AU2005220705A1 - Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof - Google Patents

Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof Download PDF

Info

Publication number
AU2005220705A1
AU2005220705A1 AU2005220705A AU2005220705A AU2005220705A1 AU 2005220705 A1 AU2005220705 A1 AU 2005220705A1 AU 2005220705 A AU2005220705 A AU 2005220705A AU 2005220705 A AU2005220705 A AU 2005220705A AU 2005220705 A1 AU2005220705 A1 AU 2005220705A1
Authority
AU
Australia
Prior art keywords
hydrocarbon
saturated
unsaturated
aliphatic
heterocycle
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2005220705A
Inventor
James T. Dalton
Veeresa Gududuru
Eunju Hurh
Duane D. Miller
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ohio State University Research Foundation
Original Assignee
Ohio State University Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ohio State University Research Foundation filed Critical Ohio State University Research Foundation
Publication of AU2005220705A1 publication Critical patent/AU2005220705A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/34Oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/04Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/04Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D277/06Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Description

WO 2005/086638 PCT/US2005/004759 ANALOGS EXHIBITING INHIBITION OF CELL PROLIFERATION, METHODS OF MAKING, AND USES THEREOF The present application is a continuation-in-part and claims priority to and benefit of 5 U.S. Patent Application Serial No. 10/992,175, which claims the priority benefit of provisional U.S. Patent Application Serial No. 60/523,079, filed November 18, 2003, both of which are hereby incorporated by reference in their entirety. The present application claims priority to and benefit of provisional U.S. Patent Application Serial No. 60/543,724, filed February 11, 2004, and provisional U.S. Patent 10 Application Serial No. 60/555,803, filed March 24, 2004, both of which are hereby incorporated by reference in their entirety.. This application was made, at least in part, with funding received from the U.S. Department of Defense under grant DAMD 17-01-1-0830. The U.S. government may retain certain rights in this invention. 15 BACKGROUND Prostate cancer accounts for 33% of all newly diagnosed malignancies among men in the United States (American Cancer Society: Cancer Facts and Figures (2003)). According to the American Cancer Society, an estimated 230,110 men will be diagnosed with prostate 20 cancer in 2004, and 29,900 men will die of it (American Cancer Society: Cancer Facts and Figures (2004)). The incidence of prostate cancer varies worldwide, with the highest rates found in the United States, Canada, and Scandinavia, and the lowest rates found in China and other parts of Asia (Quilnn and Babb, "Patterns and Trends in Prostate Cancer Incidence, Survival, Prevalence and Mortality. Part: International Comparisons," BJU Int. 90:162-173 25 (2002); Gronberg, "Prostate Cancer Epidemiology," Lancet 361:859-864 (2003)). These differences are caused by genetic susceptibility, exposure to unknown external risk factors, differences in health care and cancer registration, or a combination of these factors. Cancer of the prostate is multifocal and it is conmnonly observed that the cancerous gland contains multiple independent lesions, suggesting the heterogeneity of the disease 30 (Foster et al., "Cellular and Molecular Pathology of Prostate Cancer Precursors," Scand. J. Urol. Nephrol. 205:19-43 (2000)). Determinants responsible for the pathologic growth of the prostate remain poorly understood, although steroidal androgens and peptide growth factors have been implicated (Agus et al., "Prostate Cancer Cell Cycle Regulators: Response to Androgen Withdrawal and Development of Androgen Independence," J. Natl. Cancer. Inst. 1 WO 2005/086638 PCT/US2005/004759 ANALOGS EXHIBITING INHIBITION OF CELL PROLIFERATION METHODS OF MAKING, AND USES THEREOF The present application is a continuation-in-part and claims priority to and benefit of 5 U.S. Patent Application Serial No. 10/992,175, which claims the priority benefit of provisional U.S. Patent Application Serial No. 60/523,079, filed November 18, 2003, both of which are hereby incorporated by reference in their entirety. The present application claims priority to and benefit of provisional U.S. Patent Application Serial No. 60/543,724, filed February 11, 2004, and provisional U.S. Patent 10 Application Serial No. 60/555,803, filed March 24, 2004, both of which are hereby incorporated by reference in their entirety.. This application was made, at least in part, with funding received from the U.S. Department of Defense under grant DAMD 17-01-1-0830. The U.S. government may retain certain rights in this invention. 15 BACKGROUND Prostate cancer accounts for 33% of all newly diagnosed malignancies among men in the United States (American Cancer Society: Cancer Facts and Figures (2003)). According to the American Cancer Society, an estimated 230,110 men will be diagnosed with prostate 20 cancer in 2004, and 29,900 men will die of it (American Cancer Society: Cancer Facts and Figures (2004)). The incidence of prostate cancer varies worldwide, with the highest rates found in the United States, Canada, and Scandinavia, and the lowest rates found in China and other parts of Asia (Quinn and Babb, "Patterns and Trends in Prostate Cancer Incidence, Survival, Prevalence and Mortality. Part: International Comparisons," BJU Int. 90:162-173 25 (2002); Gronberg, "Prostate Cancer Epidemiology," Lancet 361:859-864 (2003)). These differences are caused by genetic susceptibility, exposure to unknown external risk factors, differences in health care and cancer registration, or a combination of these factors. Cancer of the prostate is multifocal and it is commonly observed that the cancerous gland contains multiple independent lesions, suggesting the heterogeneity of the disease 30 (Foster et al., "Cellular and Molecular Pathology of Prostate Cancer Precursors," Scand. J Urol. Nephrol. 205:19-43 (2000)). Determinants responsible for the pathologic growth of the prostate remain poorly understood, although steroidal androgens and peptide growth factors have been implicated (Agus et al., "Prostate Cancer Cell Cycle Regulators: Response to Androgen Withdrawal and Development of Androgen Independence," J. Natl. Cancer. Inst. 1 WO 2005/086638 PCT/US2005/004759 91:1869-1876 (1999); Djakiew, "Dysregulated Expression of Growth. Factors and Their Receptors in the Development of Prostate Cancer," Prostate 42:150-160 (2000)). As long as the cancer is confined to the prostate, it can be successfully controlled by surgery or radiation, but in metastatic disease, few options are available beyond androgen ablation 5 (Frydenberg et al., "Prostate Cancer Diagnosis and Management," Lancet 349:1681-1687 (1997)), the mainstay of treatment in the case of lymph node involvement or disseminated loci. Once tumor cells have become hormone refractory, the standard cytotoxic agents are marginally effective in slowing disease progression, although they do provide some degree of palliative relief. Current chemotherapeutic regimens, typically two or more agents, afford 10 response rates in the range of only 20-30% (Beedassy et al., "Chemotherapy in Advanced Prostate Cancer," Sern. Oncol. 26:428-438 (1999); Raghavan et al., "Evolving Strategies of Cytotoxic Chemotherapy for Advanced Prostate Cancer," Eur. J. Cancer 33:566-574 (1997)). One promising drug development strategy for prostate cancer involves identifying and testing agents that interfere with growth factors and other molecules involved in the cancer 15 cell's signaling pathways. G-protein coupled receptors ("GPCRs") are a family of membrane bound proteins that are involved in the proliferation and survival of prostate cancer cells initiated by binding of lysophospholipids ("LPLs") (Raj et at., "Guanosine Phosphate Binding Protein Coupled Receptors in Prostate Cancer: A Review," J Urol. 167:1458-1463 (2002); Kue et al., "Essential Role for G Proteins in Prostate Cancer Cell Growth and 20 Signaling," J. Urol. 164:2162-2167 (2000); Guo et al., "Mitogenic Signaling in Androgen Sensitive and Insensitive Prostate Cancer Cell Lines," J. Urol. 163:1027-1032 (2000); Barki Harrington et al., "Bradykinin Induced Mitogenesis of Androgen Independent Prostate Cancer Cells," J. Urol 165:2121-2125 (2001)). The importance of G protein-dependent pathways in the regulation of growth and metastasis in vivo is corroborated by the 25 observation that the growth of androgen-independent prostate cancer cells in mice is attenuated by treatment with pertussis toxin, an inhibitor of Gi/o proteins (Hex et al., "Influence of Pertussis Toxin on Local Progression and Metastasis After Orthotopic Implantation of the Hmnan Prostate Cancer Cell Line PC3 in Nude Mice," Prostate Cancer Prostatic Dis. 2:36-40 (1999)). Lysophosphatidic acid ("LPA") and sphingosine 1-phosphate 30 ("SIP") are lipid mediators generated via the regulated breakdown of membrane phospholipids that are. known to stimulate GPCR-signaling. LPL binds to GPCRs encoded by the Edg gene family, collectively referred to as LPL receptors, to exert diverse biological effects. LPA stimulates phospholipase D activity and PC-3 prostate cell proliferation (Qi et al., "Lysophosphatidic Acid Stimulates Phospholipase 2 WO 2005/086638 PCT/US2005/004759 D Activity and Cell Proliferation in PC-3 Human Prostate Cancer Cells," J. Cell. Physiol. 174:261-272 (1998)). Further, prior studies have shown that LPA is mitogenic in prostate cancer cells and that PC-3 and DU-145 express LPA1, LPA2, and LPA3 receptors (Daaka, "Mitogenic Action of LPA in Prostate," Biochim. Biophys. Acts 1582:265-269 (2002)). 5 Advanced prostate cancers express LPL receptors and depend on phosphatidylinositol 3 kinase ("PI3K") signaling for growth and progression to androgen independence (Kue and Daaka, "Essential Role for G Proteins in Prostate Cancer Cell Growth and Signaling," J. Ural. 164:2162-2167 (2000)). Thus, these pathways are widely viewed as one of the most promising new approaches to cancer therapy (Vivanco et al., "The Phosphatidylinositol 3 10 Kinase AKT Pathway in Human Cancer," Nat. Rev. Cancer 2:489-501 (2002)) and provide an especially novel approach to the treatment of advanced, androgen-refractory prostate cancer. Despite the promise of this approach, there are no clinically available therapies that selectively exploit or inhibit LPA or PI3K signaling. The present invention is directed to overcoming these and other deficiencies in the 15 prior art. SUMMARY A first aspect of the present invention relates to compounds according to formula (I) and formula (II) X3 R2 N X R3 4,N S R4 X 20 R3 R (I) RI (II) wherein
X
1 and X 2 are each optional, and each can be oxygen;
X
3 and X 4 are each optional, and each can be oxygen or sulfur; I is an integer from 1 to 12; 25 R 1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or 3 WO 2005/086638 PCT/US2005/004759 R5 R6 (CH2)m
R
7 R R8 or-(CH 2 )m-Y' where mi is an integer from 0 to 10 and Y' is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; 5 R 2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 hydrocarbon, Rlo-N(Z)-hydrocarbon- or R 10 -hydrocarbon- where the hydrocarbon group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated R11 R12 --(CH2)n R13 10 or unsaturated mixed heterocycle, or R15 R14 or -(CH 2 )n- Y2 where n is an integer from 0 to 10 and Y 2 is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; R3 is hydrogen or an aliphatic or non-aliphatic straight- or branched-chain Cl to C 10 15 hydrocarbon; R4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl;
R
5 , R 6 , R, R 8 , R 9 , R", R 2 , R 13 , R 1 4 , and R 1 5 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to 20 C10 hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl; R"o is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, 25 hydrocarbon-O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, 4 WO 2005/086638 PCT/US2005/004759 H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-phenylalkyl-O-hydrocarbon-, phenylalkyl-O-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic 5 or non-aliphatic straight- or branched-chain C1 to C10 group, and wherein each alkyl is a C1 to C 10 alkyl; and Z is independently hydrogen or t-butoxycarbonyl. A second aspect of the present invention relates to compounds according to formula 10 (V) and formula (VI) X5 X5 R2 O R2 O XR3 X NR3 X2 R4",N S R4,N S RI (V) RI (VI) wherein
X
1 and X 2 are each optional, and each can be oxygen;
X
5 is optional, and can be oxygen; 15 R 1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocycles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or R5 R6 S- (CH2)m R7 R9 R8 or-(CH 2 )m-Y where m is an integer from 0 to 10 and Y' is a 20 saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 hydrocarbon, Rlo-N(Z)-hydrocarbon- or R1 0 -hydrocarbon- where the hydrocarbon 25 group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a 5 WO 2005/086638 PCT/US2005/004759 saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated R11 R12 S-(CH 2 )n i R13 or unsaturated mixed heterocycle, or R15 R14 or -(CHI 2 )n- y 2 where n is an integer from 0 to 10 and y 2 is a saturated or unsaturated cyclic hydrocarbon, saturated 5 or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; R3 is nothing, hydrogen or an aliphatic or non-aliphatic straight- or branched-chain C1 to C10 hydrocarbon; R4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched 10 chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl;
R
5 , R 6 , R 7 , R 8 , R 9 , R 11 , R 2 , R , R 14, and R 15 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10O hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, 15 alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl;
R
10 is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, 0 hydrocarbon-, hydrocarbon-0O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, 20 H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-phenylalkyl-O-hydrocarbon-, phenylalkyl-0O-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic or non-aliphatic straight- or branched-chain C1 to CI0 group, and wherein each alkyl is a C1 25 to C10 alkyl; and Z is independently hydrogen or t-butoxycarbonyl. A third aspect of the present invention relates to compounds according to formula (VII) 6 WO 2005/086638 PCT/US2005/004759 X3 R2 R3 0 R4N- O RI (VIi) wherein
X
3 is optional and can be oxygen;
X
6 is oxygen or nitrogen; 5 R' is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or Rs5 R 6 S--(CH2m R7 R9 R8 or-(CH2)m-Y 1 where mn is an integer from 0 to 10 and Y' is a 10 saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 hydrocarbon, Rlo-N(Z)-hydrocarbon- or R 10 -hydrocarbon- where the hydrocarbon 15 group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated RII R12 S-(CH 2 )n
R
1 3 or unsaturated mixed heterocycle, or RI5 R14 or -(CH 2 ),- y 2 where n is an integer from 0 to 10 and Y2 is a saturated or unsaturated cyclic hydrocarbon, saturated 20 or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; 7 WO 2005/086638 PCT/US2005/004759
R
3 is nothing, hydrogen or an aliphatic or non-aliphatic straight- or branched-chain Cl to C10O hydrocarbon;
R
4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl; 5 R 5 , R 6 , R 7 , R 5 , R?, R 1 , R 12 , R 3 , R 4 , and R i s are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10 hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl; 10 R 1 o is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, hydrocarbon-0O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, 15 H(Z)N-phenylalkyl-O-hydrocarbon-, phenylalkyl-0O-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic or non-aliphatic straight- or branched-chain Cl to C10 group, and wherein each alkyl is a C1 to C 10 alkyl; and 20 Z is independently hydrogen or t-butoxycarbonyl. A fourth aspect of the present invention relates to compounds of Formula (VIII) O0 R40 N NH-(CH 2 )n-NH N R4 N
N
7 RiX8 X8 R1 (VIII) wherein 25 X 8 is O or S; n is between 1 and 30;
R
1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non 30 aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or 8 WO 2005/086638 PCT/US2005/004759 R5 R6
-
(CH)m R7 R R8 or-(CH 2 )m--Y' where m is an integer from 0 to 10 and Y' is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; 5 R 4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain Cl to C10 hydrocarbon, aryl, acetyl, or mesyl; and
R
5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10 hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, 10 amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl. A fifth aspect of the present invention relates to compounds having Formula
X
9
X
9 R16 R16 HO NH X7 NH NH NH R17 R18 (IX) R17 R18 15 (X) H NH R16 NH R16 HO"X 0 (XI) 0 (XII) 20 wherein
X
7 is PO 3 H or O-benzyl;
X
9 is O or nothing; 9 WO 2005/086638 PCT/US2005/004759
R
16 is a C1 to C30 aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C1 to C30 hydrocarbon;
R
17 and R i g are independently nothing, hydrogen, -SO2R9, COR", and R1 9 ; and R1 9 is an aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or 5 unsubstituted, C1 to C30 hydrocarbon or a substituted or unsubstituted aryl. A sixth aspect of the present invention relates to a compound of Formula (XIV) and (XV) 0 0 C12H2_O O11'. 0 HO NH(CH 2
)
12 HN OH O- -PONH 4 10 NHBoc NHBoc (XIV) ONH 4 (XV). A seventh aspect of the present invention relates to a pharmaceutical composition including a pharmaceutically acceptable carrier and a compound according to the first, second, third, fourth, fifth, and sixth aspects of the present invention. 15 A eighth aspect of the present invention relates to a method of destroying a cancer cell that includes the steps of providing a compound according to the first, second, third, fourth, fifth, and sixth aspects of the present invention and contacting a cancer cell with the compound under conditions effective to destroy the contacted cancer cell. A ninth aspect of the present invention relates to a method of treating or preventing a 20 cancerous condition that includes the steps of: providing a compound according to the first, second, third, fourth, fifth, and sixth aspects of the present invention and administering an amount of the compound to a patient in a manner effective to treat or prevent a cancerous condition. A tenth aspect of the present invention relates to a method of making a compound 25 according to formula (I) that includes the steps of: reacting an intermediate according to fornnula (III), X3 1/ X4 OH RI (III) 10 WO 2005/086638 PCT/US2005/004759 where 1, R 1 , X 3 , and X 4 are defined as above, with either (i) a suitable primary or secondary amine according to the formula (HNR 2
R
3 ) where R 2 and R3 are defined as above, or (ii) ammonia in the presence of an R 2 -H containing compound, under conditions effective to form the compound according to formula (I). 5 A eleventh aspect of the present invention relates to a method of making a compound according to formula (II) that includes the steps of: reacting an intermediate according to formula (IV), X3
ICH
3 S NH RI (IV) where R I and X 3 are defined as above, with a primary or secondary amine according to the 10 formula (HNR 2
R
