Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CURCUMIN ANALOGUES FOR TREATING CANCER
Document Type and Number:
WIPO Patent Application WO/2001/040188
Kind Code:
A1
Abstract:
The present invention is directed to curcumin analogs (I), wherein Y is OH, halogen, or CF¿3?; Z is H, OH, OR¿1?, halogen, or CF¿3?; X¿1? and X¿2? are independently C or N; and A is as defined in the application; exhibiting anti-tumor and anti-angiogenic properties, pharmaceutical formulations including such compounds and methods of using such compounds.

Inventors:
SNYDER JAMES P (US)
DAVIS MATTHEW C (US)
ADAMS BRIAN (US)
SHOJI MAMORU (US)
LIOTTA DENNIS C (US)
FERSTL EVA M (US)
SUNAY USTUN B (US)
Application Number:
PCT/US2000/032870
Publication Date:
June 07, 2001
Filing Date:
December 04, 2000
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV EMORY (US)
SNYDER JAMES P (US)
DAVIS MATTHEW C (US)
ADAMS BRIAN (US)
SHOJI MAMORU (US)
LIOTTA DENNIS C (US)
FERSTL EVA M (US)
SUNAY USTUN B (US)
International Classes:
A61K31/065; A61K31/10; A61K31/121; A61K31/122; A61K31/15; A61K31/351; A61K31/352; C07D317/54; A61K31/36; A61K31/404; A61K31/4439; A61K31/45; A61K31/4545; A61K31/46; A61P35/00; A61P43/00; C07C33/28; C07C33/48; C07C43/23; C07C45/29; C07C45/62; C07C45/74; C07C49/233; C07C49/235; C07C49/248; C07C49/255; C07C49/697; C07C49/747; C07C49/753; C07C225/22; C07C235/34; C07C251/40; C07C275/24; C07C317/10; C07C317/18; C07C321/20; C07D209/34; C07D213/63; C07D213/68; C07D295/205; C07D309/20; C07D309/22; C07D309/38; C07D311/58; C07D317/50; C07D401/06; C07D401/14; C07D451/02; C07D451/06; (IPC1-7): C07D213/63; A61P35/00; C07C49/235; C07C49/248; C07C317/10; C07C33/48; C07D451/02
Other References:
EL-SUBBAGH, HUSSEIN I. ET AL: "Synthesis and biological evaluation of certain.alpha.,.beta.-unsaturated ketones and their corresponding fused pyridines as antiviral and cytotoxi agents", J. MED. CHEM. (2000), 43(15), 2915-2921, XP002162598
TEUSCHER, E. ET AL: "Potential antiangiogenic substances among.alpha.,.alpha.'- bis(amidinobenzyl)cycloalkanone derivatives and 4-amidinoanilides of.alpha.-(arylsulfonylamino)-.omega.-phenylcarboxylic acids", PHARMAZIE (1987), 42(2), 109-10, XP000942237
Attorney, Agent or Firm:
Humphrey, Christopher M. (P.O. Drawer 34009 Charlotte, NC, US)
Download PDF:
Claims:
THAT WHICH IS CLAIMED:
1. A compound of the Formula (I) wherein: Y is OH, halogen, or CF3 ; Z is H, OH, OR1, halogen, or CF3 ; X1 and X2 are independently C or N; and A is selected from the group consisting of : wherein n is 18; X3 is O, S, SO, S02, NH, or NRI ; Q is NH or NRI ; and Vl are each independently OH, OR2, or halogen; Rl and R2 are independently H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl or dialkylaminocarbonyl ; the dashed lines indicate the presence of optional double bonds; and L is the point of bonding of A to the compound structure, with the proviso that Z is not H when Y is OH, Cl or Br and A is and pharmaceutically acceptable salts thereof.
2. A compound according to Claim 1, wherein Y is flourine.
3. A compound selected from the group consisting of : 1,5Bis(2, 4difluorophenyl) penta1,4diene3one; 3,5Bis (2fluorobenzylidene)piperidin4oneacetate; and 3,5Bis (2hydroxybenzylidene) tetrahydro4Hpyran4one.
4. A pharmaceutical formulation comprising a compound of Claim 1 in a pharmaceutically acceptable carrier.
5. A method of treating cancerous tissue in a subject, comprising administering to the subject an effective amount of a compound of formula (I) wherein: Y is OH, halogen, or CF3 ; Z is H, OH, OR1, halogen, or CF3; Xi and X2 are independently C or N; and A is selected from the group consisting of : wherein n is 18; X3 is O, S, SO, S02, NH, or NRI ; Q is NH or NRI ; and VI4 are each independently OH, OR2, or halogen ; Ri and R2 are independently H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl or dialkylaminocarbonyl ; the dashed lines indicate the presence of optional double bonds ; and L is the point of bonding of A to the compound structure, with the proviso that Z is not H when Y is OH, Cl or Br and A is and pharmaceutically acceptable salts thereof.
6. A method according to Claim 5, wherein the effective amount comprises an amount sufficient to inhibit VEGF production in the cancerous tissue.
7. A method according to Claim 5, wherein the effective amount comprises an amount sufficient to inhibit TF production in the cancerous tissue.
8. A method according to Claim 5, wherein said administering step comprises administering an effective amount of the compound in a pharmaceutically acceptable carrier.
Description:
CURCUMIN ANALOGUES FOR TREATING CANCER PROPERTIES CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of U. S. Provisional No. 60/168,913, filed December 3,1999, which is incorporated by reference herein in its entirety.

FIELD OF THE INVENTION The invention relates to compounds useful for the treatment of cancer, and in particular to compounds exhibiting anti-tumor and anti-angiogenic properties and methods for using such compounds.

BACKGROUND OF THE INVENTION Tissue factor (TF) is a sedimentable, integral membrane receptor protein with an estimated molecular weight of 42-47 kDa. Peritumor fibrin deposition, which is characteristic of most types of human cancer, is the result of the local expression of potent procoagulants like tissue factor (TF) in tumor cells, tumor-associated macrophages (TAMs) and tumor-associated vascular endothelial cells (VECs). In addition to the importance of TF expression in the pathogenesis of the thrombotic complications common to cancer patients, increasing evidence links TF expression to the regulation of tumor angiogenesis, growth and metastasis. For example, angiogenesis in vivo is inhibited by TF antisense. Further, murine tumor cells transfected to overexpress TF enhance vascular permeability factor (VEGF) transcription and translation. Conversely, tumor cells transfected with TF antisense reduce VEGF transcription and translation.

VEGF acts specifically on VECs to promote vascular permeability, endothelial cell growth and angiogenesis, and has been shown to induce expression of TF activity in VECs and monocytes and is chemotactic for monocytes, osteoblasts and VECs.

Expression of TF and VEGF in cancer cells is further enhanced under hypoxic condition. Thus, there is evidence to suggest that TF is a key molecule participating in the regulation of VEGF synthesis and, hence, tumor angiogenesis in cancer.

Relatively few compounds exhibiting anti-angiogenic properties useful in the treatment of cancer have been investigated. Curcumin (diferuloylmethane), the aromatic yellow pigment in curry, turmeric and mustard, is known to have anti-angiogenic, anti- tumor, and anti-tumor promoting properties. In addition, curcumin exhibits numerous other therapeutic effects, including anti-oxidative, anti-thrombotic, anti-inflammatory, anti-cholesterol and anti-diabetic properties. Two other compounds that have received considerable attention are genistein, a soybean-derived isoflavone tyrosine kinase C inhibitor, and linomide, a quinoline-3-carboxaminde. Certain flavonoids, such as apigenin, have been shown to be more potent inhibitors of cell proliferation and in vitro angiogenesis than genistein. There remains a need in the art for compounds that exhibit anti-tumor and anti-angiogenic properties for use in cancer therapy.

SUMMARY OF THE INVENTION The present invention provides a group of curcumin analogs that inhibit TF expression and VEGF expression in cancer cells and in vascular endothelial cells in the tumor microenvironment, thereby blocking tumor angiogenesis and growth, without exhibiting a high level of toxicity with regard to normal vascular endothelial cells. The anti-angiogenic and anti-tumor compounds of the present invention can be useful in the treatment of any condition benefiting from angiogenesis inhibition, such as cancer.

In one aspect, the present invention provides compounds of Formula (I) below.

In another aspect, the present invention provides a pharmaceutical formulation comprising a compound of Formula (I) or Formula (II) below in a pharmaceutically acceptable carrier.

In a third aspect, the present invention provides a method of treating cancerous tissue in a subject, comprising administering an effective amount of a compound of Formula (I) or Formula (II) to the subject. Preferably, the compound is administered in a pharmaceutically acceptable carrier. The effective amount is preferably an amount sufficient to inhibit VEGF or TF production in the cancerous tissue.

BRIEF DESCRIPTION OF THE DRAWINGS Having thus described the invention in general terms, reference will now be made to the accompanying drawings, wherein: Figures 1A and 1B graphically illustrate the relationship between cell viability and VEGF production of human melanoma cells after treatment with the compounds of the present invention at two concentrations; Figures 2A and 2B graphically illustrate the effect of known compounds and the compounds of the present invention on human melanoma cell viability; Figures 3A and 3B graphically illustrate the effect of known compounds and the compounds of the present invention on human breast cancer cell viability; Figures 4A and 4B graphically illustrate the effect of known compounds and the compounds of the present invention on transformed murine endothelial cell viability; and Figures 5A and 5B graphically illustrate the effect of known compounds and the compounds of the present invention on human endothelial cell viability.

DETAILED DESCRIPTION OF THE INVENTION The present invention will be described more fully hereinafter, including preferred embodiments thereof. This invention may, however, be embodied in many 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.

As used herein, the term"compound"is intended to refer to a chemical entity, whether in the solid, liquid or gaseous phase, and whether in a crude mixture or purified and isolated. The terms"alkyl,""alkene,"and"alkoxy"include straight chain and branched alkyl, alkene, and alkoxy, respectively. The term"lower alkyl"refers to C1-C4 alkyl. The term"alkoxy"refers to oxygen substituted alkyl, for example, of the formulas -OR or-ROR', wherein R and Rl are each independently selected alkyl. The terms "substituted alkyl"and"substituted alkene"refer to alkyl and alkene, respectively, substituted with one or more non-interfering substituents, such as but not limited to, C3- C6 cycloalkyl, e. g., cyclopropyl, cyclobutyl, and the like ; acetylene; cyano; alkoxy, e. g.,

methoxy, ethoxy, and the like; lower alkanoyloxy, e. g., acetoxy; hydroxy; carboxyl; amino; lower alkylamino, e. g., methylamino; ketone; halo, e. g. chloro or bromo; phenyl; substituted phenyl, and the like. The term"halogen"includes fluorine, chlorine, iodine and bromine.

"Aryl"means one or more aromatic rings, each of 5 or 6 carbon atoms. Multiple aryl rings may be fused, as in naphthyl or unfused, as in biphenyl. Aryl rings may also be fused or unfused with one or more cyclic hydrocarbon, heteroaryl, or heterocyclic rings.

"Substituted aryl"is aryl having one or more non-interfering groups as substituents.

"Heteroaryl"is an aryl group containing from one to four N, O, or S atoms (s) or a combination thereof, which heteroaryl group is optionally substituted at carbon or nitrogen atom (s) with C1-6 alkyl,-CF3, phenyl, benzyl, or thienyl, or a carbon atom in the heteroaryl group together with an oxygen atom form a carbonyl group, or which heteroaryl group is optionally fused with a phenyl ring. Heteroaryl rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings.

Heteroaryl includes, but is not limited to, 5-membered heteroaryls having one hetero atom (e. g., thiophenes, pyrroles, furans); 5 membered heteroaryls having two heteroatoms in 1,2 or 1,3 positions (e. g., oxazoles, pyrazoles, imidazoles, thiazoles, purines); 5-membered heteroaryls having three heteroatoms (e. g., triazoles, thiadiazoles); 5-membered heteroaryls having 3 heteroatoms ; 6-membered heteroaryls with one heteroatom (e. g., pyridine, quinoline, isoquinoline, phenanthrine, 5,6- cycloheptenopyridine); 6-membered heteroaryls with two heteroatoms (e. g., pyridazines, cinnolines, phthalazines, pyrazines, pyrimidines, quinazolines); 6-membered heretoaryls with three heteroatoms (e. g., 1,3,5-triazine); and 6-membered heteroaryls with four heteroatoms.

"Substituted heteroaryl"is heteroaryl having one or more non-interfering groups as substituents.

"Heterocycle"or"heterocyclic"means one or more rings of 5,6 or 7 atoms with or without unsaturation or aromatic character and at least one ring atom which is not carbon. Preferred heteroatoms include sulfur, oxygen, and nitrogen. Multiple rings may be fused, as in quinoline or benzofuran.

"Substituted heterocycle"is heterocycle having one or more side chains formed from non-interfering substituents.

"Non-interfering substituents"are those groups that yield stable compounds.

