Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL CURCUMIN DERIVATIVES AND THEIR PHARMACEUTICAL USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2009/017874
Kind Code:
A2
Abstract:
Disclosed are methods relating to analogues, derivatives, and metabolites of curcumin. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.

Inventors:
OKUNIEFF PAUL (US)
LIU CHAOMEI (US)
ZHANG LURONG (US)
Application Number:
PCT/US2008/064877
Publication Date:
February 05, 2009
Filing Date:
May 27, 2008
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV ROCHESTER (US)
OKUNIEFF PAUL (US)
LIU CHAOMEI (US)
ZHANG LURONG (US)
International Classes:
A61K31/415
Foreign References:
US20020006966A1
US20050187255A1
US20040265231A1
US20070111948A1
Other References:
SHIM ET AL.: 'Hydrazinocurcumin, a Novel Synthetic Curcumin Derivative, is a Potent Inhibitor of Endothelial Cell Proliferation.' BIOORGANIC & MEDICINAL CHEMISTRY vol. 10, 2002, pages 2439 - 2444
Attorney, Agent or Firm:
CLEVELAND, Janell, T. (P.C999 Peachtree Street,Suite 100, Atlanta GA, US)
Download PDF:
Claims:

CLAIMS

What is claimed is:

1. A method of mitigating radiation toxicity in a subject comprising the step of administering to the subject a composition comprising:

wherein Z is selected from CO, CH 2 CO, CH 2 CH 2 CO, CH(OH)CH 2 CO, CH(NH 2 )CH 2 CO, CH 2 CH(OH)CO, CH 2 CH(NH 2 )CO, SO, and SO 2 ; and wherein R is selected from substituted or unsubstituted aryl, amino acid residue, substituted or unsubstituted heterocycle, and fused bicyclic or heterobicyclic moiety, or a pharmaceutically acceptable salt thereof in a dosage and amount effective to mitigate radiation toxicity in the subject.

2. The method of claim 1, wherein the subject is a mammal.

3. The method of claim 1, wherein the subject is a human.

4. The method of claim 1, wherein the subject has been diagnosed with a need for mitigating radiation toxicity prior to the administering step.

5. The method of claim 1, wherein the administering step is performed before the subject acquires radiation toxicity.

6. A method of mitigating radiation toxicity in a subject comprising the step of administering to the subject a composition comprising:

wherein R 51 is selected from:

a. substituted or unsubstituted alkyl;

b. substituted or unsubstituted sulphydrylalkyl having one or two sulphydryl group(s);

c. substituted or unsubstituted alkaryl selected from benzyl and phenyl ethyl, having from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

_

wherein R 52 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl;

d. substituted or unsubstituted aryl having a structure:

wherein R 53 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; from one to two nitro,

with the proviso that R 53 is not 3-nitro; from one to two cyano; one to two carboxyl with the proviso that R 53 is not 4-carboxylic acid; and from one to two acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

e. substituted or unsubstituted heterocycle having a structure:

Het-R 55 ,

wherein R 55 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

f. fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 57 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and

wherein R 57 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; or a pharmaceutically acceptable salt thereof in a dosage and amount effective to mitigate radiation toxicity in the subject.

7. The method of claim 6, wherein the subject is a mammal.

8. The method of claim 6, wherein the subject is a human.

9. The method of claim 6, wherein the subject has been diagnosed with a need for mitigating radiation toxicity prior to the administering step.

10. The method of claim 6, wherein the administering step is performed before the subject acquires radiation toxicity.

11. A method of treating radiation-induced gastrointestinal syndrome in a subject comprising the step of administering to the subject a compound comprising:

wherein Z is selected from CO, CH 2 CO, CH 2 CH 2 CO, CH(OH)CH 2 CO, CH(NH 2 )CH 2 CO, CH 2 CH(OH)CO, CH 2 CH(NH 2 )CO, SO, and SO 2 ; and wherein R is selected from substituted or unsubstituted aryl, amino acid residue, substituted or unsubstituted heterocycle, and fused bicyclic or heterobicyclic moiety, or a pharmaceutically acceptable salt thereof in a dosage and amount effective to treat gastrointestinal syndrome in the subject.

12. The method of claim 11, wherein the subject has been diagnosed with a need for treatment of gastrointestinal syndrome.

13. The method of claim 11, wherein the administering step is performed after the subject is exposed to irradiation.

14. The method of claim 11, wherein the subject is a mammal.

15. The method of claim 11, wherein the subject is a human.

16. A method of treating radiation-induced gastrointestinal syndrome in a subject comprising the step of administering to the subject a compound comrprising:

wherein R 51 is selected from:

a. substituted or unsubstituted alkyl;

b. substituted or unsubstituted sulphydrylalkyl having one or two sulphydryl group(s);

c. substituted or unsubstituted alkaryl selected from benzyl and phenyl ethyl, having from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

wherein R 52 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary

amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl;

d. substituted or unsubstituted aryl having a structure:

wherein R 53 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; from one to two nitro, with the proviso that R 53 is not 3-nitro; from one to two cyano; one to two carboxyl with the proviso that R 53 is not 4-carboxylic acid; and from one to two acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

e. substituted or unsubstituted heterocycle having a structure:

Het-R 55 ,

wherein R 55 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

f. fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 57 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and

wherein R 57 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; or a pharmaceutically acceptable salt thereof in a dosage and amount effective to treat gastrointestinal syndrome in the subject.

17. The method of claim 16, wherein the subject has been diagnosed with a need for treatment of gastrointestinal syndrome.

18. The method of claim 16, wherein the administering step is performed after the subject is exposed to irradiation.

19. The method of claim 16, wherein the subject is a mammal.

20. The method of claim 16, wherein the subject is a human.

21. A method for the treatment of a disease of uncontrolled cellular proliferation comprising administering to a subject having a disease of uncontrolled cellular proliferation a compound comprising:

wherein Z is selected from CO, CH 2 CO, CH 2 CH 2 CO, CH(OH)CH 2 CO, CH(NH 2 )CH 2 CO, CH 2 CH(OH)CO, CH 2 CH(NH 2 )CO, SO, and SO 2 ; and wherein R is selected from substituted or unsubstituted aryl, amino acid residue, substituted or unsubstituted heterocycle, and fused bicyclic or heterobicyclic moiety, or a pharmaceutically acceptable salt thereof in a dosage and amount effective to treat the disease of uncontrolled cellular proliferation.

22. The method of claim 21, wherein the subject has been diagnosed with a need for treatment of a disease of uncontrolled cellular proliferation.

23. The method of claim 21, wherein the disease of uncontrolled proliferation is a carcinoma, lymphoma, leukemia, or sarcoma.

24. The method of claim 21, wherein the subject is a mammal.

25. The method of claim 21, wherein the subject is a human.

26. A method for the treatment of a disease of uncontrolled cellular proliferation comprising administering to a subject having a disease of uncontrolled cellular proliferation a compound comprising:

wherein R 51 is selected from:

a. substituted or unsubstituted alkyl;

b. substituted or unsubstituted sulphydrylalkyl having one or two sulphydryl group(s);

c. substituted or unsubstituted alkaryl selected from benzyl and phenyl ethyl, having from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

wherein R 52 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl;

d. substituted or unsubstituted aryl having a structure:

wherein R 53 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; from one to two nitro, with the proviso that R 53 is not 3-nitro; from one to two cyano; one to two carboxyl with the proviso that R 53 is not 4-carboxylic acid; and from one to two acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

e. substituted or unsubstituted heterocycle having a structure:

Het-R 55 ,

wherein R 55 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

f. fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 57 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and

wherein R 57 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; or a pharmaceutically acceptable salt thereof in a dosage and amount effective to treat the disease of uncontrolled cellular proliferation.

27. The method of claim 26, wherein the subject has been diagnosed with a need for treatment of a disease of uncontrolled cellular proliferation.

Ill

28. The method of claim 26, wherein the disease of uncontrolled proliferation is a carcinoma, lymphoma, leukemia, or sarcoma.

29. The method of claim 26, wherein the subject is a mammal.

30. The method of claim 26, wherein the subject is a human.

31. A method of reducing radiation damage in a subject comprising administering to a subject an effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

32. The method of claim 31 , wherein the radiation damage is caused by radiation therapy.

33. The method of claim 32, wherein the radiation therapy is used to treat cancer.

34. The method of claim 31, wherein the radiation damage is caused by nuclear radiation.

35. The method of claim 31, wherein the radiation is caused by a weapon.

36. The method of claim 31, wherein the agent is administered following radiation exposure.

37. The method of claim 31, wherein the agent is administered lhour, 2 hours, 3 hours, 4 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours, 36 hours, 48 hours, or 72 hours following exposure to ionizing radiation (IR).

38. The method of claim 31, wherein the agent is administered one, two, three, or four times every 24 hours.

39. The method of claim 38, wherein the agent is administered every 24 hours for 1, 2, 3, 4, 5, 6, 7, 14, or 21 days.

40. The method of claim 31, wherein the agent is administered intraarticularly, intravenously, transdermally, or orally.

41. The method of claim 31, wherein the agent comprises the compound:

wherein Z is selected from CO, CH 2 CO, CH 2 CH 2 CO, CH(OH)CH 2 CO, CH(NH 2 )CH 2 CO, CH 2 CH(OH)CO, CH 2 CH(NH 2 )CO, SO, and SO 2 ; and wherein R is selected from substituted or unsubstituted aryl, amino acid residue, substituted or unsubstituted heterocycle, and fused bicyclic or heterobicyclic moiety, or a pharmaceutically acceptable salt thereof.

42. The method of claim 31, wherein the agent comprises the compound:

wherein R > 51 is selected from:

a. substituted or unsubstituted alkyl;

b. substituted or unsubstituted sulphydrylalkyl having one or two sulphydryl group(s);

c. substituted or unsubstituted alkaryl selected from benzyl and phenyl ethyl, having from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

wherein R 52 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl;

d. substituted or unsubstituted aryl having a structure:

wherein R 53 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; from one to two nitro, with the proviso that R 53 is not 3-nitro; from one to two cyano; one to two carboxyl with the proviso that R 53 is not 4-carboxylic acid; and from one to two acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

e. substituted or unsubstituted heterocycle having a structure:

Het-R 55 ,

wherein R 55 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino;

f. fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 57 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and

wherein R 57 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; or a pharmaceutically acceptable salt thereof.

43. The method of claim 31, wherein the agent comprises a compound selected from the group consisting of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6-heptadiene-3,5-dione monoisonicotinoylhydrazone (also referred to herein as D 12); l,7-bis-(4-hydroxy-3- methoxyphenyl)-l,6-heptadiene-3,5-dione mononicotinoylhydrazone (also referred to herein as D13); l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6-heptadiene-3,5-dione benzoyl monohydrazone (also referred to herein as D68); and l,7-bis-(4-hydroxy-3- methoxyphenyl)-l,6-heptadiene-3,5-dione 2'-furoyl monohydrazone (also referred to herein as D 56)

44. The method of claim 31, wherein the subject is a mammal.

45. The method of claim 44, wherein the subject is a human.

46. The method of claim 44, wherein the subject is a mouse, rat, guinea pig, or nonhuman primate.

47. The method of claim 31, further comprising administration of a therapy that reduces the effects of ionizing radiation.

48. The method of claim 47, wherein the therapy is a second agent.

49. The method of claim 48, wherein the second agent is administered before exposure to IR.

50. The method of claim 48, wherein the second agent is an amifostine.

51. A method of mitigating the damaging effects of exposure to radiation comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

52. A method of treating gastrointestinal syndrome comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

53. A method of reducing the effects of gastrointestinal syndrome comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

54. A method of preventing gastrointestinal syndrome comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

55. A method of treating bone marrow syndrome comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

56. A method of reducing the effects of bone marrow syndrome comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

57. A method of preventing bone marrow syndrome comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

58. A method of administering a sub-total body irradiation (sub-TBI) dose to a subject comprising administering to the subject a lethal dose of ionizing radiation (IR) and simultaneously shielding an keeping out of the IR field an appendage of the subject.

59. The method of claim 58, wherein the subject is a mouse.

60. The method of claim 58, wherein the appendage is a leg.

61. The method of claim 58, wherein the appendage is an arm.

62. A model for testing the mitigating effects of an agent on gastrointestinal syndrome comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure.

63. The model of claim 62, wherein the radiation dose is between 8 and 12 Gy.

64. The method of claim 63, wherein the radiation dose is between 8 and 10.5 Gy.

65. The method of claim 64, wherein the radiation dose is between 8.9 and 9.1 Gy.

66. The model of claim 62, wherein the subject is a mammal.

67. The model of claim 66, wherein the subject is a mouse.

68. A method of screening for an agent that treats gastrointestinal syndrome comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure and administering the agent following IR exposure, wherein a reduction in the toxic effects of IR relative to a control indicate an agent that treats gastrointestinal syndrome.

69. A method of screening for an agent that inhibits gastrointestinal syndrome comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure and administering the agent following IR exposure, wherein a reduction in the toxic effects

of IR relative to a control indicate an agent that treats gastrointestinal syndrome.

70. A method of screening for an agent that mitigates radiation toxicity comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure and administering the agent following IR exposure, wherein a reduction in the toxic effects of IR relative to a control indicate an agent that treats radiation toxicity.

71. A method of screening for an agent that inhibits acute radiation syndrome comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during ER exposure and administering the agent following ER exposure, wherein a reduction in the toxic effects of ER relative to a control indicate an agent that treats acute radiation syndrome.

72. A method of screening for an agent that treats acute radiation syndrome comprising administering a lethal dose of ionizing radiation (ER) to a subject, wherein on appendage of the subject is shielded and kept out of the ER field during ER exposure and administering the agent following ER exposure, wherein a reduction in the toxic effects of ER relative to a control indicate an agent that treats acute radiation syndrome.

Description:

NOVEL CURCUMIN DERIVATIVES AND THEIR PHARMACEUTICAL USES THEREOF

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 60/940,396, filed May

25, 2007 and U.S. Provisional Application No. 60/940,621, filed May 29, 2007, which are hereby incorporated herein by reference in its entirety.

BACKGROUND

[0001] The gastrointestinal (GI) injury is the second major cause of death after ionizing radiation (IR) exposure. Although the Amifostine, the only drug for anti-IR, can effectively protect the GI injury, it has to be given before IR. Due to the unpredicted timing of accidental or intentional radiation exposure or nuclear terrorist attack, the urgent need is to develop mitigation agents that can effectively reduce the severity of GI injury after IR exposure.

SUMMARY

[0002] hi accordance with the purpose(s) of this invention, as embodied and broadly described herein, this invention, in one aspect, relates to a method of reducing radiation damage in a subject comprising administering to the subject an effective amount of an agent, wherein the agent is an analog, derivative, or metabolite of curcumin.

[0003] In a further aspect, the invention relates to a method of reducing radiation damage in a subject comprising administering to the subject an effective amount of an agent, wherein the agent is administered after radiation exposure.

[0004] In a further aspect, the invention relates to compositions capable or reducing radiation damage in a subject and methods of their synthesis.

[0005] In a further aspect, the invention relates to compounds modified from curcumin, designed to have antioxidant properties to be capable of acting as radioprotective to the gastrointestinal tract and other tissues.

[0006] In a further aspect, the invention relates to a compound comprising a structure:

wherein Z is selected from CO, CH 2 CO, CH 2 CH 2 CO, CH(OH)CH 2 CO, CH(NH 2 )CH 2 CO, CH 2 CH(OH)CO, CH 2 CH(NH 2 )CO, SO, and SO 2 ; and wherein R is selected from substituted or unsubstituted aryl, amino acid residue, substituted or unsubstituted heterocycle, and fused bicyclic or heterobicyclic moiety, or a pharmaceutically acceptable salt thereof.

[0007] hi a further aspect, the invention relates to a compound comprising a structure:

wherein R 51 is selected from: substituted or unsubstituted alkyl; substituted or unsubstituted sulphydrylalkyl having one or two sulphydryl group(s); substituted or unsubstituted alkaryl selected from benzyl and phenyl ethyl, having from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

wherein R 52 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl; substituted or unsubstituted aryl having a structure:

wherein R 53 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; from one to two nitro, with the proviso that R 53 is not 3-nitro; from one to two cyano; one to two carboxyl with the proviso that R 53 is not 4-carboxylic acid; and from one to two acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; substituted or unsubstituted heterocycle having a structure:

Het-R 55 ,

wherein R 55 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 57 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and wherein R 57 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted

alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; or a pharmaceutically acceptable salt thereof.

[0008] In a further aspect, the invention relates to a pharmaceutical composition comprising a therapeutically effective amount of at least one of the disclosed compounds or disclosed products and a pharmaceutically acceptable carrier.

[0009] hi a further aspect, the invention relates to a method of preparing a curcumin derivative, the method comprising the step of reacting curcumin with a hydrazine derivative selected from a substituted or unsubstituted arylsulfonylhydrazide, a substituted or unsubstituted alkylcarbohydrazide, a substituted or unsubstituted sulphydrylalkylcarbohydrazide, a substituted or unsubstituted alkarylcarbohydrazide, a substituted or unsubstituted arylcarbohydrazide, a substituted or unsubstituted heterocyclic carbohydrazide, a substituted or unsubstituted carbohydrazide, a substituted or unsubstituted 2-aminoacetohydrazide, and a substituted or unsubstituted fused bicyclic or heterobicyclic carbohydrazide.

[0010] In a further aspect, the invention relates to a product of the disclosed methods.

[0011] In a further aspect, the invention relates to a method of mitigating radiation toxicity in a subject comprising the step of administering to the subject at least one of the disclosed compounds at least one of the disclosed pharmaceutical compositions, or at least one of the disclosed products in a dosage and amount effective to mitigate radiation toxicity in the subject.

[0012] In a further aspect, the invention relates to a method of treating radiation-induced gastrointestinal syndrome in a subject comprising the step of administering to the subject at least one of the disclosed compounds at least one of the disclosed pharmaceutical

compositions, or at least one of the disclosed products in a dosage and amount effective to treat gastrointestinal syndrome in the subject.

[0013] In a further aspect, the invention relates to a method for the treatment of a disease of uncontrolled cellular proliferation comprising administering to a subject having a disease of uncontrolled cellular proliferation at least one of the disclosed compounds at least one of the disclosed pharmaceutical compositions, or at least one of the disclosed products in a dosage and amount effective to treat the disease of uncontrolled cellular proliferation.

[0014] Additional advantages of the invention will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practice of the invention. The advantages of the invention will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.

BRIEF DESCRIPTION OF THE DRAWINGS

[0001] The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate several embodiments and together with the description illustrate the disclosed compositions and methods.

[0002] Figure 1 shows a voltammeter analysis using neutral buffer with Ag/ AgCl as reference electrode and glassy carbon as working electrode. Figure IA shows vehicle alone. Figure IB shows compounds with low redox potential. Figure 1C shows Curcumin. Figure ID shows D13. Figure IE shows D68. Figure IF shows Vitamin C. Figure IG shows D12. Figure IH shows HPLC analysis of D68.

[0003] Figure 2 shows GI protection of novel compounds D12, D13, and D68 relative to a control. Figure 2 A shows GI protection of D12 in BALB/c mice. Figure 2B shows GI protection of D12 in BALB/c mice. Figure 2C shows GI protection of D68 in BALB/c mice.

[0004] Figure 3 shows GI protection of novel compound D68 relative to a control in a different mouse model. Figure 3A shows GI protection of D68 in C57BL/6 mcie. Figure 3B shows GI protection of D68 in C57BL/6 mice.

[0005] Figure 4 shows that the disclosed compounds can rescue a mouse following total body irradiation. Figure 4A shows TBI protection of D 12, 4B shows TBI protection of D13, 4C shows TBI protection of D68. Figure 4D shows TBI protection of D68 following 7.0 Gy TBI.

[0006] Figure 5 shows a side-by-side comparison of the D12, D13, and D68 in rescuing a mouse from sub- TBI.

[0007] Figure 6 shows that compared with the normal, the numbers of proliferating crypts in all the segments of mice exposed to 10.5 Gy treated with vehicle alone were decreased dramatically. Figure 6 A shows that proliferating crypts in jejunum as seen by H&E staining. Figure 6B shows increased crypts via BrdU staining. Figures 6C-E show measured villi length in the jejunum, duodenum, and ileum by H&E and BrdU staining in Balb/C and C57BL/6 mice following treatment with D68 and sub-TDI. Figure 6F shows the effects of different treatment on Proliferating crypts.

[0008] Figure 7 shows the stool hemoccult score differed among the groups. Figure 7A shows the hemoccult score. Figure 7B and C shows the stools of IR and D68 treated mice. Figure 7D and E show that Plasma endotoxin levels were reduced in D68 treated Balb/c (D) and C57BL/6 (E) mice. Figure 7F shows loss and gain of body weight in D68 treated and control mice following IR.

