US20050203077A1 - Selenophene anti-tumor agents - Google Patents

Selenophene anti-tumor agents Download PDF

Info

Publication number
US20050203077A1
US20050203077A1 US11/103,991 US10399105A US2005203077A1 US 20050203077 A1 US20050203077 A1 US 20050203077A1 US 10399105 A US10399105 A US 10399105A US 2005203077 A1 US2005203077 A1 US 2005203077A1
Authority
US
United States
Prior art keywords
compound
solution
group
cancer
nci
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
US11/103,991
Inventor
Ching-Jer Chang
Curtis Ashendel
Darrick Kim
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.)
Individual
Original Assignee
Individual
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/US1997/009717 external-priority patent/WO1997046225A1/en
Application filed by Individual filed Critical Individual
Priority to US11/103,991 priority Critical patent/US20050203077A1/en
Publication of US20050203077A1 publication Critical patent/US20050203077A1/en
Priority to US11/593,371 priority patent/US20070197493A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: PURDUE UNIVERSITY
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D421/00Heterocyclic compounds containing two or more hetero rings, at least one ring having selenium, tellurium, or halogen atoms as ring hetero atoms
    • C07D421/14Heterocyclic compounds containing two or more hetero rings, at least one ring having selenium, tellurium, or halogen atoms as ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/40Radicals substituted by oxygen atoms
    • C07D307/46Doubly bound oxygen atoms, or two oxygen atoms singly bound to the same carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/22Radicals substituted by doubly bound hetero atoms, or by two hetero atoms other than halogen singly bound to the same carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D345/00Heterocyclic compounds containing rings having selenium or tellurium atoms as the only ring hetero atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0834Compounds having one or more O-Si linkage
    • C07F7/0838Compounds with one or more Si-O-Si sequences

Definitions

  • the present invention relates to compositions and a method for treating a patient having a tumor. More specifically, the present invention relates to the treatment of such patients with an effective amount of a selenophene derivative.
  • the control and cure of cancer represents one of our most challenging health problems.
  • the treatment of cancer can be approached by several modes of therapy including surgery, radiation, chemotherapy or a combination of any of these treatments.
  • Chemotherapy continues to be an indispensable therapy for inoperable or metastatic forms of the disease.
  • the mouse L1210 leukemia cell line was initially the preferred model system used for screening natural compounds for antitumor activity.
  • the P388 murine leukemia system was found to be more sensitive and predictive than L1210 leukemia system, and has been used as primary screen during the past decade.
  • Systematic screening for compounds exhibiting toxicity to these two leukemia cell lines has resulted in the isolation of a large number of active natural products.
  • the anticancer activities of these compounds were predominantly in leukemia, lymphoma and a few rare tumors.
  • NCI National Cancer Institute
  • This in vitro prescreening system is based on the measurement of antitumor cytotoxicity against human tumor cell line panels consisting of approximately 60 cell lines of major human tumors (including leukemia and slower growing tumor cells such as lung, colon, breast, skin, kidney, etc.).
  • the most important advantage of the new in vitro screening panels is the opportunity to identify compounds that are selectively more cytotoxic to cells of slowly growing solid tumors than to rapidly growing leukemia cells.
  • the cytotoxicity profile of the NCI human tumor cell panels displays the tumor specificity of a given compound, however the assay does not assess the toxicity of that compound to normal human cells. Accordingly a second bioassay is utilized to measure the selective cytotoxicity against certain types of tumor cells verses normal human cells.
  • the growth and differentiation of cells are regulated by signaling cascades induced by various mitogenic proteins (J. Kurdan and B. L. Taylor, “Signal Transduction,” Academic Press, New York, N.Y. 1994) that often are encoded by proto-oncogenes.
  • the overexpression, amplification or mutation of the oncoprotein is critically involved in the initiation, progression and metastasis of malignant cells (R. A. Weinberg, “Oncogenes and the Molecular Origins of Cancer,” Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989).
  • Many oncoproteins alter normal cellular growth regulation by modulating the intracellular signaling pathways from the membrane to the nucleus.
  • cancer may be considered as a disease of cellular signal transduction, which presents a novel approach for anticancer therapy.
  • One of the critical enzymes involved in the oncoprotein signal transduction is protein kinase C (U. Nishizuka, Nature, 308, 693, 1984 and Science, 233, 305, 1986).
  • protein kinase C U. Nishizuka, Nature, 308, 693, 1984 and Science, 233, 305, 1986.
  • the determination of a compound's ability to inhibit protein kinase C activity has become a good prognostic for discovering novel anticancer agents (A. Basu, Pharmac Ther, 59, 257, 1993).
  • the selenophene compounds will demonstrate selectivity for certain class members of protein kinases, including protein kinase C. Inhibition of a specific classes of protein kinases will allow the treatment of other diseases associated with defects in signaling transduction.
  • Selenophenes are selenium containing heterocyclic compounds that are analogs of naturally occurring thiophene, furan and pyrrole compounds. Selenophenes have been found to be effective antitumor agents, and exhibit enhanced cytotoxicity against slow growing tumor cells; selective cytotoxicity against human renal, ovarian tumor cells, and skin tumor cells; and exhibit inhibition of protein kinase C.
  • R 1 and R 2 are independently selected from the group consisting of; H, CHO, CH 2 OH and CH 2 NH 2 ;
  • X and Y are independently selected from the group consisting of Se, S, O, NCH 3 and NH;
  • R 3 , R 4 , R 5 and R 6 are independently selected from the group consisting of H, CHO, CH 2 OH and CH 2 NH 2 ; cyclodextrin complexes of such compounds; and when R 3 , R 4 , R 5 or R 6 is CH 2 NH 2 , the pharmaceutically acceptable salt of the compound represented thereby.
  • novel cytotoxic compounds of the above formula and chemotherapeutic pharmaceutical compositions containing said compounds in anti-tumor effective amounts are provided.
  • the present invention is directed to selenophene compounds, their pharmaceutical compositions and methods utilizing such compounds/compositions for treating patients having tumor.
  • the selenophene compounds are effective antitumor agents against slow growing tumors, and generally have been found to exhibit high selective cytotoxicity for individual tumor cell lines.
  • the compounds of the present invention are selenophene compounds of the formula I:
  • R 1 and R 2 are independently selected from the group consisting of; H, CHO, CH 2 OH and CH 2 NH 2 ;
  • X and Y are independently selected from the group consisting of Se, S, O and NR;
  • R is H or C 1 -C 7 alkyl
  • R 7 is H, CO(CH 2 ) 2 CO 2 H, (CH 2 ) 2 OCH 3 , C 1 -C 4 alkyl or COC 1 -C 17 alkyl;
  • R 8 is H or C 1 -C 7 alkyl
  • R 9 and R 10 are independently H, CN, C 1 -C 4 alkyl, or mono- or di- hydroxy C 2 -C 4 alkyl;
  • R 11 is C 1 -C 7 alkyl, or C 1 -C 7 alkenyl
  • R 12 and R 13 are independently H, C 1 -C 7 alkyl, COOR 8 , CN, CH(OR 7 )COOR 8 , Br, CO-thienyl, COC 6 H 4 ,OH(p);
  • R 14 is NHR 7 or OR 8 ;
  • R 15 is COOR 8 , CH(OR 7 )CH 2 OR 16 or CH(OCOC 1 -C 4 alkyl)CH 2 OR 8 ;
  • R 16 is H, COCH 2 CH 2 CO 2 H, or COC 1 -C 7 alkyl
  • anti-tumor selenophenes of the above formula I there is provided anti-tumor selenophenes of the above formula I,
  • X and Y are independently selected from the group consisting of S, Se and NH;
  • R 1 , R 3 , and R 6 are H;
  • R 5 is selected from the group consisting of CHO or CH 2 OH; and cyclodextrin complexes of such compounds. These compounds have been demonstrated to exhibit cytotoxic selectivity against transformed human cells (See Table 1).
  • X and Y are independently selected from the group consisting of S, Se and NH;
  • R 2 , R 3 and R 6 are H;
  • R 5 is selected from the group consisting of CHO or CH 2 OH; and cyclodextrin complexes of such compounds.
  • R 1 , and R 2 are independently selected from the group consisting of; H, CHO, CH 2 OH and CH 2 NH 2 ;
  • X and Y are independently selected from the group consisting of Se, S, O, NCH 3 , and NH;
  • R 3 , R 4 , R 5 and R 6 are independently selected from the group consisting of H, CHO, CH 2 OH and CH 2 NH 2 ; cyclodextrin complexes of such compounds; and when R 2 or R 3 is CH 2 NH 2 , the pharmaceutically acceptable salt of the compound represented thereby; with the proviso, that when R 2 is R 1 is other than and when R 1 is R 2 is other than
  • novel intermediates of Formula II are also provided: wherein W is selected from the group consisting of N(CH 3 ) 2 and
  • X and Y are independently selected from the group consisting of Se, S, O, NCH 3 and NH.
  • One aspect of the present invention is a method of preparing the compounds of Formula I through an intermediate a compound of the formula: in accordance with the general methods of schemes 1 - 4 as described hereinbelow, wherein X and Y are independently selected from the group consisting of Se, S, O, NCH 3 and NH.
  • the selenophene compounds of this invention are readily formulated into pharmaceutical compositions, also within the scope of this invention, for use in the presently described method for treatment of patients having tumors.
  • the pharmaceutical composition comprises an anti-tumor effective amount of a selenophene compound of formula I: wherein R 1 and R 2 are independently selected from the group consisting of, H, CHO, CH 2 OH and CH 2 NH 2 ;
  • X and Y are independently selected from the group consisting of Se, S, O, and NR, wherein R is H or C 1 -C 7 alkyl;
  • R 7 is H, CO(CH 2 ) 2 CO 2 H, (CH 2 ) 2 OCH 3 , C 1 -C 4 alkyl or COC 1 -C 17 alkyl;
  • R 8 is H or C 1 -C 7 alkyl
  • R 9 and R 10 are independently H, CN, C 1 -C 4 alkyl, or mono- or di- hydroxy C 2 -C 4 alkyl,
  • R 11 is C 1 -C 7 alkyl, or C 1 -C 7 alkenyl
  • R 12 and R 13 are independently H, C 1 -C 7 alkyl, COOR 8 , CN, CH(OR7)COOR 8 , Br, CO-thienyl, COC 6 H 4 OH(p);
  • R 14 is NHR 7 or OR 8 ;
  • R 15 is COOR 8 , CH(OR 7 )CH 2 OR 16 or CH(OCOC 1 -C 4 alkyl)CH 2 OR 8 ;
  • R 16 is H, COCH 2 CH 2 CO 2 H, or COC 1 -C 7 alkyl
  • Another pharmaceutical composition within the scope of this invention comprises an anti-tumor effective amount of a selenophene compound of the formula I: wherein R 1 and R 2 are independently selected from the group consisting of; H, CHO, CH 2 OH and CH 2 NH 2 ;
  • X and Y are independently selected from the group consisting of Se, S, O, NCH 3 and NH;
  • R 3 , R 4 and R 6 are H
  • R 5 is selected from the group consisting of H, CHO, CH 2 OH and CH 2 NH 2 ;
  • the present compounds are readily prepared using art-recognized chemical-synthesis procedures as exemplified hereinbelow.
  • the cytotoxic activity of the present selenophene compounds have been measured utilizing two different assays or screens.
  • the first screen measures the cytotoxicity against a panel of sixty different human tumor cell lines.
  • This assay provides data regarding the general cytotoxicity of an individual compound.
  • this type of assay is useful in identifying compounds which have enhanced cytotoxic activity against slow growing tumors as compared to faster growing tumor cells such as leukemia tumor cell lines.
  • the identification of such compounds is critical since previously identified antitumor agents have low cytotoxic activity against slower growing tumors.
  • the specificity of a compound for a limited number of tumor cell lines also indicates that such a compound will likely be less cytotoxic to normal cells.
  • the specificity of a cytotoxic compound for tumor cell lines relative to normal cells is an important characteristic of an effective antitumor agent.
  • Antitumor cytotoxicity data generated from the National Cancer Institute human tumor cell panels can also be expressed in a graphic pattern (mean graph) to display differential cell growth inhibition (K. D. Paull, R. H. Shoemaker, L. Hodes, A. Monks, D. A. Scudiero, L. Rubinstein, J. Plowman and M. R. Boyd, J. Natl. Cancer Inst., 81, 1088, 1989.)
  • the mean graph the arithmetic mean of the logarithm of the GI 50 (50% growth inhibition), TGI (total growth inhibition) or LC 50 (50% lethal concentration) values is used as an anchor point.
  • Relative cytotoxicity is displayed by projecting bars to the right or left of the mean, depending on whether cell sensitivity to a test compound is more or less than average.
  • the length of a bar is indicative of differential cytotoxicity against a specific type of tumor cells or tumor panels.
  • the antitumor cytotoxicity of the selenophene compounds tested in those in vitro -assays was measured by a microculture assay using either a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) or sulforhodamine B (SRB) based assay.
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • SRB sulforhodamine B
  • GI 50 the concentration of drug at which cell numbers are reduced to 50% of control cell culture
  • This in vitro microculture assay has an advantage over in vivo assays in that results are obtained within a week as opposed to several months.
  • the MTT assay is based on the production of a dark blue formazan product by dehydrogenase in the mitochondria of live tumor cells after exposure to drug for 6 days [M. C. Alley, D. A. Scudiero, A. Monks, M. L. Hursey, M. J. Czerwinski, D. L. Fine, B. J. Abbott, J. G. Mayo, R. H. Shoemaker and M. R. Boyd, Cancer Res., 48, 589, 1988].
  • live cells are stained and can be measured at 570 nm.
  • the SRB assay is based on the binding of the anionic group to the basic amino acid residues of cellular proteins after exposure of tumor cells to drug for 2 days [P. Skehan, R. Storeng, D. Scudiero, A. Monks, J. McMahon, D. Vistica, J. T. Warren, H. Bohesch, S. Kenney and M. R. Boyd, J. Nat. Cancer Inst., 82, 1107, 1990.] Thus, the total protein can be measured at 564 nm. Antitumor cytotoxicity is reported as GI 50 , effect drug dose at which cell growth is retarded to 50% of control culture of tumor cells.
  • the active compounds are defined as those compounds having GI 50 values that are less than 10 ⁇ 4 M or 10 ⁇ g/ml.
  • Table 1 illustrates that selenophenes generally exhibit greater cytotoxicity for human renal carcinoma cells in comparison to the normal human cells.
  • the data of Table 1 demonstrates the selective cytotoxicity of various selenophene compounds against human renal carcinoma and ras-oncogene transformed human bronchial epithelial cells [in GI 50 ( ⁇ g/ml)].
  • the following abbreviations are used for the tested cell lines:
  • A-498 human renal carcinoma
  • TBE ras-transformed human bronchial epithelial cells
  • the present invention further provides pharmaceutical formulations comprising an effective amount of a selenophene compound for treating a patient having a tumor.
  • an effective amount of the selenophene compound is defined as the amount of the compound which, upon administration to a patient, inhibits growth of tumor cells, kills malignant cells, reduces the volume or size of the tumors or eliminates the tumor entirely in the treated patient.
  • the effective amount to be administered to a patient is typically based on body surface area, patient weight, and patient condition.
  • body surface area may be approximately determined from patient height and weight (see e.g., Scientific Tables, Geigy Pharmaceuticals, Ardley, N.Y., pages 537-538 (1970)).
  • An effective amount of the selenophene compounds in the present invention can range from about 5 mg/kg to about 100 mg/kg, more preferably from about 0.25 mg/kg to about 50 mg/kg, and most preferably about 0.1 to about 10 mg/kg.
  • Effective doses will also vary, as recognized by those skilled in the art, dependent on route of administration, excipient usage and the possibility of co-usage with other therapeutic treatments including other anti-tumor agents, and radiation therapy.
  • the pharmaceutical formulation may be administered via the parenteral route, including subcutaneously, intraperitoneally, intramuscularly and intravenously.
  • parenteral dosage forms include aqueous solutions of the active agent, in a isotonic saline, 5% glucose or other well-known pharmaceutically acceptable liquid carrier.
  • the selenophene compound is dissolved in a saline solution containing 5% of dimethyl sulfoxide and 10% Cremphor EL (Sigma Chemical Company).
  • solubilizing agents such as cyclodextrins, which form specific, more soluble complexes with the present selenophene compounds, or other solubilizing agents well-known to those familiar with the art, can be utilized as pharmaceutical excipients for delivery of the selenophene compounds.
  • the present compound can also be formulated into dosage forms for other routes of administration utilizing well-known methods.
  • the pharmaceutical compositions can be formulated, for example, in dosage forms for oral administration in a capsule, a gel seal or a tablet.
  • Capsules may comprise any well-known pharmaceutically acceptable material such as gelatin or cellulose derivatives.
  • Tablets may be formulated in accordance with conventional procedure by compressing mixtures of the active polythiophene and solid carriers, and lubricants well-known to those familiar with the art. Examples of solid carriers include starch, sugar, bentonite.
  • the compounds of the present invention can also be administered in a form of a hard shell tablet or capsule containing, for example, lactose or mannitol as a binder and conventional fillers and tableting agents.
  • Bis(tricyclohexyltin)selenide can be prepared from tricyclohexyltin chloride (Aldrich Chemical Co.) and sodium selenide (Alfa Chemical Co.).
  • the functional group can be introduced through selective ⁇ -formylation using lithium diisopropylamide (LDA) and dimethylformamide (DMF), which can then be sequentially converted into hydroxylmethyl and aminomethyl functional groups.
  • LDA lithium diisopropylamide
  • DMF dimethylformamide
  • a BCl 3 solution (1.0 M solution in hexanes, 580 ⁇ L) was added dropwise to an anhydrous toluene solution (5 mL) containing 1,4-diselenophene-1,4-diketone (100 mg) and bis(tricyclohexyltin)selenide (520 mg) under N 2 at room temperature.
  • the solution was refluxed for 30 min and cooled to room temperature.
  • the reaction solution was diluted with ethyl acetate (100 mL) and washed with H 2 O (2 ⁇ 100 mL).
  • the organic layer was separated, dried over MgSO 4 , filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5′, 2′′-terselenophene in 80% yield.
  • BCl 3 (1.0 M solution in hexanes, 580 ⁇ L) was added dropwise to an anhydrous toluene solution (5 mL) containing 2′,2′′-diselenophenyl-1,4-diketone (100 mg) and bis(tricyclohexyltin)sulfide (520 mg) under N 2 at room temperature.
  • the solution was refluxed for 30 min and cooled to room temperature.
  • the reaction solution was diluted with ethyl acetate (100 mL) and washed with H 2 O (2 ⁇ 100 mL).
  • the organic layer was separated, dried over MgSO 4 , filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5,2′′-diselenophenylthiophene in 85% yield.
  • BCl 3 (1.0 M solution in hexanes, 900 ⁇ L) was added dropwise to an anhydrous toluene solution (00 mL) containing 2′,2′′-difuranyl-1,4-diketone (100 mg) and bis(tricyclohexyltin)-selenide (750 mg) under N 2 at room temperature.
  • the solution was refluxed for 30 min and cooled to room temperature.
  • the reaction solution was diluted with ethyl acetate (100 mL) and washed with H 2 O (2 ⁇ 100 mL).
  • the organic layer was separated, dried over MgSO 4 , filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5,2′′-difuranylselenophene in 80% yield.
  • BCl 3 (1.0 M solution in hexanes, 1.6 mL) was added dropwise to an anhydrous toluene solution (5 mL) containing 2′,2′′-dithienyl-1,4-diketone (200 mg) and bis(tricyclohexyltin)selenide (1.3 g) under N 2 at room temperature. The solution was refluxed for 30 min and cooled to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H 2 O (3 ⁇ 100 mL). The organic layer was separated, dried over MgSO 4 , filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5,2′′-dithienylselenophene in 90% yield.
  • an alternative synthesis strategy (Scheme 4) can be utilized to prepare numerous “hybrid” ⁇ -Terselenophenes.
  • X and Y are selected from the group consisting of Se, O, S, N(CH 3 ) and NH
  • Z is selected from the group consisting of Se, S, N(CH 3 ) and NH.
  • Various functional groups can be introduced using the approaches outlined in the synthesis of ⁇ -terselenophenes (Scheme 2).
  • 1-(2′-Selenyl)-4-(2′′-furyl)butane-1,4-dione 7 (0.25 g, 0.9 mmol) and Lawesson's reagent (0.66 g, 1.63 mmol) were refluxed overnight in 7 mL toluene.
  • the toluene was evaporated and the crude product was purified using silica flash column with ether/hexane as eluent.
  • the product 8 was recrystallized from methanol; yield: 0.22 g (88%). mp. 76-77° C.
  • 1,4-Bis-(2′-selenyl)butane-1,4-dione 11 (34.4 mg, 0.1 mmol), sodium acetate (123 mg, 0.15 mmol) and methylamine chloride (101.3 mg, 0.15 mmol) were refluxed overnight in 3 mL ethanol. 10 ml water was then added, and the product was extracted with dichloromethane. The product 12 was recrystallized from ethanol; yield: 26 mg (76%).
  • a highly polar functional group can be incorporated into the selenophene compounds in order to improve their water solubility. Addition of a carbonylic functional group through an ester linkage (Scheme 5) resulted in a transient solubility. However, the benzylic ester may be readily hydrolyzed to regenerate the water insoluble starting material.
  • Scheme 1 can be modified to produce related selenophene analogs in accordance with Scheme 7:
  • An alternative approach of enhancing the water solubility of hydrophobic drugs comprises the preparation of their polar prodrug analogs.
  • Glutamate Conjugate As mentioned above, conversion of the hydroxyl group of 2-hydroxymethyl-5,2′:5′, 2′′-terselenophene into its amino analog can moderately improve its water solubility. However, the amino analog is less stable. The amino analog may be transformed into its ⁇ -glutamate prodrug (as shown in Scheme 9) to further enhance its water solubility and stability. This conjugate may also enhance target selectivity for the treatment of kidney cancer because of the higher ⁇ -glutamyl transpeptidase activity in kidney. A modified procedure can also be designed for the preparation of glutathione conjugate.
  • cyclodextrin derivatives can form stable inclusion complexes with 2-aminomethyl substituted thiophene compounds.
  • ⁇ -Cyclodextrin (cyclic heptaamylose) derivatives are commonly used for improving water solubility because of their low costs. It is anticipated that the selenophene compounds of the present invention can be complexed with ⁇ -hydroxypropyl, dimethyl and sulfated ⁇ -cyclodextrins to enhance the water solubility of those compounds.
  • PKC Protein Kinase C screening assay utilized in the following experiments is similar to standard PKC assays used by many investigators. Its primary features are that 1) the assay utilizes a 50:50 mixture of recombinant mouse PKC ⁇ and mouse PKC ⁇ 2 ; 2) employs histone as phosphate-accepting substrate; and 3) the PKC enzymatic activity is activated with phosphatidylserine, TPA and low concentration of calcium, so that both calcium and TPA are somewhat limiting for the extent of activation. In this manner the assay is sensitive to inhibitors of PKC activation. A more detailed description of the assay is provided in the following paragraphs.
  • the recombinant PKC formulation is a mixture (equal parts by activity) of mouse PKC ⁇ and mouse PKC ⁇ 2 .
  • the enzymes are expressed in Sf9 insect cells from recombinant baculovirus and partially purified on DEAE-cellulose and Sephacryl 200 gel filtration.
  • Sufficient PKC is added to each reaction to provide approximately 4 pmols phosphate transferred in 30 minute (per total reaction.) The reaction is linear over the time when 4 pmols of phosphate is transferred and the reaction remains linear well beyond this time frame.
  • the PKC screening assay is performed in 96 well polystyrene U bottom micro titer plates, in a total reaction volume of 50 ul. Solution manipulations are performed during the assay utilize a Rainin, motorized EDP-plus M8 eight-channel micropipettor.
  • Samples were typically assayed at three dilutions, however some highly active pure compounds were assayed at six dilutions.
  • Assay samples are dissolved in DMSO at a concentration of 10 mg/ml or less for samples suspected of being more potent. In some cases 50% DMSO:water, water, or methanol is substituted (if essential) for the solvent. At least 25 ⁇ l of the highest concentration sample to be assayed is transferred to a well in a 96 well U-bottom polystyrene assay plate.
  • Serial 5-fold or 10-fold dilutions are made using the EDP-plus M8 eight-channel pipettor in dilute mode and mixing by repipetting.
  • NSC # Structure IC 50 ( ⁇ g/ml) 675347 1 ⁇ 10 0 675346 5 ⁇ 10 ⁇ 1 675345 9 ⁇ 10 ⁇ 1 675344 5 ⁇ 10 1 675343 3 ⁇ 10 1 676628 1 ⁇ 10 0 676629 2 ⁇ 10 ⁇ 1 676630 9 ⁇ 10 ⁇ 1 676631 2 ⁇ 10 0 676632 5 ⁇ 10 1 676633 6 ⁇ 10 0 676634 8 ⁇ 10 ⁇ 1 676635 8 ⁇ 10 0 674973 7 ⁇ 10 0 675244 1 ⁇ 10 0 675245 1 ⁇ 10 0 675246 3 ⁇ 10 0 675247 1 ⁇ 10 1