3 ) where R 2 and R 3 are defined as above, under conditions effective to form the compound according to formula (II). A twelfth aspect of the present invention relates to intermediate compounds according to formula (III) and formula (IV). The present invention affords a significant improvement over previously identified 15 cancer therapeutics that are known to be useful for the inhibition of prostate cancer cell growth. In a previous report, it was shown that cytotoxic compounds were obtained by replacing the glycerol backbone in LPA with serine amide in five prostate cancer cell lines (Gududuru et al., "Synthesis and Biological Evaluation of Novel Cytotoxic Phospholipids for Prostate Cancer," Btaorg. Med. Chem. Lett. 14:4919-4923 (2004), which is hereby 20 incorporated by reference in its entirety). The most potent compounds reported in Gududuru et al. (cited above) were non-selective and potently killed both prostate cancer and control cell lines. The present invention affords compounds that possess similar or even improved potency, but more importantly, improved selectivity, particularly with respect to prostate cancer cell lines. Compounds of the present invention are shown to be effective against 25 prostate cancer cells and ovarian cancer cells. 11 WO 2005/086638 PCT/US2005/004759 BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS The following detailed description of embodiments of the present invention can be best understood when read in conjunction with the following drawings, where like structure is indicated with like reference numerals and in which: 5 Figure 1 illustrates one approach (scheme 1) for the synthesis of thiazolidine carboxylic acid amides. The thiazolidine carboxylic acid intermediate (2a-v) is formed upon reacting L-cysteine with various aldehydes under reported conditions (Seki et al., "A Novel Synthesis of(+)-Biotin from L-Cysteine,"J. Org. Chem. 67:5527-5536 (2002), which is hereby incorporated by reference in its entirety). The intermediate carboxylic acid is reacted 10 with an amine to form the corresponding amide (3-27); Figure 2 illustrates one approach (scheme 2) for the synthesis of N-30 aryl and N sulfonyl derivatives of the thiazolidine carboxylic acid amides. The N-acyl and N-sulfonyl derivatives (compounds 28 and 29) were synthesized from compound 5 by standard procedures; 15 Figure 3 illustrates one approach (scheme 3) for the synthesis of thiazole carboxylic acid amides. The thiazolidine carboxylic acid methyl ester was converted to the thiazole carboxylic acid methyl ester following a reported procedure (Badr et al,, "Synthesis of Oxazolidines, Thiazolidines, and 5,6,7,8-Tetrahydro-lH, 3H-pyrrolo[1,2-c] Oxazole (or Thiazole)-l,3-diones from 0-Hydroxy- or 0-Mercapto-cc-amino Acid Esters," Bull. Chem. 20 Soc. Jpn. 54:1844-1847 (1981), which is hereby incorporated by reference in its entirety), and then converted to the alkylamide; Figures 4A-B illustrate agarose gel electrophoresis of total DNA extracted from 2 x 106 LNCaP cells following treatment with thiazolidine compounds 4 (Figure 4A) and 5 (Figure 4B) for 24 to 108 hours. The results show the effects of treatment on DNA 25 fragmentation, indicating progression of cell death. In Figure 4A, the dose and exposure time are indicated for compound 4 as follows: lane 1, 100 bp DNA marker; lane 2, 5 fM for 36 h; lane 3, 3 jM for 24 h; lane 4, 3 !tM for 24 h; lane 5, 3 1M for 48 h; lane 6, 3 JM for 72 h; lane 7, 3 AM for 108 h; and lane 8, 50 AM for 36 h. In Figure 4B, the dose and exposure time are indicated for compound 5 as follows: lane 1, 100 bp DNA marker; lane 2, 5 tM for 24 h; 30 lane 3, 5 tM for 48 h; lane 4, 5 gM for 72 h; lane 5, 5pM for 96 h; lane 6,3 jiM for 96 h; lane 7, 8 gM for 48 h; and lane 8, 8 AM for 72 h; Figures 5A-B demonstrate AKT inhibitory effects of thiazolidine compounds, as measured by inhibition of AKT phosphorylation. Figure 5A shows the immunoblot results 12 WO 2005/086638 PCT/US2005/004759 using anti-phospho AKT (5473) or anti-AKT antibodies. The immunoblots were visualized by enhanced chemiluminescence, and changes of relative levels of phospho-AKT compared to total AKT by analog treatment were quantified by densitometric analysis. Figure 5B graphically illustrates the immunological detection of AKT using anti-AKT and anti-phospo 5 AKT, shown in Figure 5A; Figure 6 illustrates one approach (scheme 4) for the synthesis of 4-thiazolidinone carboxylic acids, and their conversion to corresponding amides by reaction with primary or secondary amines (HNR2R3). As shown in this reaction scheme, different starting materials (where 1 differs) can be used to prepare various compounds of the invention; 10 Figure 7 illustrates a second approach (scheme 5) for the synthesis of 4-thiazolidinone carboxylic acids, and their conversion to corresponding amides by reaction with R 2 -CNO; Figure 8 illustrates three approaches for modifying the core structure of the thiazolidinone compounds of the present invention (scheme 6) to afford ring-bound sulfone or sulfoxide groups (steps a and b, respectively), as well as the complete reduction of 15 carbonyl groups (step c); Figure 9 illustrates a process for the synthesis of polyamine conjugates of thiazolidinone amides; Figure 10 illustrates a scheme for the synthesis of thiazolidinone ethers and esters; Figure 11 illustrates a scheme for the synthesis of oxazoline amides; 20 Figure 12 illustrates a scheme for the synthesis of thiazolidinone dimers; Figure 13 illustrates a scheme for the synthesis of serine amide alcohols and phosphates. The reagents and conditions can be: (i) CH 3
(CH
2 )nNH 2 , EDC, HOBt, CH 2
C
2 , rt, 5 h (ii) TFA, CH2C1 2 , rt, 0.5 h (iii) tetrazole, dibenzyl diisopropylphosphoramidite, CH 2
C
2 , rt, 0.5 h, H202, rt, 0.5 h (iv) H2, 10% Pd/C, EtOH, rt, 3 h; 25 Figure 14 illustrates a scheme for the synthesis of unsaturated serine amide alcohols and phosphates. The reagents and conditions can be: (i) CsH 1 7 (CH: CH)CsH 1 6
NIH
2 , EDC, HOBt, CH 2 C1 2 , rt, 5 h (ii) 2M HCl/Et20, rt, overnight (iii) tetrazole, di tert butyl diisopropylphosphoramidite, CH 2
C
2 , rt, 0.5 h, H202, rt, 0.5 h (iv) TFA, CH 2 Cl 2 , rt, 0.5 h; Figure 15 illustrates a scheme for the synthesis of serine diamide phosphates and 30 other amide analogs. The reagents and can be conditions: (i) R 2
NH
2 , EDC, HLOBt, CH2C1 2 2 , rt, 5 h (ii) TFA, CH 2 Cl 2 , rt, 0.5 h (iii) TEA, R 3
SO
2 CI or R 3 NCO or R 3 COC1 (iv) H2, 10% Pd/C, EtOH, rt, 3 h (v) tetrazole, dibenzyl diisopropylphosphloramidite, CH2C1 2 , rt, 0.5 h, 11H202, rt, 0.5 h (vi) H2, 10% Pd/C, EtOH, rt, 3 h; and 13 WO 2005/086638 PCT/US2005/004759 Figure 16 illustrates a scheme for the preparation of amino alcohol analogs. The Reagents and conditions: (i) TFA, CH 2
CI
2 , rt, 0.5 h (ii) a. LAH, Et 2 0, reflux, 7 h, b. HC1 DETAILED DESCRIPTION OF EMBODIMENTS OF THE INVENTION 5 The present invention will now be described with occasional reference to the specific embodiments of the invention. This invention may, however, be embodied in different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the invention to those skilled in the art. 10 Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The terminology used in the description of the invention herein is for describing particular embodiments only and is not intended to be limiting of the invention. As used in the description of the invention and the appended claims, the singular forms "a," "an," and 15 "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth as used in the 20 specification and claims are to be understood as being modified in all instances by the term "about." Accordingly, unless otherwise indicated, the numerical properties set forth in the following specification and claims are approximations that may vary depending on the desired properties sought to be obtained in embodiments of the present invention. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the 25 invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical values, however, inherently contain certain errors necessarily resulting from error found in their respective measurements. One aspect of the invention relates to compounds according to formulae (I) and (II) below 14 WO 2005/086638 PCT/US2005/004759 X3 X4 R2 R2' N S.X R3 R4N- S X2 1 xl R4 R3 (I) R1 (II) wherein X1 and X2 are each optional, and each can be oxygen; X and X 4 are each optional, and each can be oxygen or sulfur; 5 1 is an integer from 1 to 12;
R
1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C 1 to C30 hydrocarbons, or R5 R6 S- (CH 2 )m R7 10 R9 R8 or-(CH 2 )m-Y' where m is an integer from 0 to 10 and Y' is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 15 hydrocarbon, Rl 0 -N(Z)-hydrocarbon- or Ri 0 -hydrocarbon- where the hydrocarbon group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated R11 R12 S-(CH 2 )n R13 or unsaturated mixed heterocycle, or R15 R14 or -(CH 2
)
n - Y 2 where 20 n is an integer from 0 to 10 and Y2 is a saturated or unsaturated cyclic hydrocarbon, saturated 15 WO 2005/086638 PCT/US2005/004759 or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; R3 is hydrogen or an aliphatic or non-aliphatic straight- or branched-chain Cl to C10 hydrocarbon; 5 R4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain Cl 1 to C10 hydrocarbon, aryl, acetyl, or mesyl;
R
5 , R6, R , R , R 9 , R" 1 , R' 12 , R" 3 , R14, and Rs are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C 1 to C10O hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, 10 dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl;
R'
0 is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, hydrocarbon-O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, 15 H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-phenylalkyl-O-hydrocarbon-, phenylalkyl-O-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic 20 or non-aliphatic straight- or branched-chain Cl to C10 group, and wherein each alkyl is a C1 to C10 alkyl; and Z is independently hydrogen or t-butoxycarbonyl. As used herein, "aliphatic or non-aliphatic straight- or branched-chain hydrocarbon" refers to both alkylene groups that contain a single carbon and up to a defined upper limit, as 25 well as alkenyl groups and alkynyl groups that contain two carbons up to the upper limit, whether the carbons are present in a single chain or a branched chain. Unless specifically identified, a hydrocarbon can include up to about 30 carbons, or up to about 20 hydrocarbons, or up to about 10 hydrocarbons. As used herein, the term "alkyl" can be any straight- or branched-chain alkyl group 30 containing up to about 30 carbons unless otherwise specified. The alkyl group can be a sole constituent or it can be a component of a larger constituent, such as in an alkoxy, arylalkyl, alkylamino, etc. As used herein, "saturated or unsaturated cyclic hydrocarbons" can be any such cyclic hydrocarbon, including but not limited to phenyl, biphenyl, triphenyl, naphthyl, cycloalkyl, 16 WO 2005/086638 PCT/US2005/004759 cycloalkenyl, cyclodienyl, etc.; "saturated or unsaturated N-heterocycles" can be any such N containing heterocycle, including but not limited to aza- and diaza-cycloalkyls such as aziridinyl, azetidinyl, diazatidinyl, pyrrolidinyl, piperidinyl, piperazinyl, and azocanyl, pyrrolyl, pyrazolyl, imidazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, 5 tetrazinyl, pyrrolizinyl, indolyl, unsaturated O0-heterocycles" can be any such O-containing heterocycle including but not limited to oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, dioxanyl, furanyl, pyrylium, benzofuranyl, etc.; "saturated or unsaturated S-heterocycles" can be any such S-containing heterocycle, including but not limited to thiranyl, thietanyl, tetrahydrothiophenyl, dithiolanyl, tetrahydrothiopyranyl, thiophenyl, 10 thiepinyl, thianaphthenyl, etc.; "saturated or unsaturated mixed heterocycles" can be any. heterocycle containing two or more S-, N-, or O-heteroatoms, including but not limited to oxathiolanyl, morpholinyl, thioxanyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiaziolyl, etc. Another aspect of the present invention relates to compounds according to formula 15 (V) and formula (VI) X5 XS R2 R2 3 R4-,N SC 1R4,N S RI (V) Ri (VI) wherein
X
1 and X 2 are each optional, and each can be oxygen;
X
5 is optional, and can be oxygen; 20 R1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or R5 R6 Rs R7 -(CH2)m R7 R9 R8 or-(CH2)m-Y 1 where m is an integer from 0 to 10 and Y 1 is a 25 saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated 17 WO 2005/086638 PCT/US2005/004759 or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 hydrocarbon, R10-N(Z)-hydrocarbon- or R 10 -hydrocarbon- where the hydrocarbon 5 group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated R1 R12 -(CH2)n R13 or unsaturated mixed heterocycle, or R15 R14 or -(CH 2 )n- Y 2 where n is an integer from 0 to 10 and Y 2 is a saturated or unsaturated cyclic hydrocarbon, saturated 10 or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
3 is nothing, hydrogen or an aliphatic or non-aliphatic straight- or branched-chain Cl to C 10 hydrocarbon;
R
4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched 15 chain C 1 to C 10 hydrocarbon, aryl, acetyl, or mesyl;
R
5 , R 6 , R 7 , R 8 , R', R 11 , R 12 , R 3 , R 1 4 , and R i s are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C 1 to C10 hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, 20 alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl;
R
0 is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, hydrocarbon-O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, 25 H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-phenylalkyl-O-hydrocarbon-, phenylalkcyl-0-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalklcyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic or non-aliphatic straight- or branched-chain C1 to C10 group, and wherein each alkyl is a C1 30 to C10 alkyl; and 18 WO 2005/086638 PCT/US2005/004759 Z is independently hydrogen or t-butoxycarbonyl. Another aspect of the present invention relates to compounds according to formula (VII) X3 R2 X6 R3R4N 0 R4,N 5 R1 (VII) wherein
X
3 is optional and can be oxygen;
X
6 is oxygen or nitrogen; I is an integer from 1 to 12; 10 R 1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or Rs5 R6 S-(CH2)m R7 R R8 or-(CH2)m-Y 1 where m is an integer from 0 to 10 and Y' is a 15 saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 hydrocarbon, Rlo-N(Z)-hydrocarbon- or Ri 0 -hydrocarbon- where the hydrocarbon 20 group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated 19 WO 2005/086638 PCT/US2005/004759 R11 R12 S- (CH2)n R13 or unsaturated mixed heterocycle, or R15 R14 or -(CH 2 )n- Y 2 where n is an integer from 0 to 10 and Y2 is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; 5 R 3 is nothing, hydrogen or an aliphatic or non-aliphatic straight- or branched-chain Cl to C 10 hydrocarbon;
R
4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl; R 5 , R R, R 7 , R, R, R", R, R", RI 4 , and Ri 5 are independently selected from the 10 group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10O hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl; R" is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) 15 -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, hydrocarbon-O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-phenylalkyl-O-hydrocarbon-, phenylalkyl-O-hydrocarbon-, 20 phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic or non-aliphatic straight- or branched-chain C1 to C10 group, and wherein each alkyl is a C1 to C10 alkyl; and Z is independently hydrogen or t-butoxycarbonyl. 25 Yet another aspect of the present invention relates to compounds of Formula (VIII) 20 WO 2005/086638 PCT/US2005/004759 0 R40 \ N NH-(CH 2 )n-NH I R4 RI ( X8R1 Xa R1 (VIII) wherein
X
8 is O or S; n is between 1 and 30; 5 R' is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or R5 R6 S (CH2)m R7 R9 R8 or-(CH 2 )m-Y 1 where m is an integer from 0 to 10 and Y' is a 10 saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle;
R
4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl; and 15 R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10 hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl. 20 21 WO 2005/086638 PCT/US2005/004759 Another aspect of the present invention relates to compounds having Formula
X
9 X9 R16 R16 HO NH X7 NH NH NH R17 R18 (IX) R17 R18 (X) 5 HONH R16 XNH R16 o (XI) o (XII) wherein
X
7 is PO 3 H or O-benzyl; 10 X 9 is O or nothing;
R
1 6 is a C1 to C30 aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C1 to C30 hydrocarbon;
R
17 and R 18 are independently nothing, hydrogen, -SO 2
R"
9 , COR 9 , and R 19 ; and R1 9 is an aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or 15 unsubstituted, Cl to C30 hydrocarbon or a substituted or unsubstituted aryl. In one example, the compound of Formula (X) is limited such that R 1 6 is not C 1 4H 2 9 when X 7 is PO 3 H and X 8 is O. A further aspect of the present invention relates to a compound of Formula (XIV) and (XV) 20 0 0
C
1 2
H
2 6 - ( 0 O O12 0 HO NH(CH 2
)
12 HN OH O-P-ONH 4 NHBoc NHBoc (XIV) ONH+4 (XV). 22 WO 2005/086638 PCT/US2005/004759 It will be understood that the dotted lines in the structures of Formulae II and VII indicate the presence or absence of a bond. Preferred R' groups include benzyl, furanyl, indolyl, pyridinyl, phenyl, or substituted phenyl (with R 5
-R
9 as defined above). 5 Preferred R 2 groups include aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbons, phenyl, phenylalkyls, substituted phenyls and substituted phenylalkyls with Ru 1
-R