[0009] Figure 8 shows that D68 restores GI endocrine and exocrine ability in mice with GI syndrome of ARS. Figure 8 shows (A) plasma secretin levels, (B) Plasma CCK levels, (C) Plasma Insulin levels, and (D) Plasma Amylase levels 3.5 days following 10.5Gy sub-TBI in Balb/C mice.

[0010] Figure 9 shows the levels of various inflammatory molecules 3.5 days following exposure to 12Gy sub-TDI in Balb/C mice. Figure 9 shows that, several Ms that were increased by the 12 Gy IR could be reduced by D68, including MCP-I (A), IL6 (B), KC (C), ILl β( D), BLC (E), TNFα F), TCA-3 (G) and G-CSF (H). Figure 91 shows alterations in plasma CD30 levels.

[0011] Figure 10 shows the results of our preliminary analysis of D68 in plasma as a function of time.

[0012] Figure 11 shows a typical chromatogram obtained for a SPE-extracted plasma sample where D68 and curcumin are measured at 327 ran and 430 ran (lower panel), respectively.

[0013] Figure 12 shows further analysis of the HPLC chromatograms from the PK studies shows the presence of additional peaks that are believed to be metabolites of D68.

[0014] Figure 13 shows that when a solution of DPPH is mixed with that of a substance that can donate a hydrogen atom (such as curcumin and the D-analogs thereof), this gives rise to the reduced form of DPPH.

[0015] Figure 14 shows IC50 values which were used to compare DPPH scavenging activity. Compared are curcumin, D12, 13, D68, and Trolox.

[0016] Figure 15 shows the total antioxidant capacity of D47 and D68 compared to curcumin.

[0017] Figure 16 shows that intracellular ROS were detected by FCM in SW 480 cell line. A shows 5 Gy + Vehicle alone. B shows 5 Gy + D68 micro-emulsion. C shows the percentage of gate after 5 Gy IR and treatment. D shows X mean alternation. 5 Gy + E alone = 5 Gy IR + Vehicle. 5 Gy + D 68-E = 5 Gy IR+ D68 microemulsion lOug/mL. E shows alternation of percentage gate after treatment. F shows a decrease of X mean after treatment with D68.

[0018] Figure 17 shows the kinetic plot for CytoC-Fe +3 → Cyto-Fe +2 .

[0019] Figure 18 shows a plot of competition kinetics - CytoC versus curcumin. K cUr = 1.19 (±0.12) x 10 5 M "1 s "1 .

DETAILED DESCRIPTION

[0020] Before the present compounds, compositions, articles, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods or specific recombinant biotechnology methods unless otherwise

specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.

A. Definitions

[0021] As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a pharmaceutical carrier" includes mixtures of two or more such carriers, and the like.

[0022] Ranges can be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as "about" that particular value in addition to the value itself. For example, if the value "10" is disclosed, then "about 10" is also disclosed. It is also understood that when a value is disclosed that "less than or equal to" the value, "greater than or equal to the value" and possible ranges between values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value "10" is disclosed the "less than or equal to 10"as well as "greater than or equal to 10" is also disclosed. It is also understood that the throughout the application, data is provided in a number of different formats, and that this data, represents endpoints and starting points, and ranges for any combination of the data points. For example, if a particular data point "10" and a particular data point 15 are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.

[0023] In this specification and in the claims which follow, reference will be made to a number of terms which shall be defined to have the following meanings:

[0024] "Optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.

[0025] A "decrease" can refer to any change that results in a smaller amount of a symptom, condition, or disease such as radiation toxicity. Thus, a "decrease" can refer to a reduction in an activity as well as a reduction in the effects of a disease or condition. Also for example, a decrease can be a change in the symptoms of a disorder such that the symptoms are less than previously observed. Thus, for example, a decrease in the toxic effects of ionizing radiation can include but is not limited to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% decrease in the symptoms associated with exposure to ionizing radiation.

[0026] An "increase" can refer to any change that results in a larger amount of a symptom, condition, or disease such as radiation toxicity. Thus, for example, an increase in the amount in toxic effects of ionizing radiation can include but is not limited to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% increase in the symptoms associated with exposure to ionizing radiation.

[0027] Through extensive searching for GI protectants, agents were identified that mitigated the acute GI syndrome of acute radiation syndrome (ARS) and reduce the death rate of ARS within 7 days. The approach was to chemically synthesize agents that derived from natural compounds. The testing system was a sub-total body irradiation (sub-TBI) model in which one leg of animal was shielded from IR allowing the animal to tolerate a high IR dose that causes GI death within 5-7 days without bone marrow death. After screen about a hundred of synthetic compounds, agents were identified that had a high potential of anti-oxidant (measured by Voltammetric analysis) and were capable of mitigating the acute GI syndrome of ARS. The effectiveness of the radioprotector was demonstrated by several facts: 1) an increased a long-term survival of mice that received a GI death IR dose at LD 100 / 7 ; 2) an increased DMF for GI-IR death (about 1.16 with a potential to be higher after

formulation optimization); 3) a higher than 50% prolonged the survival time for the mice received a high TBI dose that cause BM death (LD 1OO z 30 ); 4) an increased proliferation of crypts; 5) an increased length and number of villi of small intestine; and 6) a reduced abnormalities of GI syndrome induced by IR, such as hemoccults, loose stool, decreased amylase and secretin and increased endotoximia. Beneficial effects were observed in other two strains of mouse, indicating that the new agent is effective on different genome backgrounds.

[0028] Herein disclosed are novel GI radioprotectors including for example D 12, D13, and D68. The disclosed compounds are: 1) effective when given after exposure to a GI death IR dose; 2) can be synthesized in a large quantity; 3) are small compounds that have non-antigenicity and can be used in a long-run without fear of host's developing resistance or immune reaction that is associated with most of protein regime; 4) their maximum tolerate dose (MTD) can be 15 higher than effective dose (ED 50 ), indicating that their is a big safety window (most radioprotector has a ratio of 5 to 10; 5) are stable at various hustle conditions, such as boil, and therefore they are portable by soldiers; 6) orally effective and can be self-administrated; 7) cheap to synthesize and therefore cost-effective compared with other biological regime, such as protein growth factors (G-CSF, KGF, FGF) which require a whole set of expensive processes for the production and purification.

B. Compounds and compositions

[0029] Before the present compounds, compositions, articles, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, example methods and materials are now described.

[0030] Disclosed are the components to be used to prepare the disclosed compositions as well as the compositions themselves to be used within the methods disclosed

herein. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular analogue, derivative or metabolite of curcumin is disclosed and discussed and a number of modifications that can be made, specifically contemplated is each and every combination and permutation of curcumin and the modifications that are possible unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited each is individually and collectively contemplated meaning combinations, A-E, A-F, B-D, B-E, B-F, C-D, C- E, and C-F are considered disclosed. Likewise, any subset or combination of these is also disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E would be considered disclosed. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the disclosed methods.

[0031] Radiation of tissues sets in motion a set of biological pathways that can result in progressive toxicity. To date, most or all radioprotective drugs approved by the FDA have utility only if given before and during the radiation exposure. These radioprotective agents are commonly antioxidants. More recently, however, several laboratories have identified mechanisms of radiation toxicity that can be altered well after the radiation exposure, including ongoing forms of DNA damage, DNA repair, and signal transduction related to long-term redox abnormalities.

[0032] The potential use of curcumin and its analogues or flavonoid compounds as radioprotectors is of increasing interest because of their high antioxidant activity, low toxicity and abundance in nature, and their anti-inflammatory, anti-NFKB, and cyclo- oxygenase suppression.

[0033] Curcumin [diferuloyl methane; l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione; CR] is the major constituent in the rhizome of Curcuma longa (Zingiberaceae), commonly named turmeric. The structure of curcumin can be divided into 3 parts A, B and C. Part A and C are same: />αrα-phenol with a ortho- methoxyl group. B is a α, β-unsaturated 1,3-diketone.

Curcumin

[0034] Curcumin has been reported to possess various biological and pharmacological activities, including antioxidative (Dutta Sabari et al. Bioorganic & Medicinal Chemistry Letters 15 (2005) 2738-2744; Deng S.L. et al. Food Chemistry 98 (2006) 112-119; Waylon M. Weber, Lucy A. Hunsaker, Steve F. Abcouwer, Lorraine M. Deck, and David L. Vander Jagt, Bioorg. Med. Chem. 13 (2005) 3811-3820; Venkateswarlu S. et al. Bioorganic & Medicinal Chemistry 13 (2005) 6374-6380; Daniel S. et al. J. of Inorganic Biochemistry 98 (2004) 266-275; Priyadarsini K. I. Et al. Free Radical Biology and Medicine, 35 (2003) 475-484), anti-inflammatory (Selvam C. et al. Bioorganic & Medicinal Chemistry Letters 15 (2005) 1793-1797; Chainani-Wu, N. J. of Alternative and Compementary Medicine 9 (2003) 161-168), anti-HIV-1 integrase (Di Santo R. et al. IL Farmaco 60 (2005) 409-417; Mazumder A. et al. Biochemical Pharmacology, 49 (1995) 1165-1170), anti-angiogenic and anticancer (Lin L. et al. Bioorganic & Medicinal Chemistry 14 (2006) 2527-2534; Robinson T.P. et al. Bioorganic & Medicinal Chemistry 13 (2005) 4007-4013; Woo H.B. et al. Bioorganic & Medicinal Chemistry Letters 15 (2005) 3782-3786; Adams B.K. et al. Bioorganic & Medicinal Chemistry 12 (2004) 3871-3883). These putative cancer preventive and therapeutic properties of curcumin have been considered to be associated with its antioxidant and anti-NFKB properties (Deng S.L. et al. Food Chemistry 98 (2006) 112-119; Sabari et al. Bioorganic & Medicinal Chemistry Letters 15 (2005) 2738-2744; Kelly M. R. et al. Mutation Research 485 (2001) 309-318) since the oxidative damage of DNA, lipid layer and cell membrane are believed to be

associated with a variety of chronic health problems, such as cancer, inflammatory, neurodegenerative diseases and aging. It is also believed that the antioxidant activity of curcuniin is responsible for its free radical scavenging ability.

[0035] Structurally, both of curcumin's hydroxyl groups attached to the aromatic rings (A and C parts) and the methylene CH 2 group of the β-l,3-diketone moiety (B part) are responsible for the formation of free radicals and protection of DNA, RNA, lipid and protein molecules. (Jovanovic S. V. et al. J Am. Chem. Soc. 2001, 123, 3064- 3068; Priyadarsini K.I. et al. Free Rad. Biol. Med. 2003,35,475).

Curcumin

-H * R

(Two possible sites of attack)

[0036] The β-l,3-diketone moiety alone does not have antioxidant properties. Apparently, the presence of both β-l,3-diketone and phenol functionalities is necessary for optimal antioxidant function of curcumin. The two potential sites of attack in the curcumin molecule, as shown in the above figure, are interchangeable. This is because the higher resonance stabilization afforded in the phenoxyl radical, the initially generated curcumin alkoxyl radical will undergo rapid intramolecular H-shift (Jovanovic S. V. et al. J Am. Chem. Soc. 2001, 123, 3064-3068). Also the pair of 4-

hydroxyl groups conjugated through a long olefmic double bond chain in the curcumin molecule is easily ionized to form radicals after absorption of energy from radiation. This is also facilitated by the formation of a hydrogen-bond between the A- hydroxyl group and its neighboring 3-methoxy group in benzene rings.

[0037] The antioxidant activity and other biological properties of curcumin can be decreased if its hydroxyl groups in the aryl rings are methylated or if the olefmic bonds in the β-l,3-diketone moiety are hydrogenated to give the tetrahydro derivative of curcumin. (D. L. Flynn, T. R. Belliotti, and A. M. Boctor et al. (J. Med. Chem. 1991, 34, 518-525); Deng S.L. Food Chemistry 98 (2006) 112-119).

[0038] It is also understood that the para hydroxy groups in curcumin are important for antiinflammatory activity. This activity can be enhanced when, in combination with the para hydroxy groups, the meta positions are occupied with alkyl groups (AN Nurfina, MS Reksohadiprodjo, and H Timmerman et al. Eur J Med Chem (1997)32, 321-328). The presence of olefmic double bonds and 4-hydroxyl groups can be important for the antiinflammatory activity of curcumin. In addition to the olefmic double bonds and the 4-hydroxy groups, the presence of one 3-OCH 3 group can also be important for the anti-inflammatory activity, especially when all the meta positions were substituted by methoxy groups (SS Sardjiman, MS Reksohadiprodjo, and L Hakim et al. Eur J Med Chem (1997)32, 625-630).

[0039] In 2005, Dutta S. et al. synthesized a curcumin derivative semicarbazone (CRSC) and found that this compound has a better antioxidant activity than that of curcumin. On the whole, the phenoxyl radical is the predominant species in the case of curcumin, while in the case of CRSC it is the imine carbonyl radical, which is the major species being formed in this reaction. This specie is also more reactive, according to the reactions of three radicals, N 3 *, methyl, and halocarbon peroxyl radicals with both curcumin and CRSC. Modification of the β-l,3-diketone moiety in curcumin can up-regulate its antioxidant property and free radical scavenging ability. Further, the curcumin semicarbazone theoretically has one more chemical mechanism as a radical scavenger than the curcumin itself. (Sabari Dutta et al. Bioorganic and medicinal chemistry letter 15(2005) 2738-2744):

[0040] As used herein, the term "residue" of a chemical species refers to the moiety that is the resulting product of the chemical species in a particular reaction scheme or subsequent formulation or chemical product, regardless of whether the moiety is actually obtained from the chemical species. Thus, an ethylene glycol residue in a polyester refers to one or more -OCH 2 CH 2 O- units in the polyester, regardless of whether ethylene glycol was used to prepare the polyester. Similarly, a sebacic acid residue in a polyester refers to one or more -CO(CH 2 ) 8 CO- moieties in the polyester, regardless of whether the residue is obtained by reacting sebacic acid or an ester thereof to obtain the polyester.

[0041] As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described below. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Unless explicitly disclosed, this disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds. Also, the terms "substitution" or "substituted with" include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.

[0042] In defining various terms, "A 1 ," "A 2 ," "A 3 ," and "A 4 " are used herein as generic symbols to represent various specific substituents. These symbols can be any substituent, not limited to those disclosed herein, and when they are defined to be certain substituents in one instance, they can, in another instance, be defined as some other substituents.

[0043] The term "alkyl" as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, for example 1 to 12 carbon atoms or 1 to 6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, «-butyl, isobutyl, s-butyl, t- butyl, «-pentyl, isopentyl, s-pentyl, neopentyl, hexyl, heptyl, octyl, nonyl, decyl, dode cyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like. The alkyl group can also be substituted or unsubstituted. The alkyl group can be substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein. A "lower alkyl" group is an alkyl group containing from one to six carbon atoms.

[0044] Throughout the specification "alkyl" is generally used to refer to both unsubstituted alkyl groups and substituted alkyl groups; however, substituted alkyl groups are also specifically referred to herein by identifying the specific substituent(s) on the alkyl group. For example, the term "halogenated alkyl" specifically refers to an alkyl group that is substituted with one or more halide, e.g., fluorine, chlorine, bromine, or iodine. The term "alkoxyalkyl" specifically refers to an alkyl group that is substituted with one or more alkoxy groups, as described below. The term "alkylamino" specifically refers to an alkyl group that is substituted with one or more amino groups, as described below, and the like. When "alkyl" is used in one instance and a specific term such as "alkylalcohol" is used in another, it is not meant to imply that the term "alkyl" does not also refer to specific terms such as "alkylalcohol" and the like.

[0045] This practice is also used for other groups described herein. That is, while a term such as "cycloalkyl" refers to both unsubstituted and substituted cycloalkyl moieties, the substituted moieties can, in addition, be specifically identified herein; for example, a particular substituted cycloalkyl can be referred to as, e.g., an "alkylcycloalkyl." Similarly, a substituted alkoxy can be specifically referred to as, e.g., a "halogenated alkoxy," a particular substituted alkenyl can be, e.g., an "alkenylalcohol," and the like. Again, the practice of using a general term, such as "cycloalkyl," and a specific term, such as "alkylcycloalkyl," is not meant to imply that the general term does not also include the specific term.

[0046] The term "cycloalkyl" as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, and the like. The term "heterocycloalkyl" is a type of cycloalkyl group as defined above, and is included within the meaning of the term "cycloalkyl," where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted. The cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl,

cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein.

[0047] The term "polyalkylene group" as used herein is a group having two or more CH 2 groups linked to one another. The polyalkylene group can be represented by the formula — (CH 2 ) a — , where "a" is an integer of from 2 to 500.

[0048] The terms "alkoxy" and "alkoxyl" as used herein refer to an alkyl or cycloalkyl group bonded through an ether linkage; that is, an "alkoxy" group can be defined as — OA 1 where A 1 is alkyl or cycloalkyl as defined above. "Alkoxy" also includes polymers of alkoxy groups as just described; that is, an alkoxy can be a polyether such as — OA 1 - OA 2 or — OA 1 - (OA 2 ) a — OA 3 , where "a" is an integer of from 1 to 200 and A 1 , A 2 , and A 3 are alkyl and/or cycloalkyl groups.

[0049] The term "alkenyl" as used herein is a hydrocarbon group of from 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon double bond. Asymmetric structures such as (A 1 A 2 )C=C(A 3 A 4 ) are intended to include both the E and Z isomers. This may be presumed in structural formulae herein wherein an asymmetric alkene is present, or it may be explicitly indicated by the bond symbol C=C. The alkenyl group can be substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein.

[0050] The term "cycloalkenyl" as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms and containing at least one carbon-carbon double bound, i.e., C=C. Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, norbornenyl, and the like. The term "heterocycloalkenyl" is a type of cycloalkenyl group as defined above, and is included within the meaning of the term "cycloalkenyl," where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkenyl group and heterocycloalkenyl group can be

substituted or unsubstituted. The cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein.

[0051] The term "alkynyl" as used herein is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond. The alkynyl group can be unsubstituted or substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol, as described herein.

[0052] The term "cycloalkynyl" as used herein is a non-aromatic carbon-based ring composed of at least seven carbon atoms and containing at least one carbon-carbon triple bound. Examples of cycloalkynyl groups include, but are not limited to, cycloheptynyl, cyclooctynyl, cyclononynyl, and the like. The term "heterocycloalkynyl" is a type of cycloalkenyl group as defined above, and is included within the meaning of the term "cycloalkynyl," where at least one of the carbon atoms of the ring is replaced with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkynyl group and heterocycloalkynyl group can be substituted or unsubstituted. The cycloalkynyl group and heterocycloalkynyl group can be substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein.

[0053] The term "aryl" as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, phenoxybenzene, and the like. The term "aryl" also includes "heteroaryl," which is defined as a group that contains an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus. Likewise, the term

"non-heteroaryl," which is also included in the term "aryl," defines a group that contains an aromatic group that does not contain a heteroatom. The aryl group can be substituted or unsubstituted. The aryl group can be substituted with one or more groups including, but not limited to, substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, azide, nitro, silyl, sulfo-oxo, or thiol as described herein. The term "biaryl" is a specific type of aryl group and is included in the definition of "aryl." Biaryl refers to two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl.

[0054] The term "aldehyde" as used herein is represented by the formula — C(O)H. Throughout this specification "C(O)" is a short hand notation for a carbonyl group, i.e., C=O.

[0055] The terms "amine" or "amino" as used herein are represented by the formula NA 1 A 2 A 3 , where A 1 , A 2 , and A 3 can be, independently, hydrogen or substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

[0056] The term "carboxylic acid" as used herein is represented by the formula — C(O)OH.

[0057] The term "ester" as used herein is represented by the formula — OC(O)A 1 or — C(O)OA 1 , where A 1 can be a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term "polyester" as used herein is represented by the formula — (A 1 O(O)C-A 2 - C(O)O) 2 - or — (A 1 O(O)C-A 2 -OC(O)) a — , where A 1 and A 2 can be, independently, a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein and "a" is an interger from 1 to 500. "Polyester" is the term used to describe a group that is produced by the reaction between a compound having at least two carboxylic acid groups with a compound having at least two hydroxyl groups.

[0058] The term "ether" as used herein is represented by the formula A 1 OA 2 , where A 1 and A 2 can be, independently, a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein. The term "polyether" as used herein is represented by the formula — (A 1 O- A 2 O) 3 — , where A 1 and A 2 can be, independently, a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group described herein and "a" is an integer of from 1 to 500. Examples of polyether groups include polyethylene oxide, polypropylene oxide, and polybutylene oxide.

