AU2005203131A1 - Curcumin analogues for treating cancer - Google Patents

Curcumin analogues for treating cancer Download PDF

Info

Publication number
AU2005203131A1
AU2005203131A1 AU2005203131A AU2005203131A AU2005203131A1 AU 2005203131 A1 AU2005203131 A1 AU 2005203131A1 AU 2005203131 A AU2005203131 A AU 2005203131A AU 2005203131 A AU2005203131 A AU 2005203131A AU 2005203131 A1 AU2005203131 A1 AU 2005203131A1
Authority
AU
Australia
Prior art keywords
bis
substituted
alkoxy
alkyl
halogen
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
AU2005203131A
Inventor
Brian Adams
Matthew C. Davis
Eva M. Ferstl
Dennis C. Liotta
Mamoru Shoji
James P. Snyder
Ustun B. Sunay
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.)
Emory University
Original Assignee
Emory University
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
Priority claimed from PCT/US2000/032870 external-priority patent/WO2001040188A1/en
Application filed by Emory University filed Critical Emory University
Priority to AU2005203131A priority Critical patent/AU2005203131A1/en
Publication of AU2005203131A1 publication Critical patent/AU2005203131A1/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

S&FRef: 599504D1
AUSTRALIA
PATENTS ACT 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT Name and Address of Applicant: Actual Inventor(s): Address for Service: Invention Title: Emory University, of 2009 Ridgewood Drive, Atlanta, Georgia, 30322, United States of America James P. Snyder Matthew C. Davis Brian Adams Mamoru Shoji Dennis C. Liotta Eva M. Ferstl Ustun B. Sunay Spruson Ferguson St Martins Tower Level 31 Market Street Sydney NSW 2000 (CCN 3710000177) Curcumin analogues for treating cancer The following statement is a full description of this invention, including the best method of performing it known to me/us:- 5845c Curcumin Analogues for Treating Cancer 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 of using such compounds.
Background of the Invention Tissue factor (TF) is a sedimentable, integral membrane receptor protein with an 0 estimated molecular weight of 42-47 kDa. Peritumor fibrin deposition, which is i) to characteristic of most types of human cancer, is the result of the local expression of potent Sprocoagulants 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.
187.doc:SAK SExpression 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, tumour 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, antic tumour, and anti-tumour promoting properties. In addition, curcumin exhibits numerous c other therapeutic effects, including anti-oxidative, anti-thrombotic, anti-inflammatory, C anti-cholesterol and anti-diabetic properties. Two other compounds that have received considerable attention are genistein, a soybean-derived isoflavone tyrosine kinase C 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-tumour 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 tumour microenvironment, thereby blocking tumour angiogenesis and growth, without exhibiting a high level of toxicity with regard to normal vascular endothelial cells. The anti-angiogenic and anti-tumour 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 below.
In another aspect, the present invention provides a pharmaceutical formulation comprising a compound of Formula 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 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.
According to a fourth aspect of the invention, there is provided a compound of the formula: [R:\L1BXXJ3758.doc:ae 2a Ri X
X
1
R
X R 4
R
8
R
3
R
7 wherein: c Xi is C-Yor N; Y is selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, N CF 3 alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, aryl, Ssubstituted aryl, alkaryl, arylalkyl, heteroaryl, substituted heteroaryl, heterocycle, C substituted heterocycle, amino, alkylamino, dialkylamino, carboxylic acid, carboxylic ester, carboxamide, nitro, cyano, azide, alkylcarbonyl, acyl, and trialkylammonium;
X
2 is C-R 2 or N;
X
3 is C-R 6 or N;
RI-R
8 are selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF 3 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, and trialkylammonium; A is selected from the group consisting of: 0 O 0 0 L L LL L L and x x wherein X is O, S, SO, SO 2 or NR; Q is NR; and each R is independently selected from the group consisting of hydrogen, alkyl, alkoxy, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl and dialkylaminocarbonyl; the dashed lines indicate the presence of optional double bonds; L is the point of bonding of A to the compound structure; or a pharmaceutically acceptable salt thereof, with the proviso that 1) when none of X, X 2 and X 3 are N, A is IR:\LIBXX3758.doc:ael S'x' X is O or S, and Y is hydrogen, then R 2 and R< are not hydroxyl; and 2) when none of XI, X 2 and X 3 are N, A is 0 L L X X is NR, Y is hydrogen, halogen, CF 3 alkyl, or alkoxy, then R is Snot H.
According to a fifth aspect of the invention, there is provided a method of treating cancerous tissue in a subject, comprising administering to the subject an effective amount of a compound of the formula: R, X X R
X
2 X3
R
4 Ra R3 R 7 wherein: XI is C-Y or N; Y is selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy,
CF
3 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, and trialkylammonium;
X
2 is C-R 2 or N;
X
3 is C-R 6 or N; R!-RS are selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF 3 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, and trialkylammonium; A is selected from the group consisting of: [R:\LIBXX]3758.doc:aei L L L L and x x wherein X is O, S, SO, SO 2 or NR; Q is NR; and each R is independently selected s from the group consisting of hydrogen, alkyl, alkoxy, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, Salkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl and dialkylaminocarbonyl; C the dashed lines indicate the presence of optional double bonds; and L is the point of bonding of A to the compound structure; or to a pharmaceutically acceptable salt thereof.
According to a sixth aspect of the invention, there is provided a pharmaceutical formulation, comprising a compound of the formula: R, XA X, R x 2
X
3
R
4 R 8
R
3
R
7 is wherein: X is C-Y or N; Y is selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy,
CF
3 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, and trialkylammonium;
X
2 is C-R 2 or N;
X
3 is C-R 6 or N;
RI-R
8 are selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF 3 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, and trialkylammonium; A is selected from the group consisting of: [R:\LIBXX3758.doc:acl C wherein X is O, S, SO, SO 2 or NR; Q is NR; and each R is independently selected from the group consisting of hydrogen, alkyl, alkoxy, substituted alkyl, aryl, substituted CI aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, 0alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl and dialkylaminocarbonyl; C the dashed lines indicate the presence of optional double bonds; and L is the point of bonding of A to the compound structure; or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
[R:\LIBXX3758.doc:ae
S
iBRIEF DESCRIPTION OF THE DRAWINGS Having thus described the invention in general terms, reference will now be made c to the accompanying drawings, wherein: Figures 1A and 1B graphically illustrate the relationship between cell viability O 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; M 10 Figures 3A and 3B graphically illustrate the effect of known compounds and the compounds of the present invention on human breast cancer cell viability; SFigures 4A and 4B graphically illustrate the effect of known compounds and the C 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 C 1-C4 alkyl. The term "alkoxy" refers to oxygen substituted alkyl, for example, of the formulas -OR or -ROR', wherein R and R' 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, cyclopropyl, cyclobutyl, and the like; acetylene; cyano; alkoxy, e.g.,
U
methoxy, ethoxy, and the like; lower alkanoyloxy, acetoxy; hydroxy; carboxyl; amino; lower alkylaminfo, methylamino; ketone; halo, e.g. chioro or bromo; phenyl; substituted phenyl, and the like. The term "halogen" includes fluorine, chlorine, iodine and bromine.
S 5 "Aryl" means one or more aromatic ring s. 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 0 nninterferiflg groups as substituents.
"Heteroaryl" is an aryl group containing from one to four N, 0, or S atoms(s) or a ri combination thereof, which heteroaryl group is optionally substituted at carbon or nitrogen atom(s) with C 1-6 alkyl, -CF 3 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. 1-eteroaryl rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings.
1-eteroaryl includes, but is not limited to, 5-membered heteroaryls having one hetero atom thiophefles, pyrroles, furans); 5 membered heteroaryls having two heteroatonIS in 1 ,2 or 1,3 positions oxazoles, pyrazoles, imidazoles, thiazoles, purines); 5-membered heteroaryls having three heteroatomS triazoles, thiadiazoles); 5-membered heteroaryYs having 3 heteroatonS; 6-membered heteroaryls with one heteroatom pyridine, quinoline, isoquinolifle, phenanthririe, 5,6cycloheptenoPYridine); 6-membered heteroaryls with two heteroatoms pyridazineS.
cinnolines, phthaiazines, pyrazines, pyrimidines, quinazolines); 6-memnbered heretoaryls with three heteroatonms I ,3,5-triazifle); and 6-membered heteroaryls with four heteroatoms.
"Substituted heteroaryl" is heteroaryl having one or more non-interfering groups as substituents.
"H-eterocycle" or "heterocyclic" means one or more rings of 5, 6 or 7 atoms with or without unsaturatiori or aromatic character and at least one ring atom which is not carbon. Preferred heteroatoins include sulfur, oxygen, and nitrogen. Multiple rings may be fused, as in quinoline or benzofurafl.
it "Substituted heterocycle" is heterocycle having one or more sid chains formed t~lfromn nninterferinlg substituents.
"Non-~interfering substituents" are those groups that yield stable compounds.