1 5 groups as defined above. Preferred aliphatic or non-aliphatic straight- or branched-chain hydrocarbons are C8 to C24 hydrocarbons, including C10 to C20 alkyls, more preferably C14 to C18 alkyls. 10 Preferred R 3 groups include hydrogen and C1 to C10 alkyls. Preferred R 4 groups include hydrogen, acyl, acetyl, and mesyl. Preferred Rio groups are polyarnines. The integer I is preferably from 1 to 10, more preferably 1 to 8, 1 to 6, or 1 to 4. The integer In is preferably from 0 to 8, 0 to 6, 0 to 4, or 0 to 2. The integer n is preferably from 0 15 to8, 0 to6, 0 to4,or 0 to2. Exemplary compounds according to formula (I) include, without limitation: 2-(4-oxo 2-phenylthiazolidin-3-yl)acetamide (compound 65), N-decyl-2-(4-oxo-2-phenylthiazolidin-3 yl)acetamide (compound 66), N-tetradecyl-2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 67), N-octadecyl-2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 68), 20 N-octadecyl-2-(4-oxo-2-biphenylthiazolidin-3-yl)acetamide (compound 69), 2-(2-(1 (dimethylamino)naphthalen-4-yl)-4-oxothiazol idin-3-yl)-N-octadecylacetamide (compound 70), 2-(2-(4-methoxyphenyl)-4-oxothiazolidin-3-yl)-N-octadecylacetamtnide (compound 71), 2-(2-(2,6-dichlorophenyl)-4-oxothiazolidin-3 yl)-N-octadecylacetamide (compound 72), N octadecyl-2-(4-oxo-2-phenyl-l-sulfoxide-thiazolidin-3-yl)acetamide (compound 80), N 25 octadecyl-2-(4-oxo-2-phenyl-l1-sulfonyl-thiazolidin-3-yl)acetamide (compound 81), N-(3,5 difluorophcnyl)-2-(4-oxo-2-phenylthiazolidin-3-yl)acctamide (compound 73), N-(3,5 difluorophenyl)-2-(4-oxo-2-phenylthiazolidin-3-yl)ethanethioamide, N-(3,5 bis(trifluoromethyl)phenyl)-2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 74), N (3,5-dichlorophenyl)-2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 75), N-(2,4 30 dimethoxyphenyl)-2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 76), N-* (naphthalen-l1-yl).2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 77), 3-(2 (octadecylamino)ethyl)-2-phenylthiazolidin-4-one (compound 79), N-(2-(2 phenylthiazolidin-3-yl)ethyl)octadecan-1 -amine, and salts thereof. Preferred compounds according to formula (I) include compounds 68, 71, 80, and 81. 23 WO 2005/086638 PCT/US2005/004759 Exemplary compounds according to formula (II) include, without limitation: (4R)-2 (4-methoxyphenyl)-N-oetadecylthiazolidine-4-carboxamide (compound 15); (4R)-2-(4 ethoxyphenyl)-N-octadecylthiazolidine-4-carboxamide; N-octadecyl-2-phenyithiazole-4 carboxamide (compound 34); (4R)-2-(3,5-difluorophenyl)-N-octadecylthiazolidine-4 5 carboxamide (compound 23); (4R)-2-(4-cyanophenyl)-N-octadecylthiazolidine-4 carboxamide (compound 22); (4R)-N-octadecyl-N-mesyl-2-phenylthiazolidine-4 carboxamide (compound 29); (4R)-N-octadecyl-N-acetyl-2-phenylthiazolidine-4 carboxamide (compound 28); (4R)-N-heptyl-2-phenylthiazolidine-4-carboxamide (compound 3); (4R)-N-octadecyl-2-phenylithiazolidine-4-carboxamide (compound 5, R-isomer); (4S)-N 10 octadecyl-2-phenylthiazolidine-4-carboxamide (compound 5, S-isomer); (4R)-N-tetradecyl 2-phenylithiazolidine-4-carboxamide hydrochloride (compound 4); (4R)-N-octadecyl-2 biphenylthiazolidine-4-carboxamide (compound 27); (4R)-2-dodecyi-N octadecylthiazolidine-4-carboxamide (compound 7); (4R)-N-octadecyl-2-(pyridin-3 yl)thiazolidine-4-carboxamide (compound 11); 2-(furan-3-yl)-N-octadecylthiazolidine-4 15 carboxamide (compound 12); (4R)-N-nonadecyl-2-phenylthiazolidine-4-carboxamide (compound 6); (4R)-2-(4-hydroxyphenyl)-N-octadecylthiazolidine-4-carboxamide; 2-(3 hydroxyphenyl)-N-octadecylthiazolidine-4-carboxamide (compound 14); (4R)-2-(2,4,6 dimethoxyphenyl) N-octadecylthiazolidine-4-carboxamide; 2-(3,4-dimethoxyphenyl)-N octadecylthiazolidine-4-carboxamide (compound 18); (4R)-2-(4-fluorophenyl)-N 20 octadecylthiazotidine-4-carboxamide (compound 19); (4R)-2-(2,6-dichlorophenyl)-N octadecylthiazolidine-4-carboxamide (compound 24); (4R)-2-(4-bromophenyl)-N octadecylthiazolidine-4-carboxamide (compound 20); (4R)-N-octadecyl-2-p tolylthiazolidine-4-carboxamide (compound 26); (4R)-2-cyclohexyl-N-octadecylthiazolidine 4-carboxamide (compound 8); 2-(4-nitrophenyl)-N-octadecylthiazolidine-4-carboxamide 25 (compound 21); (4R)-2-(4-(dimethylamino)phenyl)-N-octadecylthiazolidine-4-carboxamide (compound 13); (4R)-2-(lH-indol-3-yl)-N-octadecylthiazolidine-4-carboxamide (compound 10); (4R)-2-benzyl-N-octadecylthiazolidine-4-carboxamide (compound 9); (4R)-2-(3-bromo 4-fluorophenyl)-N-octadecylthiazolidine-4-carboxamide (compound 25); (4R)-2-(3,4,5 trimethoxyphenyl)-N,N-dioctylthiazolidine-4-carboxamide; and salts thereof. 30 Preferred compounds according to formula (II) include compounds 5 (R-isomer), 13, 14, 16, 17, 18, 19, 25, and 26. Compounds of Formula V include, but are not limited to: 24 WO 2005/086638 PCT/US2005/004759 o NH O- C 1 8
H
37 NH O-C8H37 S S o HN "' OH O ~' s HN- "'~O HN OH O 0 12
H
25 S MeOS 0 HN *"OH 00 0 F H HN "OH HN "OH CI HN " OH I S s s 0 HN "Ni OH S and N 5 Compounds of Formula (VII) include, but are not limited to: 0 HCI. NH- NH-(CH 2 )nCH 3 0 ,wherein n= 6, 13, and 17. Compounds of Formulae (IX), (X), (XI), and (XII) include, but are not limited to those found in Table 9 of the Examples section. The compounds of the present invention and their intermediates can be synthesized 10 using commercially available or readily synthesized reactants. By way of example, the compounds according to formula (I) can be synthesized according to scheme 4 illustrated in Figure 6. According to one approach, an intermediate acid according to formula (Ili) 25 WO 2005/086638 PCT/US2005/004759 X3 1/ X4 S N R1 OH 1 RI (III) (where 1, R', X 3 , and X 4 are as defined above) is reacted with appropriate amines in the presence of EDC/HOBt under standard conditions. The intermediate acids can be prepared initially via condensing mercaptoacetic acid, glycine methyl ester, and aromatic aldehydes in 5 a one-pot reaction, followed by basic hydrolysis of the ester (Holmes et al., "Strategies for Combinatorial Organic Synthesis: Solution and Polymer-Supported Synthesis of 4 Thiazolidinones and 4-Metathiazanones Derived from Amino Acids," . Org. Chenm. 60:7328-7333 (1995), which is hereby incorporated by reference in its entirety). By substituting glycine methyl ester with analogs containing longer carbon backbones, it 10 becomes possible to prepare compounds according to formula (III) and, ultimately, formula (I), with I being greater than 1 (i.e., containing an alkylene group that is longer than methylene). According to a second approach, the thiazolidinone amides of formula (1) can also be prepared by a simple and direct method (Schuemacher et al., "Condensation Between Isocyanates and Carboxylic Acids in the Presence of 4-Dimethylaminopyridine. (DMAP), a 15 Mild and Efficient Synthesis of Amides," Synthesis 22:243-246 (2001), which is hereby incorporated by reference in its entirety), which involves reaction of the intermediate acid with desired isocyanates in the presence of a catalytic amount of DMAP (Figure 7) (scheme 5), Further modification of the thiazolidinone compounds can be achieved by, e.g., 20 exhaustive reduction of using BH 3 THF under reflux conditions to eliminate carbonyl or sulfoxide groups X 3 and X 4 (Figure 8) (scheme 6c), as well as oxidation of a compound using
H
2 0 2 and K-MnO 4 to afford sulfoxides or sulfones, respectively, as shown in scheme 6a and 6b. Also by way of example, compounds according to formula (II) can be prepared by 25 reacting an intermediate acid according to formula (IV), X3
CH
3 S NH R1 (IV) 26 WO 2005/086638 PCT/US2005/004759 where compound (IV) can be either the R- or S-stereoisomer and R' and X 3 are defined as above, with appropriate amines in the presence of EDC/HOBt under standard conditions. The intermediate acids can be prepared via reaction of L-cysteine with desired aldehydes under reported conditions (Seldki et al., "A Novel Synthesis of (+)-Biotin from L-Cysteine," J. 02g. 5 Chem. 67:5527-5536 (2002), which is hereby incorporated by reference in its entirety). The compounds of the present invention can also be modified to contain a polymeric conjugate. Suitable polymeric conjugates include, without limitation, poly(alkyl)amines, poly(alkoxy)amine, polyamines, etc. It is also well known that polyamine containing compounds exhibit a number of biological activities and have been utilized as 10 chemotherapeutic agents. Exemplary conjugates include those containing the naturally occurring polyamines like putrescine, spermidine, and spermine, as well as synthetic polyamines. According to one approach, a compound of the present invention can be conjugated to a polyamine by reacting the intermediate acid or a nitrophenyl derivative thereof with a 15 polyamine NH 2 -R2 where R 2 is R 10 -N(Z) - hydrocarbon-. or. R 10 -hydrocarbon- , with R 10 and Z being as defined above. An exemplary synthesis scheme is illustrated in Figure 9. By way of example, compounds of Formulae (V) and (VI) can be formed in accordance with the exemplary synthesis scheme illustrated in Fig. 10. The compound can be made in any other suitable manner. 20 By way of example, oxazoline analog compounds of Formula (VII) can be formed in accordance with the scheme illustrated in Fig. 11. Additionally, the compounds of Formula (VII) can be formed using the methods outlined above with respect to the compounds of Formula (II). The compounds may also be made in any other suitable manner. By way of example, compounds of Formula (VIII) can be formed in accordance with 25 the scheme illustrated in Fig. 12. Additionally, the compounds can be made in any other suitable manner. By way of example, compounds of Formulae (IX) and (X) can be made in accordance with the general synthesis of serine amide phosphates (SAPs), serine amide alcohols (SAAs), and serine diamide phosphates (SDAPs) shown in the schemes of Figs. 13-16. Commercially available N 30 Boc-serine (R or S form) is allowed to react with an appropriate amine in presence of EDC/HOBt to form an amide. The amide is treated with TFA to give an SAA analog. Phosphorylation of the amide and concurrent removal of protecting groups under hydrogenolysis conditions using Pd/C in ethanol gave an SAP. Unsaturated analogues of SAA and SAP can synthesized by similar procedures as shown in the scheme of Fig. 14. Serine diamide phosphates (SDAPs) and other 27 WO 2005/086638 PCT/US2005/004759 amine derivatives can be synthesized starting from O-benzyl N-Boc-serine as shown in the scheme of Fig. 15. LAH mediated reduction of an amine compound gives long chain N-alkyl amino alcohols as shown in the scheme of Fig. 16. Compounds of Formula (XI) and (XII) which have an ethanolamine amide backbone rather than the serine amide backbone can be synthesized according 5 to the reported procedure Lynch, K. R. H., D. W.Carlisle, S. J.Catalano, J. G.Zhang, M.MacDonald, T. L. Mol. Pharmacol. 1997, 52, 75-81, which is incorporated by reference in its entirety. The compounds can also be in the form of a salt, preferably a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salt" refers to those salts that retain 10 the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxylie acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 15 cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcysteine and the like. Other salts are known to those of skill in the art and can readily be adapted for use in accordance with the present invention. The compounds of the present invention can be present in the form of a racemic mixture, containing substantially equivalent amounts of stereoisomers. In another 20 embodiment, the compounds of the present invention can be prepared or otherwise isolated, using known procedures, to obtain a stereoisomer substantially free of its corresponding stcreoisomer (i.e., substantially pure). By substantially pure, it is intended that a stereoisomer is at least about 95% pure, more preferably at least about 98% pure, most preferably at least about 99% pure. 25 Another aspect of the present invention relates to pharmaceutical compositions that contain one or more of the above-identified compounds of the present invention. Generally, the pharmaceutical composition of the present invention will include a compound of the present invention or its pharmaceutically acceptable salt, as well as a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" refers to any suitable 30 adjuvants, carriers, excipients, or stabilizers, and can be in solid or liquid form such as, tablets, capsules, powders, solutions, suspensions, or emulsions. In one example, the composition will contain from about 0.01 to about 99 percent or from about 20 to about 75 percent of active compound(s), together with the adjuvants, carriers and/or excipients. For example, application to mucous membranes can be achieved 28 WO 2005/086638 PCT/US2005/004759 with an aerosol spray containing small particles of a compound of. this invention in a spray or dry powder form. The solid unit dosage forms can be of any suitable type. The solid form can be a capsule and the like, such as an ordinary gelatin type containing the compounds of the present 5 invention and a carrier, for example, lubricants and inert fillers such as, lactose, sucrose, or cornstarch. In another embodiment, these compounds are tableted with conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders like acacia, cornstarch, or gelatin, disintegrating agents, such as cornstarch, potato starch, or alginic acid, and a lubricant, like stearic acid or magnesium stearate. 10 The tablets, capsules, and the like can also contain a binder such as gum tragacanth, acacia, corn starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose, or saccharin. When the dosage unit form is a capsule, it can contain, in addition to materials of the above type, a liquid carrier such as a 15 fatty oil. Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets can be coated with shellac, sugar, or both. A syrup can contain, in addition to active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye, and flavoring such as cherry or orange flavor, 20 For oral therapeutic administration, these active compounds can be incorporated with excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, and the like. Such compositions and preparations can contain at least 0.1% of active compound. The percentage of the compound in these compositions can, of course, be varied and can conveniently be between about 2% to about 60% of the weight of the unit. The amount of 25 active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained. In one example, compositions according to the present invention are prepared so that an oral dosage unit contains between about 1 mg and 800 mg of active compound. The active compounds of the present invention may be orally administered, for 30 example, with an inert diluent, or with an assimilable edible carrier, or they can be enclosed in hard or soft shell capsules, or they can be compressed into tablets, or they can be incorporated directly with the food of the diet. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable 29 WO 2005/086638 PCT/US2005/004759 solutions or dispersions. In all cases, the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for 5 example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils. The compounds or pharmaceutical compositions of the present invention may also be administered in injectable dosages by solution or suspension of these materials in a physiologically acceptable diluent with a pharmaceutical adjuvant, carrier or excipient. Such 10 adjuvants, carriers and/or excipients include, but are not limited to, sterile liquids, such as water and oils, with or without the addition of a surfactant and other pharmaceutically and physiologically acceptable components. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean ail, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols, such as propylene 15 glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions. These active compounds may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, 20 liquid polyethylene glycols, and mixtures thereof in oils. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions. Under ordinary conditions of storage and use, these preparations contain 25 a preservative to prevent the growth of microorganisms. For use as aerosols, the compounds of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants. The materials of the present invention also . may be administered in a non 30 pressurized form such as in a nebulizer or atomizer. The compounds of the present invention are particularly useful in the treatment or prevention of various forms of cancer, particularly prostate cancer, breast cancer, and ovarian cancer. It is believed that other forms of cancer will likewise be treatable or preventable upon administration of the compounds or compositions of the present invention to a patient. 30 WO 2005/086638 PCT/US2005/004759 Preferred compounds of the present invention are selectively disruptive to cancer cells, causing ablation of cancer cells but not normal cells. Significantly, harm to normal cells is minimized because the cancer cells are susceptible to disruption at much lower concentrations of the compounds of the present invention. 5 Thus, a further aspect of the present invention relates to a method of destroying a cancerous cell that includes: providing a compound of the present invention and then contacting a cancerous cell with the compound under conditions effective to destroy the contacted cancerous cell. According to various embodiments of destroying the cancerous cells, the cells to be destroyed can be located either in vivo or ex vivo (i.e., in culture). 10 A still further aspect of the present invention relates to a method of treating or preventing a cancerous condition that includes: providing a compound of the present invention and then administering an effective amount of the compound to a patient in a manner effective to treat or prevent a cancerous condition. An effective amount will be understood as referring to an amount of the compound that is effective at reducing, 15 preventing, ameliorating, or improving at least one symptom of the condition for which the compound is administered. It will be further understood that the term "prevent" shall be understood as referring to the prevention of the development of at least one symptom related to the condition for which the compound is administered. According to one embodiment, the patient to be treated is characterized by the 20 presence of a precancerous condition, and the administering of the compound is effective to prevent development of the precancerous condition into the cancerous condition. This can occur by destroying the precancerous cell prior to or concurrent with its further development into a cancerous state. According to another embodiment, the patient to be treated is characterized by the 25 presence of a cancerous condition, and the administering of the compound is effective either to cause regression of the cancerous condition or to inhibit growth of the cancerous condition. This preferably occurs by destroying cancer cells, regardless of their location in the patient body. That is, whether the cancer cells are located at a primary tumor site or whether the cancer cells have metastasized and created secondary tumors within the patient body. 30 As used herein, patient refers to any mammalian patient, including without limitation, humans and other primates, dogs, cats, horses, cows, sheep, pigs, rats, mice, and other rodents. When administering the compounds of the present invention, they can be administered systemically or, alternatively, they can be administered directly to a specific site 31 WO 2005/086638 PCT/US2005/004759 where cancer cells or precancerous cells are present. Thus, administering can be accomplished in any manner effective for delivering the compounds or the pharmaceutical compositions to the cancer cells or precancerous cells. Exemplary modes of administration include, without limitation, administering the compounds or compositions orally, topically, 5 transdermally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes. When the compounds or pharmaceutical compositions of the present invention are 10 administered to treat or prevent a cancerous condition, the pharmaceutical composition can also contain, or can be administered in conjunction with, other therapeutic agents or treatment regimen presently known or hereafter developed for the treatment of various types of cancer. Examples of other therapeutic agents or treatment regimen include, without limitation, radiation therapy, chemotherapy, surgical intervention, and combinations thereof. 15 Compositions within the scope of this invention include all compositions wherein the compound of the present invention is contained in an amount effective to achieve its intended purpose. While individual needs may vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typical dosages comprise about 0.01 to about 100 mg/kg body wt. The most preferred dosages comprise about 0.1 to about 20 100 mg/kg body wt. Treatment regimen for the administration of the compounds of the present invention can also be determined readily by those with ordinary skill in art. That is, the frequency of administration and size of the dose can be established by routine optimization, preferably while minimizing any side effects. EXAMPLES 25 The Examples set forth below are for illustrative purposes only and are not intended to limit, in any way, the scope of the present invention. Example 1 - Synthesis of Thiazolidine Carboxylic Acid Amides All reagents and solvents used were reagent grade or were purified by standard 30 methods before use. Moisture-sensitive reactions were carried under an argon atmosphere. Progress of the reactions was followed by thin-layer chromatography (TLC) analysis. Flash column chromatography was carried out using silica gel (200-425 mesh) supplied by Fisher. Melting points were measured in open capillary tubes on a Thomas-Hoover melting point apparatus and are uncorrected. All compounds were characterized by NMR and MS (ESI). 32 WO 2005/086638 PCT/US2005/004759 1H NMR spectra were recorded on a Varian 300 instrument. Chemical shifts are reported as S values relative to MeaSi as internal standard. Mass spectra were obtained in the electrospray (ES) mode using Esquire-LC (Broker) spectrometer. Elemental analyses were performed by Atlantic Microlab Inc. (Norcross, GA). 5 All the compounds described in this study were prepared following straightforward chemistry. Reaction of L-cysteine with various aldehydes under reported conditions (Seki et al., "A Novel Synthesis of (+)-Biotin from L-Cysteine," J. Org. Chem. 67:5527-5536 (2002), which is hereby incorporated by reference in its entirety) afforded corresponding acids (Figure 1, 2a-v), which were isolated as diastereomeric mixtures. These mixtures were used 10 directly for the formation of corresponding amides by reacting with appropriate alkyl amines using EDC/HOBt as shown in Scheme 1. All compounds thus prepared were characterized as diastereomeric mixtures (Table 1). A mixture of appropriate carboxylic acid (Figure 1, 2a-2v, 0.3-0.5 g), EDC (1.25 equiv) and HOBt (1 equiv) in CH2C1 2 (25-50 mL) was stirred for 10 min. To this solution, 15 appropriate alkyl amine (1 equiv) was added and stirring continued at room temperature for 6-8 h. Reaction mixture was diluted with CH 2