Abstract

Novel selenophene compounds useful as anti-tumor agents are described. Preferred compounds include compounds of formula I:
Figure US20050203077A1-20050915-C00001

wherein R1 and R2 are independently selected from the group consisting of
Figure US20050203077A1-20050915-C00002

H, CHO, CH2OH and CH2NH2; and X and Y are independently selected from the group consisting of Se, S, O, NCH3, and NH. Pharmaceutical compositions and a method for treating patients having tumors utilizing the disclosed selenophene compounds are also described.

Description

    GOVERNMENT RIGHTS
  • This invention was made with United States Government support under Grant No. UO1 CA50743, awarded by the National Cancer Institute. The United States Government has certain rights in the invention.
  • FIELD OF THE INVENTION
  • The present invention relates to compositions and a method for treating a patient having a tumor. More specifically, the present invention relates to the treatment of such patients with an effective amount of a selenophene derivative.
  • BACKGROUND AND SUMMARY OF THE INVENTION
  • The control and cure of cancer represents one of our most challenging health problems. The treatment of cancer can be approached by several modes of therapy including surgery, radiation, chemotherapy or a combination of any of these treatments. Chemotherapy continues to be an indispensable therapy for inoperable or metastatic forms of the disease.
  • The selection of natural compounds, or the synthesis of new compounds having effective anticancer activity is complicated by the still limited knowledge of cancer cell biology and biochemistry. Therefore, development of new effective anti-tumor agents will remain heavily dependent on screening compounds to discover novel compounds having cytotoxic activity. Preferably, such compounds exhibit enhanced cytotoxicity against tumor cells relative to their cytotoxicity to normal cells.
  • The success of novel antitumor drug development programs is dependent on the initial identification of antitumor agents. Thus the discovery of antitumor agents requires the systematic screening of a large number of natural products and synthetic compounds.
  • The mouse L1210 leukemia cell line was initially the preferred model system used for screening natural compounds for antitumor activity. However, the P388 murine leukemia system was found to be more sensitive and predictive than L1210 leukemia system, and has been used as primary screen during the past decade. Systematic screening for compounds exhibiting toxicity to these two leukemia cell lines has resulted in the isolation of a large number of active natural products. However, the anticancer activities of these compounds were predominantly in leukemia, lymphoma and a few rare tumors. Low clinical efficacy, or the lack of clinical efficacy of known chemotherapeutics against slower growing solid tumors, is a serious concern.
  • It has been recognized that the use of a single antileukemia screening system could bias the end results and lead to the isolation of compounds only active in the treatment of fast growing tumors. In addition, the use of a single antileukemia screening system may not detect novel compounds with high specificities for particular cell lines. It is also likely that many novel compounds with possible anti-tumor activity have remained undetected by the less sensitive in vivo models due to the low concentrations at which many active natural products occur.
  • Considering the diversity of tumors in terms of cell type, morphology, growth rate and other cellular characteristics, the U.S. National Cancer Institute (NCI) has developed a “disease-oriented” approach to antitumor activity screening (M. R. Boyd, in “Principle of Practice of Oncology” J. T. Devita, S. Hellman, S. A. Rosenberg (Eds.) Vol. 3, PPO Update, No. 10, 1989). This in vitro prescreening system is based on the measurement of antitumor cytotoxicity against human tumor cell line panels consisting of approximately 60 cell lines of major human tumors (including leukemia and slower growing tumor cells such as lung, colon, breast, skin, kidney, etc.). The most important advantage of the new in vitro screening panels is the opportunity to identify compounds that are selectively more cytotoxic to cells of slowly growing solid tumors than to rapidly growing leukemia cells.
  • The cytotoxicity profile of the NCI human tumor cell panels displays the tumor specificity of a given compound, however the assay does not assess the toxicity of that compound to normal human cells. Accordingly a second bioassay is utilized to measure the selective cytotoxicity against certain types of tumor cells verses normal human cells.
  • The growth and differentiation of cells are regulated by signaling cascades induced by various mitogenic proteins (J. Kurdan and B. L. Taylor, “Signal Transduction,” Academic Press, New York, N.Y. 1994) that often are encoded by proto-oncogenes. The overexpression, amplification or mutation of the oncoprotein is critically involved in the initiation, progression and metastasis of malignant cells (R. A. Weinberg, “Oncogenes and the Molecular Origins of Cancer,” Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). Many oncoproteins alter normal cellular growth regulation by modulating the intracellular signaling pathways from the membrane to the nucleus. Therefore, cancer may be considered as a disease of cellular signal transduction, which presents a novel approach for anticancer therapy. One of the critical enzymes involved in the oncoprotein signal transduction is protein kinase C (U. Nishizuka, Nature, 308, 693, 1984 and Science, 233, 305, 1986). Thus, the determination of a compound's ability to inhibit protein kinase C activity has become a good prognostic for discovering novel anticancer agents (A. Basu, Pharmac Ther, 59, 257, 1993). Furthermore it is anticipated that the selenophene compounds will demonstrate selectivity for certain class members of protein kinases, including protein kinase C. Inhibition of a specific classes of protein kinases will allow the treatment of other diseases associated with defects in signaling transduction.
  • Selenophenes are selenium containing heterocyclic compounds that are analogs of naturally occurring thiophene, furan and pyrrole compounds. Selenophenes have been found to be effective antitumor agents, and exhibit enhanced cytotoxicity against slow growing tumor cells; selective cytotoxicity against human renal, ovarian tumor cells, and skin tumor cells; and exhibit inhibition of protein kinase C.
  • In accordance with this invention there is provided a method for the treatment of cancer which utilizes selenophene compounds of the formula I:
    Figure US20050203077A1-20050915-C00003
  • wherein R1 and R2 are independently selected from the group consisting of;
    Figure US20050203077A1-20050915-C00004