Suitable non-interfering substituents or radicals include, but are not limited to, halo, C1-C10 alkyl, C2-C10 alkenyl, C2-Clo alkynyl, Cl-Clo alkoxy, C7-C12 aralkyl, C-C12 alkaryl, C3-CIO cycloalkyl, C3-C10 cycloalkenyl, phenyl, substituted phenyl, toluoyl, xylenyl, biphenyl, C2-CI2 alkoxyalkyl, C7-CI2 alkoxyaryl, C7-C12 aryloxyalkyl, C6-CI2 oxyaryl, Cl-C6 alkylsulfinyl, Cl-Clo alkylsulfonyl,-(CH2) m-0-(Cl-CIo alkyl) wherein m is from 1 to 8, aryl, substituted aryl, substituted alkoxy, fluoroalkyl, heterocyclic radical, substituted heterocyclic radical, nitroalkyl, -NO2, -CN, -NRC(O)-(C1-C10 alkyl), -C(O)- (C1-Clo alkyl), C2-Clo thioalkyl,-C (O) O- (CI-Clo alkyl),-OH,-S02, =S,-COOH,-NR2, carbonyl,-C(O)-(C1-C10 alkyl)-CF3, -C(O)-CF3, -C(O)NR2, -(C1-C10alkyl)-S-(C6-C12 aryl),-C (O)-(C6-C12 aryl),-(CH2)m-O-(CH2)m-O-(C1-C10 alkyl) wherein each m is from 1 to 8,-C (O) NR2,-C (S) NR2,-SO2NR2,-NRC (O) NR2,-NRC (S) NR2, salts thereof, and the like. Each R as used herein is H, alkyl or substituted alkyl, aryl or substituted aryl, aralkyl, or alkaryl.

The present invention provides compounds of Formula (I) wherein: Y is OH, halogen, or CF3 ; Z is H, OH, ORI, halogen, or CF3; Xi and X2 are independently C or N; and A is selected from the group consisting of : wherein n is 1-8; X3 is O, S, SO, SO2, NH, or NRi ; Q is NH or NR1 ; and Vlq are each independently OH, OR2, or halogen; Ri and Ra are independently H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl or dialkylaminocarbonyl; the dashed lines indicate the

presence of optional double bonds; and L is the point of bonding of A to the compound structure, with the proviso that Z is not H when Y is OH, Cl or Br and A is and pharmaceutically acceptable salts thereof.

The present invention also provides a method of treating cancerous tissue in a subject, such as a human or other mammal, comprising administering to the subject an effective amount of a compound of Formula (I) above or a compound of Formula (II) wherein: X4 is (CH2) m, O, S, SO, S02, or NRI2, where Rl2 is H, alkyl, substituted alkyl, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl or <BR> <BR> <BR> dialkylaminocarbonyl ;<BR> <BR> <BR> mis 1-7 ; each X5 is independently N or C-R, 1 ; and each R3-Rll are independently H, halogen, hydroxyl, alkoxy, CF3, alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, alkaryl, arylalkyl, heteroaryl, substituted heteroaryl, heterocycle,

substituted heterocycle, amino, alkylamino, dialkylamino, carboxylic acid, carboxylic ester, carboxamide, nitro, cyano, azide, alkylcarbonyl, acyl, or trialkylammonium ; and the dashed lines indicate optional double bonds; with the proviso that when X4 is (CH2) m, m is 2-6, and each X5 is C-RIl, R3-R are not alkoxy, and when X4 is NRz2 and each X5 is N, R3-Rlo are not alkoxy, alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, alkaryl, arylalkyl, heteroaryl, substituted heteroaryl, amino, alkylamino, dialkylamino, carboxylic acid, or alkylcarbonyl.

The present invention includes all stereoisomeric configurations of the compounds of Formula (I) and Formula (II), including both optical isomers, such as enantiomers and diastereoisomers, and geometric (cis-trans) isomers.

Examples of the compounds of the present invention include, but are not limited to: (EF2) R= 2-OH (E1F4) X= C, R= 2-OH (EF3) R 3-OH (EF31) X = C-1, R = 2-OH (EF1) R 4-OH (EF25) X= O, R= 2-OH (EF8) R=2-F (EF29) X = O, R = 2-F (EF9) R = 2, 4-F (EF30) X = O, R = 2, 4-F (EF10) R= 3, 4-F (EF36) X = O, R = 3, 4 (OMe) (EF23) R = 2, 6-F (EF28) X = O, R = 2-OMe (MD6) R = 3,4-(OMe) (EF27) X = O, R = 4-OMe (EF16) R = 2-OMe (EF34) X = NMe, R = 2-OH (EF17) R = 3-OMe (EF33) X = NMe, R = 2-F (EF18) R = 4-OMe (EF47) X = NMe, R = 2, 4-F (EF35) X = NMe, R = 3,4- (OMe) (EF24) X = NH2OAc, R = 2-F (EF26) X = NH2Cl, R = H

(°) n R S/ Oh (EF15) n=O, R=H (EF13) n = 1, R = H, (E, E) (EF14) n = 1, R = H, (E, @ (EF32) R = 2-OH (EFll) n=2, R=2-OH (EF48) R = 2-F (EF12) n=2, R=H (EF19) R=2, 4-F2 (EF20) R= 3, 4-F2 Rilo (MD279L) R = 3,4- (OMe) R= \ \ R 0 O (EF37) Rl = Me, R = 3, 4-F (MD38) Rl = Me, R = 3,4- (OMe) (EF44) Rl = Pr, R = 3, 4- (OMe) (EF21) R = 2, 4-F (EF22) R = 3, 4-F (MD279U) R = 3,4- (OMe) oo Rw R MeO MeO. OH N OH (EF46) Rz = CH2Br, R=3, 4-(OMe) v DA (EF40) Rl = Et, R= 3, 4-F 9 (EF6) 9 (EF41) Rl = Et, R= 3, 4-(OMe) (EF49) Rl = Ph, R = 2-F (EF39) R, = Ph, R = 3, 4- (OMe) (EF45) Rl = morph. R = 3, 4- (OMe) (EF43) Rl = menth., R = 3, 4-(OMe) v (EF5) 9 x Y OH "' (EF7) //I,. (EF7) (EF42) R= 3, 4-(OMe) X = Cl Y=H (EF50) R=3, 4- (OMe) X=OH Y=Me

and the like and pharmaceutically acceptable salts thereof. Additional exemplary compounds are given in the appended examples.

The compounds of the present invention may be prepared according to methods known in the art, particularly in light of the disclosure and examples set forth herein. The starting materials used to synthesize the compounds of the present invention are commercially available or capable of preparation using methods known in the art. For example, some compounds of the present invention may be prepared by reaction of an aromatic aldehyde, such as hydroxybenzaldehyde or fluoro-substituted benzaldehyde, with a ketone, such as acetone, cyclohexanone, cyclopentanone, tetrahydro-4-H-pyran-4- one, N-methyl-4-piperidone, piperidin-4-one, and the like, under basic aldol condensation conditions. Similarly, other compounds of the present invention may be prepared by reaction of an alkoxy-substituted benzaldehyde or anisaldehyde with a ketone. As would be understood, the actual ketone or aldehyde utilized will depend on the type and position of the substituents of the desired final compound. The salts of the present invention may be prepared, in general, by reaction of a compound of the invention with the desired acid or base in solution. After the reaction is complete, the salts can be crystallized from solution by the addition of an appropriate amount of solvent in which the salt is insoluble.

The compounds of Formula (I) or Formula (II) can have pharmaceutical activity and can be useful in the treatment of a subject suffering from one or more conditions that would benefit from inhibition of angiogenesis. For example, the compounds of the present invention can be used in the treatment of cancerous tissue and the tumors associated therewith, including breast, colon, prostate and skin cancer. In addition, the compounds of the present invention can be useful for mediating inflammation,

rheumatoid arthritis and certain forms of diabetes. Subjects which can be treated include animal subjects, typically vertebrates, including both mammalian (e. g., human, cat, dog, cow, horse, sheep, pig, monkey, ape, etc.) and avian subjects (e. g., chicken, turkey, duck, goose, quail, pheasant, etc.). It is believed, for example, that administering an effective amount of a compound of Formula (I) or Formula (II) to a subject can result in inhibition of angiogenesis in cancerous tissue. Thus, the present invention can provide methods for treating tumor-bearing subjects in which the compounds of the invention are administered to the subject in need of such treatment in an amount effective and in a manner effective to combat such tumors, for example, by virtue of inhibition of angiogenesis within the tumor. The anti-angiogenesis effect is believed to result, at least in part, from inhibition of TF and/or VEGF production in the tumor. In addition, it is believed that the compounds of the present invention can be used as a prophylactic treatment to prevent certain types of inflammatory skin conditions including, but not limited to, dermatitis and mild cases of skin cancer.

The compounds of Formula (I) or Formula (II) may be administered per se or in the form of a pharmaceutically acceptable salt. When used in medicine, the salts of the compounds of Formula (I) or Formula (II) should be both pharmacologically and pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare the free active compound or pharmaceutically acceptable salts thereof and are not excluded from the scope of this invention. Such pharmacologically and pharmaceutically acceptable salts can be prepared by reaction of a compound of Formula (I) or Formula (II) with an organic or inorganic acid, using standard methods detailed in the literature. Examples of useful salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicyclic, p-toluenesulfonic, tartaric, citric, methanesulphonic, formic, malonic, succinic, naphthalene-2-sulphonic and benzenesulphonic, and the like. Also, pharmaceutically acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium, or calcium salts of the carboxylic acid group.

Thus the present invention also provides pharmaceutical formulations or compositions, both for veterinary and for human medical use, which comprise the compounds of Formula (I) or Formula (II) or a pharmaceutically acceptable salt thereof

with one or more pharmaceutically acceptable carriers thereof and optionally any other therapeutic ingredients, such as other chemotherapeutic agents. The carrier (s) must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the formulation and not unduly deleterious to the recipient thereof.

The compositions includes those suitable for oral, rectal, topical, nasal, ophthalmic, or parenteral (including intraperitoneal, intravenous, subcutaneous, or intramuscular injection) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier that constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier or both, and then, if necessary, shaping the product into desired formulations.

Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets, tablets, lozenges, and the like, each containing a predetermined amount of the active agent as a powder or granules; or a suspension in an aqueous liquor or non-aqueous liquid such as a syrup, an elixir, an emulsion, a draught, and the like.

A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, with the active compound being in a free-flowing form such as a powder or granules which is optionally mixed with a binder, disintegrant, lubricant, inert diluent, surface active agent or dispersing agent. Molded tablets comprised with a suitable carrier may be made by molding in a suitable machine.

A syrup may be made by adding the active compound to a concentrated aqueous solution of a sugar, for example sucrose, to which may also be added any accessory ingredient (s). Such accessory ingredients may include flavorings, suitable preservatives, an agent to retard crystallization of the sugar, and an agent to increase the solubility of any other ingredient, such as polyhydric alcohol, for example, glycerol or sorbitol.

Formulations suitable for parental administration conveniently comprise a sterile aqueous preparation of the active compound, which can be isotonic with the blood of the

recipient.

Nasal spray formulations comprise purified aqueous solutions of the active agent with preservative agents and isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucous membranes.

Formulations for rectal administration may be presented as a suppository with a suitable carrier such as cocoa butter, or hydrogenated fats or hydrogenated fatty carboxylic acids.

Ophthalmic formulations are prepared by a similar method to the nasal spray, except that the pH and isotonic factors are preferably adjusted to match that of the eye.

Topical formulations comprise the active compound dissolved or suspended in one or more media such as mineral oil, petroleum, polyhydroxy alcohols or other bases used for topical formulations. The addition of other accessory ingredients as noted above may be desirable.

Further, the present invention provides liposomal formulations of the compounds of Formula (I) or Formula (II) and salts thereof. The technology for forming liposomal suspensions is well known in the art. When the compound of Formula (I) or Formula (II) or salt thereof is an aqueous-soluble salt, using conventional liposome technology, the same may be incorporated into lipid vesicles. In such an instance, due to the water solubility of the compound or salt, the compound or salt will be substantially entrained within the hydrophilic center or core of the liposomes. The lipid layer employed may be of any conventional composition and may either contain cholesterol or may be cholesterol-free. When the compound or salt of interest is water-insoluble, again employing conventional liposome formation technology, the salt may be substantially entrained within the hydrophobic lipid bilayer that forms the structure of the liposome.

In either instance, the liposomes that are produced may be reduced in size, as through the use of standard sonication and homogenization techniques. The liposomal formulations containing the compounds of Formula (I) or Formula (II) or salts thereof, may be lyophilized to produce a lyophilizate which may be reconstituted with a pharmaceutically acceptable carrier, such as water, to regenerate a liposomal suspension.

Pharmaceutical formulations are also provided which are suitable for administration as an aerosol, by inhalation. These formulations comprise a solution or

suspension of the desired compound of Formula (I) or Formula (II) or a salt thereof or a plurality of solid particles of the compound or salt. The desired formulation may be placed in a small chamber and nebulized. Nebulization may be accomplished by compressed air or by ultrasonic energy to form a plurality of liquid droplets or solid particles comprising the compounds or salts.

In addition to the aforementioned ingredients, the compositions of the invention may further include one or more accessory ingredient (s) selected from the group consisting of diluents, buffers, flavoring agents, binders, disintegrants, surface active agents, thickeners, lubricants, preservatives (including antioxidants) and the like.

Preferably, for purposes of cancer therapy, a compound of Formula (I) or Formula (II) is administered to the subject in an amount sufficient to inhibit production of TF or VEGF, thereby inhibiting angiogenesis. However, the therapeutically effective dosage of any specific compound will vary somewhat from compound to compound, patient to patient, and will depend upon the condition of the patient and the route of delivery. As a general proposition, a dosage from about 0.5 to about 20 mg/kg body weight, preferably from about 1.0 to about 5.0 mg/kg, will have therapeutic efficacy. When administered conjointly with other pharmaceutically active agents, even less of the compounds of Formula (I) or Formula (II) may be therapeutically effective. The compound of Formula (I) or Formula (II) may be administered once or several times a day. The duration of the treatment may be once per day for a period of from two to three weeks and may continue for a period of months or even years. The daily dose can be administered either by a single dose in the form of an individual dosage unit or several smaller dosage units or by multiple administration of subdivided dosages at certain intervals.