CL
2 (100-150 mL) and sequentially washed with water, satd. NaHCO 3 , brine and dried over Na 2
SO
4 . The solvent was removed under reduced pressure to yield a crude solid, which was purified by column chromatography. The purified compounds (3-6, 12, 15-18 & 27) were converted to corresponding hydrochlorides using 2M 20 HC1/Et20. (2RS, 4R)-2-Phenylthiazolidine-4-carboxylic acid heptylamide Hydrochloride (compound 3 .HC1): 1H NMR (DMSO-d6) 5 8.72 (s, IH), 7.65 (mn, 2H), 7,43 (mn, 3H), 5.89 (s, 0.6H), 5.84 (s, 0.4H), 4.66 (t, J= 6.3 Hz, 0.6H), 4.46 (t, J= 6.9 Hz, 0.4H), 3.55-3.71 (mn, 1H), 3.24-3.34 (in, IH), 3.13 (d, J= 5.7 Hz, 2H), 1.44 (m, 2H), 1.25 (s, 8H), 0.83 (t, J= 6.9 Hz, 25 3H); MS (ESI) m/z calcd for C 17
H
27
N
2 0S 307.47 (M+1), obsd 307.10. (2RS, 4R)-2-Phenylthiazolidine-4-carboxylic acid tetradecylamide Hydrochloride (compound 4.HCI): 1H NMR (DMSO-d 6 ) 5 8.69 (m, 1H), 7.64-7.71 (m, 2H), 7.45 (mn, 3H), 5.89 (s, 0.6H), 5.84 (s, 0.4H), 4,67 (t, J= 6.6 Hz, 0.6H), 4.47 (t, J= 7.2 Hz, 0.4H), 3.55-3.71 (m, 1H), 3.25-3.35 (m, 1H), 3.1,0-3.16 (m, 2H), 1.44 (min, 2H), 1.23 (s, 22H), 0.85 (t, J= 63 30 Hz, 3H); MS (ESI) m/z calcd for C 24
H
40
N
2 OS 404.65 (M), obsd 427.30 (M+Na). (2RS, 4R)-2-Phenylthiazolidine-4-carboxylic acid octadecylamide Hydrochloride (compound 5*HCI): 1 H NMR (DMSO-d6) 5 8.59 (d, J= 5.1 Hz, 1H), 7.63 (d, J= 3.9 Hz, 2H), 7.42-7.47 (in, 3H), 5.86 (s, 0.6H), 5.81(s, 0.4H), 4.60 (t, J= 6.3 Hz, 0.6H), 4.39 (t, J= 6.9 33 WO 2005/086638 PCT/US2005/004759 Hz, 0.4H), 3.52-3.66 (m, 1H), 3.24-3.30 (m, 1H), 3.10-3.16 (m, 2H), 1.42 (m, 2H), 1.23 (s, 30H), 0.85 (t, J= 6.3 Hz, 3H); MS (ESI) m/z calcd for C 28
H
4 9
N
2 0S 461.76 (M+I), obsd 461.50. (2RS, 4R)-2-Phenylthiazolidine-4-carboxylic acid nonadecylamide Hydrochloride 5 (compound 6*HC1): 'H NMR (DMSO-d6) 8 8.51 (s, IH), 7.62 (m, 2H), 7.41-7.46 (in, 3H), 5.83 (s, 0.6H), 5.78 (s, 0.4H), 4.53 (m, 0.6H), 4.32 (m, 0.4H), 3.48-3.61 (m, 1H), 3.24-3.29 (mn, 1H), 3.11-3.15 (m, 2H), 1.43 (m, 2H), 1.23 (s, 32H), 0.85 (t, J= 6.3 Hz, 3H); MS (EST) nm/z calcd for C 29 HsoN 2 0S 474.79 (M+), obsd 497.40 (M+Na). (2RS, 4R)-2-Dodecylthiazolidine-4-carboxylic acid octadecylamide (compound 7): 10 'H NMR (CDC1 3 ) 8 7.18 (m, 1H), 4.20-4.27 (m, 1H),3.79 (m, 0.3H), 3.54-3.59 (m, 0.7H), 3.08-3.34 (m, 4H), 1.65-1.78 (m, 2H), 1.43-1.51 (m, 4H), 1.27 (brs, 48H), 0.89 (t, J= 6 Hz, 6H); MS (ESI) m/z calcd for C 34
H
69
N
2 OS 553.98 (M+1), obsd 553.60. (2RS, 4R)-2-Cyclohexylthiazolidine-4-carboxylic acid octadecylamide (compound 8): 'H NMR (CDC1 3 ) 8 7.17 (m, IH), 4.10-4.20 (m, 1H), 3.76 (m, 0.3H), 3.54 (dd, J=1 1.1, 3.6 15 Hz, 0.7H), 2.97-3.34 (m, 4H), 2.02 (m, 1H), 1.68-1.78 (m, 4H), 1.48-1.54 (in, 2H), 1.27 (brs, 36H), 0.87 (t, J= 6.9 Hz, 3H); MS (ESI) m/z calcd for C 28
H
5 5
N
2 0S 467.81 (M+1), obsd 467.60. (2RS, 4R)-2-Benzylthiazolidine-4-carboxylic acid octadecylamide (compound 9): 1H NMR (CDC13) 8 7.28-7.33 (m, 5H), 7,03 (s, 0,7H), 6.48 (s, 0.3H), 4.55 (brs, 0.5H), 4.18 (brs, 20 0.5H), 3.82 (brs, 0.3H), 3.54 (dd, J= 11.1, 3.6 Hz, 0.7H), 2.99-3.31 On, 6H), 1.46-1.51 (inm, 2H),1.27 (brs, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) nim/z calcd for C 29 1 50
N
2 OS 475.79 (M+1), obsd 475.50. (2RS, 4R)-2-(1H-Indol-3y1l)-thiazolidine-4-carboxylic acid octadecylamide (compound 10): 1H NMR (CDCI 3 ) 8 7.86 (m, 0.6H), 7.77 (m, 0.4H), 7.41-7.48 (m, 4H), 7.29 25 7.34 (m,1 H), 6.0 (s, 0.3H), 5.69 (s, 0.7H), 4.37-4.41 (m, 0.5H), 3.76 (dd, J= 11.1, 4.2 Hz, 0.5H), 3.23-3.52 (m, 3H), 2.79-3.04 (in, 1H), 1.43 (m, 2H),1.27 (s, 30H), 0.89 (t, J= 6.6 Hz, 3H); MS (ESI) nm/z called for C 30 Hso 5
N
3 0S 500.80 (M+1), obsd 500.60. (2RS, 4R)-2-Pyridin-3-yl-thiazolidine-4-carboxylic acid octadecylamide (compound 11): 'H NMR (CDC1 3 ) b 8.74 (d, J= 2.1 Hz, 1H), 8.60 (d, J= 4.8 Hz, IH), 7.84 (d, J= 7.8 Hz, 30 1H), 7.31-7.36 (m, 1H), 7.08 (m, 1H), 5.44 (s, 0.5H), 5.40 (s, 0.5H), 4.28-4.35 (m, 1H), 3.72 (dd, J=1 1.1, 4.2 Hz, 1H), 3.27-3.45 (m, 3H), 2.57 (m, 1H), 1.53-1.57 (m, 2H), 1.26 (s, 30H), 0.89 (t, J= 6.6 Hz, 3H); MS (ESL) rn/z caled for C 27
H
49
N
3 0S 462.75 (M+1), obsd 462.40. 34 WO 2005/086638 PCT/US2005/004759 (2RS, 4 R)-2-Furan-3-yl-thiazolidine-4-carboxylic acid Hydrochloride (compound 12*HC1): 1H NMR (DMSO-4) 5 8.59 (d, J=15.6 Hz, 1H), 7.89 (d, J= 8.1 Hz, 1H), 7.72 (s, 1H), 5.86 (s, 0.71-1), 5.78 (s, 0.3H), 4.37-4.56 (m, 1H), 3.50-3.63 (nay, IH), 3.11-3.23 (in, 3H), 1.43 (m, 2H), 1.23 (s, 30H), 0.85 (t,J= 6.6 Hz, 3H); MS (ESI) nm/z calcd for 5 C 26
H
4 8
N
2 0 2 S 451.72 (M+1), obsd 451.60. (2RS, 4
R)-
2 -(4-Dimethylamino-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 13): 1H NMR (CDC1 3 ) 6 7.34-7.41 (m, 2H), 6.70-6.74 (m, 2H), 5.57 (s, 0.3H), 5.28 (s, 0.7H), 4.34 (m, 0.7H), 3.90 (m, 0.3H), 3.69 (dd, J= 11,1, 4.2 Hz, 1H), 3.41-3.47 (m, 1H), 3.20-3.33 (m, 2H), 2.97 (d, J= 3.6 Hz, 6H), 1.48-1.55 (m, 2H), 1.27 (s, 10 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) im/z calcd for C 30
H
54
N
3 0S 504.83 (M+1), obsd 504.60. (2RS, 4R)-2-(3-Hydroxy-phenyl)-thiazolidine-4-carboxylic add octadecylamide (compound 14): 1 H NMR (DMSO-d 6 ) 8 8.59 (s, 1H), 7.22 (t, J= 6.6 * Hz, 1H), 7.02 (d, J= 6.3 Hz, 2H), 6.82 (d, J = 7.5 Hz, 1H), 5.77 (s, 0.7H), 5.71 (s, 0.3H), 4.545 (m, 0.7H), 4.37 (m, 15 0.3H), 3.49-3.59 (m, 1H), 3.13-327 (m, 3H), 1.43 (brs, 2H), 1.23 (s, 30H), 0,85 (t, J= 6.3 Hz, 3H); MS (ESI) mn/z calcd for C 28
H
49
N
2 0 2 S 477.76 (M+1), obsd 477.60. (2RS, 4R)-2-(4-Methoxy-phenyl)-thiazolidine-4-carboxylic acid octadecylamide Hydrochloride (compound 15 HCI): 1H NMR (DMSO-d 6 ) 6 8.61 On, 1H), 7,57 (d, 3= 8.4 Hz, 2H), 6.98 (d, J= 9 Hz, 211), 5.83 (s, 0.711), 5.78 (s, 0.311), 4.61 (t, J= 6.3 Hz, 0.7H), 4,40 20 (m, 0.3H), 3.77 (s, 311), 3.51-3.70 (m, 1H), 3.22-3.31 (mn, 1H), 3.1.1 (m, 2H), 1.43 (m, 2H), 1.23 (s, 30H), 0.84 (t, J= 6.6 Hz, 3H); MS (ESI) m/z calcd for C 29
H
51
N
2 0 2 S 491.79 (M+1), obsd 491.60. (2RS, 4R)-2-(3,4-Dimethoxy-phenyl)-thiazolidine-4-carboxylic add octadecylamide Hydrochloride (compound 16 HC1): 'H NMR (DMSO-d 6 ) 5 8.58 (m, 1H), 7.33 (d, J= 4.2 Hz, 25 111), 7.14 (t, J= 7.5 Hz, 1H), 6.97 (d, J= 8.4 Hz, 1H), 5.81 (s, 0.8H), 5.77 (s, 0.2H), 4.62 (m, 0.711), 4.40 (m, 0.3H), 3.78 (d, J= 7.8 Hz, 6H), 3.52-3.68 (m, 1H), 3.23-3.29 On, 1H), 3.12 3.13 (m, 2H), 1.43 (min, 2H), 1.23 (s, 30H), 0.85 (t, J= 6.6 Hz, 3H); MS (ESI) mn/z calcd for
C
3 0H 53
N
2 0 3 S 521.81 (Mil), obsd 521.60. (2RS, 4R)-2-(3,4,5-Trimethoxy-phenyl)-thiazolidine-4-carboxylic acid 30 octadecylamide Hydrochloride (compound 17 HC1): 'H NMR (DMSO-d 6 ) 8 8.59 (mi, 1H), 7.01 (d, J= 5.7 Hz, 2H), 5.80 (s, 0.8H), 5.76 (s, 0.2H), 4.63 (in, 0.7H), 4.37 (m, 0.3H), 3.80 (d,J= 5.7 Hz, 611), 3.66 (s, 3H), 3.23-3.28 (m, 1H), 3.12-3.13 (m, 2H), 1.43 (m, 2Hf), 1.23 (s, 35 WO 2005/086638 PCT/US2005/004759 30H), 0.85 (t, J= 6 Hz, 3H); MS (ESI) inm/z calcd for C 31
H
55
N
2 0 4 S 551.84 (M+1), obsd 551.60. (2RS, 4 R)-2-(4-Acetylamino-phenyl)-thiazolidine-4-carboxylic acid octadecylamnide Hydrochloride (compound 18-HC1): 'H NMR (DMSO-d 6 ) 5 10.18 (s, 1H), 8.61 (m, 1H), 5 7.54-7.64 (m, 4H), 5.82 (s, 0.7H), 5.77 (s, 0.3H), 4.60 (min, 0.8H), 4.42 (m, 0.2H), 3.56-3.64 (m, 1H), 3.12-3.26 (m, 3H), 2.05 (s, 3H), 1.43 (m, 2H), 1.23 (s, 30H), 0.84 (t, J= 6 Hz, 3H); MS (ESI) m/z calcd for C 30
H
52
N
3 0 2 S 518.81 (M+1), obsd 518.70. (2RS, 4R)-2-(4-Fluoro-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 19): 1HNMR (CDC1 3 ) 8 7.46-7.54 (mn, 2H), 7.13-7.20 (m, 1H), 7.01-7.08 (m, 10 2H), 5.60 (s, 0.3H), 5.34 (s, 0.7H), 4.76 (m, 0.3H), 4.34 (m, 0.7H), 3.69 (dd, J= 11.1, 6.9 Hz, 1H), 3.21-3.52 (m, 3H), 1.49 (in, 2H), 1.26 (s, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (EST) mn/z calcd for C 2 8H 4 8FN 2 OS 479.75 (M+1), obsd 479.60. (2RS, 4R)-2-(4-Bromo-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 20): 'HNMR (CDC1 3 ) 8 7.48-7.62 (m, 2H), 7.36-7.42 (m, 2H), 7.14 (m, 0.7H), 15 6.40 (m, 0.3), 5.57 (d, J=10.2 Hz, 0.3H), 5.33 (d, J=11.1 Hz, 0.7H), 4.32 (m, 0.7H), 3.94 (in, 0.3H), 3.70 (dd, J= 11.1, 4.2 Hz, 1I), 3.20-3.44 (m, 3H), 1.49 (m, 2H), 1.27 (s, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESL) inm/z calcd for C 23
H
47 BrN 2 OS 539.66 (M+), obsd 539.70. (2RS, 4R)-2-(4-Nitro-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 21): 'H NMR (CDC1 3 ) 5 8.24 (d, J= 8.7 Hz, 2H), 7.67 (d, J= 8.7 Hz, 2H), 6.92 20 (min, 1H), 5.54 (s, 0.5H), 5.50(s, 0.5H), 4.24-4.31 (in, 1H), 3.67 (dd, J=10.8, 4.8 Hz, 1H), 3,27 3.44 (m, 3H), 1.55 On, 2.H), 1.26 (s, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) m/z called for
C
2 8
H
47
N
3 0 3 S 506.76 (M+1), obsd 506.60. (2RS, 4R)-2-(4-Cyano-phenyl)-thiazolidine-4-carboxylic acid octadccylarnide (compound 22): 1H NMR (CDC1 3 ) 8 7.60-7.70 (m, 4H), 6.94 (m, 0.6H), 6.37 (min, 0.4), 5.64 25 (s, 0.4H), 5.46 (s, 0.6H), 4.27 (m, 0.6H), 3.96 (m, 0.4H), 3.65-3.70 (m, 1H), 3.20-3.45 (m, 3H), 1.54 On, 2H), 1.26 (s, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ES1) m/z caled for
C
29
I
47
N
3 0S 485.77 (M.), obsd 508.50 (M+Na). (2RS, 4R)-2-(3,5-Difluoro-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 23): 1H NMR (CDCI 3 ) 8 7.04-7.08 (in, 2H), 6.97 (m, 1H11), 6.79 (m, 1H), 5.40 (s, 30 0.5H), 5.36 (s, 0.5H), 4.23-4.30 (m, 1H), 3.66 (dd, J=11.1, 4.5 Hz, 1H), 3.26-3.42 On, 3H), 1.33 (m, 2H), 1.26 (s, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) im/z calcd for C 28
H
47
F
2
N
2 0 2 S 497.74 (M+1), obsd 497.50. 36 WO 2005/086638 PCT/US2005/004759 (2RS, 4 R)-2-(2,6-Dichloro-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 24): 1H NMR (CDC1 3 ) 8 7.34-7.38 (m, 2H), 7.15-7.28 (m, 2H), 6.29 (s, 0.5H), 6.25 (s, 0.511), 4.25 (t, J= 5,7 Hz, 1H), 3.94 (dd, J=10.5, 1.8 Hz, 1H), 3.26-3.52 (m, 3H), 1.52 (in, 2H), 1.26 (s, 30H), 0.89 (t, J= 6 Hz, 3H); MS (ESI) m/z calcd for C 28
H
46 C1 2
N
2 0 2 S 5 529.65 (M), obsd 529.70. (2RS, 4R)-2-(3-Bromo-4-fluoro-phenyl)-thiazolidine-4-carboxylic acid octadecylamide (compound 25): 1H NMR (CDC1 3 ) 8 7.71 On, 1H), 7.42 (m, 1H), 7.06-7.16 (min, 211), 5.56 (d, J= 9.3 Hz, 0.2H), 5.34 (d, J= 10.2 Hz, 0.8H), 4.29 (d, J= 4.5 Hz, 0.811), 3.94 (m, 0.2H), 3.69 (dd, J=1 1,1, 4.2 Hz, 1H), 3.21-141 (min, 3H), 1.52 (m, 2H), 1.26 (s, 30H), 10 0.89 (t, .J= 6.3 Hz, 3H); MS (ESI) nm/z calcd for C 2 8
H
47 BrFN 2 OS 558.65 (M+1), obsd 558,70. (2RS, 4R)-2 p-Tolyl-thiazolidine-4-carboxylic acid octadecylamide (compound 26): 1H NMR (CDC1 3 ) 8 7.34-7.43 (m, 2H), 7.14-7.21 (m, 3H), 5.59 (s, 0.2H), 5.32 (s, 0.SH), 4.76 (in, 0.2H), 4.35 (m, 0.8H), 3.70 (dd, J= 11.1, 3.9 Hz, 1H), 3.21-3.43 (m, 311), 2.36 (d, J= 2.7 Hz, 3H), 1.51 (mn, 2H), 1.27 (s, 30H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) nm/z calcd for 15 C 29
H
51
N
2 0S 475.79 (M+1), obsd 475.60. (2RS, 4R)-2-Biphenyl-4-yl-thiazolidine-4-carboxylic acid octadecylamide Hydrochloride (compound 27*HC1): 1H NMR.(DMSO-d 6 ) 8 8.59 (m, 1H), 7,66-7.73 (m, 5H), 7.37-7.51 (m, 4H), 5.92 (s, 0.7H), 5.87 (s, 0.3H), 4.62 (m, 0.7H), 4.41 (m, 0.3H), 3.53 3.64 (m, 1H), 3.26-3.32 (min, 1H), 3.13-3.17 (m, 2H), 1.44 (m, 2H), 1.22 (s, 30H), 0.84 (t, J= 20 6.3 Hz, 3H); MS (ESI) nm/z calcd for C 34
H
53
N
2 0S 537.86 (M+1), obsd. 537.70. Example 2 - Synthesis of N-Acyt and N-sulfonyl Derivatives Thiazolldine Carboxylic Acid Amrides N-Acyl and N-sulfonyl derivatives (compounds 28 and 29) were synthesized from 25 compound 5 by standard procedures (scheme 2). Briefly, (2RS, 4R)-2-phenylthiazolidine-4 carboxylic acid octadecylamide (compound 5) was reacted with either acetic anhydride or methyl sulfonyl chloride, in pyridine, to afford the desired derivatives. (2RS, 4R)-3-Acetyl-2-phenylthiazolidine-4-carboxylic acid octadecylamide (compound 28): 1H NMR (CDC1 3 ) 8 7.31-7.41 (m, 5H), 6.01 (s, 1H), 5.12 (s, 1H), 3.73 (m, 30 1H), 3.40 (m, 1H), 3.31 (m, I H), 3.11-3.17 (m, IH), 2.00 (s, 3H), 1.27-1.33 (m, 32H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) nm/z calcd for C 30
H
50
N
2 0 2 S 502.80 (M ), obsd 502.60. (2RS,4R)-3-Methanesulfonyl-2-phenylthiazolidine-4-carboxylic acid octadecylamide (compound 29): IH NMR (CDCI 3 ) 6 7.65-7.68 (m, 2H), 7.32-7.36 (m, 3H), 6.20 (s, 1H), 4.63 37 WO 2005/086638 PCT/US2005/004759 (dd, J= 9, 6 Hz, 1H11), 3.67 d, J= 12, 6 Hz, 1H), 3.47 (dd, J= 12.3, 8.1 Hz, 1H), 3.04-3.13 (m, 2H), 3.02 (s, 3H), 1.27 (min, 32H), 0.89 (t, J= 6.3 Hz, 3H); MS (ESI) m/z called for
C
2 9
H
50
N
2 0 3
S
2 538.85 (M+), obsd 538.70. Based on the foregoing synthesis, it is expected that other acyl anhydrides (e.g., 5 containing larger alkyl groups) can also be prepared according to this same synthesis procedure (Badr et al., "Synthesis of Oxazolidines, Thiazolidines, and 5,6,7,8-Tetrahydro-lH, 3H-pyrrolo[1,2-c] oxazole (or thiazole)-1,3-diones from 3-Hydroxy- or 0-Mercapto--amino Acid Esters," Bull. Chem. Soc. Jpn. 54:1844-1847 (1981), which is hereby incorporated by reference in its entirety). 10 Example 3 - Synthesis of Thiazole Carboxylic Acid Amides The synthesis of thiazole derivative (compound 34) was accomplished starting from cysteine as shown in scheme 3. To a solution of DL-cysteine (3g, 24.76 rmmol) in MeOH (50 mL) at 0OC, SOC1 2 (2.76 15 mL, 37.14 mmol) was slowly added and warmed to room temperature then refluxed for 3 h. The reaction mixture was concentrated in vacuo to yield a residue. This residue was taken in to aqueous EtOH (1:1, 30 mL), NaHCO 3 (2.28 g, 27.23 mmol) was added, after 10 min benzaldehyde (2.5 mL, 24.76 mmol) was added and stirring continued for 3 h. CHC1 3 (200 mL) was added to the reaction mixture and washed with water, brine, dried (Na 2
SO
4 ) and 20 solvent was removed in vacuo. The crude product was purified by column chromatography to afford 2-phenylthiazolidine-4-carboxylic acid methyl ester (compound 31): yield 4.7 g, 85%; 1 H NMR (CDCl 3 ) 6 7.51-7.62 (m, 2H), 7.32-7.42 (m, 3H), 5.84 (s, 0.4H11), 5.58 (x, 0.4H), 4.24 (t, J= 6.3 Hz, 0.4H), 4.01 (t, J= 7.5 Hz, 0.6H), 3.83 (s, 3H), 3.39-3.55 (mn, 1H), 3.10-3.26 (m, 1H); MS (ESI) nm/z 224 (M+I). 25 Beginning with compound 31, 2-phenylthiazole-4-carboxylic acid methyl ester (compound 32) was synthesized following a reported procedure (Kue et al., "Essential Role for G Proteins in Prostate Cancer Cell Growth and Signaling" J. Urol. 164:2162-2167 (2000), which is hereby incorporated by reference in its entirety). Yield 033 g, 68%; 'H NMR (CDC1 3 ) 8 8.20 (s,11H), 8.0-8.04 (min, 2H), 7.45-7.50 (min, 3H), 4.0 (s, 3H); MS (ESI) m/z 220 30 (M+1). To a solution of compound 32 (0.5 g, 2.28 mmol) in MeOH (10 mL) at 0 0 C, IN NaOH (5 mL) was added and stirred for 2 h. To the reaction mixture EtOAc (30 mL), was added and acidified with 1N HC1. Extracted with EtOAc (3X50 mL), combined extracts were washed 38 WO 2005/086638 PCT/US2005/004759 with water, brine, dried (Na 2
SO
4 ) and solvent was - removed under vacua to give crude acid (compound 33), which was converted to 2-phcnylthiazole-4-carboxylic acid octadecylamide (compound 34) following the general procedure described in Example I above. Yield 0.30 g, 68%; 1 H NMR (CDC1 3 ) 6 8.10 (s, 1H), 7.96.7.93 (m, 2H), 7.46-7.50 (m, 3H), 3.49 (dd, J= 5 13.5, 6.9 Hz, 2H), 1.69 (m, 2H), 1.27 (m, 301-1), 0.89 (t, J= 6.3 Hz, 3H); MS (BSI) nm/z calcd for C 28
H
45
N
2 0S 457.73 (M+I), obsd 457.60. Table 1: compound R' R 2 R mp yield formula (oC) (%) 3-HCI Phenyl C7H15 H ND 80 C 1 7IH 27
CIN
2 0S 4-HCI Phenyl C 1 4
H
28 H 95 83 C 24
H
41 C1N 2 0S 5-HCI Phenyl CIS018H37 H 93 70 C 28
H
49
CIN
2 0S 6-IHCI Phenyl 0 1 9
H
39 H 85 78 C 29 Hsi 5
CIN
2 OS 7 n-dodecyl C181437 H 86 69 C 34
H
68
N
2 0S 8 Cyclohyxyl C 18
H
37 H 60 75 C 28
H
54
N
2 0S 9 Benzyl C 1 8
H
37 H 80 81 C 29 Hso 0
N
2 0S 10 3-indolyl C 18
H
37 H 125 65 C 30
H
49
N
3 0S 11 3-pyridinyl C 18
H
37 H 94 63 C 27
H
47
N
3 0S 12-HCI 3-furanyl C 1 8
H
37 H 99 60 C 26
H
47
CIN
2 0 2 S 13 4-dimethylaminophenyl C 18
H
37 H 75 75 C 30
H
53
N
3 0S 14 3-hydroxyphenyl C 1 sH 37 H 50 69 C 28
H
48
N
2 0 2 zS 15-HCI 4-methoxyphenyl C 1 8
H
37 H 95 70 C 29 HsICIN 2 0 2 S 16-HCI 3,4-dimethoxyphenyl C 1 8
H
37 H 103 83 C 30
H
53
CIN
2 0 3 S 17-HCI 3,4,5-trimethoxyphenyl C1sH37 H 115 70 C 31
H
ss
CIN
2 0 4 S 18-HCI 4-acetamidophenyl C 1 8
H
37 H 170 63 C 30
H
52
CIN
3 0 2 S 19 4-fluorophenyl ClSH37 H 65 73 C 28
H
47
FN
2 0S 20 4-bromophenyl C 1 8
H
37 H 81 77 C 28
H
47 BrN 2 OS 21 4-nicrophenyl C 18
H
37 H 115 60 C 28
H
47
N
3 0 3 S 22 4-cyanophenyl C 1 8
H
37 H 90 70 C 29
H
47
N
3 0S 23 3,5-difluorophenyl C18H37 H 113 70 C 28
H
46
F
2
N
2 OS 24 2,6-difluorophenyl CigH37 H 49 80 C 28
H
46
CI
2
N
2 0S 25 3-bromo-4-fluorophenyl Cg 18
H
37 H 100 78 C 28
H
46 BrFN 2 OS 26 4-methylphenyl C 18
H
37 H 120 73 C 29
H
50
N
2 0S 27-HCI Biphenyl CISH37 H 130 70 C 34 Hs 3
CIN
2 OS 28 Phenyl C 18
H
37 COCH3 90 95 C3 0 oHsoN 2 0 2 S 29 Phenyl CH37 SO 2 Me 55 90 C2qHsoN 2 0 3
S
2 10 Example 4 - Analysis of Selected Prostate Cancer Cell Lines by RT-PCR for LPA Receptor Expression DU-145, PC-3, and LNCaP human prostate cancer cells, and RH7777 rat hepatoma cells were obtained from American Type Culture Collection (Manassas, VA). Dr. Mitchell 39 WO 2005/086638 PCT/US2005/004759 Steiner at University of Tennessee Health Science Center, kindly provided PPG-1 and TSU PrI cells. Prostate cancer cells and RH7777 cells were maintained in RPMI 1640 medium and DMEM (Mediatech, Inc., Herndon, VA), respectively, supplemented with 10% fetal bovine serum (Gibco, Grand Island, NY) in 5% CO2 / 95% air humidified atmosphere at 37 0 C. 5 Total RNA was extracted using Trizol® reagent (Invitrogen Corp., Carlsbad, CA) according to the manufacturer's instruction. 0.5 Rg (LPAx) or 1 (LPA 2 and LPA 3 ) of total RNA was used to perform RT-PCR using SuperScriptTM One-Step RT-PCR with Platinum® Tag (Invitrogen Corp., Carlsbad, CA) with 0.2 gM of primers. The following primer pairs were used: 10 LPA 1 forward 5'-GCTCCACACACGGATGAGCAACC-3' (SEQ ID NO: 1), and LPA, reverse 5'-GTGGTCATTGCTGTGAACTCCAGC-3' (SEQ ID NO: 2);
LPA
2 forward 5'-CTGCTCAGCCGCTCCTATTTG-3' (SEQ ID NO: 3), and
LPA
2 reverse 5'-AGGAGCACCCACAAGTCATCAG-3' (SEQ ID NO: 4);
LPA