[0059] The terms "halide" and "halo" as used herein refer to the halogens fluorine, chlorine, bromine, and iodine. It is also contemplated that, in certain aspects, pseudohalides (e.g., mesyl groups, brosyl groups, and tosyl groups) can be substituted for halides.

[0060] The terms "hydroxyl" and "hydroxyl" as used herein is represented by the formula — OH.

[0061] The term "ketone" and "keto" as used herein is represented by the formula A 1 C(O)A 2 , where A 1 and A 2 can be, independently, a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

[0062] The term "azide" as used herein is represented by the formula — N 3 .

[0063] The term "nitro" as used herein is represented by the formula — NO 2 .

[0064] The terms "nitrile" and "cyano" as used herein are represented by the formula

-CN.

[0065] The term "silyl" as used herein is represented by the formula — SiA 1 A 2 A 3 , where A 1 , A 2 , and A 3 can be, independently, hydrogen or a substituted or unsubstituted alkyl, cycloalkyl, alkoxy, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

[0066] The term "sulfo-oxo" as used herein is represented by the formulas — S(O)A 1 , -S(O) 2 A 1 , -OS(O) 2 A 1 , or -OS(O) 2 OA 1 , where A 1 can be hydrogen or a

substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. Throughout this specification "S(O)" is a short hand notation for S=O. The term "sulfonyl" is used herein to refer to the sulfo-oxo group represented by the formula — S(O) 2 A 1 , where A 1 can be hydrogen or a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term "sulfone" as used herein is represented by the formula A 1 S(O) 2 A 2 , where A 1 and A 2 can be, independently, a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein. The term "sulfoxide" as used herein is represented by the formula A 1 S(O)A 2 , where A 1 and A 2 can be, independently, a substituted or unsubstituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, cycloalkynyl, aryl, or heteroaryl group as described herein.

[0067] The term "thiol" as used herein is represented by the formula — SH.

[0068] Unless stated to the contrary, a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer and diastereomer, and a mixture of isomers, such as a racemic or scalemic mixture.

[0069] The term "arylsulfonylhydrazide" as used herein refers to a sulfonylhydrazide- functionalized aromatic compound. Such structure can be represented by the formula:

[0070] The term "carbohydrazide" as used herein refers to a moiety represented by the formula:

[0071] The term "alkylcarbohydrazide" as used herein refers to a carbohydrazide- functionalized alkyl compound. Such structure can be represented by the formula:

[0072] The term "sulphydrylalkylcarbohydrazi.de" as used herein refers to a sulphhydryl- (viz., sulfhydryl- or thiol-) functionalized alkylcarbohydrazide compound. Such structure can be represented by the general formula:

Examples of a sulphydrylalkylcarbohydrazide include 2-mercaptopropanehydrazide and 3- mercaptopropanehydrazide.

[0073] The term "alkarylcarbohydrazide" as used herein refers to an aryl functionalized alkylcarbohydrazide compound. Such structure can be represented by the formula:

An example of an alkarylcarbohydrazide is 2-phenylacetohydrazide.

[0074] The term "arylcarbohydrazide" as used herein refers to a carbohydrazide- functionalized aromatic compound. Such structure can be represented by the formula:

[0075] The term "heterocyclic carbohydrazide" as used herein refers to a carbohydrazide- functionalized heterocyclic compound. Such structure can be represented by the formula:

Examples of a heterocyclic carbohydrazide include furan-2-carbohydrazide and 1- methylpiperidine-3-carbohydrazide.

[0076] The term "2-aminoacetohydrazide" as used herein refers to a compound represented by the general formula:

An example of a 2-aminoacetohydrazide is l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione L-tyrosine monohydrazone. It is also contemplated that the 2- aminoacetohydrazide can be further functionalized at the terminal amine with one or more additional amino acid residues, for example, as 2-amino-N-(2-(l-hydrazinyl-l-oxopropan-2- ylamino)-2-oxo- 1 -phenylethyl)-3-methylbutanamide:

[0077] It is further contemplated that an aminoacetohydrazide can be provided as oligopeptide- or polypeptide-functionalized carbohydrazide compounds. The compounds can be provided in protected or unprotected forms, as appropriate.

[0078] The term "fused bicyclic or heterobicyclic carbohydrazide" as used herein refers to a carbohydrazide-functionalized fused bicyclic or heterobicyclic compound. Examples include 2-naphthohydrazide and bicyclo[2.1.1]hexane-2-carbohydrazide:

[0079] Without wishing to be bound by theory, it is believed that free radical formed can be stabilized if the free amino group in curcumin's semicarbazone molecule is replaced by benzene or other aromatic heterocycles. Accordingly, a series of

substituted curcumin carbonyl hydrazones were designed and synthesized and their radiation protective effect and anticancer activities were screened.

X = SO 2 or CO; R = substituted benzene ring or aromatic heterocycles.

[0080] It was determined that the bond between the two nitrogens (N — N) was easily broken when the foreign energy deposition took place, because the biggest and base peak in APCI-MS for nearly all compounds in this series is about 365. So, without wishing to be bound by theory, it is believed that a likely chemical mechanism accounting for the radiation protection for this series of compounds is as follows:

( C 21 H 20 NO 5 = 366 )

[0081] On the other hand, some investigators have reported that some pyrazoles of curcumin have better anti-angiogenic and anti-cancer activity (WO 2006044379 A2; US20040176384 Al). Shim J. S. et al had revealed that 3 (see below) was eight-fold less potent than curcumin according to IC 50 S to the human polymorphonuclear cell (PMN) 5-LO. Conversion of curcumin to the pyrazole analogue 2 hydrazinocurcumin (HC) resulted in a more potent 5-LO inhibitor, while the reduced analogue 4 was 53- fold less active than 2. HC potently inhibited the proliferation of bovine aortic endothelial cells (BAECs) at a nanomolar concentration (IC 50 =520 nM) without cytotoxicity. In vivo and in vitro angiogenesis experiments showed HC was a new candidate anti-angiogenic agent (Joong Sup Shim, Dong Hoon Kim, and Hye Jin Jung et al. Bioorganic and Medicinal Chemistry 10(2002) 2439-2444).

1, to 5-LO, IC 50 8.0 um 2, IC 50 1.0 urn

3, IC 50 61 um 4, IC 50 53 um

[0082] hi one aspect, the invention relates to a compound comprising a structure:

wherein Z is selected from CO, CH 2 CO, CH 2 CH 2 CO, CH(OH)CH 2 CO, CH(NH 2 )CH 2 CO, CH 2 CH(OH)CO, CH 2 CH(NH 2 )CO, SO, and SO 2 ; and wherein R is selected from substituted or unsubstituted aryl, amino acid residue, substituted or unsubstituted heterocycle, and fused bicyclic or heterobicyclic moiety, or a pharmaceutically acceptable salt thereof. In a further

aspect, Z is CO, SO, or SO 2 ; and wherein R is substituted or unsubstituted aryl having a structure:

wherein R 11 is from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

wherein R . 12 i •s selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl.

[0083] For example, in one aspect, Z can be CO, and R can be 3-pyridinyl, 4- pyridinyl, phenyl, or furanoyl. In a further aspect, Z and R together comprise a nicotinoyl, isonicotinoyl, benzoyl, or furanoyl moiety. Accordingly, the disclosed compounds include those structures represented by a formula:

[0084] In a further aspect, the compound can be l,7-bis-(4-hydroxy-3- methoxyphenyl)-l,6-heptadiene-3,5-dione monoisonicotinoylhydrazone (also referred to herein as D 12); l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6-heptadiene-3,5-dione mononicotinoylhydrazone (also referred to herein as D 13); l,7-bis-(4-hydroxy-3- methoxyphenyl)-l,6-heptadiene-3,5-dione benzoyl monohydrazone (also referred to herein as D68); and l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6-heptadiene-3,5-dione 2'-furoyl monohydrazone (also referred to herein as D56).

[0085] hi a further aspect, Z is CO; and wherein Z and R together comprise an amino acid residue. For example, the amino acid residue can be selected from one or more of arginine, asparagine, cysteine, histidine, hydroxylysine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophane, tyrosine, and valine residues. In a further aspect, the amino acid residue is an oligopeptide comprising from one to about ten, from one to about fifteen, from one to about twenty, or from one to about twenty five amino acid residues, hi a yet further aspect, the amino acid residue is a polypeptide.

[0086] In a further aspect, Z is CO, SO, or SO 2 ; and wherein R is substituted or unsubstituted heterocycle having a structure:

Het-R 31 ,

wherein R 31 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 32 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl. In a further aspect, Het is selected from furan, thiophene, imidazole, thiazole, oxazole, thiadiazole, oxadiazole, triazole, pyridine, and pyrimidine.

[0087] In a further aspect, R is a fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 41 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and wherein R 31 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 42 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl. In a further aspect, Fus is selected from aromatic, partially hydrogenated, or fully hydrogenated naphthalene, quinoline, isoquinoline, indole, benzothiophene, benzoimidazole, benzothiazole, benzoxazole, benzofuran, benzopyran, and 4H-[l]benzopyran[4,3-b]thiophene.

[0088] In a further aspect, the invention relates to a compound comprising a structure:

wherein R 51 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted sulphydrylalkyl having one or two sulphydryl group(s); substituted or unsubstituted alkaryl

selected from benzyl and phenyl ethyl, having from one to three substituent(s) independently selected from one to three substituted or unsubstituted alkyl; from one to three substituted or unsubstituted alkoxyl; from one to three substituted or unsubstituted alkylthio; from one to three halogen; from one to three primary amino, secondary amino, tertiary amino; from one to two nitro; from one to two cyano; and from one to two acyl having a structure:

wherein R 52 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; primary amino, secondary amino, tertiary amino; substituted or unsubstituted aryl; and substituted or unsubstituted heteroaryl; substituted or unsubstituted aryl having a structure:

wherein R 53 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; from one to two nitro, with the proviso that R 53 is not 3-nitro; from one to two cyano; one to two carboxyl with the proviso that R 53 is not 4-carboxylic acid; and from one to two acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; substituted or unsubstituted heterocycle having a structure:

Het-R 55 ,

wherein R 55 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted

alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; from one to two cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; fused bicyclic or heterobicyclic moiety having a structure:

Fus-R 57 ,

wherein Fus is selected from aromatic, partially hydrogenated, and fully hydrogenated fused bicyclic or heterobicyclic moieties; and wherein R 57 is from one to three substituent(s) independently selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; substituted or unsubstituted alkylthio; halogen; primary amino, secondary amino, tertiary amino; nitro; cyano; carboxyl; and acyl having a structure:

wherein R 54 is selected from substituted or unsubstituted alkyl; substituted or unsubstituted alkoxyl; and primary amino, secondary amino, or tertiary amino; or a pharmaceutically acceptable salt thereof.

[0089] In a yet further aspect, R 51 is substituted or unsubstituted alkyl having one or two hydroxyl groups. In a further aspect, R 51 is 2-hydroxyethyl, 2-hydroxypropyl, 1- methyl-2-hydroxyethyl, or 3-hydroxybutyl. In a further aspect, R 51 is selected from 2- sulphydrylethyl, 2-sulphydrylpropyl, l-methyl-2-sulphydrylethyl, and 3- sulphydrylbutyl .

[0090] It can be difficult to make the products in pure states if they are in gum forms or sticky forms. Some compounds in this invention have gum, sticky solid and crystal forms. At first, they were in gum form or sticky solid form even after they had gone through the chromatographic column and were pure indicated by thin layer chromatography (TLC). But these gum or sticky form could be changed into the

amorphous or other crystal form after being dissolved in some suitable solvents, such as methanol, ethanol, w-propanol, isopropanol and «-butanol, or acetone, or ethyl acetate, or the combination of them, and heated to reflux for 0 to 2 hours with our without the catalysis of several drops of concentrated hydrochloric acid. The molecular polarity is different among the gum, sticky solid and crystal forms. So the TLC behavior is also different among these forms. The molecular polarity of crystal form is greatest, sticky solid next, the gum form smallest.

C. Methods of making the compositions

[0091] The compositions disclosed herein and the compositions necessary to perform the disclosed methods can be made using any method known to those of skill in the art for that particular reagent or compound unless otherwise specifically noted.

[0092] hi one aspect, the invention relates to a method of preparing a curcumin derivative, the method comprising the step of reacting curcumin with a hydrazine derivative selected from a substituted or unsubstituted arylsulfonylhydrazide, a substituted or unsubstituted alkylcarbohydrazide, a substituted or unsubstituted sulphydrylalkylcarbohydrazide, a substituted or unsubstituted alkarylcarbohydrazide, a substituted or unsubstituted arylcarbohydrazide, a substituted or unsubstituted heterocyclic carbohydrazide, a substituted or unsubstituted carbohydrazide, a substituted or unsubstituted 2-aminoacetohydrazide, and a substituted or unsubstituted fused bicyclic or heterobicyclic carbohydrazide. hi one aspect, the curcumin derivative is at least one of the disclosed compounds.

[0093] hi one aspect, the method produces a hydrazone:

One example is the reaction of curcumin with benzenesulfonylhydrazide:

[0094] In one aspect, the method produces a pyrazole:

One example is the reaction of curcumin with ethyl 2-hydrazinylbenzoate:

[0095] Typically, hydrazine derivatives suitable for use in the inventive methods can be obtained commercially; however, the disclosed methods are not limited to commercially available starting materials and/or intermediates. Accordingly, in a further aspect, the method further comprises the step of synthesizing the hydrazine derivative. Hydrazine derivatives are well-known in the art; on of ordinary skill in the art of organic chemical synthesis can readily prepare hydrazines without undue experimentation with reference to published procedures and techniques. In one aspect, the synthesizing step comprises reduction of an aryl diazonium salt to form a hydrazine derivative.

[0096] Aryl diazonium salts can be prepared, for example, by treatment of an amine with sodium nitrite in the presence of a mineral acid. Reduction can be accomplished, for example, by treatment with sulfite salts.

[0097] In a further aspect, the synthesizing step comprises the steps of treating a carbonyl compound with at least two molar equivalents of hydrazine to form a hydrazone, and reducing the hydrazone to form a hydrazine derivative.

The hydrazone can be reduced under common reducing conditions, including sulfite salts.

[0098] In one aspect, the hydrazine derivative is a substituted or unsubstituted arylsulfonylhydrazide selected from benzenesulfonohydrazide and p- toluenesulfonohydrazide. In a further aspect, the hydrazine derivative is a substituted or unsubstituted carbohydrazide selected from benzohydrazide, nicotinohydrazide, isonicotinohydrazide, aminobenzohydrazide, hydroxybenzohydrazide, furancarbohydrazide, and thiophenecarbohydrazide. In a further aspect, the hydrazine derivative is a substituted or unsubstituted 2-aminoacetohydrazide 2- aminoacetohydrazide comprising a residue of arginine, asparagine, cysteine, histidine, hydroxylysine, hydroxyproline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophane, tyrosine, or valine.

D. Pharmaceutical compositions

[0099] In one aspect, the invention relates to pharmaceutical compositions comprising a therapeutically effective amount of at least one of the disclosed compounds or at least one of the disclosed products and a pharmaceutically acceptable carrier.

[00100] Compounds disclosed herein and compositions thereof can be administered by various methods including, for example, orally, intravenously, enterally, parenterally, topically, nasally, vaginally, opthalinically, sublingually or by inhalation for the treatment of diseases related to uncontrolled proliferative diseases such as, Routes of administration and dosages known in the art can be found in Comprehensive Medicinal Chemistry, Volume 5, Hansch, C. Pergamon Press, 1990; incorporated herein by reference in its entirety.

[00101] Although the compounds described herein can be administered as pure chemicals either singularly or plurally, it is preferable to present the active ingredient as a pharmaceutical composition. Thus another embodiment of the invention is the use of a pharmaceutical composition comprising one or more compounds and/or a pharmaceutically acceptable salt thereof, together with one or more pharmaceutically acceptable carriers thereof and, optionally, other therapeutic and/or prophylactic ingredients. The carrier(s) should be "acceptable" in the sense of being compatible with the other ingredients of the composition and not overly deleterious to the recipient thereof. The pharmaceutical composition, is administered to an animal diagnosed as in need of treatment for a disease of uncontrolled cellular proliferation, in an amount effective to treat the disease of uncontrolled cellular proliferation, such as the various cancers and precancerous conditions described herein.

[00102] It will be further appreciated that the amount of the compound, or an active salt or derivative thereof (i.e. a prodrug), required for effective use in treatment of a disease of uncontrolled cellular proliferation, such as the various cancers and precancerous conditions described herein, will vary not only with the particular compound and/or salt selected but also with the route of administration, the nature of the condition being treated, and the age and condition of the patient. An effective amount of a compound provided herein is a substantially nontoxic but sufficient amount of the compound to provide a clinically useful degree inhibition of the growth or progression of the disease of uncontrolled cellular proliferation.

[00103] Though it is not possible to specify a single predetermined pharmaceutically effective amount of the compounds of the invention, and/or their pharmaceutical compositions, for each and every disease condition to be treated, determining such pharmaceutically effective amounts are within the skill of, and ultimately at the discretion of an attendant physician or clinician of ordinary skill. In some embodiments, the active compounds of the invention are administered to achieve peak plasma concentrations of the active compound of from typically about 0.1 to about 100 μM, about 1 to 50 μM, or about 2 to about 30 μM. This can be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 0.5-500 mg of the active ingredient. Desirable blood levels can be maintained

by continuous infusion to provide about 0.01-5.0 mg/kg/hr or by intermittent infusions containing about 0.4-15 mg/kg of the active compounds of the invention.

[00104] Pharmaceutical compositions include those suitable for oral, enteral, parental (including intramuscular, subcutaneous and intravenous), topical, nasal, vaginal, ophthalinical, sublingually or by inhalation administration. The compositions can, where appropriate, be conveniently presented in discrete unit dosage forms and can be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with liquid carriers, solid matrices, semi-solid carriers, finely divided solid carriers or combination thereof, and then, if necessary, shaping the product into the desired delivery system.

[00105] When desired, the above-described compositions can be adapted to provide sustained release of the active ingredient employed, e.g., by combination thereof with certain hydrophilic polymer matrices, e.g., comprising natural gels, synthetic polymer gels or mixtures thereof.

[00106] The compounds of the invention can have oral bioavailability as exhibited by blood levels after oral dosing, either alone or in the presence of an excipient. Oral bioavailability allows oral dosing for use in chronic diseases, with the advantage of self-administration and decreased cost over other means of administration.

[00107] Pharmaceutical compositions suitable for oral administration can be presented as discrete unit dosage forms such as hard or soft gelatin capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or as granules; as a solution, a suspension or as an emulsion. The active ingredient can also be presented as a bolus, electuary or paste. Tablets and capsules for oral administration can contain conventional excipients such as binding agents, fillers, lubricants, disintegrants, or wetting agents. The tablets can be coated according to methods well known in the art., e.g., with enteric coatings.

[00108] Oral liquid preparations can be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or can be presented as a dry

product for constitution with water or other suitable vehicle before use. Such liquid preparations can contain conventional additives such as suspending agents, emulsifying agents, non-aqueous vehicles (which can include edible oils), or one or more preservative.

[00109] The compounds can also be formulated for parenteral administration

(e.g., by injection, for example, bolus injection or continuous infusion) and can be presented in unit dose form in ampules, pre- filled syringes, small bolus infusion containers or in multi-does containers with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient can be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen- free water, before use.

[00110] For topical administration to the epidermis, the compounds can be formulated as ointments, creams or lotions, or as the active ingredient of a transdermal patch. Suitable transdermal delivery systems are disclosed, for example, in Fisher et al. (U.S. Patent (No. 4,788,603, incorporated herein by reference) or Bawas et al. (U.S. Patent No. 4,931,279, 4,668,504 and 4,713,224; all incorporated herein by reference). Ointments and creams can, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions can be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents. The active ingredient can also be delivered via iontophoresis, e.g., as disclosed in U.S. Patent Nos. 4,140,122, 4383,529, or 4,051,842; incorporated herein by reference.

[00111] Compositions suitable for topical administration in the mouth include unit dosage forms such as lozenges comprising active ingredient in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; mucoadherent gels, and mouthwashes comprising the active ingredient in a suitable liquid carrier.

[00112] When desired, the above-described compositions can be adapted to provide sustained release of the active ingredient employed, e.g., by combination thereof with certain hydrophilic polymer matrices, e.g., comprising natural gels, synthetic polymer gels or mixtures thereof.

[00113] The pharmaceutical compositions according to the invention can also contain other adjuvants such as flavorings, coloring, antimicrobial agents, or preservatives.

[00114] It will be further appreciated that the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.

