such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH) , thyroid stimulating hormone (TSH) , and luteinizing hormone (LH) ; hepatic growth factor; prostaglandin, fibroblast growth factor; prolactin; placental lactogen, OB protein; tumor necrosis factors -a and ~β; mullerian- inhibiting substance; mouse gonadotropin- associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO) ; nerve growth factors such as NGF-β; platelet-growth factor; insulin-like growth factor-I and -II; erythropoietin (EPO) ; osteoinductive factors; interferons such as interferonoi, β, and -γ; colony stimulating factors (CSFs) such as macrophage-CSF (M-CSF) ; granulocyte/macrophage-CSF (GM- CSF) ; and granulocyte-CSF (G-CSF) ; interleukins (ILs) such as IL-1, IL- la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12; IL-13, IL-14, IL-15, IL-16, IL-17, IL- 18. As used herein, the term growth and/or differentiation factors include proteins from natural sources or from recombinant cell culture and biologically active equivalents of the native sequence, including synthetic molecules and mimetics.

[ 0088 ] By way of illustration, further differentiation into ectodermal cells can be achieved by culturing cells in a culture medium containing GDF-9. Further differentiation into endodermal cells can be achieved by culturing cells in a culture medium containing wnt . Further differentiation into mesodermal cells can be achieved by culturing cells in a culture medium containing Activin or Nodal . [0089] The present disclosure provides methods of inducing the expression of one or more of SOX9, COL2A1, TGF-βΙ, TGF- β2, TGF^3, BMP2, BMP4 , BMPRII, SMAD3, SMAD4, SMAD6, SMAD7 , TIMP-1 or TIMP-2, such as in a stem/progenitor cell, and the prevention or treatment of disease. In some embodiments, the expression of one or more of SOX9 , COL2A1 , TGF-βΙ, GF^2, TGF^3, BMP2, BMP4 , BMPRII, SMAD3, SMAD4 , SMAD6, SMAD7, TIMP-1 or TIMP-2 in the stem/progenitor cell induces differentiation. For example, the results presented below demonstrate that CDDO-Imidazolide (CDDO-Im) and CDDO- Ethyl amide (CDDO-EA) induce expression of each of the above -reference genes and induce chondrogenesis in mesenchymal stem cells and organ cultures of newborn mouse calvaria. Accordingly, cells exposed to synthetic triterpenoids under in vitro, in vivo, or ex vivo conditions are of use in the treatment of diseases, conditions or disorders wherein altered gene expression has been shown to provide an advantage and/or transplantation of differentiated cells would provide a benefit. Examples of synthetic triterpenoid-mediated gene induction in particular stem cells and use of the same are as follows.

Synthetic Trxterpenoid-Mediated Induction of SOX9.

[0090] Sox9 is a member of the Sox gene family, which is characterized by the presence of an HMG box with more than 50% homology to the sex determining gene Sry (Schepers, et al. (2002) Dev. Cell 3:167-170) . The HMG box can bind and bend DNA, and it has been proposed that Sox genes encode architectural DNA binding proteins. In addition to the HMG box, Sox9 possesses a transactivation domain at its C terminus (Sudbeck, et al . (1996) Nat. Genet. 13:230-232) , and it has been shown that it can activate the genes Mis in testis (De Santa Barbara, et al . (1998) Mol . Cell Biol. 18:6653-6665; Arango, et al . (1999) Cell 99:409-419) and Col2aI (Bell, et al . (1997) Nat. Genet. 16:174-178) during chondrogenesis in vitro and in vivo. Moreover, in vitro studies suggest that Sox genes may also have a role in RNA splicing (Ohe, et al . (2002) Proc . Natl. Acad. Sci . USA 99 : 1146-51) .

[ 0091] Recombinant expression of Sox9 in cultured outer root sheath (ORS) cells has been shown to induce the expression of keratin 15, increase the proliferation of ORS cells in vitro, and enhance colony- forming activity (Shi, et al . (2011) Annals Dermatol. 23:183-143). In this respect, Sox9 has potential importance in the regulation of hair follicle homeostasis. Accordingly, the induction of Sox9 in ORS cells by synthetic triterpenoids could be of use in modulating hair follicle homeostasis.

[ 0092 ] The neural crest is a transient migratory population of stem cells derived from the dorsal neural folds at the border between neural and non-neural ectoderm. Following induction, prospective neural crest cells are segregated within the neuroepithelium and then delaminate from the neural tube and migrate into the periphery, where they generate multiple differentiated cell types. Group E Sox genes (Sox8, Sox9 and SoxlO) are expressed in the prospective neural crest and Sox9 expression precedes expression of premigratory neural crest markers. Forced expression of Sox9 promotes neural-crest-like properties in neural tube progenitors at the expense of central nervous system neuronal differentiation. Subsequently, in migratory neural crest cells, SoxE gene expression biases cells towards glial cell and melanocyte fate, and away from neuronal lineages (Cheung & Briscoe (2003) Development 130:5681-93) . In this respect, Sox9 and SoxlO promote oligodendrocyte specification and terminal differentiation, respectively (Finzsch, et al . (2008) Development 13:637- 646) . Therefore, synthetic triterpenoid-mediated induction of Sox9 in NC cells could be useful in the production of oligodendrocytes and therapies for diseases such as familial dysautonomia, which affects the development and function of nerves throughout the body.

Synthetic Triterpenoid-Mediated Induction of BMP-2.

[0093] TGF-β and BMP-2 applied to undifferentiated murine embryonic stem cells have been shown to up-regulate mRNA of mesodermal- (Brachyury) and cardiac-specific transcription factors (Nkx2.5, MEF2C) (Behfar, et al . (2002) FASEB J. 16:1558-66) . Embryoid bodies generated from stem cells primed with these growth factors demonstrated an increased potential for cardiac differentiation with a significant increase in beating areas and enhanced myofibrillogenesis . In an environment of post -mitotic cardiomyocytes , stem cells engineered to express a fluorescent protein under the control of a cardiac promoter differentiated into fluorescent ventricular myocytes beating in synchrony with host cells, a process significantly enhanced by TGF-β or BMP-2. In vitro, disruption of the TGF-β/ΒΜΡ signaling pathways by latency-associated peptide and/or noggin prevented differentiation of stem cells. In vivo, transplantation of stem cells into heart also resulted in cardiac differentiation provided that TGF- /BMP-2 signaling was intact. In infarcted myocardium, grafted stem cells differentiated into functional cardiomyocytes integrated with surrounding tissue, improving contractile performance. Thus, embryonic stem cells are directed to differentiate into cardiomyocytes by signaling mediated through TGF- β/ΒΜΡ-2, a cardiac paracrine pathway required for therapeutic benefit of stem cell transplantation in diseased heart (Behfar, et al . (2002) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-2 and TGF- β in stem cells is of use in stimulating cardiomyocyte differentiation in vitro or in vivo for treatment of heart disease .

[ 0094 ] Atrioventricular (AV) endocardium transforms into the cushion mesenchyme, the primordia of the valves and membranous septa, through epithelial -mesenchymal transformation (EMT) . BMP-2 is known to be critical for AV EMT and post-EMT AV valvulogenesis . In particular, using stage-24 AV cushion mesenchymal cell aggregates cultured on 3D-collagen gels, exogenous BMP-2 was shown to induce migration of the mesenchymal cells into the collagen gels, and promote expression of periostin, a known valvulogenic matrix maturation mediator. Moreover, transcripts of Twist and Idl, which have been implicated in cell migration in embryogenesis and activation of the periostin promoter, were induced by BMP-2 (Inai, et al . (2008) Dev. Biol. 315:383-96) . Therefore, synthetic triterpenoid-mediated induction of BMP-2 in AV cushion mesenchymal cell aggregates is of use in post-EMT AV cushion tissue maturation and differentiation.

[ 0095 ] The atrioventricular heart valve leaflets and chordae tendineae are composed of diverse cell lineages and highly organized extracellular matrices that share characteristics with cartilage and tendon cell types in the limb buds and somites. During embryonic chicken valvulogenesis, aggrecan and sox9, characteristic of cartilage cells, are observed in the AV valve leaflets, in contrast to tendon-associated genes scleraxis and tenascin, present in the chordae tendineae. In the limb buds and somites, cartilage cell lineage differentiation is regulated by BMP2 , while FGF4 controls tendon cell fate. In this respect, it has been demonstrated BMP2 is sufficient to activate Smadl/5/8 phosphorylation and induce sox9 and aggrecan expression in cells from prefused endocardial cushions (Lincoln, et al . (2006) Dev. Biol. 292:292-302) . Therefore, synthetic triterpenoid-mediated induction of BMP-2 is of use in atrioventricular valve progenitor cell diversification into leaflets.

[ 0096 ] Myocardial and coronary development are both critically dependent on epicardial cells. During cardiomorphogenesis , a subset of epicardial cells undergoes an epithelial -to-mesenchymal transition (EMT) and invades the myocardium to differentiate into various cell types, including coronary smooth muscle cells and perivascular and cardiac interstitial fibroblasts. It has been demonstrated that epicardium-derived cells (EPDCs) treated with TGF-βΙ or BMP-2 obtain characteristics of smooth muscle cells (van Tuyn, et al . (2007) Stem Cells 25:271-8) . Therefore, synthetic triterpenoid-mediated induction of BMP-2 and TGF- βΐ in epicardium-derived cells may be of use in stimulating coronary smooth muscle cell differentiation.