Suitable non-intterferng substituents or radicals include, but are not limited to. halo,
C
1 -CwO alkyl,
C
2 -Cio alenC-Coayyl Cr-Clo alkoxy,C-C2aaylCC, lkarl, C-Ciocycloalkyl
C
3 -CIO cycloalkel phenyl, substituted phenyl. oluoYl, xylenyl, biphenyl,
C
2
-C
1 2 alkoxyalkyl,
C
7
-C
1 2 alkoxyaryl,
C
7 -C12 arixalkyl wherein oxyaryl, CrC6 alkylsulfiyll CICtO alkylsulfofl
(CH
2 )m11O4_CPCOaky) he in is from I to 8, aryl, substituted aryl, substituted alkoxy, fluoroalkyl, heterocyclic radical, substituted heterocyclic radical, nitroalkyl, -N02 -CN, NRC(O)(C1Clo alkyl), alkyl), C2-Cto thioalkyl, _c(o)O_(Ci-Co alkyl), -OH, -S02, G00H, -NR2, carbonyl, -CIO alkyl)-CF3, -C(O)-CF3, -C(O)NR2, jCjO akyl)-S(C6-C 2 aryl, -(O)(C6CI2 ryl, -CH2m-o.CH2,-O(CiCjoalkyl) wherein each m is from
I
to 8, C(O)NR2, -C(S)NR2,
-SO
2 NR2, 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 (1) Y
Y
z X1 xl z
X
2
X
2 wherein: Y is 01-1, halogen, or CF 3 Z is H, 01-1, ORi, halogen, or CF 3 X I and X 2 are independently C or N; and A is selected frorn the group consisting of: 0 N
N'L
R
1
R
2 1 N~
N
SR
1 L
-Y
e" R 2 0 L'
L
1
R
2 L
L
I
1
R
2 wherein n1 is 1-8; X 3 isO0, S, SO, S0O, NH, or NRi; Q is NH or NRI; and V 1 -4 are each independently OH, OR 2 or halogen; R, and R 2 are independently H, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarboflyl, aminocboflyl, alkylamiflocarboflYl or dialkylamilocabonyl; the dashed lines indicate the presence of optional double bonds; and L is the point of bonding of A to the Ocompound structure, with the proviso that Z is not H when Y is OH, Cl or Br and A is nL L L L L. L o oo
LL
(CH
2 0 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 above or a compound of Formula (11) R3
O
R4a Rlo R 5sR wherein: Xi is (CH 2 0, S, SO, S02, or NRt2, where
R
1 2 is H, alkyl, substituted alkyl, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl or dialkylarinocarbonyl; mis 1-7; cacb X 5 is independently N or C-RI; halogen, hydroxyl, alkoxy,
CF,
and each RrRi, are independently H, halogen, hydroxyl alkoxy,
CF
3 alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, ary!, substituted aryl, alkaryl, aryalkyl, heteroaryl, substituted heteroaryl, heterocycle, I
I
substituted heterocycle, amino, alkylamfino, dialkylainfo, carboxylic acid. carboxyfic est er, carboxamide, nitro, cyano, azide. alkylcarbofll acyl, or trialkyl ammufonium: and the dashed lines indicate optional double bonds; with the proviso that when X4 is (CIH26, mn is 2-6, and each X 5 is C-R 1
R
3
-RI
1 are not alkoxy, anid when X4 is NR 12 and each X 5 is N, f( 3 -RIO are not alkoxy, alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, aryl.. substituted aryl, alkaryl, arylalkyl, heteroaryl, substituted heteroaryl, amino, alkylainfo, dialkylamidno, carboxylic acid, or alkylcarboflyl.
The present invention includes all stereoiSomeric configurations of the compounds of Formula and Formula including both optical isomers, such as enantiornerS and diastcreoisome1s, and geometric (cis-trans) isomers.
Examples of the compounds of thc present invention include, but are not limited 0 ®r
N
(EF2) (EF3) (EF 1) (EF8) (EF9) (E 10) (EF23) (MD6) (EF 16) (EF 17) (EF 18) R =2-OH R =3-OH R =4-OH R =2-F R =274-F R =3,4-F R =2,6-F R =3,4-(OMe) R =2-OMe R =3-OMe R 4_OMe (EF4) X R =2-OH (EF3 1) X C-1,R 2-OH (EF29) X R =2-FH (EF3O) X 0, R =2-F (EF36) X 0, R 3 ,44Oe (EF2S) X 0, R =2-4OMe) (EF27) X R =4-OMe (EF34) X N R -OHe (EF33) X NMe, R 2-F1 (EF47) x= NMe, R =2.4-F (EF3 5) X =NMe, R 3,4-(OMe) (EF24) X NH 2 OAc, R =2-F (EF26) X NH1C, R =H -8- I
I
(EF15)n0,
R=I
(EF13)n R E) (EF14) n R Z) (EFI 1) n R 2-OH (EFI2)n R H
R>R
(EF37) R, Me, R 3,4-F (MD38) R, Me, R 3,4.(OMe) (EF44) R, =Pr, R =3,4-(OMe)
RIN
0.N
OH
R-
(FF32) R 2-OH (EF48) R =2-F (EF 19) R 2,4-F 2 (EF20) R 3,4-F 2 (MD279 L) R =3,4-(OMe) 0 (EF2I1) R =2,4-F (EF22) R 3,4-F (MD279U)R 3,4-(OMe) OH N OH '(EF6) Oil 01H OH (EF46) (EF4O) (EF4 1) (EF49) (EF39) (EF43) R, C 2 Br, R =3,4-(OMe) R, =Et, R =3,4-F RI= Ei, R =3,4-(OMe) R, =Ph, R=2-F R,=Ph, R 3,4-(OMe) morph. R 3,4-(OMe) R,=menth., R =3,4-(OMe) x Y R (EF42) R =3,4-(OMC) X CI
Y=H
R =3,4-(OMe)
X=OH
Y =Me 2005203131 19 Jul 2005 H~Ho MD239
HO
H3CVO O CHS Hcoo 0CH,
B
0
H
1 00 0CP% -1I1I- BA7 BA8 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-4one, N-mcthyl-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 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, -12rheumatoid arthritis and certain forms of diabetes. Subjects which can be treated include ifn animal subjects, typically vertebrates, including both mammalian human, cat, dog, 8 cow, horse, sheep, pig, monkey, ape, etc.) and avian subjects chicken, turkey, duck, Sgoose, quail, pheasant, etc.). It is believed, for example, that administering an effective n 5 amount of a compound of Formula or Formula (II) to a subject can result in inhibition Sof angiogenesis in cancerous tissue. Thus, the present invention can provide methods for treating tumor-bearing subjects in which the compounds of the invention are Sadministered to the subject in need of such treatment in an amount effective and in a Smanner effective to combat such tumors, for example, by virtue of inhibition of 10 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 Sbelieved that the compounds of the present invention can be used as a prophylactic i 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 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 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 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 or Formula (II) or a pharmaceutically acceptable salt thereof 13 with one or more pharmaceutically acceptable carriers thereof and optionally any other in 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 uniformldy and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier or both, and then, if CK1 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 tab .let 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 -14recipient.
SNasal spray formulations comprise purified aqueous solutions of the active agent with preservative agents and isotonic agents. Such formulations are preferably adjusted Sto a pH and isotonic state compatible with the nasal mucous membranes.
Formulations for rectal administration may be presented as a suppository with a O 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.
M 10 Topical formulations comprise the active compound dissolved or suspended in Ci one or more media such as mineral oil, petroleum, polyhydroxy alcohols or other bases S3 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 or Formula (II) and salts thereof. The technology for forming liposomal suspensions is well known in the art. When the compound of Formula 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 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 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 or Formula (II) is administered to the subject in an amount sufficient to inhibit production ofTF 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 or Formula (II) may be therapeutically effective. The compound of Formula 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 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 370 C and under -16- C0 2 195% air. SVR cells were cultured in DMEM (Mediatech cellgro) containing t) FBS and 2mM L-glutamine. Complete HUVEC media was a gift from the Cell Culture SCenter 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 were then added and the plates were Cc incubated for 72 hours. Supematant from each well was either aspirated or collected and 10 media containing Neutral Red (GIBCO-BRL, Long Island, NY) at a concentration of Sl/ml was then added to each well. The plates were then incubated at 370 C for Sminutes. Next, the cells were washed with twice with PBS and alcoholic-HCI 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 were then added and the plates were incubated for 72 hours. Superatant was then collected from each well and frozen in a -80 0 C freezer until needed. Cell viability was determined by Neutral Red Assay. VEOF ELISA Kit (R D, Minneapolis, MN) was used to determine the amount of VEGF in the culture supematants. 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 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 40 C overnight to solubilize TF. Supematant was then collected from each well and frozen until needed.
-17- IMUBIND Tissue Factor ELISA Kit (American Diagnostica Inc, Greenwich, CT) was used to determnine TF concentration in each sample. The ELISA was carried out according to manufacturer's procedure.
S 5
EXPERJIMIENTAL
Example 1 ~Preparation of EFI, EF2.EF3, EK4 EF25, EF31, EF34 compound The compounds of this series were all synthesized by the following procedure: S 10 Aqueous NaOH (20 wt%, 15m], 75mmol) was added dropwise to a vigorously stirred solution of hydroxybenzaldehyde (51 mniol) and ketone (25 mnmol) in EtOH abs (20 mL).
The reaction was stirred at room tempcrature for 48 his, H 2 0 dist (100 rnL) was added, and the purple solution was neutralized by gently bubbling C0 2 through it. The precipitating yellow solid was filtered off, washed with H 2 0 dist and dried under vacuum. The products were purified by recrystallization. A description of each compound obtained by the above process is given below.
I ,5-Bis(4-hydroxvpheflyl)peftal ,4-dien-3-onC (EFI): yellow solid mp 236'C (acetone
H
2 'H NMR (400 MHz, CD 3 OD) 8 7.71 (211, 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). 1 3 C NMR (100 MI-z, CD 3
OD)
191.9,161.8,145.3, 131.8, 127.8, 123.6,117.1. EIHRMS: ni/z 266.0943 (M C17HI4i03 requires 266.0943).
1 ,5-Bis(2-hydroxyphenyl)penta-l ,4-dien-3-ofle (EF2): yellow solid mp 1 55 0
C
(acetone
H
2 1 H NMR (400 M14z, CD 3 OD) 868.09 (2 H, d, J3 16 7.63 (2H, dd, J 8.4 Hz, J 1.6 Hz), 7.31 d, J 16 Hz), 7.24 td, J 7.6 Hz., J =1.6 Hz), 6.88 4H, J =7.2Hz). "C NMR (100 Mz, CD 3 OD) 6192.8, 158.9, 141.0, 133.1, 130.2, 126.3, 123.2, 121.0, 117.2. Anal. Calcd for C1 7 H1403: C, 78.68; H, 5.30. Found:
C,
76.56; H, 5.32. ElHRMS; ml/z 248.0837
C,
1
H,
2 0 2 requires 248.0837).