For the following examples, RPMI-7951 human melanoma, MDA-MB-231 and MDA-MB-435 human breast cancer cell lines were purchased from American Type Cell Collection (Rockville, MD). HUVECs were obtained from the Department of Dermatology, Emory University. Murine endothelial cells infected with simian virus 40 (SV40) large T antigen and activated H-ras (SVR), were a kind gift from Dr. Jack Arbiser at Emory. RPMI-7951, MDA-MB-231 and MDA-MB-435 cell lines were cultured in MEM-alpha medium (GIBCO-BRL, Long Island, NY) containing 10% fetal bovine serum (RPMI-7951, MDA-MB-231) or 5% FBS (MDA-MB-435) at 37° C and under 5%

C02/95% air. SVR cells were cultured in DMEM (Mediatech cellgro) containing 10% FBS and 2mM L-glutamine. Complete HUVEC media was a gift from the Cell Culture Center in the Department of Dermatology, Emory University. The cells were cultured in 48 well plates in all of the experiments described.

Neutral Red Assay was utilized to determine the effect of the compounds of the present invention on cell viability. Neutral Red was purchased from GIBCO-BRL (Long Island, NY). Cells were plated at a concentration of 20,000 cells/well and cultured overnight. Compounds or vehicle (DMSO. 1%) were then added and the plates were incubated for 72 hours. Supernatant from each well was either aspirated or collected and media containing Neutral Red (GIBCO-BRL, Long Island, NY) at a concentration of 15 gl/ml was then added to each well. The plates were then incubated at 37° C for 30 minutes. Next, the cells were washed with twice with PBS and alcoholic-HCl (0. 5N- HC1/35% ethanol) was added to each well. The plates were then placed on a plate shaker until all residues were solubilized (pink color). The solubilized mixtures were then transferred to a 96 well plate and the absorbances were read on a micro-test plate reader at a wavelength of 570nM.

A VEGF enzyme-linked immunosorbent assay (ELISA) was utilized to determine the effect of the compounds of the present invention on VEGF production of a variety of human cancer cell lines. For the VEGF assay, cells were plated at a concentration of 80,000 cells/well and cultured overnight. Compounds or vehicle (DMSO. 1%) were then added and the plates were incubated for 72 hours. Supernatant was then collected from each well and frozen in a-80°C freezer until needed. Cell viability was determined by Neutral Red Assay. VEGF ELISA Kit (R & D, Minneapolis, MN) was used to determine the amount of VEGF in the culture supernatants. The ELISA was carried out according to the manufacturer's procedure.

A TF ELISA assay was utilized to determine the effect of the compounds of the present invention on TF production of human cancer cell lines. For the TF assay, cells were plated at a concentration of 80,000 cells/well and cultured overnight. Compounds or vehicle (DMSO. 1%) were then added and the plates were incubated for 72 hours.

Cells were treated with 1% Triton X-100 in PBS and left overnight at 4° C overnight to solubilize TF. Supernatant was then collected from each well and frozen until needed.

IMUBIND Tissue Factor ELISA Kit (American Diagnostica Inc, Greenwich, CT) was used to determine TF concentration in each sample. The ELISA was carried out according to manufacturer's procedure.

EXPERIMENTAL Example 1 Preparation of EFI, EF2, EF3, EF4, EF25, EF31, EF34 compounds The compounds of this series were all synthesized by the following procedure: Aqueous NaOH (20 wt%, 15ml, 75mmol) was added dropwise to a vigorously stirred solution of hydroxybenzaldehyde (51 mmol) and ketone (25 mmol) in EtOH abs (20 mL).

The reaction was stirred at room temperature for 48 hrs, Ha0 dist (100 mL) was added, and the purple solution was neutralized by gently bubbling C02 through it. The precipitating yellow solid was filtered off, washed with H20 dist and dried under vacuum. The products were purified by recrystallization. A description of each compound obtained by the above process is given below.

1, 5-Bis (4-hydroxyphenyl) penta-1, 4-dien-3-one (EF1) : yellow solid (6%), mp 236°C (acetone/H20). 1H NMR (400 MHz, CD30D) 6 7.71 (2H, d, J = 16 Hz), 7.58 (4H, d, J = 8.8 Hz), 7.07 (2H, d, J = 16 Hz), 6.84 (4H, d, J = 8.4 Hz). 13 C NMR (100 MHz, CD30D) S 191. 9,161.8,145. 3,131.8,127.8,123.6,117.1. EIHRMS: m/z 266. 0943 (M+, C17H1403 requires 266.0943).

1, 5-Bis (2-hydroxyphenyl) penta-1, 4-dien-3-one (EF2): yellow solid (75%), mp 155°C (acetone/H20). IH NMR (400 MHz, CD30D) 8 8.09 (2 H, d, J = 16 Hz,), 7.63 (2H, dd, J = 8. 4 Hz, J = 1. 6 Hz), 7.31 (2H, d, J = 16 Hz), 7.24 (2H, td, J = 7. 6 Hz, J = 1. 6 Hz), 6.88 (t, 4H, J = 7. 2 Hz). 13C NMR (100 MHz, CD30D) 8 192. 8,158.9,141.0,133.1,130.2, 126.3,123.2,121.0,117.2. Anal. Calcd for C1H14O3 : C, 78.68; H, 5.30. Found: C, 76.56 ; H, 5.32. EIHRMS: m/z 248.0837 ((M-H20) +, Cl7Hl202 requires 248.0837).

1, 5-Bis (3-hydroxyphenyl) penta-1, 4-dien-3-one (EF3): yellow solid (15%), mp 198- 200°C (acetone/H20). IH NMR (400 MHz, CD30D) 8 7.70 (2H, d, J = 16 Hz), 7.24 (2H, t, J = 7. 6 Hz), 7.17 (2H, d, J = 16 Hz), 7.17 (2H, d, J = 8 Hz), 7.11 (2H, s), 6.73 (2H, dd, J = 8 Hz, J = 2.4 Hz). 13C NMR (100 MHz, CD30D) 8 191.6,159.3,145.5,137.6,

131.2,126.4,121.4,119.1,115.9. Anal. CalcdforCiyOs : C, 78.68; H, 5.30. Found: C, 76.41: H, 5.48. EIHRMS: m/z 266.0943 (M+, C17H1403 requires 266.0943).

2,6-Bis (2-hydroxybenzylidene) cyclohexanone (EF4): yellow solid (70%, recrystallized from acetone/H20).'H NMR (400 MHz, CD30D) 8 7.98 (2H, s,), 7.32 (2H, dd, J = 7.6 Hz, J = 1. 2 Hz), 7.19 (2H, td, J = 7. 6Hz, J = 1. 2Hz), 6.86 (4H, m), 2.86 (4H, m), 1.75 (2H, m). 13C (100 MHz, CD30D) 8 192.8,158.2,137.3,134.5,131.5,124.5,120.2, 116.6,29.9,24.8. Anal. Calcd for C20HI803 : C, 78.41; H, 5.92. Found: C, 78.15; H, 6.03. EIHRMS: m/z 306.1263 (M+, C20Hz803 requires 306.1256).

3,5-Bis (2-hyrdoxybenzylidene) tetrahydro-4-H-pyran-4-one (EF25): yellow solid (60%, recrystallized from acetone/H20). IH NMR (400 MHz, CD30D) 8 8.08 (2H, s), 7.24 (2H, td, J = 8. 4 Hz, J = 1.6 Hz), 7.09 (2H, dd, J = 7. 6 Hz, J = 1.6 Hz), 7.90-7.86 (4H, m), 4.84 (4H, d, J = 1. 6 Hz). 13C NMR (100 MHz, CD30D) 8 187.8,158.5,133.9,132.5, 131.9,123.3,120.5,116.8,70.0. Anal. Calcd for C19H16O4 : C, 74.01; H, 5.23. Found: C, 73.23; H, 5.23. EIHRMS: m/z 290.0933 (M+, Cl9Hz604 requires 290.0943).

2,5-Bis (2-hydroxyphenyl) cyclopentanone (EF31): yellow solid (81%, recrystallized from acetone). 1H NMR (400 MHz, CD30D) 8 8.00 (2H, s), 7.57 (2H, dd, J = 8.0 Hz, J = 1.2 Hz), 7.22 (2H, td, J = 8.0 Hz, J = 1.6 Hz), 6.88 (4H, m), 3.06 (4H, s).

3,5-Bis (2-hydroxybenzylidene) l-methyl-4-piperidone (EF34): yellow solid (75%, recrystallized from methanol/H20). tH NMR (400 MHz, CD30D) 8 8.11 (2H, s), 7.23 (4H, t, J = 7.6 Hz), 6.88 (4H, t, J = 8.0 Hz), 3.76 (4H, d, J = 1. 2 Hz), 2.42 (3H, s). 13C NMR (100 MHz, CD30D) 8 188.40,158.58,135.09,133.09,132.27,131.64,123.59, 120.38,116.87. HREIMS: m/z 303.1259 (M+-H20, C20Hz7No2 requires 303.1259).

Example 2 Preparation of EF8, EF9, EF10, EF23, EF29, EF30, EF33 The compounds of this series were all synthesized by the following procedure: A solution of fluoro-substituted benzaldehyde (5.00 mmol) in ethanol abs. (1 mL) was added at room temperature over a period of 5 min, with stirring, to a solution of NaOH (0.75 mmol) and ketone (acetone, tetrahydro-4-H-pyran-4-one, N-methyl-piperodin-4- one) (2.50 mmol) in a mixture of ethanol abs (7 mL) and H20 dist. (7 mL). The solution turns yellow immediately, and usually a yellow precipitate starts forming within 10 min

(except EF8, where an oil forms). The reaction was stirred at room temperature for 3 hrs., the yellow solid filtered off, washed with water and hexanes and dried under vacuum. The product was obtained in analytically pure form, further purification was only necessary where indicated. A description of each compound obtained by the above process is given below.

1, 5-Bis (2-fluorophenyl) penta-1, 4-dien-3-one (EF8) : yellow solid (50%). 1H NMR (400 MHz, CDC13,) 6 7.86 (2H, d, J = 16 Hz), 7.63 (2H, td, J = 7.6 Hz, J = 1. 6 Hz), 7.42- 7.35 (2H, m), 7. 18 (2H, d, J = 16 Hz), 7.26-7.10 (4H, m). 13C NMR (100 MHz, CDC13) 8 189.2,161.9 (d, J = 253.4 Hz), 136.3 (d, J = 2.3 Hz), 132.1 (d, J = 9.1 Hz), 129.5 (d, J = 2.2 Hz), 127.8 (d, J = 6.1 Hz), 124.7 (d, J = 3.8 Hz), 123.1 (d, J = 11.4 Hz), 116.4 (d, J = 22.0 Hz). Anal. Calcd for Cl7HI2F2O : C, 75.55; H, 4.48; F, 14. 06. Found: C 75.30,; H 4.55. EIHRMS: m/z 270.0865 (M+, C17H12F2O requires 270.0856).

1, 5-Bis (2,4-difluorophenyl) penta-1, 4-dien-3-one (EF9): yellow solid (72%). 1H NMR (300 MHz, CDCl3) # 7.79 (2H, d, J = 16. 2 Hz), 7.62 (2H, dd, J = 15 Hz, J = 8.4 Hz,), 7.10 (2H, d, J = 16 Hz), 8.85-8.97 (4H, m). 13C NMR (75 MHz, CDC13) 8 188.5, 164.8 (dd, J= = 169. 6 Hz, J = 12. 2 Hz), 161. 3 (dd, J= 160.3 Hz, J= 12.2 Hz), 135.3,130.7 (q, J = 10.0 Hz, J = 4. 6 Hz), 127.3,119.5, 112.3 (d, J = 21.9 Hz), 104.9 (t, J = 25. 4 Hz).

Anal. Calcd for Cl7HIoF4O : C, 66.67; H, 3.29. Found: C, 66.38; H, 3.41. EIHRMS: m/z 306.0654 (M+, Cl7HIOOF4 requires 306.0668).

1, 5-Bis (3,4-difluorophenyl) penta-1, 4-dien-3-one (EF10): yellow solid (86%). 1H NMR (400 MHz, CDC13) 6 7.64 (2H, d, J = 16 Hz), 7.44 (2H, ddd, J = 9.6 Hz, 7.6 Hz, J = 2 Hz), 7.32-7.36 (2H, m), 7.18-7.25 (2H, m), 6.96 (2H, d, J = 16 Hz). 13C NMR (100 MHz, CDCl3) # 180.0,152.7 (dd, J = 105. 4 Hz, J = 12.9 Hz), 150.2 (dd, J= 101. 7 Hz, J= 12. 9 Hz), 141.5,132.1 (t, J = 5.2 Hz), 126.2,125.5 (q, J = 6 Hz, J = 3 Hz), 118.2 (d, J = 17. 5 Hz), 116.7 (d, J = 20.5 Hz). Anal. Calcd for Cl7HloF4O : C, 66.67; H, 3.29.

Found: C, 66.54; H, 3.28. EIHRMS: m/z 306.0671 (M+, Cl7HloOF4 requires 306.0668).