3 forward 5'-CCATAGCAACCTGACCAAAAAGAG-3' (SEQ ID NO: 5), and 15 LPA 3 reverse 5'-TCCTTGTAGGAGTAGATGATGGGG-3' (SEQ ID NO: 6); f3-actin forward 5'-GCTCGTCGTCGACAACGGCTC-3' (SEQ ID NO: 7), and 0-actin reverse 5'-CAAACATGATCTGGGTCATCTTCTC-3' (SEQ ID NO: 8). PCR conditions were as follows: After 2 min denaturation step at 94 oC, samples were subjected to 34 to 40 cycles at 94 'C for 30 see, 60 'C (LPA 1 ) or 58 oC (LPA 2 and LPA 3 ) for 20 30 see, and 72 oC for I min, followed by an additional elongation step at 72 'C for 7 min. Primers were selected to span at least one intron of the genomic sequence to detect genomic DNA contamination. The PCR products were separated on 1.5% agarose gels, stained with ethidium bromide, and the band intensity was quantified using Quantity One Software (Bio Rad Laboratories, Inc., Hercules, CA). Expression levels of each receptor subtype in different 25 cell lines were expressed as ratios compared to i-actin mRNA level. LPL receptor expression in these cell lines was determined to validate their use as in vitro models (see Table 2 below). 1 Rg of total RNA was subjected to RT-PCR, the PCR products were separated on agarose gels, and relative expression level of each receptor subtype compared to /3-actin was quantified by Quantity One Software (Bio-Rad). LPA 1 was 30 the predominant LPL receptor expressed in these cell lines. However, LNCaP cells did not express this receptor subtype. LPA 3 receptor was uniquely expressed in prostate cancer cell lines. RH7777 cells do not express any of the known LPL receptors. 40 WO 2005/086638 PCT/US2005/004759 Table 2: LPL Receptor mRNA Expression LPL Old Expression level relative to /3-actin Receptor Name RH777 DU145 PC-3 LNCaP PPC-1 TSU-P1S
LPA
1 EDG-2 UDa 2.16 2.53 UD 2.29 2.13
LPA
2 EDG-4 UD 0.33 0.43 0.32 0.41 0.19
LPA
3 EDG-7 UD 0.07 0.27 0.28 0.15 UD Sum LPA 1
-
3 0 2.56 3.23 0.60 2.85 2.32 aUD=under detection limit Example 5 - Cytotoxicity Assay in Prostate Cancer Cells 5 For in vitro cytotoxicity screening, 1000 to 5000 cells were plated into each well of 96-well plates depending on growth rate, and exposed to different concentrations of a test compound for 96 h in three to five replicates. All the compounds were dissolved in dimethyl sulfoxide at 5 to 20 mM, and diluted to desired concentrations in complete culture medium. Cell numbers at the end of the drug treatment were measured by the SRB assay (Gududuru et 10 al., "Synthesis and Biological Evaluation of Novel Cytotoxic Phospholipids for Prostate Cancer," Bioorg. Med. Chemn. Lett. 14:4919-4923 (2004); Rubinstein et at., "Comparison of in vitro Anticancer-Drug-Screening Data Generated with a Tetrazolium Assay Versus a Protein Assay Against a Diverse Panel of Human Tumor Cell Lines," J Naatl, Cancer Inst. 82:1113-1118 (1990), each of which is hereby incorporated by reference in its entirety). 15 Briefly, the cells were fixed with 10% of trichioroacetic acid, stained with 0.4% SRB, and the absorbances at 540 mn was measured using a plate reader (DYNEX Technologies, Chantilly, VA). Percentages of cell survival versus drug concentrations were plotted and the IC 50 (concentration that inhibited cell growth by 50% of untreated control) values were obtained by nonlinear regression analysis using WinNonlin (Pharsight Corporation, Mountain View, 20 CA). 5-fluorouracil was used as a positive control to compare potencies of the new compounds. A sandwich ELISA (Roche, Mannheim, Genrmany) utilizing monoclonal antibodies specific for DNA and histones was used to quantify degree of apoptosis induced by the analogs after 72 h exposure. This assay measures DNA-histone complexes (mono- and 25 oligonucleosomes) released into cytoplasm from the nucleus during apoptosis. RH7777 cells were employed because of nonspecific cytotoxicity of compound 4 in receptor-negative cells as well as receptor-positive prostate cancer cells. The ability of 2-aryl-thiazolidine derivatives (ATCAAs) to inhibit the growth of five human prostate cancer cell lines (DU-145, PC-3, LNCaP, PPG-1, and TSU-Prl) was assessed 41 WO 2005/086638 PCT/US2005/004759 using the sulforhodamine B (SRB) assay (described above). A control cell line (RH7777) that does not express LPL receptors (Svetlov et al., "EDG Receptors and Hepatic Pathophysiology of LPA and EDG-ology of Liver Injury," Biochimnica et Biophysica ACT 1582:251-256 (2002), which is hereby incorporated by reference in its entirety) was also 5 utilized to understand whether the antiproliferative activity of these derivatives is mediated through inhibition of LPL receptors. The diastereomeric mixtures of the target compounds 3-29 were used as such to evaluate their in vitro inhibitory activity against prostate cancer cell lines, and the results are summarized in Tables 3 and 4 below. 5-Fluorouracil was used as the reference drug. To 10 deduce sound structure-activity relationships, ICs 50 s should on principle be determined on pure isomers. One drawback of testing mixtures of stereoisomers, unavoidable in this case, was that the effect of each stereoisomer on the biological activity could not be assessed. On the other hand, the IC 50 values calculated can be used as a screening method to select promising selective cytotoxic agents and to identify the diastereomeric mixture with the best 15 availability to inhibit the growth of prostate cancer cells. Many of these thiazolidine analogs were very effective in killing prostate cancer cell lines with IC50 values in the low/sub micromolar range (Table 3). Examination of the cytotoxic effects of compounds 3-5 shows that as the chain length increases from C7 to C,8, the potency also increases. However, a further increase in the alkyl chain length by one carbon unit (i.e., Cs 18 to C 19 ) caused a 20 significant loss in cytotoxicity. Interestingly, C 14 derivative (compound 4) demonstrated higher potency than compound 5, but was 8-fold less selective against RH7777 cell line. Thus, an alkyl chain with a Cis unit is optimal for maintaining the potency and selectivity observed in this series of compounds. N-Acyl and N-sulfonyl derivatives (compounds 28 and 29) were less cytotoxic than parent compound 5. Replacement of the phenyl ring with an 25 alkyl or cyclohexyl group reduced the potency (compounds 7 and 8) relative to the thiazolidine (compound 5) derivative. Introduction of a methylene spacer separating the phenyl ring and the thiazolidine ring furnished a compound 9, which was less active than the parent compound 5. 42 WO 2005/086638 PCT/US2005/004759 Table 3: Antiproliferative effects of compounds 3-17 on prostate cancer cell lines Compd a IC 5 0 (gM) RH777 7 a DU-145 PC-3 b LNCaPb PPClb TSU-Prlb 3-HCI 52.2 44.9 38.5 12.4 34.7 28.0 4-HCI 3.4 2.4 3.0 1.4 1.3 2.0 5-HCI 25.6 5.4 7.8 2.1 2.0 5.0 6-HCI NA >20 NA 13.6 16.8 >20 7 -20 8.9 15.0 11.9 13.0 10.7 8 >20 >20 >20 12.8 9.3 >20 9 >20 15.3 16.4 4.4 4.0 11.2 10 >20 8.9 11.5 2.1 1.3 4.4 11 10.5 7.5 9.2 3.6 2.9 7.8 12-HCI 10.4 6.6 8.1 1.7 1.1 4.2 13 >20 5.3 6.0 1.6 1.1 3.0 14 31.0 5.7 6.7 1.7 1.2 4.0 15-HCI >20 8.7 -20 2.1 1.5 ND 16-HCI 10.3 4.5 5.2 0.85 0/58 2.4 17-HCI 11.4 3.9 4.0 0.82 0/48 2.4 5-FU ND 11.9 12.0 4.9 6.4 3.6 aControl cell line. bProstate cancer cell lines. ND--=not detectable. NA=no activity. Table 4: Antiproliferative effects of compounds 18-29 and 34 on prostate cancer cell line
IC
50 (RM) Compd RH7777a DU-145b PC-3b LNCaPb ppC.lb TSU-Prlb 18-HCI 21.1 3.1 5.6 1.3 0.55 0.94 19 17.4 5.7 6/8 1.9 2.1 5.4 20 >20 13.8 17.3 5.1 3.7 18.3 21 -20 15.3 -20 8.4 15.3 15.9 22 >20 >20 >20 5.9 5.0 >20 23 >20 >20 >20 11.2 10.6 >20 24 >20 >20 >20 13.1 17.1 ~20 25 -20 11.3 13.5 3.0 4.7 14.0 26 >20 10.5 12.8 1.9 1.9 8.0 27-HCI >20 >20 >20 >20 >20 >20 28 >20 ~20 ~20 16.1 12.6 >20 29 >20 >20 >20 >20 >20 >20 34 >20 >20 >20 >20 >20 >20 5-FU ND 11.9 12.0 4.9 6.4 3.6 5 aControl cell line. bprostate cancer cell lines. To understand the effect of unsaturation on potency and selectivity, and to overcome the problems associated with stereoisomers, the central thiazolidine core in compound 5 was replaced with a thiazole ring. However, thiazole derivative (compound 34) did not show any 10 activity below 20 pgM in both prostate and RH7777 cells, which indicates that thiazolidine 43 WO 2005/086638 PCT/US2005/004759 ring with two chiral centers plays an important role in providing potency and selectivity. Replacements of the phenyl ring with a heterocycle, such as an indole, pyridine or furan ring was investigated by synthesizing analogs (compounds 10-12). The furanyl derivative (compound 12) showed equivalent cytotoxicity as compound 5, but was 3-fold less selective 5 against RH7777 cells. The cytotoxicity data of compounds 13-27 provides a summary of a broad survey of phenyl ring substituted analogs, Examination of the IC 5 0 values of these analogs demonstrates a greater tolerance for diverse substituents in the phenyl ring. In general, the most potent analogues possessed electron-donating substituents, as exemplified by comparison of 10 compound 13, and compounds 16-18, relative to compound 5. One of the most active compounds (compound 18) with an IC 5 0 of 0.55 IM was 38-fold more selective in PPC-1 cells compared to RH7777 cells. On the other hand, thiazolidine analogs (compounds 19-25), with electron-withdrawing substituents demonstrated less cytotoxicity. Comparison of the potencies of compound 26 and compound 27, suggest that substitution of the phenyl ring with 15 a bulky group reduces the activity. From the LPL receptor mRNA expression studies (Table 2), it was evident that these cell lines serve as an excellent model system to explore the effects of LPL receptor. Given the structural similarity of SAPs to ceramide (and the known ability of ceramide to induce apoptosis), it was then determined whether the antiproliferative effects of thiazolidine analogs 20 were mediated via apoptotla events. The ability of the analogs to induce apoptosis in LNCaP, PC-3, and RH7777 cells was examined using a quantitative sandwich ELISA that measures DNA-histone complex released during apoptosis. The enrichment factor calculated (as ratio of OD405 in treated and un-treated cells) provides a quantitative assessment of the degree of apoptosis induced. Initially, only two compounds (4 & 5) were used for this study. Apoptotic 25 activity of analog (compound 4) was selective in prostate cancer cells despite nonselective cytotoxicity in RH7777 negative control cells (see Table 5 below). Analog compound 5 induced apoptosis in PC-3 and LNCaP cells, but to a lesser extent in PC-3 cells perhaps due to lower potency in this cell line. This data suggests that thiazolidine analogs may act as potent inducers of apoptosis and selectively kill a variety of prostate cancer cell lines. 30 44 WO 2005/086638 PCT/US2005/004759 Table 5: Thiazolidine Amides-Induced Apoptosis Compound for 72 h PC-3 LNCaP RH7777 2 gtM 1.8 14.1 2.6 4 5 gM 18.7 75.4 3.2 10 gM 54.0 80.7 2.5 2 gM 1.4 4.5 5 5 tM 2.3 45.2 ND 10 gM 3.4 37.1 20 !iM 12.7 26.1 These results are consistent with the assay testing LNCaP cells for DNA 5 fragmentation by agarose gel electrophoresis. LNCaP cells were treated with a thiazolidine derivative (compound 4 or 5) for 24 to 108 hours, and then total DNA was extracted from 2 x 106 cells by simple centrifugation method, treated with RNase and Proteinase K. After precipication in ethanol, DNA was reconstituted in Tris-EDTA buffer, separated on agarose gels, and visualized by ethidium bromide staining * (Herrmann et al., "A Rapid and Simple 10 Method for the Isolation of Apoptotic DNA Fragments," Nucl. Acids Res. 22:5506-5507 (1994), which is hereby incorporated by reference in its entirety). The results, shown in Figures 4A-B, demonstrate that both of these compounds induce cell apoptosis in the LNCaP prostate cancer cell line. As another assessment of cytotoxicity, AKT inhibition was measured. 30 pg of total 15 cellular protein from untreated control cells and compound-treated cells were separated by SDS-PAGE, transferred to nitrocellulose membrane, and total AKT and phospho-AKT were probed with anti-AKT and anti-phospho AKT antibody specific for AKT phosphorylated at Ser 473, respectively (Cell Signaling Technology, Beverly, MA). The immunoblots were visualized by enhanced, chemiluminescence, and changes of relative levels of phospho-AKT 20 compared to total AKT by analog treatment were quantified by densitometric analysis. Figure 5B graphically illustrates the immunological detection of AKT using anti-AKT and anti phospo-AKT, shown in Figure 5A. From the foregoing, it should be appreciated that the introduction off ring activating groups on the phenyl ring resulted in increasing potencies for prostate cancer cell lines. The 25 above results demonstrate several new anticancer agents (represented by compounds 16, 17, and 18) with low/sub micromolar cytoxicity and high selectivity. From this study, compound 18 emerged as one of the most potent and selective cytotoxic agents with an IC 5 0 of 0.55 tM 45 WO 2005/086638 PCT/US2005/004759 and 38-fold selectivity in PPC-1 cells. Further, the ability of these analogs to induce apoptosis in LNCaP, PC-3 and RH7777 cells provides an important clue to understand their mechanism of action. 5 Example 6 - Synthesis of Thiazolidinone Amides The synthesis of thiazolidinone derivatives (comnounds 65 -72) utilized straightforward chemistry as shown in scheme 4 (Figure 6), where 1 is 1. Various 4 thiazolidinones were synthesized following a reported procedure of condensing mercaptoacetic acid, glycine methyl ester, and aromatic aldehydes in a one-pot reaction, 10 followed by basic hydrolysis of the ester (Holmes et al., "Strategies for Combinatorial Organic Synthesis: Solution and Polymer-supported Synthesis of 4-thiazolidinones and 4 metathiazanones Derived from Amino Acids," J Org. Chemn. 60:7328-7333 (1995), which is hereby incorporated by reference in its entirety). Thiazolidinone amides were obtained by the treatment with appropriate amines in the presence of EDC/HOBt under standard conditions. 15 Compound 65 that has no side chain was synthesized from the corresponding acid as shown in Figure 6 (scheme 4). Thiazolidinone amides (compounds 73 -77) were synthesized by a simple and direct method (Schuemacher et al., "Condensation Between Isocyanates and Carboxylic Acids in the Presence of 4-dimethylaminopyridine (DMAP), a Mild and Efficient Synthesis of Amides," Synthesis 22:243-246 (2001), which is hereby incorporated by 20 reference in its entirety), which involves reaction of the acid compound 64a with different isocyanates in the presence of a catalytic amount of DMAP (Figure 7)(scheme 5). Exhaustive reduction of compound 68 using BH 3 THF under reflex conditions gave compound 79 (Figure 8) (scheme 6). Oxidation of 68 using H 2 0 2 and with KMnO 4 afforded sulfoxide (compound 80) and sulfone (compound 81), respectively, as shown in scheme 6. All 25 compounds were characterized by 1 H and 1 3 C NMR, mass spectroscopy and, in certain cases, elemental analysis. Compounds were obtained as mixtures of diastereomers and were used as such for the biological studies. Characteristic data for exemplary compounds 68, 71, 72, and 81 are provided below. 30 N-oetadecyl-2-(4-oxo-2-phenylthiazolidin-3-yl)acetamide (compound 68): 'H NMR (300 MHz, CDC1 3 ): 8 0.89 (t, J= 6.0 Hz, 3H), 1.26 (br s, 30H), 1.46 (m, 2H), 3.16-3.29 (m, 3H), 3,82 (d, J= 1.5 Hz, 2H), 4.20 (s, 0.5H), 4,25 (s, 0.5H), 5.83-5.85 (mn, 2H), 7.27-7.41 (m, 5H); 13C NMR (300 MHz, CDCI 3 ): 8 13.55, 22.13, 26.30, 28.69, 28.80, 28.88, 28.99, 29.03, 46 WO 2005/086638 PCT/US2005/004759 29.10, 29.14, 31.37, 32.13, 39.08, 45.88, 63.67, 127.05, 128.58, 128.96, 137.61, 166.30, 171.61; MS (ESI) m/z 511 [M+Na). Anal. Calcd for C29H 4 8
N
2 0 2 S; C, 71.26; H, 9.90; N, 5.73. Found: C, 71.18; H, 10.03; N, 5.79. 2
-(
2 -4methoxyphenyl)-4-oxothiazolidin-3-yl)-N-ocatadecylacetamide (compound 5 71): 1 H NMR (300 MHz, CDC1 3 ): 8 0.89 (t, J= 6.0 Hz, 3H), 1.26 (br s, 30H), 1.33 (s, 2H), 3.16-3.19 (m, 1H), 3.2-3.29 (mn, 2H), 3,80 (d, J= 0.9 Hz, 2H), 3.83 (s, 3H), 4.16 (s, 0.5H), 4.21 (s, 0.47H), 5.82 (s, 1H), 6.9 (dd, J= 1.8 Hz, 2H), 7.29 (dd, J= 1.5 Hz, 2H); 13C NMR (300 MHz, CDC13): 8 13.53, 22.12, 26.31, 28.70, 28.74, 28.79, 28.89, 28.99, 29.03, 29.09, 29.13, 31.36, 32.23, 39.06, 45.74, 54.79, 63.44, 128.64, 129.11, 159.97, 166.41, 171.47; MS 10 (ESI) m/z 541 [M+Na]. Anal. Calcd for C 30 H50 N 2 03 S: C, 69.45; H, 9.71; N, 5.40. Found: C, 69.30; H, 9.86; N, 5.43. 2-(2-(2,6-dichlorophenyl)-4-oxothiazolidin-3-yl)-N-octadecylacetamide (compound 72): 1H NMR (300 MHz, CDC1 3 ): 8 3.54 (d, J= 15.3 Hz, IH), 3.87 (s, 2H), 4.25 (d, J= 15.3 Hz, 1H), 5.88 (s, 1H), 7.10 (t, J= 1.8 Hz, 1H), 7.36-7.43 (m, 7H), 8.29 (s, 1H); 1 3 C NMR (300 15 MHz, CDC1 3 ): 8 32.35, 46.73, 64.40, 117,37, 123.85, 127.29, 128.74, 129.32, 134.59, 136.87, 138.61,165.14, 172.60; MS (ESI) nm/z 403 [M+Na]. Anal. Calcd for C 17 H14 C 12
N
2 02 S: C, 53.55; H, 3.70; N, 7.35. Found: C, 53.39; H, 3.47; N, 7.36. N-octadecyl-2-(4-oxo-2-phenyl-l-sulfonyl-thiazolidin-3-yl)acetamide (compound 81): 1H NMR (300 MHz, CDC1 3 ): 8 0.89 (t, J= 6.0 Hz, 3H), 1.26 (br s, 32H), 3.19-3.34 (m, 3H), 20 3.88-4.03 (dd, J= 16.5 Hz, 2H), 4.66 (s, 0.51), 4.72 (s, 0.5H), 5.67 (br s, 1H), 5.95 (s, 1H), 7.38 (m, 2H), 7.50-7.53 (m, 3H); " 3 C NMR (300 MHz, CDC1 3 ): 6 13.54, 22.12, 26.26, 28.66, 28.79, 28.96, 29.02, 29.09, 29,14, 31.36, 39.30, 44.35, 49.85, 81.32, 125.77, 128.43, 128.91, 130.55, 163.23, 165.30; MS (ESI) m/z 519 [M-H]. Anal. Calcd for C 29 H48 N 2 04 S: C, 66.88; H, 9.29; N, 5.38. Found: C, 66.68; H, 9.27; N, 5.41. 25 Example 7 - Cytotoxicity Assay The antiproliferative activity of all the synthesized compounds was evaluated against five human prostate cancer cell lines and in RH7777 cells (negative control) using the sulforhodamine B (SRB) assay (see description in Example 5 above). 5-Fluorouracil (5-FU) 30 was used as reference drug. As shown in Table 6, 4-thiazolidinone carboxylic acids (compounds 64a and 64b) were unable to inhibit the growth of any of the five prostate cancer cells below 50 AM. However, the corresponding amides (compounds 66 -68) showed higher activities. It was observed that an increase in the alkyl chain length [compounds 66 (10), 67 47 WO 2005/086638 PCT/US2005/004759 (C14), and 68 (C 18)] enhances the antiproliferative activity of these analogs in prostate cancer cells. Interestingly, the simple amide 65 without any long alkyl chain is not cytotoxic below 100 PtM, which indicates that the absence of an alkyl side chain causes a considerable decrease in antiproliferative effect. On the other hand, replacement of the alkyl chain with 5 various aryl side chains (compounds 73 -78) reduced the biological activity. Among this series, compound 73 is moderately cytotoxic, where as analogs (compounds 76 -78) displayed poor cytotoxicity in several prostate cancer cell lines. However, it is noteworthy to mention that thiazolidinone amides (compounds 74 and 75), with electron-withdrawing substituents on the aryl ring showed cytotoxicity in the range of 13-29 gM against all five 10 prostate cancer cell lines. Table 6; Antiproliferative effects of compounds 64a --64b and 65 -78 R1 Y IC 50 (PM) Compd R Y RH7777 a DU-145b PC-3 0 LNCaPb PPC- 1 TSU-Pr1b 64a phenyl OH ND >50 >50 >50 >50 >50 64b biphenyl OH >100 >100 >100 >100 >100 >100 65 phenyl NHz >100 >100 >100 >100 >100 >100 66 phenyl NIH-CloH 21 20.0 22.4 20.3 14.1 15.8 19.7 67 phenyl NIH-C 14
H
29 16.4 19.6 13.5 14.1 10.1 13.4 68 phenyl NH-ClaH 37 39.6 12.6 11.1 9.3 7.1 8.5 69 biphenyl NH-Cs 8 H37 >50 >50 >50 >50 >50 >50 70 dimethylamino NH-C 18
H
37 >50 >50 >50 >50 >50 >50 naphtalen-4-yl 71 4-methoxy NH"-Ca 8
H