3 -hydroxyphcnyl)penta-1,4-dien 3 -oe (EF3): yellow solid mp 198- 200*C (acetone
H
2 'H NMR (400 MHz, CD 3 OD) 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 6.73 (2H, dd, J 8 Hz, J 2.4 Hz). 1 3 C NMR (100 MHz, CD OD) 6 191.6, 159.3, 145.5, 137.6, 18- 131.2,126.4, 121.4,119.1,115.9. Anal. Calcd for C 17 HI403: C, 78.68; H, 5.30. Found: C, 76.41:1-H,5.48. EIHRMS: -L/z 266.0943 C1 7 HI1403 requires 266.0943).
2 6 -Bis(2.hydroxbcE1zylidefle)cyclohexa none (EF4): yellow solid recrystallized from acetonefH2O). 'H NMR (400 MHz, CD 3 OD) 8 7.98 (2H, 7.32 (2HI dd, J 7.T6 S 5 Hz,]J 1.2 Hz), 7.19 (2H, id, J 7.6Hz,]J 1.2Hz), 6.86 (4H, in), 2.86 in), 1.75 (2H, in). 3 C (100 MHz, CD 3 OD) 8 192.8, 158.2.,137.3, 134.5, 131.5, 124.5, 120.2, 116.6, 29.9, 24.8. Anal. Calcd for C 2 0H 1 8 0 3 C, 7 8.4 1; H, 5.92. Found: C, 78.15;9 H, 6.03. EIHRMS: mInz 30 6 1263
C
2 oHIgO 3 requires 306.1256).
3 ,5-Bis(2-hyrdoxybflzylidfe)tetnbydro.bHpyran4-oe (EF25): yellow solid recrystallized from acetonelH2O). 'H NMR (400 MHz, CD 3 OD) 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, in), 4.84 (4H, d,J =1.6 Hz). 3 C NMR (100MHz, CD3OD) 5 187.8, 158.5, 133.9, 132.5, ri 131.9, 123.3, 120.5,116.8,70.0. Anal. Calcd for C 19
HI
6 04: C, 74.01; H, 5.23. Found: C, 73.23; 5.23. EIHRMS: ml/z 290.0933
C
19
H
16 0 4 requires 290.0943).
2,5-Bis(2-hydroxyphenfl)Cyclopentanone (EF3 yellow solid recrystallized from acetone). 'H NMR (400 MHz, CD 3 OD) 5 8.00 (2H, 7.57 (2H, dd, J 8.0 Hz, J= 1.2 Hz), 7.22 (2H, td, J3 8.0 Hz, J3 1.6 Hz), 6.88 (4H, mn), 3.06 (4H, s).
3,S-Bis(2-hydroxybeflzylidefle) 1 -iethyIl4-piperidoflC (EF34): yellow solid recrystallized from methanol/Hl2O). 'H NMR (400 MHz, CD 3 OD) 8.11 (2H, 7.23 (4H, t, 3 7.6 Hz), 6.88 (4H, t, 3 8.0 Hz), 3.76 (4H, d, J 1.2 Hz), 2.42 (3H, 1 3 c NMR (100 MHz, CD 3 OD) 5 188.40, 158.58, 135.09, 133.09, 132.27, 131.64,123.59, 120.38, 116.87. HREIMS: m/z 303.1259
(M+-H
2 0, C 2 oH, 7 N02 requires 303.1259).
Example 2 Peaain o EF8, E 10El, EF23, EF29, EF3O, EF33 The compounds of this series were all synthesized by the following procedure: A solution of fluoro-substitu.ted 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 minol) and ketone (acetone, tetrahydro-4-H-pyran-4-ofe, N-methyl-piperodil- 4 one) (2.50 mmol) in a mixture of ethanol abs (7 inL) and H 2 0 dist. (7 The solution turns yellow immediately, and usually a yellow precipitate starts forming within 10 mnn -19- (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 S 5 process is given below.
i(-noohnl~et-,-de--n (EF8): yellow solid
NMR
(400 MHz, CDCI3,) 8 7.86 d, J 16 Hz), 7.63 td, J3 7.6 Hz, J 1.6 Hz), 7.42- __7.35 (2H, in), 7.18 (2H, d, J1 16 Hz), 7.26-7.10 (4H, in). 1 3 C NMR (100 MHz, CDC]I 3 8 189.2, 161.9 3 253.4 Hz), 136.3 J 2.3 Hz), 132.1 J 9.1 Hz), 129.5 J S 10 2.2 Hz), 127.8 J=6.1 Hz), 124.7 =3.8 Hz), 123.1 J =11.4 Hz), 116.4
J
22.0 Hz). Anal. Calcd for C I 7 H 1 2
F
2 0: C, 75.55; H, 4.48; F, 14.06. Found: C 75.30, H 4.55. EII-RMS: m/z 270.0865
C
17
H,
2
F
2 0 requires 270.0856).
1,-i(,-iloohnlpna14de--n (EF9): yellow solid NMR (300 MHz, CDC 1 3 8 7.79 (2H, d, J3 16.2 Hz), 7.62 (2H, dd, J 15 Hz, J 8.4 7.10 (2H, d, J 16 Hz), 8.85 8.97(4H, in). 3 C NMR (75 MI-z, CDCI1.) 6 188.5, 164.8 (dd, J3 169.6 Hz, J 12.2 Hz), 161.3 (dd, J 160.3 Hz, J 12.2 Hz), 135.3, 130.7 J 10.0 Hz, J =4.6 Hz), 127.3, 119.5, 112.3 J 21.9 Hz), 104.9 J3=25.4 Hz).
Anal. Calcd for C,,1-IwF4O: C, 66.67; H, 3.29. Found: C, 66.38; H, 3.4 1. EIHR.MS: in/z 306.0654 C1 7
H,
0 0F 4 requires 306.0668).
l,5-Bis(3,4-difluoropheflyl)penta1,4-dieno-3-one (EF yellow solid
IH-
NMR (400 MI-z, CDC]I 3 5 7.64 (2H, d, J 16 Hz), 7.44 (214, ddd, J =9.6 1-Iz, 7.6 Hz, J 2 Hz), 7.32 -7.36 (2H, in), 7.18 7.25 (2H, in), 6.96 (2H, d, J 16 Hz). 3 C NMR (100 MHz, CDCG1 3 8 180.0, 152.7 (dd, J 105.4 Hz, J 12.9 H4z), 15 0.2 (dd, J 10 1. 7 Hiz, J1 12.9 Hz), 141.5, 132.1 J 5.2 Hz), 126.2, 125.5 J1 6 Hz, J 3 Hz), 118.2 J1 17.5 Hz), 116.7 J 20.5 Hz). Anal. Calcd for C17HIOF 4 0: C, 66.67; H, 3.29.
Found: C, 66.54; H, 3.28. EIHRMS: m/z 306.0671 C1 7
H,
0 0F4 requires 306.0668).
1 5-Bis(2,6-difluoropbel)penta-I ,4-dieu-3-one (EF23): yellow solid
'H
NMR (400 MI-z, CDCI 3) 8 7.82 (2H, d, J3 16.4 Hz), 7.35 (2H, d, J 16 Hz), 7.33 (2H, td, J =89Hz, J =2.4 Hz), 6.96 (4H, t, i 8.8 Hz). 3 C NMR (100 MHz, CD)C1 3 8 189.5, 162.2 (dd, J =10 17.6 Hz, J 29.2 Hz), 131.6 J= 10.9 Hz), 13 1.0 (t,J 8.7 Hz), 129.9, 113.0 112.1 J 25.6 Hz). Anal. Calcd for C 1 7 H IOF04: C, 66.67; H, 3.29.
I I Found: C, 66.46; H, 3.26, EIHRMS: nih 306.0657
C
17
HI
0 F0 4 requires 306.0668).
3,-i(-looezldnettayrAHprn-n (EF29): recrystallized from hot ethanol). 'H NMR (400 MHz, CDD]3)-8 7.92 (2H, d, J 1.2 Hz), 7.40-7.36 in), 7.20-7.18 (4H, mn), 7. 13 (2H, t, J 9.6 Hz), 4.80 4H, CH2). 3 C NMR (100 MHz, CDC 1) 8 185.1, 16 1.0 J =250.7 Hz), 134.8, 131.5 J =8.7 Hzl), 13 1.1 J 2.2 Hz), 129.6 J 3.7 Hz), 124.3 J 3.7 Hz), 122.8 J 13.8 Hz), 116.2 I 21.9 Hz), 68.9 J =5.1 Hz). Anal. Calcd for C1 9
H
14
F
2 02: C, 73.07; H, 4.52. Found: C, 73,07; H, 4.47. ElHRMS; m/z 312.0950
C,
9
H,
4 F7 2 0 2 requires 312,0962).
3,5-Bis(2 ,4-difluorobefzlyidcfl)tetrhdo-4Hpyran4-one (EF3O): recryst.
S 10 from ethanol/I
H
2 'H NMR (400 MHz, CDCI3) 6 7.85 (2H, 7.21-7.14 in), 6.98-6:87 (2H,mi), 4.77 (4H, 13C NMR (100 MHz, CDC1 3 184.81, 163.95 (dd, J1 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 J 25.5 Hz), 68.81 J 4.4 Hz). Anal. Calcd for C, 9
HI
2 F7 4 02; C, 65.52; H, 3.47. Found: C, 65.67; H, 3.43. EIHRMS: mhz 348.0761
C
19
HI
4 F7 2 02 requires 348.0773).
3,-i(-looezldn~-ehl4pprdn (EF33): (82 yellow solid). 'H NMR (400 MHz, CDC 13) 6 7.90 (2H, 7.40-7.33 (2H, mn), 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, 2.40 (3 H, 3 C NMR (100 MHz, CDCI1 3 6 186.3, 16 1.1 J 251 Hz), 134.9,13 1.1 J =8.1 Hz), 13 1.0 J 3.0OHz), 129.7 J=3.6 Hz), 124.1 J =2.9 Hz), 123.4 J 13.9 Hz), 116.1 J =21.8 Hz), 57.2, 45.8. Anal. Calcd for
C
2 0HI 7
F
2 0N: C, 73.83; H, 5.27; N, 4.30. Found: C, 73.59; H, 5.32; N, 4.39. EIHRMS: m/z 325.1278
C
20 H 1 7 7 2 0N requires 325.1278).
Example 3 Preparation of EF11, EF12, EF13, EF14, The compounds of this series were all synthesizcd by the following procedure: Bjis-diethylphosphorylinethylsul fide, -sulfoxide and sulfone were obtained according to literature procedures (Tetrahedron, 1992, 48, 8065-8072; Phosphorus Sulfur 1981, *369-3 74). A solution of phosphonate (0.60 mniol) and aldehyde (1.25 Mmol) in CH 2 CI2 (3 mL) was added to the heterogeneous mixture of 50 aqueous NaOH (2 niL) and -21-
CH
2 C1 1 (2 mL), containing triethylbenzylajmonium chloride (TEBA, 0.06 mmol). The reaction was stirred at room temperature over night, the product was extracted from the 0 reaction mixture with CH 2 CI2 and purified by column chromatography. A description of each compound obtained by the above process is given below.
5 3 ,S..Bis(2-hydroxybeflzylidene)-sulfone (EFI yellow solid EIOAc/hexafleS). 1I-H NMR (400 MHz, CD 3 OD) 8 7.77 (21H, d, J =15.6 Hz), 7.48 (21H, dd, J =8.4 Hz, J3 2.0 Hz), 7.25 td, J 8.0 Hz, J1 2.5 Hz), 7.22 (2H, d, J 15.6 Hz), 6.88-6.84 (41,mi), 4.91 '3C NMR (100 MHz, CD3OD) 6 158.94, 140.21, 133.55, 131.63, 127.84, 121.15, 121.03, 117.23.
3 ,5-BisbelYidcnesulfone (EFI2): white solid 20% EtOAcI hexanes). 'H NMR (400 MHz, CDC] 13) 8 7.65 d, J3 15.2 Hz, H3), 7.53-7.50 mn), 7.44-7.40 (6H rn), 6.86 (2H, d, J 15.6 Hz, 1 3 CNMR (100 MHz, CDCl 3 8 143.7, 132.7, 131.5, ri129.3, 128.8, 126.5. Anal. Calcd for C 1 6
H
14 S02: C, 71.09; H, 5.22, S, 11.86. Found:
C,
70.88; H, 5.21, S, 12.01. EIHRMS: mlz 270.0715
C
16 H,1 4 S0 2 requires 270.0731).
EE-3,5-BisbeI1zylidenesulfoxide (EFI3): white solid 20% EtOAc, hexanes).
'H
NMR (300 MHz, CDCI3) 8 7.52-7.46 (4H, mn), 7.42-7.35 (6H, in), 7.31 (211, d, J 15.3 Hz), 6.87 (2H, d, J1 15.3 Hz). 3 C NMVR (75 M.1-z, CDC1 3 8 137.0, 133.9, 131.1, 130.0, 129.1, 127.9. Anal. Calcd for C1 6
H,
4 S0: C, 75.56; H, 5.55, S, 12.60. Found: C, 75.33; H, 5.60, S, 12.60.
E,Z-3,5-Bisbeflzyfidenesulfoxide (EFI4): white solid 20% EtOAc, hexanes). 1H NMR (300 MHz, CDC13) 6 7.52-7,40 (10OH, in), 7.33 (11H, d, J3 15.3 Hz), 7.12 (1 H, d, J 10.5 Hz), 6.93 (1 H, d, J 15.6 Hz), 6.39 (1 H, d7 J 10.5 Hz). 3 C NMR (75 MHz, CDCI3) 5 138.5, 136.3, 135.5, 134.1, 130.1, 129.8,129.6, 129.0, 128.8.,127.8. Anal.
Calcd for C1 6
H,
4 S0: C, 75.56; H, 5.55, S, 12.60. Found: C, 75.34; H, 5.54, S, 12.57.
3,S-Bisbenzylideflcsulflde (EFI white solid 5% EtOAc/hexafles) mixture of E,E and E,Z (ca. 2.5: NMR (400 MHz, CDC 1 3 6 7.40-7.10 (20H, mn), 6.79 (2H, d, J3 15.6 Hz), 6.76 (1 H, d, J1 15.6 Hz), 6.62 d, J 15.6 Hz), 6.61 (1IH, d, J 15.6 Hz), 6.53 (IH, J1 10.8 Hz), 6.41 (IH, d, J 10.8 Hz). Anal. Calcd for C, 6 1-l, 4 S: C, 80.63; H, 5.92; S, 13.45. Found. C, 80.40; H, 5.93; S, 13.39.
22 Example 4 Preparation of EF16, EF17, EF18, EF27,,EF28 Thle compounds of this series were all synthesized by the following procedure: NaOH- 10 mmol) was added as a solid to a stirred solution of methoxy-substituted benzaldehyde anisaldehyde (2.50 mmol) and ketone (acetone, tetrahydro4-H-pymnJone) in EtOH abs (5 mL). A yellow solid started forming within I hr. The reaction was stirred at room temperature for 20 hrs, the product filtered off, washed with cold EtOHabs and H20 dist and dried under vacuum. A description of each compound obtained by the above process is given below.
V' ,-i(-etopey~eta14dc--n (EF16): yellow solid Mp 123- 124 -C (EtOI-). 'H NMR (400 MHz. CDCI3) 5 8.07 (2H, d, J 16 7.63 (2H, dd, J =7.6 Hz, J 1.6 Hiz), 7.37 ddd, J1=7.2 H-z, J 1.6 Hz, J 1.2 Hz), 7.18 d, J= 16 llz), 6.99 (2H, t, J3 7.6 Hz), 6.93 (2H, d. J 8.4 Hz). 3 C NMR (100 MHz, CDC1 3 8 190.2, 158.7, 138.4, 131.8, 128.9, 126.4, 124.1, 120.9, 111.4, 55.7. Anal. Calcd for C1 9 HIS03: C, 77.53; H, 6.16. Found: C, 77.26; H, 6.17. EIHRMS: ml/z 294.1256
C
19
H,
8 0 3 requires 294.1256).
l,5-Bis(3-methoxyphenlY)pecta-1,4-dien-3-ofle (EFi yellow solid (40%0/, chromatography using 20 EtOAc/hexafles). 'H NMR (300 MHz, CDCI 3 5 7.70 (211, d, J 15.9 Hz), 7.33 (2H, t, J 7.6 Hz), 7.21 (211, d, J =7.8 Hz), 7.13 (211, t, J3 2.4 Hz), 7.06 (211, d, J 15.9 Hz), 6.96 (2H, ddd, J 8.1 Hiz, J1 2.4 Hz, J 0.9 Hz). 3 C NMR MI-z, CDC1 3 6 188.83, 159.96, 143.31, 136.21, 130.03, 125.72, 121.20, 116.46, 113.36, 55.55.