    H, CHO, CH2OH and CH2NH2;
  • X and Y are independently selected from the group consisting of Se, S, O, NCH3 and NH;
  • R3, R4, R5 and R6 are independently selected from the group consisting of H, CHO, CH2OH and CH2NH2; cyclodextrin complexes of such compounds; and when R3, R4, R5 or R6 is CH2NH2, the pharmaceutically acceptable salt of the compound represented thereby.
  • Further in accordance with this invention there are provided novel cytotoxic compounds of the above formula and chemotherapeutic pharmaceutical compositions containing said compounds in anti-tumor effective amounts.
  • Additional objects, features, and advantages of the invention will become apparent to those skilled in the art upon consideration of the following detailed description of preferred embodiments exemplifying the best mode of the invention as presently perceived.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to selenophene compounds, their pharmaceutical compositions and methods utilizing such compounds/compositions for treating patients having tumor. The selenophene compounds are effective antitumor agents against slow growing tumors, and generally have been found to exhibit high selective cytotoxicity for individual tumor cell lines.
  • The compounds of the present invention are selenophene compounds of the formula I:
    Figure US20050203077A1-20050915-C00005
  • wherein R1 and R2 are independently selected from the group consisting of;
    Figure US20050203077A1-20050915-C00006

    H, CHO, CH2OH and CH2NH2;
  • X and Y are independently selected from the group consisting of Se, S, O and NR;
  • R is H or C1-C7 alkyl;
  • R3, R4, R5 and R6 are independently selected from the group consisting of nitro, amino, alkoxy, cyano, chloro, bromo, iodo, C1-C7 alkyl or haloalkyl, C1-C7 alkenyl or haloalkenyl, C1-C4 alkanoyloxy methyl, CH2OR7, COR8, CH2NR9R10, CH(OR7)R11, CH=CR12R13, CH=NR14, CH2SC(NH)NH2 and C≡CR15 wherein
  • R7 is H, CO(CH2)2CO2H, (CH2)2OCH3, C1-C4 alkyl or COC1-C17 alkyl;
  • R8 is H or C1-C7 alkyl;
  • R9 and R10 are independently H, CN, C1-C4 alkyl, or mono- or di- hydroxy C2-C4 alkyl;
  • R11 is C1-C7 alkyl, or C1-C7 alkenyl;
  • R12 and R13 are independently H, C1-C7 alkyl, COOR8, CN, CH(OR7)COOR8, Br, CO-thienyl, COC6H4,OH(p);
  • R14 is NHR7 or OR8;
  • R15 is COOR8, CH(OR7)CH2OR16 or CH(OCOC1-C4 alkyl)CH2OR8;
  • R16 is H, COCH2CH2CO2H, or COC1-C7 alkyl;
    • cyclodextrin complexes of such compound and when R3, R4, R1 or R6 is CH2NR6R7, the pharmaceutically acceptable salt of the compound represented thereby.
  • In one preferred embodiment of this invention there is provided anti-tumor selenophenes of the above formula I,
    • wherein R2 is
      Figure US20050203077A1-20050915-C00007
  • X and Y are independently selected from the group consisting of S, Se and NH;
  • R1, R3, and R6 are H; and
  • R5 is selected from the group consisting of CHO or CH2OH; and cyclodextrin complexes of such compounds. These compounds have been demonstrated to exhibit cytotoxic selectivity against transformed human cells (See Table 1).
  • In another preferred embodiment of this invention there is provided anti-tumor selenophenes of the above formula I wherein R1 is
    Figure US20050203077A1-20050915-C00008
  • X and Y are independently selected from the group consisting of S, Se and NH;
  • R2, R3 and R6 are H;
  • R5 is selected from the group consisting of CHO or CH2OH; and cyclodextrin complexes of such compounds.
  • Other preferred compounds in accordance with this invention are selenophenes of formula I:
    Figure US20050203077A1-20050915-C00009

    wherein R1, and R2 are independently selected from the group consisting of;
    Figure US20050203077A1-20050915-C00010

    H, CHO, CH2OH and CH2NH2;
  • X and Y are independently selected from the group consisting of Se, S, O, NCH3, and NH;
  • R3, R4, R5 and R6 are independently selected from the group consisting of H, CHO, CH2OH and CH2NH2; cyclodextrin complexes of such compounds; and when R2 or R3 is CH2NH2, the pharmaceutically acceptable salt of the compound represented thereby; with the proviso, that when R2 is
    Figure US20050203077A1-20050915-C00011

    R1 is other than
    Figure US20050203077A1-20050915-C00012

    and when R1 is
    Figure US20050203077A1-20050915-C00013

    R2 is other than
    Figure US20050203077A1-20050915-C00014
  • In accordance with another embodiment of the present invention novel intermediates of Formula II are also provided:
    Figure US20050203077A1-20050915-C00015

    wherein W is selected from the group consisting of N(CH3)2 and
    Figure US20050203077A1-20050915-C00016
  • and X and Y are independently selected from the group consisting of Se, S, O, NCH3 and NH.
  • One aspect of the present invention is a method of preparing the compounds of Formula I through an intermediate a compound of the formula:
    Figure US20050203077A1-20050915-C00017

    in accordance with the general methods of schemes 1-4 as described hereinbelow, wherein X and Y are independently selected from the group consisting of Se, S, O, NCH3 and NH.
  • The selenophene compounds of this invention are readily formulated into pharmaceutical compositions, also within the scope of this invention, for use in the presently described method for treatment of patients having tumors. In one preferred embodiment of this invention, the pharmaceutical composition comprises an anti-tumor effective amount of a selenophene compound of formula I:
    Figure US20050203077A1-20050915-C00018

    wherein R1 and R2 are independently selected from the group consisting of,
    Figure US20050203077A1-20050915-C00019

    H, CHO, CH2OH and CH2NH2;
  • X and Y are independently selected from the group consisting of Se, S, O, and NR, wherein R is H or C1-C7 alkyl;
  • R3, R4, R1 and R6 are independently selected from the group consisting of nitro, amino, alkoxy, cyano, chloro, bromo, iodo, C1-C7 alkyl or haloalkyl, C1-C7 alkenyl or haloalkenyl, C1-C4 alkanoyloxy methyl, CH2OR7, COR8, CH2NR9R10, CH(OR7)R11, CH=CR12R13, CH=NR14, CH2SC(NH)NH2 and C=CR15 wherein
  • R7 is H, CO(CH2)2CO2H, (CH2)2OCH3, C1-C4 alkyl or COC1-C17 alkyl;
  • R8 is H or C1-C7 alkyl;
  • R9 and R10 are independently H, CN, C1-C4 alkyl, or mono- or di- hydroxy C2-C4 alkyl,
  • R11 is C 1-C7 alkyl, or C1-C7 alkenyl;
  • R12 and R13 are independently H, C1-C7 alkyl, COOR8, CN, CH(OR7)COOR8, Br, CO-thienyl, COC6H4OH(p);
  • R14 is NHR7 or OR8;
  • R15 is COOR8, CH(OR7)CH2OR16 or CH(OCOC1-C4 alkyl)CH2OR8;
  • R16 is H, COCH2CH2CO2H, or COC1-C7 alkyl;
    • cyclodextrin complexes of such compound and when R3, R4, R5 or R6 is CH2NR6R7, the pharmaceutically acceptable salt of the compound represented thereby, and a pharmaceutically acceptable carrier.
  • Another pharmaceutical composition within the scope of this invention comprises an anti-tumor effective amount of a selenophene compound of the formula I:
    Figure US20050203077A1-20050915-C00020

    wherein R1 and R2 are independently selected from the group consisting of;
    Figure US20050203077A1-20050915-C00021

    H, CHO, CH2OH and CH2NH2;
  • X and Y are independently selected from the group consisting of Se, S, O, NCH3 and NH;
  • R3, R4 and R6 are H;
  • R5 is selected from the group consisting of H, CHO, CH2OH and CH2NH2;
    • cyclodextrin complexes of such compounds; and when R3, R4, R5 or R6 is CH2NH2, the pharmaceutically acceptable salt of the compound represented thereby; with the proviso, that when R2 is
      Figure US20050203077A1-20050915-C00022

