Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED 2-AMINOBENZAMIDE CASPASE INHIBITORS AND THE USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2000/055114
Kind Code:
A1
Abstract:
The present invention is directed to novel 2-aminobenzamide thereof, represented by general Formula (I) where R¿1?-R¿3?, X and A-D are defined herein. The present invention also relates to the discovery that compounds having Formula (I) are potent inhibitors of caspases and apoptotic cell death. Therefore, the inhibitors of this invention can retard or block cell death in a variety of clinical conditions in which the loss of cells, tissues or entire organs occurs.

Inventors:
CAI SUI XIONG
WANG YAN
WEBER ECKARD
MILLS GORDON B
GREEN DOUGLAS R
Application Number:
PCT/US2000/006398
Publication Date:
September 21, 2000
Filing Date:
March 15, 2000
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CYTOVIA INC (US)
International Classes:
A61K9/02; A61K9/06; A61K9/08; A61K9/22; A61K31/27; A61K31/381; C12N9/99; A61K38/00; A61P1/00; A61P1/02; A61P1/04; A61P1/16; A61P1/18; A61P3/10; A61P7/00; A61P7/06; A61P9/00; A61P13/10; A61P13/12; A61P15/08; A61P17/00; A61P17/02; A61P17/06; A61P17/14; A61P17/16; A61P19/02; A61P19/08; A61P25/00; A61P25/32; A61P27/02; A61P29/00; A61P37/00; A61P39/00; A61P43/00; C07C271/22; C07C271/24; C07C271/28; C07D333/38; C07D333/40; C07K5/023; C07K7/02; C12N5/04; C12N5/07; C12N5/0793; (IPC1-7): C07C229/00; C07C69/74; C07C205/00; A01N37/10; A01N37/12; A01N37/00
Other References:
CHEMICAL ABSTRACTS, vol. 132, 1999, Columbus, Ohio, US; abstract no. 273983P, WATAYA, Y ET AL: "Cytotoxic Mechanism of 1-(3-C-ethynyl-,beta,-D-ribo-pentofuranosyl) cytosine (ECyd)" XP002930807
CHEMICAL ABSTRACTS, vol. 127, 1997, Columbus, Ohio, US; abstract no. 315124P, PELEG, S ET AL: "1,25-Dihydroxyvitamin D3 and its Analogs Inhibit Acute Myelogenous Leukemia Progemitor Proliferation by Suppressing Interleukin-1.beta Production" XP002930808
Attorney, Agent or Firm:
Esmond, Robert W. (Kessler Goldstein & Fox P.L.L.C. Suite 600 1100 New York Avenu, N.W. Washington DC, US)
Download PDF:
Claims:
What Is Claimed Is :
1. A compound having the Formulae I or II or III: or a pharmaceutically acceptable salt or prodrug thereof, wherein: R, is an optionally substituted alkyl or hydrogen; R3 is an Nprotecting group; R2 is hydrogen or optionally substituted alkyl; Q is an optionally substituted saturated or partially saturated carbocycle or heterocycle; X is a peptide of 14 amino acids or a bond; Y is a peptide of 14 amino acids or a bond; A is CR6 or nitrogen; B is CR7 or nitrogen; C is CR8 or nitrogen; D is CRg or nitrogen; provided that not more than two of A, B, C or D is nitrogen; and R6R9 <BR> <BR> independently are hydrogen, halo, C,C6 haloalkyl, C6Cl 0 aryl, C4C7 cycloalkyl, C1C6 alkyl, C2C6 alkenyl, C2C6 alkynyl, C6CI0 aryl (C,C6) alkyl, C6CI0 aryl (C2C6) alkenyl, C6CI0 aryl (C2C6) alkynyl, ClC6 hydroxyalkyl, nitro, amino, cyano, C,C6 acylamino, hydroxy, ClC6 acyloxy, CC6 alkoxy, alkylthio, or carboxy; or one of R6 and R7, or R, and R8, or Rg and R9 are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle; E is C, 4, nitrogen, oxygen or sulfur; F is C, 5, nitrogen, oxygen or sulfur; G is Cl6, nitrogen, oxygen or sulfur; provided that only one of E, F, G is nitrogen, oxygen or sulfur and R, 4R, 6 are independently hydrogen, halo, C,C6 haloalkyl, C6Cl, aryl, C4C7 cycloalkyl, ClC6 alkyl, ClC6 alkenyl, C2C6 alkynyl, C6C, 0 aryl (ClC6) alkyl, C6Cl0 aryl (C2C6) alkenyl, C6C10 aryl (C2C6) alkynyl, C,C6 hydroxyalkyl, nitro, amino, cyano, C,C6 acylamino, hydroxy, ClC6 acyloxy, ClC6 alkoxy, alkylthio, or carboxy; or one of R14 and Rl5, or R, 5 and R, 6, are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle.
2. A compound according to claim 1, wherein R3 is t butyloxycarbonyl, acetyl or benzyloxycarbonyl.
3. A compound according to claim 1, wherein R, is H, Me, Et or acetoxymethyl.
4. A compound according to claim 1, wherein R2 is hydrogen, fluoromethyl, acyloxymethyl, arylacyloxymethyl or aminomethyl.
5. A compound according to claim 1, wherein X is a bond.
6. A compound according to claim 1, wherein A, B, C and D are CH.
7. A compound according to claim 1, wherein A is nitrogen, and B, C and D are CH.
8. A compound according to claim 1, wherein G is sulfur, and E and F are CH.
9. A compound according to claim 1, wherein Q is cyclohexyl or cyclopentyl.
10. A compound according to claim 1, wherein said compound has the Formula IV: or a pharmaceutically acceptable salt or prodrug thereof, wherein R2 is hydrogen or optionally substituted alkyl, wherein the substituent is halo, hydroxy, alkoxy, aryloxy, alkylthio, arylthio, amino, acyloxy, or arylacyloxy; R6R9 independently are hydrogen, halo, C,C6 haloalkyl, C6Cl0 aryl, C4C7 cycloalkyl, C,C6 alkyl, C2C6 alkenyl, C2C6 alkynyl, C6Cl0 aryl (ClC6) alkyl, C6Cl0 aryl (C2C6) alkenyl, C6C, 0 aryl (C2C6) alkynyl, CC6 hydroxyalkyl, nitro, amino, cyano, C,C6 acylamino, hydroxy, CC6 acyloxy, C,C6 alkoxy, alkylthio, or carboxy; or one of R6 and R,, or R, and R8, or R8 and R, are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle, selected from the group consisting ofOCH2O,OCF20, (CH,(CH,OCHCHO,CH,N(R)CH, CH2CH2N(R13)CH2, CH2N (R, 3) CH2CH2and CH=CHCH=CH; wherein R, 3 is hydrogen, alkyl or cycloalkyl ; R, o is hydrogen, C,C6 alkoxy, C,C6 alkyl, C,C6 haloalkyl, C6C, 0 aryl, C4C7<BR> <BR> <BR> <BR> <BR> <BR> cycloalkyl, C6C, o aryl (C,C6) alkyl, benzyloxy, substituted benzyloxy, or NRlIR12 ; wherein R and R, 2 independently are hydrogen, CjCg alkyl, C,C6 <BR> <BR> <BR> <BR> haloalkyl, C6C, o aryl, C4C7 cycloalkyl, C6C, o aryl (CC6) alkyl, or R and R, 2 are combined to form a heterocyclic ring system selected from the group consisting of pyrrolidine, piperidine, piperazine, and morpholine.
11. A compound according to claim 10, wherein R2 is hydrogen, fluoromethyl, acyloxymethyl, arylacyloxymethyl or aminomethyl.
12. A compound according to claim 10, wherein R, o is benzyloxy.
13. A compound according to claim 10, wherein R, is H, Me or acetoxymethyl.
14. A compound according to claim 10, wherein X is a peptide of 12 amino acids or a bond.
15. A compound according to claim 1, wherein said compound is selected from the group consisting of : 2 (Zamino) benzoylAspfmk, <BR> <BR> <BR> <BR> 2 (Zamino)3methylbenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> 2 (Zamino)3, 5dimethylbenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> <BR> 2 (Zamino)4chlorobenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> <BR> 2 (Zamino)5chlorobenzoylAspfink,<BR> <BR> <BR> <BR> <BR> 2 (Zamino)5fluorobenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> <BR> 2 (Zamino)6fluorobenzoylAspfmk, cis2 (Zamino)cyclohexanecarboxyAspfmk, <BR> <BR> <BR> 2 (Zamino)5methylbenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> <BR> 2 (Zamino)6methylbenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> 2 (Zamino)6chlorobenzoylAspfmk,<BR> <BR> <BR> <BR> <BR> <BR> 2 (Zamino)3methoxybenzoylAspfmk, 3 (Zamino) thiophene2carboxylAspfmk,<BR> <BR> <BR> <BR> <BR> <BR> <BR> 3(methoxycarbonylamino) thiophene2carboxylAspfmk, cis2 (Zamino) cyclopentanecarboxylAspfmk, trans2 (Zamino) cyclopentanecarboxylAspfmk, 2 (Zamino) benzoylAspDCBmethylketone, methoxycarbonylVal(2aminobenzoyl)Aspfmk, ZGlu (2aminobenzoyl)Aspfmk, and ZVal (2aminobenzoyl)Aspfmk.
16. A pharmaceutical composition, comprising a compound of claim 1, and a pharmaceutically acceptable carrier.
17. A method of inhibiting cell death of a cell or tissue, comprising contacting said cell or tissue with an effective amount of a compound of claim 1.
18. A method of treating or ameliorating cell death in the central or peripheral nervous system, retinal neurons, cardiac muscle or immune system cells of an animal, comprising administering to the animal in need of such treatment or ameliorating an effective amount of a compound of claim.
19. The method of claim 18, wherein said cell death is in the central or peripheral nervous system, and is due to one of : (a) a condition of ischemia and excitotoxicity selected from the group consisting of focal ischemia due to stroke and global ischemia due to cardiac arrest; (b) traumatic injury; (c) viral infection; (d) radiationinduced nerve cell death; (e) a neurodegenerative disorder selected from the group consisting of Alzheimer's disease, Parkinson's Disease, a prion disease, multiple sclerosis, amyotrophic lateral sclerosis, and spinobulbar atrophy; (f) spinal cord injury; or (g) acute bacterial meningitis.
20. The method of claim 18, wherein said cell death is in the central or peripheral nervous system, and is due to expansion of trinucleotide repeats of specific genes.
21. The method of claim 18, wherein said cell death is due to Huntington's Disease.
22. The method of claim 18, wherein said cell death is in cardiac muscle tissue, and is due to myocardial infarction, congestive heart failure, cardiomyopathy or viral infection of the heart.
23. The method of claim 18, wherein said cell death is in retinal neurons and is due to increased intraocular pressure, agerelated macular degeneration or retinitis pigmentosa.
24. The method of claim 18, wherein said cell death is in the immune system, and is due to an immune deficiency disorder selected from the group consisting of acquired immune deficiency syndrome, severe combined immune deficiency syndrome and radiationinduced immune suppression.
25. The method of claim 18, wherein said cell death is due to an autoimmune disorder selected from the group consisting of lupus erythematosus, rheumatoid arthritis and type I diabetes.
26. The method of claim 18, wherein said cell death is due to type I diabetes.
27. A method of treating or preventing polycystic kidney disease, renal amyloidosis, acute renal failure, cyclosporine A induced tubular epithelial cell death, hypoxiainduced necrosis of renal proximal tubules, HIV induced nephropathy or anemia/erythropoiesis in an animal, comprising administering to the animal in need of such treatment an effective amount of a compound of claim 1.
28. A method of protecting a mammalian organ or tissue from cell death due to deprivation of normal blood supply, comprising contacting said organ or tissue with an effective amount of a compound of claim 1.
29. The method of claim 28, wherein said organ or tissue is present in a storage medium prior to transplant into a mammal.
30. The method of claim 28, wherein said contacting comprises infusion of said compound into the organ or tissue, or bathing of said organ or tissue in a storage medium which comprises said compound.
31. A method of reducing or preventing cell death in a donor organ or tissue after it has been transplanted into a host due to the effects of reperfusion injury or due to the effects of host immune cells, comprising administering to said host in need thereof an effective amount of a compound of claim 1.
32. A method of reducing or preventing the death of mammalian sperm or eggs used in in vitro fertilization procedures, comprising contacting said sperm or egg with an effective amount of a compound of claim 1.
33. A method of extending the lifespan of a mammalian or yeast cell line, comprising contacting said cell line with a compound of claim 1.
34. The method of claim 33, wherein said contacting comprises including said compound in a cell growth medium.
35. A method of treating or ameliorating hair loss or premature graying of the hair in a mammal, comprising contacting the hair or hair follicles of the mammal in need thereof with a compound of claim 1.
36. The method of claim 35, wherein hair loss is treated, and said hair loss is due to malepattern baldness, radiation, chemotherapy or emotional stress.
37. A method of treating or ameliorating skin damage of a mammal due to exposure to high levels of radiation, heat or chemicals, comprising applying to the skin of the mammal in need thereof with a compound of claim 1.
38. The method of claim 37, wherein said compound is applied as part of an ointment.
39. The method of claim 37, wherein said skin damage is due to acute overexposure to the sun, and wherein said treating reduces blistering and peeling of the skin.
40. A method of treating or ameliorating sepsis or multiorgan failure in an animal, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
41. A method of treating or ameliorating hepatitis in an animal, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
42. A method of treating or ameliorating hereditary tyrosinemia type 1 in an animal, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
43. A method of treating or ameliorating chronic alcohol ingestion induced buccal mucosa cell death in an animal, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
44. A method of treating or ameliorating cell death in plants or flowers, comprising administering to the plants or flowers in need thereof an effective amount of a compound of claim 1.
45. A method of treating or ameliorating radiation or ultraviolet irradiation induced cell death in an animal, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
46. A method of treating or ameliorating apoptotic death of bone marrow cells in myelodysplastic syndromes (MDS), comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
47. A method of treating or ameliorating apoptotic cell death in acute pancreatitis, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
48. A method of treating or preventing the inflammatory response in psoriasis or inflammatory bowel disease, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
49. A method of treating or ameliorating organ apoptosis after burn injury, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
50. A method of treating or ameliorating small bowel tissue injury after intestinal ischemiareperfusion, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
51. A method of treating, ameliorating or preventing oral mucositis, gastrointestinal mucositis, bladder mucositis, proctitis, bone marrow cell death, skin cell death or hair loss resulting from chemotherapy or radiation therapy of cancer in an animal, comprising administering to the animal in need thereof an effective amount of a compound of claim 1.
52. The method of claim 51, wherein said compound is administered topically or orally.
53. The method of claim 52, wherein said compound is formulated as part of a mouthwash for the treatment, amelioration or prevention of oral mucositis.
54. The method of claim 52, wherein said compound is formulated as part of a slow release buccal lozenge.
55. The method of claim 52, wherein said compound is formulated as part of a suppository.
56. The method of claim 52, wherein said compound is formulated as part of a gel.
57. The method of claim 52, wherein said compound is administered through a bladder catheter for the treatment, amelioration or prevention of bladder mucositis.
58. The method of claim 52, wherein said compound is administered as part of an enema for the treatment, amelioration or prevention of proctitis.
59. The method of claim 52, wherein said compound is formulated as an oral formulation which is capable of coating the gastrointestinal surfaces for the treatment, amelioration or prevention of gastrointestinal mucositis.
60. The method of claim 59, wherein said gastrointestinal mucositis is esophageal mucositis, gastric mucositis, or intestinal mucositis.
61. The method of claim 51, wherein said compound is administered by i. v. injection for the treatment, amelioration or prevention of bone marrow cell death.
62. The method of claim 51, wherein said compound is administered as part of a pharmaceutical composition comprising a pharmaceutically acceptable carrier.
63. The method of claim 51, wherein said compound is administered after chemotherapy or radiation therapy of cancer in said animal.
64. The method of claim 51, wherein said compound is administered during chemotherapy or radiation therapy of cancer in said animal.
65. The method of claim 51, wherein said compound is administered prior to chemotherapy or radiation therapy of cancer in said animal.
Description:
Substituted 2-Aminobenzamide Caspase Inhibitors and the Use Thereof Background of the Invention Field of the Invention This invention is in the field of medicinal chemistry. In particular, the invention relates to substituted 2-aminobenzamides and analogs that are inhibitors of caspases. The invention also relates to the use of these 2- aminobenzamides and analogs for reducing or treating apoptotic cell death and/or reducing interleukin 1-P production.