[00115] m general, one of skill in the art understands how to extrapolate in vivo data obtained in a model organism, such as athymic nude mice inoculated with human tumor cell lines, to another mammal, such as a human. These extrapolations are not simply based on the weights of the two organisms, but rather incorporate differences in metabolism, differences in pharmacological delivery, and administrative routes. Based on these types of considerations, a suitable dose will, in alternative embodiments, typically be in the range of from about 0.5 to about 10 mg/kg/day, or from about 1 to about 20 mg/kg of body weight per day, or from about 5 to about 50 mg/kg/day.

[00116] The desired dose can conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose, as necessary by one skilled in the art, can itself be further divided, e.g., into a number of discrete loosely spaced administrations.

[00117] One skilled in the art will recognize that dosage and dosage forms outside these typical ranges can be tested and, where appropriate, be used in the methods of this invention.

[00118] According to another aspect of the invention, pharmaceutical compositions of matter useful for the treatment of cancer are provided that contain, in addition to the aforementioned compounds, an additional therapeutic agent. Such agents can be chemotherapeutic agents, ablation or other therapeutic hormones, antineoplastic agents, monoclonal antibodies useful against cancers and angiogenesis inhibitors. The following discussion highlights some agents in this respect, which are illustrative, not limitative. A wide variety of other effective agents also can be used.

[00119] Among hormones which can be used in combination with the present inventive compounds, diethylstilbestrol (DES), leuprolide, fiutamide, cyproterone acetate, ketoconazole and amino glutethimide.

[00120] Among antineoplastic and anticancer agents that can be used in combination with the inventive compounds, 5-fluorouracil, vinblastine sulfate, estramustine phosphate, suramin and strontium-89. Other chemotherapeutics useful in combination and within the scope of the present invention are buserelin, chlorotranisene, chromic phosphate, cisplatin, cyclophosphamide, dexamethasone, doxorubicin, estradiol, estradiol valerate, estrogens conjugated and esterified, estrone, ethinyl estradiol, floxuridine, goserelin, hydroxyurea, melphalan, methotrexate, mitomycin, prednisone and tamoxifen.

E. Pharmaceutical carriers/Delivery of pharamceutical products [00121] As described above, the compositions can also be administered in vivo in a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to a subject, along with the nucleic acid or vector, without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained. The carrier would naturally be selected to minimize any degradation of the active ingredient and to minimize any adverse side effects in the subject, as would be well known to one of skill in the art.

[00122] The compositions maybe administered orally, parenterally (e.g., intravenously), by intramuscular injection, by intraperitoneal injection, transdermally,

extracorporeally, topically or the like, including topical intranasal administration or administration by inhalant. As used herein, "topical intranasal administration" means delivery of the compositions into the nose and nasal passages through one or both of the nares and can comprise delivery by a spraying mechanism or droplet mechanism, or through aerosolization of the nucleic acid or vector. Administration of the compositions by inhalant can be through the nose or mouth via delivery by a spraying or droplet mechanism. Delivery can also be directly to any area of the respiratory system (e.g., lungs) via intubation. The exact amount of the compositions required will vary from subject to subject, depending on the species, age, weight and general condition of the subject, the severity of the allergic disorder being treated, the particular nucleic acid or vector used, its mode of administration and the like. Thus, it is not possible to specify an exact amount for every composition. However, an appropriate amount can be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein.

[00123] Parenteral administration of the composition, if used, is generally characterized by injection. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution of suspension in liquid prior to injection, or as emulsions. A more recently revised approach for parenteral administration involves use of a slow release or sustained release system such that a constant dosage is maintained. See, e.g., U.S. Patent No. 3,610,795, which is incorporated by reference herein.

[00124] The materials may be in solution, suspension (for example, incorporated into microparticles, liposomes, or cells). These may be targeted to a particular cell type via antibodies, receptors, or receptor ligands. The following references are examples of the use of this technology to target specific proteins to tumor tissue (Senter, et al., Bioconjugate Chem., 2:447-451, (1991); Bagshawe, K.D., Br. J. Cancer, 60:275-281, (1989); Bagshawe, et al., Br. J. Cancer, 58:700-703, (1988); Senter, et al., Bioconjugate Chem., 4:3-9, (1993); Battelli, et al., Cancer Immunol. Immunother., 35:421-425, (1992); Pietersz and McKenzie, Immunolog. Reviews, 129:57-80, (1992); and Roffler, et al., Biochem. Pharmacol, 42:2062-2065, (1991)). Vehicles such as "stealth" and other antibody conjugated liposomes (including lipid mediated drug targeting to colonic carcinoma), receptor mediated targeting of DNA

through cell specific ligands, lymphocyte directed tumor targeting, and highly specific therapeutic retroviral targeting of murine glioma cells in vivo. The following references are examples of the use of this technology to target specific proteins to tumor tissue (Hughes et al., Cancer Research, 49:6214-6220, (1989); and Litzinger and Huang, Biochimica et Biophysica Acta, 1104: 179-187, (1992)). In general, receptors are involved in pathways of endocytosis, either constitutive or ligand induced. These receptors cluster in clathrin-coated pits, enter the cell via clathrin- coated vesicles, pass through an acidified endosome in which the receptors are sorted, and then either recycle to the cell surface, become stored intracellularly, or are degraded in lysosomes. The internalization pathways serve a variety of functions, such as nutrient uptake, removal of activated proteins, clearance of macromolecules, opportunistic entry of viruses and toxins, dissociation and degradation of ligand, and receptor-level regulation. Many receptors follow more than one intracellular pathway, depending on the cell type, receptor concentration, type of ligand, ligand valency, and ligand concentration. Molecular and cellular mechanisms of receptor-mediated endocytosis has been reviewed (Brown and Greene, DNA and Cell Biology 10:6, 399- 409 (1991)).

1. Pharmaceutically Acceptable Carriers

[00125] The compositions, including antibodies, can be used therapeutically in combination with a pharmaceutically acceptable carrier.

[00126] Suitable carriers and their formulations are described in Remington: The Science and Practice of Pharmacy (19th ed.) ed. A.R. Gennaro, Mack Publishing Company, Easton, PA 1995. Typically, an appropriate amount of a pharmaceutically- acceptable salt is used in the formulation to render the formulation isotonic. Examples of the pharmaceutically- acceptable carrier include, but are not limited to, saline, Ringer's solution and dextrose solution. The pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5. Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon,

for instance, the route of administration and concentration of composition being administered.

[00127] Pharmaceutical carriers are known to those skilled in the art. These most typically would be standard carriers for administration of drugs to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH. The compositions can be administered intramuscularly or subcutaneously. Other compounds will be administered according to standard procedures used by those skilled in the art.

[00128] Pharmaceutical compositions may include carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like in addition to the molecule of choice. Pharmaceutical compositions may also include one or more active ingredients such as antimicrobial agents, antiinflammatory agents, anesthetics, and the like.

[00129] The pharmaceutical composition may be administered in a number of ways depending on whether local or systemic treatment is desired, and on the area to be treated. Administration may be topically (including ophthalmically, vaginally, rectally, intranasally), orally, by inhalation, or parenterally, for example by intravenous drip, subcutaneous, intraperitoneal or intramuscular injection. The disclosed antibodies can be administered intravenously, intraperitoneally, intramuscularly, subcutaneously, intracavity, or transdermally.

[00130] Preparations for parenteral administration include sterile aqueous or nonaqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.

[00131] Formulations for topical administration may include ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.

[00132] Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets, or tablets. Thickeners, flavorings, diluents, emulsifiers, dispersing aids or binders may be desirable..

[00133] Some of the compositions may potentially be administered as a pharmaceutically acceptable acid- or base- addition salt, formed by reaction with inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base such as sodium hydroxide, ammonium hydroxide, potassium hydroxide, and organic bases such as mono-, di-, trialkyl and aryl amines and substituted ethanolamines.

2. Therapeutic Uses

[00134] Effective dosages and schedules for administering the compositions may be determined empirically, and making such determinations is within the skill in the art. The dosage ranges for the administration of the compositions are those large enough to produce the desired effect in which the symptoms of the disorder are effected. The dosage should not be so large as to cause adverse side effects, such as unwanted cross-reactions, anaphylactic reactions, and the like. Generally, the dosage will vary with the age, condition, sex and extent of the disease in the patient, route of administration, or whether other drugs are included in the regimen, and can be determined by one of skill in the art. The dosage can be adjusted by the individual physician in the event of any counter-indications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. Guidance can be found in the literature for appropriate dosages for given classes of pharmaceutical products. For example, guidance in selecting appropriate doses for

antibodies can be found in the literature on therapeutic uses of antibodies, e.g., Handbook of Monoclonal Antibodies, Ferrone et al., eds., Noges Publications, Park Ridge, N. J., (1985) ch. 22 and pp. 303-357; Smith et al., Antibodies in Human Diagnosis and Therapy, Haber et al., eds., Raven Press, New York (1977) pp. 365- 389. A typical daily dosage of the antibody used alone might range from about 1 μg/kg to up to 100 mg/kg of body weight or more per day, depending on the factors mentioned above.

[00135] Following administration of a disclosed composition, for treating, inhibiting, or preventing a GI syndrome, BM syndrome, or inflammation of ARS, the efficacy of the therapeutic compositions can be assessed in various ways well known to the skilled practitioner. For instance, one of ordinary skill in the art will understand that a composition, disclosed herein is efficacious in treating or inhibiting a GI syndrome, BM syndrome, or inflammation of ARS in a subject by observing that the composition reduces death, cytokine levels, or increases the health of the subject.

F. Methods of using the compositions

[00136] Disclosed herein are methods of treating radiation damage. Ionizing radiation (PR) remains a main stream therapy for cancer, since it controls both primary and metastatic cancer without significant systemic damage. However, radiation therapy does cause IR-induced local damage of normal tissue (radiation toxicity), leading to a temporary or persistent impairment of irradiated tissues, which lowers the life quality of cancer patients. Some severe side effects such as the acute radiation syndrome conditions of gastrointestinal syndrome and bone marrow syndrome can even result in the discontinuation of the life-saving radiation therapy (Johansen et al. Radiother Oncol. 40: 101-9 (1996), Niemierko et al. IntJRadiat Oncol Biol Phys. 25: 135-45, 1993., Wiess et al. Toxicology 15;189(l-2):l-20 (2003 JuI). Radiation damage can also occur by exposure to nuclear radiation, or exposure to a weapon that causes radiation. It is understood and herein contemplated that by "weapon" is meant any bomb, machine, or other device capable of being used in convention warfare, nonconventional warfare, or terrorist activities.

[00137] Disclosed herein are methods of reducing radiation damage in a subject comprising administering to the subject an effective amount of an analogue,

derivative, or metabolite of curcumin or other compositions disclosed herein. As disclosed above, the radiation damage can be caused by radiation therapy, such as that used to treat cancer. The radiation damage can also be caused by nuclear radiation, or by a weapon, such as a bomb, terrorist agent. The compositions herein can be admininstered prior to, after, or during exposure to radiation. Thus, disclosed herein are methods of treating, inhibiting, preventing, or mitigating radiation toxicity, radiation induced gastroinstestinal (GI) syndrome or bone marrow (BM) syndrome, acute radiation syndrome (ARS), lethal brain bleeding, or the effects associated with any of the above conditions, wherein the agent is administered following exposure to radiation. It is understood and herein contemplated that the disclosed compositions can be used to prevent gastroinstestinal (GI) syndrome or bone marrow (BM) syndrome, acute radiation syndrome (ARS), lethal brain bleeding, or the effects associated with any of the above conditions. It is further understood that the disclosed compositions can be administered one, two, three, or four times every 24 hours. It is further understood that the agent of the disclosed methods can be administered every 24 hours for 1, 2, 3, 4, 5, 6, 7, 14, or 21 days or at any time point in between.

[00138] As used herein, "mitigate" means to reduce the damage associated with a symptom, disease, or condition relative to the untreated state. It is also understood that "mitigation" can be in reference to a symptom, disease, or condition, in addition to or alternatively to damage associated with the symptom, disease, or condition. It is understood and herein contemplated that the reduction is not limited to the complete ablation of the damage, symptom, disease, or condition, but may include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to untreated, native, or control levels. It is also understood that "mitigation" has occurred if the further damage due to disease progression or symptoms are reduced without a reduction in the state prior to treatment. Thus, for example, in a subject with damage from radiation toxicity prior to treatment, the radiation toxicity would be "mitigated" if, following treatment, further damage from progression of toxicity was reduced relative to a control, even if the level of damage in the subject was not reduced relative to pre treatment levels. By way of example, if a subject had a radiation toxicity damage at a

level of X when treatment was started, a composition or treatment method would be understood to mitigate the damage from radiation toxicity even if the damage from radiation toxity increased (e.g., X+10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%) provided untreated controls increased more (e.g., X+15%, 25%, 35%, 45%, 55%, 65%, 75%, 85%, 95%).

[00139] "Inhibit," "inhibiting," and "inhibition" mean to decrease an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.

[00140] "Treatment," "treat," or "treating" mean a method of reducing the effects of a disease or condition. Treatment can also refer to a method of reducing the disease or condition itself rather than just the symptoms. The treatment can be any reduction from native levels and can be but is not limited to the complete ablation of the disease, condition, or the symptoms of the disease or condition. Therefore, in the disclosed methods, treatment" can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in the severity of an established disease or the disease progression. For example, a disclosed method for reducing the effects of radiation toxicity, gastrointestinal syndrome, bone marrow syndrome, inflammation, or uncontrolled cellular proliferation is considered to be a treatment if there is a 10% reduction in one or more symptoms of the disease in a subject with the disease when compared to native levels in the same subject or control subjects. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels. It is understood and herein contemplated that "treatment" does not necessarily refer to a cure of the disease or condition, but an improvement in the outlook of a disease or condition.

[00141] Therefore, in one aspect, the invention relates to a method of mitigating radiation toxicity in a subject comprising the step of administering to the subject at least one of the disclosed compounds at least one of the disclosed pharmaceutical

compositions, or at least one of the disclosed products in a dosage and amount effective to mitigate radiation toxicity in the subject. In one aspect, the subject is a mammal, for example, a human. In a further aspect, the subject has been diagnosed with a need for mitigating radiation toxicity prior to the administering step. In a yet further aspect, the administering step is performed before the subject acquires radiation toxicity.

[00142] Pn one aspect, the invention relates to a method of treating radiation- induced gastrointestinal syndrome in a subject comprising the step of administering to the subject at least one of the disclosed compounds at least one of the disclosed pharmaceutical compositions, or at least one of the disclosed products in a dosage and amount effective to treat gastrointestinal syndrome in the subject, hi one aspect, the subject is a mammal, for example, a human, hi a further aspect, the subject has been diagnosed with a need for treatment of gastrointestinal syndrome, hi a further aspect, the administering step is performed after the subject is exposed to irradiation.

[00143] hi one aspect, the invention relates to a method for the treatment of a disease of uncontrolled cellular proliferation comprising administering to a subject having a disease of uncontrolled cellular proliferation at least one of the disclosed compounds at least one of the disclosed pharmaceutical compositions, or at least one of the disclosed products in a dosage and amount effective to treat the disease of uncontrolled cellular proliferation, hi one aspect, the subject is a mammal, for example, a human. In a further aspect, the subject has been diagnosed with a need for treatment of a disease of uncontrolled cellular proliferation, hi a further aspect, the disease of uncontrolled proliferation is a carcinoma, lymphoma, leukemia, or sarcoma.

[00144] The disease of uncontrolled cellular proliferation treated can be a carcinoma, lymphoma, leukemia, or sarcoma. The types of cancer treated by methods of the invention include but are not limited to Hodgkin's Disease, meyloid leukemia, polycystic kidney disease, bladder cancer, brain cancer, head and neck cancer, kidney cancer, lung cancer, myeloma, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, colon cancer, cervical carcinoma, breast cancer, epithelial cancer, and leukemia. The compositions can also be used as regulators in diseases of uncontrolled proliferation and/or

precancerous conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias.

[00145] The effectiveness of the methods for treating the diseases of uncontrolled cellular proliferation can vary as a function of several variables, including the specific genetic nature of disease or cancer, the details of the method of administration of the compound, the exact structure of the compounds administered, and other factors which are known to those of ordinary skill in the art.

[00146] The compounds disclosed herein can be either used singularly, or plurally, in mixtures of one or more compounds, tautomers, isomers, or enantiomers, and in pharmaceutical compositions thereof, for the treatment of mammalian diseases of uncontrolled cellular proliferatio, particularly those diseases related to humans.

[00147] Disclosed herein are methods of treating, preventing, or reducing radiation- induced gastrointestinal syndrome, radiation toxicity, or the damaging effects associate with either condition comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin. It is understood and herein contemplated that the agent can be administered one, two, three, or four times every 24 hours. It is further understood that the agent of the disclosed methods can be administered every 24 hours for 1, 2, 3, 4, 5, 6, 7, 14, or 21 days. Also disclosed are methods further comprising the administration of a therapy that reduces the effects of ionizing radiation. It is understood and herein contemplated that the therapy can comprise any procedure or composition known by those of skill in the art to reduce the effects of ionizing radiation. It is further understood that the therapy can comprise a second agent. It is contemplated herein that the agent can be administered before or after the exposure to ionizing irradiation. It is further contemplated that the agent is an amifostine. Thus, for example, disclosed herein are methods wherein the second agent is an amifostine administered prior to exposure to IR.

[00148] Also disclosed are methods of mitigating, treating, preventing, or reducing radiation-induced bone marrow syndrome or the effects thereof comprising

administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

[00149] Further disclosed are are methods of mitigating, treating, preventing, or reducing lethal brain bleeding or the damaging effects thereof comprising administering to a subject and effective amount of an agent, wherein the agent is a derivative, analogue, or metabolite of curcumin.

[00150] Disclosed herein are methods of administering a sub-total body irradiation (sub-TBI) dose to a subject comprising administering to the subject a lethal dose of ionizing radiation (IR) and simultaneously shielding an keeping out of the IR field an appendage of the subject. It is understood and herein contemplated that the appendage can be any appendage comprising a bone comprising bone marrow. Thus, for example, the appendage can be an arm or a leg. It if further understood and herein contemplated that the dose of radiation can be any dose between 5 and 15 Gy. Thus, for example disclosed are methods wherein the dose of radiation is between 8 and 12 Gy. Also disclosed are methods wherein the dose of radiation is between 8 and 10.5 Gy. Also disclosed are methods wherein the radiation dose is between 8.9 and 9.1 Gy. It is understood that the subject can be any animal that is effected by radiation. Thus, for example, the subject can be a mouse.

[00151] It is understood and herein contemplated that the disclosed methods of administering sub total body irradiation can be used to make a model for radiation induced gastrointestinal syndrome. It is further understood that such a model can be used to screen for an agent or test a known agent for effectiveness in treating, reducing, preventing, or inhibiting gastrointestinal syndrome. It is further understood that such a model can be used to screen for an agent or test an known agent for efficacy in treating, reducing, preventing, inhibiting, or mitigating radiation toxicity.

[00152] Screening for agents that mitigate radiation toxicity can be useful for subjects exposed to ionizing radiation. Therefore, disclosed herein are methods of screening for an agent that mitigate, inhibit, treat, or reduce radiation toxicity comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure

and administering the agent following IR exposure, wherein a reduction in the toxic effects of IR relative to a control indicate an agent that treats radiation toxicity. It is understood that the same methods can be used to screen for agents effective against GI syndrome. Thus, for example disclosed herein are methods of screening for an agent that mitigate, inhibit, treat, or reduce radiation toxicity comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure and administering the agent following IR exposure, wherein a reduction in the toxic effects of IR relative to a control indicate an agent that treats gastrointestinal syndrome. It is further understood that same methods can be used to screen for agents effective against acute radiation syndrome. Thus, for example, disclosed herein are methods of screening for an agent that mitigate, inhibit, treat, or reduce radiation toxicity comprising administering a lethal dose of ionizing radiation (IR) to a subject, wherein on appendage of the subject is shielded and kept out of the IR field during IR exposure and administering the agent following IR exposure, wherein a reduction in the toxic effects of IR relative to a control indicate an agent that treats acute radiation syndrome.

[00153] Radiation toxicity can be divided into two main stages: early toxicity and late toxicity (MacKay et al. Radiother Oncol. 46:215-6 (1998), Rubin et al. Radiother Oncol. 35: 9-10, (199?), Dubray et al. Cancer Radiother. 1: 744-52 (1997), Vozenin- Brotons et al. RadiatRes. 152: 332-7 (1999), Lefaix et al. Br J Radiol Suppl. 19: 109- 13 (1986), Lefaix et al. Soc Biol FiI. 191: 777-95 (1997), Verola et al. Br J Radiol Suppl. 19: 104-8 (1986)). For example, in the irradiated soft tissue, there is early radiation dermatitis (ERD) that occurs within one month after IR, and late radiation fibrosis (LRF) which develops two months later. In the irradiated lung, there is pnuemonitis (early) and lung fibrosis (late) (Chen et al. Int J Radiat Oncol Biol Phys. 49: 641-8 (2001), Chen et al. Seminars in Radiation Oncology 12: 26-33 (2002), Marks et al. Int J Radiat Biol. 76: 469-75 (2000)). In the irradiated brain, there is brain edema (early) and brain degeneration (late). The pathophysiological mechanisms underlying these phenomena have been studied, but remain unclear.