      R1 is other than
      Figure US20050203077A1-20050915-C00023

      and when R1 is
      Figure US20050203077A1-20050915-C00024

      R2is other than
      Figure US20050203077A1-20050915-C00025

      and a pharmaceutically acceptable carrier.
  • The present compounds are readily prepared using art-recognized chemical-synthesis procedures as exemplified hereinbelow.
  • The cytotoxic activity of the present selenophene compounds have been measured utilizing two different assays or screens. The first screen measures the cytotoxicity against a panel of sixty different human tumor cell lines. This assay provides data regarding the general cytotoxicity of an individual compound. In particular this type of assay is useful in identifying compounds which have enhanced cytotoxic activity against slow growing tumors as compared to faster growing tumor cells such as leukemia tumor cell lines. The identification of such compounds is critical since previously identified antitumor agents have low cytotoxic activity against slower growing tumors. The specificity of a compound for a limited number of tumor cell lines also indicates that such a compound will likely be less cytotoxic to normal cells. The specificity of a cytotoxic compound for tumor cell lines relative to normal cells is an important characteristic of an effective antitumor agent.
  • Antitumor cytotoxicity data generated from the National Cancer Institute human tumor cell panels can also be expressed in a graphic pattern (mean graph) to display differential cell growth inhibition (K. D. Paull, R. H. Shoemaker, L. Hodes, A. Monks, D. A. Scudiero, L. Rubinstein, J. Plowman and M. R. Boyd, J. Natl. Cancer Inst., 81, 1088, 1989.) In the mean graph, the arithmetic mean of the logarithm of the GI50 (50% growth inhibition), TGI (total growth inhibition) or LC50 (50% lethal concentration) values is used as an anchor point. Relative cytotoxicity is displayed by projecting bars to the right or left of the mean, depending on whether cell sensitivity to a test compound is more or less than average. The length of a bar is indicative of differential cytotoxicity against a specific type of tumor cells or tumor panels.
  • In a second assay, the cytotoxic selectivity is assessed by comparing compound cytotoxicity against human renal carcinoma cells (A-498), ras-transformed human bronchial epithelial cells (TBE) and normal human renal cells (RPTEC). IC50 values were compared between treated TBE cells and RPTEC cells and the selective cytotoxicity index (SCI) was determined [SCI=GI50(RPTEC)/GI50 (A-498)].
  • The antitumor cytotoxicity of the selenophene compounds tested in those in vitro -assays was measured by a microculture assay using either a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) or sulforhodamine B (SRB) based assay. [M. R. Boyd in “Principles and Practices of Oncology,” V. T. DeVita, Jr.]. The experiments were conducted at Purdue University in 96-well microtiter plates and the cytotoxic effects of the selenophene compounds on those cells were measured by cell count using a Coulter Z. F. counter (Hialeah, Fla.). The results are expressed as GI50, the concentration of drug at which cell numbers are reduced to 50% of control cell culture [T. C. K. Chan, C. J. Chang, N. M. Koonchanok and R. L. Geahlen, Biochem. Biophys. Res. Commun., 193, 1152, (1993); S. Hellman and S. A. Rosenberg (Eds.), Vol. 3, PPO Updates, Number 10, (1989).]
  • This in vitro microculture assay has an advantage over in vivo assays in that results are obtained within a week as opposed to several months. The MTT assay is based on the production of a dark blue formazan product by dehydrogenase in the mitochondria of live tumor cells after exposure to drug for 6 days [M. C. Alley, D. A. Scudiero, A. Monks, M. L. Hursey, M. J. Czerwinski, D. L. Fine, B. J. Abbott, J. G. Mayo, R. H. Shoemaker and M. R. Boyd, Cancer Res., 48, 589, 1988]. Thus, only live cells are stained and can be measured at 570 nm. The SRB assay is based on the binding of the anionic group to the basic amino acid residues of cellular proteins after exposure of tumor cells to drug for 2 days [P. Skehan, R. Storeng, D. Scudiero, A. Monks, J. McMahon, D. Vistica, J. T. Warren, H. Bohesch, S. Kenney and M. R. Boyd, J. Nat. Cancer Inst., 82, 1107, 1990.] Thus, the total protein can be measured at 564 nm. Antitumor cytotoxicity is reported as GI50, effect drug dose at which cell growth is retarded to 50% of control culture of tumor cells. The active compounds are defined as those compounds having GI50 values that are less than 10−4 M or 10 μg/ml.
  • The data presented in Table 1 illustrates that selenophenes generally exhibit greater cytotoxicity for human renal carcinoma cells in comparison to the normal human cells. The data of Table 1 demonstrates the selective cytotoxicity of various selenophene compounds against human renal carcinoma and ras-oncogene transformed human bronchial epithelial cells [in GI50(μg/ml)]. The following abbreviations are used for the tested cell lines:
  • RPTEC: normal human renal cells
  • A-498: human renal carcinoma
  • TBE: ras-transformed human bronchial epithelial cells
  • SCI: selectively cytotoxicity index GI50 (RPTEC)/GI50 (A-498)
    TABLE 1
    NSC GI50 (μg/ml)
    Number RPTEC A-498 TBE SCI Structure
    674973 4 × 100 3 × 100 4 × 100 1
    Figure US20050203077A1-20050915-C00026
    675246 1 × 10−13 × 10−2 3 × 10−63 × 10−6 3 × 10−32 × 10−3 >1000 >1000
    Figure US20050203077A1-20050915-C00027
    675247 2 × 10−18 × 100 7 × 10−52 × 10−6 3 × 1012 × 101 >1000 >1000
    Figure US20050203077A1-20050915-C00028
    676628 4 × 102 8 × 101 1 × 102 5
    Figure US20050203077A1-20050915-C00029
    676632 2 × 10−33 × 10−4 3 × 10−72 × 10−7 <10−32 × 10−4 >1000 >1000
    Figure US20050203077A1-20050915-C00030
    675347 2 × 101 3 × 101 1 × 101 <1
    Figure US20050203077A1-20050915-C00031
    675344 <10−21 × 10−4 3 × 10−76 × 10−8 <10−27 × 10−6 >1000 >1000
    Figure US20050203077A1-20050915-C00032
    676633 2 × 101 1 × 102 1 × 101 <1
    Figure US20050203077A1-20050915-C00033
    676634 1 × 1014 × 10−1 6 × 10−43 × 10−3 3 × 10−46 × 10−3 >1000 >100
    Figure US20050203077A1-20050915-C00034
    676635 2 × 100 <10−3 2 × 101 >1000
    Figure US20050203077A1-20050915-C00035
    123127 5 × 10−2 5 × 10−2 3 × 10−2 1 Adriamycin
  • The present invention further provides pharmaceutical formulations comprising an effective amount of a selenophene compound for treating a patient having a tumor. As used herein, an effective amount of the selenophene compound is defined as the amount of the compound which, upon administration to a patient, inhibits growth of tumor cells, kills malignant cells, reduces the volume or size of the tumors or eliminates the tumor entirely in the treated patient.
  • The effective amount to be administered to a patient is typically based on body surface area, patient weight, and patient condition. The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described by Freireich, E. J., et al., Cancer Chemother. Rep., 50 (4):219 (1966). Body surface area may be approximately determined from patient height and weight (see e.g., Scientific Tables, Geigy Pharmaceuticals, Ardley, N.Y., pages 537-538 (1970)). An effective amount of the selenophene compounds in the present invention can range from about 5 mg/kg to about 100 mg/kg, more preferably from about 0.25 mg/kg to about 50 mg/kg, and most preferably about 0.1 to about 10 mg/kg.
  • Effective doses will also vary, as recognized by those skilled in the art, dependent on route of administration, excipient usage and the possibility of co-usage with other therapeutic treatments including other anti-tumor agents, and radiation therapy.
  • The pharmaceutical formulation may be administered via the parenteral route, including subcutaneously, intraperitoneally, intramuscularly and intravenously. Examples of parenteral dosage forms include aqueous solutions of the active agent, in a isotonic saline, 5% glucose or other well-known pharmaceutically acceptable liquid carrier. In one preferred aspect of the present embodiment, the selenophene compound is dissolved in a saline solution containing 5% of dimethyl sulfoxide and 10% Cremphor EL (Sigma Chemical Company). Additional solubilizing agents such as cyclodextrins, which form specific, more soluble complexes with the present selenophene compounds, or other solubilizing agents well-known to those familiar with the art, can be utilized as pharmaceutical excipients for delivery of the selenophene compounds.
  • The present compound can also be formulated into dosage forms for other routes of administration utilizing well-known methods. The pharmaceutical compositions can be formulated, for example, in dosage forms for oral administration in a capsule, a gel seal or a tablet. Capsules may comprise any well-known pharmaceutically acceptable material such as gelatin or cellulose derivatives. Tablets may be formulated in accordance with conventional procedure by compressing mixtures of the active polythiophene and solid carriers, and lubricants well-known to those familiar with the art. Examples of solid carriers include starch, sugar, bentonite. The compounds of the present invention can also be administered in a form of a hard shell tablet or capsule containing, for example, lactose or mannitol as a binder and conventional fillers and tableting agents.
  • The following examples are provided to illustrate various embodiments of Applicants' invention, and are not intended to in any way limit the scope of the invention as set forth in this specification and appended claims.
  • EXAMPLE 1
  • Synthesis of ∝-Terselenophenes
  • A two-step total synthesis of ∝-terselenophene from selenophene (Aldrich Chemical Co.) has been developed (See Scheme 1).
    Figure US20050203077A1-20050915-C00036
  • Bis(tricyclohexyltin)selenide can be prepared from tricyclohexyltin chloride (Aldrich Chemical Co.) and sodium selenide (Alfa Chemical Co.). The functional group can be introduced through selective ∝-formylation using lithium diisopropylamide (LDA) and dimethylformamide (DMF), which can then be sequentially converted into hydroxylmethyl and aminomethyl functional groups. These functional groups can provide required starting points for further chemical modifications, see Scheme 2 as follows:
    Figure US20050203077A1-20050915-C00037
  • EXAMPLE 2
  • Synthesis of Hybrid ∝-Terselenophenes
  • The synthetic strategy designed for the preparation of ∝-terselenophene can be readily modified for the synthesis of numerous “hybrid” ∝-selenophenes containing other five-membered heterocycles (See Scheme 3).
    Figure US20050203077A1-20050915-C00038

    Wherein X, and Y are selected from the group consisting of Se, O, S, NCH3 and NH2, and Z is selected from the group consisting of Se, S, NCH3 and NH2. Various functional groups can be introduced using the approaches outlined in the synthesis of ∝-terselenophenes (Scheme 2).
  • EXAMPLE 3
  • Preparation of 1,4-Diselenophene-1,4-Diketone.
  • A CH2Cl2 solution containing selenophene (5 g) and succinyl chloride (2 g) was added dropwise to an anhydrous CH2Cl2 solution (60 mL) containing AlCl3 (5 g) under N2 at 0° C. The reaction mixture was stirred at 0° C. for 1 h, slowly warmed to room temperature, and stirred for 4 h at room temperature. The reaction mixture was poured into a beaker containing ice. Ethyl acetate (200 mL) was added and the organic layer was separated out using a separatory funnel. The aqueous layer was back washed with ethyl acetate (2×100 mL). The combined organic layer was washed with H2O (2×300 mL). The organic layer was collected, dried over MgSO4, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (10:1) hexanes/ethyl acetate to afford the product in 25% yield.
  • EXAMPLE 4
  • Preparation of 2,2′:5′,2″-Terselenophene.
  • A BCl3 solution (1.0 M solution in hexanes, 580 μL) was added dropwise to an anhydrous toluene solution (5 mL) containing 1,4-diselenophene-1,4-diketone (100 mg) and bis(tricyclohexyltin)selenide (520 mg) under N2 at room temperature. The solution was refluxed for 30 min and cooled to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (2×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5′, 2″-terselenophene in 80% yield.
  • EXAMPLE 5
  • Preparation of 2-Formyl-5,2′:5′,2″-Terselenophene.
  • LDA (1.0 M solution in THF, 310 μL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5′,2″-terselenophene (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (1 mL) was added, stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (4×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using CH2Cl2 to afford 2-formyl-5,2′:5′,2″-terselenophene in 75% yield.
  • EXAMPLE 6
  • Preparation of 2,5″-Diformyl-5,2′:5′,2″-Terselenophene.
  • LDA (1.0 M solution in THF, 1.0 mL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5′,2″-terselenophene (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (2 mL) was added, stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (4×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (5:1) CH2Cl2/ethyl acetate to afford 2,5″-diformyl-5,2′:5′,2″-terselenophene in 75% yield.
  • EXAMPLE 7
  • Preparation of 2-Hydroxymethyl-5,2′:5′,2″-Terselenophene.
  • NaBH4 (10 mg) was added to a THF solution (2 mL) 2-formyl-5,2′:5′,2″-terselenophene (15 mg) and stirred at room temperature for 2 h. The reaction solution was diluted with ethyl acetate (50 mL), washed with 2N HCl (5 mL), and then washed with H2O (3×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford 2-hydroxymethyl-5,2′:5′, 2″-terselenophene in 98% yield.
  • EXAMPLE 8
  • Preparation of 2,5″-Dihydroxymethyl-5,2′:5′,2″-Terselenophene.
  • NaBH4 (10 mg) was added to a THF solution (2 mL) containing 2,5″-diformyl-5,2′:5′, 2″-terselenophene (15 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (50 mL), washed with 2N HCl (5 mL), and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (1:1) hexanes/ethyl acetate to afford 2,5″-dihydroxymethyl-5,2′:5′, 2″-terselenophene in 98% yield.
  • EXAMPLE 9
  • Preparation of 2,4-Diselenophenylfuran.
  • d- 10-camphorsulfonic acid (2 g) was added to an ethanolic solution (15 mL) containing 2′,2″-diselenophene-1,4-diketone (100 mg) and refluxed for 2 days. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (10:1) hexanes/ethyl acetate to afford 2,2′:5,2″-diselenophenylfuran in 90% yield.
  • EXAMPLE 10
  • Preparation of 5′-Formyl-2,2′:5,2″-Diselenophenylfuran.
  • LDA (1 molar solution in THF, 350 μL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-diselenophenylfuran (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (excess) was added, stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (4:1) hexanes/ethyl acetate to afford 5′-formyl-2,2′:5,2″-diselenophenylfuran.
  • EXAMPLE 11
  • Preparation of 5′,5″-Diformyl-2,2′:5,2″-Diselenophenylfuran.
  • LDA (1 molar solution in THF, 1 mL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-diselenophenylfuran (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (excess) was added, stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (4:1) hexanes/ethyl acetate to afford 5,5″-diformyl-2,2′:5,2″-diselenophenylfuran.
  • EXAMPLE 12
  • Preparation of 5′-Hydroxymethyl-2,2′:5,2″-Diselenophenylfuran.
  • NaBH4 (excess) was added to a THF solution (2 mL) containing 5′-formyl-2,2′:5,2″-diselenophenylfuran (15 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford 5′-hydroxymethyl-2,2′:5,2″-diselenophenylfuran.
  • EXAMPLE 13
  • Preparation of 5′,5″-dihydroxymethyl-2,2′:5,2″-diselenophenylfuran.
  • NaBH4 (excess) was added to a THF solution (2 mL) containing 5′,5″-diformyl-2,2′:5,2″-diselenophenylfuran (15 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (1:1) hexanes/ethyl acetate to afford 5′,5″-dihydroxymethyl-2,2′:5,2″-diselenophenylfuran.
  • EXAMPLE 14
  • Preparation of 2,2′:5,2″-diselenophenylthiophene.
  • BCl3 (1.0 M solution in hexanes, 580 μL) was added dropwise to an anhydrous toluene solution (5 mL) containing 2′,2″-diselenophenyl-1,4-diketone (100 mg) and bis(tricyclohexyltin)sulfide (520 mg) under N2 at room temperature. The solution was refluxed for 30 min and cooled to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (2×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5,2″-diselenophenylthiophene in 85% yield.
  • EXAMPLE 15
  • Preparation of 5′-formyl-2,2′:5,2″-diselenophenylthiophene.
  • LDA (1.0 M solution in THF, 350 μL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-diselenophenylthiophene under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (1 mL) was added, stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using CH2Cl2 to afford 5′-formyl-2,2′:5,2″-diselenophenylthiophene in 80% yield.
  • EXAMPLE 16
  • Preparation of 5′,5″-Diformyl-2,2′:5,2″-Diselenophenylthiophene.
  • LDA (1.0 M solution in THF, 1 mL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-diselenophenylthiophene under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (2 mL) was added, the solution was stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (5:1) CH2Cl2/ethyl acetate to afford 5′,5″-diformyl-2,2′:5,2″-diselenophenylthiophene in 80% yield.
  • EXAMPLE 17
  • Preparation of 5′-Hydroxymethyl-2,2′:5,2″-Diselenophenylthiophene.
  • NaBH4 (10 mg) was added to a THF solution (3 mL) containing 5′-formyl-2,2′:5,2″-diselenophenylthiophene (20 mg) and stirred at room temperature for 2 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (5×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) CH2Cl2/ethyl acetate to afford 5′-hydroxymethyl-2,2′:5,2″-diselenophenylthiophene in 98% yield.
  • EXAMPLE 18
  • Preparation of 5,5″-Dihydroxymethyl-2,2′:5,2″-Diselenophenylthiophene.
  • NaBH4 (10 mg) was added to a THF solution (3 mL) containing 5′,5″-diformyl-2,2′:5,2″-diselenophenylthiophene (20 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (5×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (1:1) hexanes/ethyl acetate to afford 5′,5″-dihydroxymethyl-2,2′:5,2″-diselenophenylthiophene in 98% yield.
  • EXAMPLE 19
  • Preparation of 2,2′:5,2″-Diselenophenylpyrrole.
  • An ethanolic solution (20 mL) containing 2′,2″-diselenophenyl-1,4-diketone (200 mg) and ammonium acetate (500 mg) and sodium acetate (200 mg) was refluxed overnight. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (10:1) hexanes/ethyl acetate to afford 2,2′:5,2″-diselenophenylpyrrole in 94% yield.
  • EXAMPLE 20
  • Preparation of 5′-Formyl-2,2′:5,2″-Diselenophenylpyrrole.
  • LDA (1.0 M solution in THF, 760 μL) was added to an anhydrous THF solution (5 mL) containing 2,2′:5,2 ″-diselenophenylpyrrole (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (1.5 mL) was added, the solution was slowly warmed to room temperature, and stirred at room temperature for 2 h. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (3:1) hexanes/ethyl acetate to afford 5′-formyl-2,2′:5,2″-diselenophenylpyrrole in 75% yield.
  • EXAMPLE 21
  • Preparation of 5′-Hydroxymethyl-2,2′:5,2″-Diselenophenylpyrrole.
  • NaBH4 (20 mg) was added to a THF solution (2 mL) containing 5′-formyl-2,2′:5,2″-diselenophenylpyrrole (20 mg) and stirred at room temperature for 2 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (3×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford 5′-hydroxymethyl-2,2′:5,2″-diselenophenylpyrrole in 98% yield.
  • EXAMPLE 22
  • Preparation of 2′,2″-Difuranyl-1,4-Diketone.
  • A CH2Cl2 solution containing furan (10 mL) and succinyl chloride (2 g) was added dropwise to an anhydrous CH2Cl2 solution (100 mL) containing AlCl3 (10 g) under N2 at 0° C. The reaction mixture was stirred at 0° C. for 2 h, slowly warmed to room temperature, and stirred for 4 h. The reaction mixture was poured into a beaker containing ice. Ethyl acetate (300 mL) was added and the organic layer was separated out using a separatory funnel. The aqueous layer was back washed with ethyl acetate (2×100 mL). The combined organic layer was washed with H2O (2×300 mL). The organic layer was collected, dried over MgSO4, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (3:1) hexanes/ethyl acetate to afford 2′,2″-difuranyl-1,4-diketone in 25% yield.
  • EXAMPLE 23
  • Preparation of 2,2′:5,2″-Difuranylselenophene.
  • BCl3 (1.0 M solution in hexanes, 900 μL) was added dropwise to an anhydrous toluene solution (00 mL) containing 2′,2″-difuranyl-1,4-diketone (100 mg) and bis(tricyclohexyltin)-selenide (750 mg) under N2 at room temperature. The solution was refluxed for 30 min and cooled to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (2×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5,2″-difuranylselenophene in 80% yield.
  • EXAMPLE 24
  • Preparation of 5′-Formyl-2,2′:5,2″-Difuranylselenophene.
  • LDA (1.0 M solution in THF, 420 μL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-difuranylselenophene (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (1 mL) was added, the solution was stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (3:1) hexanes/ethyl acetate to afford 5′-formyl-2,2′:5,2″-difuranylselenophene in 75% yield.
  • EXAMPLE 25
  • Preparation of 5′,5″-Diformyl-2,2′:5,2″-Difuranylselenophene.
  • LDA (1.0 M solution in THF, 1 mL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-difuranylselenophene (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, added anhydrous DMF (2 mL), stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford 5′,5″-diformyl-2,2′:5,2″-difuranylselenophene in 80% yield.
  • EXAMPLE 26
  • Preparation of 5′-Hydroxymethyl-2,2′:5,2″-Difuranylselenophene.
  • NaBH4 (10 mg) was added to a THF solution (2 mL) containing 5′-formyl-2,2′:5,2″-difuranylselenophene (20 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (3×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford 5′-hydroxymethyl-2,2′:5,2″-difuranylselenophene in 98% yield.
  • EXAMPLE 27
  • Preparation of 5′,5″-Dihydroxymethyl-2,2′:5,2″-Difuranylselenophene.
  • NaBH4 (10 mg) was added to a THF solution (2 mL) containing 5′,5″-diformyl-2,2′:5,2″-difuranylselenophene (20 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (3×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (1:1) hexanes/ethyl acetate to afford 5′,5″-dihydroxymethyl-2,2′:5,2″-difuranylselenophene in 98% yield.
  • EXAMPLE 28
  • Preparation of 2′,2″-Dithienyl-1,4-Diketone.
  • A CH2Cl2 solution containing thiophene (10 mL) and succinyl chloride (2 g) was added dropwisely to an anhydrous CH2Cl2 solution (100 mL) containing AlCl3 (10 g) under N2 at 0° C. The reaction mixture was stirred at 0° C. for 2 h, slowly warmed to room temperature, and stirred for 4 h. The reaction mixture was poured into a beaker containing ice. Ethyl acetate (300 mL) was added and the organic layer was separated out using a separatory funnel. The aqueous layer was back washed with ethyl acetate (2×100 mL). The combined organic layer was washed with H2O (2×300 mL). The organic layer was collected, dried over MgSO4, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (3:1) hexanes/ethyl acetate to afford 2′,2″-dithienyl-1,4-diketone in 25% yield.
  • EXAMPLE 29
  • Preparation of 2,2′:5,2″-Dithienylselenophene.
  • BCl3 (1.0 M solution in hexanes, 1.6 mL) was added dropwise to an anhydrous toluene solution (5 mL) containing 2′,2″-dithienyl-1,4-diketone (200 mg) and bis(tricyclohexyltin)selenide (1.3 g) under N2 at room temperature. The solution was refluxed for 30 min and cooled to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using hexanes to afford 2,2′:5,2″-dithienylselenophene in 90% yield.
  • EXAMPLE 30
  • Preparation of 5′-Formyl-2,2′:5,2″-Dithienylselenophene.
  • LDA (1.0 M solution in THF, 380 μL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-dithienylselenophene (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (1 mL) was added, the solution stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (3:1) hexanes/ethyl acetate to afford 5′-formyl-2,2′:5,2″-dithienylselenophene in 75% yield.
  • EXAMPLE 31
  • Preparation of 5′,5″-Diformyl-2,2′:5,2″-Dithienylselenophene.
  • LDA (1.0 M solution in THF, 1.0 mL) was added to an anhydrous THF solution (4 mL) containing 2,2′:5,2″-dithienylselenophene (100 mg) under N2 at −78° C. The solution was stirred at −78° C. for 3 h, anhydrous DMF (2 mL) was added, the solution stirred at −78° C. for 1 h, and slowly warmed to room temperature. The reaction solution was diluted with ethyl acetate (100 mL) and washed with H2O (3×100 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford 5′,5″-diformyl-2,2′:5,2″-dithienylselenophene in 85% yield.
  • EXAMPLE 32
  • Preparation of 5′-Hydroxymethyl-2,2′:5,2″-Dithienylselenophene.
  • NaBH4 (10 mg) was added to a THF solution (2 mL) containing 5′-formyl-2,2′:5,2″-dithienylselenophene, (20 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (3×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (2:1) hexanes/ethyl acetate to afford. 5′-hydroxymethyl-2,2′:5,2″-dithienylselenophene in 98% yield.
  • EXAMPLE 33
  • Preparation of 5′,5″-Dihydroxymethyl-2,2′:5,2″-Dithienylselenophene.
  • NaBH4 (10 mg) was added to a THF solution (2 mL) containing 5′,5″-diformyl-2,2′:5,2″-dithienylselenophene (20 mg) and stirred at room temperature for 5 h. The reaction solution was diluted with ethyl acetate (50 mL) and washed with H2O (3×50 mL). The organic layer was separated, dried over MgSO4, filtered, and the solvent was removed under vacuum. The residue was chromatographed over silica gel using (1:1) hexanes/ethyl acetate to afford 5′,5″-dihydroxymethyl-2,2′:5,2″-dithienylselenophene in 98% yield.
  • EXAMPLE 34
  • Alternative Method of Synthesizing Hybrid α-Terselenophenes
  • In addition to the method of synthesis described in Example 2, an alternative synthesis strategy (Scheme 4) can be utilized to prepare numerous “hybrid” α-Terselenophenes.
    Figure US20050203077A1-20050915-C00039