Description of Background Art Organisms eliminate unwanted cells by a process variously known as regulated cell death, programme cell death or apoptosis. Such cell death occurs as a normal aspect of animal development as well as in tissue homeostasis and aging (Glucksmann, A., Biol. Rev. Cambridge Philos. Soc. <BR> <BR> <BR> <BR> <P>26: 59-86 (1951); Glucksmann, A., Archives de Biologie 76: 419-437 (1965);<BR> <BR> <BR> <BR> <BR> Ellis et al., Dev. 112: 591-603 (1991); Vaux et al., Cell 76 : 777-779 (1994)).

Apoptosis regulates cell number, facilitates morphogenesis, removes harmful or otherwise abnormal cells and eliminates cells that have already performed their function. Additionally, apoptosis occurs in response to various physiological stresses, such as hypoxia or ischemia (PCT published application W096/20721).

There are a number of morphological changes shared by cells experiencing regulated cell death, including plasma and nuclear membrane blebbing, cell shrinkage (condensation of nucleoplasm and cytoplasm), organelle relocalization and compaction, chromatin condensation and

production of apoptotic bodies (membrane enclosed particles containing intracellular material) (Orrenius, S., J. Internal Medicine 237: 529-536 (1995)).

Apoptosis is achieved through an endogenous mechanism of cellular suicide (Wyllie, A. H., in Cell Death in Biology and Pathology, Bowen and Lockshin, eds., Chapman and Hall (1981), pp. 9-34). A cell activates its internally encoded suicide program as a result of either internal or external signals. The suicide program is executed through the activation of a carefully regulated genetic program (Wylie et al., Int. Rev. Cyt. 68 : 251 (1980); Ellis et al., Ann. Rev. Cell Bio. 7 : 663 (1991)). Apoptotic cells and bodies are usually recognized and cleared by neighboring cells or macrophages before lysis.

Because of this clearance mechanism, inflammation is not induced despite the clearance of great numbers of cells (Orrenius, S., J. Internal Medicine 237: 529-536 (1995)).

Mammalian interleukin-lß (IL-lß) plays an important role in various pathologic processes, including chronic and acute inflammation and autoimmune diseases (Oppenheim, J. H. et. al. Immunology Today, 7,45-56 (1986)). IL-1 is synthesized as a cell associated precursor polypeptide (pro- IL-1 (3) that is unable to bind IL-1 receptors and is biologically inactive (Mosley et al., J. Biol. Chem. 262: 2941-2944 (1987)). By inhibiting conversion of precursor IL-1B to mature IL-113, the activity of interleukin-1 can be inhibited. Interleukin-lß converting enzyme (ICE) is a protease responsible for the activation of interleukin-lß (IL-lß) (Thornberry, N. A., et al., Nature 356: 768 (1992); Yuan, J., et al., Cell 75: 641 (1993)). ICE is a substrate-specific cysteine protease that cleaves the inactive prointerleukin-1 to produce the mature IL-1. The genes that encode for ICE and CPP32 are members of the mammalian ICE/Ced-3 family of genes which presently includes at least twelve members: ICE, CPP32/Yama/Apopain, mICE2, ICE4, ICH1, TX/ICH-2, MCH2, MCH3, MCH4, FLICE/MACH/MCH5, ICE-LAP6 and ICEre, III. The proteolytic activity of this family of cysteine proteases, whose active site (a cysteine residue) is essential for ICE-mediated apoptosis, appears critical in mediating cell death (Miura et al., Cell 75: 653-660 (1993)).

This gene family has recently been named caspases (Alnernri, E. S. et. al. Cell,

87,171 (1996), and Thornberry, N. A. et. al. J. Biol. Chem. 2 72,17907-17911 (1997)) and divided into three groups according to its known functions. Table 1 summarizes these known caspases.

Table 1 Enzyme* Group I: mediators of inflammation Caspase-1 (ICE)<BR> Caspase-4 (ICE-II TX, ICH-2)<BR> Caspase-5 (ICE-III TY) Group II: effectors of apoptosis Caspase-2 (ICH-1, mNEDD2)<BR> Caspase-3 (apopain, CPP-32, YAMA)<BR> Caspase-7 (Mch-3, ICE-LAP3, CMH-1) Group III: activators of apoptosis Caspase-6 (Mch2)<BR> Caspase-8 (MACH, FLICE, Mch5)<BR> Caspase-9 (ICE-LAP6, Mch6)<BR> Caspase-10 IL-1 is also a cytokine involved in mediating a wide range of biological responses including inflammation, septic shock, wound healing, hematopoiesis and growth of certain leukemias (Dinarello, C. A., Blood 77: 1627-1652 (1991); diGiovine et al., Immunology Today 11: 13 (1990)).

Many potent caspase inhibitors have been prepared based on the peptide substrate structures of caspases. However, in contrast to their potency in vitro, no inhibitors with good efficacy (IC50 < 1 pM) in whole-cell models of apoptosis have been reported (Thornberry, N. A. Chem. Biol. 5 : R97-103 (1998)). Therefore the need exists for cell death inhibitors that are efficacy in whole-cell models of apoptosis and active in animal model of apoptosis. These inhibitors thus can be employed as therapeutic agents to treat disease states in which regulated cell death and the cytokine activity of IL-1 play a role.

WO 93/05071 discloses peptide ICE inhibitors with the formula: Z-Q2AspQ1

wherein Z is an N-terminal protecting group; Q2 is 0 to 4 amino acids such that the sequence Q2-Asp corresponds to at least a portion of the sequence Ala-Tyr- Val-His-Asp; Q, comprises an electronegative leaving group.

WO 96/03982 discloses aspartic acid analogs as ICE inhibitors with the formula:

wherein R2 is H or alkyl; R3 is a leaving group such as halogen; R, is heteroaryl-CO or an amino acid residue.

U. S. patent 5,585,357 discloses peptidic ketones as ICE inhibitors with the formula:

wherein n is 0-2; each AA is independently L-valine or L-alanine; R, is selected from the group consisting of N-benzyloxycarbonyl and other groups; R8, RgS Rlo are each independently hydrogen, lower alkyl and other groups.