37 31.1 14.8 12.6 11.8 10.7 17.5 phenyl 72 2,6-dichloro NH-CsH 37 >50 >50 >50 >50 >50 >50 phynyl 73 phenyl NH-3,5- 70.9 69.0 74.1 24.1 46.2 53.2 difluoro phenyl 74 phenyl NH-3,5-di(tri 25.4 16.2 18.1 14.5 13.1 16.1 fluoromethyl) phenyl 75 phenyl NH-3,5-di 34.9 24.0 28.6 13.2 20.5 17.2 chlorophenyl 76 phenyl NH-2,4- >100 >100 >100 82.5 >100 60/8 dimethoxy phenyl 77 phenyl NH-naphthyl >100 >100 >100 31.4 >100 69.9 78 phenyl 2,4dimethoxy >100 >100 >100 >100 >100 >100 phenylethyl 5-FU ND 11.9 12.0 4.9 6.4 3.6 aControl cell line. bProstate cancer cell lines. 48 WO 2005/086638 PCT/US2005/004759 Table 7: Antiproliferative effects of compounds 79-81 IC50 (tM) Compd RH77772 DU-145b PC-3b LNCaPb PPC-1b TSUb 79 >20 15.8 >20 >20 12.0 6.1 80 11.5 11.2 6.5 7.9 5.4 6.4 81 22.1 15.5 8.5 10.9 5.5 9.3 5-FU ND 11.9 12.0 4.9 6.4 3.6 aControl cell line. b Prostate cancer cell lines. ND=not detectable. NA--no activity. Thiazolidinone derivatives (compounds 69 and 70) with bulky biphenyl or 5 naphthalene groups demonstrated low cytotoxicity compared to compound 68 (Table 6). Compounds 71 and 72 were synthesized to understand the effects of aromatic ring substitution in compound 68. It was observed that electron-donating substituents maintained good activity while the ortho electron-withdrawing substituents substantially decrease the antiproliferative activity of these derivatives (Table 6). Compound 79, which has no amide 10 groups, showed significantly good potency in all five prostate cancer cell lines. Notably, compounds 80 and 81 bearing sulfoxide or sulfone moiety displayed higher cytotoxic potency comparable to that of the reference drug 5-FU against both PC-3 and PPC-I cell lines (Table 7). In summary, a series of novel and cytotoxic 4-thiazolidinone amides were prepared 15 and identified. Among this series, detailed structure activity relationship studies of type I compounds (Figure 6) were performed to evaluate their antiproliferative activity against five prostate cancer cell lines and RH7777 cells (negative controls). The cytotoxicity study shows that the antiproliferative activity is sensitive to 2-aryl ring substitutions, the length of the alkyl side chain, and the removal or replacements of the lipophilic alkyl side chain. Sulfur 20 oxidation is well tolerated as compounds 80 and 81 showed significant cytotoxicity compared to 5-FU. This study resulted in the discovery of potent cytotoxic 4-thiazolidinones (compounds 68, 80, and 81), which inhibit the growth of all five human prostate cancer cell lines (DU-145, PC-3, LNCaP, PPC-1, and TSU) with 2-5-fold lower selectivity compared to RH7777 cell line. These 4-thiazolidinone derivatives are a significant improvement on the 25 SAP moiety in that they are less cytotoxic but demonstrated improved selectivity in non tumor cells. 49 WO 2005/086638 PCT/US2005/004759 Example 8 - Cvtotoxicity Assay in Breast and Ovarian Cancer Cells The most potent compounds from each structural formula were selected and tested for their growth inhibitory activity in a human breast cancer cell line (MCF-7) and three human 5 ovarian cancer cell lines (CHO-I, CaOv-3, SKOv-3, and OVCAR-3). In vitro cytotoxicity assay was performed by the same sulforhodamine B (SRB) assay (described above). The compounds shown in Table 8 below where tested for activity against the breast cancer and ovarian cancer cell lines. 10 Table 8: Antiproliferative effects of compounds on breast and ovarian cancer cell lines Compd
IC
50 (tM) ompMCF.-7a CHO-b CaOv-3b OVCAR-3b SKOv-3b 3-HCI 50.3 NT 19.2 34.0 47.8 4-HCI 2.4 NT 13.9 1.6 2.1 5-HCI (R) 4.2 NT 2.5 4.5 8.5 5-IICI (S) 7.4 NT 18.0 5.2 18.0 6-HCI >20 NT NT NT NT 7 10.4 NT NT NT NT 8 -20 NT NT NT NT 9 18.7 NT NT NT NT 10 1-/7 NT NT NT NT 11 9.3 NT NT NT NT 12 NT NT 7.7 2.3 5.4 13 13.5 NT NT NT NT 14-HCI NT NT 18.3 8.1 11.0 15-HCI 16.3 NT NT NT NT 16-HCI NT NT 5.5 1.2 3.6 17-HCI NT NT 4.4 1.4 2.7 18-HCI NT NT 4.9 2.0 2.6 19 8.8 NT 5.5 2.3 4.2 20 16.6 NT NT NT NT 21 16.3 NT NT NT NT 24 17.7 NT NT NT NT 25 15.3 NT NT NT NT 26 10.3 NT NT NT NT 27-HCI >20 NT NT NT NT 28 16.3 NT NT NT NT 29 >20 NT NT NT NT 34 >20 NT NT NT NT 66 13.5 21.0 NT NT NT 67 8.9 11.4 NT NT NT 68 15.4 23.5 NT NT NT 69 >20 >20 NT NT NT 70 >20 >20 NT NT NT 50 WO 2005/086638 PCT/US2005/004759 71 13.0 15.2 NT NT NT 72 -30 >30 NT NT NT 80 14.3 11.6 NT NT NT 81 8.9 9.8 NT NT NT aBreast cancer cell line. bOvarian cancer cell lines. NT=not tested. Stereoselectivity of compound S was observed (compare the (R) and (S) isomers) in CaOV-3 and SKOv-3 cells. Substitutions on 2-phenyl ring generally increased cytotoxicity of 5 the compounds. Example 9 - Synthesis and Testing of Spermine-conjugated Thiazolidine Amide As illustrated in Figure 9, a mixture of 4-thiazolidinone acid (where R 1 is phenyl and 1 is 1) (1.5 g, 6.32 m mol), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride 10 (1.51 g, 7.9 m mol) and 1-hydroxybenzotriazole (0.85 g, 6.32 m mol) in CH 2 C1 2 was cooled in an ice bath was stirred for 10 min. To this solution 4-nitrophenol (0.78 g, 5.61 in mot) was added and stirred for 2h. The reaction mixture was diluted with CH 2 C1 2 washed sequentially with cold 5% HCt, saturated NaHCO 3 , water, brine, dried (anhydrous Na 2
SO
4 ) and solvent was removed in vacuo. The nitrophenyl ester product (compound 100) was purified by flash 15 chromatography (silica gel) using EtOAc/Hexanes to afford 1.76 g (78%). 1H NMR (CDC1 3 ) 6 3.70 (d, J= 18 Hz, 1H), 3.85 (d, J=1.2 Hz, 2H), 4.64 (d, J=17.7 Hz, 1H), 5.88 (s, 1H), 7.24 (d, J=2.1 Hz, 1H), 7.26 (d, J=2.4 Hz, 1H), 7.40-7.46 (min, 5H), 8.26 (d, J=1.8 Hz, 1H), 8.28 (d, J=2.1 Hz, 1H). To a solution of the nitrophenyl ester (compound 100) (0.5 g, 1.39 m mol) in CH 3 OH 20 (35 mL) at room temperature, a solution of spermine (0.33 g, 1.63 min mol, in CH 3 OH) was added slowly and stirred for lh. The reaction mixture was concentrated in vacuo, and to the concentrated reaction mixture 1:1 (CHC 13: CH 3 0OH) was added and filtered through celite. Solvent was removed in vacuo and the residue was purified by flash column chromatography (silica gel) using CHC13:CH 3 OH/i-PrNH 2 to give 0.2 g (50%) of spermine conjugate 25 (compound 101), which was converted to the corresponding hydrochloride salt using 2M HCI/Et 2 0. 'H NMR (DMSO-d 6 ) 8 1.71-1.76 (min, 6H), L95-2.0 (min, 2H), 2.89-3.0 (m, 10H), 3.0-3.15 (m, 4H), 3,74 (d, J=15.6 Hz, 1H), 3.87 (d, J=15.3 Hz, 1H), 4,10(d, J=16.5 Hz, IH), 7.35-7.44 (m, 5H), 8.0-8.18 (m, 4H), 8.89 (Ins, 2H), 9.15 (brs, 2H). ESIMS m/z 422.4 (M++1). 30 Compound 101 demonstrated more potent activity against prostate cancer cells compared to ovarian and MCF-7 breast cancer cells, with ICs 5 0 (M) values as follows: 51 WO 2005/086638 PCT/US2005/004759 RH7777 (>100), DU145 (12.4), PC-3 (11.1), LNCaP (26.2), PPC-1 (11.7), TSU-Prl (5.0), MCF-7 (>100), CaOv-3 (39.3), OVCAR-3 (39.7), and SKOv-3 (>100). Example 10 5 The antiproliferative effects of 3, 4, 5R, and 5S were compared to that observed with four active serine amide phosphates (SAPs) derivatives and 5-fluorouracil (5-FU, positive control) in human prostate cancer cell lines (PC-3, DU 145, LNCaP, PPC-1, TSU-Prl). A control cell line (RH7777) that does not express LPL receptors 4 and MCF-7 (a human breast cancer cell line) was included to gauge their selectivity. The chosen cell lines represent 10 different basal levels of active AKT and LPL receptor expression (discussed later). Cells were exposed to a wide range of concentrations (0 to 100 gM) of the indicated compound for 96 h. Cell numbers at the end of treatment were measured using the sulforhodamine B (SRB) assay 5 . ICs 50 (i.e., concentration that inhibited cell growth by 50% of untreated control) values were obtained by nonlinear regression analysis (WinNonlin, Pharsight Corp.). 15 As previously observed in our laboratory, the SAP derivatives (compounds S1-S4 in the Table below were potent inhibitors of tumor cell proliferation with IC 50 values ranging from 1.1 to ~ 20 pM (ND= not determined). Differences in SAP and thiazolidine antiproliferative activity were observed. The thiazolidine derivatives (3, 4, 5R, and 5S) also potently inhibited prostate and breast cancer 20 cell growth, but were 2- to 12-fold less potent in LPL receptor negative RH7777 cells, suggesting that thiazolidine analogs demonstrate more potent and selective antiproliferative activity. Two important structure-activity relationships were suggested in this small series of compounds. First, analogs containing long alkyl chains (i.e., Cs 18 ; 5R, and 5S) were more potent and selective than derivatives with shorter alkyl chain lengths (i.e., C 7 and C 1 4 ; 3 and 25 4). Secondly, the IC 5 0 for the R-isomer (5R) were less than the IC 5 0 for the S-isomer (5S) in all of the tumor cell lines, except for RH7777. This suggests a stereospecific interaction with a molecular target that is absent or less critical in RH7777 cells. Importantly, analogs 4, 5R, and SS were as potent inhibitors of tumor cell proliferation as 5-FU, and were measurably better in many cell lines. 30 52 WO 2005/086638 PCT/US2005/004759 ID Structure RH7777 DU-145 PC-3 LNCaP PC TSU-Pr MCF-7 0 S1 9 0 ND 5.7 15.3 5.8 1.8 5.0 5.8 HO-P-O N-CHIH 3 9 OH NH _H S2 00 S2 ND 10.8 -20 2.6 1.6 11.1 -20 HO-P-_ -. N-CzoH41 6 H NH,'H $3 "Y('N-c'°H4 S3 No H-CH 4 2.5 3.2 2.4 3.3 1.6 1.1 2.9 HO-_',r_'N- C2oH4, S4 HCIoo 2.9 4.1 2.6 5.1 1.9 2.2 4.6 0 UH. HN -C7 3 -C H 52.2 44.9 38,5 12.4 34.7 28.0 50.3 o CIH HN 111 N-C 4H29 -- 4 H 3.4 2.4 3.0 1.4 1.3 2.0 4.2 S 5R H N - c
,
8 25.6 5.4 7.8 2.1 2.0 5.0 4.2 o CIH.HN N-CIBH 37 5S H- ' 19.1 7.1 > 10 6.3 4.0 ~10 7.4 5-FU ND 11.9 12.0 4.9 6.4 3.6 ND Example 11 5 The cytotoxicity of thiazolidine and SAP derivatives in five human prostate cancer cell lines (DU-145, PC-3, LNCaP, PPC-1, TSU-Prl) and in a negative control cell line (RH7777) that lacks LPL receptor was examined using the sulforhodamine B (SRB) assay. Cells were exposed to a wide range of concentrations (0 to 100 pM) of the particular compound for 96 h in 96 well plates. Cells were fixed with 10% trichloroacetic acid, washed 10 five times with water. The plates were air dried overnight and fixed cells were stained with SRB solution. The cellular protein-bound SRB was measured at 540 nm using a plate reader. Cell numbers at the end of the treatment were measured. IC50 (i.e. concentration that inhibited cell growth by 50% of untreated control) values were obtained by nonlinear regression analysis using WinNonlin. For comparative purposes and to understand the 53 WO 2005/086638 PCT/US2005/004759 degree of cytotoxicity 5-fluorouracil (5-FU) was tested against all five prostate cancer cell lines. Compounds showing more potent antiproliferative activity will display low IC 5 0 values comparable to that of 5-fluorouracil.The results are summarized below.
IC
50 (pM) Structure CHO- DU PPC- TSU- MCF RH7777 PC-3 LNCaP K1 145 1 Prl 7 O K Not HN "' -OH >20 ~ 20 16.1 12.6 ~20 16.3
D
1 2
H
25 tested 20
C
1 2
H
25 S C12H25-- <O Not > No No > >
_ONH
4 . > 100 > 100 NH4 tested 100 toxicity toxicity 100 100 "ONH+4 0 HN '"OH Not > > > S > 100 > 100 71.0 ~ 100 tested 100 100 100 O HN ."%"OH Not - > > >100 >100 ~50 ~100 I S tested 100 100 100 MeO 0 HN O H Not > > > F S > 100 > 100 > 100 > 100 tested 100 100 100 F O HN "' OH Not > > > >100 >100 >100 >100 S tested 100 100 100 NC 54 WO 2005/086638 PCT/US2005/004759 O HN ' OH Not > > > > 100 > 100 > 100 > 100 S tested 100 100 100 F 0 Cl HN ")" OH Not > > > D > 100 > 100 > 100 > 100 S tested 100 100 100 0 l HN '' OH Not > > -- > 100 > 100 > 100 > 100 S tested 100 100 100 (N)
C
18
H
37 HO , NH c 18H37 Not > > > >100 >100 >100 >100 NHMe tested 100 100 100 HCI
C
18 Ha HO NH' 2.5 1.7 3.2 2.4 3.3 1.6 1.1 2.9
NH
2 HCI O Not > HO " NH(CH 2
)
12 HN OH 8.5 > 20 12.5 > 20 3.7 10.6 NHBoc NHBoc tested 20 Example 12 Prostate cell line LNCaP cells were treated with 30 RM of the compound of Formula: O HO
NH
C 18H37 NH 0 5 0 55 WO 2005/086638 PCT/US2005/004759 for the indicated period and time. The active form of AKT (Pi-AKT) and the O-Actin were quantified by Western blot analysis. The compound inhibited AKT phosphorylation to 50% by 12 hours of treatment. The compound of Fommla VIII had an IC 5 0 equal to 10.3 pM. The results of the experiment are given below. 5 Treatment with Relative abundance 30 gM (Pi-AKT//0-Actin) (-) control 1.00 (+) control 2.01 1 h 1.29 2h 1.01 6h 0.71 12 h 0.50 24 h 0.32 48 h <0.10 72 h <0.10 Example 13 Prostate cell line LNCaP cells were treated with 10 gM of the compound of Formula: HO/' NH
.
C
1 8
H
37 HO NH CygH37 NHMe HCI 10 for the indicated period and time. The active form of AKT (Pi-AKT) and AKT were quantified by Western blot analysis. The compound of Formula IX almost completely inhibited AKT phosphorylation within 6 hours of treatment. The compound an IC 5 0 equal to 3.3 pM. The results of the experiment are given below. Treatment with Relative abundance 10 pgM Formula IX (Pi-AKT/AKT) (-) control 1.00 6 h 0.07 12h 0.31 24 h 1.02 36h 0.63 48 h 0.94 15 56 WO 2005/086638 PCT/US2005/004759 Example 14 Prostate cell line LNCaP cells were treated with 10 gM of the compound of Formula HO ' NH .C1Hay NHMe HCI for the indicated period and time. The active form of AKT (Pi-AKT), AKT and 3-Actin were 5 quantified by Western blot analysis. The compound almost completely inhibited AKT phosphorylation by 1 hour of treatment. The compound had an IC 50 equal to 3.3 gM. The results of the experiment are given below. Treatment with Relative abundance Relative abundance 10 gM Formula IX (Pi-AKT/AKT) (Pi-AKT/03-Actin) (-) control 1.00 1.00 lh 0.07 0.08 2h 0.06 0.06 3h 0.04 0.04 4h 0.12 0.11 6h 0.12 0.10 8h 0.14 0.11 10 h 0.14 0.43 EXAMPLE 15 10 The cytotoxicity of synthesized compounds in five human prostate cancer cell lines (DU-145, PC-3, LNCaP, PPC-1, and TSU) and in two negative control cell lines (CHO and RH7777) was examined using the sulforhodamine B (SRB) assay (Rubinstein, L. V. S., R. H.Paull, K. D.Simon, R. M.Tosini, S.Skehan, P.Scudiero, D. A.Monks, A.Boyd, M. R. J Natl. Cancer. Inst. 1990, 82, 1113-1118, which is incorporated by reference herein). Cells were exposed to a wide range of 15 concentrations (0 to 100 rtM) of the particular compound for 96 h in 96-well plates. Cells were fixed with 10% trichloroacetic acid, washed five times with water. The plates were air dried overnight and fixed cells were stained with SRB solution. The cellular protein-bound SRB was measured at 540 nm using a plate reader. Cell numbers at the end of the treatment were calculated as a percentage of untreated control. IC 50 (i.e. concentration that inhibited cell growth by 50% of 20 untreated control) values were obtained by nonlinear regression analysis using WinNonlin. For 57 WO 2005/086638 PCT/US2005/004759 comparative purposes and to understand the degree of cytotoxicity 5-fluorouracil was tested against all five prostate cancer cell lines. The results are summarized in Table 1. From the cytotoxicity data it is clear that most of the compounds tested showed good anticancer activity against all five prostate cancer cell lines. SAAs (306b, 306e, 306f) without a 5 phosphate group are as effective as SAPs. A direct relationship was observed between length of the alkyl chain and cytotoxicity of the tested compounds. Accordingly, all of these compounds showed an alkyl chain length dependent cytotoxicity. Compounds with shorter alkyl chains (302a, 306b, 315d, 316d) are less cytotoxic than analogues with longer alkyl chains (see Table 1). Compound 302f emerged as one of the most potent SAPs tested so far with an IC 5 0 of 1.8 pM against PPC-1 10 cell line. However, SAAs are more potent than corresponding SAPs when the alkyl chain length is below 18C, but no significant difference in the cytotoxicity is observed between SAAs and SAPs with alkyl chain more than 18C. IC 5 0 values for enantiomers of SAAs (306c, 306d) and SAPs (302b, 302c) are approximately equivalent which suggests that chirality is not important for the antiproliferative activity of these compounds in prostate cancer. Introduction of a double bond in 15 the alkyl chain lowered the potency of both SAA 309 and SAP 311. To understand the importance of the amine functionality we derivatized the amine group to the corresponding Set B amide, sulfonamide and urea derivatives. Serine diamide phosphate 316d with a shorter alkyl chains failed to demonstrate cytotoxicity below 100 pM in four prostate cancer cell lines except TSU prostate cell line. The inhibitory activity of sulfonamide derivatives 315b and 316b and urea derivative 20 315c in all five prostate cancer cell lines showed a general decreasing trend suggesting that derivatization of C2 amine group is not tolerable for their ability to kill prostate cancer cells. To further investigate the extent of structural tolerance permitted in the serine amide backbone region we replaced the serine amide group with simple ethanolamine amide by synthesizing compounds 319 and 320. However, these ethanolamine amide analogs were less 25 potent and particularly compound 319 did not show any activity against DU-145, PC-3, and LNCaP prostate cancer cell lines. When the amide group in SAAs was reduced to produce long chain N-alkyl amino alcohols 317 and 318, these analogues retained cytotoxicity and were very effective in killing prostate cancer cell lines with low micromolar cytotoxicity. To determine the selectivity, several of synthesized compounds were also examined for their cytotoxicity in CHO 30 and RH7777 cells as negative controls. Many of the potent compounds showed similar cytotoxicity and were non selective in their action against prostate cancer cell lines and non-tumor negative control cells. 58 WO 2005/086638 PCT/US2005/004759 Table 9: ICs 50 (pM) of various compounds 0 o H
R
1 0 NHR 2
R
1 0 NHR 2
R
1 0 y NHR2 R 1 O N R 2 SH3 NHR3 NH2 R, + O SetA A Set B Set C Set D
IC
50 so (4M) Compd Set RH DU- LNCa
R
1 R2 R 3 CHO D PC-3 PPC-1 TSU 7777 145 P 302a (2R) PO3H C 1 oH 21 - ND ND 50.2 36.0 44.7 22.1 31.5 302b(2R) PO 3 H C 14
H
29 - ND ND 20.6 >50 10.1 >10 >10 302c(2S) PO3H C 14
H
29 - ND ND 32.0 >50 19.7 >10 >10 302d(2R) PO 3 H C 18
H
37 - ND ND 11.7 19.1 7.2 5.6 4.8 302e(2R) PO 3 H C 19
H
3 9 - 3.7 ND 5.7 15.3 5.8 1.8 5.0 302f(2S) PO 3 H C 20
H
4 1 - 7.8 ND 10.8 >20 3.6 1.8 11.1 306a(2S) H C 8
H
17 - >100 ND >100 >100 >100 >100 >100 306b(2R) H CloH 21 - ND ND 52.2 35.0 31.0 15.9 26.0 A 306c(2R) H C 1 4
H
29 - ND ND 8.2 10.2 8.1 6.3 7.5 306d(2S) H C 1 4
H
29 - ND ND 6.9 10.3 10.0 6.2 9.2 306e(2R) H Cs 18
H
37 - 2.5 2.6 5.4 5.2 3.8 2.2 4.4 306f(2R) H C19H3 9 - 2.4 3.2 5.1 5.3 5.3 1.8 3.9 306g(2S) H C 20
H
41 - 4.1 ND 7.0 6.6 3.9 2.6 6.6
CSH
1 7 309(2S) H CH:CH- - 5.2 6.8 6.9 5.9 6.6 5.1 5.5 CsHi6
C
8
H
17 311(2S) P0 3 1H CH:CHI- - 11.9 28.6 16.0 39.2 12.2 21.1 12.4 C8H16 314a(2S) OBn C 18
H
37 H 3.0 9.9 11.2 6.2 10.9 2.9 6.8 Not 314b(2S) OBn C 18
H
37
SO
2 Me >50 >50 >50 47.3 Active 16.7 >50 Active CO NiH 314c(2S) OBn C 18
H
37 Ph (3,5- 18.5 >20 20 >20 >20 >20 15.9 difluoro) 314d(2S) OBn C 8
H
17
COC
7
H
1 5 9.2 12.9 22.9 31.3 35.0 4.0 10.0 B 315b(2S) H Cs 18
H
37
SO
2 Me 12.9 9.2 23.1 13.6 16.0 10.2 20.5 CO NH 315c(2S) H C 8
H
37 Ph (3,5- 20 >20 20 >20 >20 >20 15.3 difluoro) 315d(2S) H C 8
H
17
COCH
15 >100 ND >100 81.5 >100 81.2 93.8 316b(2S) PO 3 H C 18
H
37 SOzMe >50 50 43.2 >50 15.1 17.8 35.7 Not 316d(2S) PO 3 H C 8
H
1 7
COC
7
H
15 >100 >100 >100 >100 Nt >100 79.0 Active 317(2R) H C 18
H
37 H 2.2 2.9 4.1 2.6 5.1 1.9 2.2 C S 318(2R) H C 1
IH
37 Me 1.7 2.5 3.2 2.4 3.3 1.6 1.1 59 WO 2005/086638 PCT/US2005/004759 CsH7- Not Not Not Not 319 H CH:CH- >20 >20 >20 319 H CH:CH- - >20 Active Active Active Active
C
8
H
1 7
-
Not 320 PO3H CH:CH- - >50 >50 >50 >50 50 >50
SC
7 11 14 Active 5-FU - - - - - 11.9 12.0 4.9 6.4 3.6 Although various embodiments have been depicted and described in detail herein, it will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be made without departing from the spirit of the invention and 5 these are therefore considered to be within the scope of the invention as defined in the claims which follow. 60