1,-i(-ebxpeylpna14de--n (EFIS): yellow solid Mp 129- 130 0 C (EtOH). 'H NMR (300 MI-z, CDC1 3 5 7.69 (211, d, J =15.9 Hz, 7.56 d, J =9 Hz), 6.95 (2H1, d, J 15.6 Hz), 6.92 d, J 8.7 Hz). 3 C NMR (75 MHz, CDCI3) 5 188.8, 161.5, 142.7, 130.2, 127.7, 126.6, 114.5, 55.6 Anal. Calcd for C 19 14I1803: C, 77.53; H, 6.16. Found: C. 77.31; H, 6.2 17. EIHRMS: mlz. 294.1268 C1 9 HI803 requires 294.1256).
3,-i(-ebxbnyiee~erhdo4Hprn-n (EF2 yellow solid 'H NMR (400 MHz, C~DC 3 8 7.79 (2H, 7.29 (4H, d, J 8.8 6.95 (411, 23 d, 9. Hz, 493 4H d, J 1.6 Hz), 3.85 (6H, 3 C NMR (100 MHz. CDC1 3 8 185.6, 160.8, 136.2, 132.7, 131.4. 127.7, 114.4, 68.9, 55.6. Anal.CldfrCHzO:, 74.98;H-, 5.99. Found: C, 74.81; H, 6.01. EIHRMS: mlz336.1362 (Ma,C 2 iH 20 04 requires 336.1361).
S 5 3 ,5-Bis(2-InethoxybenzylidenC)t etrahydro4HPaN oe(F):ylosli 'H NMR (400 MHz, CDCI 3 8 8.09 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 d, I =l.6 Hz), 3.87 (6H, 13 CNMR (100 MHz, CDC13) 8 186.0, 158.6, 133.3, 132.5, 131.1, 130.7, 124.1,120.3,111.0,69.1, 55.7. Anal. Calcd for C 2 1
H
20 04: C, 74.98; H, 5.99. Found: C, 74.84; H, 5.92. EIHR.MS: mfz'336.1370)
C
2 1
H
20 04 requires 336.1361).
Example Preparation of EF19, F2O, EF32 is 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 concent-ration 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.
l,5-Bis(2,4-difluorophenylY)penan-oI (EF19): white solid 'H NMR (300 Mliz, CDC1 3 8 7.17-7.09 (2H, in), 6.82-6.72 (414, in), 3.60 (1IH, in), 2.84-2.60 (4H, mn), 1.80-1.75 (4H, in), 1.59 (1IH, s-br). 3 C NMR (75 MHz, CDC1 3 6 162.9 (dd, J1 34.4 Hz, J =12 Hz), 159.7 (dd, J3 35 Hz, J =12 Hz), 13 1.1 J 9 Hz), 124.6 (dd, J3 19.7 Hz, J 3.8 Hz), 111.2 (dd, J 20.5 Hz, J 3.4 Hz), 103.8 J 25.7 Hz), 70.6, 3 8.0, 25. 1.
Anal. Calcd for C1 7
H
36 F04: C, 65.38; H, 5.16. Found: C, 65.64; 5.24. EIHRMS: m/z 312.1137 (MC, C I 7 H 1 6 F04 requires 312.1137).
I,5-Bis(3,4-difluoropheflyl).peftan 3 -o (EF2O): white solid (8 'H NMR (300 MHz, CDCI3) 5 7,07-6.91 (4H, in), 6.87-6.80 (2H, in), 3.57 (1 H, in), 2.77-2.54 (4H, in), 1.73 (4H, 1.41 (11-1, s-br). 1 3 C NMR (75 MI-z, CDC1 3 8 151.2 (dd, J1 107.6 Hz, .1 12.5 Hz), 148.0 (dd, J 105.8 Hz, J1 12.5 Hz), 138.9 124.3 J 5.8 Hz), 117.3, 24 117.1, 70.4, 39.3, 31.5. EIHRMS: mlz312.1138
(M
t Cl 7 HF0 4 requires 312.1137).
l,5-Bis(2-bydroxypheyl)pentan 3 -o (EF32): white solid 'H NMR (300 MHz, CDCI3) 6 7.56 (2H, s-br), 7.09-7.05 (4H, in), 6.87-6.80 (4H, in), 3.54 (11-H, in), 2.92-2.84 (2H, mn), 2.69-2.62 (2H, mn), 1.80-1.71 (4H, in). 3 C NMR (75 MHz, CDCI 3 5 154.11, 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 all1 synthesized by the following procedure: PCC (107 mg, 0.50 iniol) was added in one portion at room temperature to a stirred solution of alcohol (103 mng. 0.33 inol) in CH 2 CI2 (5mL). The reaction was stirred at room temperature for 14 hns (TLC-analysis, no sm left), filtered over CELITE and concentrated. The crude product was purified by chromatography on silica gel using EtOActhexafles, A description of each compound obtained by the above process is given below.
l,S-Bis(2,4difluorophflyl)peton 3 on (EF2I): white solid 'H NMR (400 MHz, CDCI 3 8 7.16-7.09 (2H, mn), 6.80-6.72 (4H, in), 2.87 (4H, t, J 7.6 2.69 (4H, t, J3 7.6 Hz). 1 3 C NMR (100 MHz, CDCI 3 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 J 15.9), 111.17 (dd, J 20.8 Hz, 3 3.8 Hz), 103.84 J 25.5 Hz), 42.93, 23.08.
l,S-Bis(3,4-difluorophenlY)peftan--on (EF22): white solid 'H NMR (400 MHz, CDCI,) 6 7.07:7.00 (2H. ddd), 6.98-6.92 (2H, in), 6.87-6.80 (2H, in), 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(2hydroxybenzylidele)acetone (800 mg. 3. 00 inmol) in DMF (10 ML) was added imidazole~ (545 mng, 7.56 inmol) and DMAP (10 mng). The bright yellow solution was cooled to 0 0
C
and tbutyl'diphenylchlorosi lane (1.75 mL, 6.73 inmol) was a dded 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 The combined organic layers were washed with brine dried over MgSO4 and S 5 concentrated. The crude product was purified by plug chromatography on silica gel 0\ EtOAc/hexanes). The product (108 was obtained as a yellow foam and contains some tbutyldiphenylsilylalcohol. It was used without further purification. 'H NMR (400 Mhz, CDCI3) 6 8.48 (2H, d, J 16.0 Hz), 7.75-7.71 (8H, 7.66 (2H, dd, J 8.0 Hz, J SHz), 7.46-7.35 (12H, 7.18 (2H, d, J =16.4 Hz), 6.96 (21H, td), 6.89 (2H, td), 6.50 (2H, dd,J= 8.0 Hz,J 1.2 Hz), 1.08 (18H, "C NMR(100 MHz, CDCI 3 8 189.93, 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.
r, Disilylprotected ketone (1.74g, 2.34 mmol) was dissolved in THF and cooled to 78 0 C, where CH 3 Li (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
NH
4 CI, the layers separated and the aqueous phase extracted with ether. The combined organic layers were washed with brine, dried over MgSO4 and concentrated. The crude product was purified by chromatography on silica gel EtOAc/hexanes). 60 white foam. 'H NMR (400 MHz, CDCI3) 6 7.72 (81H, dd, J 6.4 Hz, J 1.6 Hz), 7.53 (2H, dd, J 7.2 Hz, J 2.4 Hz), 7.41 (4H, 7.36-7.32 (10H, m), 6.82 (4H, 6.45 (2H, d, J 16.4 Hz), 6.43 (2H, dd, J 7.2 Hz, J 2.8 Hz), 1.67 (3H, 1.09 (18H, 3 C NMR (100 MHz, CDCI3) 6 152.96, 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, CDCI 3 7.32 (1H, dd, J 7.6 Hz, J 1.6 Hz), 7.14-7.07 (2H, 7.00 (1H, dd, J 8.0 Hz, J 1.6 Hz), 6.89-6.82 (3H, 6.75 (1I, dd, J 8 Hz, J 1.2 Hz), 6.47 (1H, d, J 9.6 Hz), 6.31 (IH, d, J 16.4 Hz), 5.69 (1H, d, J= 10 Hz), 5.00 (1H, s-br), 1.66 (3H, "C NMR (100 MHz, CDC 3 6 153.1, 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 C 1 sH 1 602: C, 81.79; H, -26- 6.10. Found: C, 81.61; H,6.16. EIHRMS: rnJz 264.1 150 CjgH 1 602 requires 264.1150).
Example 8 Prcparation of EFS 51,5-Bis(2-hydroxypheyl)pental-en 3 -o (EF5): To a solution of 1,5-bis(2hydroxyphefl)pefta 1,4-dien-3 -one (EFI1) (109 mg, 0.41 minol) in THF/methanol (10/1) (2.5 mnL) was added NaBI{ 4 (40mg, 1.30 mmol) in one portion at 0 0 C. After stirring for 30 in at this temperature the reaction was quenched with H 2 0 dist and cold brine, diluted with Et 2 O (10 mL) and neutralized by bubbling C02 through the dark-red S 10 solution (color change to pale yellow). The aqueous phase was extracted with ether, the combined ether layers washed with brine, dried over MgSO4 and concentrated. The crude product was purified by chromatography on silica gel using 30% EtOAc/hexanes.
The product was obtained as a white solid 'H4 NMR (300 MHz, CDCI 3 8 7.27 (2H, dd, J 8.0 Hz, J 2.4 Hz), 7.21 (11-1, s-br), 7.14-7.06 (3H, in), 6.90-6.83 (3H, in), 6.80-6.74 (2H, in), 6.18 (1I-H, dd, J =20.8 Hz, J 8.8 Hz), 5.99 (1IH, s-br), 4.24 (1IH, tdeformed), 3.23 (1IH, s-br), 2.86 (1 H, in), 2.71 (1IH, in), 1.91 (2H, 3 C NMR MHz, CDC13) 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-hydroxyphelyl)pefta-1 ,4-dien-3-imifle (EF6): Methoxyl-amifle hydrochloride (30-35 wt%/ in H 2 0, 0.30 m.L, 1.12 minol) was added in one portion to a solution of 1,5-bis(2-hydroxyphenyl)pelta ,4dien 3 one (EF l) in methanol/CHC13 (5 rnL). The reaction proceeded at room temperature for 24 hrs, then additional methoxylamine hydrochloride 15 mL, 0.56 inmol) 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 'H NMR (400 MHz, CDC1 3 8 7.56 (1 H, dd, J 8.0 H~z, J 7.48 (1 H, dd, J 8.0 Hz, J 1.6 Hz), 7.41 (11-H, d, 3 16.0 Hz), 27 I
I
7.39 (1IH, d, J =16.8 Hz), 7.28 (1 H d, J 16.8 Hz), 7.20-7.13 (2H, in), 6.97-6.90 (2H, inr) 6 9 4 (1IH, d, J 16.0 Hz), 6.80 (1 H, dd, J 8.0 Hz, J =1.6 Hz), 5.53 (1 H, s-br), 5.36 (1I, s-br), 4.02 3 C NMR (100 MI-z, CDC1 3 8 155.43, 153.86. 153.70,132.28, 130.25, 130.10,129.71, 128.00,. 12i.91, 124.16, 123.88, 123.57, 121.40, 121.30,118.45, S 5 116.37, 116.33, 62.41.
Example Preparation of MD)279U and MD279L A solution of 1, ,5bi s(3,4-dimcthoxypheflyl)pen- 1,3-dien-3 -one in a mixture of S 10 EtOH abs and THF was subject to hydrogenation at 50 psi using Raney Nickel as ri the catalyst for 8 his. Filtration through CELITE and concentration under vacuum yielded the crude product, which was purified by chromatography on silica gel using CI EtOAc/hexafles.
I,-i(,-iebxpey~et--n (MD279U): 37%, white solid. 'H NMR (400 MHz, CDCI 3 8 6.77 (2H1, d, J 8.8 Hz), 6.69 (4H, in), 3.86 (6H, 3.85 s), 2.84 t, J 7.6 Hz), 2.70 (4H, t, J 7.6 Hiz). 3 C NMR (100 MHz, CDCI,) 5 209.6 1, 149.00, 147.50, 133.77, 120.22, 111.82, 111.38, 56.07, 55.98, 45.01, 29.54. Ri=0.28 (EtOAc/hexales 1/2).
l,5-Bis(3,4-dimethoxyphenyl)penta- 3 -ol (MD279L): 51%, white solid. 'H NMR (400 MHz, CDC1 3 8 6.79 (2H, d, J 8.8 Hz), 6.73 (4H, in), 3.87 (6H, 3.86 3.68 (1i, in), 2.78-2.71 (2H, in), 2.63 (2H, ddd, J 14.0 Hz, 3 =9.2 Hz, J3 6.8 Hz), 1.79 (4H..
in), 1.58 (lH, s-br). 3 C NMR (100 MHz, CDCI 3 8 149.02, 147.34, 134.80, 120.29, 111.84, 111.39,71.04.
Example I1I 3,-i-c~~-rfur--oulezldn)pprdn-n acetate
CF
3 0 CF3
N
I1l .t~c 28 A suspension of 1.76 g (11.49 mmol) of 4-piperidone hydrate, HCI salt, in 60.0 mL of in glacial acetic acid was saturated with dry HCI gas and to the resultant solution was added 5.0 g 8 (28.72 mmol) of a,a,a-trifluoro-2-tolualdehyde. The mixture was allowed to stir at room Stemperature for 72 hrs and then diluted with 50.0 mL of toluene and evaporated under vacuum.
5 The residue was diluted twice more with 50.0 mL portions of toluene and evaporated under 0> 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 S collected by suction filtration and dried under high vacuum to afford 3.63 g (67%)of a bright r yellow solid.
Example 12 4 -one 0
N
H
1 To a solution of 1.00 g (6.52 mmol) of4-piperidone hydrate, HCI 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 MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to afford 1.25g of a red-yellow solid.
Example 13 3,5-Bis-(2-fluoro-3-a c a-tri fl uoro m ethylb e nzylidene)-piperid in -4-one F 0
F
F