Mjalli et al. (Bioorg Med. Chem. Lett., 3,2689-2692,1993) report the preparation of peptide phenylalkyl ketones as reversible inhibitors of ICE, such as: Thornberry et al. (Biochemistry, 33,3934-3940,1994) report the irreversible inactivation of ICE by peptide acyloxymethyl ketones:

wherein Ar is COPh-2,6- (CF3) 2, COPh-2, 6-(CH3) 2, Ph-F5 and other groups.

Dolle et al. (J Med Chem. 37,563-564,1994) report the preparation of P, aspartate-based peptide oc- ( (2,6-dichlorobenzoyl) oxy) methyl ketones as potent time-dependent inhibitors of ICE, such as: Mjalli et al. (Bioorg Med. Chem. Lett., 4,1965-1968,1994) report the preparation of activated ketones as potent reversible inhibitors of ICE: wherein X is NH (CH2) 2, OCO (CH2) 2, S (CH2) 3 and other groups.

Dolle et al. (J. Med. Chem. 37,3863-3866,1994) report the preparation of a- ((1-phenyl-3-(trifluoromethyl)-pyrazol-5-yl) oxy) methyl ketones as irreversible inhibitor of ICE, such as: Mjalli et al. (Bioorg Med. Chem. Lett., 5,1405-1408,1995) report inhibition of ICE by N-acyl-Aspartic acid ketones: wherein XR2 is NH (CH2) 2Ph, OCO (CH2) 2cyclohexyl and other groups.

Mjalli et al. (Bioorg. Med. Chem. Lett., 5,1409-1414,1995) report inhibition of ICE by N-acyl-aspartyl aryloxymethyl ketones, such as: Dolle et al. (J. Med. Chem. 38,220-222,1995) report the preparation of aspartyl a- ( (diphenylphosphinyl) oxy) methyl ketones as irreversible inhibitors of ICE, such as:

Graybill et al. (Bioorg Med. Chem. Lett., 7,41-46,1997) report the preparation of a- ((tetronoyl) oxy)-and a-((tetramoyl) oxy) methyl ketones as inhibitors of ICE, such as:

Semple et al. (Bioorg. Med. Chem. Lett., 8,959-964,1998) report the preparation of peptidomimetic aminomethylene ketones as inhibitors of ICE, such as: Summary of the Invention The invention relates to compound of Formulae I, II and III:

wherein R, is an optionally substituted alkyl or hydrogen; R3 is an N-protecting group; R2 is hydrogen or optionally substituted alkyl; A is CR6 or nitrogen; B is CR, or nitrogen; C is CRg or nitrogen; D is CR9 or nitrogen; provided that not more than two of A, B, C or D is nitrogen; and R6-R9 independently are hydrogen, halo, C,-C haloalkyl, C6-C, o aryl, C4-C7 cycloalkyl, C,-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C6-C, o aryl (C,-C6) alkyl, C6-CI0 aryl (C2-C6) alkenyl, C6-CI0 aryl (C2-C6) alkynyl, C,-C6 hydroxyalkyl, nitro, amino, cyano, C,-C6 acylamino, hydroxy, C,-C6 acyloxy, C,-C6 alkoxy, alkylthio, or carboxy; or one of R6 and R7, or R. and R8, or R, and R9 are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle; E is CR, 4, nitrogen, oxygen or sulfur;

F is CR, 5, nitrogen, oxygen or sulfur; G is C, 6, nitrogen, oxygen or sulfur; provided that only one of E, F, G is nitrogen, oxygen or sulfur; where R, 4-R, 6 are independently hydrogen, halo, C,-C6 haloalkyl, C6-C, 0 aryl, C4-C7 cycloalkyl, Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cg-Cjo aryl (C,-C6) alkyl, C6-C, o aryl (C2-C6) alkenyl, C6-C, o aryl (C2-C6) alkynyl, Cl-C6 hydroxyalkyl, nitro, amino, cyano, Cl-C6 acylamino, hydroxy, Cl-C6 acyloxy, C,-C6 alkoxy, alkylthio, or carboxy; or one of R, 4 and Rl5, or R, 5 and R, 6, are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle; Q represents an optionally substituted saturated or partially saturated carbocycle or heterocycle; X is a peptide of 1-4 amino acids or a bond; and Y is a peptide of 1-4 amino acids or a bond.

The invention relates to the discovery that the compounds represented by Formulae I, II and III are inhibitors of caspases. The invention also relates to the use of the compounds of the invention for reducing, preventing or treating maladies in which apoptotic cell death is either a causative factor or a result. Examples of uses for the present invention include protecting the nervous system following focal ischemia and global ischemia; treating neurodegenerative disorders such as Alzheimer's disease, Huntington's Disease, prion diseases, Parkinson's Disease, multiple sclerosis, amyotrophic lateral sclerosis, ataxia, telangiectasia, and spinobulbar atrophy; treating heart disease including myocardial infarction, congestive heart failure and cardiomyopathy; treating retinal disorders; treating autoimmune disorders including lupus erythematosus, rheumatoid arthritis, type I diabetes, Sjögren's syndrome and glomerulonephritis; treating polycystic kidney disease and anemia/erythropoiesis; treating immune system disorders, including AIDS and SCIDS; treating or ameliorating sepsis or multi-organ failure in an animal; reducing or preventing cell, tissue and organ damage during transplantation; reducing or preventing cell line death in industrial biotechnology; reducing or preventing alopecia (hair loss); reducing the premature death of skin cells;

treating or ameliorating apoptotic cell death in acute pancreatitus; treating or preventing the inflammatory response in psoriasis or inflammatory bowel disease; and treating or ameliorating organ apoptosis after burn injury.

The present invention provides pharmaceutical compositions comprising a compound of Formulae I, II and III in an effective amount to reduce apoptotic cell death in an animal.

The present invention also provides preservation or storage solutions for mammalian organs or tissue, or growth media for mammalian or yeast cells, wherein an effective amount of a compound of Formula I, II and III is included in said solutions or media in order to reduce apoptotic cell death in said organs, tissue or cells.

The invention also relates to the use of caspase inhibitors for treating, ameliorating, and preventing non-cancer cell death during chemotherapy and radiation therapy and for treating and ameliorating the side effects of chemotherapy and radiation therapy of cancer.

In particular, the invention relates to a method of treating, ameliorating or preventing oral mucositis, gastrointestinal mucositis, bladder mucositis, proctitis, bone marrow cell death, skin cell death and hair loss resulting from chemotherapy or radiation therapy of cancer in an animal, comprising administering to the animal in need thereof an effective amount of a caspase inhibitor.

Detailed Description of the Invention The inhibitors of caspases and apoptotic cell death of the present invention are compounds having the general Formulae I, II and III:

or pharmaceutically acceptable salts or prodrugs thereof, wherein: R, is an optionally substituted alkyl or hydrogen; R3 is a N-protecting group including t-butyloxycarbonyl, acetyl, and benzyloxycarbonyl; R2 is hydrogen or optionally substituted alkyl; A is CR6 or nitrogen; B is CR7 or nitrogen; C is CR8 or nitrogen; D is CR, or nitrogen; provided that not more than two of A, B, C or D is nitrogen; and R6-R9 independently are hydrogen, halo, C,-C6 haloalkyl, C6-C10 aryl, C4-C7 cycloalkyl, C,-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C6-CI0 <BR> <BR> aryl (C,-C6) alkyl, C6-C, o aryl (C2-C6) alkenyl, C6-C1o aryl (C2-C6) alkynyl, C,-C6 hydroxyalkyl, nitro, amino, cyano, Cl-C6 acylamino, hydroxy, Cl-C6 acyloxy, Cl-C6 alkoxy, alkylthio, or carboxy; or one of R6 and R7, or R7 and R8, or R8 and R, are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle; E is Cl4, nitrogen, oxygen or sulfur; F is Cl5, nitrogen, oxygen or sulfur; G is C, 6, nitrogen, oxygen or sulfur; provided that only one of E, F, G is nitrogen, oxygen or sulfur; where R, 4-RI6 are independently hydrogen, halo,

C,-C6 haloalkyl, C6-Cl0 aryl, C4-C7 cycloalkyl, C,-C6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C6-C, o aryl (Cl-C6) alkyl, C6-Cl0 aryl (C2-C6) alkenyl, C6-Cl0 aryl (C2- C6) alkynyl, Cl-C6 hydroxyalkyl, nitro, amino, cyano, C,-C6 acylamino, hydroxy, C,-C6 acyloxy, Cl-C6 alkoxy, alkylthio, or carboxy; or one of Rl4 and Rl5, or Rl, and R, 6 are taken together with the carbon atoms to which they are attached to form a carbocycle or heterocycle; Q represents an optionally substituted saturated or partially saturated carbocycle or heterocycle; X is a peptide of 1-4 amino acids or a bond; And Y is a peptide of 1-4 amino acids or a bond. Where X or Y is one amino acid, it may be any one of the common 20 amino acids e. g., Ala, Val, Leu, Ile, Pro, Phe, Trp, Met, Gly, Ser, Thr, Cys, Tyr, Asp, Asn, Glu, Asn, Lys, Arg and His.

Where X is a peptide, it may be Asp-Glu, Asp-Ala, Asp-Phe, Val-Glu, Leu- Glu, Thr-Glu, Ile-Glu, Tyr-Glu, Trp-Glu. Where Y is a peptide, it may be Glu-His, Glu-Ile, Glu-Thr, Glu-Val, Glu-Phe, Thr-His, Val-His, Ala-His and Glu-Pro.

With respect to R,, preferred alkyl groups are Cl 6 alkyl groups, e. g. methyl, ethyl, propyl, isopropyl, isobutyl, pentyl and hexyl groups; and substituted Cl 6alkyl groups, e. g. CH2OCH3 and CH2OCOCH3 (AM).

The invention relates to the discovery that the compounds represented by Formulae I, II and III are inhibitors of caspases. These inhibitors slow or block cell death in a variety of clinical conditions and industrial applications in which the loss of cells, tissues or entire organs occurs. Therefore, the invention is also related to methods of treating, preventing or reducing conditions in which apoptosis plays a role. These conditions are more fully described below.

The methods comprise administering to an animal in need of such treatment an inhibitor of the present invention, or a pharmaceutically acceptable salt or prodrug thereof, in an amount effective to inhibit apoptotic cell death.

Preferred embodiments of the compounds of Formulae I, II and III that may be employed as inhibitors of caspases are represented by Formula IV:

or pharmaceutically acceptable salts or prodrugs thereof wherein R, R2-, R6-R9 and X are as defined previously with respect to Formula I.

Examples of bridges formed by R6 and R,, or R, and R8, or R8 and Rg taken together are -OCH2O-, -OCF2O-, -(CH2)3-, -(CH2)4-, <BR> <BR> <BR> <BR> -OCH2CH20-,-CH2N (R, 3) CHZ,-CH2CH2N (R, 3)CH2-,<BR> <BR> <BR> <BR> <BR> -CH2N (R, 3) CHIzCH2 and-CH=CH-CH=CH-; where R13 is hydrogen, alkyl or cycloalkyl. <BR> <BR> <BR> <BR> <P>R, o is hydrogen, C,-C6 alkoxy, C,-C6 alkyl, Cl-C6 haloalkyl, C6-C, o aryl, C4-C7 cycloalkyl, C6-C, o aryl (C,-C6) alkyl, benzyloxy, substituted benzyloxy, or <BR> <BR> <BR> <BR> NRllRl2, wherein Rll and Rl2 independently are hydrogen, Cl-C6 alkyl, C,-C6<BR> <BR> <BR> <BR> <BR> haloalkyl, C6-C10 aryl, C4-C7 cycloalkyl, C6-C, o aryl (C,-C6) alkyl, or R,, and R, 2 are combined to form a heterocyclic ring system including pyrrolidine, piperidine, piperazine, or morpholine.