[ 0097 ] Oligodendrocyte precursor cells (OPCs) can differentiate into oligodendrocytes or astrocytes, depending on cellular microenvironments . OPCs, cultured in medium supplemented with 10% (v/v) fetal bovine serum

(FBS) , give rise to type II astrocytes that express glial fibrillary acidic protein and a cell surface ganglioside that is recognized by A2B5 monoclonal antibody. Moreover, BMPs have been reported to be involved in astrocyte differentiation of neural progenitor cells. Therefore, it was determined whether BMPs are responsible for the serum- mediated astrocyte differentiation from OPCs (Hu, et al .

(2010) Tohoku J. Exp. Med. 222:195-200) . When OPCs were cultured in the medium containing 10% FBS, cells (more than 95%) differentiated into type II astrocytes. However, when OPCs were pretreated with noggin, the degree of astrocyte differentiation was markedly decreased from 95.39 to 38.36% (Hu, et al . (2010) supra) . Taken together, these results indicate that BMP signaling may be responsible for the serum-mediated astrocyte differentiation of OPCs. Therefore, synthetic triterpenoid-mediated induction of BMP-2 and BMP-4 in OPCs may be of use in stimulating serum- mediated astrocyte differentiation.

Synthetic Triterpenoid-Mediated Induction of BMP-4.

[0098] The neural crest-derived cell population that colonizes the bowel (ENCDC) contains proliferating neural/glial progenitors. Enteric gliogenesis has been analyzed in mice that overexpress the BMP antagonist, noggin, or BMP-4 in the primordial ENS. Noggin- induced loss-of -function decreased, while BMP4-induced gain-of- function increased the glial density and glia/neuron ratio. When added to immunoisolated ENCDC, BMPs provoked nuclear translocation of phosphorylated SMAD proteins and enhanced both glial differentiation and expression of the neuregulin receptor ErbB3. This analysis indicated that BMPs are required for enteric gliogenesis and act by promoting responsiveness of ENCDC to ErbB3 ligands such as GGF2

(Chalazonitis, et al . (2011) Dev. Biol. 350:64-79) . Therefore, synthetic triterpenoid-mediated induction of BMP-2 and BMP-4 in neural crest-derived cells is of use in stimulating enteric gliogenesis.

[0099] Bone morphogenetic proteins promote astrocytic differentiation of cultured subventricular zone stem cells. In addition, it has been demonstrated that BMPs regulate the astrocytic lineage in vivo (Gomex, et al . (2003) Dev. Biol. 255:164-77) . Transgenic mice were constructed that overexpress BMP-4 under control of the neuron-specific enolase (NSE) promoter. The overexpression of BMP-4 resulted in a remarkable 40% increase in the density of astrocytes in multiple brain regions accompanied by a decrease in the density of oligodendrocytes ranging between 11 and 26%, depending on the brain region and the developmental stage. No changes in neuron numbers or the pattern of myelination were detected, and there were no gross structural abnormalities. Coculture of transgenic neurons with neural progenitor cells significantly enhanced astrocytic lineage commitment by the progenitors; an effect which was blocked by the BMP inhibitor Noggin, indicating that the stimulation of astrogliogenesis was due to BMP-4 release by the transgenic neurons. These observations indicate that BMP-4 directs progenitor cells in vivo to commit to the astrocytic rather than the oligodendroglial lineage (Gomex, et al . (2003) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-4 in neural progenitor cells may be of use in stimulating astrocyte development .

[ 00100 ] It has been shown that that keratinocytes treated with ESC-conditioned medium (CM) change their morphology and are stimulated to express the pluripotency regulator, Oct-4, and its target transcripts, Sox-2, Nanog, Utfl and Rex-l; an effect that is mediated by BMP-4 (Grinnell & Bickenbach (2007) Cell Prolif. 40:685-70) . Pre-treated keratinocytes could be specifically directed to differentiate into cells of the neuronal lineage. The majority of responsive keratinocytes were the epidermal stem cell population, with a small percentage of transit- amplifying cells also being affected. These results indicate that ESC-CM contains a number of factors, including BMP-4, which are capable of reprogramming mouse skin keratinocytes to make them more developmentally potent, as evidenced by their ability to be re- differentiated into cells of the neuronal lineage (Grinnell & Bickenbach (2007) supra) . An increase in developmental potential combined with directed differentiation could increase the therapeutic relevancy of somatic cells. In this respect, synthetic triterpenoid-mediated induction of BMP-4 in ESCs may be of use in reprogramming keratinocytes .

[ 00101] When differentiated in the presence of activin A in serum- free conditions, mouse embryonic stem cells efficiently generate an endoderm progenitor population defined by the coexpression of either Brachyury, Foxa2 and c-Kit, or c-Kit and Cxcr4. Specification of these progenitors with BMP-4 in combination with basic fibroblast growth factor (FGF) and activin A results in the development of hepatic populations highly enriched (45-70%) for cells that express the alpha- fetoprotein and albumin proteins (Gouon-Evans (2006) Nat. Biotechnol . 24:1402-11) . These cells also express transcripts of Afp, Albl, Tat, Cpsl, Cyp7al and Cyp3all; they secrete albumin, store glycogen, show ultrastructural characteristics of mature hepatocytes, and are able to integrate into and proliferate in injured livers in vivo and mature into hepatocytes expressing dipeptidyl peptidase IV or fumarylacetoacetate hydrolase (Gouon-Evans (2006) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-4 in ESCs cultured in the presence of activin A and FGF may be of use in generating cells with an immature hepatocytic phenotype and in the treatment of injured livers.

[ 00102 ] When neural progenitors induced from ES cells in a serum- free suspension culture are subsequently treated with BMP-4 and Wnt3a, a significant proportion of these neural cells become Mathl+ (Su, et al . (2006) Dev. Biol. 290:287- 96) . The induced Mathl+ cells are mitotically active and express markers characteristic of granule cell precursors (Pax6, Zicl, and Ziprol) . After purification and coculture with postnatal cerebellar neurons, ES cell -derived Mathl+ cells exhibit typical features of neurons of the external granule cell layer, including extensive motility and a T- shaped morphology. In addition, differentiation of L7+/Calbindin-D28K+ neurons (characteristic of Purkinje cells) is induced under similar culture conditions but exhibits a higher degree of enhancement by Fgf8 rather than by Wnt3a (Su, et al . (2006) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-4 in ES cell -derived Mathl+ cells cultured in the presence of Wnt3a or Fgf8 may be of use in generating Mathl+ cerebellar granule cell precursors and Purkinje cells.

[ 00103 ] The potential of different growth factors (basic fibroblast growth factor (bFGF) , TGF-βΙ, activin-A, BMP-4, hepatocyte growth factor (HGF) , epidermal growth factor (EGF) , beta nerve growth factor (PNGF), and retinoic acid) to direct the differentiation of human ES-derived cells in vitro has been analyzed (Schuldiner, et al . (2000) PNAS 97:11307-12). Differentiation of the cells was assayed by expression of 24 cell -specific molecular markers that cover all embryonic germ layers and 11 different tissues. Each growth factor had a unique effect that may have resulted from directed differentiation and/or cell selection, and could be divided into three categories : growth factors that mainly induce mesodermal cells (Activin-A and TGF-βΙ); factors that activate ectodermal and mesodermal markers (retinoic acid, EGF, BMP-4, and bFGF); and factors that allow differentiation into the three embryonic germ layers, including endoderm (NGF and HGF) (Schuldiner, et al . (2000) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-4 and/or TGF-βΙ in human ES cells may be of use in generating mesodermal cells and/or ectodermal cells .

[ 00104 ] The limited clinical response observed in many patients with colorectal cancer may be related to the presence of chemoresistant colorectal cancer stem cells (CRC-SCs) . There is evidence that many common cancers, including skin cancer, breast cancer and leukemia, can result from transforming events that occur in adult stem cells (Perez-Losada & Balmain (2003) Nat. .Rev. Cancer 3:434-443; Al-Hajj, et al . (2003) Proc . Natl. Acad. Sci . USA 100:3983-3988; Reya, et al . (2001) Nature 414:105-111). Indeed, functional parallels exist between tumorigenic and normal stem cells. Both cell types demonstrate significant proliferative potential, the ability to self-renew, and the ability to generate new tissues. However, tumorigenic stem cells lack the normal growth regulatory mechanisms that limit the uncontrolled proliferation of stem cells (Reya, et al . (2001) supra) . Tumorigenic stem cells arise in normal adult stem cell populations through the accumulation of multiple transforming mutations. As adult stem cells can persist and self -renew for the lifespan of the individual, these cells are more likely to accrue the genetic lesions necessary for malignant transformation. Such transformed tumorigenic stem cells, arising in normal adult stem cell populations, can initiate cancer development (Reya, et al . (2001) supra) . Furthermore, tumorigenic stem cells may also play important roles in tumor evolution, metastatic invasion and local recurrence following treatment.

[ 00105 ] Cancer stem cells constitute only a small proportion of a tumor or a cancerous tissue. But the cancer stem cells have a unique ability to establish new colonies of cancer cells. For example, when mouse myeloma cells are obtained from mouse ascites, separated from normal hematopoietic cells, and put into in vitro colony- forming assays, only 1 in 10,000 to 1 in 100 cancer cells were able to form colonies (Park, et al . (1971) J " . Nat. Cancer Inst. 46:411- 422) . Even when leukemic cells are transplanted in vivo, only 1-4% of cells form spleen colonies (Bruce, et al . (1963) Nature 199:79-80; Bergsagel, et al . (1968) Cancer Res. 28 : 2187-2196) .