[00154] hi general, IR kills cell through the production of free radicals. The IR toxicity is a result of counteraction of host defense system that responds to IR physical insult. Upon IR, the cells are damaged by free radicals, and undergo either repair or

apoptosis/death, which initiates the cascade of signal transduction pathways (such as Nuclear factor-κB (NFKjS), etc.). As a result, IR up-regulates the expression of inflammatory mediators (such as cytokines, lymphokines and chemokines) and immunomodulatory molecules (MHC, co-stimulatory molecules, adhesion molecules, death receptors, heat shock proteins) in irradiated tumor, stromal, and vascular endothelial cells (Friedman et al.). Among them, for example, ILl β, IL6, MCP-I, COX-2 and TGFα play critical roles in IR toxicity (Chen et al. (2001) Hallahan et al. Important Adv Oncol. :71-80 (1993)). The accumulated cytokines and chemokines attract the immune cells (such as macrophages, dendritic cells, T cells and B cells) to the irradiated spot to engulf the apoptotic and necrotic cellular debris. After internalizing the debris, some of the mutated normal tissue "self antigens can be presented by dendritic cells to T cells (McBride et al. Radiat Res.162(1): 1-19 (2004 JuI)). The interaction of sensitized T cells with the existing IR-induced mutated "wrong proteins" or "wrong genes" (which can pass to daughter cells) in irradiated normal tissues triggers a new wave of mass production of cytokines, which occurs a few months after IR, which may be the driving force for the late toxicity (chronic inflammation). This process is evidenced by the several waves of mass production of secretory molecules (cytokines and inflammatory mediators) at the stages of early and late toxicity.

[00155] A network exists between IR-induced molecules, such as interaction among NO, NF-KjS, cytokines and COX. An interaction loop and feed-back control exists among these molecules. Upon IR, the NO and the signaling of DNA breakage directly activate NF-kj3, which induces IL1/3. The ILIjS binds to its receptors, which again triggers NFkjS and P38 pathways to enhance its production, a positive feed-back to amplify the inflammation signaling. ILl β is a key cytokine in the IR inflammation process. As one of the effects, ILIjS enhances the expression of COX-2, and together they markedly induce inflammatory angiogenesis (Kuwano et al. FASEB J. 18(2):300- 10 (2004)), a critical process in IR inflammation (toxicity). As a control, IL-IB- induced activation of the COX-2 gene is modulated by NF-kj3 (Kirtikara et al. (2000), Crofford et al. Arthritis Rheum. 40,226-236 (1997)). The COX-2 selective inhibitors can block ILIjS induced angiogenesis but only partially block VEGF-induced angiogenesis. Similarly, the ILIjS induced angiogenesis is much less in the COX-2

knockout mice than wild-type mice (Kuwano et al. (2004)). Overexpression of COX-2 also is accompanied by up-regulation of nitric oxide synthases (Tsuji et al. Nippon Rinsho. 56: 2247-2252 (1998)), which can intensify local damage.

[00156] Cyclooxygenase is the rate-limiting step in the conversion of arachidonic acid to prostaglandins. There are two known genes of cyclooxygenase, COX-I and COX-2. COX-I is constitutively expressed at low levels in many cell types. Specifically, COX-I is known to be essential for maintaining the integrity of the gastrointestinal epithelium. COX-2 expression is stimulated by growth factors, cytokines, and endotoxins. The cyclooxygenase 2 isoform (COX-2) is not expressed in most tissues (e.g., liver) under physiological conditions but is highly upregulated in inflammatory processes and cancer, for example. Up-regulation of COX-2 is responsible for the increased formation of prostaglandins associated with inflammation.

[00157] Inflammation is a complex stereotypical reaction of the body expressing the response to damage of its cells and vascularized tissues. The discovery of the detailed processes of inflammation has revealed a close relationship between inflammation and the immune response. The main features of the inflammatory response are vasodilation, i.e. widening of the blood vessels to increase the blood flow to the infected area; increased vascular permeability, which allows diffusible components to enter the site; cellular infiltration by chemotaxis, or the directed movement of inflammatory cells through the walls of blood vessels into the site of injury; changes in biosynthetic, metabolic, and catabolic profiles of many organs; and activation of cells of the immune system as well as of complex enzymatic systems of blood plasma.

[00158] There are two forms of inflammation, acute and chronic. Acute inflammation can be divided into several phases. The earliest, gross event of an inflammatory response is temporary vasoconstriction, i.e. narrowing of blood vessels caused by contraction of smooth muscle in the vessel walls, which can be seen as blanching (whitening) of the skin. This is followed by several phases that occur over minutes, hours and days later. The first is the acute vascular response, which follows within seconds of the tissue injury and lasts for several minutes. This results from vasodilation and increased capillary permeability due to alterations in the vascular

endothelium, which leads to increased blood flow {hyperemia) that causes redness {erythema) and the entry of fluid into the tissues {edema).

[00159] Examples of chronic inflammatory diseases include tuberculosis, chronic cholecystitis, bronchiectasis, rheumatoid arthritis, Hashimoto's thyroiditis, inflammatory bowel disease (ulcerative colitis and Crohn's disease), silicosis and other pneumoconiosis, and implanted foreign body in a wound.

[00160] Activated cells can also be identified at the site of inflammation. "Activated cells" are defined as cells that participate in the inflammatory response. Examples of such cells include, but are not limited to, T-cells and B-cells , macrophages, NK cells, mast cells, eosinophils, neutrophils, Kupffer cells, antigen presenting cells, as well as vascular endothelial cells.

[00161] Macrophages release cytokines (e.g., tumor necrosis factor, interleukin-1), which heighten the intensity of inflammation by stimulating inflammatory endothelial responses; these endothelial changes help recruit large numbers of T cells to the inflammatory site.

[00162] Damaged tissues release pro-inflammatory mediators (e.g., Hageman factor (factor XIl) that trigger several biochemical cascades. The clotting cascade induces fibrin and several related fibrinopeptides, which promote local vascular permeability and attract neutrophils and macrophages. The kinin cascade principally produces bradykinin, which promotes vasodilation, smooth muscle contraction, and increased vascular permeability.

[00163] Dislcosed herein are methods of treating inflammation in a subject by administering to the subject an effective amount of a derivative, analogue, or metabolite of curcumin. "Inflammation" or "inflammatory" herein, is defined as the reaction of living tissues to injury, infection, or irritation. Anything that stimulates an inflammatory response is said to be inflammatory.

[00164] "Inflammatory disease" is defined as any disease state associated with inflammation. The inflammation can be associated with an inflammatory disease. Examples of inflammatory disease include, but are not limited to, asthma, systemic

lupus erythematosus, rheumatoid arthritis, reactive arthritis, spondyarthritis, systemic vasculitis, insulin dependent diabetes mellitus, multiple sclerosis, experimental allergic encephalomyelitis, Sjogren's syndrome, graft versus host disease, inflammatory bowel disease (including Crohn's disease and ulcerative colitis) and scleroderma, myasthenia gravis, Guillain-Barre disease, primary biliary cirrhosis, hepatitis, hemolytic anemia, uveitis, Grave's disease, pernicious anemia, thrombocytopenia, Hashimoto's thyroiditis, oophoritis, orchitis, adrenal gland diseases, anti-phospholipid syndrome, Wegener's granulomatosis, Behcet's disease, polymyositis, dermatomyositis, multiple sclerosis, vitiligo, ankylosing spondylitis, Pemphigus vulgaris, psoriasis, and dermatitis herpetiformis.

[00165] It is understood and herein contemplated that primary mediators of inflammation include cytokines. Thus, disclosed are methods of inhibiting cytokines in a subject exposed to radiation. Examples of cytokines involved in inflammation include by are not limited to ILl β, IL6, KC, TNFα, TGF/3, VEGF, TCA-3, G-CSF, and MCPl or any combination thereof.

[00166] Inflammation can be associated with a number of different diseases and disorders. Examples of inflammation include, but are not limited to, inflammation associated with hepatitis, inflammation associated with the lungs, and inflammation associated with an infectious process. Inflammation can also be associated with liver toxicity, which can be associated in turn with cancer therapy, such as apoptosis induction or chemotherapy, or a combination of the two, for example.

[00167] When the inflammation is associated with an infectious process, the infectious process can be associated with a viral infection. Examples of viral infections include, but are not limited to, Herpes simplex virus type-1, Herpes simplex virus type-2, Cytomegalovirus, Epstein-Barr virus, Varicella-zoster virus, Human herpesvirus 6, Human herpesvirus 7, Human herpesvirus 8, Variola virus, Vesicular stomatitis virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C virus, Hepatitis D virus, Hepatitis E virus, Rhinovirus, Coronavirus, Influenza virus A, Influenza virus B, Measles virus, Polyomavirus, Human Papilomavirus, Respiratory syncytial virus, Adenovirus, Coxsackie virus, Dengue virus, Mumps virus, Poliovirus, Rabies virus, Rous sarcoma virus, Yellow fever virus, Ebola virus, Marburg virus, Lassa fever

virus, Eastern Equine Encephalitis virus, Japanese Encephalitis virus, St. Louis Encephalitis virus, Murray Valley fever virus, West Nile virus, Rift Valley fever virus, Rotavirus A, Rotavirus B, Rotavirus C, Sindbis virus, Simian Immunodeficiency cirus, Human T-cell Leukemia virus type-1, Hantavirus, Rubella virus, Simian Immunodeficiency virus, Human Immunodeficiency virus type-1, and Human Immunodeficiency virus type-2.

[00168] The infectious process can also be associated with a bacterial infection. Examples of bacterial infections include, but are not limited to, M. tuberculosis, M. bovis, M. bovis strain BCG, BCG substrains, M. avium, M. intracellular e, M. africanum, M. kansasii, M. marinum, M. ulcerans, M. avium subspecies paratuberculosis, Nocardia asteroides, other Nocardia species, Legionella pneumophila, other Legionella species, Salmonella typhi, other Salmonella species, Shigella species, Yersinia pestis, Pasteurella haemolytica, Pasteurella multocida, other Pasteurella species, Actinobacillus pleuropneumoniae, Listeria monocytogenes, Listeria ivanovii, Brucella abortus, other Brucella species, Cowdria ruminantium, Chlamydia pneumoniae, Chlamydia trachomatis, Chlamydia psittaci, Coxiella burnetti, other Rickettsial species, Ehrlichia species, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, Streptococcus agalactiae, Bacillus anthracis, Escherichia coli, Vibrio cholerae, Campylobacter species, Neiserria meningitidis, Neiserria gonorrhea, Pseudomonas aeruginosa, other Pseudomonas species, Haemophilus influenzae, Haemophilus ducreyi, other Hemophilus species, Clostridium tetani, other Clostridium species, Yersinia enterolitica, and other Yersinia species.

[00169] The infectious process can also be associated with a parasitic infection. Examples of parasitic infections include, but are not limited to, Toxoplasma gondii, Plasmodium species such as Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae, and other Plasmodium species, Trypanosoma brucei, Trypanosoma cruzi, Leishmania species such as Leishmania major, Schistosoma such as Schistosoma mansoni and other Shistosoma species, and Entamoeba histolytica.

[00170] The infectious process can also be associated with a fungal infection. Examples of fungal infections include, but are not limited to, Candida albicans,

Ctyptococcus neoformans, Histoplama capsulatum, Aspergillus fumigatus, Coccidiodes immitis, Paracoccidiodes brasiliensis, Blastomyces dermitidis, Pneumocystis carnii, Penicillium marneffi, and Alternaria alternata.

[00171] The inflammation can be associated with cancer. Examples of types of cancer include, but are not limited to, lymphoma (Hodgkins and non-Hodgkins) B-cell lymphoma, T-cell lymphoma, leukemia such as myeloid leukemia and other types of leukemia, mycosis fungoide, carcinoma, adenocarcinoma, sarcoma, glioma, blastoma, neuroblastoma, plasmacytoma, histiocytoma, melanoma, adenoma, hypoxic tumor, myeloma, AIDS-related lymphoma or AIDS-related sarcoma, metastatic cancer, bladder cancer, brain cancer, nervous system cancer, squamous cell carcinoma of the head and neck, neuroblastoma, glioblastoma, ovarian cancer, skin cancer, liver cancer, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer, breast cancer, cervical carcinoma, epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, head and neck carcinoma, hematopoietic cancer, testicular cancer, colo-rectal cancer, prostatic cancer, and pancreatic cancer.

[00172] The disclosed compositions can be used to treat any disease where uncontrolled cellular proliferation occurs such as cancers. A non-limiting list of different types of cancers is as follows: lymphomas (Hodgkins and non-Hodgkins), leukemias, carcinomas, carcinomas of solid tissues, squamous cell carcinomas, adenocarcinomas, sarcomas, gliomas, high grade gliomas, blastomas, neuroblastomas, plasmacytomas, histiocytomas, melanomas, adenomas, hypoxic tumours, myelomas, AIDS-related lymphomas or sarcomas, metastatic cancers, or cancers in general.

[00173] A representative but non-limiting list of cancers that the disclosed compositions can be used to treat is the following: lymphoma, B cell lymphoma, T cell lymphoma, mycosis fungoides, Hodgkin's Disease, myeloid leukemia, bladder cancer, brain cancer, nervous system cancer, head and neck cancer, squamous cell carcinoma of head and neck, kidney cancer, lung cancers such as small cell lung cancer and non-small cell lung cancer, neuroblastoma/glioblastoma, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, liver cancer, melanoma, squamous cell carcinomas of the mouth, throat, larynx, and lung, colon cancer, cervical cancer,

cervical carcinoma, breast cancer, and epithelial cancer, renal cancer, genitourinary cancer, pulmonary cancer, esophageal carcinoma, head and neck carcinoma, large bowel cancer, hematopoietic cancers; testicular cancer; colon and rectal cancers, prostatic cancer, or pancreatic cancer.

[00174] Compounds disclosed herein may also be used for the treatment of precancer conditions such as cervical and anal dysplasias, other dysplasias, severe dysplasias, hyperplasias, atypical hyperplasias, and neoplasias.

[00175] Also disclosed are methods of reducing transplant rejection in a recipient by administering to the recipient a composition as disclosed herein, for example, a derivative, analogue, or metabolite of curcumin. Inflammation is associated with transplant rejection in a transplant recipient. In one example of transplant rejection, antibodies are formed against foreign antigens on the transplanted material. The transplantation can be, for example, organ transplantation, such as liver, kidney, skin, eyes, heart, or any other transplantable organ of the body or part thereof.

[00176] Transplantation immunology refers to an extensive sequence of events that occurs after an allograft or a xenograft is removed from a donor and then transplanted into a recipient. Tissue is damaged at both the graft and the transplantation sites. An inflammatory reaction follows immediately, as does activation of biochemical cascades. A series of specific and nonspecific cellular responses ensues as antigens are recognized. Antigen-independent causes of tissue damage (i.e., ischemia, hypothermia, reperfusion injury) are the result of mechanical trauma as well as disruption of the blood supply as the graft is harvested. In contrast, antigen-dependent causes of tissue damage involve immune-mediated damage.

[00177] Rejection is the consequence of the recipient's alloimmune response to the nonself antigens expressed by donor tissues. In hyperacute rejection, transplant subjects are serologically presensitized to alloantigens (i.e., graft antigens are recognized as nonself). Histologically, numerous polymorphonuclear leukocytes (PMNs) exist within the graft vasculature and are associated with widespread microthrombin formation and platelet accumulation. Little or no leukocyte infiltration occurs. Hyperacute rejection manifests within minutes to hours of graft implantation. Hyperacute rejection has

become relatively rare since the introduction of routine pretransplantation screening of graft recipients for antidonor antibodies.

[00178] In acute rejection, graft antigens are recognized by T cells; the resulting cytokine release eventually leads to tissue distortion, vascular insufficiency, and cell destruction. Histologically, leukocytes are present, dominated by equivalent numbers of macrophages and T cells within the interstitium. These processes can occur within 24 hours of transplantation and occur over a period of days to weeks.

[00179] hi chronic rejection, pathologic tissue remodeling results from peritransplant and posttransplant trauma. Cytokines and tissue growth factor induce smooth muscle cells to proliferate, to migrate, and to produce new matrix material. Interstitial fibroblasts are also induced to produce collagen. Histologically, progressive neointimal formation occurs within large and medium arteries and, to a lesser extent, within veins of the graft. Leukocyte infiltration usually is mild or even absent. All these result in reduced blood flow, with subsequent regional tissue ischemia, fibrosis, and cell death.

[00180] Transplant rejection may occur within 1-10 minutes of transplantation, or within 10 minutes to 1 hour of transplantation, or within 1 hour to 10 hours of transplantation, or within 10 hours to 24 hours of transplantation, within 24 hours to 48 hours of transplantation, within 48 hours to 1 month of transplantation, within 1 month to 1 year of transplantation, within 1 year to 5 years of transplantation, or even longer after transplantation.

[00181] As used throughout, by a "subject" is meant an individual. Thus, the "subject" can include domesticated animals, such as cats, dogs, etc., livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), laboratory animals (e.g., mouse, rabbit, rat, guinea pig, etc.) and birds. Preferably, the subject is a mammal such as a primate such as a monkey or chimpanzee, and, more preferably, a human.

1. Methods of using the compositions as research tools

[00182] The disclosed compositions can be used in a variety of ways as research tools. For example, the disclosed compositions, such as D68, D 12, and Dl 3 can be used to study the effects of ionizing radiation, and in particular the effects of ionizing

radiation in the onset of gastroinstestinal injury (GI) syndrome or bone marrow (BM) syndrome of acute radiation syndrome (ARS).

[00183] The disclosed compositions can be used as discussed herein as either reagents in micro arrays or as reagents to probe or analyze existing microarrays to identify genes or proteins critical to the formation of ARS, GI syndrome, or BM syndrome. The compositions can also be used in any known method of screening assays, related to chip/micro arrays. The compositions can also be used in any known way of using the computer readable embodiments of the disclosed compositions, for example, to study relatedness or to perform molecular modeling analysis related to the disclosed compositions.

G. Kits

[00184] Disclosed herein are kits that are drawn to reagents that can be used in practicing the methods disclosed herein. The kits can include any reagent or combination of reagent discussed herein or that would be understood to be required or beneficial in the practice of the disclosed methods. For example, the kits could include analogues, derivatives, or metabolites of curcumin, such as, for example D 12, D 13, or D68, to perform the methods disclosed herein. For example, disclosed is a kit for preventing GI syndrome or BM syndrome of ARS following exposure to IR.

H. Compositions with similar functions

[00185] It is understood that the compositions disclosed herein have certain functions, such as inhibiting COX-2. Disclosed herein are certain structural requirements for performing the disclosed functions, and it is understood that there are a variety of structures which can perform the same function which are related to the disclosed structures, and that these structures will ultimately achieve the same result, for example, preventing or delaying the onset of GI syndrome or BM syndrome or ARS.

I. Examples

[00186] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and

evaluated, and are intended to be purely exemplary and are not intended to limit the disclosure. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in 0 C or is at ambient temperature, and pressure is at or near atmospheric.

1. Example 1: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione mono-p-toluenesulfonylhydrazone

[00187] The concentrated hydrochloric acid (about 10 drops) was added to the solution of curcumin (3.68g, lOmmol) and p-toluenesulfonylhydrazide (2.23g, 12mmol) in 80 ml of ethanol. The mixture was stirred at refluxing for 6 hours. Then the TLC check indicated that there were still some starting material (curcumin) in the mixture. The p-toluenesulfonylhydrazide (0.37g, 2mmole) was added to the mixture and the mixture was refiuxed again for another 2 hours. The refluxing solution was monitored by the TLC check. The final solution was decolored with some charcoal and concentrated to 30ml. After overnight refridgeration, the yellow solid was filtered and recrystallized with methanol and ethyl acetate (5:1) to afford the title compound (3.38g), 63.06% yield, mp 256-8 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.51(2H, d, J=8 Hz), 7.15(6H, m), 6.98(4H, m), 6.81(2H, d, J=8.4 Hz), 6.72(2H, m), 3.85(6H, s, - OCH 3 X2), 2.32(3H, s, -CH 3 ). APCIMS: m/z 365.4, 364.3.