    Wherein X and Y are selected from the group consisting of Se, O, S, N(CH3) and NH, and Z is selected from the group consisting of Se, S, N(CH3) and NH. Various functional groups can be introduced using the approaches outlined in the synthesis of ∝-terselenophenes (Scheme 2).
  • EXAMPLE 35
  • Preparation of 3-Dimethylamino-1-(2′-Selenyl)Propanone
    Figure US20050203077A1-20050915-C00040
  • Synthesis of 2-acetylselenophene(1): A solution of selenophene (2.0 g, 15 mmol), acetic anhydride (2.34 g, 23 mmol) and tin (IV) chloride (0.06-g, 0.23 mmol) in 30 mL of dry methene chloride was stirred under Argon for two days until TLC plate showed completion of the reaction.
  • A mixture of crude 2-acetylselenophene (2.6 g, 15 mmol), paraformaldehyde (0.59 g, 19.6 mmol), dimethylamine hydrochloride (1.6 g, 19.5 mmol) and 0. 15 mL of HCl was refluxed for 16 h in 7 mL of ethanol. The reaction mixture was cooled and the precipitate was filtered, washed with ether and dried; yield 2.77 g (69.3%). This Mannich base hydrochloride (2 g) was basified using ammonium hydroxide. The solution was extracted (3×15 mL) with diethyl ether. The organic layer was washed with water and dried with sodium sulfate. Evaporation of ether gave 1.6 g of product 2.
    Figure US20050203077A1-20050915-C00041