[ 00106 ] It has been demonstrated that BMP-4 promotes the differentiation of normal colonic stem cells. Therefore, it was determined whether BMP-4 might be used to induce differentiation of CRC-SCs for therapeutic purposes (Lombardo, et al . (2011) Gastroenterology 140:297-309) . CRC-SCs were isolated from tumor samples based on expression of CD133 or using a selection culture medium. BMP-4 expression and activity on CRC-SCs were evaluated and it was found that CRC-SCs did not express BMP-4 whereas differentiated cells did. Recombinant BMP-4 promoted differentiation and apoptosis of CRC-SCs; this effect did not depend on Smad4 expression level or microsatellite stability. BMP-4 activated the canonical and noncanonical BMP signaling pathways, including phospholnositide 3 -kinase (PI3K) and PKB (protein kinase B) /AK . The potential therapeutic effect of BMP-4 was also assessed in immunocompromised mice after injection of CRC-SCs that responded to chemotherapy or that did not. Administration of BMP-4 to immunocompromised mice with tumors that arose from CRC-SCs increased the antitumor effects of 5- fluorouracil and oxaliplatin. BMP-4 promoted terminal differentiation, apoptosis, and chemosensitization of CRC- SCs in tumors that did not have simultaneous mutations in Smad4 and constitutive activation of PI3K (Lombardo, et al . (2011) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-4 in CRC-SCs may be of use in combination with anti-cancer agents in the treatment of advanced colorectal tumors .

[ 00107 ] Human glioblastoma is a heterogenous tumor composed of tumor cells and a small portion of cancer stem cells, which have a high tumorigenic potential and a low proliferation rate. Glioma cancer stem cells are phenotypically similar to the normal stem cells, they express CD133 gene and other genes characteristic of neural stem cells and posses the self -renewal potential. Cancer stem cells derived from glioblastoma are capable of recapitulating original polyclonal tumors when xenografted to nude mice. They are chemoresistant and radioresistant and therefore responsible for tumor progression and recurrence after conventional glioblastoma therapy. BMP-4 has been shown to trigger a significant reduction in the stem-like, tumor-initiating precursors of human glioblastomas (GBMs) (Piccirillo, et al . (2006) Nature 444:761-765) . Transient in vitro exposure to BMP-4 abolishes the capacity of transplanted GBM cells to establish intracerebral GBMs. Most importantly, in vivo delivery of BMP-4 effectively blocks the tumor growth and associated mortality that occur in 100% of mice after intracerebral grafting of human GBM cells. BMP-4 activates its cognate receptor and triggers the Smad signaling cascade in cells isolated from human GBMs. This is followed by a reduction in proliferation, and increased expression of markers of neural differentiation, with no effect on cell viability. The concomitant reduction in clonogenic ability, in the size of the CD133 + population and in the growth kinetics of GBM cells indicates that BMP-4 reduces the tumor-initiating cell pool of GBMs (Piccirillo, et al . (2006) supra) . Therefore, synthetic triterpenoid-mediated induction of BMP-4 in chemoresistant GBM cells may be of use in the treatment of GBM.

Synthetic Triterpenoid-Mediated Induction of TGF-βΙ.

[00108] Engineering vascularized tissue is crucial for its successful implantation, survival, and integration with the host tissue. Vascular smooth muscle cells (v-SMCs) provide physical support to the vasculature and aid in maintaining endothelial viability. By supplementing ESCs with platelet- derived growth factor-BB (PDGF-BB) and TGF-βΙ, human ESCs could be differentiated into smooth-muscle-like cells

(SMLCs) (Vo, et al . (2010) Stew Cell Rev. 6:237-47) . The SMLCs highly expressed specific smooth muscle cell (SMC) markers including alpha-smooth muscle actin, calponin, SM22, and smooth muscle myosin heavy chain; secreted fibronectin and collagen; and contracted in response to carbachol . In vitro tubulogenesis assays revealed that these hESC-derived SMLCs interacted with human endothelial progenitor cell (EPCs) to form longer and thicker cord- like structures in vitro thereby supporting and augmenting capillary-like structures (Vo, et al . (2010) supra) . Therefore, synthetic triterpenoid-mediated induction of TGF-βΙ in ESCS may be of use in combination with PDGF-BB to produce smooth-muscle-like cells for therapeutic vascular tissue engineering.

[00109] Stem cell factor is essential to the migration and differentiation of melanocytes during embryogenesis based on the observation that mutations in either the stem cell factor gene, or its ligand, KIT, result in defects in coat pigmentation in mice. Stem cell factor is also required for the survival of melanocyte precursors while they are migrating towards the skin. Using an immature melanocyte precursor cell line, it has been demonstrated that TGF-βΙ promotes melanocyte precursor proliferation in autocrine and/or paracrine regulation (Kawakami, et al . (2002) J " . Invest. Dermatol. 118:471-8) . In addition, an anti-TGF-βΙ antibody decreased the number of KIT-positive neural crest cells in a neural crest cell primary culture system, and abolished the growth of the neural crest cells in a wild- type neural crest explant. These results indicate that TGF- βΐ affects melanocyte precursor proliferation and differentiation in the presence of stem cell factor/KIT in an autocrine/paracrine manner (Kawakami, et al . (2002) supra) . Therefore, synthetic triterpenoid-mediated induction of TGF-βΙ in neural crest stem cells or melanocyte precursor cells may be of use in combination with stem cell factor and KIT to produce melanocytes for therapeutic strategies in the treatment of pigment cell disorders .

Synthetic Trxterpenoid-Mediated Inductxon of TGF-p2.

[00110] Oligodendrocyte progenitor cells derived from human embryonic stem cells have been reported to remyelinate axons and improve locomotor function in a rodent model of spinal cord injury. It has been demonstrated that human ES cell -derived OPCs express functional levels of midkine, hepatocyte growth factor (HGF) , activin A, TGF-P2, and brain-derived neurotrophic factor (BDNF) , proteins with reported trophic effects on neurons (Zhang, et al . (2006) Stem Cells Dev. 15:943-52) . The neurotrophic activity of hES cell -derived OPCs was further demonstrated by stimulatory effects on neurite outgrowth of adult rat sensory neurons in vitro (Zhang, et al . (2006) supra) . Therefore, synthetic triterpenoid-mediated induction of TGF^2 in human ES cell -derived OPCs may be of use in remyelinating axons and improving locomotor function for therapeutic strategies in the treatment of spinal cord injury.

[00111] The effects of TGF- 2 and glial cell line-derived neurotrophic factor (GDNF) in the differentiation of midbrain progenitors toward a dopaminergic phenotype have been analyzed (Roussa, et al . (2008) Stem Cells 26:1683- 94) . This analysis indicated that neurturin and persephin

(PSPN) , but not GDNF, were capable of transient induction of dopaminergic neurons in vitro. This process, however, required the presence of endogenous TGF-β. In vitro, combined TGF- /PSPN treatment achieved a yield of approximately 20% TH-positive cells that were less vulnerable against 1 -methyl -4 -phenyl pyridinium ion toxicity. These results indicate that TGF-β is required for the induction of dopaminergic neurons, whereas GDNF is required for regulating and/or maintaining a differentiated neuronal phenotype (Roussa, et al . (2008) supra). Therefore, synthetic triterpenoid-mediated induction of TGF- 2 in midbrain progenitor cells, in combination with PSPN, may be of use as a potent inductive cocktail for the generation of dopaminergic neurons of use in tissue engineering and cell replacement therapies for Parkinson's disease .

Synthetic Triterpenoid-Mediated Induction of TGF- 3.

[00112] Skin grafts are frequently used for a variety of indications in plastic and reconstructive surgery. Their necrosis is a common complication, while different therapies have been proposed. Currently, ASCs hold great promise for their angiogenic potential and role during tissue repair. In this respect, it has been demonstrated that autologous transplantation of ASCs can increase angiogenesis , skin-graft survival and wound healing (Zografou, et al . (2011) J " . Plast. Reconstr. Aesthet Surg. 64:1647-56). ASCs were isolated, cultured, labelled with fluorescent dye and injected under full -thickness skin grafts in rats. Collagen ' s framework and angiogenesis were assessed, as were levels of vascular endothelial growth factor (VEGF) and TGF-p3. Statistically significant increases of microvessel density, collagen density, VEGF and TGF- 3 expression were noted in the ASC-treated group. Therefore, synthetic triterpenoid-mediated induction of TGF- 3 in ASCs may be of use in increasing full -thickness skin-graft survival in skin-graft surgery.

[ 00113 ] Many inductive factors participate in the initial endocardial to mesenchymal transformation event necessary to form the prevalvular cushion. Using embryonic valve progenitor cells in in vitro three-dimensional tissue- engineered models of embryonic valvular remodeling with in vivo analysis, the roles of three prominent growth factors during avian mitral valvulogenesis have been determined (Chiu, et al . (2010) Tissue Engin. Part A 16:3375-83). This analysis indicated that TGF-p3, BMP-2, and vascular endothelial growth factor A (VEGFA) are expressed in spatiotemporally distinct patterns and at different levels within remodeling embryonic valves in vivo. TGF-p3 induced cell migration, invasion, and matrix condensation; BMP-2 induced invasion; and VEGFA inhibited invasion but increased migration (Chiu, et al . (2010) supra) . Therefore, synthetic triterpenoid-mediated induction of TGF-P3 and BMP-2 in embryonic valve progenitor cells may be of use in facilitating valvulogenic remodeling and phenotype maturation, which can be integrated into clinically needed regenerative strategies for heart valve disease and to accelerate the development of engineered tissue valves. [00114] In culture, BMP-2 and TGF-β promote neurogenesis at the expense of a smooth-muscle-like fate in clusters of neural-crest-derived multipotent progenitor cells. In this respect, neurons generated by TGF-β factors belong to the autonomic lineage and cells within the developing sympathetic ganglia express TGF^-type II receptor. It has been suggested that TGF-β functions in a biphasic manner during autonomic gangliogenesis to control both neurogenesis and subsequently the number of neurons generated from progenitor cells (Hagedorn, et al . (2000) Dev. Biol. 228:57-72) . Therefore, synthetic triterpenoid- mediated induction of TGF- 3 in neural-crest-derived multipotent progenitor cells may be of use in facilitating neurogenesis and the treatment of neurodegenerative diseases such as Parkinson's disease.