2. Example 2: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione monobenzenesulfonylhydrazone

[00188] The concentrated hydrochloric acid (about 10 drops) was added to the solution of curcumin (3.68g, lOmmol) and benzenesulfonylhydrazide (2.07g, 12mmol) in 80 ml of ethanol. The mixture was stirred at refluxing for 8 hours. The refluxing solution was monitered by the TLC check. The reaction solution was evaporated in vacuo and the residue was purified by silica gel column chromatography with the elute of n-heptane and ethyl acetate to give a light yellow powder (2.37g), 45.40% yield, mp 245-6 0 C. 1 R NMR (400 MHz, DMSO-d6)δ 9.99 (3H, s), 9.79(4H, s), 7.66(9H, m), 3.35(6H, s, -OCH 3 X2). APCIMS: m/z 365.2.

[00189] In accordance with the procedures used for the above two compounds, the following compounds were synthesized in this invention.

3. Example 3: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione monoisonicotinoylhydrazone

[00190] Yellow solid (68.38%), mp 263-4 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 8.85(2H, d, J=2.4 Hz), 8.03(2H, d, J=2.4 Hz), 6.98(12H, m), 3.82(6H, s, -OCH 3 X2). APCIMS: m/z 366.1, 365.2, 335.0.

4. Example 4: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione mononicotinoylhydrazone

[00191] Yellow solid (43.94%), mp 265-6°C. 1 H NMR (400 MHz, DMSO-d6)δ 7.19(6H, m), 6.99(4H, m), 6.98(2H, d, J=8 Hz), 6.83(2H, d, J=8 Hz), 6.78(2H, m), 3.86(6H, s, -OCH 3 X2). APCMS: m/z 367.3, 366.3, 365.2.

5. Example 5: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-hydroxybenzoyl monohydrazone

[00192] White solid (73.31%), mp >270°C. 1 H NMR (400 MHz, DMSO-d6)δ 10.08(5H, m), 7.78(6H, m), 6.83(6H, m), 3.33(6H, s, -OCH 3 X2). APCMS: m/z 367.3.

6. Example 6: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 2'-furoyl monohydrazone

[00193] Yellow solid (64.92%), mp 256-7°C. 1 B. NMR (400 MHz, DMSO-d6)δ 7.16(5H, m), 6.99(6H, m), 6.82(2H, d, J=8 Hz), 6.75(2H, m), 3.87(6H, s, -OCH 3 X2). APCIMS: m/z 365.10.

7. Example 7: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione thiophene-2'-carboxyI monohydrazone

[00194] Yellow solid (54.88%), mp 192-3 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 8.37(1H, dd, Hz, J 2 =I.2 Hz), 8.21(1H, dd, Ji=4.8 Hz, J 2 =0.8 Hz), 7.75(1H, d, J=16.4 Hz), 7.39(lH,d, J=16.4 Hz), 7.35(3H, m), 7.32(2H, d, J=7.2 Hz), 7.18(2H, m), 7.07(2H, d, J=7.2 Hz), 6.86(2H, t, J=8 Hz), 3.90( 3H, s, -OCH 3 ), 3.88(3H, s, -OCH 3 ). APCMS: m/z 365.10.

8. Example 8: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'H-[l]-benzopyrano[4,3-b]thiophene-2'-carboxyl monohydrazone

[00195] Yellow solid (89.77%), mp 212-3°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.43(1H, s, -OH), 9.38(1H, s, -OH), 8.22(1H, s), 7.76(1H, d, J=16.4 Hz), 7.64(1H, dd, Ji=7.2 Hz, J 2 =I.2 Hz), 7.36(5H, m), 7.19(2H,t, J=2.4 Hz), 7.08(5H, m), 6.87(2H, dd, Ji=8 Hz, J 2 =4.4 Hz), 5.40(2H, s, -CH 2 O-), 3.92 (3H, s, -OCH 3 ), 3.89(3H, s, -OCH 3 ).

9. Example 9: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-methyl-l',2',3'-thiadiazole-5'-carboxyI monohydrazone

[00196] Yellow solid (86.61%), mp 221-2 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.49(1H, s, -OH), 9.42(1H, s, -OH), 7.75(1H, d, J=16 Hz), 7.40(4H, m), 7.15(5H, m), 6.87(2H,t, J=8 Hz), 3.91(3H, s, -OCH 3 ), 3.89(3H, s, -OCH 3 ), 3.05(3H, s, -CH 3 ).

10. Example 10: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-tert-butylbenzoyl monohydrazone

[00197] Burgundy solid (59.04%), mp 266-7 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.46(1H, s, -OH), 9.38(1H, s, -OH), 7.35(1H, m), 7.31(2H, m), 7.21(3H, d, J=1.6 Hz), 7.01(5H, m), 6.96(1H, m), 6.88(2H, d, J=4.8 Hz), 6.86(1H, m), 3.87(6H, s, - OCH 3 X2), 1.38(9H, s).

11. Example 11: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-aminobenzoyI monohydrazone

[00198] Yellow solid (79.29%), mp 260-l°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.12(4H, bs, -OHX2, -NH 2 ), 7.23(1H, m), 7.19(4H, d, J=2.4 Hz), 7.01(1H, d, J=1.6 Hz), 6.99(3H, m), 6.95(1H, m), 6.83(3H, d, J=8.4 Hz), 6.79(1H, m), 3.87(6H, s, - OCH 3 X2).

12. Example 12: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione benzoyl monohydrazone

[00199] Yellow solid (69.75%), mp 258-9 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.29(1H, d, J=16.4Hz), 7.20(2H, d, J=1.6 Hz), 7.02(4H, m), 6.98(1H, d, J=16.8 Hz), 6.85(5H, m), 3.87(6H, s, -OCH 3 X2).

13. Example 13: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-methylbenzoyl monohydrazone

[00200] Yellow solid (59.60%), mp 262-4 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.25(2H, d, J=16.4 Hz), 7.20(2H, d, J=2.0 Hz), 7.02(4H, m), 6.97(2H, d, J=16.4 Hz), 6.83(4H, m), 3.87(6H, s, -OCH 3 X2), 2.54(3H, s, CH 3 ).

14. Example 14: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-nitrobenzoyl monohydrazone

[00201] Yellow solid (75.71%), mp 248-50°C. 1 H NMR (400 MHz, DMSO-d6)δ 8.40(2H, d, J=7.2 Hz), 8.15(2H, d, J=7.2Hz), 7.67(1H, d, J=I 6.4 Hz), 7.34(3H, m), 7.17(2H, d, J=16.4 Hz), 6.88(6H, m), 3.87(6H, s, -OCH 3 X2).

15. Example 15: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 3'-hydroxybenzoyl monohydrazone

[00202] Yellow solid (66.33%), mp 246-7 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.26(2H, d, J=I 6.4 Hz), 7.20(3H, m), 7.02(3H, m), 6.97(2H, d, J=I 6.4 Hz), 6.84(5H, m), 3.87(6H, s, -OCH 3 X2).

16. Example 16: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 2'-chlorobenzoyl monohydrazone

[00203] Yellow solid (48.61%), nip 248-9 0 C. 1 R NMR (400 MHz, DMSO-d6)δ 7.24(2H, d, J=16.4 Hz), 7.20(3H, m), 7.01(3H, m), 6.97(2H, d, J=16.4 Hz), 6.83(5H, m), 3.87(6H, s, -OCH 3 X2).

17. Example 17: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-chlorobenzoyl monohydrazone

[00204] Yellow solid (60.13%), mp 256-7 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.97(2H, d, J=7.2 Hz), 7.66(2H, d, J=7.2 Hz), 7.20(3H, m), 7.18(2H, d, J=8.4 Hz), 7.00(3H, m), 6.83(2H, d, J=8 Hz), 6.77(2H, m), 3.87(6H, s, -OCH 3 X2).

18. Example 18: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 2'-nitrobenzoyl monohydrazone

[00205] Yellow solid (82.86%), mp 208-9°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.47(1H, s, -OH), 9.32(1H, s, -OH), 8.33(1H, d, J=7.6 Hz), 8.2O(1H, d, J=7.6Hz), 7.92(2H, m), 7.81(1H, d, J=16.4 Hz), 7.42(2H, m), 7.25(3H, m), 7.11(1H, dd, Jl=8.4Hz, J2=1.6Hz), 6.97(1H, dd, Jl=8.4Hz, J2=1.6Hz), 6.89(1H, d, J=8 Hz), 6.82(1H, d, J=16.4 Hz), 6.79(1H, d, J=8 Hz), 3.89(3H, s, -OCH 3 ), 3.83(3H, s, -OCH 3 ).

19. Example 19: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 2'-hydroxy-4'-methoxybenzoyl monohydrazone

[00206] Yellow solid (51.88%), mp 261°C (decomposition). 1 H NMR (400 MHz, DMSO-d6)δ 7.28(2H, d, J=16.4 Hz), 7.20(3H, m), 6.99(5H, m), 6.85(4H, m), 3.87(6H, s, -OCH 3 X2).

20. Example 20: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione 4'-trifluoromethylitrobenzoyl monohydrazone

[00207] yellow solid (51.10%), mp 267-8 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.25(2H, d, J=16.4 Hz), 7.20(4H, m), 7.01(3H, m), 6.98(2H, d, J=16.4 Hz), 6.84(2H, d, J=8 Hz), 6.82(2H, d, J=7.2 Hz), 3.87(6H, s, -OCH 3 X2).

21. Example 21: Preparation of l,7-bis-(4-hydroxy-3-methoxyphenyl)-l,6- heptadiene-3,5-dione L-tyrosine monohydrazone

[00208] Yellow solid (62.30%), mp 253-4 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.25(2H, d, J=16.4 Hz), 7.20(3H, m), 7.01(4H, m), 6.98(2H, d, J=16.4 Hz), 6.84(2H, d, J=8 Hz), 6.82(2H, d, J=9.2 Hz), 3.87(6H, s, -OCH 3 X2).

22. Example 22: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- methylpyrazole

[00209] Curcumin (3.68g, lOmmol) and methylhydrazine (0.88g, 20mmol) were added to absolute ethanol (80ml) containing some concentrated hydrochloric acid (0.4ml). The reaction mixture was stirred at room temperature for 1 hour, then heated under refluxing for 20 hours. The mixture was allowed to cool to room temperature and then concentrated using a rotary evaporator under reduced pressure. The residue was purified by silica gel column chromatography with the elute of combination of n- heptane and ethyl acetate to give a light yellow powder (0.4Og), 10.58% yield, mp 228-3O 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.30 (IH, s, -OH), 9.26(1H, s, -OH), 7.28(2H, d, J=16 Hz), 7.06(6H, m), 6.75(4H, m), 3.90(3H, s, -OCH 3 ), 3.88(3H, s, - OCH 3 ), 3.78(3H, s, -CH 3 ). APCIMS: m/z 379.3, 378.3, 377.2.

23. Example 23: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyi)-ethylene-l- (2-hydroxyethyl)pyrazole

[00210] Light yellow powder (17.89%), mp 225-7°C. 1 H NMR (400 MHz, DMSO- d6)δ 9.26 (IH, s, -OH), 9.14(1H, s, -OH), 7.42(1H, d, J=8.4 Hz), 7.24(2H, d, J=16 Hz), 7.05(5H, m), 6.80(3H, m), 4.93(1H, bs, -OH), 4.29(2H, m, -CH 2 -), 3.88(3H, s, - OCH 3 ), 3.86(3H, s, -OCH 3 ), 3.78(3H, m, -CH 3 ). APCIMS: m/z 410.2, 409.3.

24. Example 24: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2-hydroxycarbonylphenyl)pyrazole

[00211] Yellow powder (46.69%), mp 203-5°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.29(1H, s, -OH), 9.18(1H, s, -OH), 7.94(1H, dd, Ji=7.6 Hz, J 2 -1.6 Hz), 7.75(1H, t, J=7.2 Hz), 7.66(1H, t, J=7.6 Hz), 7.54(1H, d, J=7.6 Hz), 7.04(1OH, m), 6.53(1H, d, J =16 Hz), 3.87(3H, s, -OCH 3 ), 3.79(3H, s, -OCH 3 ). APCIMS: m/z 485.4.

25. Example 25: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyi)-ethylene-l- (2-ethoxycarbonylphenyl)pyrazole

[00212] Yellow powder (29.30%), mp 81-2 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.3O(1H, s, -OH), 9.19(1H, s, -OH), 7.91(1H, dd, J 1 =S Hz, J 2 =I.6 Hz), 7.81(1H, t, J=7.6 Hz), 7.67(1H, t, J=7.6 Hz), 7.61(1H, d, J=8 Hz), 6.96(1OH, m), 6.55(1H, d, J =16.4 Hz), 4.02(2H, q, J=3.2 Hz), 3.87(3H, s, -OCH 3 ), 3.80(3H, s, -OCH 3 ), 1.01(3H, t, J=3.2 Hz). APCMS: m/z 515.5, 514.5, 513.5.

26. Example 26: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethyIene-l- (imidazolin-2-yl)pyrazole

[00213] Yellow powder (90.74%), mp >260°C. 1 H NMR (400 MHz, DMSO-d6)δ 1O.23(1H, bs, -NH), 9.53(1H, bs, -OH), 9.43(1H, bs, -OH), 7.40(3H, m), 7.28(1H, d, J=I.2 Hz), 7.24(1H, m), 7.08(4H, m), 6.87(2H, t, J =8 Hz), 4.02(3H, s, -OCH 3 ), 3.88(3H, s, -OCH 3 ). APCIMS: m/z 435.4, 434.4, 433.3.

27. Example 27: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2-nitrophenyl) pyrazole

[00214] Yellow powder (39.18%), mp 197-8°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.35(1H, s, -OH), 9.23(1H, s, -OH), 8.18(1H, d, J=8 Hz), 7.95(1H, m), 7.81(2H, m), 7.20(5H, m), 6.97(3H, m), 6.80(2H, dd, Hz), 6.68(1H, d, J=I 6 Hz), 3.87(3H, s, -OCH 3 ), 3.81(3H, s, -OCH 3 ). APCIMS: m/z 486.10.

28. Example 28: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (4-nitrophenyl) pyrazole

[00215] Yellow powder (75.46%), mp 218-9 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.36(1H, s, -OH), 9.26(1H, s, -OH), 8.46(2H, d, J=9.2 Hz), 7.88(2H, d, J=9.2 Hz), 7.26(5H, m), 7.05(4H, m), 6.82(2H, dd, J α =8 Hz, J 2 =I.6 Hz), 3.88(3H, s, -OCH 3 ), 3.84(3H, s, -OCH 3 ). APCMS: m/z 486.10.

29. Example 29: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (4-methylphenyl) pyrazole

[00216] White powder (62.36%), mp 116-7 0 C. 1 U NMR (400 MHz, DMSO-d6)δ 9.3O(1H, s, -OH), 9.19(1H, s, -OH), 7.43(4H, q, J,=19.2 Hz, J 2 =8.4 Hz), 7.20(3H, m), 7.08(3H, m), 6.96(2H, m), 6.81(2H, dd, Ji=8 Hz, J 2 =2 Hz), 6.76(1H, m), 3.87(3H, s, - OCH 3 ), 3.82(3H, s, -OCH 3 ). APCIMS: m/z 455.10.

30. Example 30: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2-methylphenyl) pyrazole

[00217] White powder (37.00%), mp 96-8 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.35(1H, s, -OH), 9.27(1H, s, -OH), 7.48(4H, m), 7.36(1H, m), 7.12(2H, m), 7.05(4H, m), 6.72(3H, m), 6.38(2H, dd, Ji=8.4 Hz, J 2 =2 Hz), 3.86(3H, s, -OCH 3 ), 3.78(3H, s, - OCH 3 ). APCMS: m/z 455.10.

31. Example 31: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2-pyridyl) pyrazole

[00218] Yellow powder (52.83%), mp >260°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.58(1H, s, -OH), 9.46(1H, s, -OH), 8.66(2H, m), 8.39(1H, d, J=8 Hz), 7.81(1H, t, J=8.4 Hz), 7.43(1H, d, J=16.4 Hz), 7.32(1H, d, J=1.6 Hz), 7.10(5H, m), 6.85(2H, dd, Ji=8 Hz, J 2 =2.4 Hz), 6.59(1H, dd, Ji=8 Hz, J 2 =2.4 Hz), 6.28(1H, t, J=6 Hz), 3.88(3H, s, -OCH 3 ), 3.77(3H, s, -OCH 3 ). APCIMS: m/z 440.35.

32. Example 32: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2,4-dinitrophenyl) pyrazole

[00219] Yellow powder (52.83%), mp 205-6°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.38(1H, s, -OH), 9.26(1H, s, -OH), 8.96(1H, d, J=2.4 Hz), 8.7O(1H, dd, Jl=8.8 Hz, J2=2.4 Hz), 8.06(1H, d, J=8.4 Hz), 7.22(5H, m), 7.04(2H, m), 6.98(1H, d, J=I 6.4 Hz), 6.82(3H, m), 3.87(3H, s, -OCH 3 ), 3.82(3H, s, -OCH 3 ). APCIMS: m/z 530.95.

33. Example 33: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2-trifluoromethylphenyI) pyrazole

[00220] White powder (53.74%), mp 212-3°C. 1 B. NMR (400 MHz, DMSO-d6)δ 9.29(1H, s, -OH), 9.19(1H, s, -OH), 8.03(1H, d, J=7.6 Hz), 7.93(1H, t, J=7.6 Hz), 7.86(1H, t, J=8.6 Hz), 7.63(1H, d, J=7.6 Hz), 7.24(1H, d, J=I.2 Hz), 7.17(1H, d, J=16.4 Hz), 7.14(2H, m), 7.01(3H, m), 6.85(1H, dd, Jl=8 Hz, J2=1.6 Hz), 6.81(1H, d, J=8 Hz), 6.76(1H, d, J=8.4 Hz), 6.37(1H, d, J=16 Hz), 3.87(3H, s, -OCH 3 ), 3.78(3H, s, -OCH 3 ). APCMS: m/z 509.00.

34. Example 34: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (2,6-dichloro-4-trifiuoromethylphenyl) pyrazole

[00221] White powder (67.23%), mp 163-5 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 9.33(1H, s, -OH), 9.22(1H, s, -OH), 8.30(2H, s), 7.26(1H, d, J=I.6 Hz), 7.19(1H, d, J=16.4 Hz), 7.18(1H, d, J=16.4 Hz), 7.14(1H, m), 7.03(4H, m), 6.82(1H, d, J=8 Hz), 6.78(1H, d, J=8 Hz), 6.47(1H, d, J=16.4 Hz), 3.87(3H, s, -OCH 3 ), 3.81(3H, s, -OCH 3 ). APCMS: m/z 576.90.

35. Example 35: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (4-trifluoromethylpyrimidin-2-yl) pyrazole

[00222] Yellow powder (23.46%), mp 194-5°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.37(1H, s, -OH), 9.28(1H, s, -OH), 9.33(1H, d, J=7.2 Hz, 5'-H), 8.03(1H, d, J=7.2 Hz, 6'-H), 7.8O(1H, d, J=16.4 Hz), 7.30(4H, m), 7.18(1H, m), 7.16(1H, d, J=I 6.4 Hz), 6.84(2H, dd, Jl=8 Hz, J2=3.6 Hz), 3.89(3H, s, -OCH 3 ), 3.85(3H, s, -OCH 3 ). APCMS: m/z 510.85.

36. Example 36: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (benzothiazol-2-yl) pyrazole

[00223] Yellow powder (75.45%), mp 209-210.5 0 C. 1 B. NMR (400 MHz, DMSO- d6)δ 9.47(1H, s, -OH), 9.33(1H, s, -OH), 8.27(1H, d, J=16.4 Hz), 8.12(1H, d, J=8 Hz), 8.02(1H, d, J=8 Hz), 7.57(1H, m), 7.47(2H, m), 7.40(2H, m), 7.34(2H, d, J=16.4 Hz), 7.24(1H, d, J=I.6 Hz), 7.14(2H, m), 7.07(1H, dd, Jl=8 Hz, J2=1.6Hz), 6.9O(1H, d, J=8 Hz), 6.84(1H, d, J=8 Hz), 3.93(3H, s, -OCH 3 ), 3.90(3H, s, -OCH 3 ). APCIMS: m/z 510.85.

37. Example 37: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (3.5-difluorophenyl) pyrazole

[00224] Yellow powder (62.74%), mp 105-6°C. 1 H NMR (400 MHz, DMSO-d6)δ 7.38(3H, m), 7.25(2H, m), 7.19(2H, m), 7.12(1H, m), 7.04(3H, m), 6.93(1H, d, J=16.4 Hz), 6.82(2H, dd, Jl=8 Hz, J2=2.8 Hz), 3.88(3H, s, -OCH 3 ), 3.83(3H, s, -OCH 3 ). APCIMS: m/z 476.95.