1, 5-Bis (2,6-difluorophenyl) penta-1, 4-dien-3-one (EF23): yellow solid (91%). IH NMR (400 MHz, CDC13) 8 7.82 (2H, d, J = 16. 4 Hz), 7.35 (2H, d, J = 16 Hz), 7.33 (2H, td, J = 8 Hz, J = 2.4 Hz), 6.96 (4H, t, J = 8.8 Hz). 13C NMR (100 MHz, CDC13) 8 189.5, 162.2 (dd, J = 1017. 6 Hz, J = 29.2 Hz), 131.6 (t, J = 10. 9 Hz), 131. 0 (t, J = 8. 7 Hz), 129.9,113.0 (t), 112. 1 (d, J = 25.6 Hz). Anal. Calcd for C17H10FO4 : C, 66.67; H, 3.29.

Found: C, 66.46; H, 3.26, EIHRMS: m/z 306.0657 (M+, CI7H1oF04 requires 306.0668).

3,5-Bis (2-fluorobenzylidene) tetrahydro-4-H-pyran-4-one (EF29): (84%, recrystallized from hot ethanol). IH NMR (400 MHz, CDD13) 8 7.92 (2H, d, J = 1.2 Hz), 7.40-7.36 (2H, m), 7.20-7.18 (4H, m), 7.13 (2H, t, J = 9.6 Hz), 4.80 (s, 4H, CH2). 13c NMR (100 MHz, CDC13) 8 185.1,161.0 (d, J = 250.7 Hz), 134.8,131.5 (d, J = 8.7 Hz), 131. 1 (d, J = 2.2 Hz), 129.6 (d, J = 3.7 Hz), 124. 3 (d, J = 3.7 Hz), 122.8 (d, J = 13.8 Hz), 116.2 (d, J-.

21.9 Hz), 68.9 (d, J = 5.1 Hz). Anal. Calcd for ClgHl4F202 : C, 73.07; H, 4.52. Found: C, 73.07; H, 4.47. E1HRMS ; m/z 312.0950 (M+, Cl9HI4F202 requires 312.0962).

3,5-Bis (2,4-difluorobenzylidene) tetrahydro-4-H-pyran-4-one (EF30): (82%, recryst. from ethanol/H2O). IH NMR (400 MHz, CDC13) 8 7.85 (2H, s), 7.21-7.14 (1H, m), 6.98-6.87 (2H, m), 4.77 (4H, s). 13C NMR (100 MHz, CDC13) 6 184.81,163.95 (dd, J = 240 Hz, J = 12 Hz), 161.43 (dd, J = 265 Hz, J = 12 Hz), 134.48,132.02 (dd, J = 9.5 Hz, J = 4. 3 Hz), 128.62,119.19,112.00 (dd, J = 3. 7 Hz, J = 21. 9. Hz), 104.85 (t, J = 25. 5 Hz), 68.81 (d, J = 4.4 Hz). Anal. Calcd for Cl9Hl2F402 ; C, 65.52; H, 3.47. Found: C, 65.67; H, 3.43. EIHRMS: m/z 348.0761 (M+, Cl9Hl4F202 requires 348.0773).

3,5-Bis (2-fluorobenzylidene) l-methyl-4-piperidone (EF33): (82 %, yellow solid). 1H NMR (400 MHz, CDC13) 8 7.90 (2H, s), 7.40-7.33 (2H, m), 7.28 (2H, td, J = 7.6 Hz, J = 1.2 Hz), 7.18 (2H, td, J = 7.6 Hz, J = 0.8 Hz), 7.12 (2H, td, J = 10.0 Hz, J = 0.8 Hz), 3.64 (4H, s), 2.40 (3H, s). 13C NMR (100 MHz, CDC13) 6 186.3,161.1 (d, J = 251 Hz), 134.9,131.1 (d, J = 8. 1 Hz), 131. 0 (d, J = 3. 0 Hz), 129.7 (d, J = 3. 6 Hz), 124.1 (d, J = 2. 9 Hz), 123.4 (d, J = 13.9 Hz), 116.1 (d, J = 21. 8 Hz), 57. 2,45.8. Anal. Calcd for C2oHl7F20N : C, 73.83; H, 5.27; N, 4.30. Found: C, 73.59; H, 5.32; N, 4.39. EIHRMS: m/z 325.1278 (M+, C2oHl7F20N requires 325.1278).

Example 3 Preparation of EFll, EF12, EF13, EF14, EF15 The compounds of this series were all synthesized by the following procedure: Bis-diethylphosphorylmethylsulfide,-sulfoxide and-sulfone were obtained according to literature procedures (Tetrahedron, 1992,48,8065-8072; Phosphorus Sulfur 1981,10, 369-374). A solution of phosphonate (0.60 mmol) and aldehyde (1.25 mmol) in CH2C12 (3 mL) was added to the heterogeneous mixture of 50 % aqueous NaOH (2 mL) and

CH2C12 (2 mL), containing triethylbenzylammonium chloride (TEBA, 0.06 mmol). The reaction was stirred at room temperature over night, the product was extracted from the reaction mixture with CH2C12 and purified by column chromatography. A description of each compound obtained by the above process is given below.

3,5-Bis (2-hydroxybenzylidene)-sulfone (EF11) : yellow solid (45%, 30% EtOAc/hexanes). lH NMR (400 MHz, CD30D) 8 7.77 (2H, d, J = 15. 6 Hz), 7.48 (2H, dd, J = 8. 4 Hz, J = 2. 0 Hz), 7.25 (2H, td, J = 8. 0 Hz, J = 2. 5 Hz), 7.22 (2H, d, J = 15.6 Hz), 6.88-6.84 (4H, m), 4.91 (2H, s-br). 13C NMR (100 MHz, CD30D) 8 158.94,140.21, 133.55,131.63,127.84,121.15,121.03,117.23.

3,5-Bisbenzylidenesulfone (EF12): white solid (78%, 20% EtOAc/hexanes). lH NMR (400 MHz, CDC13) 6 7.65 (2H, d, J = 15. 2 Hz, H3), 7.53-7.50 (4H, m), 7.44-7.40 (6H m), 6.86 (2H, d, J = 15.6 Hz, H2). 13C NMR (100 MHz, CDC13) 8 143.7,132.7,131.5, 129.3,128.8,126.5. Anal. Calcd for Cl6Hl4SO2 : C, 71.09; H, 5.22, S, 11. 86. Found: C, 70.88 ; H, 5.21, S, 12.01. EIHRMS: m/z 270.0715 (M+, C16Hl4SO2 requires 270.0731).

E, E-3,5-Bisbenzylidenesulfoxide (EF13): white solid (33%, 20% EtOAc, hexanes).'H NMR (300 MHz, CDC13) 8 7.52-7.46 (4H, m), 7.42-7.35 (6H, m), 7.31 (2H, d, J = 15. 3 Hz), 6.87 (2H, d, J = 15.3 Hz). 13C NMR (75 MHz, CDC13) 8 137.0,133.9,131.1,130.0, 129.1,127.9. Anal. Calcd for CI6HI4SO : C, 75.56; H, 5.55, S, 12.60. Found: C, 75.33; H, 5.60, S, 12.60.

E, Z-3,5-Bisbenzylidenesulfoxide (EF14): white solid (15%, 20% EtOAc, hexanes).'H NMR (300 MHz, CDC13) 6 7.52-7.40 (10H, m), 7.33 (1H, d, J = 15.3 Hz), 7.12 (1H, d, J = 10.5 Hz), 6.93 (1H, d, J = 15. 6 Hz), 6.39 (1H, d, J = 10.5 Hz). 13C NMR (75 MHz, CDC13) 8 138.5,136.3,135.5,134.1,130.1,129.8,129.6,129.0,128.8,127.8. Anal.

Calcd for Cl6HI4SO : C, 75.56 ; H, 5.55, S, 12.60. Found: C, 75.34; H, 5.54, S, 12.57.

3,5-Bisbenzylidenesulfide (EF15): white solid (20%, 5% EtOAc/hexanes) mixture of E, E and E, Z (ca. 2. 5 : 1).'H NMR (400 MHz, CDC13) 8 7.40-7.10 (20H, m), 6.79 (2H, d, J = 15.6 Hz), 6.76 (1H, d, J = 15.6 Hz), 6.62 (2H, d, J = 15.6 Hz), 6.61 (lH, d, J = 15. 6 Hz), 6.53 (1H, J = 10. 8 Hz), 6.41 (1H, d, J = 10.8 Hz). Anal. Calcd for C16Hl4S : C, 80.63 ; H, 5.92; S, 13.45. Found: C, 80.40 ; H, 5.93 ; S, 13. 39.

Example 4 Preparation of EF16, EF17, EF18, EF27, EF28 The compounds of this series were all synthesized by the following procedure: NaOH (0.10 mmol) was added as a solid to a stirred solution of methoxy-substituted benzaldehyde/anisaldehyde (2.50 mmol) and ketone (acetone, tetrahydro-4-H-pyranone) in EtOH abs (5 mL). A yellow solid started forming within 1 hr. The reaction was stirred at room temperature for 20 hrs, the product filtered off, washed with cold EtOH abs and H20 dist and dried under vacuum. A description of each compound obtained by the above process is given below.

1, 5-Bis (2-methoxphenyl) penta-1, 4-dien-3-one (EF16): yellow solid (60%). Mp 123- 124 °C (EtOH). tHNMR (400 MHz, CDCl3) 8 8.07 (2H, d, J = 16 Hz,), 7.63 (2H, dd, J =7. 6Hz, J= 1. 6Hz), 7.37 (2H, ddd, J = 7.2 Hz, J = 1.6 Hz, J = 1.2 Hz), 7.18 (2H, d, J = 16 Hz), 6.99 (2H, t, J = 7.6 Hz), 6.93 (2H, d, J = 8.4 Hz). i3C NMR (100 MHz, CDCl3) 8 190. 2,158.7,138.4,131.8,128.9,126.4,124.1,120.9,111.4,55.7. Anal. Calcdfor Cl9HI803 : C, 77.53; H, 6.16. Found: C, 77. 26; H, 6.17. EIHRMS: m/z 294.1256 (M+, C19HI803 requires 294.1256).

1, 5-Bis (3-methoxyphenyl) penta-1, 4-dien-3-one (EF17): yellow solid (40%, chromatography using 20 % EtOAc/hexanes).'H NMR (300 MHz, CDC13) 8 7.70 (2H, d, J = 15.9 Hz), 7.33 (2H, t, J = 7. 6 Hz), 7.21 (2H, d, J = 7. 8 Hz), 7.13 (2H, t, J = 2. 4 Hz), 7.06 (2H, d, J = 15.9 Hz), 6.96 (2H, ddd, J = 8.1 Hz, J = 2.4 Hz, J = 0. 9 Hz). 13c NMR (75 MHz, CDC13) 8 188.83,159.96,143.31,136.21,130.03,125.72,121.20,116.46, 113.36,55.55.

1, 5-Bis (4-methoxyphenyl) penta-1, 4-dien-3-one (EF18): yellow solid (93%). Mp 129- 130 °C (EtOH). IH NMR (300 MHz, CDC13) 8 7.69 (2H, d, J = 15.9 Hz, 3), 7.56 (4H, d, J = 9 Hz), 6.95 (2H, d, J = 15. 6 Hz), 6.92 (4H, d, J = 8.7 Hz). 13C NMR (75 MHz, CDC13) 8 188. 8,161.5,142.7,130.2,127.7,126.6,114.5,55.6 Anal. Calcd for ClgHIsO3 : C, 77.53; H, 6.16. Found: C, 77.31; H, 6.217. EIHRMS: m/z 294.1268 (M+, CloHlsO3 requires 294.1256).

3,5-Bis (4-methoxybenzylidene) tetrahydro-4-H-pyran-4-one (EF27): yellow solid (40%). 1H NMR (400 MHz, CDC13) 8 7.79 (2H, s), 7.29 (4H, d, J = 8.8 Hz,), 6.95 (4H,

d, J = 9.2 Hz), 4.93 (4H, d, J = 1.6 Hz), 3.85 (6H, s). 13C NMR (100 MHz, CDCl3) 8 185.6,160.8,136.2,132.7,131.4,127.7,114.4,68.9,55.6. Anal. Calcd for C2lH2004 : C, 74.98; H, 5.99. Found: C, 74.81; H, 6.01. EIHRMS: m/z 336.1362 (M+, C21H20O4 requires 336.1361).

3,5-Bis (2-methoxybenzylidene) tetrahydro-4-H-pyran-4-one (EF28) : yellow solid (67%). 1H NMR (400 MHz, CDCl3) 8 8.09 (2H, s), 7.36 (2H, td, J = 8.8 Hz, J = 1.6 Hz), 7.07 (2H, dd, J = 7. 6 Hz, J = 1. 6 Hz), 6.97 (2H, t, J = 7.6 Hz), 6.93 (2H, d, J = 8 Hz), 4.81 (4H, d, J = 1.6 Hz), 3.87 (6H, s). 13C NMR (100 MHz, CDCl3) 8 186.0,158.6,133.3, 132.5,131.1,130.7,124.1,120.3,111.0,69.1,55.7. Anal. CalcdforC2lH20o4 : C, 74.98; H, 5.99. Found : C, 74.84 ; H, 5.92. EIHRMS: m/z 336.1370) (M', C21H2004 requires 336.1361).