Synthetic Triterpenoid-Mediated Induction of Smad3.

[00115] It has been demonstrated that Smad3-null mice have decreased muscle mass and pronounced skeletal muscle atrophy (Ge, et al . (2011) Cell Res. 21:1591-604). Consistent with this, increases in protein ubiquitination and elevated levels of the ubiquitin E3 ligase uRFl is observed in muscle tissue isolated from Smad3-null mice. Loss of Smad3 also led to defective satellite cell (SC) functionality. Smad3-null SCs showed reduced propensity for self -renewal , which may lead to a progressive loss of SC number. Decreased SC number was observed in skeletal muscle from Smad3-null mice showing signs of severe muscle wasting. Further in vitro analysis of primary myoblast cultures identified that Smad3-null myoblasts exhibit impaired proliferation, differentiation and fusion, resulting in the formation of atrophied myotubes . A search for the molecular mechanism revealed that loss of Smad3 results in increased myostatin expression in Smad3-null muscle and myoblasts. Given that myostatin is a negative regulator, increased myostatin levels appear to be responsible for the atrophic phenotype in Smad3-null mice. Consistent with this theory, inactivation of myostatin in Smad3-null mice rescues the muscle atrophy phenotype (Ge, et al. (2011) Cell Res. 21:1591-604) . Given the role of Smad3 in satellite cell function, synthetic triterpenoid- mediated induction of Smad3 in satellite cells may be of use in facilitating muscle differentiation and fusion to augment existing muscle fibers and to form new fibers following injury or disease.

[ 00116 ] It has been shown that treatment of mouse embryonic stem cells and human MSCs from fetal membranes of term placenta with hyaluronan mixed esters of butyric and retinoic acids (HBR) facilitates stem cell cardiogenesis

(Maioli, et al . (2010) PLoS One 5:el5151) . HBR differentially affects the patterning of Smad proteins by enhancing gene and protein expression of Smadl, 3, and 4, while down-regulating Smad7. Chromatin immune precipitation and transcriptional analyses showed that HBR increased the transcription of the cardiogenic gene Nkx-2.5 through Smad4 binding to its own consensus Smad site. Treatment of mouse embryonic stem cells and MSCs with HBR led to the concomitant overexpression of both Smad4 and a-sarcomeric actinin. Smad4 silencing by the aid of lentiviral -mediated Smad4 shRNA confirmed a dominant role of Smad4 in HBR- induced cardiogenesis (Maioli, et al . (2010) supra) . Therefore, synthetic triterpenoid-mediated induction of Smad4 in embryonic stem cells or MSCs is of use in facilitating stem cell cardiogenesis in the treatment of cardiac disease and injury. [ 00117 ] Neuronal differentiation mechanisms following treatment of neural stem/progenitor cells (NPCs) with a Gl/S-phase cell cycle blocker have been investigated

(Misumi, et al . (2008) Eur. J. Neurosci . 28:1049-59). The addition of deferoxamine (DFO) or aphidicolin (Aph) to neurospheres for 8 hours, followed by 3 days of differentiation, resulted in an increased number of neurons and neurite outgrowth. DFO induced enhanced expression of TGF-βΙ and cdk5 at 24 hours after differentiation, whereas Aph only increased TGF-βΙ expression. DFO-induced neurogenesis and neurite outgrowth were attenuated by administration of a cdk5 inhibitor, roscovitine, suggesting that the neurogenic mechanisms differ between DFO and Aph. TGF-βΙ did not increase neurite outgrowth but rather the number of beta-tubulin III -positive cells, which was accompanied by enhanced p27(kipl) mRNA expression. In addition, TGF-β receptor type II expression was observed in nestin-positive NPCs. Results indicated that DFO-induced TGF-βΙ signaling activated Smad3 translocation from the cytoplasm to the nucleus. In contrast, TGF-βΙ signaling inhibition, via a TGF-β receptor type I inhibitor (SB- 505124), resulted in decreased DFO-induced neurogenesis, in conjunction with decreased p27 (kipl) protein expression and Smad3 translocation to the nucleus. These results indicate that cell cycle arrest during Gl/S-phase induces TGF-βΙ expression. This, in turn, prompts enhanced neuronal differentiation via Smad3 translocation to the nucleus and subsequent p27 (kipl) activation in NPCs (Misumi, et al . (2008) supra) . Therefore, synthetic triterpenoid-mediated induction of Smad3 and/or TGF-βΙ in neural progenitor cells is of use in facilitating neurogenesis and neurite outgrowth in the treatment of neurodegenerative disease or nerve injury. Synthetic Triterpenoid-Mediated Induction of Smad4.

[00118] In the mammalian brain, neurogenesis continues only in few regions of the forebrain. It has been shown that BMP-mediated signaling is active in adult neural stem cells and is crucial to initiate the neurogenic lineage in the adult mouse subependymal zone (Colak, et al . (2008) J. Neurosci . 28:434-46) . Conditional deletion of Smad4 in adult neural stem cells severely impairs neurogenesis, and this is phenocopied by infusion of Noggin, an extracellular antagonist of BMP. Smad4 deletion in stem, but not progenitor cells, as well as Noggin infusion lead to an increased number of 01ig2 -expressing progeny that migrate to the corpus callosum and differentiate into oligodendrocytes. Transplantation experiments further verified the cell -autonomous nature of this phenotype . Thus, BMP-mediated signaling via Smad4 is required to initiate neurogenesis from adult neural stem cells and suppress the alternative fate of oligodendrogliogenesis (Colak, et al . (2008) supra) . Accordingly, synthetic triterpenoid-mediated induction of Smad4 in adult neural stem cells may be of use in facilitating neurogenesis in the treatment of neurodegenerative disease or nerve injury.

Synthetic Triterpenoid-Mediated Induction of Smad6.

[00119] Neural stem cells were differentiated into astrocytes to analyze gene expression patterns (Park, et al . (2012) Neurosci. Lett. 506:50-4) . Amongst the analyzed genes, 1898 genes were up-regulated relative to the neural stem cells, whereas 1642 genes were down-regulated. The up- regulated genes included Gfap, Smad6, Fst, Tgfb2 and Cdkn2. The down-regulated genes included Ccnbl, Ccndl and Ccnd2. In addition, gene networks associated with BMP and signaling pathways were identified using Ingenuity Pathway Analysis. These results suggest that the differentiation of neural stem cells into astrocytes is based on a combinatorial network of various signaling pathways, including cell cycle, BMP and TGF- 2 signaling (Park, et al . (2012) supra) . Accordingly, synthetic triterpenoid- mediated induction of Smad6 and/or BMP and/or TGF- 2 in adult neural stem cells may be of use in facilitating astrocyte formation in the repair of the brain or spinal cord following traumatic injury.

Synthetic Triterpenoxd-Mediated Induction of Smad7.

[00120] The role of Smad7 during skeletal muscle cell differentiation has been assessed (Kollias, et al . (2006) Mol . Cell Biol. 26:6248-60) . In these studies, increased expression of Smad7 has been shown to abrogate myostatin- but not TGF- βΐ-mediated repression of myogenesis. Further, constitutive expression of exogenous Smad7 potently enhances skeletal muscle differentiation and cellular hypertrophy. Conversely, targeting of endogenous Smad7 by small interfering RNA inhibits muscle cell differentiation, indicating an essential role for Smad7 during myogenesis. Congruent with a role for Smad7 in myogenesis, it has been observed that the muscle regulatory factor (MyoD) binds to and transact ivates the Smad7 proximal promoter region. Furthermore, Smad7 directly interacts with MyoD and enhances MyoD transcriptional activity. Thus, Smad7 cooperates with MyoD, creating a positive loop to induce Smad7 expression and to promote MyoD driven myogenesis. Taken together, these data implicate Smad7 as a fundamental regulator of differentiation in skeletal muscle cells (Kollias, et al . (2006) supra) . Accordingly, synthetic triterpenoid-mediated induction of Smad7 may be of use in facilitating skeletal muscle cell differentiation in the repair of the muscle following traumatic injury or surgery.

[ 00121] As indicated herein, synthetic triterpenoids induce gene expression and stem/progenitor cell differentiation. In this respect, the present invention also provides a method for the treatment of a patient suffering from a degenerative disease or injury by transplantation into said patient of a population of stem/progenitor cells treated with a synthetic triterpenoid . Transplantation can be achieved using any conventional stem cell transplant procedure with or without a scaffold or matrix. In accordance with some embodiments, degenerative diseases or injuries that may be treated in accordance with this method of the invention, include but are not limited to, skin wounds or injury, muscle injury, Parkinson's disease, diabetes, nerve injury including spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, cardiac injury or disease, baldness, myocardial infarction, muscular dystrophy, or liver disease or injury. In addition, the synthetic triterpenoids of the instant invention find application in the treatment of congenital disorders. In some embodiments, the congenital disorder is familial dysautonomia, a congenital heart defect {e.g., congenital heart valve disease) , a dysganglionic disorder in the gastrointestinal tract such as hypoganglionosis or Hirschsprung's disease, or a pigment cell disorder such as vitiligo. Furthermore, the synthetic triterpenoids of the invention find application in the treatment of resistant cancers such as chemoresistant colorectal cancer or gliomas. Embodiments of this method of the invention, therefore extend to the use of cells prepared according the present invention in the preparation of a medicament for the treatment of a degenerative disease, injury or congenital disorder.