3 C ~CF 3 -29- To a solution of 400 mg (2.61 mmol) of 4-piperidorle hydrate, HCI salt, and 1.00 g (5.21 mniol)of 2-luoro- 3 ca,ac -trifluoromethiylbenzaldehyde 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 mniol) of cetyltrimethylammfoflium chloride. The mixture was allowed to stir vigorously at room S 5 temperature for 48 hours, diluted with 100 mal of brine and extracted with two 50 ml, portions of methylene chloride. The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo to afford 1 .02g of a yellow foam.
Example 14 3 ,5-BisA2--ChlorebeflYidefl)pipeldinA I I -one
N
To a solution of 1.0 g (6.52 mmol) of 4-piperidofle hydrate, HCI salt, and 1.88 g (13.37 nunol) of 2-chioro 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 cetyltrimethylalfloflium 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 MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to give 0.92g (41 of a pale yellow powder.
Example
CF
2 CF3 H36 To a solution of 1.0 g (8.84 mmol) of I -methylpiperidifle 4 -one and 3.08 g (13.37 nmmol) of a,cL~a-trifluoro-2-tolu aldehyde in 88 ml of a 0.25M solution of aqueous NaOH (22.09 mmrol) was added 1.16 mL (0.88 mmol) of a25% w/w aqueous solution of cetyltrimethylaflmonium 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 MgSO4 and concentrated in vacuo to give 3.56g of a pale yellow powder.
S 5 Example 16 3 ,S-Bis(-pyrilidiflydele1methylpiperidin4one c-I tI to To a solution of 1.0 g (8.84 mmol) of I methylpiperidifle-4ofe and 1.89 g (16.70 ninol) of 2-pyridine carboxaldehyde in 88 mnL of a 0.25M solution of aqueous NaOH (22.09 mmol) was added 1.16 niL (0.88 mmol) of a 25% wfw aqueous solution of cetyltrimethylammofliumn 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 MgSO4 and concentrated in vacuo to give 2.50g of a pale yellow powder.
Example 17 3,5-Bis-(4-py ridinylidefle)-l -methylpiperidinl 4 -one To a solution of 1.0 g (8.84 rmol) of I -methylpiperidine4 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 mnL 88 minol) of a 25% w/w aqueous solution of cetyltrimethylanuflonim chloride. The mixture was allowed to stir vigorously at room -31temperature for 3 hours, diluted with 100 ml of brine and extracted with two 60 mL portions of methylenle chloride. The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo to give 2.1 5g of a yellow-orange powder.
Example 18 3 ,S-Bis-(2,6-difluorobetkzylidene)-rn ethylpiperidifl 4 -one
CH
3 To a solution of 1.0 g (8.84 mmol) of 1-methylpiperidifle-4-ne and 2.57 g (18,09 mimol) 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 cetyltrinethylalnu1iul 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 anhydrouls MgSQ4 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 3.01lg of a bright yellow solid.
32 Example 19 3 5 -Bis4( 2 ,6difluorobeUzylidene)tropin- 4 -one
H,C.NF
FF To a solution of 0.50 g.(3.
5 9 mmol) of tropinone and 1.05 g (7.39 mmol) of 2,6difluoro benzaldehyde in 36 mL of a 0.25M solution of aqueous NaOH (9.04 mmol) was added 0.71 mL (0.54 mniol) of a 25% w/w aqueous solution of cetyltrimethylarnmonhilm 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 MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 840 mg of a brighit yellow solid.
Example 20 3 ,5-Bis(2fluoroenzylidene)tropin- 4 -one -33 To a solution of 0.50 g (3.59 rnmol) of tropinone and 0.9 14 g (7.36 mmol) of 2fluoro 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 cetylftimethylanunofliWu 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 mfl, portions of methylene chloride. The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide Example 21 3,5-Bis-(2-pyridiflylidcf)tropin 4 -ote
\/N
0 To a solution of 0.75 g (5.39 rmcl) of tropinone and 1. 18 g (7.39 mmol) of 2pyridine carboxaldehyde in 54 mL ofa 0,25M solution of aqueous NaOH (13.47 mmcl) was added 0.71 mnL (0.54 mmcl) of a 25% w/w aqueous solution of cetyltrimethylanlioiflI 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 miL portions of methylene chloride. The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 180 mg (I of a tan, powder.
-34- Example 22 tO 1,5-Bis-(2,3-dimethyoxyphenyl)-penta-1,4-dien-3-one SOCK 0 OCHS cn oc Oa, H3CO OCHn, To a solution of 0.5 g (8.61 mmol) of acetone and 2.86 g (17.22 mmol) of 2,3- Sdimethoxy 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 Sportions of methylene chloride. The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.99 g of a bright yellow solid.
Example 23 1,5-Bis-(2,3-methylenedioxypbhenyl)-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 2,3methylenedioxy 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 MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.03 g of a yellow solid.
I I Example 24 1 ,5-Bis-(4-d im ethylaminopbeflyl)-penta- 1,4-dien-3-one 0 (HIC)2N
N(CH))?
To a solution of 1.0 g (17.22 mmol) of acetone and 5.26 g (35.30 mmol) of 4dimethylaniino benzaldehyde in 172 mL of a 0.25M solution of aqueous NaOH (43.05 mmol) was added 2.26 mL (1.72 mrnol) of a 25% w/w aqueous solution of cetyltrimethylafmmofium chloride. The mixture was allowed to stir vigorously at room S 10 temperature for 72 hours, diluted with 100 ml of brine and extracted with two 75 mL portions of methylene chioride. The organic phase was dried over anhiydrous MgSO 4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1.21 g of a dark red powder.
Example 1 ,5-Bis-(2,6-d imethoxypbenyl)-peflta-1 ,4-dien-3-one OCHS 0
OCH,
OCH
3 H3CO To a solution of 0.25 g (4.31 mmol) of acetone and 1.47 g (8.85 mmol) of 2,6dimethoxy benzaldehyde in 43 mL of a 0.25M solution of aqueous NaOH (10. 80 mmol) was added 0.57 mnL (0.43 mniol) of a 25% w/w aqueous solution of cetyltrimethylaxnmonium 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 chloridc. The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 1. g of a yellow powder.
36 I I Example 26 1,5-Bis-(2,3-difuorophenyl)-penta-1,4-dien-3-one F 0 F F
F
To a solution of 0.5 g (8.61 mmol) of acetone and 2.51 g (17.65 mmol) of 2,3difluoro benzaldehyde in 86 mL of a 0.25M solution of aqueous NaOH (21.52 mmol) m was added 1.13 mL (0.86 mmol) of a 25% w/w aqueous solution of r t cetyltrimethylammonium chloride. The mixture was allowed to stir vigorously at room S 10 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 MgSO4 and concentrated in vacuo. The residue was recrystallized from ethyl acetate to provide 110 mg of a yellow powder.
Example 27 (E)-3-(2-fluorobenzylidenyl)indolin-2-one
F
N
H
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 of a bright yellow powder.
-37- Example 28 To a solution containing 2.0 g (15.0 2 mmol) of 2-oxildole and 1.77 g (16.52 mmol) of 2-pyridinc carboxaldehyde in 30 ml of abs. Ethanol was added 192 mg (2.25 mmol) piperidirie and the mixturc 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 recovercd material was dried under high vacuum for 12 hrs to afford 2.82 g of a pale red powder.
Example 29 1S (E)-3-(2,3-difluorobeflzylidcflyl)indolin- 2 -ol 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 m-L of abs. ethanol was added 192 mg (2.25 minol) 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 -38- I I washed with two 25 mL portions of cold abs. ethanol. The recovered material was dried under high vacuum for 12 hrs.
Example I ,3-Bis-(2-fluorobelzlidne)ildaf-2-ofle A solution of 1.88g (15.13 nimol) 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 nimol) 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-fluorobenzylidefl)pipeidi4onesectate EF24 4-Piperidone hydrochloride monohydrate (307 mg, 2.00 mmol) was suspended in glacial acetic acid (8 mL) and saturated with HCI gas at roonm 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 9 'H NMR (400 MHz, d 6 -DMSO) 8 10. 11 (1lH, s-br), 7.90 (2H, s), 7.57 (2H, qd, J3=7.6 Hz, J =1.6 Hz), 7.51 (2H, tdJ 8.0 Hz, J1 1.2 Hz), 7.37 q, J =10.0 Hz), 4.37 3.60-3.20 (1 H, s-br), 1.91 (3H, 3 C NMR (100 MHz, d 6 DMSO) 8 181.95, 172.04, 160.33 J =249 Hz), 132.57 J =9Hz), 131.81 J= 4 Hz), 131.05, 129.86, 124.96 J 3 Hz), 121.49 J 13 Hz), 116.1 J 21 Hz), 43.79, 21.11. 1-REIMS: m/z 311.1123 (M+-HOAc, C, 9 1 5 N0F 2 requires 311.1122).
-39- I I 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 antiangiogenic 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 ELISA' and TF ELISAd Assays; Comparison with Melanoma and Anti-Angiogenesis Agents Curcumin 100 14 Series I MD6 97 6 MDIO 82 6 EF-1 92 8 EF-2 46 7 EF-3 77 8 EF-4 98 4 EF-8 97 8 EF-9 88 11 ND 8 Series II MD279L 100 100 MD279U 100 100 100 100 Melanoma Chemotherapeutic Agents Decabazine 100 100 Cisplatin 100 100 2826 273 1753 ND 1585 315 1793 ND 1923 234 ND ND 1295 84 1009 ND 2092 230 ND ND 2159 199 ND ND 2208 319 ND ND 1868 705 ND ND ND 257 ND ND 2235 2511 2361 1603 779 894 5791 7200 7506
ND
5858 5316 7663
ND
1136 1531 I I Known Anti-angiogenic Agent Thalidomide 100 100 1810 2827 ND ND C, Measurement of the extent of suppression of the vascular endothelial growth factor (VEGF) vs. DMSO control and curcumin.
b %of control.
SIn concentration units of pg/ml/well.
d Measurement of the extent of suppression of tissue factor (TF) vs. DMSO control and curcumin.
SAs indicated in the table and accompanying figures, two series of analogs were discovered. The Series I analogs proved to inhibit VEGF production and simultaneously Sinhibit 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 EF15, EF19-22, 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.
-41 Example 33 Table 2 Vascular Endothelial Growth Factor (VEGF) Production by Human Melanoma Cell Line, RPMI-7951 measured by VEOF ELISA assay Cell Viabilit)y VEQF Concentrations of compounds 5 WM _20.pM 5 u-M 20 itM DMSO CUTcumin f(NO 100 100 105 47 (]p/mi) 4197 4111 4197 100 221 Our synthetic curcumnin analogs which EF-1 98 43 EF-2 68 42 EF-3 88 43 EF-4 102 38 EF-5 n~d 84 EF-6 rd. 72 EF-7 n.d 78 EF-8 94 54 EF-9 92 70 EF-10 n.d 54 EF-1 I n.d 93 EF-12 n.d 92 EF-13 n.d 92 EF-14 n.d. 86 EF-25 12 n. d.
A231L n.d 77 A231U n.d 80 A232 n.d 78 A239 nd 86 inhibit VEGF production and cell growth.
2573 61 0 0 1522 36 0 0 2827 67 0 0 2920 70 0 0 n.d n.d n.d 41.
m d n.d n.d n.d n.d n.d 27 2276 54 35 1 1925 46 595 14 a d nd 217 nd n.d. n.d 51 a d n.d n.d n.d n. d. nL'd. 53 n.d n.d n.d. 36 216* 21 n n.d n.d n.d n.d. n.d n~d n.d 34 n,d n.d n.d n.d n.d n.d Our synthetic curcunin analogs which inhibit VEGF production, but not cell growth.