Example 5 Preparation of EF19, EF20, EF32 The compounds of this series were all synthesized by the following procedure: A solution of substituted dienone (0.69 mmol) in EtOH abs (29 mL) was subject to hydrogenation at 33 psi using Raney Nickel as the catalyst for 4 hrs. Filtration through CELITE and concentration under vacuum yielded the crude product, which was purified by chromatography on silica gel using 25 % EtOAc/hexanes. A description of each compound obtained by the above process is given below.

1, 5-Bis (2,4-difluorophenyl)-pentan-3-ol (EF19): white solid (92%). IH NMR (300 MHz, CDC13) 8 7.17-7.09 (2H, m), 6.82-6.72 (4H, m), 3.60 (1H, m), 2.84-2.60 (4H, m), 1.80-1.75 (4H, m), 1.59 (1H, s-br). 13C NMR (75 MHz, CDC13) 8 162.9 (dd, J = 34.4 Hz, J = 12 Hz), 159.7 (dd, J = 35 Hz, J = 12 Hz), 131. 1 (t, J = 9 Hz), 124.6 (dd, J = 19.7 Hz, J = 3. 8 Hz), 111.2 (dd, J = 20.5 Hz, J = 3.4 Hz), 103.8 (t, J = 25. 7 Hz), 70.6,38.0,25.1.

Anal. Calcd for C17Hl6FO4 : C, 65.38 ; H, 5.16. Found: C, 65.64; H, 5.24. EIHRMS: m/z 312.1137 (M+, C17Hl6FO4 requires 312.1137).

1, 5-Bis (3, 4-difluorophenyl)-pentan-3-ol (EF20): white solid (81%). 1H NMR (300 MHz, CDC13) 8 7.07-6.91 (4H, m), 6.87-6.80 (2H, m), 3.57 (1H, m), 2.77-2.54 (4H, m), 1.73 (4H, t), 1. 41 (1H, s-br). 13C NMR (75 MHz, CDC13) 8 151.2 (dd, J = 107.6 Hz, J = 12.5 Hz), 148.0 (dd, J = 105. 8 Hz, J = 12. 5 Hz), 138.9 (t), 124.3 (d, J = 5.8 Hz), 117.3,

117.1,70.4,39.3,31.5. EIHRMS: m/z 312. 1138 (M+, C17Hl6FO4 requires 312.1137). l, 5-Bis (2-hydroxyphenyI)-pentan-3-ol (EF32): white solid (45%). IH NMR (300 MHz, CDC13) 8 7.56 (2H, s-br), 7.09-7.05 (4H, m), 6.87-6.80 (4H, m), 3.54 (1H, m), 2.92-2.84 (2H, m), 2.69-2.62 (2H, m), 1.80-1.71 (4H, m). 13C NMR (75 MHz, CDCl3) 8 154.11, 130.84,127.91,127.77,121.14,116.05,69.28,37.89,25.87.

Example 6 Preparation of EF21, EF22 The compounds of this series were all synthesized by the following procedure: PCC (107 mg, 0.50 mmol) was added in one portion at room temperature to a stirred solution of alcohol (103 mg. 0.33 mmol) in CH2C12 (5mL). The reaction was stirred at room temperature for 14 hrs (TLC-analysis, no sm left), filtered over CELITE and concentrated. The crude product was purified by chromatography on silica gel using 20 % EtOAc/hexanes. A description of each compound obtained by the above process is given below.

1, 5-Bis (2,4-difluorophenyl)-pentan-3-on (EF21): white solid (92%).'H NMR (400 MHz, CDC13) 8 7.16-7.09 (2H, m), 6.80-6.72 (4H, m), 2.87 (4H, t, J = 7.6 Hz), 2.69 (4H, t, J = 7.6 Hz). 13C NMR (100 MHz, CDC13) 8 208.08,163.00 (dd, J = 48 Hz, J = 11. 7 Hz), 159.73 (dd, J = 49 Hz, J = 12 Hz), 131.39,123.54 (d, J = 15.9), 111.17 (dd, J = 20.8 Hz, J = 3.8 Hz), 103.84 (t, J = 25.5 Hz), 42.93,23.08.

1, 5-Bis (3,4-difluorophenyl)-pentan-3-on (EF22): white solid (86%). IH NMR (400 MHz, CDC13) 8 7.07-7.00 (2H, ddd), 6.98-6.92 (2H, m), 6.87-6.80 (2H, m), 2.84 (4H, t, J = 7.2 Hz), 2.68 (4H, t, J = 7. 2 Hz).

Example 7 Preparation of EF7 EF7 was obtained in a three-step synthesis. To a solution of 2, 5-bis (2- hydroxybenzylidene) acetone (800 mg. 3.00 mmol) in DMF (10 mL) was added imidazole (545 mg, 7.56 mmol) and DMAP (10 mg). The bright yellow solution was cooled to 0°C and tbutyldiphenylchlorosilane (1.75 mL, 6.73 mmol) was added dropwise. After stirring for 30 minutes the cooling bath was removed, and the reaction proceeded at room

temperature until no more starting material or monoprotected alcohol were detectable by TLC. (hexanes/EtOAc = 2/1, Rf (starting material) = 0. 17, Rf (mono) = 0.44, Rf (di) = 0.78). The orange solution was poured into ice water (50 mL) and extracted with ether (3x). The combined organic layers were washed with brine (3x), dried over MgS04 and concentrated. The crude product was purified by plug chromatography on silica gel (15% EtOAc/hexanes). The product (108 %) was obtained as a yellow foam and contains some tbutyldiphenylsilylalcohol. It was used without further purification. lH NMR (400 Mhz, CDC13) 8 8. 48 (2H, d, J = 16.0 Hz), 7.75-7.71 (8H, m), 7.66 (2H, dd, J = 8. 0 Hz, J = 2. 0 Hz), 7.46-7.35 (12H, m), 7.18 (2H, d, J = 16.4 Hz), 6.96 (2H, td), 6.89 (2H, td), 6.50 (2H, dd, J = 8.0 Hz, J = 1.2 Hz), 1.08 (18H, s). 13C NMR (100 MHz, CDC13) b 189.93, 154.83,138.24,135.63,135.38,132. 35,130.30,128.14,127.92,127.26,125.43,121.54, 120.08,26.81,19.88.

Disilylprotected ketone (1.74g, 2.34 mmol) was dissolved in THF and cooled to- 78°C, where CH3Li (1.90 mL, 2.66 mmol, 1.4 M/ether) was added dropwise. After stirring for 10 min the originally bright yellow solution completely cleared, it was quenched with saturated NH4C1, the layers separated and the aqueous phase extracted with ether. The combined organic layers were washed with brine, dried over MgS04 and concentrated. The crude product was purified by chromatography on silica gel (10% EtOAc/hexanes). 60 %, white foam. 1H NMR (400 MHz, CDC13) 5 7.72 (8H, dd, J = 6.4 Hz, J = 1.6 Hz), 7.53 (2H, dd, J = 7. 2 Hz, J = 2. 4 Hz), 7.41 (4H, m), 7.36-7.32 (10H, m), 6.82 (4H, m), 6.45 (2H, d, J = 16. 4 Hz), 6.43 (2H, dd, J = 7.2 Hz, J = 2.8 Hz), 1.67 (3H, s), 1.09 (18H, s). 13C NMR (100 MHz, CDC13) 8 152.96,135.66,135.33,132.81,130.12, 128.33,128.01,127.72,126.42,123.37,121.37,119.62,74.02,28.39 ,26.73,19.79.

EF7: Deprotection of the alcohol was carried out with tetrabutylammonium fluoride (2.2 equiv) in THF. The product was obtained after column chromatography on silica gel (30% EtOAc/hexanes) as a white solid (48 %).'H NMR (400 MHz, CDC13) 8 7. 32 (1H, dd, J = 7. 6 Hz, J = 1.6 Hz), 7.14-7.07 (2H, m), 7.00 (1H, dd, J = 8. 0 Hz, J = 1.6 Hz), 6.89-6.82 (3H, m), 6.75 (1H, dd, J = 8 Hz, J = 1.2 Hz), 6.47 (1H, d, J = 9. 6 Hz), 6.31 (1H, d, J = 16. 4 Hz), 5.69 (1H, d, J = 10 Hz), 5.00 (1H, s-br), 1.66 (3H, s). 13C NMR (100 MHz, CDC13) 8 153.1,153.0,133.9,129.5,129.0,128.1,127.8,126.7,123.9,123.7, 121.4,121.2,121.1,116.5,116.0,78.0,27. 6. Anal. Calcd for Cl8Hl602 C, 81.79; H,

6.10. Found: C, 81.61; H, 6.16. EIHRMS: m/z 264. 1150 (M+, Cz8Hz602 requires 264.1150).

Example 8 Preparation of EF5 1, 5-Bis (2-hydroxyphenyl) penta-1-en-3-ol (EF5): To a solution of 1, 5-bis (2- hydroxyphenyl) penta-1, 4-dien-3-one (EF1) (109 mg, 0.41 mmol) in THF/methanol (10/1) (2.5 mL) was added NaBH4 (40mg, 1.30 mmol) in one portion at 0°C. After stirring for 30 min at this temperature the reaction was quenched with H20 dist and cold brine, diluted with Et2O (10 mL) and neutralized by bubbling COx through the dark-red solution (color change to pale yellow). The aqueous phase was extracted with ether, the combined ether layers washed with brine, dried over MgS04 and concentrated. The crude product was purified by chromatography on silica gel using 30% EtOAc/hexanes.

The product was obtained as a white solid (66%). IH NMR (300 MHz, CDC13) 8 7.27 (2H, dd, J = 8.0 Hz, J = 2.4 Hz), 7.21 (1H, s-br), 7.14-7.06 (3H, m), 6.90-6.83 (3H, m), 6.80-6.74 (2H, m), 6.18 (1H, dd, J = 20. 8 Hz, J = 8. 8 Hz), 5.99 (1H, s-br), 4.24 (1H, t- deformed), 3.23 (1H, s-br), 2.86 (1H, m), 2.71 (1H, m), 1.91 (2H, m). 13C NMR (75 MHz, CDC13) 5 154.25,152.96,132.84,130.75,129.03,127.79,127.62,127.46,125. 71, 123.97,121.12,121.07,116.42,116.22,72.09,37.57,25.76.

Example 9 Preparation of EF6 N (Methoxy)-1, 5-bis (2-hydroxyphenyl) penta-1, 4-dien-3-imine (EF6): Methoxyl-amine hydrochloride (30-35 wt% in H2O, 0.30 mL, 1.12 mmol) was added in one portion to a solution of 1, 5-bis (2-hydroxyphenyl) penta-1, 4-dien-3-one (EF1) in methanol/CHCl3 (2/3) (5 mL). The reaction proceeded at room temperature for 24 hrs, then additional methoxylamine hydrochloride (0.15 mL, 0.56 mmol) was added and the reaction stirred for an additional 24 hrs. After completion (TLC) the solvent was evaporated, the residue dissolved in methanol, stirred with silica gel and purified after concentration by chromatography on silica gel using 30% EtOAc/hexanes. The product was obtained as a pale yellow foam (86%). H NMR (400 MHz, CDC13) 8 7.56 (1H, dd, J = 8.0 Hz, J = 1.6 Hz), 7.48 (1H, dd, J = 8.0 Hz, J = 1.6 Hz), 7.41 (lH, d, J = 16.0 Hz),

7.39 (1H, d, J = 16. 8 Hz), 7.28 (1H, d, J = 16.8 Hz), 7.20-7.13 (2H, m), 6.97-6.90 (2H, m), 6.94 (1H, d, J = 16. 0 Hz), 6.80 (1H, dd, J = 8. 0 Hz, J = 1.6 Hz), 5.53 (1H, s-br), 5.36 (1H, s-br), 4.02 (3H, s). 63CNMR (100 MHz, CDC13) 5 155.43,153.86,153.70,132.28, 130.25,130.10,129.71,128.00,127.91,124.16,123.88,123.57,121. 40,121.30,118.45, 116.37,116.33,62.41.

Example 10 Preparation of MD279U and MD279L A solution of 1, 5-bis (3,4-dimethoxyphenyl) penta-1, 3-dien-3-one in a mixture of EtOH abs and THF (5/1) was subject to hydrogenation at 50 psi using Raney Nickel as the catalyst for 8 hrs. Filtration through CELITE and concentration under vacuum yielded the crude product, which was purified by chromatography on silica gel using 25% EtOAc/hexanes.

1, 5-Bis (3,4-dimethoxyphenyl) penta-3-one (MD279U) : 37%, white solid. 1H NMR (400 MHz, CDC13) 8 6.77 (2H, d, J = 8.8 Hz), 6.69 (4H, m), 3.86 (6H, s), 3.85 (6H, s), 2.84 (4H, t, J = 7.6 Hz), 2.70 (4H, t, J = 7.6 Hz). C NMR (100 MHz, CDCI3) 5 209.61, 149. 00, 147.50,133.77,120.22,111.82,111.38,56.07,55.98,45.01,29.54. Rf=0. 28 (EtOAc/hexanes = 1/2).

1, 5-Bis (3,4-dimethoxyphenyl) penta-3-ol (MD279L): 51%, white solid. IH NMR (400 MHz, CDC13) 66. 79 (2H, d, J = 8. 8 Hz), 6.73 (4H, m), 3.87 (6H, s), 3.86 (6H, s), 3.68 (1H, m), 2.78-2.71 (2H, m), 2.63 (2H, ddd, J = 14.0 Hz, J = 9.2 Hz, J = 6.8 Hz), 1.79 (4H, m), 1.58 (1H, s-br). 13C NMR (100 MHz, CDC13) 8 149.02,147.34,134.80,120.29, 111.84,111.39,71.04.