[ 00122 ] Exemplary stem/progenitor cells, which can be treated with a synthetic triterpenoid, the result of treatment, and the therapeutic use of said cells are listed in Table 1.

TABLE 1

Result of Treatment

Stem/

with a Synthetic Therapeutic Uses Progenitor Cell

Triterpenoid

Adipose tissue- Induce differentiation

Increase skin- derived of skin cells and

graft survival in mesenchymal stimulation of

skin-graft surgery stem cells angiogenesis

Atrioventricular

Repair of cardiac diversification into

inj ury

Valve leaflets

progenitor Valve tissue

Valvulogenic remodeling

cells engineering and and phenotype

treatment of heart maturation

valve disease

Differentiation and Treatment of

Chemoresistant

apoptosis of advanced

cancer stem

chemoresistant cancer colorectal or GBM cells

stem cells tumors

Induce cardiomyocyte Treatment of heart differentiation disease and injury

Reprogram keratinocytes Treatment of for differentiation Neurodegenerative into neuronal lineage disease

Produce cells with

Treatment of liver immature hepatocytic

injury or disease phenotype

Embryonic stem

Treatment of cells

Induce neural cell neurodegenerative differentiation disease or brain injury

Induce mesodermal cell Treatment of and/or ectodermal cell muscle, skin or differentiation neuronal injury

Induce production of

Vascular tissue smooth-muscle-like

engineering cells Epicardium-

Stimulate coronary Treatment of heart derived

smooth muscle cell disease or cardiac progenitor

differentiation injury

cells

Treatment of

Melanocyte Induce production of

pigment cell precursor cells melanocytes

disorders

Induce cardiogenesis

or atrioventricular Treatment of heart

Mesenchymal

cushion tissue disease or cardiac stem cell

maturation and injury

differentiation

Midbrain Treatment of

Induce production of

progenitor neurodegenerative dopaminergic neurons

cells disease

Treatment of

Induce production of

familial

oligodendrocytes

dysautonomia

Treatment of

Induce production of

pigment cell melanocytes

Neural crest disorders

cells Treatment of

Stimulate enteric

neurodegenerative gliogenesis

disease

Treatment of

Induce neurogenesis neurodegenerative diseases

Induce astrocyte Treatment of

Neural development neurodegenerative progenitor disease or nerve,

Induce neurogenesis and

cells brain or spinal neurite outgrowth

cord injury

Treatment of

Induce neurogenesis neurodegenerative

Neural stem

disease or nerve, cells Induce astrocyte brain or spinal development cord injury

Induce serum-mediated

astrocyte

Oligodendrocyte Treatment of

differentiation

progenitor neural or spinal

Induce remyelination of

cells cord injury

axons and improve

locomotor function

Treatment of

Outer root Maintain hair follicle

aberrant hair sheath cells homeostasis

growth

Induce muscle Treatment of

Satellite cells

differentiation and muscle injury or fusion disease

Repair of the

Skeletal muscle

Induce skeletal muscle muscle following cell

cell differentiation traumatic injury progenitors

or surgery

[ 00123 ] For the purposes of the present invention, "prevention" or "preventing" includes 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 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.

[ 00124 ] "Treatment" or "treating" includes 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) , 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 effecting any measurable decrease in a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease.

[ 00125 ] An "effective amount," "therapeutically effective amount" or "pharmaceutically effective amount" means that amount which, when administered to a subject or patient for treating a disease, is sufficient to effect such treatment for the disease.

[ 0012 6 ] In addition to the above-described novel compounds, one or more of the following synthetic triterpenoids can be used in the methods of this invention. In particular embodiments, additional synthetic triterpenoids of use in the instant methods have the structure of Formula I, which includes hydrates, isomers, prodrugs or pharmaceutically acceptable salts of Formula I :

Formula I

wherein,

X 1 and X 2 are independently

hydrogen, OR a , NR a R b , or SR a , wherein

R a is a hydrogen, cyano, -CF 3 , nitro, amino, or substituted or unsubstituted heteroaryl group;

R b is hydrogen, hydroxyl , alkyl, aryl , aralkyl, acyl, alkoxy, aryloxy, acyloxy, alkylamino, arylamino, amido, or a substituted version of any of these groups;

or a substituent convertible in vivo to hydrogen; provided that R a is absent when the atom to which it is bound is part of a double bond, further provided that when R a is absent the atom to which it is bound is part of a double bond;

Y is CH 2 or CH 2 -C¾;

Z is a covalent bond, -C(=0)-, alkanediyl, alkenediyl, alkynediyl, or a substituted version of any of these groups ;

the dashed bonds can be independently present or absent ; R 1 , R 2 , R 3 and R 4 are each independently a hydrogen, hydroxyl, alkyl, substituted alkyl, alkoxy or substituted alkoxy group;

R 5 , R 6 , R 7 , R 8 , R 9 and R 10 are independently hydrogen, hydroxyl, halo, cyano, -C≡CR a , -C0 2 R a , -COR a , alkyl, alkenyl, alkynyl, aryl , aralkyl, heteroaryl , heteroaralkyl , acyl , alkoxy, aryloxy, acyloxy, alkylamino, arylamino, nitro, amino, amido, -C(0)R c or a substituted version of any of these groups, wherein

R c is hydrogen, hydroxy, halo, amino, hydroxyamino, azido or mercapto; or Ci-C 15 -alkyl , C 2 -Ci 5 -alkenyl , C 2 -Ci 5 - alkynyl, C 6 -Ci 5 -aryl, C 7 -C ls -aralkyl , Ci-Ci S -heteroaryl , C 2 - Ci 5 -heteroaralkyl , Ci~C 15 -acyl , C 1 -C15- alkoxy, C 2 -Ci 5 - alkenyloxy, C 2 -Ci 5 -alkynyloxy, C 6 -Ci 5 -aryloxy, C 7 -C 15 - aralkyloxy, Ci-Cis-heteroaryloxy, C 2 -C 15 -heteroaralkyloxy, C x - Ci 5 -acyloxy, Cx-Cxs-alkylamino , C 2 -Ci 5 -dialkylamino, Cx-Cis- alkoxyamino, C 2 -C 15 -alkenylamino, C 2 -C 15 -alkynylamino , C 6 -Ci 5 - arylamino, C 7 -Ci 5 -aralkylamino, Ci-C 15 -heteroarylamino, C 2 - Ci 5 -heteroaralkylamino, Cx-Cis-alkylsulfonylamino, C 1 -C15- amido, Ci-Ci 5 -alkylsilyloxy, or substituted versions of any of these groups;

R 5 and R 6 , R 7 and R 8 , or R 9 and R 10 are independently taken together as =0;

R 11 and R 12 are each independently hydrogen, hydroxyl, halo, alkyl , alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, acyl, alkoxy, aryloxy, aralkoxy, heteroaryloxy, hetero-aralkoxy, acyloxy, alkylamino, dialkylamino , arylamino, aralkylamino, heteroarylamino, heteroaralkylamino, amido, or a substituted version of any of these groups, or

R 11 and R 12 are taken together and are alkanediyl, alkenediyl, arenediyl , alkoxydiyl, alkenyloxydiyl, alkylaminodiyl , alkenylaminodiyl , or alkenylaminooxydiyl; R is hydrogen, hydroxy or oxo;

R 14 is hydrogen or hydroxy1 ; and

R 15 is a hydrogen, hydroxy1 , -NR d R e , cyano, halo, azido, phosphate, 1 , 3 -dioxoisoindolin-2 -yl , mercapto, silyl or - COOH group,

substituted or unsubstituted versions of C 1 -C15- alkyl, C 2 -Ci 5 -alkenyl , C 2 -C 15 -alkynyl , C 6 -C 15 -aryl, C 7 -C 15 - aralkyl, Ci-Ci 5 -heteroaryl , C 2 -Ci 5 -heteroaralkyl , Ci-Ci 5 -acyl, Ci-Ci 5 -alkoxy, C 2 -Ci 5 -alkenyloxy, C 2 -Ci 5 -alkynyloxy, C 6 -C 15 - aryloxy, C 7 -Ci S -aralkyloxy, Cx-Cis-heteroaryloxy, C 2 -Ci 5 - heteroaralkyloxy, Ci-Ci 5 -acyloxy, Ci-Ci 5 -alkylamino, C 2 -Ci 5 - alkenylamino, C 2 -Ci 5 -alkynylamino, C 6 -Ci 5 -arylamino, C 7 -Ci 5 - aralkylamino, Ci-Cxs-heteroarylamino, C 2 -Ci 5 - heteroaralkylamino, Ci-Ci 5 -amido, Ci-Cas-alkylthio, C 2 -Ci 5 ~ alkenylthio, C 2 -Ci 5 -alkynylthio, C e -Ci 5 -arylthio, C 7 -Ci 5 - aralkylthio, Cx-Cis-heteroarylthio, C 2 -C 15 -heteroaralkylthio , Ci-Ci 5 -acylthio, Ci-Ci 2 -thioacyl , C ! -C 12 -alkylsulfonyl , C 2 -C 12 - alkenylsulfonyl , C 2 -Ci 2 -alkynylsulfonyl , C 3 -Ci 2 ~arylsulfonyl , C 7 -C 12 -aralkylsulfonyl , Ci-Ci 2 -heteroarylsulfonyl , C x -C 12 - heteroaralkylsulfonyl , C ! -C 12 -alkylsulfinyl , C 2 -C X2 - alkenylsulfinyl , C 2 -Ci 2 -alkynylsulfinyl , C 6 -Ci 2 -aryl sulfinyl, C 7 -Ci 2 -aralkylsulfinyl , Ci-Ci 2 -heteroarylsulfinyl , Ci-Ci 2 -heteroaralkylsulfinyl , Cx-C^-alkylphosphonyl , C -C 12 - alkylphosphate , C 2 -Ci 2 -dialkylphosphate, Ci-Ci 2 - alkylammonium, Ci-Ci 2 -alkylsulfonium, Ci-C 15 -alkylsilyl , or a substituted version of any of these groups,

a -C0 2 Me, carbonyl imidazole, -CO-D-Glu (OAc) 4 , CONH 2 , -CONHNH 2 , -CONHCH 2 CF 3 , or -C (=0) -heteroaryl group, or