Preferred R, is H, Me, Et, t-Bu or AM. Preferred R2 is fluoromethyl, acyloxymethyl, arylacyloxymethyl and aminomethyl. Preferred R, o is benzyloxy and substituted benzyloxy. Preferred X is a peptide of 1-2 amino acids or a bond.

Exemplary preferred inhibitors of caspases having Formulae I-IV include, without limitation: 2- (Z-amino) benzoyl-Asp-fmk, <BR> <BR> <BR> 2- (Z-amino)-3-methylbenzoyl-Asp-fmk,<BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-3,5-dimethylbenzoyl-Asp-fmk,<BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-4-chlorobenzoyl-Asp-fmk,<BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-5-chlorobenzoyl-Asp-fmk,<BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-5-fluorobenzoyl-Asp-fmk,

2- (Z-amino)-6-fluorobenzoyl-Asp-fink, cis-2- (Z-amino)-cyclohexanecarboxyl-Asp-fmk, <BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-5-methylbenzoyl-Asp-fink,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-6-methylbenzoyl-Asp-fink,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-6-chlorobenzoyl-Asp-fink,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> 2- (Z-amino)-3-methoxybenzoyl-Asp-fink, 3- (Z-amino) thiophene-2-carboxyl-Asp-fmk, <BR> <BR> <BR> <BR> <BR> 3- (methoxycarbonylamino) thiophene-2-carboxyl-Asp-fmk,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> cis-2- (Z-amino) cyclopentanecarboxyl-Asp-fink,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> trans-2- (Z-amino) cyclopentanecarboxyl-Asp-fink, 2- (Z-amino) benzoyl-Asp-DCB-methylketone, methoxycarbonyl-Val- (2-aminobenzoyl)-Asp-fmk, Z-Glu- (2-aminobenzoyl)-Asp-fmk, and Z-Val-(2-aminobenzoyl)-Asp-fmk. where Z is benzyloxycarbonyl, fmk is fluoromethylketone and DCB is 2,6- dichlorobenzoyloxy.

Useful aryl groups are C6, 4 aryl, especially C6 l0 aryl. Typical C6 l4 aryl groups include phenyl, naphthyl, phenanthrenyl, anthracenyl, indenyl, azulenyl, biphenyl, biphenylenyl and fluorenyl groups.

Useful cycloalkyl groups are C38 cycloalkyl. Typical cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.

Useful saturated or partially saturated carbocyclic groups are cycloalkyl groups as defined above, as well as cycloalkenyl groups, such as cyclopentenyl, cycloheptenyl and cyclooctenyl.

Useful halo or halogen groups include fluorine, chlorine, bromine and iodine.

Useful alkyl groups include straight-chained and branched Cl l0 alkyl groups, more preferably C, _6 alkyl groups. Typical Cl, 0 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, 3-pentyl, hexyl and octyl groups. Also contemplated is a trimethylene group substituted on

two adjoining positions on the benzene ring of the compounds of the invention.

Useful arylalkyl groups include any of the above-mentioned Cl l0 alkyl groups substituted by any of the above-mentioned C6, 4 aryl groups. Useful values include benzyl, phenethyl and naphthylmethyl.

Useful haloalkyl groups include Cl l0 alkyl groups substituted by one or more fluorine, chlorine, bromine or iodine atoms, e. g. fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl, chloromethyl, chlorofluoromethyl and trichloromethyl groups.

Useful alkoxy groups include oxygen substituted by one of the Cl, 0 alkyl groups mentioned above.

Useful alkylthio groups include sulphur substituted by one of the C,., o alkyl groups mentioned above. Also included are the sulfoxides and sulfones of such alkylthio groups.

Useful acylamino groups are any Cl 6 acyl (alkanoyl) attached to an amino nitrogen, e. g. acetamido, propionamido, butanoylamido, pentanoylamido, hexanoylamido as well as aryl-substituted C2 6 substituted acyl groups.

Useful acyloxy groups are any C,. 6 acyl (alkanoyl) attached to an oxy (- O-) group, e. g. formyloxy, acetoxy, propionoyloxy, butanoyloxy, pentanoyloxy, hexanoyloxy and the like.

Useful arylacyloxy groups include any of the aryl groups mentioned above substituted on any of the acyloxy groups mentioned above, e. g. 2,6- dichlorobenzoyloxy, 2,6-difluorobenzoyloxy and 2,6-di- (trifluoromethyl)- benzoyloxy groups.

Useful amino groups include-NH2,-NHRIl, and-NR"R, 2, wherein R,, and R12are C I-10 alkyl or cycloalkyl groups as defined above.

Useful saturated or partially saturated heterocyclic groups include tetrahydrofuranyl, pyranyl, piperidinyl, piperizinyl, pyrrolidinyl, imidazolidinyl, imidazolinyl, indolinyl, isoindolinyl, quinuclidinyl,

morpholinyl, isochromanyl, chromanyl, pyrazolidinyl pyrazolinyl, tetronoyl and tetramoyl groups.

Useful heteroaryl groups include any one of the following: thienyl, benzo [b] thienyl, naphtho [2,3-b] thienyl, thianthrenyl, furyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxanthiinyl, 2H-pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, 4H-quinolizinyl, isoquinolyl, quinolyl, phthalzinyl, naphthyridinyl, quinozalinyl, cinnolinyl, pteridinyl, carbazolyl, p-carbolinyl, phenanthridinyl, acrindinyl, perimidinyl, phenanthrolinyl, phenazinyl, isothiazolyl, phenothiazinyl, isoxazolyl, furazanyl, phenoxazinyl, 1,4-dihydroquinoxaline-2,3-dione, 7-aminoisocoumarin, pyrido [1,2- a] pyrimidin-4-one, 1,2-benzoisoxazol-3-yl, benzimidazolyl, 2-oxindolyl and 2-oxobenzimidazolyl. Where the heteroaryl group contains a nitrogen atom in a ring, such nitrogen atom may be in the form of an N-oxide, e. g. a pyridyl N-oxide, pyrazinyl N-oxide, pyrimidinyl N-oxide and the like.

Optional substituents include one or more alkyl; halo; haloalkyl; cycloalkyl; aryl optionally substituted with one or more lower alkyl, halo, haloalkyl or heteroaryl groups; aryloxy optionally substituted with one or more lower alkyl, halo, haloalkyl or heteroaryl groups; aralkyl; heteroaryl optionally substitued with one or more lower alkyl, haloalkyl and aryl groups; heteroaryloxy optionally substitued with one or more lower alkyl, haloalkyl and aryl groups; alkoxy; alkylthio; arylthio; amino; acyloxy; arylacyloxy optionally substitued with one or more lower alkyl, halo alkyl and aryl groups; Diphenylphosphinyloxy optionally substituted with one or more lower alkyl, halo or haloalkyl groups; heterocyclo optionally substitued with one or more lower alkyl, halo alkyl and aryl groups; heterocycloalkyloxy optionally substitued with one or more lower alkyl, halo alkyl and aryl groups; partially unsaturated heterocycloalkyl optionally substitued with one or more lower alkyl, halo alkyl and aryl groups; or partially unsaturated heterocycloalkyloxy optionally substitued with one or more lower alkyl, halo alkyl and aryl groups.

Particular examples of such optional substituents that may be present at R2

include, without limitation, 3-pyrazolyloxy optionally substituted at the 2,4 and 5-positions with lower alkyl; 3- (l-phenyl-3-trifluoromethyl) pyrazolyloxy; 2,6-di (trifluoromethyl) benzoyloxy; 2,6-dimethylbenzoyloxy, pentafluoro- phenoxy; 2,6-dichlorobenzoyloxy; 2- (3- (2-imidazolyl) naphthyl) oxy; diphenylphosphinyloxy; tetronyloxy; and tetramoyloxy.

Certain of the compounds of the present invention may exist as stereoisomers including optical isomers. The invention includes all stereoisomers and both the racemic mixtures of such stereoisomers as well as the individual enantiomers that may be separated according to methods that are well known to those of ordinary skill in the art.

Examples of pharmaceutically acceptable addition salts include inorganic and organic acid addition salts such as hydrochloride, hydrobromide, phosphate, sulphate, citrate, lactate, tartrate, maleate, fumarate, mandelate and oxalate; and inorganic and organic base addition salts with bases such as sodium hydroxy and Tris (hydroxymethyl) aminomethane (TRIS, tromethane).

Examples of prodrugs include compounds of Formulae I-IV wherein R, is an alkyl group or substituted alkyl group such as CH2OCH3 and CH2OCOCH3 (AM ester).

The invention is also directed to a method for treating disorders responsive to the inhibition of caspases in animals suffering thereof. Particular preferred embodiments of compounds for use in the method of this invention are represented by previously defined Formulae I-IV.

The compounds of this invention may be prepared using methods known to those skilled in the art. Specifically, compounds with Formulae I-IV can be prepared as illustrated by exemplary reactions in Scheme 1. The intermediate 1 was prepared according to Revesz et al. (Tetrahedron Lett. 35, 9693-9696,1994). Coupling of 1 with a N-protected 2-aminobenzoic acid, which is either commercially available or which can be prepared from a commercially available 2-aminobenzoic acid, such as 2-Z-aminobenzoic acid, gave amide 2. Oxidation of 2 by Dess-Martin reagent according to Revesz et al. (Tetrahedron Lett. 35,9693-9696,1994) gave 3. Acid catalyzed cleavage of the ester gave the free acid 4.

Scheme 1 ci ? Y OH "1 TT" / + \ OH O NH O NH2° O O OH HN OH N F *Co2B.-t Dess-Martin 01 NH O COZBu-t EDCI, HOBT, DMAP O 2 O O N F \ I N F "TUA" 0 NH 0 0 NH 0 Xzo C02Bu-t ar C02H \ O \ O 3 4

An important aspect of the present invention is the discovery that compounds having Formulae I-IV are inhibitors of caspases. Therefore, these inhibitors are expected to slow or block cell death in a variety of clinical conditions in which the loss of cells, tissues or entire organs occurs.

The cell death inhibitors of the present invention can be used to reduce or prevent cell death in the nervous system (brain, spinal cord, and peripheral nervous system) under various conditions of ischemia and excitotoxicity, including, but not limited to, focal ischemia due to stroke and global ischemia due to cardiac arrest, as well as spinal cord injury (Emery et al. J.

Neurosurgery, 89. 911-920 (1998)). One particular usage is to treat the effects of oxygen deprivation which can occur during the birth of infants in high-risk labors or drowning. The cell death inhibitors can also be used to reduce or prevent cell death in the nervous system due to traumatic injury (such as head trauma), viral infection or radiation-induced nerve cell death (for example, as a side-effect of cancer radiotherapy). The cell death inhibitors can also be used

to reduce or prevent cell death in a range of neurodegenerative disorders, including but not limited to Alzheimer's disease (Mattson et al. Brain Res.

807: 167-176 (1998)), Huntington's Disease, Parkinson's Disease, multiple sclerosis, amyotrophic lateral sclerosis, and spinobulbar atrophy. The in vivo neuroprotective properties of cell death inhibitors of the invention can be tested in a rat transient focal brain ischemia model (Xue et al., Stroke 21: 166 (1990)). The cell death inhibitors may also be used to treat or ameliorate cell death in acute bacterial meningitis.