Example 11 3,5-Bis- (a, a, a-trifluoro-2-toluylbenzylidene)-piperidin-4-one acetate

A suspension of 1.76 g (11.49 mmol) of 4-piperidone hydrate, HC1 salt, in 60.0 mL of glacial acetic acid was saturated with dry HC1 gas and to the resultant solution was added 5.0 g (28.72 mmol) of a, a, a-trifluoro-2-tolualdehyde. The mixture was allowed to stir at room temperature for 72 hrs and then diluted with 50.0 mL of toluene and evaporated under vacuum.

The residue was diluted twice more with 50.0 mL portions of toluene and evaporated under vacuum. The gummy residue was suspended in 50 mL of toluene containing 5.0 mL of ethyl acetate, heated briefly to reflux and allowed to cool to room temperature. The solids formed were collected by suction filtration and dried under high vacuum to afford 3.63 g (67%) of a bright yellow solid.

Example 12 3-5-Bis-(pyridinylidene)-piperidin4-one To a solution of 1. 00 g (6.52 mmol) of 4-piperidone hydrate, HCl salt, and 1.40 g (13.05 mmol) of 2-pyridine carboxaldehyde in 91 mL of a 0.25M solution of aqueous NaOH (22.82 mmol) was added 0.86 mL of a 25% w/w solution (0.65 mmol) of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for three hours, diluted with 100 ml of brine and extracted with three 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to afford 1. 25g (69%) of a red-yellow solid.

Example 13 3,5-Bis- (2-fluoro-3-a, a, a-trifluoromethylbenzylidene)-piperidin-4-one

To a solution of 400 mg (2.61 mmol) of 4-piperidone hydrate, HCl salt, and 1.00 g (5.21 mmol) of 2-fluoro-3-a, a, a-trifluoromethylbenzaldehyde in 36 mL of a 0.25M solution of aqueous NaOH (9.11 mmol) was added 0.35 mL of a 25% w/w solution (0.26 mmol) of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 48 hours, diluted with 100 ml of brine and extracted with two 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo to afford 1.02g (86%) of a yellow foam.

Example 14 3,5-Bis-(2-chlorobenzylidene)-piperidin - one To a solution of 1.0 g (6.52 mmol) of 4-piperidone hydrate, HCl salt, and 1.88 g (13. 37 mmol) of 2-chloro benzaldehyde in 95 mL of a 0.25M solution of aqueous NaOH (22.82 mmol) was added 0.90 mL of a 25% w/w aqueous solution (0.65 mmol) of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 48 hours, diluted with 100 ml of brine and extracted with two 35 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to give 0.92g (41%) of a pale yellow powder.

Example 15 3,5-Bis- (2-a, a, a-trifluoromethylbenzylidiene) - 1-methylpiperidin-4-one To a solution of 1.0 g (8.84 mmol) of 1-methylpiperidine-4-one and 3.08 g (13.37 mmol) of a, a, a-trifluoro-2-tolu aldehyde in 88 mL of a 0.25M solution of aqueous NaOH (22.09 mmol) was added 1.16 mL (0.88 mmol) of a 25% w/w aqueous solution of

cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 3 hours, diluted with 100 ml of brine and extracted with three 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo to give 3.56g (95%) of a pale yellow powder.

Example 16 3,5-Bis-(2-pyrilidinyldene)-1-methylpiperidin-4-one

To a solution of 1.0 g (8.84 mmol) of 1-methylpiperidine-4-one and 1.89 g (16.70 mmol) of 2-pyridine carboxaldehyde in 88 mL of a 0.25M solution of aqueous NaOH (22.09 mmol) was added 1.16 mL (0.88 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 3 hours, diluted with 100 ml of brine and extracted with three 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo to give 2.50g (97%) of a pale yellow powder.

Example 17 3,5-Bis- (4-pyridinylidene)-1-methylpiperidin-4-one

To a solution of 1.0 g (8. 84 mmol) of 1-methylpiperidine-4-one and 1.89 g (16. 70 mmol) of 4-pyridine carboxaldehyde in 88 mL of a 0. 25M solution of aqueous NaOH (22.09 mmol) was added 1.16 mL (0.88 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room

temperature for 3 hours, diluted with 100 ml of brine and extracted with two 60 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo to give 2.15g (84%) of a yellow-orange powder.

Example 18 3,5-Bis-(2, 6-difluorobenzylidene)-1-methylpiperidin-4-one

To a solution of 1.0 g (8.84 mmol) of 1-methylpiperidine-4-one and 2.57 g (18.09 mmol) of 2, 6-difluoro benzaldehyde in 90 mL of a 0.25M solution of aqueous NaOH (22.59 mmol) was added 1.16 mL (0.88 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 12 hours, diluted with 100 ml of brine and extracted with three 40 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was slurried in 100 ml of boiling ethyl acetate and the insolubles were removed by rapid suction filtration. The filtrate was concentrated in vacuo and recrystallized from ethyl acetate to provide 3.01g (94%) of a bright yellow solid.

Example 19 3,5-Bis-(2, 6-difluorobenzylidene)-tropin-4-one

To a solution of 0.50 g (3.59 mmol) of tropinone and 1.05 g (7. 39 mmol) of 2,6- difluor benzaldehyde in 36 mL of a 0. 25M solution of aqueous NaOH (9.04 mmol) was added 0.71 mL (0.54 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 4 hours, diluted with 100 ml of brine and extracted with two 25 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 840 mg (60%) of a bright yellow solid.

Example 20 3,5-Bis- (2-fluorobenzylidene)-tropin-4-one

To a solution of 0.50 g (3.59 mmol) of tropinone and 0.914 g (7.36 mmol) of 2- fluoro benzaldehyde in 36 mL of a 0. 25M solution of aqueous NaOH (9. 04 mmol) was added 0.47 mL (0.36 mmol) of a 25% w/w aqueous solution of cetyltrimethylammoniutn chloride. The mixture was allowed to stir vigorously at room temperature for 12 hours, diluted with 100 ml of brine and extracted with two 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide Example 21 3,5-Bis- (2-pyridinylidene)-tropin-4-one To a solution of 0.75 g (5.39 mmol) of tropinone and 1. 18 g (7.39 mmol) of 2- pyridine carboxaldehyde in 54 mL of a 0.25M solution of aqueous NaOH (13.47 mmol) was added 0.71 mL (0.54 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 48 hours, diluted with 150 ml of brine and extracted with three 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 180 mg (11%) of a tan powder.

Example 22 1, 5-Bis- (2, 3-dimethyoxyphenyl)-penta-1, 4-dien-3-one

To a solution of 0.5 g (8.61 mmol) of acetone and 2.86 g (17.22 mmol) of 2,3- dimethoxy benzaldehyde in 86 mL of a 0.25M solution of aqueous NaOH (21.59 mmol) was added 2.83 mL (2.15 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 72 hours, diluted with 100 ml of brine and extracted with three 50 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.99 g (65%) of a bright yellow solid.

Example 23 1, 5-Bis- (2, 3-methylenedioxyphenyl)-penta-1, 4-dien-3-one To a solution of 0.19 g (3.27 mmol) of acetone and 1.00 g (6.66 mmol) of 2,3- methylenedioxy benzaldehyde in 35 mL of a 0.25M solution of aqueous NaOH (8.79 mmol) was added 0.65 mL (0.49 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 2 hours, at which point it was diluted with 15 mL of 95% ethanol, and stirring was continued for an additional 2 hours. The solution was saturated with sodium chloride and extracted with two 35 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.03 g (98%) of a yellow solid.

Example 24 1, 5-Bis- (4-dimethylaminophenyl)-penta-1, 4-dien-3-one

To a solution of 1.0 g (17.22 mmol) of acetone and 5.26 g (35.30 mmol) of 4- dimethylamino benzaldehyde in 172 mL of a 0.25M solution of aqueous NaOH (43.05 mmol) was added 2.26 mL (1.72 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 72 hours, diluted with 100 ml of brine and extracted with two 75 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.21 g (22%) of a dark red powder.

Example 25 1, 5-Bis- (2, 6-dimethoxyphenyl)-penta-1, 4-dien-3-one To a solution of 0.25 g (4. 31 mmol) of acetone and 1.47 g (8.85 mmol) of 2,6- dimethoxy benzaldehyde in 43 mL of a 0.25M solution of aqueous NaOH (10.80 mmol) was added 0.57 mL (0.43 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 72 hours, diluted with 100 ml of brine and extracted with two 75 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.10 g (72%) of a yellow powder.

Example 26 1, 5-Bis- (2, 3-difluorophenyl)-penta-1, 4-dien-3-one

To a solution of 0.5 g (8.61 mmol) of acetone and 2.51 g (17.65 mmol) of 2,3- difluor benzaldehyde in 86 mL of a 0.25M solution of aqueous NaOH (21.52 mmol) was added 1.13 mL (0.86 mmol) of a 25% w/w aqueous solution of cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room temperature for 48 hours, at which point it was diluted with 100 ml of brine and extracted with two 75 mL portions of methylene chloride. The organic phase was dried over anhydrous MgS04 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 110 mg (4%) of a yellow powder.

Example 27 (E)-3- (2-fluorobenzylidenyl) indolin-2-one To a solution containing 2.0 g (15.02 mmol) of 2-oxindole and 2.05 g (16.52 mmol) of 2-fluoro benzaldehyde in 30 mL of abs. ethanol was added 190 mg (2.25 mmol) piperidine and the mixture was refluxed for 12 hrs. The mixture was allowed to cool to room temperature and the solids formed were collected by suction filtration and washed with two 25 mL portions of cold abs. ethanol. The recovered material was dried under high vacuum for 12 hrs. to afford 3.22 g (90%) of a bright yellow powder.

Example 28 <BR> <BR> (E)-3- (2-pyridinylidenyl) indolin-2-one

To a solution containing 2.0 g (15.02 mmol) of 2-oxindole and 1.77 g (16.52 mmol) of 2-pyridine carboxaldehyde in 30 mL of abs. Ethanol was added 192 mg (2.25 mmol) piperidine and the mixture was refluxed for 12 hrs. The mixture was allowed to cool to room temperature and the solids formed were collected by suction filtration and washed with two 25 mL portions of cold abs. ethanol. The recovered material was dried under high vacuum for 12 hrs to afford 2.82 g (84%) of a pale red powder.

Example 29 (E)-3- (2, 3-difluorobenzylidenyl) indolin-2-one To a solution containing 2.0 g (15.02 mmol) of 2-oxindole and 2.20 g (16.52 mmol) of 2, 3-difluoro benzaldehyde in 30 mL of abs. ethanol was added 192 mg (2.25 mmol) piperidine and the mixture was refluxed for 12 hrs. The mixture was allowed to cool to room temperature and the solids formed were collected by suction filtration and

washed with two 25 mL portions of cold abs. ethanol. The recovered material was dried under high vacuum for 12 hrs.

Example 30 1, 3-Bis- (2-fluorobenzylidene) indan-2-one

A solution of 1.88g (15.13 mmol) of 2-fluorobenzaldehyde in 3.0 mL of abs. ethanol was added over a period of 5 min to a solution containing 1.00 g (7.57 mmol) of 2-indanone and 90 mg (2.27 mmol) of NaOH in 40 mL of a 1: 1 mixture of abs. ethanol and water at room temperature. The mixture was allowed to stir for 12 hrs and solids formed were collected by suction filtration and washed with cold ethanol and dried under high vacuum.

Example 31 3,5-Bis- (2-fluorobenzylidene)-piperidin-4-one-acetate-EF24 4-Piperidone hydrochloride monohydrate (307 mg, 2.00 mmol) was suspended in glacial acetic acid (8 mL) and saturated with HCl gas at room temperature. To the resulting clear solution 2-fluorobenzaldehyde (0.59 mL, 5.60 mmol) was added and the reaction allowed to stand at room temperature for 48 h. The forming yellow crystals were filtered off, washed with EtOH abs and dried under vacuum. Further purification was not necessary.

Yellow crystals (91%). 1H NMR (400 MHz, d6-DMSO) 8 10.11 (1H, s-br), 7.90 (2H, s), 7.57 (2H, qd, J = 7. 6 Hz, J = 1. 6 Hz), 7.51 (2H, td, J = 8. 0 Hz, J = 1. 2 Hz), 7.37 (4H, q, J = 10.0 Hz), 4.37 (4H, s), 3.60-3.20 (1H, s-br), 1.91 (3H, s). 13C NMR (100 MHz, d6- DMSO) 8 181.95,172.04,160.33 (d, J = 249 Hz), 132.57 (d, J = 9 Hz), 131.81 (d, J = 4 Hz), 131. 05,129.86,124.96 (d, J = 3 Hz), 121.49 (d, J = 13 Hz), 116.1 (d, J = 21 Hz), 43.79,21.11. HREIMS: m/z311. 1123 (M+-HOAc, ClgHI5NOF2requires 311.1122).

Example 32 Cell Viability and VEGF/TF Inhibition Analysis As described above, the effect of the compounds of the present invention on Neutral Red uptake, VEGF production, and TF production was measured for a variety of human cancer cell lines. Some of this data is also summarized in Figures 1-5. Note that, as applied to compounds EF4, MD6 and MD10 in the figures, the term"known"means that the listed compounds have appeared in the literature, but without any suggestion that those compounds exhibit anti-angiogenic properties or usefulness as a cancer treatment.