Z and R 15 form a three to seven-membered ring, such that Z and R 15 are further connected to one another through one or more of -0- and alkanediyl, further wherein Z is - CH- and R 15 is -CH 2 - or Z, R 15 , and carbon numbers 13, 17 and 18 form a ring such that R 15 is bound to carbon 13, wherein Y is methanediyl or substituted methanediyl and R is -0-, wherein

R d and R e are independently hydrogen, hydroxyl , alkyl, alkenyl , alkynyl, aryl , aralkyl, heteroaryl, heteroaralkyl , acyl , alkoxy, alkenyloxy, alkynyloxy, aryloxy, aralkoxy, heteroaryloxy , heteroaralkoxy, thioacyl , alkylsulfonyl , alkenylsulfonyl , alkynylsulfonyl , arylsulfonyl , aralkylsulfonyl , heteroaryl sulfonyl , or heteroaralkylsulfonyl , or a substituted version of any of these groups.

[00127] In certain embodiments, the bond between C 2 and C 3 in the A-ring is a double bond. In other embodiments, the bond between C 2 and C 3 in the A-ring is a single bond.

[00128] Definitions of alkyl, aryl, etc. are as defined for the substituents of Formula I and Formula II above. In addition, the term "alkylsulfonium" refers to the group - SRR ' , in which R and R' can be the same or different alkyl groups, or R and R 1 can be taken together to represent an alkanediyl . Non-limiting examples of alkylsulfonium groups include -SH(CH 3 ) , -SH(CH 2 CH 3 ) , -SH (CH 2 CH 2 CH 3 ) , -S(CH 3 ) 2 , S(CH 2 CH 3 ) 2 , -S (CH 2 CH 2 CH 3 ) 2 , -SH (cyclopentyl) , and SH (cyclohexyl) .

[00129] Exemplary compounds of use in the methods of the present invention include those shown below.

CDDO CDDO-Me CDDO-MA

CDDO-TFEA CDDO-EA CDDO-Im

[00130] The synthetic triterpenoids of the present disclosure may be provided to a stem/progenitor cell ex vivo, in vitro or in vivo. When used under ex vivo or in vivo conditions, the stem/progenitor cell may be cultured under conventional cell culture conditions in the presence of a synthetic triterpenoid to induce the expression of one or more the genes described herein. As described herein, additional cell growth and differentiation factors may be employed to facilitate differentiation of the stem/progenitor cells. Stem/progenitor cells treated in such a manner can then be transplanted into a subject for therapeutic purposes.

[00131] The present invention also extends to articles coated with one or more synthetic triterpenoids, such as tissue culture dishes, multi-well plates {e.g., 1, 2, 4, 8, 24, 48, 96-wells, etc) , PETRI -dishes , tissue culture flasks, fermentors, bioreactors, etc. for differentiating stem/progenitor cells. Such articles may be composed of any generally suitable material, such as a plastics material, for example polypropylene, or other materials such as glass, metal, etc. Suitable metals include mirror-polished metals, e.g., mirror-polished stainless steel.

[ 00132 ] When used in vivo, i.e., administered directly to a subject, the triterpenoid compounds of this invention may be administered in a pharmaceutical composition by various routes including, but not limited to, oral, subcutaneous, intravenous, or intraperitoneal administration (e.g. by injection) . Depending on the route of administration, the active compound 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 into a subject or wound site.

[ 00133 ] To administer the therapeutic compound by other than parenteral administration, it may be necessary 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, et al . (1984) J " . Neuroimmunol . 7:27).

[ 00134 ] The therapeutic compound may also be administered parenterally, intraperitoneally, intraspinally, or intracerebrall . Dispersions can be prepared in glycerol, liquid polyethylene 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.

[ 00135 ] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. 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 (for example, 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 polyalcohols 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.

[ 00136 ] Sterile injectable solutions can be prepared by incorporating the synthetic triterpenoid 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 synthetic triterpenoid into a sterile carrier which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case 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 synthetic triterpenoid) plus any additional desired ingredient from a previously sterile-filtered solution thereof.

[00137] The synthetic triterpenoid can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The synthetic triterpenoid and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the patient's diet. For oral therapeutic administration, the synthetic triterpenoid 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 synthetic triterpenoid in the compositions and preparations may, of course, be varied. The amount of the synthetic triterpenoid in such therapeutically useful compositions is such that a suitable dosage will be obtained.

[00138] 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 patients to be treated; each unit containing a predetermined quantity of synthetic triterpenoid 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 synthetic triterpenoid and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a synthetic triterpenoid for the treatment of a selected condition in a patient.

[ 00139 ] The synthetic triterpenoid may also be administered topically to the skin, eye, or mucosa. Alternatively, if local delivery to the lungs is desired the synthetic triterpenoid may be administered by inhalation in a dry- powder or aerosol formulation.

[ 00140 ] In yet another embodiment, the synthetic triterpenoid can be administered as a coating on an article for implantation. Such articles include polymer scaffolds containing stem/progenitor cells, stents, shunts, and the like.

[ 00141] Active compounds are administered at a therapeutically effective dosage sufficient to treat a condition associated with a given patient. For example, the efficacy of a synthetic triterpenoid 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 described herein. A "therapeutically effective dosage" preferably reduces the amount of symptoms of the condition in the infected subject by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. 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.

[ 00142 ] The actual dosage amount of a synthetic triterpenoid of the present disclosure or composition comprising a synthetic triterpenoid of the present disclosure administered to a patient 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 patient 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 patient. The dosage may be adjusted by the individual physician in the event of any complication.

[ 00143 ] 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 administrations daily, for one or several days (depending of course of the mode of administration and the factors discussed above) . Other suitable dose ranges include 1 mg 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.

[ 00144 ] 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. In other non-limiting examples, a dose may also comprise from about 1 microgram/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 milligram/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 milligram/kg/body weight, etc., can be administered, based on the numbers described above.

[00145] Single or multiple doses of the synthetic triterpenoids 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, patients may be administered two doses daily at approximately 12 hour intervals. In some embodiments, the synthetic triterpenoid is administered once a day.

[00146] The synthetic triterpenoid 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 there-between . 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 synthetic triterpenoid may taken orally and that the timing of which is or is not dependent upon food intake. Thus, for example, the synthetic triterpenoid can be taken every morning and/or every evening, regardless of when the patient has eaten or will eat .

[00147] The invention is described in greater detail by the following non-limiting examples.

Example 1: Materials and Methods

[00148] Reagents. The synthesis of the triterpenoids has been described (Liby, et al . (2007) Nat. Rev. Cancer 7:357- 69; Sporn, et al . (2011) J " . Nat. Prod. 74:537-45) . All other chemicals were from Sigma-Aldrich .

[00149] Calvarial Organ Cultures . Details have been published previously (Garrett, et al . (2003) J. Clin. Invest. 111:1771-82) . Calvaria were cultured in BGJ medium supplemented with 1 mg/mL of bovine serum albumin (Cohn fraction V) , 100 U/ml each of penicillin/streptomycin, and 0.292 mg/mL of glutamine . On Day 1, the calvaria were treated with synthetic oleanane triterpenoids. On Day 4, the medium was replaced with fresh medium, again containing synthetic oleanane triterpenoids. On Day 7, calvaria were collected, either stored at -80°C for further RNA analysis or fixed in 10% buffered formalin for 24 hours and transferred to 80% ethanol for histologic analysis.

[00150] Histologic Analysis of Calvaria . After fixation for 24 hours, calvaria were decalcified in EDTA, embedded in paraffin and sectioned at 4 μτη. Sections were stained either with modified hematoxylin and eosin (H&E) or with toluidine blue (1% in 70% ethanol for 20 minutes, followed by destaining in 70%, 90% and 100% ethanol for 15 seconds) , placed in xylene twice, and then mounted. Procedures for immunofluorescence staining have been described (Medici, et al . (2010) Nat. Med. 74:537-45). Primary antibodies against collagen type II (AB746P, Millipore) , were used at 1:100 dilution; ALEXFLUOR secondary antibodies (Invitrogen) at 1:200 dilution. For nuclear staining, To-PRO-3 Iodide (T3605, Invitrogen) was used.

[ 00151] Bone Marrow-Derived Stem Cell Culture and Immunoblotting. Human bone marrow-derived stromal cells, which contain a population of responsive mesenchymal stem cells (ScienCell Research Laboratories) , were grown in mesenchymal stem cell medium (ScienCell Research Laboratories) . Cells were serum-starved 24 hours prior to all experimental conditions. Immunoblotting was performed using the following antibodies at concentrations (and using protocols) recommended by the respective manufacturers: SOX9 (sc-20095, Santa Cruz Biotechnology) , collagen Hal (sc-7764 and sc-28887, Santa Cruz), aggrecan (ab3778 and ab36861, Abeam), BMP-2 (abl4933, Abeam), phospho-Smad5 (9516, Cell Signaling Technology) , Smad5 (9517, Cell Signaling Technology), β-actin (A1978, SigmaeAldrich) . HRP- conjugated IgG TRUEBLOT reagents (18-8814, eBioscience) were used at a dilution of 1:1000.