Claims (30)

1. A compound according to formula (V) or formula (VI) X5 Xs R2 R2 OaX ... ,X2R4,<- R :-X2 R4 Y SX R4, N -S R 1 (V) RI (VI) 5 wherein X 1 and X 2 are each optional, and each can be oxygen; X 5 is optional, and can be oxygen; R 1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or 10 unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or R5 R6 S-(CH 2 m R7 R9 R8 or-(CH 2 )m-Y 1 where m is an integer from 0 to 10 and Yi is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or 15 unsaturated mixed heterocycle; R 2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 hydrocarbon, Rlo-N(Z)-hydrocarbon- or Ri 0 -hydrocarbon- where the hydrocarbon group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a 20 saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated R11 R12 -(CH2)n R13 or unsaturated mixed heterocycle, or RIS R14 or -(CH 2 )n- y 2 where 61 WO 2005/086638 PCT/US2005/004759 n is an integer from 0 to 10 and Y2 is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; R 3 is nothing, hydrogen or an aliphatic or non-aliphatic straight- or branched-chain Cl 5 to C10 hydrocarbon; R 4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl; R 5 , R 6 , R 7 , R 5 , R 9 , R", R 12, R" 3 , R 14 , and R" 5 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to 10 C10 hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl; R i o 0 is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, 15 hydrocarbon-0O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-phenylalkyl-0O-hydrocarbon-, phenylalkyl-0O-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or 20 phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic or non-aliphatic straight- or branched-chain C1 to C10 group, and wherein each alkyl is a C1 to C10 alkyl; and Z is independently hydrogen or t-butoxycarbonyl. 25
2. The compound as claimed in claim 1 having the formula O NH_ O- C1,H37 NH O- C 18 H 37 S S 62 WO 2005/086638 PCT/US2005/004759 o HN OH 0 S HN " OH C 1 2 H 25 ' S 0 o jo 0 HN " LoH 0 HN - O H F OHN " OH MeOF ; NC 0 0 0 O O O HN "'OH CI HN "N OH HN OH F CS ;or N 5
3. A compound according to formula (VII) X3 R2 R3 R4-~ 0 R3 R4,N- O R1 (VII) wherein X 3 is optional and each can be oxygen; X 6 is oxygen or nitrogen; 10 R 1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or R5 R6 S-(CH2)m R7 R9 R8 or-(CH 2 )m--Y 1 where m is an integer from 0 to 10 and Y 1 is a 63 WO 2005/086638 PCT/US2005/004759 saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; R 2 is hydrogen, an aliphatic or non-aliphatic straight- or branched-chain C1 to C30 5 hydrocarbon, R 10 -N(Z)-hydrocarbon- or R'o-hydrocarbon- where the hydrocarbon group is an aliphatic or non-aliphatic straight- or branched-chain Cl to C30 hydrocarbon, a saturated or unsaturated cyclic hydrocarbon, a saturated or unsaturated N-heterocycle, a saturated or unsaturated 0-heterocycle, a saturated or unsaturated S-heterocycle, a saturated R1 R12 S- (CH2)n R13 or unsaturated mixed heterocycle, or R15 R14 or -(CH 2 )n- y 2 where 10 n is an integer from 0 to 10 and Y2 is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or unsaturated mixed heterocycle; R 3 is nothing, hydrogen or an aliphatic or non-aliphatic straight- or branched-chain C1 to C10 hydrocarbon; 15 R 4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl; R 5 , R 6 , R, R 8 , R 9 , R 11 , R 12 , R" , and R 5 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10I hydrocarbon, alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, 20 dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl; R 1 0 is H(Z)N-, H(Z)N-hydrocarbon-, H(Z)N-hydrocarbon-N(Z) -hydrocarbon-, H(Z)N-hydrocarbon-, O hydrocarbon-, hydrocarbon-O-hydrocarbon-, hydrocarbon-N(Z) hydrocarbon-, 25 H(Z)N-hydrocarbon-carbonyl-hydrocarbon-, hydrocarbon-carbonyl-hydrocarbon, H(Z)N-phenyl-, H(Z)N-phenylalkyi-, H(Z)N-phenylalkyl-N(Z)-hydrocarbon-, H(Z)N-pheny1alkyl-O-hydrocarbon-, phenylalkyl-O-hydrocarbon-, phenylalkyl-N(Z) -hydrocarbon-, H(Z)N-phenylalkyl-carbonyl-hydrocarbon-, or phenylalkyl-carbonyl-hydrocarbon-, wherein each hydrocarbon is independently an aliphatic 64 WO 2005/086638 PCT/US2005/004759 or non-aliphatic straight- or branched-chain C1 to C10 group, and wherein each alkyl is a C1 to C 10 alkyl; and Z is independently hydrogen or t-butoxycarbonyl. 5
4. The compound as claimed in claim 3 having the formula O HCI. NH NH-(CH 2 )nCH 3 0 ,wherein n= 6, 13, or 17.
5. A compound according to formula (VIII) O 0 R4N NH-(CH 2 )n-NH R4 NN 7 RI X8R X8 RI (VIII) 10 wherein X 8 is O or S; n is between I and 30; R 1 is selected from the group of saturated or unsaturated cyclic hydrocarbons, saturated or unsaturated N-heterocyeles, saturated or unsaturated O-heterocycles, saturated or 15 unsaturated S-heterocycles, saturated or unsaturated mixed heterocycles, aliphatic or non aliphatic straight- or branched-chain C1 to C30 hydrocarbons, or Rs5 R6 S- (CH)m R7 R9 R or-(CH 2 )m-Y' 1 where min is an integer from 0 to 10 and Y' is a saturated or unsaturated cyclic hydrocarbon, saturated or unsaturated N-heterocycle, saturated or unsaturated O-heterocycle, saturated or unsaturated S-heterocycle, or saturated or 20 unsaturated mixed heterocycle; 65 WO 2005/086638 PCT/US2005/004759 R4 is optional, or can be hydrogen, an aliphatic or non-aliphatic straight- or branched chain C1 to C10 hydrocarbon, aryl, acetyl, or mesyl; and R 5 , R 6 , R 7 , R 8 , and R 9 are independently selected from the group of hydrogen, hydroxyl, an aliphatic or non-aliphatic straight-or branched-chain C1 to C10 hydrocarbon, 5 alkoxy, aryloxy, nitro, cyano, chloro, fluoro, bromo, iodo, haloalkyl, dihaloalkyl, trihaloalkyl, amino, alkylamino, dialkylamino, acylamino, arylamido, amido, alkylamido, dialkylamido, arylamido, aryl, C5 to C7 cycloalkyl, arylalkyl.
6. A compound according to formula (IX), formula (X), formula (XI), or formula (XII) X9 X9 R16 Ri6 HO NH X7 NH NH NH 10 R17 R18 (IX) R17 R 18 (X) HONH R16 NH R16 0 (XI) 0 (XII) 15 wherein X 7 is PO 3 H or O-benzyl; X 9 is O or nothing; R 16 is a C1 to C30 aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C 1 to C30 hydrocarbon; 20 R and R 18 are independently nothing, hydrogen, -SO 2 R" 9 , COR" 9 , and R 9 ; and R1 9 is an aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, Cl1 to C30 hydrocarbon or a substituted or unsubstituted aryl, with the proviso that that R1 6 is not C 1 4 H 29 when X 7 is PO 3 H and X 8 is O. 25
7. The compound as claimed in claim 6 wherein X 9 is O, X 7 is PO 3 H, and R 16 is a C7 to C20 straight-chain or branched, aliphatic or non-aliphatic hydrocarbon. 66 WO 2005/086638 PCT/US2005/004759
8. The compound as claimed in claim 6 wherein X 9 is O.
9. The compound as claimed in claim 6 wherein X 7 is PO 3 H. 5
10. The compound as claimed in claim 6 wherein R 16 is a C7 to C20 straight-chain or branched, aliphatic or non-aliphatic hydrocarbon.
11. The compound as claimed in claim 6 wherein R 17 and R 18 are hydrogen. 10
12. A compound according to formula (XIV) and formula (XV) O O CygH25-;, O Ow 0 HO NH(CH 2 ) 12 HN OH O O-P--ONH 4 I + NHBoc NHBoc (XIV) ONH+ 4 (XV).
13. A pharmaceutical composition comprising: 15 a pharmaceutically acceptable carrier and a compound according to any one of claims 1, 3, 5, and 12 or salt thereof.
14. A pharmaceutical composition comprising: a pharmaceutically acceptable carrier and 20 a compound according to formula (IX), formula (X), formula (XI), or formula (XII) X9 X9 R16 R16 HO NH X7 NH NH NH R17 RI8 (TX) R17 RIB (X) 67 WO 2005/086638 PCT/US2005/004759 HO NH R16 X7 NH R16 0 (XI) 0 (XII) wherein X 7 is PO 3 H or O-benzyl; 5 X 9 is O or nothing; R is a C1 to C30 aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C1 to C30 hydrocarbon; R 1 7 and R 1 8 are independently nothing, hydrogen, -SO 2 R' 9 , COR 9 , and R 9 ; and R1 9 is an aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or 10 unsubstituted, CI to C30 hydrocarbon or a substituted or unsubstituted aryl or salt thereof.
15. A method of destroying a cancer cell comprising: providing a compound according to any one of claims 1, 3, 5, and 12 and 15 contacting a cancer cell with the compound under conditions effective to destroy the contacted cancer cell.
16. The method as claimed in claim 15 wherein said contacting occurs ex vivo. 20
17. The method as claimed in claim 15 wherein said contacting occurs in vivo.
18. The method according to claim 15 wherein said cancer cell is selected from a prostate cancer cell, a breast cancer cell, and an ovarian cancer cell. 25
19. A method of destroying a cancer cell comprising: providing a compound according to formula (IX), formula (X), formula (XI), or formula (XII) 68 WO 2005/086638 PCT/US2005/004759 X9 X9 R16 R16 HO NH X7 NH NH NH R17 N R18 (IX) R17 R18 (X) HONH R16 X7 NH R16 HO v "X7v S(XI) 0 5 (XII) wherein X 7 is PO 3 H or O-benzyl; X 9 is O or nothing; R 6 is a C1 to C30 aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, 10 substituted or unsubstituted, C1 to C30 hydrocarbon; R17 and R 18 are independently nothing, hydrogen, -SO 2 R 1 9 , COR" 9 , and R 19 ; and R19 is an aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C1 to C30 hydrocarbon or a substituted or unsubstituted aryl; and contacting a cancer cell with the compound under conditions effective to destroy the 15 contacted cancer cell.
20. The method as claimed in claim 19 wherein said contacting occurs ex vivo.
21. The method as claimed in claim 19 wherein said contacting occurs in vivo. 20
22. The method according to claim 19 wherein said cancer cell is selected from a prostate cancer cell, a breast cancer cell, and an ovarian cancer cell.
23. A method of treating or preventing a cancerous condition comprising: 25 providing a compound according to at least one of claims 1, 3, 5, and 12; administering an amount of the compound to a patient in a manner effective to treat or prevent a cancerous condition. 69 WO 2005/086638 PCT/US2005/004759
24. The method as claimed in claim 23 wherein the cancerous condition is prostate cancer, breast cancer, or ovarian cancer. 5
25. The method as claimed in claim 23 wherein the patient is characterized by the presence of a precancerous condition, and said administering is effective to prevent or slow the development of the precancerous condition into the cancerous condition.
26. The method as claimed in claim 23 wherein the patient is characterized by the 10 presence of a cancerous condition, and said administering is effective either to cause regression of the cancerous condition or to inhibit progression of the cancerous condition.
27. A method of treating or preventing a cancerous condition comprising: 15 providing a compound according to formula (IX), formula (X), formula (XI), or formula (XII) X9 X9 R 16 R1 6 HO NH X7 NH NH NH R17 " R1 8 (IX) R17 N R18 (X) HONH R NH R16 HO o(X7 20 O (XI) (XII) wherein X 7 is PO 3 H or O-benzyl; X 9 is O or nothing; 25 R 16 is a C1 to C30 aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C1 to C30 hydrocarbon; R 17 and R is are independently nothing, hydrogen, -SO 2 R 19 , COR 1 9 , and R' 9 ; and 70 WO 2005/086638 PCT/US2005/004759 R1 9 is an aliphatic or non-aliphatic, straight-, cyclic- or branched-chain, substituted or unsubstituted, C1 to C30 hydrocarbon or a substituted or unsubstituted aryl; administering an amount of the compound to a patient in a manner effective to treat or prevent a cancerous condition. 5
28. The method as claimed in claim 27 wherein the cancerous condition is prostate cancer, breast cancer, or ovarian cancer.
29. The method as claimed in claim 27 wherein the patient is characterized by the 10 presence of a precancerous condition, and said administering is effective to prevent development of the precancerous condition into the cancerous condition.
30. The method as claimed in claim 27 wherein the patient is characterized by the presence of a cancerous condition, and said administering is effective either to cause 15 regression of the cancerous condition or to inhibit growth of the cancerous condition. 71
AU2005220705A 2004-02-11 2005-02-11 Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof Abandoned AU2005220705A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US54372404P 2004-02-11 2004-02-11
US60/543,724 2004-02-11
US55580304P 2004-03-24 2004-03-24
US60/555,803 2004-03-24
PCT/US2005/004759 WO2005086638A2 (en) 2004-02-11 2005-02-11 Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof

Publications (1)

Publication Number Publication Date
AU2005220705A1 true AU2005220705A1 (en) 2005-09-22

Family

ID=34976061

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005220705A Abandoned AU2005220705A1 (en) 2004-02-11 2005-02-11 Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof

Country Status (8)

Country Link
EP (1) EP1723127A4 (en)
JP (1) JP2007522247A (en)
AU (1) AU2005220705A1 (en)
BR (1) BRPI0507612A (en)
CA (1) CA2559333A1 (en)
EA (1) EA200601471A1 (en)
IL (1) IL177444A0 (en)
WO (1) WO2005086638A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115894398A (en) * 2022-12-02 2023-04-04 青岛科技大学 Fluorine-containing thiazole amide insecticide and acaricide

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101395146B1 (en) * 2011-02-09 2014-05-16 부산대학교 산학협력단 New compounds having skin whitening, antioxidant and PPAR activity, and medical use thereof
WO2014066613A2 (en) * 2012-10-26 2014-05-01 Zhejiang University Sphingolipid metabolite mimetics
EP2968346B1 (en) * 2013-03-15 2024-02-07 Cancer Research Technology, LLC Methods and compositions for gamma-glutamyl cycle modulation
WO2023170024A1 (en) * 2022-03-11 2023-09-14 University Of Copenhagen Camk2 modulators and their use in medicine

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4962096A (en) * 1987-03-24 1990-10-09 Nippon Chemiphar Co., Ltd. Heterocyclic glycerol derivatives and their use as anti-hypertensive agents
US4826990A (en) * 1987-09-30 1989-05-02 American Home Products Corporation 2-aryl substituted heterocyclic compounds as antiallergic and antiinflammatory agents
US5149704A (en) * 1991-05-03 1992-09-22 Abbott Laboratories Platelet activating antagonists
FR2678938B1 (en) * 1991-07-10 1993-10-08 Rhone Poulenc Rorer Sa PYRROLIDINE DERIVATIVES, THEIR PREPARATION AND THE MEDICINAL PRODUCTS CONTAINING THEM.
FR2700168B1 (en) * 1993-01-07 1995-02-03 Rhone Poulenc Rorer Sa Thiazolidine derivatives, their preparation and the drugs containing them.
DE19546918A1 (en) * 1995-12-15 1997-06-19 Bayer Ag Bicyclic heterocycles

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115894398A (en) * 2022-12-02 2023-04-04 青岛科技大学 Fluorine-containing thiazole amide insecticide and acaricide
CN115894398B (en) * 2022-12-02 2024-03-12 青岛科技大学 Fluothiazole amide-containing insecticidal acaricide

Also Published As

Publication number Publication date
WO2005086638A3 (en) 2005-11-24
CA2559333A1 (en) 2005-09-22
BRPI0507612A (en) 2007-07-03
EP1723127A4 (en) 2009-10-28
EA200601471A1 (en) 2007-10-26
JP2007522247A (en) 2007-08-09
WO2005086638A2 (en) 2005-09-22
EP1723127A2 (en) 2006-11-22
IL177444A0 (en) 2006-12-10

Similar Documents

Publication Publication Date Title
US20060014740A1 (en) Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof
US20090143446A1 (en) Thiazolidinone amides, thiazolidine carboxylic acid amides, methods of making, and uses thereof
CA2962524C (en) (2-(1-h-indolyl)imidazol-4-yl)(3,4,5-trimethoxyphenyl)methanone useful for the treatment of cancer
CA2657107C (en) Human protein tyrosine phosphatase inhibitors and methods of use
US20090324581A1 (en) Heteroarylamide lower carboxylic acid derivative
MXPA02005456A (en) Substituted oxazoles and thiazoles derivatives as hppar alpha activators.
EP2744807A1 (en) Lysophosphatidic acid receptor antagonists
JP5947724B2 (en) New inhibitors of cyclophilin and their use
US20080255213A1 (en) Thiazolidinone amides, thiazolidine carboxylic acid amides, and serine amides, including polyamine conjugates thereof, as selective anti-cancer agents
AU2005220705A1 (en) Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof
EP3773566A1 (en) Ox2r compounds
EP1926487A2 (en) Thiazolidinone amides, thiazolidine carboxylic acid amides, and serine amides, including polyamine conjugates thereof, as selective anti-cancer agents
JP6795525B2 (en) Arylsulfonamide compounds as carbonic anhydrase inhibitors and their therapeutic use
MXPA06009242A (en) Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof
KR20170072226A (en) Benzylhydroxyde derivatives, preparation thereof and therapeutic use thereof

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period