Table 1 below lists the results for selected curcumin analogs of the present invention in comparison to the results for curcumin and other known chemotherapeutic and anti- angiogenic agents for RPMI-7951 cells.

Table 1 Characteristics of Selected Novel Curcumin Analogs in the Human Melanoma Cell Line RPMI-7951 as Measured by the Neutral Red, VEGF ELISAa and TF ELISAd Assays; Comparison with Melanoma and Anti-Angiogenesis Agents Neutral Red Uptake, % VEGF'ELISA TF'ELISA Compound 5 20jnM 5M 20 FM 5 nM 20 FtM DMSO (0.1%) 100 3713 7054 Curcumin 100 14 3989 714 7974 1026 SeriesI MD6 97 6 2826 273 1753 ND MD10 82 6 1585 315 1793 ND EF-1 92 8 1923 234 ND ND EF-2 46 7 1295 84 1009 ND EF-3 77 8 2092 230 ND ND EF-4 98 4 2159 199 ND ND EF-8 97 8 2208 319 ND ND EF-9 88 11 1868 705 ND ND EF-10 ND 8 ND 257 ND ND SeriesII MD279L 100 100 2235 1603 5791 5858 MD279U 100 100 2511 779 7200 5316 EF-15 100 100 2361 894 7506 7663 MelanomaChemotherapeutic Agents Decabazine 100 100 1136 1531 ND ND Cisplatin 100 100

Known Anti-angiogenic Agent Thalidomide 100 100 1810 2827 ND ND a Measurement of the extent of suppression of the vascular endothelial growth factor (VEGF) vs. DMSO control and curcumin.

''% of control.

° In concentration units of pg/ml/well. d Measurement of the extent of suppression of tissue factor (TF) vs. DMSO control and curcumin.

As indicated in the table and accompanying figures, two series of analogs were discovered. The Series I analogs proved to inhibit VEGF production and simultaneously inhibit cell growth for several cancer cell lines. Some of the compounds within this group were also more efficacious than TAXOL in preventing growth of a human breast cancer cell line and more potent than curcumin and CISPLATIN in inhibiting proliferation of normal human and transformed murine VECs.

The Series II analogs, which include EF 15, EF 19-22, MD279L and MD279U, selectively blocked VEGF production without causing cell death. These compounds were also not cytotoxic to normal or malignant VECs.

These results indicate that the analogs of the present invention can directly inhibit tumor and vascular endothelial cell growth as well as shut down the production of VEGF which is vital for tumor-induced angiogenesis. Thus, the results suggest that the novel Series I analogs are potential anti-cancer/anti-angiogenic agents, while the Series II compounds are promising anti-angiogenic drugs with little toxicity to normal VECs.

Example 33 Table 2 Vascular Endothelial Growth Factor (VEGF) Production by Human Melanoma Cell Line, RPMI-7951 measured by VEGF ELISA assay Cell Viability VEGF Concentrations of compounds 5 µM 20 µM 5 1M 20 I1M (%) (%) (pg/ml) (%) (pg/ml) (% DMSO (0.1%) 100 100 4197 100 4197 100 Curcumin 105 47 4111 98 221 5 Our synthetic curcumin analogs which inhibit VEGF production and cell growth.

EF-1 98 43 2573 61 0 0 EF-2 68 42 1522 36 0 0 EF-3 88 43 2827 67 0 0 EF-4 102 38 2920 70 0 0 EF-5 n. d. 84 n.d. n.d. n.d. 41 EF-6 n. d. 72 n. d. n.d. n.d. 45 EF-7 n. d. 78 n. d. n. d. n. d. 27 EF-8 94 54 2276 54 35 1 EF-9 92 70 1925 46 595 14 EF-10 n. d. 54 n. d. n. d. 217 5 EF-11 n.d. 93 n. d. n. d. n. d. 51 EF-12 n. d. 92 n. d n. d n. d. 70 EF-13 n. d. 92 n. d. n. d. n. d. 53 EF-14 n. d. 86 n. d. n. d. n.d. 36 EF-25 12 n. d. 216* 21 n. d. n. d.

A231L n. d. 77 n. d. n. d. n. d. 25 A231U U n.d. 80 n.d. n.d. n.d. 34 A232 n. d. 78 n. d. n. d. n. d. 40 A239 n. d 86 n. d n. d. n. d. 40 Our synthetic curcumin analogs which inhibit VEGF production, but not cell growth.

EF-15 112 111 1187 28 938 22 A279L 110 109 904 22 0 0 A279U 112 113 1197 28 1179 18 Drugs Currently in Market ** Decarbazine 91 99 1171 28 1293 31 *** Thalidomide 105 97 1865 44 2387 57 Other Tested Compounds

MD6 101 42 3200 76 30 1 MD10 91 42 1866 44 0 0 BA 3 93 102 4860 116 2035 48 BA 4 90 84 4708 112 3348 80 Values are a mean of the duplicate assays.

*DMSO (0.1%): VEGF 1045 pg/ml= 100% **Decarbazine: Chemotherapeutic drug currently used for treatment of human melanoma.

***Thalidomide : Anti-angiogenic drug currently under clinical trial. n. d. : not done Example 34 Table 3 Vascular Endothelial Growth Factor (VEGF) and Tissue Factor (TF) Production by Human Prostate Cancer Cell lines, DU-145 and PC-3 measured by VEGF ELISA and TF ELISA, respectively All compounds are used at 20 KM and DMSO (solvent control), 0.1 % at a final concentration.

VEGF TF DU-145 PC-3 DU-145 PC-3 pg/ml pg/ml pg/ml pg/ml DMSO 18760 ~ 2633 2103 ~ 100 7699 ~ 406 203 ~ 6 (100%) (100%) (100%) (100%) Curcumin 17957 ~ 1910 2088 ~ 103 7134 ~ 301 138 ~ 10 (96%) (99%) (93%) (68%) Our synthetic curcumin analogs A 26565+ 9818 1729+ 59 n. d. n. d.

(142%) (82%) B 153421 ~ 5607 2168 ~ 279 n. d. n. d.

(82%) (99%) C 20559+ 9224 1734 +397 n. d. n. d.

(110%) (82%) EF-1 19616 ~ 4624 325 ~ 75 12879 ~ 149 107 ~ 9 (105%) (15%) (167%) (53%)

EF-2 7516+1915 26+ 44 5540+364 73+10 (40%) (1%) (72%) (36%) EF-3 25901 ~ 620 917 ~ 261 6687 ~ 188 140 ~ 8 (138%) (44%) (87%) (69%) EF-4 10274 _ 4467 48 + 83 5180 +420 33 + 7 (55%) (2%) (67%) (16%) A231U 27875 ~ 4446 984 ~ 349 n.d. n.d.

(149%) (47%) A271a 24190 ~ 2160 1196 ~ 438 n.d. n.d.

(129%) (57%) MD6 18428 ~ 3377 1820 ~ 283 n.d. n.d.

(98%) (87%) MD10 7830 ~ 2262 611 ~ 248 5886 ~ 332 0 ~ 0 (42%) (29%) (76%) (0%) BA-8 40883 +6639 1648 +229 n. d n. d (218%) (78%) Values are a mean of the triplicate assays and S. D. n. d. : not done As you can see, DU-145 has a 20-fold higher level of VEGF production than PC-3 cells.

This level is seen only one other cell line, MDA-MB-231 breast cancer cell. Greater the VEGF and TF production, higher concentration of compounds will be required to inhibit the same percentage, e. g., effect of E-2 on DU-145 and PC-3 cells.

Both cell lines do not produce basic FGF (bFGF) at all.

Example 35 Table 4 Vascular Endothelial Growth Factor (VEGF) Production by Human Prostate Cancer Cell Lines, DU-145 & PC-3 measured by VEGF ELISA assay All compounds are used at 20 uM and DMSO (solvent control), 0.1 % at a final concentration.

DU-145 PC-3 VEGF VEGF (pg/ml) (%) pg/ml) (%) DMSO 18760 +2633 100 2103 + 100 100 Curcumin 17957 ~ 1910 96 2088 + 103 99 Our synthetic curcumin analogs A 26565 + 9818 142 1729 + 59 82 B 15321 ~ 5607 82 2168 ~ 279 103 C 20559 ~ 9224 110 1734 ~ 397 82 EF-1 19616 +4624 105 325 ~ 75 15 EF-2 7516 ~ 1915 40 26 ~ 1 EF-3 25901 + 620 149 917 261 44 EF-4 10274 ~ 4467 55 48+ 83 2 EF-5 20140+ 2874 107 2748 ~ 416 131 A231L 27927+ 4466 149 1687 +250 80 A231U 27875 ~ 4446 149 984 ~ 49 47 A232 25308 ~ 4722 135 1955 ~ 437 93 A239 272292148 145 1678 +293 80 A271a 24190+ 2160 129 1196 + 438 57 A272 24155 ~ 4635 129 1864 ~ 339 89 A277a 27403 ~ 3143 146 2007 ~ 30 95 A277b 27124 ~ 1346 145 1868 ~ 38 89 MD6 18428 ~ 3377 98 1820 +283 87 MD10 7830 +2262 42 611 ~ 248 29 BA-8 40883 ~ 6639 218 1648 +229 78 As you can see, DU-145 has a 20-fold higher level of VEGF production than PC-3 cells.

This level is seen only one other cell line, MDA-MB-231 breast cancer cell. Greater the VEGF production, higher concentration of compounds will be required to inhibit the same percentage, e. g., effect of E-2 on DU-145 and PC-3 cells. Both cell lines do not produce basic FGF at all.

Values are a mean of the triplicate assays and S. D.

Example 36 Table 5 Tissue Factor (TF) Production by Human Melanoma Cell Line, RPMI-7951 measured by TF ELISA assay TF Concentrations of compounds 1 uM 5 µM 20 µM (pg/ml) (%) (pg/ml) (%) (pg/ml)) DMSO (0.1%) 7054 100 7054 100 7054 100 Curcumin n. d. 8871 126 1026 15 Our synthetic curcumin analogs EF-2 n. d. 5780 82 1009 14 EF-15 n. d. 7506 106 7663 109 EF-32 n. d. n. d. 8654 123 A231U n. d. 5473 78 5981 85 A279L n. d. 5791 82 5858 83 A279U n. d. 7200 102 5316 75 MD6 6400 91 1753 25 n. d.

MD10 7966 113 1793 25 n. d.

BA-1 n. d. n. d. 6669 95 BA-3 n. d. n. d. 2088 30 BA-4 n. d. 7043 100 2120 30 n. d.: not done. Values are a mean of the duplicate assays.

Example 37 Table 6 Tissue Factor (TF) Production by Human Breast Cancer Cell Line, MDA-MB-231 measured by TF ELISA assay TF Concentrations of compounds 0.5 MM 5 u. M 10 u. M 20 uM (pg/ml)) (pg/ml) (%) (pg/ml) (%) (pg/ml) (%) DMSO (0.1%) 45753 100 45753 100 45753 100 45753 100 Curcumin n. d. 45814 100 44727 98 n. d.

Our synthetic curcumin analogs EF-2 37697 82 n.d. n.d. n.d.

A279L n. d. n. d. n. d. 42601 93 A279L n. d. n. d. n. d. 42190 92 EF-15 n. d. n. d. n. d. 46394 101 n. d. : not done. Values are a mean of the duplicate assays.

Example 38 A number of the compounds of the present invention were screened for activity using the NCI Anti-Tumor Screen. The results are given in Tables 7-9 below. Sixty human tumor cell lines were treated for 48 hours with 10-fold dilutions of compounds at a minimum of five concentrations (. 01 pM-100pM). Sulforhodamine B (SRB) assay was used to calculate cell viability or growth. GI50 refers to the concentration at which the drug inhibits tumor cell growth by 50%. LC50 refers to the concentration at which the drug causes 50% tumor cell death. EF 24 and EF 25 represent the average of three separate experiments.