[ 00152 ] Quantitative RT-PCR (qRT-PCR) . Detailed procedures for qRT-PCR are known in the art (Lee, et al . (2006) Biochem. Pharmacol. 72:332-43). In brief, 30 ng of RNA was reverse transcribed to cDNA using the random primers and Applied Biosystems' High Capacity cDNA Archive Kit in a 96- well format MASTERCYCLER Gradient from EPPENDORF . Subsequently, cDNA was amplified with ASSAYS-ON-DEMAND Products containing two gene specific primers and one TAQMAN MGB probe (6-FAM dye-labeled) using the TAQMAN Universal PCR Master Mix in an ABI PRISM 7000 Sequencing Detector (Applied Biosystems) . All labeled primers were obtained from Applied Biosystems. Example 2 : Synthesis of Triterpenoids

[00153] The triterpenoids of the invention can be generally produced from natural compounds such as oleanolic acid, ursolic acid, betulinic acid, or hederagenin, or derivatives thereof that include additional A and/or C ring modifications. Synthesis of the compounds can be achieved using any conventional method of synthesizing similar triterpenoids such as CDDO or CDDO-Me. See, e.g., US 6,326,507, US 6,552,075, US 6,974,801, US 7,288,568, 7,863,327, US 7,915,402, US 7,943,778, US 8,034,955, US 8,071,632, US 8,124,656, US 8,124,799, US 8,129,429 and WO 2009/146216.

[00154] As one example, triterpenoid compounds of the invention can be synthesized by (a) methylating the carboxylic acid group of a compound of Formula III to afford a methyl ester (Formula IV) ; oxidizing the hydroxyl group of a compound of Formula IV with an oxidizing agent to form a double bond in Ring A (Formula V) , epoxidating Ring C of the enone to form an epoxide, and forming a Coring enol and halogenating the A-ring enone to yield a compound of Formula VI (Scheme 1)

Formula III Formula IV

Oxidizing Agent

Formula VI Formula V

SCHEME 1

[ 00155 ] According to this example, the methylating agent is an electrophilic methyl source including, but not limited to iodomethane, dimethyl sulfate, dimethyl carbonate, diazomethane , or with methylating reagents such as methyl triflate or methyl fluorosulfonate , optionally in the presence of a base such as K 2 C0 3 or Li 2 C0 3 . Further, the oxidizing agent can be an iodine oxidizing agent such as o- iodoxybenzoic acid (IBX) (Nicolaou, et al . (2002) J. Am. Chem. Soc. 124:2245-2258), diacetoxyiodobenzene (DAIB) , fluorous DAIB (F-DAIB) , Dess-Martin-Periodinane (DMP) , or a stabilized formulation of IBX (SIBX; Ozanne, et al . (2003) Org. Lett. 5:2903) in one or a combination of suitable solvents such as DMSO and phenyl fluoride ( fluorobenzene) . Furthermore, epoxidation of Ring C can be carried out with an oxidant such as a peroxyacid, e.g., meta- chloroperoxybenzoic acid (.mCPBA) , peroxyacetic acid, or potassium peroxymonosulfate (Oxone) . Acid catalyzed opening of the epoxide and bromination of the A Ring can be achieved with a hydrogen halide such as (HBr or HI) and a diatomic halogen molecule such as Br 2 or I 2 .

[ 00156 ] As used in the context of the present invention, Formula III includes naturally occurring starting materials such as oleanolic acid, ursolic acid, betulinic acid, or hederagenin, or derivatives thereof.

Betulinic Acid Hederagenin

[00157] As a specific example of using this synthetic method in the synthesis of triterpenoids , CDDO-Me was synthesized from oleanolic acid. As shown in Scheme 2, the natural triterpenoid oleanolic acid (1) was used as the starting material in the synthesis of CDDO-Me. The method commences with methylation of the carboxylic acid of oleanolic acid (1) to afford methyl ester 2 in quantitative yield. With ester 2, activation of the A-ring is fulfilled by 2- iodoxybenzoic acid-mediated two- fold oxidation to give enone 3. Epoxidation with meta-chloroperoxybenzoic acid, followed by direct C-ring enolization and A-ring enone bromination with bromine and hydrobromic acid, affords key intermediate 4. With bromide 4 in hand, a cross-coupling reaction with copper cyanide provides CDDO-Me (5) (Scheme 2) . Intermediate 4 was prepared in high yield and few overall steps, thereby providing a base compound for development of the analogs and derivatives described herein .

SCHEME 2

[00158] Unless otherwise specified, the reagents used in the instant methods are conventionally known in the art . For example, Mel refers to methyl iodide, DMF refers to dimethylformamide , IBX is 2-iodoxybenzoic acid, DMSO is dimethyl sulfoxide, PhF is phenyl fluoride, mCPBA refers to ff!eta-chloroperoxybenzoic acid, HBr is hydrogen bromide, DCM is dichloromethane , AcOH is acetic acid, and CuCN is copper cyanide .

Example 3 : Ring A Modifications

[00159] Given the reactivity of Ring A halogen, Formula VI serves as a substrate for the synthesis of the triterpenoid derivatives of the invention. For example, contact of a compound of Formula VI with a cyanide ion source such as K 4 [Fe(CN) 6 ], KCN, NaCN, ZnCN, CuCN, (CH 3 ) 2 C ( -OH) CN or TMSCN results in the displacement of the aromatic halide with a cyanide ion. In other embodiments, the compound of Formula VI can be reacted with a wide variety of reagents to replace the halogen on Ring A. For example, the compound of Formula VI can be aminated or coupled or cross-coupled with an alkyl, alkenyl, alkynyl or aryl group to provide a variety of substituents on Ring A. For example, Formula VI can be aminated via Buchwald-Hartwig amination (Buchwald & Muci (2002) Top. Curr. Chem. 219:133-209; Hartwig (1999) Pure Appl. Chem. 71:1417; Buchwald & Yang (1999) J. Org et. Chem. 576:125; Hartwig (1998) ACIEE 37:2046; Hartwig (1998) Acc. Chem. Res. 31:852; Buchwald et al . (1998) Acc . Chem. Res. 31:805) to provide amides and amines 6 (R 16 = H and R 17 = H or CHO) . Moreover, when 6 is a formamide (R 16 = H and R 17 = CHO) , isonitrile 7 can be readily synthesized under mild conditions (Porcheddu, et al . (2005) J. Org. Chem. 70:2361-3) . In addition, Sonogashira coupling (Sonogashira, et al . (1975) Tetrahedron Lett. 16 : 4467 -70) provides alkynes 8 and 10 . Likewise, Suzuki (Miyuara, et al . (1979) Tetrahedron Lett. 20:3437-40; Miyaura & Suzuki (1979) Chem. Comm. 19:866-7; Miyaura & Suzuki (1995) Chem. Rev. 95:2457- 2483), Stille (Kosugi, et al . (1977) Chem. Lett. 301; Milstein & Stille (1978) J. Am. Chem. Soc . 100:3636), and Negishi (King, et al . (1977) J. Chem. Soc. Chem. Commun. 19:683) cross -coupling reactions provide compounds having the structure of compound 9 and dimers such as compounds 10 - 17 are readily produced when the halogen of Formula VI is iodide.

7

6

(R=alkyl, aryl , alkenyl, alkynyl)

11

(R=H, alkyl, aryl , alkenyl, alkynyl)

15

17

Example 4: Derivatives with Modifications at C-17

[00160] Amides (Formula VI) , ethers (Formula VII) , and esters (Formula VIII and Formula IX) are readily obtained using the instant method in combination with techniques known in the art. See US 6,974,801 and US 2008/0233195.

Formula VI Formula VII

Formula VIII Formula IX

[ 00161] In accordance with Formulae VI-IX,

R 11 and R 12 are as defined for Formula I;

R 1B is -OMs, -CH 2 OMs, -C(=0)C≡CR a , -C≡CC0 2 R a , -C≡CS0 2 R a , - C≡CC(=0)R a , -S0 2 R a , =0 or =CR c R d , wherein

R d is hydrogen, halo, alkylthiyl, or substituted or unsubstituted alkylsulfonyl or alkylsulfonyl -O- ;

R 22 and R 23 are independently a hydrogen, hydroxyl , halo, alkyl, nitro or amino group;