The cell death inhibitors of the invention can be used to reduce or prevent cell death in any condition which potentially results in the death of cardiac muscle (Black et al., J. Mol. Cel. Card. 30 : 733-742 (1998) and Maulik et al. Free Radic. Biol. Med. 24 : 869-875 (1998)). This includes myocardial infarction due to myocardial ischemia and reperfusion, congestive heart failure and cardiomyopathy. One particular application is to reduce or prevent myocardial cell death as occurs in certain viral infections of the heart.

The in vivo activity of the cell death inhibitors of the invention can be tested using the"mouse liver apoptosis"model described by Rodriguez et al.

(Rodriguez et al., J. Exp. Med., 184: 2067-2072 (1996)). In this model, mice are treated intravenously (IV) with an antiFas antibody which induces massive apoptosis in the liver and other organs, leading to generalized organ failure and death. This model is useful for indirectly testing the systemic bioavailability of the cell death inhibitors of the invention, as well as their in vivo anti-apoptotic properties. The cell death inhibitors of the invention therefore can be used to reduce or prevent apoptosis of liver cells (Jones et al.

Hepatology 27: 1632-42 (1998)) such as in sepsis (Jaeschke et al. J. Immunol.

160 : 3480-3486 (1998)) and hereditary tyrosinemia type 1 (HT1) (Kubo et al.

Prov. Natl. Acad. Sci. USA, 95 : 9552-9557 (1998). The cell death inhibitors of the invention also can be used to treat hepatitis (Suzuki, Proc. Soc. Exp. Biol.

Med. 217 : 450-454 (1998)); treat or ameliorate apoptotic cell death in acute pancreatitus; and treat or ameliorate organ apoptosis after burn injury.

The cell death inhibitors of the invention can be used to reduce or prevent cell death of retinal neurons (Kermer et al. J. Neurosci. 18: 4656-4662

(1998) and Miller et al. Am. J. Vet. Res. 59 : 149-152 (1998)) as can occur in disorders which increase intraocular pressure (such as glaucoma) or retinal disorders associated with the aging process (such as age-related macular degeneration). The inhibitors can also be used to treat hereditary degenerative disorders of the retina, such as retinitis pigmentosa.

The cell death inhibitors of the invention can also be used to reduce or prevent cell death in the kidney. This includes renal amyloidosis (Hiraoka et al. Nippon Jinzo Gakkai Shi, 40 : 276-83 (1998)), acute renal failure (Lieberthal et al. Semin Nephrol. 18: 505-518 (1998)), murine tubular epithelial cell death induced by cyclosporine A (Ortiz et al. Kidney International Supp. 68 : S25-S29 (1998)) and HIV-induced nephropathy (Conaldi et al. J Clin. Invest. 102 : 2041-2049 (1998)).

The cell death inhibitors of the invention can also be used to reduce or prevent cell death of buccal mucosa due to chronic alcohol ingestion (Slomiany et al. Biochem. Mol. Biol. Int. 45 : 1199-1209 (1998)).

The cell death inhibitors of the invention can also be used to reduce or prevent cell death in plants (Richberg et al. Curr. Opin. Plant Biol. l : 480-485 (1998)), such as plant cell death due to pathogens (Pozo et al. Curr. Biol. 8.

1129-1132 (1998) and Greenberg et al. Cell, 77: 551-563 (1994)).

The cell death inhibitors of the invention can also be used to reduce or prevent cell death due to radiation and ultraviolet-irradiation (Sheikh et al.

Oncogene, 17 : 2555-2563 (1998)).

The cell death inhibitors of the invention can also be used to reduce or prevent apoptotic death of bone marrow cells in myelodysplastic syndromes (MDS) (Mundle et al., Am. J. Hematol. 60: 36-47 (1999)).

The cell death inhibitors of the invention can also be used to reduce or prevent premature death of cells of the immune system, and are particularly useful in treating immune deficiency disorders, such as acquired immune deficiency syndrome (AIDS), severe combined immune deficiency syndrome (SCIDS) and related diseases. The cell death inhibitors can also be used to treat radiation-induced immune suppression.

Transplantation of human organs and tissues is a common treatment for organ failure. However, during the transplantation process, the donor organ or tissue is at risk for cell death since it is deprived of its normal blood supply prior to being implanted in the host. This ischemic state can be treated with cell death inhibitors by infusion into the donor organ or tissue, or by direct addition of the cell death inhibitors to the organ/tissue storage medium.

Cell death inhibitors may also be used to reduce or prevent cell death in the donor organ/tissue after it has been transplanted to protect it from the effects of reperfusion injury and/or effects of host immune cells which kill their targets by triggering apoptosis. The cytoprotective effects of cell death inhibitors can also be used to prevent the death of human or animal sperm and eggs used in in vitro fertilization procedures. These inhibitors can be used during the harvesting process and can also be included in the storage medium.

Mammalian cell lines, insect cells and yeast cells are commonly used to produce large amounts of recombinant proteins (such as antibodies, enzymes or hormones) for industrial or medicinal use. The lifespan of some of these cell lines is limited due to growth conditions, the nature of the recombinant molecule being expressed (some are toxic) and other unknown factors. The lifespans of industrial cell lines can be extended by including these cell death inhibitors in the growth medium in a concentration range of 1- 100 1M.

The factors governing hair growth and loss are largely unknown.

There is some evidence, however, that hair follicle regression (referred to as catagen) may be due at least partially to apoptosis. Therefore, it is contemplated that the cell death inhibitors of the present invention can be used to treat hair loss that occurs due to various conditions, including but not limited to male-pattern baldness, radiation-induced or chemotherapy-induced hair loss, and hair loss due to emotional stress. There is also evidence that apoptosis may play a role in the loss of hair color. Therefore, it is contemplated that the cell death inhibitors of the present invention can also be used in treating or preventing cases of premature graying of the hair.

The death of skin epithelial cells can occur after exposure to high levels of radiation, heat or chemicals. It is contemplated that the cell death inhibitors of the present invention can be used to treat, reduce or prevent this type of skin damage. In one particular application, the cell death inhibitors can be applied as part of a topical formulation, e. g. an ointment, to treat acute over-exposure to the sun and to prevent blistering and peeling of the skin.

Goldberg et al. (Nature Genetics 13: 442-449 (1996)) reported recently that huntingtin, a protein product of Huntington's disease (HD) gene, can be cleaved by CPP32 but not ICE. The mutation underlying HD is an expansion of a CAG trinucleotide at the 5'end of the HD gene. The trinucleotide expansion exceeding 36 repeats is associated with the clinical presentation of HD. The CAG expansion promotes cleavage of huntingtin by CPP32, thus links the role of CPP32 in the apoptotic cell death in HD. Compounds of the present invention with CPP32 inhibiting activity will be useful in blocking CPP32 induced apoptotic cell death, thus in preventing and treating HD and other disorders characterized by expansion of trinucleotide repeats such as myotonic dystrophy, fragile X mental retardation, spinobulbar muscular atrophy, spinocerebellar atoxia type I and Dentato-Rubro pallidoluysian atrophy.

The invention relates to a method of treating, ameliorating or preventing oral mucositis, gastrointestinal mucositis, bladder mucositis, proctitis, bone marrow cell death, skin cell death and hair loss resulting from chemotherapy or radiation therapy of cancer in an animal, comprising administering to the animal in need thereof an effective amount of a caspase inhibitor.

When animals are treated with chemotherapeutic agents and/or radiation to kill cancer cells, an unwanted side effect is the apoptotic death of rapidly dividing non-cancer cells. Such non-cancer cells include cells of the gastrointestinal tract, skin, hair, and bone marrow cells. According to the present invention, caspase inhibitors are administered to such non-cancer cells to prevent apoptosis of such cells. In a preferred embodiment, the caspase inhibitors are administered locally, e. g. to the gastrointestinal tract, mouth,

skin or scalp to prevent apoptosis of the gastrointestinal, mouth, skin or hair cells but allowing for the death of the cancer cells. Thus, in one example, it is possible to treat brain cancer with chemotherapy or radiation therapy and protect the outer skin, hair cells, gastrointestinal tract and bone marrow by local administration of a caspase inhibitor. In the case of oral mucositis, the caspase inhibitor can be applied, for example, in the form of a mouth wash or mouth rinse, in a gel, or in the form of an oral slow release lozenge to prevent activation of caspases and apoptotic cell death induced by the chemotherapeutic agent or by radiation. In the case of gastrointestinal mucositis, the caspase inhibitor can be applied in a form such that it is not absorbed systemically or in a form that coats the surface of the gastrointestinal tract, or a suppository formulation for the treatment of gastrointestinal mucositis. In the case of proctitis, the capsase inhibitor may be applied as part of an enema or suppository. In the case of bladder mucositis, the caspase inhibitor may be applied though a bladder catheter. For prevention of radiation or chemotherapy-induced hair loss, the caspase inhibitor can be applied, for example, to the scalp in the form of a hair rinse, hair gel, shampoo or hair conditioner. Importantly, the caspase inhibitor can be applied prior to the administration of the chemotherapeutic agent or radiation, thus preventing the onset of the damaging effects of the chemotherapeutic agent or radiation to the normal cells.

The cell death inhibitors of the present invention may also be used to treat or prevent the inflammatory response in psoriasis or inflammatory bowel disease.

Compositions within the scope of this invention include all compositions wherein the compounds of the present invention are contained in an amount which is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typically, the compounds may be administered to mammals, e. g. humans, orally at a dose of 0.0025 to 50 mg/kg, or an equivalent amount of the pharmaceutically acceptable salt thereof, per day of the body weight of the mammal being treated for apoptosis-

mediated disorders, e. g., neuronal cell death, heart disease, retinal disorders, polycystic kidney disease, immune system disorders and sepsis. Preferably, about 0.01 to about 10 mg/kg is orally administered to treat or prevent such disorders. For intramuscular injection, the dose is generally about one-half of the oral dose. For example, for treatment or prevention of neuronal cell death, a suitable intramuscular dose would be about 0.0025 to about 25 mg/kg, and most preferably, from about 0.01 to about 5 mg/kg.

The unit oral dose may comprise from about 0.01 to about 50 mg, preferably about 0.1 to about 10 mg of the compound. The unit dose may be administered one or more times daily as one or more tablets each containing from about 0.1 to about 10, conveniently about 0.25 to 50 mg of the compound or its solvates.

In a topical formulation, the compound may be present at a concentration of about 0.01 to 100 mg per gram of carrier. In a preferred embodiment, the compound is present at a concentration of about 0.07-1.0 mg/ml, more preferably, about 0.1-0.5 mg/ml, most preferably, about 0.4 mg/ml.

In addition to administering the compound as a raw chemical, the compounds of the invention may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the compounds into preparations which can be used pharmaceutically. Preferably, the preparations, particularly those preparations which can be administered orally or topically and which can be used for the preferred type of administration, such as tablets, dragees, slow release lozenges and capsules, mouth rinses and mouth washes, gels, liquid suspensions, hair rinses, hair gels, shampoos and also preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration by injection, topically or orally, contain from about 0.01 to 99 percent, preferably from about 0.25 to 75 percent of active compound (s), together with the excipient.