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 Thalidomide 105 Other Tested Compounds 1171 28 1865 44 1293 31 2387 57 42 MD6 MD 10 BA 3 BA 4 101 42 3200 91 42 1866 93 102 4860 90 84 4708 76 30 1 44 0 0 116 2035 48 112 3348 Values are a mean of the duplicate assays.
*DMSO VEGF 1045 pg/mfr= 100% Decarbazine: Chemotherapeutic drug currently used for treatment of human melaunomfa.
***Thalidomide: Anti-angiogenic drug currently under clinical trial.
n.d.:not done Example 34 Table 3 Vascular Endothelial Growth Factor (VEOF) 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 uM and DMSO (solvent control), 0.1% at a final concentration.
VEGF
TF
DU-145 PC-3 DU-145 PC-3 pg/mI pg/mI -pg/mI Pg/mI
DMSO
18760 2633 (I100%) Curcumin 17957 +1910 (96%) Our synthetic curcumnin analogs A 26565+ 9818 (I142%) 2103 +100 (100%) 2088 +103 (99%) 1729+ 59 (82%) 2168 +279 (99%) 1734 +397 (82-) 7699 +406 (I100%) 7134 +301 (93%) 203+ 6 (100%l.) 138 (68%) 15321 5607 (82%) 20559 9224 (I010% 19616+ 4624 (I105%) EF- I 325 75 15%j/) 12879+ 149 (167%) 107+ 9 (53%) -43- EF-2 7516+ 1915 26+ 44 5540 +364 73+ C1 5 EF-3 25901 620 917+261 6687+ 188 140+ 8 (138%) (69%) EF-4 10274 +4467 48+ 83 5180 +420 33+ 7 (16%) A231U 27875 +4446 984 +349 n.d n.d.
(149%) (47%) A271a 24190 +2160 1196 +438 nd n.d (129%) (570) MD6 18428 +3377 1820+ 283 n.d n.d (87%) MDIO. 7830+2262 611 +24M5886 +332 0+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-3cls As you can see, DU-145 has a 20-fold higher level of VEGF production than PC-3cls 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, effect of E-2 on DU- 145 and PC-3 cells.
Both ccll lincs do not produce basic FOF (bFGF) at all.
-44- Example Table 4 Vascular Endothelial Growth Factor (VEGF) Production by Human Prostate Cancer Gell Lines, DU-145 PC-3 measured by VEOF ELISA assay All compounds are used at 20 uM and DMSO (solvent control), 0. 1% at a final concentration.
DU-145
P-
VEOF
VG
(pg/mi) (00pg/mi) No) DMS0 18760 +2633 Curcumnin 17957 +1910 Our synthetic curcumin analogs A 26565 +9818 B 15321 +5607 C 20559 +9224 EF-1 19616 +4624 EF-2 7516 +1915 EF-3 25901 620 EF-4 10274+T4467 EF-5 20140 +2874 A231 L 27927 +4466 A231U 27875 +4446 A232 25308 +4722 A239 27229 +2148 A271a 24190+ 2160 A272 24155 +4635 A277a 27403 +3143 A277b 27124 +1346 MD6 18428 +3377 MD1O 7830 +2262 BA-8 40883 +6639 100 96 142 82 110 105 40 149 55 107 149 149 135 145 129 129 146 145 98 42 218 2103 +100 208+ 103.
1729+ 59 2168 +279 1734 397 325+ 75 26+ 44 917T+ 261 48+ 83 274 416 1687 +250 984+--349 1955+ 437 1678 +293 1196 +438 1864+339 2007 +30 1868 +38 1820 +283 611 +;248 1648+229 100 99 82 103 82 1 4-4 2 131 47 93 57 89 9 87 29 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-231I breast cancer cell. Greater the VEGF production, higher concentration of compounds will be required to irdhibit the same percentage, 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.
I I Example 36 Table Tissue Factor (TF) Production by Human Melanoma Cell Line, RPMI-795 I measured by TF ELISA assay
TF
Concentrations of compounds 1 M iM 0IiM DMSO 1%) Curcumin 7054 100 Our synthetic curcumin analogs EF-2 n.d.
n.d.
EF-32 n.d.
A231 U n.d.
A279L n.d.
A279U n.d.
MD6 6400 9' MDIO 7966 11: BA-I n.d.
BA-3 n A.
BA-4 n.d.
7054 100 8871 126 5780 82 7506 106 n.d.
5473 78 5791 82 7200 102 1753 25 1793 25 7054 100 1026 1009 7663 8654 5981 5858 5316 n.d.
n.d.
6669 2088 2120 nd.
7043 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-231I measured by TF ELISA assay
TF
Concentrations of compounds 0.5 UM 51 0-11M 20 &t (pg/mi) N% (Pg/mI) (ON (pg/mi) (pg/mI) ON) DMS0 Curcumin 45753 100 45753 n.d 45814 100 45753 100 44727 45753 100 Our synthetic curcumin analogs EF-2 37697 82 nLd -46- A279L n.d n.d n.d.
A279L n.d n.d n.d.
tnd. n.d n.d.
n.d. not done. Values are a mean of the duplicate assays.
42601 42190 46394 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 (.01M-100iM). 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 Leukemia EF4 EF7 EF9 EFII CCRF-CEM 1.4 12.5 3.1 22.0 1.9 17.7 4.2 21.3 K-562 2.5 14.9 3.5 27.4 MOLT-4 2.2 7.7 9.3 19.7 RPMI-8226 0.6 14.8 2.4 18.2 SR 1.6 15.2 1.2 21.5 Non-Small Cell Lung Cancer A549 2.6 14.9 8.9 38.7 EKVX 2.3 17.3 11.8 30.6 HOP-62 2.9 16.3 16.6 83.3 HOP-92 2.8 12.7 16.3 23.5 NCI-H226 19.0 18.7 24.3 >10C NCI-H23 1.8 17.6 12.3 35.8
COMPOUNDS
EF15 41.5 49.9 78.5 >100 49.8 52.0 >100 64.0 >100 >100 >100 S>100 EF19 18.2 18.1 19.0 2.6 21.7 22.7 18.0 17.5 18.0 17.5 19.9 22.8 EF24 0.2 0.4 0.3 0.5 0.2 0.1 1.9 0.9 0.7 3.0 3.6 0.9 EF25 0.3 1.1 0.6 0.4 0.2 0.2 2.5 1.4 1.5 2.4 2.2 1.7 Curcumin CISPLATIN 3.2 0.2 7.9 0.1 3.2 5.0 0.3 2.0 3.2 0.1 12.6 0.8 15.8 1.6 4.0 0.3 ND 0.6 20.0 0.8 5.0 0.1 -47- NCI-H322M 2.7 NCI-H-460 2.3 NCI-H-522 2.9 Colon Cancer COLO 205 2.0 HCC-2998 1.7 HCT-1 16 2.2 2.2 HT29 2.2 KM12 1.6 SW-620 2.7 CNS Cancer SF-268 3.0 SF-295 3.4 SF-539 2.0 SNB-19 3.2 2.0 U251 2.0 16.9 17.6 16.2 17.5 18.4 17.6 17.2 17.8 17.1 16.2 19.8 15.5 17.3 17.5 11.8 16.1 16.7 36.5 >100 12.7 30.7 >1 00 13.4 33.3 69.9 7.6 3.3 1.9 4.9 4.1 2.3 3.2 13.7 14.2 2.6 14.7 11.2 2.1 39.6 20.5 36.8 42.6 45.9 19.1 32.5 24.6 53.8 39. 9 48.C 20.: 35.~ >100 31.6 >100 49.1 46.1 55.7 79.8 >100 80.9 P>100 >100 98.8 1 95.1 20.3 17.9 18.2 17.5 17.7 17.8 24.8 17.9 17.6 17.7 21.5 18.7 16.9 16.1 19.0 17.4 1.4 81.0 0.9 0.8 1.1 0.2 0.2 0.3 0.2 0.2 0.6 85.0 0.3 0.7 2.0 0.2 1.9 1.2 0.9 2.0 1.5 0.8 0.7 2.0 0.3 0.5 1.4 1.7 0.5 2.2 1.8 0.7 15.8 7.9 5.0 12.6 3.2 3.2 4.0 5.0 5.0 4.0 6.3 7.9 3.2 7.9 6.3 5.0 12.6 15.8 7.9 12.6 6.3 1.3 0.1 0.8 0.1 1.3 1.6 1.3 0.8 0.2 0.3 0.4 0.6 0.4 Melanoma LOX IMVI 2.2 17.2 3.4 40.3 100 20.0 MALME-3M 2.3 17.5 17.8 40.7 >100 18.0 M14 1.9 15.7 4.1 33.9 >100 18.3 SK-MEL-2 2.4 16.8 14.5 24.0 31.7 18.4 SK-MEL-28 2.9 17.4 18.6 42.5 83.7 17.1 SK-MEL-5 1.6 15.5 12.1 23.2 53.9 17.1 UACC-257 2.9 16.9 16.0 29.8 55.4 17.9 UACC-62 1.5 12.2 13.9 21.3 66.7 15.1 Ovarian Cancer IGROVI 2.9 15.9 3.8 35.9 100 17.2 0.8 7.9 48 OVCAR-3 3.7 OVCAR-4 2.5 2.6 OVCAR-8 2.9 SK-OV-3 6.7 Renal Cancer 786-0 2.3 A498 2.2 ACHfN 3.3 CAKI-1 3.1 RXF 393 0.5 SN12C 2.3 3.0 UO0-31 1.6 Prostate Cancer PC-3 2.1 DU-145 1.4 Breast Cancer MCF-7 2.7 NCI/ADR-RES 2.7 MDA-MB-23 1 3.5 HS 578T 5.1 MDA-MB-435 1.8 MDA-N 1.6 BT-549 2.4 T-47D 1.7 Mean 2.4 15.6 14.1 16.7 17.0 16.6 16,4 17.2 15.7 14.3 11.0 15.2 17.4 13.5 14.1 15.2 20.0 3.4 15.4~ 3.5 2.4 16.5 15.0 2.3 17.1 17.9 9.6 41.8 100 17.9 28.7 100 17.4 89.2 100 18.5 47.1 >100 18.0 >100 >10018.5 41.3 34.2 41.0 29.2 39.0 34.2 41.2 28.4 >100 17.0 23.3 16.3 100 17.8 >100 21.8 23.5 18.8 100 17.4 100 17.4 96.8 18.2 1.A 0.7 1 .3 0.4 1.7 0.2 0.9 1.1 0.8 0.6 1.0 3.1 0.6 0.3 0.7 0.2 0.9 1.0 0.6 0.5 0.7 1.3 0.7 0.9 2.0 1.7 0.8 2.4 0.4 1.3 1.5 1.8 0.6 1.4 2.2 1.2 0.5 1.2 0.3 1.6 1.9 5.1 1.0 1.0 1.5 1.3 1.1 6.3 10.0 15.8 7.9 7.9 3.2 15.8 12.6 12.6 3.2 7.9 15.8 12.6 7.9 15.8 3.2 7.9 20.0 10.0 3.2 2.5 5.0 7.9 6.7 0.3 1.3 1.3 0.3 0.3 0.8 1.3 0.8 1.6 0.4 0.4 0.8 3.2 1.3 1.6 0.8 1.3 0.7 17.2 4.8 34.9 100 17.5 16.4 2. 1 29.9 >1 00 18.1 18.0 19.3 19.8 22.4 17.5 16.1 17.1 7 .7 15.8 3.0 30.7 14.1 9.9 3.2 2.9 6.5 4.4 7.2 28.7 44.6 17.7 86.1 >100 25.3 25.6 >100 22.4 52.1 100 21.9 31.4 61.2 17.1 36.9 67.8 16.9 32.9 >100 17.9 24.7 18.5 15.5 34.7 75.9 17.8 ND- Not Detcrmined As shown, EF4, EF24 and EF 25 exhibited a lower G150 than the chemotherapeutic agent CISPLATIN for several cell types.
-49 Table 8 Median Lethal Concentration (LCSO, 11M) of Compounds in NCI Anti-Tumor Screen Panel/Cell Line Leukemia
CCRF-CEM
(TB)
K-562 MOLT-4 RPMI-8226
SR
EF4 E F7 59.1 79.5 53.5 76.0 91.4 70.7 69.5 64.7 >100 92.7 >100 76.8
COMPOUNDS
EF9 EF19 EF24 EF25 Curcun'in
CISPLATIN
>100 72.3 70.9 >100 <100 39.8 >100 66.8 >100 >100 <100 50.1 Non-Small Cell Lung Cancer A549 >100 53.3 EKVX 37.2 56.6 HOP-62 69.1 63.1 HOP-92 41.4 54.9 NCI-H226 >100 78.8 NCI-H23 9.4 62.9 NCI-H322M 42.3 56.0 NCI-H460 40.2 66.8 NCI-H522 >100 >100 Colon Cancer COLO 205 8.7 55.9 H-CT-2998 5.9 56.8 HCT-1 16 13.2 57.6 >100 68.9 HT29 29.1 60.6 >100 >100 95.6 77.9 71.0 49.1 60.7 70.4 >100 62.3 55.6 60.9 57.1 52.8 32.2 7.7 64.7 41.9 14.6 62.6 90.1 75.4 75.6 66.2 >100 54.9 82.6 68.2 65.8 89.4 57.7 9.4 58.7 28.8 59.3 23.3 75.7 48.3 96.8 5.3 59.2 17.0 57.5 50.1 75.4 22.9 55.9 8.0 56.1 5.0 56.3 1.8 >100 13.6 62.6 57.3 56.1 3.4 63.7 4.4 >100 70.9 76.3 71.8 24.2 37.2 35.4 58.0 12.9 32.2 38.9 14.1 33.4 5.3 11.1 16.1 67.5 10.7 75.3 79.4 <1 00 79.4 79.4 79.4 63.1
ND
<100 <100 63.1 63.1 79.4 63.1 31.6 50.1 79.4 <100 63.1 79.4 91.8 <100 50.1 50.1 50.1 50.1 50.1 50.1 50.1 50.1 50.1 50.1 39.8 50.1 50.1 19.9 50.1 50.1 50.1 50.1 50.1 50.1 KM 12 SW-620 CNS Cancer SF-268 SF-295 6.5 55.5 76.3 65.9 53.2 >100 >100 85.3 55.6 79.5 55.1 58.8 69.7 20.6 43.3 <100 35.0 50.1 50 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 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 LOX IMVI >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 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 IGROVI 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 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 786-0 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 CAK1- I >100 52.3 93.4 67.8 27.2 53.7 50.1 50.1 RXF 393 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 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 -51- -?ni t L 50 1 AJ*A IA "j DU-145 7.0 54.8 9.0 56.6 a l~ Breast Cancer SMCF-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 HS578T >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 SMDA-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 10 T-47D 50.8 55.4 56.1 68.9 51.9 92.9 <100 50.1 CMean 33.9 61.7 52.5 64.6 13.2 24.2 66.0 46.0 SEF4, 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 .1 M-40 1 M. 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 MDIO 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 1.7ND 1.5 ND 3.8 14.0 3.1 2.3 6.8 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 -52presented 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 0 and that modifications and other embodiments are intended to be included within the CI 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.
O~~
aO
O-
-53