Table 7 Median Growth Inhibitory Concentration (GI50, uM) of Compounds in NCI Anti-Tumor Screen Panel/Cell Line COMPOUNDS Leukemia EF4 EF7 EF9 EF11 EF15 EF19 EF24 EF25 Curcumin CISPLATIN CCRF-CEM 1.4 12.5 3.1 22.0 41.5 18.2 0.2 0.3 3.2 0.2 HL-60 (TB) 1.9 17.7 4.2 21.3 49.9 18. 1 0.4 1.1 7.9 0.1 K-562 2.5 14.9 3.5 27.4 78.5 19.0 0.3 0.6 3.2 1.0 MOLT-4 2.2 7.7 9.3 19.7 >100 2.6 0.5 0.4 5.0 0.3 RPMI-8226 0.6 14.8 2.4 18.2 49.8 21.7 0.2 0.2 2.0 0.5 SR 1.6 15.2 1.2 21.5 52.0 22. 7 0.1 0.2 3.2 0.1 Non-Small Cell Lung Cancer A549 2.6 14.9 8.9 38.7 >100 18.0 1.9 2.5 12.6 0.8 EKVX 2.3 17.3 11.8 30.6 64.0 17.5 0.9 1.4 15.8 1.6 HOP-62 2.9 16.3 16.6 83.3 >100 18. 0 0.7 1.5 4.0 0.3 HOP-92 2.8 12.7 16.3 23.5 >100 17.5 3.0 2.4 ND 0.6 NCI-H226 19.0 18.7 24.3 >100 >100 19.9 3.6 2.2 20.0 0.8 NCI-H23 1.8 17.6 12.3 35.8 >100 22.8 0.9 1.7 5.0 0.1

NCI-H322M 2. 7 16.9 16.7 36.5 >100 20.3 1.4 1.9 15.8 1.3 NCI-H460 2.3 17.6 12.730.7 >100 17.9 81.0 1.2 7.9 0.1 NCI-H522 2.9 16.2 13.433.3 69.9 18.2 0.9 0.9 5.0 0.8 Colon Cancer COLO 205 2.0 17.5 7.6 39.6 >100 17.5 0.8 2.0 12.6 4.0 HCC-2998 1.7 18.4 3.3 20.5 31.6 17.7 1.1 1.5 3.2 0.1 HCT-116 2.2 17.6 1.9 36.8 >100 17.8 0.2 0.8 3.2 1.3 HCT-15 2.2 17.2 4.9 42.6 49.1 24.8 0.2 0.7 4.0 1.6 HT29 2.2 17.8 4.1 45.9 46.1 17.9 0.3 2.0 5.0 1.3 KM12 1.6 17.1 2. 3 19.1 55.7 17.6 0.2 0.3 5.0 2.0 SW-620 2.7 16.2 3.2 32.5 79.8 17.7 0.2 0.5 4.0 0.8 CNS Cancer SF-268 3.0 19.8 13.7 24.6 >100 21.5 0.6 1.4 6.3 0.2 SF-295 3.4 15.5 14.2 53.8 80.9 18.7 85.0 1.7 7.9 0.3 SF-539 2.0 17.3 2.6 39.9 >100 16.9 0.3 0.5 3.2 0.4 SNB-19 3.2 17.5 14.7 48.0 >100 16.1 0.7 2.2 7.9 1.0 SNB-75 2.0 11.8 11.2 20.3 98.8 19.0 2.0 1.8 6.3 0.6 U251 2.0 16.1 2.1 35.7 95.1 17.4 0.2 0.7 5.0 0.4 Melanoma LOXIMVI 2.2 17.2 3.4 40.3 >100 20.0 0.2 0.2 2.5 0.3 MALME-3M 2. 3 17.5 17.8 40.7 >100 18.0 2.3 1.9 12.6 0.2 M14 1.9 15.7 4.1 33.9 >100 18.3 1.2 1.8 4.0 0.3 SK-MEL-2 2.4 16.8 14.5 24.0 31.7 18.4 2.6 2.0 15.8 1.3 SK-MEL-28 2.9 17.4 18.6 42.5 83.7 17.1 2.1 2.4 5.0 0.8 SK-MEL-5 1.6 15.5 12.1 23.2 53.9 17.1 1.3 1.5 7.9 0.5 UACC-257 2.9 16.9 16.0 29.8 55.4 17.9 1.8 1.6 12.6 1.0 UACC-62 1.5 12.2 13.9 21.3 66.7 15.1 1.6 1.7 6.3 0.3 Ovarian Cancer IGROV1 2.9 15.9 3.8 35.9 >100 17. 2 0.8 0.8 7.9 0.6

OVCAR-3 3.7 15.6 14.1 41.8 >100 17. 9 1. 1 0.9 6.3 1.0 OVCAR-4 2.5 14.1 15.2 28.7 >10017. 4 0.7 2.0 10.0 0.3 OVCAR-5 2.6 16.7 20.0 89.2 >10018. 5 1.3 1.7 15.8 1.3 OVCAR-8 2.9 17.0 3.4 47.1 >100 18. 0 0.4 0.8 7.9 1.0 SK-OV-3 6.7 16.6 15.4>100 >10018.5 1.7 2.4 7.9 1.3 Renal Cancer 786-0 2.3 16.4 3.5 41.3 >10017.0 0.2 0.4 3.2 0.3 A498 2.2 17.2 2.4 34.2 23.3 16.3 0.9 1.3 15.8 2.0 ACHN 3.3 15.7 16.5 41.0 >10017. 8 1.1 1.5 12.6 0.5 CAKI-1 3.1 14.3 15.0 29.2 >10021. 8 0.8 1.8 12.6 0.3 RXF393 0.5 11.0 2.3 39.0 23.5 18.8 0.6 0.6 3.2 0.8 SN12C 2.3 15.2 17.1 34.2 >100 17. 4 1.0 1.4 7.9 1.0 TK-10 3.0 17.4 17.9 41.2 >10017. 4 3.1 2.2 15.8 1.3 UO-31 1.6 13.5 9.6 28.4 96.8 18.2 0.6 1.2 12.6 0.8 Prostate Cancer PC-3 2.1 17.2 4.8 34.9 >10017. 5 0.3 0.5 7.9 1.6 DU-145 1.4 16.4 2.1 29.9 >10018. 1 0.7 1.2 15.8 0.4 Breast Cancer MCF-7 2.7 18.0 3.0 28.7 44.6 17.7 0.2 0.3 3.2 0.4 NCI/ADR-RES2. 7 19.3 30.7 86.1 >100 25.3 0.9 1.6 7.9 0.8 MDA-MB-231 3. 5 19.8 14.1 25.6 >100 22.4 1.3 1.9 20.0 3.2 HS 578T 5.1 22.4 9.9 52.1 >100 21.9 1.0 5.1 10.0 1. 3 MDA-MB-435 1.8 17.5 3.2 31.4 61.2 17.1 0.6 1.0 3.2 1.6 MDA-N 1.6 16.1 2.9 36.9 67.8 16.9 0.5 1.0 2.5 0.8 BT-549 2.4 17.1 6.5 32.9 >100 17. 9 0.7 1.5 5.0 1.3 T-47D 1.7 7.7 4.4 24.7 18.5 15.5 1.3 1.3 7.9 2.0 Mean 2.4 15.8 7.2 34.7 75.9 17.8 0.7 1. 1 6.7 0.7 ND-Not Determined As shown, EF4, EF24 and EF 25 exhibited a lower GI50 than the chemotherapeutic agent CISPLATIN for several cell types.

Table 8 Median Lethal Concentration (LC50, pM) of Compounds in NCI Anti-Tumor Screen Panel/Cell Line COMPOUNDS Leukemia EF4 EF7 EF9 EF19 EF24 EF25 Curcumin CISPLATIN CCRF-CEM 59.1 79.5 >100 72.3 70.9 >100 <100 39.8 HL-60 (TB) 53.5 76.0 >100 66.8 >100 >100 <100 50.1# K-562 91.4 70.7 >100 90.1 75.4 91.8 <100 50.1 MOLT-4 69.5 64.7 >100 75. 6 66.2 >100 79.4 50.1 RPMI-8226 >100 92.7 95.6 >100 54.9 70.9 <100 50.1 SR >100 76.8 77.9 82. 6 68.2 76.3 79.4 50.1 Non-Small Cell Lung Cancer A549 >100 53.3 71.0 65.8 89.4 71.8 79.4 50.1 EKVX 37.2 56.6 49.1 57.7 9.4 24.2 79.4 50.1 HOP-62 69.1 63.1 60.7 58.7 28.8 37.2 63.1 50.1 HOP-92 41.4 54.9 70.4 59.3 23.3 35.4 ND 50.1 NCI-H226 >100 78.8 >100 75.7 48.3 58.0 <100 50.1 NCI-H23 9.4 62.9 62. 3 96.8 5.3 12.9 <100 50.1 NCI-H322M 42.3 56.0 55.6 59.2 17.0 32.2 63.1 50.1 NCI-H460 40.2 66.8 60.9 57.5 50.1 38. 9 63.1 39.8 NCI-H522 >100 >100 57.1 75.4 22.9 14.1 79.4 50.1 Colon Cancer COLO 205 8.7 55.9 52.8 55.9 8.0 33.4 63.1 50.1 HCT-2998 5.9 56.8 32.2 56.1 5.0 5.3 31.6 19.9 HCT-116 13.2 57.6 7.7 56.3 1.8 11.1 50.1 50.1 HCT-15 >100 68.9 64.7 >100 13.6 16.1 79.4 50.1 HT29 29.1 60.6 41.9 62.6 57. 3 67.5 <100 50.1 KM12 6.5 55. 5 14.6 56.1 3.4 10.7 63.1 50.1 SW-620 76.3 65.9 62.6 63.7 4.4 75.3 79.4 50.1 CNS Cancer SF-268 53.2 >100 >100 85.3 55.6 43.3 <100 50.1 SF-295 79.5 55.1 58.8 69.7 20.6 35.0 50.1 50.1

SF-539 24.7 55.7 23.1 55.3 7.6 16.8 31.6 50.1 SNB-19 42.3 55.9 58.8 57.3 19. 3 37.3 63.1 50.1 SNB-75 21.5 49.0 48.8 57.5 8.8 14.0 50.1 50.1 U251 18.5 54.4 12.9 55.8 0.8 5.8 50.1 39.8 Melanoma LOXIMVI >100 73.4 67.6 74.4 6.7 3.6 <100 50.1 MALME-3M 24.3 68.8 73.4 59.6 25.2 19.3 50.1 39.8 M14 13.2 54.0 48.6 68.7 27.7 19.4 50.1 50.1 SK-MEL-2 25. 3 57.0 58. 9 57.0 19.5 12.6 79.4 50.1 SK-MEL-28 33.8 62.9 61.1 55.6 33.1 6.6 50.1 50.1 SK-MEL-5 5.7 53.7 53.1 55.5 6.4 6.6 50.1 7.9 UACC-257 57.8 55.2 62.1 56.3 13.5 8.2 50.1 50.1 UACC-62 6.1 49.6 52.8 53.2 4.5 14.4 50.1 15.8 Ovarian Cancer IGROV1 43.9 60.9 46.2 55.6 7.1 14.5 79.4 50. 1 OVCAR-3 40.7 54.5 52. 1 59. 3 16.2 23. 5 63.1 50.1 OVCAR-4 18.4 53.1 61.6 65.1 17.2 23.3 <100 39.8 OVCAR-5 55.9 58.4 63. 3 61.1 9.2 8.4 79.4 50.1 OVCAR-8 >100 86.4 85.3 67.3 31.3 23.0 79.4 50.1 SK-OV-3 63.6 56.2 53.7 65.9 12.8 26.9 50.1 50.1 Renal Cancer 786-O 15.9 54.8 49.6 55.4 1.6 32.6 39.8 50.1 A498 16.2 55.6 23.0 54.7 3.4 6.0 63.1 50.1 ACHN 32.7 54.0 54.8 56. 3 5.6 18.2 63.1 50.1 CAKI-1 >100 52. 3 93.4 67.8 27.2 53.7 50.1 50.1 RXF393 30.8 49.2 17.9 58.4 3.6 4.6 50.1 50.1 SN12C 21.5 54.4 62. 3 55.9 5.4 12.0 63.1 50.1 TK-10 22.6 55.9 56.5 56. 3 27.8 24.6 63.1 50.1 UO-31 6.2 51. 3 49.5 56.6 4.5 68.1 50.1 25.1 Prostate Cancer PC-3 28.6 55. 6 41.0 56.7 7.2 24.8 63.1 50.1

DU-145 7.0 54.8 29.0 56.6 5.4 20.1 63.1 50.1 Breast Cancer MCF-7 44.1 65.9 42.2 57.1 5.0 44.7 <100 50.1 NCI/ADR-RES >100 >100 >100 >100 60.6 71.0 <100 50.1 MDA-MB-231 >100 74.6 57.7 66.3 9.1 17.2 <100 50.1 HS 578T >100 >100 >100 >100 81.4 72.3 <100 50.1 MDA-MB-435 8.0 56.0 35.9 55.6 7.4 16.3 39.8 39.8 MDA-N 20.2 56.9 33.2 55.2 5.6 20.0 31.6 39.8 BT-549 26.1 56.8 47.3 64.7 4.7 13.6 39.8 50.1 T-47D 50.8 55.4 56.1 68.9 51.9 92.9 <100 50.1 Mean 33.9 61.7 52.5 64.6 13.2 24.2 66.0 46.0 EF4, EF9, EF24, and EF25 exhibited a lower LC50 than the chemotherapeutic agent CISPLATIN for several cell types.

Human tumor and endothelial cell lines were treated for 72 hours with compounds at a minimum of four concentrations between. lllM-40, uM. Neutral Red assay was used to calculate cell viability. The numbers in Table 9 are representative of at least three separate experiments.

Table 9 Median Growth Inhibitory Concentration (GI50, uM) of Compounds in Emory Laboratory Cell Screen Panel/Cell Line COMPOUNDS Melanoma EF2 EF4 EF25 EF34 MD6 MD10 MD283 MD286 MD287 Curcumin RPMI 7951 0.8 3.6 0.6 ND 1.9 2.2 1.4 1.0 0.7 6.3 Breast Cancer MDA-MB-231 1. 5 ND 0.8 0.8 ND ND 1.8 ND ND 11.6 MDA-MB-435 3. 3 ND 1.8 ND ND ND 1.9 ND ND 16.3 HUVECS 1.7 ND 1.5 ND 3.8 14.0 3.1 2.3 6.8 25 Mean 1.8 3.6 1.2 0.8 2.9 8.1 2.1 1.7 3.8 14.8 Many modifications and other embodiments of the invention will come to mind to one skilled in the art to which this invention pertains having the benefit of the teachings

presented in the foregoing descriptions and the associated drawings. Therefore, it is to be understood that the invention is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.