Also included within the scope of the present invention are the non- toxic pharmaceutically acceptable salts of the compounds of the present

invention. Acid addition salts are formed by mixing a solution of the particular cell death inhibitors of the present invention with a solution of a pharmaceutically acceptable non-toxic acid such as hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid, oxalic acid, and the like. Basic salts are formed by mixing a solution of the particular cell death inhibitors of the present invention with a solution of a pharmaceutically acceptable non-toxic base such as sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate Tris and the like.

The pharmaceutical compositions of the invention may be administered to any animal which may experience the beneficial effects of the compounds of the invention. Foremost among such animals are mammals, e. g., humans, although the invention is not intended to be so limited.

The caspase inhibitors and pharmaceutical compositions thereof may be administered by any means that achieve their intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired. In general, the caspase inhibitors are administered locally to the tissues that are to be protected from apoptosis and separately from the chemotherapeutic agent. For example, cisplatin may be administered by i. v. injection to treat a cancer such as brain, lung, breast, liver, kidney, pancreatic, ovarian, prostatic cancer, and the caspase inhibitor administered locally to treat, ameliorate, or prevent apototic cell death in the mouth or gastrointestinal tract, such as a mouth wash for the treatment of oral mucositis; and IV injectable aqueous solution for the treatment of bone marrow cell death; and an oral formulation suitable for coating the gastrointestinal surfaces or an emema or suppository formulation for the treatment of gastrointestinal mucositis including proctitis. The caspase inhibitors may also be applied through a bladder catheter for the treatment,

amelioration or prevention of bladder mucositis. Alternatively or concurrently, the caspase inhibitors may be applied topically to the skin and/or scalp to treat, ameliorate or prevent apoptotic cell death of hair and skin cells.

In a further embodiment, the chemotherapeutic agent or radiation may be applied locally to treat a localized cancer such as brain, lung, breast, liver, kidney, pancreatic, ovarian, prostatic cancer, and the caspase inhibitor administered systemically, e. g. by i. v. injection, to treat, ameliorate or prevent apoptotic cell death of the gastrointestinal tract cells, mouth epithelial cells, bone marrow cells, skin cells and hair cells. In the case of oral mucositis in brain cancer treatment, for example, a caspase inhibitor that does not penetrate the blood-brain barrier can be applied, for example, systemically by i. v. injection followed by irradiation of the brain tumor. This would protect the oral mucosa from the harmful effects of radiation but the caspase inhibitor would not protect the brain tumor from the therapeutic effects of radiation.

Importantly, the caspase inhibitor can be applied prior to administration of the radiation, thus preventing the onset of the damaging effects of the radiation to the normal mucosa cells.

The pharmaceutical preparations of the present invention are manufactured in a manner which is itself known, for example, by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. Thus, pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.

Suitable excipients are, in particular, fillers such as saccharides, for example lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, as well as binders such as starch paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added such as the above-mentioned starches and also carboxymethyl-starch, cross-

linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries are, above all, flow-regulating agents and lubricants, for example, silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropymethyl-cellulose phthalate, are used.

Dye stuffs or pigments may be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.

Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain the active compounds in the form of granules which may be mixed with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin. In addition, stabilizers may be added.

Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of one or more of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.

Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-

soluble salts and alkaline solutions. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered.

Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides or polyethylene glycol-400 (the compounds are soluble in PEG-400). Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers.

In accordance with one aspect of the present invention, compounds of the invention are employed in topical and parenteral formulations and are used for the treatment of skin damage, such as that caused by exposure to high levels of radiation, including ultraviolet radiation, heat or chemicals.

One or more additional substances which have therapeutic effects on the skin may also be incorporated in the compositions. Thus, the composition may also contain one or more compounds capable of increasing cyclic-AMP levels in the skin. Suitable compounds include adenosine or a nucleic acid hydrolysate in an amount of about 0.1-1% and papaverine, in an amount of about 0.5-5%, both by weight based on the weight of the composition. Also suitable are P-adrenergic agonists such as isoproterenol, in an amount of about 0.1-2% or cyclic-AMP, in an amount of about 0.1-1%, again both by weight based on the weight of the composition. Other suitable types of additional active ingredients which may be incorporated in the compositions of this invention include any compounds known to have a beneficial effect on skin.

Such compounds include retinoids such as Vitamin A, in an amount of about 0.003-0.3% by weight and chromanols such as Vitamin E or a derivative thereof in an amount of about 0.1-10% by weight, both based on the weight of the composition. Additionally, anti-inflammatory agents and keratoplastic agents may be incorporated in the cosmetic composition. A typical anti- inflammatory agent is a corticosteroid such as hydrocortisone or its acetate in an amount of about 0.25-5% by weight, or a corticosteroid such as dexamethasone in an amount of about 0.025-0.5% by weight, both based on

the weight of the composition. A typical keratoplastic agent is coal tar in an amount of about 0.1-20% or anthralin in an amount of about 0.05-2% by weight, both based on the weight of the composition.

The topical compositions of this invention are formulated preferably as oils, creams, lotions, ointments and the like by choice of appropriate carriers.

Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C, 2). The preferred carriers are those in which the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Additionally, transdermal penetration enhancers can be employed in these topical formulations. Examples of such enhancers can be found in U. S. Patent Nos. 3,989,816 and 4,444,762.

Creams are preferably formulated from a mixture of mineral oil, self- emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of an oil such as almond oil, is admixed. A typical example of such a cream is one which includes about 40 parts water, about 20 parts beeswax, about 40 parts mineral oil and about 1 part almond oil.

Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil such as almond oil with warm soft paraffin and allowing the mixture to cool. A typical example of such an ointment is one which includes about 30% almond oil and about 70% white soft paraffin by weight.

Lotions may be conveniently prepared by dissolving the active ingredient, in a suitable high molecular weight alcohol such as propylene glycol or polyethylene glycol.

In addition, these compositions may include other medicinal agents, growth factors, wound sealants, carriers, etc., that are known or apparent to those skilled in the art. The compositions of the invention are administered to a warm-blooded animal, such as human, already suffering from a skin damage, such as a burn, in an amount sufficient to allow the healing process to proceed

more quickly than if the host were not treated. Amounts effective for this use will depend on the severity of the skin damage and the general state of health of the patient being treated. Maintenance dosages over a prolonged period of time may be adjusted as necessary. For veterinary uses, higher levels may be administered as necessary.

In the case of an animal suffering from decreased hair growth, the compositions of the invention are administered in an amount sufficient to increase the rate of hair growth. Amounts effective for this use will depend on the extent of decreased hair growth, and the general state of health of the patient being treated. Maintenance dosages over a prolonged period of time may be adjusted as necessary. For veterinary uses, higher levels may be administered as necessary.

When the compounds are to be administered to plants, they may be applied to the leaves and/or stems and/or flowers of the plant, e. g. by spraying.

The compounds may be spayed in particulate form or dissolved or suspended in an appropriate carrier, e. g. in water or an oil-water emulsion. The compounds may also be combined with the soil of the plant. In this embodiment, the compounds are taken up by the roots of the plant.

In a preferred embodiment, the caspase inhibitor is formulated as part of a mouthwash for the treatment, amelioration or prevention of oral mucositis. Such mouthwashes are aqueous solutions of the caspase inhibitor which may also contain alcohol, glycerin, synthetic sweeteners and surface- active, flavoring and coloring agents. They may also contain anti-infective agents such as hexetidine and cetylpyridinium chloride. The mouthwashes may also contain topical anesthetics (e. g. benzocaine, cocaine, dyclonine hydrochloride, lidocaine, proparacaine hydrochloride or teracaine hydrochloride), for example, for relieving pain of radiation or chemotherapy- induced sores. The mouth washes may have either acidic or basic pH. See Remington's Pharmaceutical Sciences, A. R. Gennaro (ed.), Mack Publishing Company, pp. 1045,1046,1526 and 1965 (1990).

In another preferred embodiment, the caspase inhibitor is formulated as an oral formulation which is capable of coating the gastrointestinal surfaces

for the treatment, amelioration or prevention of gastrointestinal mucositis.

Examples of gastrointestinal mucositis include esophageal mucositis, gastric mucositis, and intestinal mucositis. Such formulations may comprise gastric antacids such as aluminum carbonate, aluminum hydroxide gel, bismuth subnitrate, bismuth subsalicylate, calcium carbonate, dihydroxyaluminum sodium carbonate, magaldrate, magnesium carbonate, magnesium hydroxide, magnesium oxide, sodium bicarbonate, milk of bismuth, dihydroxyaluminum aminoacetate, magnesium phosphate, magnesium trisilicate and mixtures thereof. Other additives include without limitation H2-receptor antagonists, digestants, anti-emetics, adsorbants, and miscellaneous agents. See Remington's Pharmaceutical Sciences, A. R. Gennaro (ed.), Mack Publishing Company, pp. 774-778 (1990).

Chemotherapy agents such as cisplatin and radiation therapy often induce early and late onset emesis in the patient. Thus, in one embodiment an antiemetic is coadminstered together with the caspase inhibitor to avoid emesis and retain contact of the caspase inhibitor with the gastrointestinal tract. Examples of such antiemetics include without limitation compounds that block the dopaminergic emetic receptors such as metoclopramide and trimethobenzamide, and cannabinoids. Metoclopramide may be administered orally prior to and/or during chemotherapy/radiation therapy/caspase inhibitor therapy to prevent the early emesis response and then later by intranasal administration according to U. S. Patent Nos. 5,760,086 and 4,536,386 to prevent delayed onset emesis. During the period after chemotherapy/radiation therapy, both the caspase inhibitor and the antiemetic may be coadministered to treat, ameliorate or prevent gastrointestinal mucositis.

In a further embodiment, the caspase inhibitor may be formulated as an IV injectable solution for the treatment, amelioration or prevention of bone marrow cell death.

The compositions may be administered to a warm-blooded animal, such as human, already suffering from chemotherapy or radiation therapy- induced non-cancer cell death, or, more preferably, before or during therapy with chemotherapy or radiation.

The following examples are illustrative, but not limiting, of the method and compositions of the present invention. Other suitable modifications and adaptations of the variety of conditions and parameters normally encountered in clinical therapy and which are obvious to those skilled in the art are within the spirit and scope of the invention. <BR> <BR> <BR> <BR> <BR> <BR> <BR> <P> Example 1<BR> <BR> <BR> <BR> 2-(Z-Amino) benzoyl-Asp-fmk Step A. 2-Z-Aminobenzoic acid. To a solution of 2-aminobenzoic acid (0.30 g, 2.2 mmol) in pyridine (2 mL) was added benzyl chloroformate (0.6 mL, 4.2 mmol) at 0 °C. The mixture was then stirred at room temperature for 1 h, diluted with EtOAc (50 mL), washed with 2N HCI, water and brine, dried over Na2SO4 and concentrated in vacuo. The crude solid was washed with 4: 1 hexane/EtOAc twice and dried in vacuo to yield the title compound as a white solid (270 mg, 1.0 mmol, 45%).'H NMR (DMSO-d3): 8 11.49 (br s, 1H), 8.22 (d, J = 7.5,1 H), 7.95 (d, J = 7.5,1H), 7.51 (t, J = 7.5,1H), 7.39-7.32 (m, 5 H), 7.05 (d, J = 7.5,1 H), 5.15 (s, 2 H).