Claims (39)

1. A compound of the formula: ^-e R X X R R4 RE )R 3 R 7 O wherein: X, is C-Y or N; Y is selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF 3 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, and trialkylammonium; X 2 is C-R 2 or N; X3 is C-R6 or N; RI-R are selected from the group consisting of hydrogen, 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, and trialkylammonium; A is selected from the group consisting of: o 0 0L L L and x wherein X is O, S, SO, SO2, or NR; Q is NR; and each R is independently selected from the group consisting of hydrogen, alkyl, alkoxy, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl and dialkylaminocarbonyl; the dashed lines indicate the presence of optional double bonds; L is the point of bonding of A to the compound structure; or a pharmaceutically acceptable salt thereof, with the proviso that 1) when none of X, X 2 and X 3 are N, A is x X is O or S, and Y is hydrogen, then R2 and R6 are not hydroxyl; and 2) when none ofXi, X2, and X 3 are N, A is O L L x X is NR, Y is hydrogen, halogen, CF 3 alkyl, or alkoxy, then R is not H.
2. A compound according to Claim 1, wherein Xi is N.
3. A compound according to Claim 2, wherein the optional double bonds are present, A is and X is as defined above.
4. A compound according to Claim 3, wherein X is NR, wherein R is as defined above. A compound according to Claim 4, wherein R is hydrogen, alkyl, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl or dialkylaminocarbonyl.
6. A compound according to Claim 3, wherein Y is hydroxyl, halogen, alkoxy, or CF 3 56 0 L L is and X is as defined above.
8. A compound according to Claim 7, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
9. A compound according to Claim 1, wherein the optional double bonds are present, A 0 L L is x and X is NR, and R is as defined above. A compound according to Claim 9, wherein R is hydrogen, alkyl, acyl, alkoxycarbonyl, arninocarbonyl, alkylaminocarbonyl or dialkylaminocarbonyl.
11. A compound according to Claim 9, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
12. A compound according to Claim 1, wherein X 2 and X 3 are N.
13. A compound according to Claim 1, wherein Y is hydroxyl, halogen, alkoxy, or CF3.
14. A compound according to Claim 1, wherein the optional double bonds are present, A O L L is is and X is S. A compound according to Claim 14, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
16. A compound according to Claim 1, wherein the optional double bonds are present, A is and XisO0.
17. A compound according to Claim 16, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
18. A compound according to Claim 1, selected from the group consisting of:- 3,5 -Bis-(2-hydroxybenzylidene)-tetrahydro-4-H-pyran-4-one, 3,5-Bis-(2-hydroxybenzylidene)- 1 -methyl-4-piperidone, 3,5-Bis-(2-fluorobenzylidene)-tetrahydro-4-H-pyran-4-one, 3,5-Bis-(2,4-difluorobenzylidene)-tetrahydro-4-H-pyran-4-one, 3 ,5-Bis-(2-fluorobenzylidene)- I -methyl-4-piperidone, 3,5-Bis-(4-methoxybenzylidene)-tetrahydro-4-H-pyran-4-one, -Bis-(2-methoxybenzylidene)-tetrahydro-4-H-pyran-4-one, 3,5-Bis-(benzylidene)-tetrahydro-4-H-thi opyran-4-one, -Bis-(c(x,a(-tri fluoro-2-toluylbenzylidene)-piperidin-4-one, 3 ,5-Bis-(pyridinylidene)-piperidin-4-one, 3 ,5-Bis-(2-fluoro-3-a,a,c-trifluoromethylbenzylidene)-piperidin-4-one, 3,5-Bis-(2-a,a,x-trifluoromethylbenzylidiene)- I -metbylpiperidin-4-one, 3,5-Bis-(2-pyridinylidene)-1I -methylpiperidin-4-one, -Bis-(4-pyridinyl idene)- 1 -methylpiperidin-4-one, -Bis-(2,6-difluorobenzylidene)-l1-methylpiperidin-4-one, 3,5-B is-(2,6-di fluorobenzylidene)-tropin-4-one, 3 ,5-Bi fluorobenzylidene)-tropin-4-one, and pharmaceutically acceptable salts thereof.
19. A method of treating cancerous tissue in a subject, comprising administering to the subject an effective amount of a compound of the formula: R 3 "7 wherein: SXI is C-Y or N; Ng Y is selected from the group consisting of hydrogen, 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, and trialkylammonium; X2 is C-R 2 or N; X 3 is C-R6 or N; Ri-Rs are selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF 3 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, and trialkylammonium; A is selected from the group consisting of: 0 0 L LL L L and x wherein X is O, S, SO, SO 2 or NR; Q is NR; and each R is independently selected from the group consisting of hydrogen, alkyl, alkoxy, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl and dialkylaminocarbonyl; the dashed lines indicate the presence of optional double bonds; and L is the point of bonding of A to the compound structure; or 59 O (N a pharmaceutically acceptable salt thereof. A method according to Claim 19, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
21. A method according to Claim 19, wherein Xi is N. O 0 L L x and X is as defined above.
23. A method according to Claim 22, wherein X is NR, wherein R is as defined above.
24. A method according to Claim 23, wherein R is hydrogen, alkyl, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, or dialkylaminocarbonyl.
25. A method according to Claim 22, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
26. A method according to Claim 19, wherein the optional double bonds are present, A is 0 L L x and X is S.
27. A method according to Claim 26, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
28. A method according to Claim 19, wherein the optional double bonds are present, A is 0 X is N, and R is as defined above.L x X is NR, and R is as defined above.
29. A method according to Claim 28, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
30. A method according to Claim 19, wherein the optional double bonds are present, A is 0 0 and XisO0. 3 1. A method according to Claim 30, wherein Y is hydroxyl, halogen, alkoxy, or CF3.
32. A method according to Claim 19, wherein said compound is selected from the group consisting of- 3,5-Bis(2-hydroxybenzylidene)tetrahydro-4-H-pyran-4-one, 3,5-Bis(2-hydroxybenzylidene) 1 -methyl-4-piperidone, 3 ,5-Bis(2-fluorobenzylidene)tetrahydro-4-H-pyran-4-one, 3, 5-Bis(2,4-di fluorobenzylidene)tetrahydro-4-H-pyran-4-one, 3,5-Bis(2-fluorobenzylidene) 1 -methyl-4-piperidone, 3 ,5-Bis(4-methoxybenzylidene)tetrahydro-4-H-pyran-4-one, 3,5-Bis(2-methoxybernzylidene)tetrahydro-4-H-pyral-4-one, 3 ,5-Bis-(benzylidene)-tetrahydro-4-H-thiopyran-4-one, 3 ,5-Bis-(cx,ac,c-tri fluoro-2-toluylbenzylidene)-piperidin-4-one, 3,5-Bis-(pyridinylidene)-piperidin-4-one, -Bis-(2-fluorobenzylidene)-piperidin-4-one, 3,5-Bis-(benzylidene)-piperidin-4-one, 3 ,5-Bis-(2-fluoro-3-a,a,a-tri fluoromethylbenzylidene)-piperfidin-4-one,. -Bi s-(2-chlorobenzylidene)-piperidin-4-one, 3 ,5-Bis-(2-c,a,L-tri fluoromethylbenzyl idiene)- I -methylpiperidin-4-one, 3,5-Bis-(2-pyridinylidene)- I -methylpipefidin-4-one, 3,5-Bis-(4-pyridinylidene)- I -methylpiperidin-4-one, 3 ,5-Bis-(2,6-di fluorobenzylidene)- I -methylpiperidin-4-one, 3,5-Bis-(2,6-difluorobenzylidene)-tropin-4-one, 3 ,5-Bis-(2-fluorobenzylidene)-tropin-4-one, and pharmaceutically acceptable salts thereof.
33. A pharmaceutical formulation, comprising a compound of the formula: x R R 3 wherein: X 1 is C-Y orN; Y is selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF3, alkyl, substituted alkyl, alkenyl, alkynyl, cycloalkyl, substituted cycloalkyl, aryl, io substituted aryl, alkaryl, arylalkyl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, amino, alkylamino, dialkylamino, carboxylic acid, carboxylic ester, carboxamide, nitro, cyano, azide, alkylcarbonyl, acyl, and tri alkyl ammonium; X2 is C-R2 or N; X 3 is C-.6 orN; Ri-R8 are selected from the group consisting of hydrogen, halogen, hydroxyl, alkoxy, CF 3 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, and trialkylammonium; A is selected from the group consisting of: 0 0 0 LL L L wherein X is O, S, SO, SO2, or NR; Q is NR; and each R is independently selected from the group consisting of hydrogen, alkyl, alkoxy, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, substituted heterocycle, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl and dialkylaminocarbonyl; the dashed lines indicate the presence of optional double bonds; and ¢c€ 0 L is the point of bonding of A to the compound structure; or i) a pharmaceutically acceptable salt thereof; and Sa pharmaceutically acceptable carrier.
34. A pharmaceutical formulation according to claim 33, wherein Y is hydroxyl, to halogen, alkoxy, or CF 3 A pharmaceutical formulation according to Claim 33, wherein Xi is N.
36. A pharmaceutical formulation according to Claim 35, wherein the optional double 0 L L bonds are present, A is and X is as defined above.
37. A pharmaceutical formulation according to Claim 36, wherein X is NR, wherein R is as defined above.
38. A pharmaceutical formulation according to Claim 37, wherein R is hydrogen, alkyl, acyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, or dialkylaminocarbonyl.
39. A pharmaceutical formulation according to Claim 36, wherein Y is hydroxyl, halogen, alkoxy, or CF3. A pharmaceutical formulation according to Claim 33, wherein the optional double bonds are present, A is and X is S.
41. A pharmaceutical formulation according to Claim 40, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
42. A pharmaceutical formulation according to Claim 33, wherein the optional double o bonds are present, A is X is NR, and R is as defined above.
43. A pharmaceutical formulation according to Claim 42, wherein Y is hydroxyl, halogen, alkoxy, or CF 3 to 44. A pharmaceutical formulation according to Claim 33, wherein the optional double bonds are present, A is and X is O. A pharmaceutical formulation according to Claim 44, wherein Y is hydroxyl, halogen, alkoxy, or CF 3
46. A pharmaceutical formulation according to Claim 33, wherein said compound is selected from the group consisting of: 3,5-Bis(2-hydroxybenzylidene)tetrahydro-4-H-pyran-4-one, 3,5-Bis(2-hydroxybenzylidene)l -methyl-4-piperidone, 3,5-Bis(2-fluorobenzylidene)tetrahydro-4-H-pyran-4-one, 3,5-Bis(2,4-difluorobenzylidene)tetrahydro-4-H-pyran-4-one, 3,5-Bis(2-fluorobenzylidene) I methyl -4-piperid one, 3 ,5 -B is(4-methox ybenzyl idene)tetrahydro-4- H.pyran..4-one, -B is(2-methoxybenzyl idene)tetrahydro-4..H.pyan-4-one, -B i s-(benzyl idene)-tet rahyd ro-4 -H-thi opyran -4-one, 3 ,5-Bis-(a,cx,cx-trifluoro-2-toluylbenzylidene)-piperidin..4-one, 3,5-Bis-(pyridinylidenc)-pipeidii-4-one, 3,5-Bis-(2-fluorobenzylidene)-piperidin-4-one, -Bis-(benzylidenc)-piperidin-4-one, 3,5-Bis-(2-fiuoro-3-axa,a-tri fluoromethylbenzyl idene)-piperidin-4-one, I 0 3 ,5-B is-(2-ch lorobenzyl idene)-p iperi d in-4 -one, s-( 2 -a,ca,c-tri tluoromethylbenzylidiene)- I -methylpiperidin-4-one, 3,5-Bis-(2-pyridinylidcne)- I -methylpiperidin-4 -one, 3 ,5-Bis-(4-pyridinylidene)-lI-methylpiperidin-4-one, 3,5-Bis-(2,6-difluorobenzylidency I -mcthylpiperidin-4-one, 1535Bs(26dfurbez-Vn)trpn4oe is 3,5-Bis-(2,-difluorobenzylidene)-tropin-4-one,n pharmaceutically acceptable salts thereof.
47. A compound as claimed in claim 1, substantially as hereinbefore described with reference to any one of the examples.
48. A method of treating cancerous tissue in a subject, substantially as hereinbefore described with reference to any one of the examples.
49. A pharmaceutical formulation, substantially as hereinbefore described with reference to any one of the examples. A compound as claimed in claim 1, when used for treating cancerous tissue in a subject.
51. The use of a compound as claimed in claim 1 in the manufacture of a medicament for the treatment of cancerous tissue. Dated 19 July, 2005 Emory University Patent Attorneys for the Applicant/Nominated Person SPRUSON FERGUSON
AU2005203131A 1999-12-03 2005-07-19 Curcumin analogues for treating cancer Abandoned AU2005203131A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2005203131A AU2005203131A1 (en) 1999-12-03 2005-07-19 Curcumin analogues for treating cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US16891399P 1999-12-03 1999-12-03
US60/168913 1999-12-03
PCT/US2000/032870 WO2001040188A1 (en) 1999-12-03 2000-12-04 Curcumin analogues for treating cancer
AU2005203131A AU2005203131A1 (en) 1999-12-03 2005-07-19 Curcumin analogues for treating cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU19428/01A Division AU779230C (en) 1999-12-03 2000-12-04 Curcumin analogues for treating cancer