R 19 , R 20 , R 21 , R 24 and R 25 are independently a hydrogen, hydroxyl, -NR f R 9 , cyano, halo, azido, phosphate, 1,3- dioxoisoindolin-2 -yl , mercapto, silyl or -COOH group,

substituted or unsubstituted versions of C1-C15- alkyl, C 2 -C 15 -alkenyl , C 2 -Ci 5 -alkynyl , C 6 -C 15 -aryl, C 7 -Ci 5 - aralkyl , Ci-C 15 -heteroaryl , C 2 -C 15 -heteroaralkyl , Ci-Cis-acyl, Ci-Ci 5 -alkoxy, C 2 -C 15 -alkenyloxy, C 2 -Ci 5 -alkynyloxy, C 6 -Ci 5 - aryloxy, C 7 -Ci 5 -aralkyloxy, Ci-Ci 5 -heteroaryloxy, C 2 -C 15 - heteroaralkyloxy, Ci-Ci 5 -acyloxy, Cx-Cis-alkylamino , C 2 -Ci 5 - alkenylamino, C 2 -Ci 5 -alkynylamino, C 6 -Ci 5 -arylamino, C 7 -C 15 - aralkylamino, Ci-Ci 5 -heteroarylamino, C 2 -Ci 5 - heteroaralkylamino , Ci-Ci 5 -amido , Ci-C 15 -alkylthio, C 2 -Ci 5 - alkenylthio, C 2 -Ci 5 -alkynylthio, C 6 -Ci 5 -arylthio, C 7 -Ci 5 - aralkylthio, Ci-C 15 -heteroarylthio , C 2 -Ci 5 -heteroaralkylthio, Ci-C 15 -acylthio, Ci-Ci 2 -thioacyl , Ci-Ci 2 -alkylsulfonyl , C 2 -C 12 - alkenylsulfonyl , C -Ci 2 -alkynylsulfonyl , C 6 -Ci 2 -arylsulfonyl , C 7 -Ci 2 -aralkylsulfonyl , C ! -Ci 2 -heteroarylsulfonyl , Cx-C 12 - heteroaralkylsulfonyl , Ci-Ci 2 -alkylsulfinyl , C 2 -C 12 ~ alkenylsulfinyl , C 2 -C 12 -alkynylsulfinyl , C 6 -Ci 2 -aryl sulfinyl, C 7 -Ci 2 -aralkylsulfinyl , Ci-Ci 2 -heteroarylsulfinyl , Ci-Ci 2 -heteroaralkylsulfinyl , Ci~Ci 2 -alkylphosphonyl , Ci-Ci 2 - alkylphosphate, C 2 -Ci 2 -dialkylphosphate , Ci-Ci 2 - alkylammonium, Ci-Ci 2 -alkylsulfonium, Ci-Ci 5 -alkylsilyl , or a substituted version of any of these groups,

a -C0 2 Me, carbonyl imidazole, -CO-D-Glu (OAc) 4 , -CONH 2 , -CONHNH 2 , -CONHCH 2 CF 3 , or -C (=0) -heteroaryl group.

Example 5: CDDO-Me Derivatives with A-Ring Modifications

[00162] CDDO-Me derivatives within the scope of Formula I and containing A-Ring modifications are as follows.

Formula X Formula XI

Formula XII , Formula XIII

[00163] In accordance with Formulae X-XIII,

R 11 and R 12 are as defined for Formula I;

X is =0 or -OMe;

dashed bonds are present or absent; R 26 and R 27 are independently a hydrogen, halo {e.g., CI or F) , alkylthiyl, or substituted or unsubstxtuted alkylsulfonyl or alkylsulfonyl -O- ;

R 28 and R 29 are independently -H, or together are =0.

[ 00164 ] Exemplary compounds of Formulae X-XIII include compounds 18 - 82 :

27 28 29

Example 6: Mesylate Derivatives of CDDO-Me

[00165] Mesylate derivatives of CDDO-Me of Formula XIV-XVI are also included with the scope of this invention.

Formula XIV Formula XV

Formula XVI

[00166] In accordance with Formulae XIV-XVI:

R 11 and R 12 are as defined for Formula I;

R 30 , R31L , R32 and R33 are each independently a hydrogen, cyano, -OMs, or -CH 2 OMs group; or

R 30 and R 31 or R 32 and R 33 together are =C¾; and dashed bonds are either present or absent .

[00167] Exemplary compounds of Formulae XIV-XVI include compounds 62-75:

71 72 73

74 75

[00168] Mesylate derivatives having the structure of Formula XVII are also embodied by the present invention.

Formula XVII

wherein R 11 and R 12 are as defined for Formula I; and at least one of R y is OMs and the remaining R y are a hydrogen, cyano, -OMs, -C¾OMs, or =CH 2 group.

Example 7 : Gene Expression and Cellular Differentiation

[00169] For the results described here, more than 300 individual calvarial organ cultures were performed. The analysis presented herein indicated that both CDDO-Im and CDDO-EA have marked ability to induce chondrogenesis in newborn mouse calvaria. Because it is membranous bone, the newborn calvarium does not manifest the chondrogenic phenotype, except for a very thin margin at suture lines; care was taken to avoid using suture areas of calvaria in any of the analyses described below. Treatment with either triterpenoid (200 nM) for 7 days clearly had a profound chondrogenic effect on the calvaria. No new cartilage was seen on control sections stained with either H&E or toluidine blue. In contrast, the metachromatic toluidine blue purple staining was indicative of the ability of CDDO- Im and CDDO-EA to induce the formation of proteoglycans, such as aggrecan, which are characteristic of cartilage (Roughley (2001) Arthritis Res. 3:342-7). With toluidine blue, bone stained orthochromatically (blue) .

[ 00170 ] With respect to dose-response to either synthetic oleanane, 200 nM appeared to be optimal. Treatment with 50 nM triterpenoid yielded only marginal induction of chondrogenesis , while treatment with 500 nM synthetic oleanane gave somewhat variable results. Treatment with 1 mM triterpenoid was invariably toxic to the organ cultures . Results with 200 nM CDDO-Im and CDDO-EA were obtained in at least three sets of replicate experiments and immunohistochemistry showed that CDDO-Im and CDDO-EA (200 nM) both induced the formation of type II collagen (collagen Hal) , which was not seen in the control sections .

[ 00171 ] In addition to the histologic analysis of the calvarial cultures, mechanistic aspects of the action of both CDDO-EA and CDDO-Im were investigated in the calvaria. After 7 days of culture, RNA was isolated from the calvaria and quantitative RT-PCR analysis was performed for more than 15 different markers, including: SOX9, collagen Hal, all three isoforms of TGF-β, BMPs 2 and 4, BMP receptor II, Smads 3, 4, 6, and 7, tissue inhibitors of metalloproteinases (TIMP-1 and TIMP-2) , and matrix metalloproteinase-9 (MMP-9) . Tables 2 and 3 show that essentially all of these markers (except MMP-9) were significantly up-regulated by both triterpenoids, when the calvaria were treated at either the 200 or the 500 nM dose. The 50 nM dose was generally ineffective. In contrast, both triterpenoids were strong inhibitors of the expression of MMP-9; CDDO-EA (200 nM) caused almost 80% inhibition of the expression of this metalloproteinase , which is known to be involved in the degradation of cartilage (Shinoda, et al . (2008) J. Biol. Chem. 283:24632-9).

Table 2

CDDO-Im

Gene

50 nM 200 nM 500 nM

S0X9 1.26+0.08 1.33+0.05 1.83+0.13**

COL2A1 1.45+0.09 1.32+0.23 1.72+0.39

TGF-βΙ 0.96+0.07 1.08+0.12 1.46±0.12* GF- 2 1.17+0.05 1.15+0.09 1.17+0.10

TGF^3 1.30+0.07 1.52+0.16** 1.66±0.13**

BMP-2 1.17±0.07 1.60+0.19 3.14±0.53**

BMP-4 0.97+0.05 0.98+0.06 1.35±0.22

BMPRII 1.09±0.06 1.22+0.11 1.46+0.14*

Smad3 0.97±0.07 0.90±0.05 1.35+0.19

Smad4 0.96+0.06 1.03±0.09 1.26+0.09

Smad6 0.95+0.05 1.23+0.14 1.77+0.19**

Smad7 1.05±0.07 1.320.14 1.78+0.18**

TIMP-1 1.43+0.19 1.83±0.18 3.00+0.65**

TIMP-2 1.24+0.10 1.67+0.19 2.62+0.29**

MMP-9 0.73±0.16 0.44+0.12** 0.26+0.03**

Table 3

CDDO-ea

Gene

50 nM 200 nM 500 nM

SOX9 1.64±0.17 2.62+0.33** 3.12+0.38**

COL2A1 4.12±1.78* 5.45+1.13** 3.18±0.71

TGF-βΙ 1.17+0.09 1.47±0.24* 1.74±0.17**

TGF- 2 1.36±0.13* 1.61+0.11** 1.88+0.09**

TGF^3 1.39±0.07* 1.56±0.13** 1.44+0.11**

BMP-2 1.53+0.20 2.52±0.29** 5.66+0.67**

BMP-4 1.28±0.12 1.47+0.13 2.49±0.35**

BMPRII 1.50+0.14* 1.71+0.19** 2.24+0.22**

Smad3 1.14+0.12 1.35±0.07 1.93±0.16**

Smad4 1.06+0.03 1.37+0.12** 1.89+0.12**

Smad6 1.09±0.04 1.77+0.24** 2.85+0.31**

Smad7 1.28±0.10 1.86+0.19** 3.06+0.28** TIMP-1 1.88+0.34 2.85±0.38** 4.65+0.38**

TIMP-2 1.78+0.18* 2.44+0.31** 4.09+0.36**

MMP-9 0.49±0.09** 0.22+0.03** 0.19+0.06**

[ 00172 ] Further studies on mechanism were pursued in human bone marrow stem cell (BMSC) cultures. Western blot analysis showed that CDDO-Im and CDDO-EA (each 100 nM for 7 days) induced expression of the chondrocyte markers, SOX9, collagen type 2, and aggrecan, none of which were detectable in the control cultured stem cells. Furthermore, both triterpenoids (also at 100 nM) rapidly induced expression of both BMP-2 and its relevant signal transduction protein, phospho-Smad5 (P-Smad5) , in these stem cells. BMP-2 is known to induce chondrogenic lineage development of human mesenchymal stem cells in culture

(Schmitt, et al . (2003) Differentiation 71:567-77) .