Step B. tert-Butyl S-fluoro-3-[2-Z-aminobenzoylamido./-4- hydroxypentanoate. A mixture of 2-Z-aminobenzoic acid (90 mg, 0.33 mmol), 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI, 61 mg, 0.40 mmol), 1-hydroxybenzotriazole hydrate (HOBT, 73 mg, 0.38 mmol), dimethylaminopyridine (DMAP, 22 mg, 0.18 mmol) and tert-butyl 3-amino-5- fluoro-4-hydroxypentanoate (79 mg, 0.38 mmol) in THF (10 mL) was stirred at room temperature for 20 h, diluted with 1: 1 hexane/EtOAc (75 mL), washed with water, 2N HCI, water, 2N NaOH and brine, dried over Na2SO4 and cocentrated in vacuo. The residue was purified by chromatography (3: 2 Hexane/EtOAc) to yield the title compound as a yellow hydroscopic solid (52 mg, 0.11 mmol, 3 3%).

Step C. 2- (Z-Amino) benzoyl-Asp (OBu-t)-fmk. A mixture of periodinane (0.41 g, 0.97 mmol) and tert-butyl 5-fluoro-3- [2-Z- aminobenzoylamido]-4-hydroxypentanoate (52 mg, 0.11 mmol) in

dichloromethane (15 mL) was refluxed for 20 h, cooled to room temperature, and 25 mL of saturated sodium bicarbonate aqueous solution containing 1.0 g of Na2S203was added. The resulting mixture was stirred for 2 h, diluted with 1: 1 hexane/EtOAc (75 mL), washed with water and brine, dried over Na2SO4 and concentrated in vacuo. The residue was purified by chromatography (3: 2 hexane/EtOAc) to yield the title compound as a hydroscopic yellow solid (45 mg, 0.10 mmol, 91%).'H NMR (CDC13): 8 10.48 (s, 1H), 8.42 (d, J = 8.7, 1H), 7.51-7.33 (m, 7H), 7.07 (m, 1 H), (m, 1H), 5.21 (s, 2H), 3.11- 2.83 (m, 2 H), 1.44 (s, 9 H).

Step D. 2- (Z-Amino) benzoyl-Asp-fink To a solution of 2- (Z- Amino) benzoyl-Asp (OBu-t)-fmk (45 mg, 0.10 mmol) in 5 mL of CH2Cl2 was added 1 mL of TFA. The resulting solution was allowed to stir at rt for 1 hr, diluted with EtOAc (75 ml), washed with water, aqueous Na2HPO4 to pH 5 and then brine, dried over Na2SO4 and concentrated in vacuo to give the title compound as a white solid (15 mg, 0.037 mmol, 38%).'H NMR (DMSO- d6): 6 10.63 (s, 1H), 9.08 (s, 1 H), 8.14 (d, J = 7.8,1 H), 7.76 (d, J = 7.8,1 H), 7.54 (t, J = 7.8,1 H), 7.41-7.35 (m, 5 H), 7.15 (t, J = 7.8,1H), 5.16 (s, 2H), 5.26-4.95 (m, 2H), 4.83 (m, 1H), 3.00-2.64 (m, 2 H).

Example 2 2- (Z Amino)-6-methylbenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example 1 from 2-amino-6-methylbenzoic acid.'H NMR (DMSO-d6): 8 8.85 (s, 1H), 8.68 (s, 1H), 7.49-7.01 (m, 8H), 5.12 (s, 2H), 4.82 (m, 1H), 5.26-4.95 (m, 2H), 3.00-2.64 (m. 2H), 2.26 (s, 3H).

Example 3 2- (Z-Amino)-5-methylbenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example <BR> <BR> <BR> 1 from 2-amino-5-methylbenzoic acid.'H NMR (DMSO-d6): 6 10.48 (s, 1H),

9.10 (d, J = 9,1H), 8.00 (d, J = 8.7,1H), 7.89 (s, 1H), 7.58 (s, 1H), 7.41-7.34 (m, 5H), 5.14 (s, 2H), 4.83 (m, 1H), 5.39-4.41 (m, 2H), 2.94-2.80 (m. 2H), 2.30 (s, 3H).

Example 4 2- (Z-Amino)-3-methylbenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example <BR> <BR> <BR> 1 from 2-amino-3-methylbenzoic acid.'H NMR (DMSO-d6): 8 9.05 (s, 1H), 8.73 (s, 1H), (m, 8H), 5.08 (s, 2H), 4.72 (m, 1H), 5.32 (bs, 2H), 2.81-2.66 (m. 2H), 2.21 (s, 3H).

Example 5 2- (Z Amino)-3-methoxybenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example <BR> <BR> <BR> 1 from 2-amino-3-methoxybenzoic acid.'H NMR (DMSO-d6): 8 8.73 (bs, 1H), 8.64 (bs, 1H), 7.57-7.06 (m, 9H), 5.06 (s, 2H), 4.72 (bs, 1H), 5.26- 4.97 (m, 2H), 3.78 (s, 3H), 2.78-2.66 (m. 2H).

Example 6 2- (Z-Amino)-5-fluorobenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example <BR> <BR> <BR> 1 from 2-amino-5-fluorobenzoic acid.'H NMR (DMSO-d6): 6 10.40 (bs, 1H), 9.13 (bs, 1H), 8.07 (q, J = 5.1,1H), 7.61 (d, J = 6.6,1H), 7.46-7.33 (m, 6H), 5.15 (s, 2H), 4.81 (bs, 1H), 2.84-2.72 (m. 2H).

<BR> <P> Example 7<BR> <BR> <BR> <BR> cis-2-(Z-Amino) cyclohexanecarboxyl-Asp-fmk The title compound was prepared in four-steps as described in Example 1 from cis-2-aminocyclohexanecarboxylic acid.'H NMR (DMSO-d6): 8 8.28 (bs, 1H), 7.39-7. 09 (m, 5H), 4.98 (s, 2H), 4.52-4.45 (m, 1H), 3.99 (bs, 1H), 2.62-2.53 (m, 4H), 1.77-1.23 (m, 8H).

Example 8 2-(Z-Amino)-3, 5-dimethylbenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example <BR> <BR> <BR> 1 from 2-amino-3,5-dimethylbenzoic acid.'H NMR (DMSO-d6): 8 8.05 (s, 1H), 7.42-7.17 (m, 8H), 5.15 (s, 2H), 5.19-5.03 (m, 2H), 4.87 (m, 1H), 2.30 (s, 3H), 2.26 (s, 3H).

Example 9 2- (Z-Amino)-5-chlorobenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example <BR> <BR> <BR> 1 from 2-amino-5-chlorobenzoic acid.'H NMR (DMSO-d6): 8 10.56 (s, 1H), 9.19 (s, 1H), 8.15 (d, J = 9.0,1H), 7.84 (s, 1H), 7.61 (d, J = 9.0,1H), 7.41-7.37 (m, 5H), 5.16 (s, 2H), 4.81 (m, 1H), 5.41-4.80 (m, 2H), 2.84-2.73 (m, 2H).

Example 10 2- (Z-Amino)-6-chlorobenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example 1 from 2-amino-6-chlorobenzoic acid.'H NMR (DMSO-d6): 8 9.17 (d, J = 4.2,1H), 8.95 (s, 1H), 7.74-7.24 (m, 8H), 5.50-5.21 (m, 2H), 5.15 (s, 2H), 4.85-4.78 (m, 1H), 2.98-2.65 (m, 2H).

Example 11 2- (Z-Amino)-4-chlorobenzoyl-Asp-fmk The title compound was prepared in four-steps as described in Example 1 from 2-amino-3,5-dimethylbenzoic acid.'H NMR (DMSO-d6): 8 10.84 (s, 1H), 9.19 (s, 1H), 8.24 (s, 1H), 7.81 (d, J= 8.4,1H), 7.42-7.24 (m, 6H), 5.18 (s, 2H), 5.25-5.20 (m, 2H), 4.82 (m, 1H), 2.94-2.63 (m, 2H).

Example 12 3-(Z-Amino) thiophene-2-carboxyl-Asp-fmk Step A. 3- (Z-Amino) thiophene-2-carboxylic acid. The mixture of methyl 3-aminothiophene-2-carboxylate (0.2g, 1.27mmol) in 2N NaOH (lOmL) was heated at 90°C for 15 min, then cooled to 0°C. To the resulting solution was added benzyl chloroformate (1.5 mL, 10.5 mmol) and THF (10 mL). The mixture was then stirred at room temperature for lh, washed with 3: 1 hexane: ethyl acetate (2 x 15 mL). The aqueous phase was acidified with 2N HCl to pH-1-2, extracted with ethyl acetate (3 x 15 mL), washed with water and brine, dried over Na2SO4 and concentrated in vacuo to yield the title compound as a white solid (70 mg).

The title compound was then prepared in three steps as described in Example 1 (B-D).'H NMR (DMSO-d6): 8 10.43 (s, 1H), 8.74 (s, 1H), 7.81 (d, J = 5.4,1H), 7.73 (d, J = 5.4,1H), 7.44-7.35 (m, 5H), 5.18 (s, 2H), 5.32-5.04 (m, 2H), 4.79 (m, 1H), 2.88-2.67 (m, 2H). <BR> <BR> <BR> <BR> <BR> <BR> <P> Example 13<BR> <BR> <BR> <BR> <BR> <BR> 3- (Methoxycarbonylamino) thiophene-2-carboxyl-Asp-fmk The title compound was prepared in four-steps as described in Example 12 from methyl 3-aminothiophene-2-carboxylate and methyl chloroformate.

'H NMR (DMSO-d6): 6 10.34 (s, 1H), 8.70 (s, 1H), 7.79 (d, J = 5.7,1H), 7.72 (d, J = 5.7,1H), 5.31-4.80 (m, 3H), 3.70 (s, 3H), 2.96-2.73 (m, 2H).

Example 14 Cis-2- (Z-Amino) cyclopentanecarboxyl-Asp-fmk The title compound was prepared in four-steps as described in Example 1 from cis-2-aminocyclopentanecarboxylic acid.'H NMR (DMSO-d6): 8 8.35 (s, 1H), 7.34-7.28 (m, SH), 7.09 (m, 1H), 5.13-4.50 (m, 5H), 4.11 (m, 1H), 2.81 (m, 1H), 2.73-2.51 (m, 2H), 1.91-1.40 (m, 6H). <BR> <BR> <P> Example 15<BR> Trans-2- (Z-Amino) cyclohexanecarboxyl-Asp-fmk The title compound was prepared in four-steps as described in Example 1 from trans-2-aminocyclohexanecarboxylic acid.'H NMR (DMSO-d6): S 12.48 (s, 1H), 8.26-8.15 (m, 1H), 7.38-7.17 (m, 5H), 5.18-4.47 (m, 5H), 2.67- 2.50 (m, 2H), 2.19 (m, 1H), 1.83-1.06 (m, 10H).

Example 16 Z-Glu- (2-aminobenzoyl)-Asp-fmk<BR> <BR> Step A. Z-Glu (OBu-t)-2-aminobenzoic acid. To a solution of Z- Glu (OBu-t) OH (272 mg, 0.81 mmol) in THF (5 mL) was added N- methylmorpholine (110 pL, 1.1 mmol) at-45 °C, followed by isobutyl chloroformate (105 uL, 0.81 mmol). The mixture was stirred at-45 °C for 30 min. and a solution of anthranllic acid (127 mg, 0.93 mmol) in THF (5 mL) was added, followed by more N-methylmorpholine (200 u. L, 1.82 mmol). The resulting mixture was stirred overnight and the cooling bath was allowed to slowly warm to room temperature. After diluted with EtOAc (100 mL), the mixture was washed with 2N HCI, water, saturated NaHC03, water, 2N HCI, water and brine, dried over Na2SO4 and concentrated in vacuo to yield the

product as a highly hydroscopic white solid (330 mg, 0.72 mmol, 89%).'H NMR (DMSO-d6): 8 11.79 (s, 1H), 8.59 (d, J = 8.6,1H), 8.00 (t, J = 6.0,1H), 7.60 (t, J= 8.6,1H), 7.39-7.31 (m, 5H), 7.17 (t, J = 7.8,1H), 5.16-4.99 (m, 2H), 4.08 (m, 1H), 2.33 (m, 2H), 2.08-1.75 (m, 2H), 1.38 (s, 9H).