    1H NMR (CDCl3) δ 8.37 (dd, 1H,H-5 of selenophene ring, J=5.52, 0.99),7.95 (dd, 1H, H-3 of selenophene ring, J=0.99, 3.99), 7.40 (dd, 1H, H4 of selenophene ring, J=5.52, 3.99), 3.10 (t, 2H, CO-CH2, J=7.6), 2.76 (t, 2H, CH2-NMe2, J=7.6), 2.29 (s, 6H, NMe2).
  • EXAMPLE 36
  • Preparation of 1-(2′-Thienyl)-4-(2″-Selenyl)butane-1,4-Dione(3).
    Figure US20050203077A1-20050915-C00042
  • A solution of 2-formylthiophene (1.05 g, 9.4 mmol) in 4 mL dry DMF was added to a suspension of sodium cyanide (0.16 g, 3.4 mmol) in 4 mL, dry DMF After stirring for 10 min, the 3-dimethylamino-1-(2′-selenyl)propanone 2 (1.73 g, 7.52 mmol) in 10 mL DMF was added slowly. The mixture was stirred overnight. Water was added (30 ML), and the product was extracted with chloroform (3×30 mL). The extract was washed with water, dried over sodium sulfate, and evaporated. The product 3 was recrystallized from ethanol; yield: 1.97 g (88.3%). mp: 121-122.40° C. 1H NMR (500 MHz, CDCl3) δ 8.37 (dd, 1H, H-5 of selenophene ring, J=5.46, 0.91), 8.03 (dd, 1H, H-3 of selenophene ring, J=3.92, 0.91), 7.81 (dd, 1H, H-5 of thiophene ring, J=0.94, 3.82), 7.63 (dd, 1H, H-3 of thiophene ring, J=4.91,0.94), 7.40 (dd, 1H, H-4 selenophene ring, J=5.46, 0.91), 7.14 (dd, 1H, H-4 of thiophene ring, J=3.82, 4.91), 3.40 (m, 4H, CH2-CH2). Anal. Calcd. for C12H10O2SSe:C, 48.49; H, 3.39; S, 10.79. Found: C, 48.83; H, 3.38; S, 10.46.
  • EXAMPLE 37
  • Preparation of 2-(2′-Selenyl)-5-(2″-Thienyl)Thiophene (4).
    Figure US20050203077A1-20050915-C00043
  • 1-(2′-Thienyl)-4-(2″-selenyl)butane-1,4-dione 3 (1.1 g, 3.70 mmol) and Lawesson's reagent (0.99 g, 2.44 mmol) were refluxed overnight in 15 mL toluene. The toluene was evaporated and the crude product was purified using silica flash column with ether/hexane as eluent. The product 4 was recrystallized from methanol; yield: 0.9 g (82.3 %). mp. 103-104° C. 1H NMR (CDCl3) δ 7.84 (dd, 1H, J=5.57, 1.04) 7.30 (dd, 1H, J=3.78,1.04), 7.20 (m, 2H), 7.15 (dd, 1H, J=3.52, 1.1), 7.00 (m, 3H); 13 C NM (300 Mhz, CDCl3) δ 142.09 (weak), 138.36 (weak), 137.01 (weak), 136.32 (weak), 130.29, 129.60, 127.84, 125.73, 124.96, 124.45, 124.29, 123.63. Anal. Calcd for C12H8S2Se: C, 48.81; H, 2.73; S, 21.72. Found: C, 49.19; H, 2.58; S, 21.68.
  • EXAMPLE 38
  • Preparation of 2-(2′-Selenyl)-5-(2″-Thienyl)Furene (5).
    Figure US20050203077A1-20050915-C00044
  • 1-(2′-Thienyl)-4-(2″-selenyl)butane-1,4-dione 3 (0.76 g, 2.56 mmol) was added to 35 mL of acetic anhydride, then slowly added 3.0 ml of HCl After 4 h at room temperature, the reaction mixture was poured into ice water and extracted with ether. The organic layer was washed with NaHCO3 and water, dried over sodium sulfate. After evaporation of the solvent, the crude material was subjected to silica column purification to give the product 5. Yield: 0.51 g (75.5%). The yellowish white solid was recrystallized from methanol. mp. 85-87° C. 1H NMR (CDCl3) δ 7.89 (dd, 1H, J=4.51, 1.03), 7.44 (dd, 1H, J=3.82, 1.01), 7.29(dd, 1H, J=3.72, 1.08), 7.26 (dd, 1H, J=4.51, 3.82), 7.22 (dd, 1H, J=5.03, 1.08), 7.03 (dd, 1H, J=5.02, 3.70), 6.53 (m, 2H).
  • EXAMPLE 39
  • Preparation of 2-(2′-Selenyl)-5-(2″-Thienyl)Pyrrol (6).
    Figure US20050203077A1-20050915-C00045
  • 1-(2′-Thienyl)-4-(2″-selenyl)butane-1,4-dione 3 (0.4 g, 1.35 mmol), sodium acetate (0.33 g, 4.0 mmol) and ammonium acetate (0.78 g, 10.1 mmol) were refluxed at 95° C. overnight in 20 mL ethanol. The solvent was evaporated and the crude product 6 was purified using silica flash column with ether/hexane as eluent; yield: 0.27 g (73%). mp. 82-83.5° C. 1H NMR δ 8.26 (br, 1H), 7.81 (d, 1H, J=5.27), 7.25 (dd, 1H, J=3.78, 5.27), 7.20(d, 1H, J=3.78), 7.16 (d, 1H, J=5.01), 7.07 (d, 1H, J=3.60), 7.02 (dd, 1H, J=5.01, 3.60), 6.40 (m, 2H).
  • EXAMPLE 40
  • Preparation of 1-(2′-Selenyl)-4-(2″-Furyl)Butane-1,4-Dione (7).
    Figure US20050203077A1-20050915-C00046
  • A solution of 2-formylfurene (2.27 g, 23.65 mmol) in 20 mL dry DMF was added to a suspension of sodium cyanide (0.42 g, 8.45 mmol) in 10 mL dry DMF After stirring for 10 min, 3-dimethylamino-1-(2′-selenyl)propanone 2 (4.3 g, 18.8 mmol) in 20 mL DMF was added slowly. The mixture was stirred overnight. Water was added (100 mL), and the product was extracted with chloroform (3×100 mL). The extract was washed with water,.dried over sodium sulfate, and evaporated. The product 7 was recrystallized from ethanol; yield: 3.52 g (66.7%). mp. 82-83.5° C. 1H NMR (CDCl3) δ 8.35 (dd, 1H, H-5 of selenophene ring, J=5.51, 0.78), 8.01 (dd, 1H, H-3 of selenophene ring, J=3.99, 0.79), 7.58 (d, 1H, H-5 of furene ring, J=1.71), 7.39 (dd, 1H, H-4 of selenophene ring, J=5.52, 3.99), 7.23 (d, 1H, H-3 of furene ring, J=3.54), 6.53 (dd, 1H, H-4 of furene ring, J=3.54, 1.70), 3.33 (m, 4H, CH2-CH2)
  • EXAMPLE 41
  • Preparation of 2-(2′-Selenyl)-5-(2″-Furyl)Thiophene (8).
    Figure US20050203077A1-20050915-C00047
  • 1-(2′-Selenyl)-4-(2″-furyl)butane-1,4-dione 7 (0.25 g, 0.9 mmol) and Lawesson's reagent (0.66 g, 1.63 mmol) were refluxed overnight in 7 mL toluene. The toluene was evaporated and the crude product was purified using silica flash column with ether/hexane as eluent. The product 8 was recrystallized from methanol; yield: 0.22 g (88%). mp. 76-77° C. 1H NMR δ 7.86 (dd, 1H, J=5.58, 1.00), 7.40 (d, 1H, J=1.76), 7.31(dd, 1H, J=3.87, 1.00), 7.23 (dd, 1H, J=5.59, 3.87), 7.11 (d, 1H, J=3.78), 7.03 (d, 1H, J=3.81), 6.49 (d, 1H, J=3.36), 6.43(dd, 1H, J=1.77, 3.36).
  • EXAMPLE 42
  • Preparation of 2-(2′-Selenyl)-5-(2″-Furyl)Pyrrol (9).
    Figure US20050203077A1-20050915-C00048
  • 1-(2′-Thienyl)-4-(2″-furyl)butane-1,4-dione 7 (0.20 g, 0.71 mmol), sodium acetate (0.18 g, 2,1 mmol) and ammonium acetate (0.41 g, 5.3 mmol) were refluxed at 95° C. overnight in 12 mL ethanol. The solvent was evaporated and the crude product 9 was purified using silica flash column with ether/hexane as eluent; yield: 0.15 g (80%). mp. 73-74° C. 1HNMR δ 8.50 (br, 1H), 7.80 (d, 1H, J=5.45), 7.36 (dd, 1H, J=1.02, 0.78), 7.22(m, 2H), 6.40 (in, 4H).
  • EXAMPLE 43
  • Preparation of 1,4-Bis-(2′-Selenyl)Butane-1,4-Dione (11).
    Figure US20050203077A1-20050915-C00049
  • Synthesis of 2-formylselenophene (10): A solution of selenophene (1.31 g, 10 mmol) in 10 ml dichloroethane was added to a mixture of freshly distilled phosphorus oxychloride (2.0 g, 13 mmol) and DMF (1.10 g, 15 mmol). After stirring for 12 hr at 60° C., 2 ml water solution of sodium acetate (2.04 g, 15 mmol) was added to the reaction mixture, and the mixture was allowed to react for another hour. Water was added (20 mL), and the product was extracted with dichloromethane (3×20 mL). The extract was washed with water, dried over sodium sulfate, and carefully evaporated.
  • A solution of crude 2-formylselenophene (454 mg, 2.9 mmol) in 0.6 mL dry DMF was added to a suspension of sodium cyanide (34.3 mg, 0.7 mmol) in 0.4 mL dry DMF. After stirring for 5 min, the Mannich base, 3-dimethylamino-1-(2-selenyl)-propanone 2 (368 mg, 1.6 mmol) in 1.2 mL DMF was added slowly. The mixture was stirred overnight. Water was added (4 mL, and the product was extracted with dichloromethane (3×6 mL). The extract was washed with water, dried over sodium sulfate, and evaporated. The product was purified from silica gel chromatography with THF/hexane as eluent. Yield: 105 mg (20%). 1HNMR (CDCl3) δ 8.37 (dd, 1H×2, H-5 of selenophene ring, J=5.53, 1.01), 8.02 (dd, 1H×2, H-3 of selenophene ring, J=1.00, 3.97), 7.40 (dd, 1H×2, H-4 of selenophene ring, J=3.97, 5.50 ), 3.39 (s, 2H×2, CH2-CH2).
  • EXAMPLE 44
  • Preparation of 2,5-Bis-(2′-Selenyl)-N-Methylpyrrol (12).
    Figure US20050203077A1-20050915-C00050
  • 1,4-Bis-(2′-selenyl)butane-1,4-dione 11 (34.4 mg, 0.1 mmol), sodium acetate (123 mg, 0.15 mmol) and methylamine chloride (101.3 mg, 0.15 mmol) were refluxed overnight in 3 mL ethanol. 10 ml water was then added, and the product was extracted with dichloromethane. The product 12 was recrystallized from ethanol; yield: 26 mg (76%). 1H NMR (CDCl3) δ 7.96 (dd, 1H×2, H-5 of selenophene ring J=5.64, 1.64), 7.31 (dd, 1H×2, H-4 of selenophene ring J=5.64, 3.78), 7.20 (dd, 1H×2, H-3 of selenophene ring J=3.78, 1.64), 6.32 (s, 1H×2, H-¾ of pyrrol ring), 3.74 (s, 3H, N-Me).
  • EXAMPLE 45
  • Preparation of 2,5-Bis-(2′-Thienyl)Selenophene (13).
    Figure US20050203077A1-20050915-C00051
  • Selenophene (22 mg, 0.28 mmol) and sodium (19.2 mg, 0.83 mmol) were stirred under argon in dry DMF (10 ml at 100° C. until the solution decolorized, forming a brown suspension (2 h). The mixture of MeOH and EtOH (1:1,2 mL) was added to the suspension at 0° C., followed by addition of 1,4-Bis(2-thienyl)butadiyne (30 mg, 0.139 mmol) in solution of THF (3 mL). After half hour, the mixture was then poured into water (20 mL) and extracted with ether (3×15 mL). The concentrated organic layer yielded 28 mg of 13 (68.7%) after silica chromatography. 1NMR was identical with the literature. (R. Shabana et al. Phosphorus, Sulfur, and Silicon, 1990, 48, 239-244).
  • EXAMPLE 46
  • Preparation of 2,5-Bis-(2′-Furyl)Selenophene (14).
    Figure US20050203077A1-20050915-C00052
  • Selenophene (868 mg 11 mmol) and sodium (757 mg, 33 mmol) were stirred under argon in dry DMF (15 mL) at 100° C. until the solution decolorized, forming a brown suspension (2 h). The mixture of MeOH and EtOH (1:1,3 mL) was added to the suspension at 0° C., followed by addition of 1,4-Bis-(2′-furyl)butadiyne (1 g, 5.5 mmol) in solution of THF (3 mL). After half hour, the mixture was poured into water (20 mL) and extracted with ether (3×20 mL). The concentrated organic layer yielded 0.343 g (24%) of 14 after silica chromatography with hexane as eluent. 1H NMR was identical with the literature.(R. Shabana et al. Phosphorus, Sulfur, and Silicon, 1990, 48, 239-244).
  • EXAMPLE 47
  • Preparation of 5′-Formyl-2,5-Bis-(2′-Furyl)Selenophene (15).
    Figure US20050203077A1-20050915-C00053
  • To a solution of 2,5-bis-(2′-furyl)selenophene 14 (0.12 g, 0.456 mmol) in THF, lithium diisopropyl amide (0.73 mmol) was added at −78° C. under argon. The mixture was stirred below −20° C. for 3 h. A large excess of DMF (6.5 mmol) was added at −78° C., and the mixture was allowed to gradually rise to room temperature. Ether (10 mL) was added, and the organic solution was washed with water, dried over sodium sulfate, and evaporated. The crude solid was purified by flash column chromatography over silica gel (ether/hexane) to give monosubstituted aldehydes 15. Yield: 78 mg (60%), which was recrystallized from THF/Hexane to provide pure product. mp: 87.5-89.2 °C.1H NMR (CDCl3) δ 9.58 (s, 1H), 7.58 (d, 1H, J=4.04), 7.43 (s, 1H), 7.35 (d, 1H, J=4.04), 7.26 (d, 1H, J=3.69), 6.64 (d, 1H, J=3.69), 6.57 (d, 1H, J=3.16), 6.46 (m, 1H).
  • EXAMPLE 48
  • Preparation of 5′-Hydroxymethyl-2,5-Bis-(2′-Furyl)Selenophene (16).
    Figure US20050203077A1-20050915-C00054