Publications (1)

Publication Number Publication Date
AU2005203131A1 true AU2005203131A1 (en) 2005-08-11

Family

ID=34839222

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005203131A Abandoned AU2005203131A1 (en) 1999-12-03 2005-07-19 Curcumin analogues for treating cancer

Country Status (1)

Country Link
AU (1) AU2005203131A1 (en)

Similar Documents

Publication Publication Date Title
AU779230B2 (en) Curcumin analogues for treating cancer
JP2003515590A5 (en)
AU710070B2 (en) Coralyne analogs as topoisomerase inhibitors
JP5337104B2 (en) Solubilized topoisomerase poison
KR100852965B1 (en) Piperazinedione compounds
JP2013502381A (en) Substituted hydrazide compounds and their applications
JP2002508359A (en) Heterocyclic cytotoxic agent
JPH0667924B2 (en) Substituted indole, benzofuran and benzothiophene derivatives as 5-lipoxygenase inhibitors
HU186650B (en) Process for producing phenol-esters
US7009052B2 (en) Sulfonamide derivatives
JPH0390064A (en) (quinoline-2-yl-methoxy) phenylacetic acid derivative containing cyclic substituent
JP3848158B2 (en) Novel xanthone compounds, their production and use as pharmaceuticals
AU2005203131A1 (en) Curcumin analogues for treating cancer
US9783547B2 (en) Water soluble 4-azapodophyllotoxin analogs
KR100839214B1 (en) Novel dithiolopyrrolones with therapeutic activity
JPS625160B2 (en)
US9309247B2 (en) 2-substituted imidazo[4,5-D]phenanthroline derivatives and their use in the treatment of cancer
IE920704A1 (en) Nitrogen oxides of aza- and diaza-anthracenedione¹derivatives as antitumor agents
WO2024006977A1 (en) Analgesic delta opioid receptor bitopic ligands
CN116589405A (en) Isoquinoline alkaloid derivative capable of efficiently inhibiting autophagy and reversing tumor multidrug resistance as well as preparation method and application thereof
WO2019232665A1 (en) Thiolized indolizine compound having anticancer activity and derivative thereof
KR20000061873A (en) Piperazine derivatives and process for the preparation thereof

Legal Events

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