Step B. tert-Butyl 5 fluoro-3-Z Glu (OBu-t)- (2-aminobenzoyl) amidoJ- 4-hydroxypentanoate. A mixture of Z-Glu (OBu-t)-2-aminobenzoic acid (330 mg, 0.72 mmol), EDCI (129 mg, 0.67 mmol), HOBT (104 mg, 0.68 mmol), DMAP (46 mg, 0.38 mmol) and tert-butyl 3-amino-5-fluoro-4- hydroxypentanoate (136 mg, 0.66 mmol) in THF (6 mL) was stirred at room temperature for 20 h. After diluted with 1: 1 hexane/EtOAc (75 mL), the mixture was washed with water, 2N HCI, water, 2N NaOH and brine, dried over Na2SO4 and concentrated in vacuo. The residue was purified by chromatography (3: 1 then 3: 2 hexane/EtOAc) to yield the title compound as a white solid (45 mg, 0.068 mmol, 10%).

Step C. ZGlu (OBu-t)-[2-aminobenzoyl/-Asp (OBu-t)-fmk. The title compound was synthesized by a similar procedure as described in Step C, Example 1 in 58% yield.

Step D. Z-Glu- [2-aminobenzoyl7-Asp-fmk. The title compound was synthesized by a similar procedure as described in Step D, Example 1 in 14% <BR> <BR> <BR> yield.'H NMR (DMSO-d6): 8 11.46 (s, 1H), 9.18 (s, 1H), 8.57-7.20 (m, 6H), 5.36-4.84 (m, 5H), 4.04 (br s, 1H), 2.95-1.81 (m, 6H). <BR> <BR> <BR> <BR> <BR> <BR> <BR> <P> Example 17<BR> <BR> <BR> <BR> Z-Val-(2-Aminobenzoyl)-Asp-fmk The title compound was synthesized as described in Example 16 from <BR> <BR> <BR> Z-Val.'H NMR (DMSO-d6): 8 11.34-11.25 (m, 1H), 9.17-7.17 (m, 11H), 5.42-4. 30 (m, 5H), 3.95-3.75 (m, 1H), 2.95-2.57 (m, 2H), 1.92 (m, 1H), 0.91- 0.84 (m, 6H).

Example 18<BR> <BR> <BR> <BR> <BR> 2- (Z-Amino) benzoyl-Asp-DCB-methylketone Step A. Z-Asp (OBu-t)-DCB-methylketone To a solution of Z- Asp (OBu-t)-bromomethylketone (500 mg, 1.24 mmol) in DMF (10 ml) was added potassium fluoride (320 mg, 5.50 mmol), and 2,6-dichlorobenzoic acid (348 mg, 1.82 mmol). The mixture was stirred at room temperature for 12 h, and then was diluted with 25 ml of ethyl acetate, washed with aqueous NH4Cl and brine, dried over Na2SO4 and concentrated in vacuo. The title compound was obtained as white solid (0.78 g, 2.62 mmol, 69%).'H NMR (CDCl3): 7.34 (m, 8H), 5.96 (d, J = 8.7,1H), 5.21 (d, J = 6.6,2H), 5.16 (s, 2H), 4.70 (m, 1H), 2.88 (m, 2H), 1.27 (s, 9H).

Step B. Asp (OBu-t)-DCB-methylketone-HCl To a solution of Z- Asp (OBu-t)-DCB-methylketone (572 mg, 1.14 mmol) in ethanol (15 ml) was added Pd/C (50 mg) and 6N HCl (0.2 ml). The mixture was stirred at room temperature under H2 atmosphere (1 atm) for 12 h, then it was filtered and concentrated. The title compound was obtained as pale white solid (416 mg, 1.04 mmol, 90%).'H NMR (CDCl3): 7.27 (m, 3H), 5.28 (m, 2H), 4.94 (m, 1H), 3.27 (m, 2H), 1.42 (s, 9H).

Step C. 2- (Z-Amino) benzoyl-Asp (OBu-t)-DCB-methylketone To a solution of 2- (Z-amino) benzoic acid (140 mg, 0.52 mmol) in THF (5 mL) was added N-methylmorpholine (65 I1L, 0.59 mmol), followed by 2-methylpropyl chloroformate (70 L, 0.54 mmol) at-45 °C. After 30 min., a solution of Asp (OBu-t)-DCB-methylketone-HCl (121 mg, 0.27 mmol) in THF (5 mL) was added to the solution. The resulting mixture was further stirred overnight and the cooling bath was allowed to slowly warm to room temperature. It was then diluted with 1: 1 hexane/EtOAc (100 mL), washed with water, 2N NaOH and brine, dried over Na2SO4 and concentrated in vacuo. The residue was purified by chromatography (3: 1 hexane/EtOAc) to yield the title compound as a white solid (365 mg, 0.05 mmol, 19%).'H NMR (CDCL): 10.90 (s, 1H), 8.49 (d, J = 7.5,1H), 7.87 (dd, J = 8.1,1.8, lH), 7.56-7.32 (m, 9H), 7.08 (t, J =

6.9,1H), 5.23 (s, 2H), 5.28 (m, 1H), 4.90 (d, J = 1.8,2H), 3.16-2.91 (m, 1H), 1.43 (m, 9H).

Step D. 2- (Z-Amino) benzoyl-Asp-DCB-methylketone A solution of 2- (Z-amino) benzoyl-Asp (OBu-t)-DCB-methylketone (35 mg) and TFA (1 mL) in methylenechloride (3 mL) was stirred at room temperature for 2 h. The mixture was diluted with EtOAc (70 mL), washed with saturated Na2HPO4 to pH-5, and further washed with water, and brine, dried over Na2SO4 and concentrated in vacuo to yield the title compound as a white solid (10 mg, 0.016 mmol, 33%).'H NMR (CDCl3): 10.94 (s, 1H), 8.49 (d, J = 8.4,1H), 7.87 (dd, J = 8.1,1.5,1H), 7.53 (m, 1H), 7.45-7.33 (m, 8H), 7.07 (m, 1H), 5.22 (s, 2H), 5.10-5.07 (m, 1H), 4.90 (m, 2H), 3.11 (dd, J = 8.4,19.0,1H), 2.95 (dd, J= 1.8,19.0,1H)). <BR> <BR> <BR> <BR> <BR> <BR> <BR> <P> Example 19<BR> <BR> <BR> <BR> <BR> <BR> <BR> Methoxycarbonyl-Val-(2-aminobenzoyl)-Asp-fmk Step A. tert-Butyl 5 fluoro-3- (2-aminobenzoylamido)-4- hydroxypentanoate. HCl. A mixture of tert-butyl 5-fluoro-3- (2-Z- aminobenzoylamido)-4-hydroxypentanoate (80 mg, 0.174 mmol), Pd-C (23mg) and 6N HCl (0.087 mL) in ethanol (5 mL) was stirred under hydrogen atmosphere at room temperature for 2 h. The mixture was filtered and the solvent was evaporated to yield the title product. It was used in next step without further purification.

Step B. tert-Butyl 5-fluoro-3- (methoxycarbonyl-Val-2- aminobenzoylamido)-4-hydroxypentanoate. To a solution of methoxycarbonyl-Val-OH (31mg, 0.17mmol) in THF (5 mL) was added N- methylmorpholine (38 uL, 0.34 mmol) at-45°C, followed by isobutyl chloroformate (45 L, 0.034 mmol). The mixture was stirred at-45°C for 30 min and a solution of tert-butyl 5-fluoro-3-(2-aminobenzoylamido)-4- hydroxypentanoate. HCl in THF (5 mL) was added, followed by more N- methylmorpholine (50 pL, 0.45 mmol). The resulting mixture was stirred overnight and the cooling bath was allowed to slowly warm to room

temperature. After dilution with ethyl acetate (50 mL), the mixture was washed with water and brine, dried over Na2SO4 and concentrated in vacuo.

The residue was purified by chromatography (3: 2 hexane: ethyl acetate) to yield the title compound as a white hydroscopic solid (20 mg, 0.041 mmol, 24%).'H NMR (CDCl3): 8 11.36-11.24 (m, 1H), 8.54 (d, J = 8.7,1H), 7.55- 7.05 (m, 4H), 5.40 (m, 1H), 4.89-3.85 (m, 6H), 3.71 (d, J = 1.8,3H), 2.84-2.61 (m, 2H), 2.29 (m, 1H), 1.46-1.44 (m, 9H), 1.05-0.98 (m, 6H).

Step C, D. Methoxycarbonyl-Val- (2-aminobenzoyl)-Asp-fmk. The title compound was synthesized with a similar procedure as described in Step C <BR> <BR> <BR> and D of Example 1. IH NMR (DMSO-d6): 8 12.52 (s, 1H), 11.27 (d, J = 6.0, 1H), 9.18 (m, 1H), 7.88-7.54 (m, 4H), 7.20 (t, J = 7.5,1H), 5.39-4.44 (m, 3H), 3.83 (m, 1H), 3.57 (d, J = 2.4,3H), 2.96-2.65 (m, 2H), 2.18 (m, 1H), 0.92 (t, J = 6.3,6H).

Example 20 Enzyme Activity The activity of 2- (Z-amino) benzoyl-Asp-fmk as an inhibitor of caspase-3 was measured in a fluorometric enzyme assay. Enzyme activity was measured using synthetic peptide substrates attached to a fluorogenic leaving group. Cleavage of the synthetic substrate by the enzyme results in a fluorescent signal which is read in a spectrofluorometer or in a fluorometric microtiter plate reader.

12 concentrations of the testing compound ranged from 30 pM to 10 u. M were tested in the enzyme assay. The enzyme reaction was conducted in the presence of 2 ng rCaspase 3 (purchased from PharMingen, a Becton division company, San Diego, CA), various concentrations of testing compound, 10 uM caspase 3 substrate Ac-DEVD-AMC (purchased from Quality Controlled Biochemicals, Inc. Hopkinton, MA) and caspase buffer (20 mM PIPES, 100 mM NaCl, 10 mM DTT, I mM EDTA, 0.1 % CHAPS and 10 % sucrose, pH 7.2) in a total volume of 100 uL. The enzyme reaction was carried out in a 96-well plate and incubated at 37 °C for 30 minutes. The

plate was then read with a fluorescence plate reader (EG&G WALLAG 1420- 002) using excitation filter at 355nm/emission filter at 460 nm. The data was analyzed using GraphPrism software to give an ICSO value of 0.2 pM.

Having now fully described this invention, it will be understood by those of ordinary skill in the art that the same can be performed within a wide and equivalent range of conditions, formulations and other parameters without affecting the scope of the invention or any embodiment thereof. All patents, patent applications and publications cited herein are fully incorporated by reference herein in their entirety.