  • To a solution of 5′-formyl-2,5-bis-(2′-furyl)selenophene (15 mg, 0.05 mmol) in 5 ml THF/MeOH (1:1), excess NaBH4 was added at room temperature. The solution was stirred for 2 h. Ethyl acetate was added, and the organic solution was washed with water, dried over sodium sulfate, and evaporated. The crude solid was purified by recrystallization from THF/Hexane to provide pure product 16. Yield: 14 mg (93.4%). mp: 75.0-77.4° C.1H NMR (CDCl3) δ0.39 (in, 1H), 7.31 (in, 2H), 6.48 (in, 1H), 6.43 (in, 2H), 6.33 (in, 1H). 13C NMR (THF-d8) δ 153.36 (weak), 150.99 (weak), 141.81, 136.66 (weak), 136.19 (weak), 125.46 (weak), 124.99, 124.71, 111.99, 105.89, 105.15, 57.43.
  • EXAMPLE 49
  • Preparation of 5′,5″-Diformyl-2-(2′-Selenyl)-5-(2″-Thienyl)Thiophene (17).
    Figure US20050203077A1-20050915-C00055
  • To a solution of 2-(2′-selenyl)-5-(2″-thienyl)thiophene 4 (0.45 g, 1.53 mmol) in THF was added lithium diisopropyl amide (2.44 mmol) at −78° C. under argon. The mixture was stirred below −20° C. for 3 h. Large excess of DMF (13 mmol) was added at −78° C., and the mixture was allowed to gradually rise to room temperature. Ether (30 mL) was added, and the organic solution was washed with water, dried over sodium sulfate, and evaporated. The crude solid was purified by flash column chromatography over silica gel (ether/hexane) to give disubstituted aldehydes 17. Yield: 135 mg (27.3%), which was recrystallized from THF/Hexane to provide pure product. mp: 197.8-199.0° C. 1H NNIR (CDCl3) δ 9.88 (s, 1H), 9.75 (s, 1H), 7.92 (d, III, J=4.28), 7.69 (d, 1H, J=3.87), 7.46 (d, 1H, J=4.28), 7.30 (d, 1H, J=1.93), 7.29 (d, 1H, J=1.93), 7.26 (d, III, J=3.87).
  • EXAMPLE 50
  • Preparation of 5′,5″-Dihydroxymethyl-2-(2′-Selenyl)-5-(2″-Thienyl)-Thiophene (18).
    Figure US20050203077A1-20050915-C00056
  • To a solution of 5′,5″-diformyl-2-(2′-selenyl)-5-(2″-thienyl)thiophene (12 mg, 0.03 mmol) in 1.5 ml THF/MeOH (1:1), excessive NaBH4 was added at room temperature. The solution was stirred for 4 h. Ethyl acetate was added, and the organic solution was washed with water, dried over sodium sulfate, and evaporated. The crude solid was purified by recrystallization from THF/Hexane to provide pure product 18. Yield: 8.2 mg (68.3%). mp: 187.1-188.8° C. 1H NNM (CDCl3) δ 7.20 (d,1H, J=3.76), 7.07 (m, 3H), 7.00 (d, 1H, J=3.66), 6.88 (d, 1H, J=3.39), 5.56 (t, 1H, OH), 5.45 (t, 1H,OH), 4.65 (m, 4H, 2CH2).
  • EXAMPLE 51
  • Synthesis of Water Soluble Analogs
  • A highly polar functional group can be incorporated into the selenophene compounds in order to improve their water solubility. Addition of a carbonylic functional group through an ester linkage (Scheme 5) resulted in a transient solubility. However, the benzylic ester may be readily hydrolyzed to regenerate the water insoluble starting material.
    Figure US20050203077A1-20050915-C00057
  • On the basis of the synthesis for hybrid ∝-terselenophenes (Scheme 3), a nitrogen atom can be introduced into the five-membered ring system (Scheme 6). Conversion of the hydroxyl group of the intermediate compound of scheme 3 into an amino group can improve water solubility. Further modification of its formulation may further enhance solubility to>1 mg/ml H2O; The ammonium analog should be highly water soluble.
    Figure US20050203077A1-20050915-C00058
  • To maximize the efficiency of synthesizing hybrid ∝-terselenophenes, Scheme 1 can be modified to produce related selenophene analogs in accordance with Scheme 7:
    Figure US20050203077A1-20050915-C00059
  • EXAMPLE 52
  • Synthesis of Prodrugs
  • An alternative approach of enhancing the water solubility of hydrophobic drugs comprises the preparation of their polar prodrug analogs.
  • a. Glycosides: Preliminary results indicate that β-D-glucoside of 2-hydroxymethyl-∝-terthiophene retains both its in vitro and in vivo activities. Scheme 8 illustrates a procedure utilized for the synthesis of glucoside, galactoside or, glucuronic acid analogs of ∝-terselenophene:
    Figure US20050203077A1-20050915-C00060
  • b. Glutamate Conjugate: As mentioned above, conversion of the hydroxyl group of 2-hydroxymethyl-5,2′:5′, 2″-terselenophene into its amino analog can moderately improve its water solubility. However, the amino analog is less stable. The amino analog may be transformed into its γ-glutamate prodrug (as shown in Scheme 9) to further enhance its water solubility and stability. This conjugate may also enhance target selectivity for the treatment of kidney cancer because of the higher γ-glutamyl transpeptidase activity in kidney. A modified procedure can also be designed for the preparation of glutathione conjugate.
    Figure US20050203077A1-20050915-C00061
    Figure US20050203077A1-20050915-C00062
  • c. Formation of Inclusion Complexes
  • The hydrophobic cavity of cyclodextrin derivatives can form stable inclusion complexes with 2-aminomethyl substituted thiophene compounds. β-Cyclodextrin (cyclic heptaamylose) derivatives are commonly used for improving water solubility because of their low costs. It is anticipated that the selenophene compounds of the present invention can be complexed with β-hydroxypropyl, dimethyl and sulfated β-cyclodextrins to enhance the water solubility of those compounds.
  • Example 53
  • Additional National Cancer Institute data demonstrating selenophene growth inhibition of human cancer cell lines is represented in the following tables. The compound must exhibit a Log10 GI50 value of<−4.00 to be considered active against the tested cell line.
    NSC: 688829
    Figure US20050203077A1-20050915-C00063
    Panel/Cell Line Log10GI50 Log10TGI Log10LC50
    Leukemia
    CCRF-CEM −5.10 >−4.00 >−4.00
    K-562 −6.60 >−4.00 >−4.00
    MOLT-4 −4.24 >−4.00 >−4.00
    RPMI-8226 −4.41 >−4.00 >−4.00
    SR −4.61 −4.14 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 −6.60 −4.80 >−4.00
    HOP-92 −4.01 >−4.00 >−4.00
    NCI-H226 −6.05 >−4.00 >−4.00
    NCI-H23 >−4.00 >−4.00 >−4.00
    NCI-H322M −4.56 −4.05 >−4.00
    NCI-H460 −6.85 >−4.00 >−4.00
    NCI-H522 −4.95 −4.40 >−4.00
    Colon Cancer
    COLO 205 −4.92 −4.48 −4.04
    HCC-2998 −6.41 >−4.00 >−4.00
    HCT-116 −6.53 >−4.00 >−4.00
    HCT-15 −4.64 >−4.00 >−4.00
    HT29 −4.63 >−4.00 >−4.00
    KM12 >−4.00 >−4.00
    SW-620 −6.08 >−4.00 >−4.00
    CNS Cancer
    SF-295 >−4.00 >−4.00 >−4.00
    SF-539 −4.56 >−4.00 >−4.00
    SNB-19 >−4.00 >−4.00 >−4.00
    SNB-75 −4.12 >−4.00 >−4.00
    U251 −6.61 −4.59 >−4.00
    Melanoma
    LOX IMVI −4.78 >−4.00 >−4.00
    MALME-3M −4.54 >−4.00 >−4.00
    M14 −4.70 >−4.00 >−4.00
    SK-MEL-2 −4.47 >−4.00 >−4.00
    SK-MEL-28 −4.18 >−4.00 >−4.00
    SK-MEL-5 −4.63 >−4.00 >−4.00
    UACC-257 −6.71 −6.29 >−4.00
    UACC-62 −6.85 >−4.00 >−4.00
    Ovarian Cancer
    IGROVI −6.75 −5.61 −4.04
    OVCAR-3 −6.89 −6.20 >−4.00
    OVCAR-4 −6.73 >−4.00
    OVCAR-5 −6.91 −6.30 >−4.00
    OVCAR-8 −4.82 −4.03 >−4.00
    SK-OV-3 −4.58 >−4.00 >−4.00
    Renal Cancer
    786-0 −4.57 −4.19
    A498 −7.67 −7.10 −6.48
    ACHN >−4.00 >−4.00 >−4.00
    CAKI-1 −6.72 −6.30 −4.53
    SN12C −4.55 −4.07 >−4.00
    TK-10 −7.55 −6.68 −4.21
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 −4.55 >−4.00 >−4.00
    DU-145 >−4.00 >−4.00 >−4.00
    Breast Cancer
    MCF7 −6.72 >−4.00 >−4.00
    MCF7/ADR-RES −4.52 >−4.00 >−4.00
    MDA-MB-231/ATCC −4.63 −4.17 >−4.00
    HS 578T −5.54 >−4.00 >−4.00
    MDA-MB-435 −4.48 >−4.00 >−4.00
    MDA-N −4.68 >−4.00 >−4.00
    BT-549 −4.25 >−4.00 >−4.00
    T-47D −6.47 >−4.00 >−4.00
    MG_MID −5.27 −4.35 −4.06
    Delta 2.40 2.75 2.42
    Range 3.67 3.10 2.48
    NSC: 688830
    Figure US20050203077A1-20050915-C00064
    Leukemia
    CCRF-CEM −4.59 >−4.00 >−4.00
    K-562 −7.14 −4.76 −4.01
    MOLT-4 −4.41 >−4.00 >−4.00
    RPMI-8226 −4.43 >−4.00 >−4.00
    SR −4.61 −4.14 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −5.22 −4.48 >−4.00
    EKVX −4.71 −4.18 >−4.00
    HOP-62 −7.21 −4.55 −4.07
    HOP-92 −4.55 >−4.00 >−4.00
    NCI-H322M −7.22 >−4.00
    NCI-H23 −4.07 >−4.00 >−4.00
    NCI-H322M −5.47 −4.70 >−4.00
    NCI-H460 −7.77 −6.19 −4.54
    NCI-H522 >−4.00 >−4.00 >−4.00
    Colon Cancer
    COLO 205 −6.35 −5.26 −4.21
    HCC-2998 −7.20 −4.94 −4.35
    HCT-116 −7.56 −4.76 −4.09
    HCT-15 −4.54 >−4.00 >−4.00
    HT29 −6.65 −4.36 >−4.00
    KM12 −4.32 >−4.00 >−4.00
    SW-620 −7.40 −4.60 >−4.00
    CNS Cancer
    SF-295 −4.68 −4.25 >−4.00
    SF-539 −4.81 −4.44 −4.08
    SNB-19 −4.47 −4.06 >−4.00
    SNB-75 −4.59 >−4.00 >−4.00
    U251 −7.08 −4.68 −4.14
    Melanoma
    LOXIMVI −4.87 −4.56 −4.25
    MALME-3M −4.36 >−4.00 >−4.00
    M14 −4.43 >−4.00 >−4.00
    SK-MEL-2 −4.40 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 −4.44 >−4.00 >−4.00
    UACC-257 −7.78 −7.34 −6.62
    UACC-62 −7.87 >−4.00 >−4.00
    Ovarian Cancer
    IGROVI −7.85 −6.76 −4.55
    OVCAR-3 <−8.00 −7.25 −4.30
    OVCAR-4 −6.98 −4.09
    OVCAR-5 −7.64 >−4.00
    OVCAR-8 −5.16 −4.51 >−4.00
    SK-OV-3 −5.49 −4.71 −4.29
    Renal Cancer
    786-0 −5.19 −4.64 −4.24
    A498 <−8.00 −7.61 −7.14
    ACHN −4.68 >−4.00 >−4.00
    CAKI-1 <−8.00 −7.55
    SN12C −4.31 >−4.00 >−4.00
    TK-10 <−8.00 −7.43 −4.19
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 −4.51 >−4.00 >−4.00
    DU-145 −4.64 >−4.00 >−4.00
    Breast Cancer
    MCF7 −7.78 >−4.00 >−4.00
    MCF7/ADR-RES −4.91 >−4.00 >−4.00
    MDA-MB-231/ATCC −4.90 −4.45 −4.01
    HS 578T >−4.00 >−4.00
    MDA-MB-435 >−4.00 >−4.00 >−4.00
    MDA-N −4.01 >−4.00 >−4.00
    BT-549 >−4.00 >−4.00 >−4.00
    T-47D −6.54 −4.24
    MG_MID −5.64 −4.62 −4.16
    Delta 2.36 2.99 2.98
    Range 4.00 3.61 3.14
    NSC: 676631
    Figure US20050203077A1-20050915-C00065
    Leukemia
    CCRF-CEM >−4.00 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −6.99 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −4.23 >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 >−4.00 >−4.00
    HOP-92 >−4.00 >−4.00 >−4.00
    NCI-H226 −7.30 −6.74 −6.28
    NCI-H23 −4.78 −4.41 −4.04
    NCI-H322M −4.66 >−4.00 >−4.00
    NCI-H460 >−4.00 >−4.00
    NCI-H522 −5.28 −4.67 −4.26
    Colon Cancer
    COLO 205 −6.54 −4.90 −4.18
    HCC-2998 −4.37 >−4.00 >−4.00
    HCT-116 −7.27 >−4.00 >−4.00
    HCT-15 −4.56 >−4.00 >−4.00
    HT29 >−4.00 >−4.00
    KM12 −4.57 >−4.00 >−4.00
    SW-620 −6.45 >−4.00 >−4.00
    CNS Cancer
    SF-268 −4.63 −4.19 >−4.00
    SF-295 −4.26 >−4.00 >−4.00
    SF-539 −4.91 −4.24 >−4.00
    SNB-19 −4.76 >−4.00 >−4.00
    SNB-75 −4.38 >−4.00 >−4.00
    U251 −7.11 −4.70 −4.35
    Melanoma
    LOX IMVI −4.82 −4.44 −4.07
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 −4.70 >−4.00 >−4.00
    SK-MEL-2 >−4.00 >−4.00 >−4.00
    SK-MEL-28 −4.18 >−4.00 >−4.00
    SK-MEL-5 >−4.00 >−4.00 >−4.00
    UACC-257 −7.50 >−4.00 −6.34
    UACC-62 −7.58 −6.94 >−4.00
    Ovarian Cancer
    IGROVI −7.09 −6.22 −4.29
    OVCAR-3 −7.56 −6.86 −4.39
    OVCAR-4 >−4.00 >−4.00
    OVCAR-5 −7.50 −6.68
    OVCAR-8 −4.61 >−4.00 >−4.00
    SK-OV-3 −4.34 >−4.00 >−4.00
    Renal Cancer
    786-0 >−4.00 >−4.00 >−4.00
    A498 −7.56 −7.08 −6.52
    ACHN >−4.00 >−4.00 >−4.00
    CAKI-1 −7.51 −6.72 −4.19
    RXF-393 −4.14 >−4.00 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 −7.30 −6.43 −4.14
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 −4.28 >−4.00 >−4.00
    DU-145 −4.70 −4.11 >−4.00
    Breast Cancer
    MCF7 −7.03 −4.66 >−4.00
    MCF7/ADR-RES −5.00 −4.50 −4.01
    MDA-MB-231/ATCC −4.72 −4.06 >−4.00
    HS 578T >−4.00 >−4.00 >−4.00