38. Example 38: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-etliylene-l- (4-trifluoromethoxyphenyl) pyrazole

[00225] Yellow powder (21.80%), mp 96-8 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.73(2H, d, J=8.4 Hz), 7.71(2H, d, J=8.4 Hz), 7.24(1H, m), 7.21(1H, d, J=7.2 Hz), 7.19(1H, d, j=16.4 Hz), 7.13(2H, d, J=16.4 Hz), 7.01(3H, m), 6.82(3H, m), 3.87(3H, s, -OCH 3 ), 3.82(3H, s, -OCH 3 ). APCIMS: m/z 524.90.

39. Example 39: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- benzyl pyrazole

[00226] Yellow powder (24.01%), nip 137-8°C. 1 H NMR (400 MHz, DMSO-d6)δ 9.27(1H, s, -OH), 9.14(1H, s, -OH), 7.37(2H, t, J=7.6 Hz), 7.25(5H, m), 7.12(1H, d, J=5.6 Hz), 7.08(1H, d, J=16.4 Hz), 7.00(4H, m), 6.89(1H, m), 6.80(2H, dd, Jl=8 Hz, J2=5.6 Hz), 5.55(2H, s, -CH2-), 3.86(3H, s, -OCH 3 ), 3.85(3H, s, -OCH 3 ). APCMS: m/z 455.00.

40. Example 40: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l- (3-trifiuoromethylphenyl) pyrazole

[00227] Yellow powder (25.90%), mp 135-6 0 C. 1 H NMR (400 MHz, DMSO-d6)δ 7.86(4H, m), 7.27(1H, m), 7.24(1H, d, J=16.4 Hz), 7.22(1H, d, J=I 6.4 Hz), 7.15(2H, m), 7.09(1H, d, J=16.4 Hz), 7.01(2H, m), 6.89(1H, d, J=16 Hz), 6.82(2H, d, J=8.4 Hz), 3.88(3H, s, -OCH 3 ), 3.81(3H, s, -OCH 3 ). APCIMS: m/z 509.05.

41. Example 41: Preparation of 3,5-bis-(4-hydroxy-3-methoxyphenyi)-ethylene-l- (4-iodophenyl) pyrazole

[00228] Yellow powder (24.53%), mp 148-9°C. 1 H NMR (400 MHz, DMSO-d6)δ 7.94(2H, d, J=7.6 Hz), 7.39(2H, d, J=8.4 Hz), 7.20(4H, m), 7.08(2H, d, J=7.2 Hz), 7.02(2H, m), 6.84(2H, d, J=16.4 Hz), 6.81(1H, d, J=8 Hz), 3.87QH, s, -OCH 3 ), 3.83(3H, s, -OCH 3 ). APCIMS: m/z 566.90.

42. Example 42: Preparation of amorphous crystal form of 3,5-bis-(4-hydroxy-3- methoxyphenyl)-ethylene-l -(4-methyIphenyl) pyrazole

[00229] The yellow gum of 3,5-bis-(4-hydroxy-3-methoxyphenyl)-ethylene-l-(4- methylphenyl) pyrazole (1.Og) from column chromatography was dissolved in methanol (30ml). One piece of boiling chip was added to the mixture and the mixture was heated under refluxing for 1 hour. When the mixture was then allowed to cool to room temperature, a white crystal formed. About 12 hours later, at room temperature, the crystal was filtered and dried. The crystal weighed 0.904g (90.4% yield) and had a m.p. 116-7°C.

43. Example 43: Characterization and synthesis of anti-oxidants derived from natural compounds

[00230] The starting compound library for screening the anti-radiation /oxidants is the purified compound from naturally existing plants. This idea is based on the fact that the majority of anti-cancer drug is originally discovered from natural products (such as Taxol, Mitomysin C, Adrimycin, etc), and therefore, mother "nature" should give a birth of anti-radiation compounds since the earth might experience some period of high radiation exposure and many livings have adapted to the irradiated environment via producing their own anti-oxidants. For example, some plants that survive from high dose IR environment (such as plants from very high attitude

mountain exposed to extensive UV, or plants growing in the high level of underground radiation area) might contain some components that are resistant to radiation. More than 20 compounds were collected that were purified (>95%) from different plants grown in different areas and attitudes, and then screen for their antioxidant activity.

[00231] The easy and fast in vitro method to characterize the anti-oxidant property was to use the electronic chemical detector (model CHI830B from CH Instruments Lie, Austin, TX). It is a voltammeter analysis using neutral buffer with Ag/ AgCl as reference electrode and glassy carbon as working electrode, which can sensitively determine if the agent releases electronics under un-trigged conditions (ready to against free radices) or under voltage-trigged condition (anti-free radices under certain circumstance). The background redox potential for the vehicle alone is shown in Fig IA. After screening more than 20 purified compounds from different plants with electronic chemical detector, it was found that most of them had a low potential of redox as represented in Fig IB (which required the voltage >0.6 V to release electronics). However, curcumin, a spice food namely curcumin, required a low voltage (about 0.2 V) to generate redox (Fig 1C). It was decided to use curcumin as a parental material to chemically synthesize new compounds that possess high redox potential.

[00232] More than 100 derivatives have been obtained and screened. Among them, the redox potential of some new compounds (such as D 12, D13 and D68) was improved as evidenced in voltametric graph that under a very low voltage, the compounds easily release electronics which can transport to surrounding redox and stabilize the microenvironment (Fig ID and E). The redox potential of the synthetic compounds was around -0.028V to 0.064V (see Table 1), which was much lower than the parental compound (0.26V). It is worth to mention that ascorbic acid (Vitamin C) a well-known anti-oxidant drug had a redox potential of 0.42 V in the measurement system (Fig IF). The agents have a much better anti-oxidant effect than Vitamin C. The first line anti-radiation drug, Amifostine in saline, released electronics at 0.084 V at this in vitro assay system with present oxygen. Amifostine is activated in vivo with enzymatic reaction. At least, at the same assay condition, the compounds disclosed herein have a similar redox potential to Amifostine, and higher than Vitamin C.

Table 1

[00233] Interestingly, if new compound was mixed with low potential parental compound, there was a high peak of released electronics under 0 V (Fig IG), indicating that there was an electronic transportation between the lower one to higher one, which provides a molecular mechanism for combined use of the agent with other low potential agents, such as vitamin C to enhance the regeneration of anti-oxidant action.

[00234] With the promising anti-oxidant potential, a few grams of D 12, D13 and D68 were synthesized, which had a purity of 94-98% as determined by MS and HPLC (Fig IH), allowing for in vivo testing.

44. Example 44: In vivo mitigation effect of the synthetic agents on GI syndrome of acute radiation syndrome (ARS) a) Capability of rescuing mice from death of GI syndrome of ARS

[00235] It is well-known that the BM is much more sensitive to IR than GI. The LD 5 ( V3O for BALB/c (6-8 weeks old male, 6-8 mice/group) was about 5.5 Gy given in a TBI (total body irradiation) fashion. The irradiation was delivered with a Shepherd Mark-I unit operating 137 Cs sources. The dose rate was 1.84 Gy/min. When IR dose was raised to 7-7.5 Gy TBI, 100% mice died within 5-12 days. However, at 7-7.5 Gy TBI dose, lethal GI damage was undetectable under microscope. In most cases, it was found that mice died of infection and bleeding (especially a lethal brain bleeding), indicating that under 7.5 Gy the death is mainly due to BM failure. Obviously, the TBI is not a good model to study agents that mitigate the lethal GI IR damage. To deliver mouse with an IR dose that is high enough to cause a lethal GI damage without BM

failure, a sub-TBI model was created in which one leg of mouse was shielded and out of the IR field, hi this system, 8.5 Gy were delivered to BALB/c mice without any death. When sub-TBI IR dose was increased to 9 Gy, around half of ER. mice died, and at 10.5 Gy, 66-100% mice died within 5-7 days with devastating gut damage observable under microscope. It seems that in the GI (sub-TBI) model, the LD 50 / 7 for BALB/c mice used was around 8.9-9.1 Gy. Once GI model was developed, the mitigation effect of the agents was tested on GI syndrome. All the agents were given orally and started 5-10 minutes after IR and continued to given one dose per day for 3- 5 days. The agent doses ranged from 70 to 150 mg/kg. It was observed that even the mice that were obtained from the same MH vendor, the response to the same ER dose in the control mice slightly differ from batch to batch and from time to time. Therefore each animal experiment had its own vehicle mock treatment group as control. The parental material curcumin was used at different doses in the model system, but there is not improvement of survival (a similar death curve as control). This may be due to the relatively high radiation dose used in the system and the low mitigation potential of the parental agent. However, the results of the synthetic agents were very promising. When BALB/c mice sub-TBI with 10.5 Gy (Tasking towards MDF of 1.2), 67% of control mice died within 7 days (LD 50/? ) while 83% mice treated with D12 survived up to now (4 months, Fig 2A). Similar mitigation results were obtained with D13, in which 50% of control mice died within 7 day and all of the rest died withinl3 days, 50% of Dl 3 treated mice survived up to now (4 months, Fig 2B). Recently, D68, which had a better mitigation effect, was obtained. When the sub-TBI dose was raised to 12 Gy, 100% untreated mice died of GI syndrome while 43% of mice treated with D68 survived (up to now, 3.5 months, Fig 2C). This gave a MDF of 1.33. The Kaplan Meier analysis indicated that all the differences between the vehicle alone and the agent treatment groups were statistically significant (P<0.05). All the data strongly indicate that the agents given after GI lethal dose exert their mitigation effect.

b) Capability of rescuing mice from death of GI syndrome of ARS in different mouse models:

[00236] To determine if the agent works in mice with different genome backgrounds, D68 (150 mg/kg) was given to C57BL/6 mice 10 min after they were

exposed to 13 Gy sub-TBI. While 33% of untreated mice survived, 83% of mice treated with D68 survived (Fig 3 A). When the IR dose increased to 15 Gy, all the control mice died within 5 days, while 33% of D68 treated mice survived up to now (1.5 month, Fig 3 B). The third strain of mouse, C3H/NeH was also used. The results showed that the D68 rescued mice from death caused by sub-TBI, while untreated mice died. The data from three strains of mice were consistent with each other and statistically significant (P<0.05), indicating that the D68 is indeed a radio-mitigator and is likely to work in different genome backgrounds.

c) Capability of rescuing mice from death of BM (bone marrow) syndrome of ARS

[00237] The total body exposure to radiation is the most likely scenario in radiation exposure events, since the enormous IR energy will penetrate all barriers to some extent. Therefore, the ability of the agents to prolong the life of mice after their total body irradiation (TBI) to a 100% lethal IR dose (LDioozio-3o, a death due to BM failure) was investigated. The agents that rescued host from GI death also prolong the life from BM and other tissue injury, when working as an anti-oxidant. To test this, the TBI in BALB/c mouse model was used. As indicated before, the LD 5 o/ 3 o for BALB/c mouse was about 5.5 Gy. The cutoff for MDF >1.2 was 6.0 Gy. The test was carried out with a "high bar", in which mice were given 7 Gy TBI (LD 1OO z 10-3O ), and then the agents were given orally 5-10 min after IR and for 3-5 days thereafter. The results were quite promising, although the days of survival of controls varied with experiments (due to the animal batch variations, each batch has its control). At the 7.0 Gy TBI, when the control mice all dead on day 7, 83% of mice that treated with D12 (70 mg/kg) survived to day 11 (Fig 4A), a 57% increase of prolong survival time. Similar result was obtained with D13 (120 mg/kg, Fig 4B). Amazingly, D68 (120 mg/kg) rescued mice from death after 7Gy TBI (Fig 4C, up to now 4 months). When TBI IR dose was raised to 7.5 Gy, the LD 50 for control was on day 6, while the LD 50 for D68 treated mice was on day 10 (Fig 4D). hi most cases, the prolonged survival time was >50%. Again, all the difference between vehicle alone and the agent treatment groups were statistically significant (P<0.05). Since the LD 5O / 3 o of BALB/c mice in the study was 5.5 Gy, the rescuing mice at 7.0Gy indicated a DMF of D68 to be 1.27. The data also indicates that the TBI at LD 1OO Zi 0-30 knocks out all the BM stem

cells, resulting in host highly susceptible to infection and bleeding, a complicate clinical situation which has to been treated with multiple approaches. The prolong of life by the agents provides the time for other treatments to exert their effect and for the survival cells to regenerate.

[00238] Importantly, when the agent was delivered much later after IR exposure, it was found that even given 8 or 24 hr after high dose 7.0 Gy TBI, Dl 3 could still prolong the life of majority of mice for more than 2 days.

[00239] To determine which one of the agents had better potential to be developed as new radioprotector, side-by-side comparison studies were perforemed. The results from 12 Gy sub-TBI (LD 10 <w) model indicated that D68 had a better effect than D 12 and D13 (Fig 5), therefore, the focus was shifted to D68.

45. Example 45: Direct effect of D68 on GI tract a) Enhanced proliferation of crypts in small intestine [00240] Due to its physiological function and hostile micro-environment, the epithelium lying on the small intestine has a rapid turnover, approximately 3-5 days. The stem cells in the bottom of villi actively divide, especially when IR causes a serious damage that triggers a very active process of repair, manifesting as a strong staining of hemotoxilin in the nuclear-enriched crypts.

[00241] To determine the effect of D68 on the crypt regeneration, mice exposed to 10.5 Gy sub-TBI were randomly divided into two group and orally given either NS alone or D68 (120 mg/kg) 10 minutes after IR and thereafter once a day for 2 days. At 80 hr post-PR, the BrdU (120 mg/kg) was i.p. injected into each mouse and 4 hours later (about 3.5 days post-IR), the mice were sacrificed and the duodenal, jejunal and ileal segments (3-4 pieces/segment) were harvested, fixed in formalin, processed in paraffin blocks, cut in transverse sections of the full segment circumference (5 μm thick) and stained with H&E or anti-BrdU following the standard immunochemistry procedure with antigen retrieve. The proliferating crypts were defined as containing 10 or more adjacent chromophilic non-Paneth cells and a lumen. The circumference of a transverse cross-section of the intestine was used as a unit. The number of proliferating crypts was counted in each circumference. At least 10 circumferences

were scored per mouse and 4-5 mice were used to generate each data point. The results (Fig 6) showed that compared with the normal, the numbers of proliferating crypts in all the segments of mice exposed to 10.5 Gy treated with vehicle alone were decreased dramatically (around 110-130 vs 10-15). However, when treated with the agents, the numbers of proliferating crypts in jejunum (Fig 6A) were increased with statistic significance (P<0.05). Similar tendency was detected with BrdU staining (Fig 6B, P<0.05). The results from two methods were consistent to each other, again, supporting that the agents enhancement of the proliferating GI crypts.

[00242] When IR dose was raised to 12 Gy, the proliferating crypts in mice treated with vehicle alone dropped significantly as compared to 10.5Gy counterparts (around 15 vs 4), while there was a high numbers of proliferating crypts in duodenum, jejunum and ileum in agent treated group (Fig 6C, P<0.05). Again, similar result was obtained with BrdU staining method (Fig 6D, P<0.05).

[00243] The villi length of duodenum, jejunum and ileum was measured using Imaging Pro program, the data showed that the 16 Gy sub-TBI mice had a reduced villi length compared to the normal mice, however, treatment with D68 was able to reduce the villi shorting of duodenum, jejunum (P<0.05, Fig 6E), another evidence of D68 directly act on GI tract.

[00244] Importantly, the numbers of proliferating crypts were consistent with the survival data. The increased crypts were associated with an enhanced survival rate. AU these results indicate that the agents directly act on GI tract.

[00245] Interestingly, the side-by-side comparison with Amifostine, a current first line radioprotector used as a positive control given via iv at 200mg/kg once 30 min before IR, the results showed that Amifostine had a higher jejunum crypts, but a similar number of crypts in duodenum and ileum as D68 (Fig 6F). The Amifostine used after IR had no effect on promoting crypts, indicating that it is not a GI mitigation agent.

[00246] hi addition, the effect of functional peptide of fibroblast growth factor 2 (FGF2, a known mitogenic biological factor for all epithelium including stem cells) on crypts was compared with D68. The results showed that the FGF2-P had a similar or

slightly better stimulatory effect compared to D68 (Fig 6F). Since these two agents have different action mechanisms (D68 is an anti-oxidant and FGF2-P is a mitogen), they can be used in combination to enhance restoration of crypts, a most critical index for GI regeneration after IR injury. Taken together, although the agents did not bring the crypts to normal value, the partially increased crypts enabled the recovery with time, leading to a long-term survival.

b) D68 partially restores the GI physiological function of mice with GI syndrome of deadly ARS

[00247] Besides of restoring the GI stem cell proliferation (crypt formation), other indices representing the GI function were also examined for the following reasons and methods: 1) bloody stool: the IR-induced necrosis/apoptosis of GI epithelium and endothelium impairs the integrity of GI, leading to bloody stool which could be detected by hemoccult analysis with a commercial kit (BD Cat# 64151) at day 3 post- IR; 2) stool form: the GI functions of enzyme digestion, absorption of nutrition and water and the regular movement of intestine muscle results in a formation of normal stool. The IR damaged GI has a loose stool or diarrhea, which were pictured at day 3.5 post-IR; 3) endotoximia: the lost of normal GI barrier and function results in the overgrowth of GI bacterial and their penetration into circulation causing sepsis, a major cause of GI death of ARS. The level of plasma endotoxin on 3.5 days post-IR was measured with a commercialized kit that utilized tachypleus amebocyte lysate to sensitively quantitate endotoxin even <0.01 EU/ml; and 4) body weight (BW): the loss of nutrition and water due to GI IR-damage reduces the body weight. The survivor can gradually re-gain their body weights, reflecting the restoring GI function.

[00248] The results are summarized as follows: The stool hemoccult score differed among the groups (Fig 7A). The higher score (more bloody stool) was detected in mice receiving 12 Gy sub-TBI with vehicle mock treatment as compared to normal mice. The irradiated mice that treated with D68 and Amifostine (positive control) had a reduced hemoccult score in their stool (P<0.05), indicating that D68 is capable to reduce the GI bleeding induced by IR. Similar results were obtained from second strain of mice.

[00249] A more serious loose (diarrhea) and bloody stool was observed in vehicle group than that in D68 group (Fig 7B and C). Also, the plasma endotoxin level of BALB/c mice exposed to 10.5 Gy in vehicle treated groups was elevated significantly compared to normal mice (mean 0.45 v.s. 3.32), while the IR mice treated with D68 had a reduced endotoxin (mean 2.12, Fig 7D). The Amifostine (as positive control) that was delivered via i.v. injection 30 min before 10.5 Gy IR resulted in a relatively low level of plasma endotoxin (mean 0.93), suggesting that it protected the integrity of gut and prevented the accumulation of bacterial and their entry into blood. However, it is well-known that it must be used before IR, even 10 min post-IR delivery resulted in a high endotoxin level (mean 2.87). Similar results were obtained in C57BL/6 mice that had exposed to up to 16 Gy sub-TBI, a lethal dose even that the BM was spared (Fig 7E). The consistent data from two strains of mouse strongly suggest that the D68 does improve the GI barrier and reduce the endotoximia, which counts for its effect on rescuing the mice from GI death.

[00250] The follow-up observation of mouse body weight (BW) indicated that in the first 6 days after 10.5Gy sub-TBI, the injury was overweighing the repair, and all mice had a similar BW loss up to 1/3 of original BW. However, if the mice did not die on 5-7 days due to the individual resistance to IR (33% in control) or the treatment with D68 (83%), their BW gradually re-gained. The mice that treated with D68 had a relatively quick re-gaining compared to that of a few survivors from controls, although BW finally reached similar level (Fig 7F). BW does not predict the death or not, but its regain indicates a good recovery.

[00251] Above data strongly indicate that the D68 is capable of partially restoring IR-damaged GI function, which accounts for why the D68 rescued the mice from GI syndrome of ARS due to the high dose radiation.

c) D68 partially restores the GI endocrine and exocrine ability in mice with deadly GI syndrome of ARS

[00252] The main physical function of GI tract is to absolve the nutrition. The tight regulation of this process is relied on the production of hormone and enzymes at right time with right amount.

[00253] In the endocrine side, the two critical hormones produced by epithelial cells lining on small intestine are 1) secretin, that stimulates secretion of a bicarbonate-rich fluids from the pancreas and liver; and 2) cholecystokinin (CCK), that stimulates secretion of pancreatic enzymes and bile. Another a critical hormone, insulin produced by pancreatic β cell is, controls the level of blood glucose in the normal condition. Due its anatomy position, the pancreas is likely to exposed to radiation when GI tract is irradiated. In the exocrine side, one of the major enzymes to digest food is amylase that is secreted by epithelial cells along the GI system, including pancreas. All these endocrine and exocrine molecules are affected when the GI tract is injured by radiation.

[00254] Considering the blood sample is the most feasible sample that can be easily collected from human, ELISA kits were used for most of above biomarkers, since ELISA kits are sensitive enough to detect any settle change in plasma. Currently, most of commercially available kits are for human use. Whether they can be used for mouse sample depends on the molecular homology between the human and mouse. The protein sequence of cholecystokinin (CCK) is 100% conserved among all the species, therefore, the kit (Peninsula Laboratories, cat# S- 1205) for human can be used for this purpose. For secretin sequence (HSDGTFTSELSRLQDSARLQRLLQGLV (SEQ ID NO: I)), there is one amino acid change (M vs. T at position 5). The kit for human secretin (Peninsula Laboratories, cat# S- 1229) has been tested and it recognizes mouse form. The mouse insulin kit is available in the market (Linco Inc. Cat# EZRMI- 13 K). The EnzChek UHr Amylase Assay kit (Molecular Probes, Cat # E33651) can be used for all species, since it measures for the enzymatic activity. The blood glucose level is measured with Sigma kits based on enzymatic activity (Cat# GAGO-20 and GAHK-20), which is universal to all species too.

[00255] Tasked with all the kits, the alteration in plasma of mice that had exposed to 10.5 Gy sub-TBI and thereafter received mitigation treatment of vehicle alone or D68 (120 mg/ml) for 3 doses (10 min, 1 and 2 day post IR) was measured. The samples were collected on 3.5 days. The data were compared among the groups (5 mice/group) of normal mice, IR + vehicle and IR + D68. The results demonstrated that: 1) compared to the normal mice, the plasma level of secretin was reduced by 10

Gy sub-TBI due to the IR damage of epithelial cells lining in small intestine, which was reversed by treatment with D68 (Fig 8A); 2) the D12, D13 and D68 increased the secretion of CCK into plasma (Fig 8B). It was noted that in this particular experiment the purity of D68 was a little bit low (87%), therefore, its effect on survival was lower than D12 and Dl 3. When the purity of D68 was reached 98%, its effect was better than D12 and Dl 3; 3) the plasma insulin lever decreased on 3.5 days post-IR while D68 partially restored it (Fig 8C); 4) the similar results of plasma amylase was obtained, the IR-reduced amylase was increased when D68 was given right post-IR (Fig 8D). However, no significant differences of plasma glucose levels were observed between the IR and D68 treated group on 3.5 days post-IR. This may be due to the complication and compensation mechanisms triggered by IR. The reduced insulin in IR mice can increase the blood glucose level, however, due to the serious GI damage, the IR mice can not take food, which reduces the glucose levels. Alternatively, fat metabolism kicking in to make up the body energy can compensate for the level of blood glucose.

d) Effect of D68 on plasma inflammatory molecules after exposed to deadly sub-TBI

[00256] It is well-known that radiation as an insult (like other pathogens) can stimulate the expression of different inflammatory molecules (EVIs, mainly cytokines and chemokines), which represents body's response to IR stress and is the major underlying molecular mechanism for the progression of GI syndromes of ARS. So far, more than 100 IMs have been identified. Although many of them are not the initial target of IR but in the down-stream of IR stress, they are drugable targets that can be modified and alter the pathophysiological process of ARS.

[00257] Considering the factors that: 1) only about 0.4 ml of plasma could be collected from each mouse tested, which is not be enough to determine the level of many IMs if the standard ELISA is used; 2) the time taken to do ELISA one by one is unrealistically long (at least 4-5 hours per assay and days for a few IMs); and 3) ELISA technique is infeasible to fit the needs of large body of victim (or warfighter) under urgent IR exposure events, Luminex bead array was used. The paired monoclonal antibodies, highly-purified standard protein and more than 50 different regions of bead were purchased. The unlabeled antibodies were covalently conjugated

onto surface of beads according to manufacture's instruction. The amounts of antibody-coated beads, biotin-antibody and streptavidin-PE used for each well in different assay were individually optimized. The standard of each IM was calibrated with commercially available kits from Upstate Inc or R&D system Inc. The advantages of using techniques of Luminex bead array are: 1) it allows a high- throughput screen of a panel of biomarker; 2) it takes only 2.5—3.5 hours to finish the assay; 3) it requires only 25-50 μl of plasma which can be easily obtained from figure of IR victim; and 4) the equipment is portable, reliable and durable. Biomarkers, identified from this project, can be easily translated into human use, which provides a good window to monitor the ARS course and to guide the treatment protocol for individual victim.

[00258] The studies disclosed herein were carried out with the model of BALB/c mice that had 12 Gy sub-TBI and then received p.o. administration of vehicle alone (0.2 ml of saline) or D68 (120 mg/kg at 10 min post-IR and daily for two more days) or FGF-P (10 ug/mouse given in the same fashion as D68) or Amifostine (200 mg/kg, either 30 min before IR, or immediately after IR to compared with the anti-oxidant D68 which was given after IR). The samples were collected 3.5 days after IR, which was chosen for the purpose of determine if there was any correlation between the plasma markers and pathohistological changes.

[00259] The results were promising. As shown in Fig 9, several IMs that were increased by the 12 Gy IR were be reduced by D68, including MCP-I (Fig 9A), IL6 (Fig 9B), KC (Fig 9C), ILl β (Fig 9D), BLC (Fig 9E), TNFα (Fig 9F), TCA-3 (Fig 9G) and G-CSF (Fig 9H). In most cases, the D68 given p.o. after IR had the same tendency as Amifostine given i.v. before IR. The data again proved that Amifostine given after IR lost its anti-oxidant activity. Interestingly, FGF-P had a similar effect as D68 although their action mechanism was completely different.

[00260] Most of IMs measured were increased by IR and decreased (towards normal) by given D68, even G-CSF. One exception was that CD30 was reduced in IR group while the treatments had tendency to increase it. Most of the altered IMs were produced to enhance or attract immune cells to clean up the dead cells. Since the interfering with the pathological process at any point, such as D68 or Amifostine for

anti-IR induced free radices or FGF-P for enhancement of tissue repair, results in a reduced secondary tissue damage and cell death, therefore, the IMs were reduced. These data at molecular level, especially its mitigation effect occurs after IR exposure, again prove that D68 is a new anti-IR agent.

46. Example 46: Toxicity of D68

[00261] The parental compound, curcumin, is the major component of curry, spice food sold in grocery stores worldwide. The purified curcumin is very safe for human use, up to 8 g/day with no detectable toxicity. It is in clinical trail for cancer prevention and treatment as well as other anti-oxidation and anti-inflammation purposes.

Most of anti -radiation agents have a therapeutic index (TI=LD 5 o/ED 5 o) of about 10-15.

[00262] To determine the TI of D68, BALB/c mice (6/group) were p.o. given D68, starting with dose of 1500 mg/kg, a dose 10 times higher than the ED 50 (120-150 mg/kg). So far, doses up to 2,000 mg/kg (13-16 times higher than ED 50 ) have been administered and none of testing mice died.

47. Example 47: Pharmacokinetic (PK) study of D68

[00263] The absorption, distribution, metabolism and excretion of D68 are important information for optimization of treatment schedule and the formulation of this compound.

a) Selection of Methods:

[00264] There are two methods can be used for PK of D68 : the HPLC or radiolabled ( 3 H or 14 C) D68. The advantages of HPLC are: 1) the assay can be used in human clinical trials; and 2) avoids the special handling and disposal requirements for use of long-lived radioisotopes in the laboratory. Radiolabeled D68 can sensitively trace the movement of D68 in animal models, but cannot be used in human studies. HPLC was used first and if needed, alternative radiolabel assays were used to confirm the results obtained from HPLC -based studies in animal models.

b) Optimization of sample extraction and HPLC analytical methods:

[00265] The current approach to quantifying D68 in plasma and tissues is a single stage extraction method using solid phase extraction (SPE) techniques followed by HPLC analysis. Recoveries of D68 from human plasma spiked at concentrations of 0.04, 0.2, 1 or 5 μg/mL D68 ranged from 74.6-98.1% for D68. The recoveries of D68 from spiked human plasma increased with decreasing D68 concentration. The percent relative standard deviation (RSD) in the inter-day recovery of D68 from spiked plasma (5 μg/mL) on three separate days was 1.13.

[00266] Using the sample extraction and HPLC analytical methods described in section D.1.2 and D.1.3, good resolving power is achieved allowing for the identification and quantification of D68 and curcumin (internal standard) in plasma and liver extracts tested to date. Figure 11 shows a typical chromatogram obtained for a SPE-extracted plasma sample where D68 and curcumin are measured at 327 run (upper panel) and 430 nm (lower panel), respectively. In this example, human plasma is spiked with D68 and curcumin at concentrations of 0.2 and 1.0 μg/mL, respectively. In the upper panel, the chromatogram shows D68 eluting at 7.2 min, whereas in the lower panel curcumin elutes at 16.2 min. These chromatograms demonstrate the ability to adequately resolve D68 and curcumin from plasma components.

[00267] The limits of detection (LOD), as calculated from the calibration-design- dependent approach, for D68 in plasma are defined as 5 and 10 times of the background in the chromatograms, respectively. Using this definition, the LOD and LOQ for D68 in spiked plasma are 0.05 μg/ml and 0.10 μg/ml, respectively. Based a preliminary C max of 0.25 μg/ml for D68 in plasma at 3.3 h for a single 120 mg/kg (p.o.) dose in BALB/c mice, the peak height for this concentration is 5 times greater than the LOQ for the current assay. The LOD/LOQ of the D68 quantitative assay is expected to improve by an order of magnitude as the wash steps in the SPE extraction phase of the assay are optimized.

[00268] The results of the analysis of D68 in plasma as a function of time are shown in Fig. 10. The mass of D68 recovered from the plasma was found to be highly variable. Based on a crude fit of the D68 data by eye, a C max of approximately 0.25

μg/mL was found to occur at 3.3 hours (W) in plasma. An AUC t of 2.2 μg # h/mL was calculated by the linear trapezoid rule as applied to the fitted curve.

[00269] Further analysis of the HPLC chromatograms from the PK studies shows the presence of additional peaks that are believed to be metabolites of D68. These peaks are clearly seen in Fig. 12 (denoted as D68*) and amount to a much larger combined area in the chromatogram than D68, assuming similar extinction coefficients for the D68 metabolites and the parent compound. These peaks represent the glucuronide and glucuronide/sulfate conjugates of D68, based on the retention times of these peaks relative to the D68 peak in the chromatograms and their UV spectra. The formation of glururonide and glucuronide/sulfate conjugates of D68 are consistent with what is known about the metabolism of D68's parent compound, curcumin. hi the case of curcumin, conjugation at one or both of the phenoxy groups is known to be a major metabolic pathway for the compound. Curcumin also undergoes reduction of the heptatrienone chain and cleavage at or near the diketone function. Whether these additional metabolic processes occur in the metabolism of D68 has yet to be established.

[00270] From an analysis of the combined concentrations of the D68 metabolites as a function of time (Fig. 11, panel B), the C max , t max and AUQ were determined for the combined metabolites. The t max for these metabolites of 3.3 hr coincides well with the tmax for D68. Comparison of the C max (2.0 μg/mL) and AUC t (12.7 μg » h/mL) calculated for the D68 metabolites to the values obtained for D68 indicates that D68 is rapidly conjugated (85%). It remains to be determined the extent to which the conjugation occurs in the intestine and in the liver. Also, additional study is needed to ascertain whether the conjugation of D68 decreases or eliminates the therapeutic effect of the compound or, as is the case for curcumin, the conjugated forms of the compound show similar activity in comparison to the parent towards inhibiting COX- 2 activity.

[00271] Because D68 is intended to be given orally, the effect that irradiation of the GI tract has on the PK of the agent was examined. For these experiments, mice where γ-irradiated to a dose of 12 Gy (sub-TBI) and then administered D68 (120 mg/kg, po) within 15 min following irradiation. Analysis of the concentrations of D68 and the

combined D68 metabolites in plasma as a function of time from administration of D68 post-radiation yielded AUC t values of 0.38 μg » h/ml for D68 and 6.5 μg # h/ml for the D68 metabolites. These values are lower than the AUC t values obtained in unirradiated mice by factors of 5.8 and 2.0, respectively. Thus, it appears that uptake of D68 from the GI tract is acutely affected when the gut is irradiated to a dose of 12 Gy sub-TBI.

[00272] Taken together, it is disclosed herein and demonstrated that: 1) the sub- TBI mouse (BM of leg was shielded, which allow a high dose that cause GI death without BM death) was good to study mitigation agent for GI syndromes of ARS; 2) the agents disclosed herein and in particular D68 have a high redox potential as measured with electronic chemical detector, which rendered the agents with a strong anti-oxidant property; 3) unlike Amifostine, the agents disclosed herein exerted its mitigation effect after IR exposure; 4) D68 agent was administrated via p.o. and D68 and the other agents disclosed herein can be pocketed by warfighter since it is a dried small stable molecule; 5) the agents, especially D68, rescued mice from GI syndromes of ARS after exposure to an IR dose of LD 50/7 (83%) and LD i OO / 7 (43%). The GI mitigation effect was observed in at lease two strains, suggesting the universal effect regardless genome background; 6) the agents were also capable of rescuing mice from a lethal BM dose, 7Gy (DMF>1.2) TBI; 7) the direct effect of the agent on GI tract was evidenced by the enhancement of crypts proliferation (deep chromophilic hemotoxilin staining and positive BrdU staining) and the preservation of length of villi; 8) D68 partially restores the GI endocrine and exocrine ability in mice with deadly GI syndrome of ARS, as evidenced by the low stool hemoccult score, reduced severity of loose and bloody stool and endotoximia; 9) D68 partially restores the GI endocrine and exocrine ability in mice with deadly GI syndrome of ARS; 10) several IMs that were increased by the 12 Gy IR were reduced by D68, including MCP-I, IL6, KC, ILl β, BLC, TNFα, TCA-3 and G-CSF. The effective pattern of D68 on IMs was similar to that of Amifostine; 11) D68 was well tolerated by the mice. The therapeutic index was >15, which is relatively safe; and 12) the PK pilot study indicated that the established procedure for sample extraction, HPLC assay running condition and the reference marker worked together sensitive enough to detect the trace amount of D68

in plasma and tissues. The peak of absorption occurred at 3 hours after D68 was taken orally.

[00273] All the data (compound redox potential, survival assay performed at LD 5 0/7 and LD 100/7 for GI and BM death, GI integrity of anatomy and function, systemic reaction of Ms) strongly suggest that D68 is a very promising agent to be developed as a novel mitigation drug for treatment of GI syndromes of ARS.

48. Example 48: DPPH (2,2-Diphenyl-l-Picrylhydrazyl ) Free Radical Scavenging Activity

[00274] DPPH (1 , below left) is characterized as a stable free radical.

I: Dipheny lpicrylhyd razy I (free radical) 2: Diphenylpkrylhydrazine (nonradical)

[00275] When a solution of DPPH is mixed with that of a substance that can donate a hydrogen atom, this gives rise to the reduced form (2, above right). When in the presence of curcumin or a derivative thereof, the percentage of free radical is reduced dramatically (Figure 13). The equation for calculating % inhibition of DPPH activity is as follows: % inhibition = (1 - Ab515 Sample/Ab515 Bland ) x 100. The IC50 value was used to compare DPPH scavenging activity. The results can be seen in Figure 14.

Table 2: IC50 values of Curcumin, Ds and Trolox

Compounds in Bold: p < 0.05 compare with curcumin.

[00276] The IC50 value indicated that the scavenging activity of curcumin was comparable to Trolox, D12, D13, D47, D68, D71, D72, D73, D75, D76 and D81 were more potent than curcumin. Other D compounds listed were less potent.

49. Example 49: Evaluation of Total Antioxidant Capacity (TAC)

[00277] The method used to evaluate TAC was the phosphomolybdenum method. This method is based on the reduction of Mo(VI) / Mo(V) by the sample analyte and subsequent formation of green phosphate/Mo(V) complex with a maximal absorption at 695 run.

[00278] The results were presented as water-soluble ascorbic acid equivalents (μmol/g of sample) at lmg/ml concentration. The results are shown in Figure 15.

[00279] D12, D13, D47, D68, D71, D72, D73, D75, D76 and D81 showed more antioxidant ability in both of these two assays. Among these tested compounds, D 12, Dl 3 and D68 showed protective effects in the in vivo animal study. D47 was remarkably much more potent than curcumin in vitro.

Table 3: Total Antioxidant Capacity of Ds (Equivalent of ascorbic acid μmol/g sample).

Compounds in bold: p < 0.05 compare with curcumin.

50. Example 50: Intracellular ROS measurement

[00280] The biological effects of radiation are due to the production of reactive oxygen species (ROS). ROS can induce oxidative damage to vital cellular molecules. ROS play an important role in irradiation-induced cell damage. To determine the antioxidant effect of Ds involve in ROS, intracellular ROS were detected by FCM in SW 480 cell line (Figure 16).

[00281] The data showed many of the Ds, for example D12, D13, D47, D68, etc. had a better free radical scavenging activity and total antioxidant capacity than curcumin. They are therefore useful as antioxidants.

51. Example 51: Properties of Curcumin and D-Compounds Thereof

[00282] Curcumin and the D-compounds described herein have many useful properties. This includes lipophilic properties. Protective effects can be found in membranes, and good radical scavengers can also be found in cytosolic compartments. Curcumin and D-compounds contain conjugated pi-bonding systems,

which provides an advantage by stabilizing free radicals formed in scavenging reactions. Redox potentials are within a range where these compounds function as antioxidants; reducing ROS.

[00283] Curcumin possesses a pro-oxidant activity as mitigation agent. It has been shown to increase ROS -mediated cellular damage (nDNA, mtDNA, proteins, membranes); and this has been shown to be effective at 20-40 μg/mL, with a 1-6 hour exposure time in tumor cell lines. Pro-oxidant activity is mediated through production of superoxide radicals; this reduces thioredoxin reductase activity through alkylation of protein; and induction of NAPDH oxidase activity takes place. Normal tissues show some elevation in ROS when treated with curcumin. This is offset by increased antioxidant capacity (i.e. thiols, catalase, SOD, etc.)

[00284] Curcumin and derivatives thereof have been shown to have radical scavenging activity - hydroxyl radical, lipid radicals (carbon radicals, peroxyl, alkoxyl). The rate constants are > 10 7 M "1 s "1 . Curcumin has + O 2~ ; 4.7 x 10 5 M '1 s "1 . Oxidation of O 2- to form O 2 .

[00285] The D-compounds (i.e. D47) are comparable to curcumin as oxidizing agents of O 2- . Direct oxidation occurs as follows:

1) D47 + O 2~ → D47 «~ + O 2

52. Example 52: Cytochrome C Assay [00286] The cytochrome C assay is a way to indirectly measure O 2 .

O 2 production occurs as follows:

xanthine oxidase + xanthine + O 2 → O 2 -.

The rate of O formation followed by production of Fe :

O 2 + CytoC-(Fe +3 ) → 02 + CytoC-(Fe +2 )

Inhibition of Fe +2 production by D-compounds:

O 2~ + D47 → O 2 + D47 »~

a) Methods:

[00287] Reaction buffer: 50 niM Tris « HCl (pH 7.4), 600 μM EDTA, 100 μM xanthine with 37.5% DMSO. Cytochrome C (horse heart) at 190 μM in 50 mM Tris'HCl (pH 7.4), 600 μM EDTA. Xanthine oxidase (bovine milk) at 0.2 units/mL 50 mM Tris'HCl (pH 7.4), 600 μM EDTA.

[00288] Reaction mixture: 96-well microtiter plate; 3 blank replicates (270 μL reaction buffer + 30 μL cytoC); 3 sample replicates (250 μL reaction buffer + 30 μL cytoC + 15 μL xanthine oxidase + 5 μL test compound.

[00289] Final concentration of reactants: 90 μM xanthine, 0.01 units xanthine oxidase, 19 μM cytoC 9.5-76 μM Curcumin/D-compounds (molar ratios of 0.5,1,2,3, and 4 relative to cytoC. The results are found in Figure 17.

[00290] A competition kinetics assay was also used. The second order rate constant for a compound from competition kinetics was measured with a common reactant and known rate constant from a reference compound. Two competing reactions:

O 2~ + CytoC-(Fe +3 ) → O 2 + CytoC-(Fe +2 )

O 2~ + cur → O 2 + cur »~

(m cyt oc/m cur ) - 1 = kcur/kcytoc [cur]/[cytoC]

Linear plot of (m cyto C/m cur ) - 1 vs. [cur]/[cytoC]

Slope x k c yt o c (5.8 x 10 5 M "1 s "1 ) = k^

[00291] The results can be seen in Figure 18.

Table 4: Second Order Rate Constants D compounds > Curcumin

53. Example 53: Structures of Compounds D71-D76 and D81

D71

D74

[00292] Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.

[00293] It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.