    MDA-MB-435 −5.09 −4.26 >−4.00
    MDA-N −4.78 −4.36 >−4.00
    BT-549 −4.77 −4.42 −4.06
    T-47D −6.17 >−4.00 >−4.00
    MG_MID −5.18 −4.47 −4.15
    Delta 2.40 2.61 2.37
    Range 3.58 3.08 2.52
    NSC: 675246
    Figure US20050203077A1-20050915-C00066
    Leukemia
    CCRF-CEM −5.50 −4.91 >−4.00
    HL-60(TB) −5.24 >−4.00 >−4.00
    K-562 −6.43 −5.12 >−4.00
    MOLT-4 −5.49 −4.92 >−4.00
    RPMI-8226 −5.13 >−4.00 >−4.00
    SR −5.18 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −4.98 −4.65 −4.33
    EKVX −5.26 −4.76 −4.37
    HOP-62 −5.14 −4.69 −4.35
    HOP-92 −5.58 −4.95 −4.42
    NCI-H322M −6.21 −5.63
    NCI-H23 −4.88 −4.51 −4.15
    NCI-H322M −4.97 −4.65 −4.32
    NCI-H460 −6.49 −5.44 −4.64
    NCI-H52 −5.61 −5.18 −4.63
    Colon Cancer
    COLO 205 −6.00 −5.39 >−4.00
    HCC-2998 −5.91 −4.93 −4.27
    HCT-116 −7.19 −4.97 −4.49
    HCT-15 −5.37 −4.75 >−4.00
    HT29 −6.07 −4.97 −4.23
    KM12 −5.38 −4.77 −4.28
    SW-620 −6.38 −4.99 −4.45
    CNS Cancer
    SF-268 −5.39 >−4.00 >−4.00
    SF-295 −4.97 −4.56 −4.14
    SF-539 −4.91 −4.60 −4.29
    SNB-19 −4.91 −4.28 >−4.00
    SNB-75 −5.41 >−4.00 >−4.00
    U251 −7.15 −4.91 −4.26
    Melanoma
    LOXIMVI −5.44 −5.01 −4.43
    MALME-3M −5.21 −4.71 −4.30
    M14 −5.06 −4.62 −4.21
    SK-MEL-2 −5.04 −4.59 −4.16
    SK-MEL-28 −5.13 −4.66 −4.27
    SK-MEL-5 −5.58 −5.05 −4.53
    UACC-257 −7.30 −6.65
    UACC-62 −7.99 −4.95 −4.17
    Ovarian Cancer
    IGROVI −7.27 −5.65 −4.91
    OVCAR-3 −7.22 −5.89 −5.16
    OVCAR-4 −6.24 −4.91 −4.32
    OVCAR-5 −6.74 −4.60 >−4.00
    OVCAR-8 −5.30 −4.48 >−4.00
    Renal Cancer
    786-0 −5.66 −5.28 −4.76
    A498 −7.41 −6.78 −6.16
    CAKI-1 −5.27 −4.73 −4.29
    RXF-393 −7.68 −7.04
    TK-10 −5.72 −4.97 >−4.00
    UO-31 −5.21 −4.41 >−4.00
    −7.50 −6.65 −4.05
    −4.92 −4.61 −4.29
    Prostate Cancer
    PC-3 −5.42 −4.88 −4.44
    DU-145 −4.99 −4.66 −4.33
    Breast Cancer
    MCF7 −6.84 −5.42 −4.53
    MCF7/ADR-RES −5.29 −4.33 >−4.00
    MDA-MB-231/ATCC −5.36 −4.47 >−4.00
    MDA-N −5.01 −4.25 >−4.00
    T-47D −5.42 −4.82 −4.35
    −5.56 −4.90 −4.40
    −5.20 −4.68 −4.26
    −5.63 −4.33 >−4.00
    MG_MID −5.78 −4.91 −4.29
    Delta 2.22 2.14 1.88
    Range 3.11 3.04 2.16
    NSC: 675247
    Figure US20050203077A1-20050915-C00067
    Leukemia
    CCRF-CEM −5.47 −5.00 >−4.00
    HL-60(TB) −5.39
    K-562 −5.88 −5.30 −4.15
    MOLT-4 −5.48 −5.08 >−4.00
    RPMI-8226 −5.39 >−4.00 >−4.00
    SR −5.43 >−4.00 >−4.00
    >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −5.34 −4.71 −4.27
    EKVX −5.30 −4.46 >−4.00
    HOP-62 −5.26 −4.67 −4.24
    HOP-92 −5.62 −5.12 −4.26
    NCI-H226 −5.77 −5.41 −5.04
    NCI-H23 −5.18 −4.60 −4.02
    NCI-H322M −4.95 −4.61 −4.28
    NCI-H460 −6.28 −5.00 −4.09
    NCI-H522 −5.78 −5.47 −5.15
    Colon Cancer
    COLO 205 −5.71 −5.29 −4.75
    HCC-2998 −6.07 −5.49 −4.90
    HCT-116 −6.27 −4.95 >−4.00
    HCT-15 −5.42 −4.91 −4.26
    HT29 −5.79 −5.12 −4.18
    KM12 −5.35 −4.82 −4.30
    SW-620 −5.87 −5.44 −5.02
    CNS Cancer
    SF-268 −5.76 −5.36 −4.68
    SF-295 −4.93 −4.54 −4.15
    SF-539 −4.76 −4.39 −4.02
    SNB-19 −5.26 −4.60 −4.04
    SNB-75 −4.83 −4.41 >−4.00
    U251 −6.33 −4.91 −4.30
    Melanoma
    LOXIMVI −5.52 −5.15 >−4.00
    MALME-3M −5.54 −4.97 −4.25
    M14 −5.43 −4.89 −4.31
    SK-MEL-2 −5.15 −4.62 −4.16
    SK-MEL-28 −5.29 >−4.00 >−4.00
    SK-MEL-5 −5.84 −5.52 −5.20
    UACC-257 −6.36 −5.70 −4.50
    UACC-62 −6.81 −5.61 −4.35
    Ovarian Cancer
    IGROVI −5.86 −5.19 −4.52
    OVCAR-3 −6.68 −5.93 −5.27
    OVCAR-4 −5.77 −5.16 −4.31
    OVCAR-5 −5.88 −4.89 >−4.00
    OVCAR-8 −5.43 −4.75 −4.14
    Renal Cancer
    786-0 −5.56 −5.20 −4.15
    A498 −6.42 −5.85 −5.14
    ACHN −5.40 >−4.00 >−4.00
    CAKI-1 −7.13 −6.33 −4.88
    RXF-393 −5.80 −5.43 −5.06
    SN12C −5.56 −4.93 >−4.00
    TK-10 −6.86 −6.18 −4.80
    UO-31 −5.28 −4.83 −4.41
    Prostate Cancer
    PC-3 −5.45 −4.91 −4.45
    DU-145 −5.16 −4.71 −4.33
    Breast Cancer
    MCF7 −6.80 −4.95 −4.32
    MCF7/ADR-RES −5.42 −4.58 >−4.00
    MDA-MB- −5.30 −4.79 −4.32
    3221/ATCC −5.27 −4.44 >−4.00
    HS 578T −5.49 −4.95 −4.33
    MDA-MB-435 −5.53 −5.01 −4.31
    MDA-N −5.11 −4.60 −4.13
    BT-549 −5.47 −4.85 >−4.00
    T-47D
    MG_MID −5.65 −4.98 −4.34
    Delta 1.48 1.36 0.93
    Range 2.37 2.33 1.27
    NSC: 675343
    Figure US20050203077A1-20050915-C00068
    Leukemia
    CCRF-CEM −4.65 −4.22 >−4.00
    HL-60(TB) −4.21 >−4.00 >−4.00
    K-562 <−8.00 −4.58 >−4.00
    MOLT-4 −4.54 −4.06 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC >−4.00 >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 >−4.00 >−4.00 >−4.00
    HOP-92 −4.39 >−4.00 >−4.00
    NCI-H226 <−8.00 −7.14 >−4.00
    NCI-H23 >−4.00 >−4.00 >−4.00
    NCI-H322M −5.01 >−4.00 >−4.00
    NCI-H460 <−8.00 >−4.00 >−4.00
    NCI-H522 −4.00 >−4.00 >−4.00
    Colon Cancer
    COLO 205 <−8.00 −5.98 −4.64
    HCC-2998 <−8.00 −5.47 >−4.00
    HCT-116 <−8.00 >−4.00 >−4.00
    HCT-15 >−4.00 >−4.00 >−4.00
    HT29 >−4.00 >−4.00
    KM12 >−4.00 >−4.00 >−4.00
    SW-620 <−8.00 >−4.00 >−4.00
    CNS Cancer
    SF-268 >−4.00 >−4.00 >−4.00
    SF-295 >−4.00 >−4.00 >−4.00
    SF-539 >−4.00 >−4.00 >−4.00
    SNB-19 >−4.00 >−4.00 >−4.00
    SNB-75
    U251 <−8.00 >−4.00 >−4.00
    Melanoma
    LOX IMVI >−4.00 >−4.00 >−4.00
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 >−4.00 >−4.00 >−4.00
    SK-MEL-2 >−4.00 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 >−4.00 >−4.00
    UACC-257 <−8.00 <−8.00
    UACC-62 <−8.00 >−4.00 >−4.00
    Ovarian Cancer
    OVCAR-3 −7.59 −4.49 >−4.00
    OVCAR-4 <−8.00 >−4.00 >−4.00
    OVCAR-5 −6.80 >−4.00 >−4.00
    OVCAR-8 <−8.00 >−4.00 >−4.00
    SK-OV-3 >−4.00 >−4.00 >−4.00
    Renal Cancer
    786-0 >−4.00 >−4.00 >−4.00
    A498 <−8.00 <−8.00 −6.80
    ACHN >−4.00 >−4.00 >−4.00
    CAKI-1 <−8.00 <−8.00 >−4.00
    RXF-393 −4.68 >−4.00 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 <−8.00 <−8.00 >−4.00
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 >−4.00 >−4.00 >−4.00
    DU-145 >−4.00 >−4.00 >−4.00
    Breast Cancer
    MCF7 <−8.00 >−4.00 >−4.00
    MCF7/ADR-RES >−4.00 >−4.00 >−4.00
    MDA-MB-231/ATCC >−4.00 >−4.00 >−4.00
    HS 578T >−4.00 >−4.00 >−4.00
    MDA-MB-435 >−4.00 >−4.00 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 >−4.00 >−4.00 >−4.00
    T-47D −6.14 >−4.00 >−4.00
    MG_MID
    Delta −5.36 −4.41 −4.06
    Range 2.64 3.59 2.74
    4.00 4.00 2.80
    NSC: 676632
    Figure US20050203077A1-20050915-C00069
    Leukemia
    CCRF-CEM −4.06 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −7.32 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −5.58 >−4.00 >−4.00
    EKVX −4.30 >−4.00 >−4.00
    HOP-62 −7.12 >−4.00 >−4.00
    HOP-92 >−4.00 >−4.00 >−4.00
    NCI-H226 −7.71 −7.27 −6.49
    NCI-H23 >−4.00 >−4.00 >−4.00
    NCI-H322M >−4.00 >−4.00 >−4.00
    NCI-H460 −7.33 >−4.00 >−4.00
    NCI-H522 >−4.00 >−4.00 >−4.00
    Colon Cancer
    HCC-2998 −6.75 −6.26 −5.61
    HCT-116 >−4.00 >−4.00 >−4.00
    HCT-15 −7.35 >−4.00 >−4.00
    HT29 >−4.00 >−4.00 >−4.00
    KM12 −6.27 >−4.00 >−4.00
    SW-620 >−4.00 >−4.00 >−4.00
    −6.82 >−4.00 >−4.00
    CNS Cancer
    SF-268 >−4.00 >−4.00 >−4.00
    SF-295 >−4.00 >−4.00 >−4.00
    SF-539 >−4.00 >−4.00 >−4.00
    SNB-19 −4.37 >−4.00 >−4.00
    SNB-75 >−4.00 >−4.00 >−4.00
    U251 −7.45 −4.92 −4.33
    Melanoma
    LOX IMVI >−4.00 <−4.00 >−4.00
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 >−4.00 >−4.00 >−4.00
    SK-MEL-2 >−4.00 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 −4.20 >−4.00 >−4.00
    UACC-257 −7.67 >−4.00
    UACC-62 −7.65 −7.25 >−4.00
    Ovarian Cancer
    IGROV1 −7.49 −6.56 >−4.00
    OVCAR-3 −7.71 −7.17 >−4.00
    OVCAR-4 −6.87 >−4.00
    OVCAR-5 −7.88 −7.11 −6.11
    OVCAR-8 >−4.00 >−4.00 >−4.00
    >−4.00 >−4.00 >−4.00
    Renal Cancer
    786-0 >−4.00 >−4.00 >−4.00
    A498 −7.73 −7.37 −7.02
    ACHN >−4.00 >−4.00 >−4.00
    CAKI-1 −7.90 −6.91 >−4.00
    RXF-393 >−4.00 >−4.00 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 −7.55 −7.09 >−4.00
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 >−4.00 >−4.00 >−4.00
    DU-145 −4.02 >−4.00 >−4.00
    Breast Cancer
    MCF7 −7.91 >−4.00 >−4.00
    MCF7/ADR-RES >−4.00 >−4.00 >−4.00
    MDA-MB- >−4.00 >−4.00 >−4.00
    231/ATCC >−4.00 >−4.00 >−4.00
    HS578T >−4.00 >−4.00 >−4.00
    MIDA-MB-435 >−4.00 >−4.00 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 −6.71 >−4.00 >−4.00
    T-47D
    MG_MID −5.16 −4.48 −4.16
    Delta 2.75 2.89 2.86
    Range 3.91 3.37 3.02
    NSC: 675344
    Figure US20050203077A1-20050915-C00070
    Leukemia
    CCRF-CEM >−4.00 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −7.36 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8826 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −4.34 >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 −4.46 >−4.00 >−4.00
    HOP-92 −4.56 −4.05 >−4.00
    NCI-H226 <−8.00 <−8.00 −6.65
    NCI-H23 −4.69 >−4.00 >−4.00
    NCI-H322M −4.68 >−4.00 >−4.00
    NCI-H460 <−8.00 >−4.00 >−4.00
    NCI-H522 >−4.00 >−4.00 >−4.00
    Colon Cancer
    HCC-2998 −6.59 −5.79 −5.01
    HCT-116 <−8.00 −7.38 −5.53
    HCT-15 −7.59 >4.00 >−4.00
    HT29 >−4.00 >−4.00 >−4.00
    KM12 >−4.00 >−4.00 >−4.00
    SW-620 >−4.00 >−4.00 >−4.00
    −7.06 >−4.00 >−4.00
    CNS Cancer
    SF-268 −4.63 >−4.00 >−4.00
    SF-295 −4.57 >−4.00 >−4.00
    SF-539 >−4.00 >−4.00 >−4.00
    SNB-19 −4.53 >−4.00 >−4.00
    SNB-75 −4.78 −4.31 >−4.00
    U251 −7.60 −4.58 >−4.00
    Melanoma
    LOXIMVI −4.46 >−4.00 >−4.00
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 >−4.00 >−4.00 >−4.00
    SK-MEL-2 >−4.00 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 >−4.00 >−4.00 >−4.00
    UACC-257 <−8.00 −7.73 −7.28
    UACC-62 <−8.00 −7.85 >−4.00
    Ovarian Cancer
    IGROV1 −7.91 −7.37 −4.79
    OVCAR-3 <−8.00 >−4.00 >−4.00
    OVCAR-4 −7.38 >−4.00 >−4.00
    OVCAR-5 <−8.00 −7.06 >−4.00
    OVCAR-8 >−4.00 >−4.00 >−4.00
    SK-OV-3 −4.78 >−4.00 >−4.00
    Renal Cancer
    786-0 −4.91 −4.17 >−4.00
    A498 <−8.00 −7.74 −7.18
    ACHN −4.89 −4.08 >−4.00
    RXF-393 −4.80 −4.28 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 <−8.00 −7.30 >−4.00
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 >−4.00 >−4.00 >−4.00
    DU-145 −4.44 >−4.00 >−4.00
    Breast Cancer
    MCF7 −6.90 >−4.00 >−4.00
    MCF7/ADR-RES >−4.00 >−4.00 >−4.00
    MDA-MB-231/ATCC −4.72 >−4.00 >−4.00
    MS 578T −4.27 >−4.00 >−4.00
    MDA-MB-435 >−4.00 >−4.00 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 −4.37 >−4.00 >−4.00
    T-47D −6.21 >−4.00 >−4.00
    MG_MID −5.28 −4.54 −4.21
    Delta 2.72 3.46 3.07
    Range 4.00 4.00 3.28
    NSC: 676630
    Figure US20050203077A1-20050915-C00071
    Leukemia
    CCRF-CEM >−4.00 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −5.47 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −4.06 >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 >−4.00 >−4.00
    HOP-92 >4.00 >−4.00 >−4.00
    NCI-H226 −6.72 −6.31 −5.61
    NCI-H23 >−4.00 >−4.00
    NCI-H322M >−4.00 >−4.00 >−4.00
    NCI-H460 −6.89 >−4.00 >−4.00
    NCI-H522 −4.68 −4.27 >−4.00
    Colon Cancer
    COLO 205 −4.03 >−4.00 >−4.00
    HCC-2998 −4.07 >−4.00
    HCT-116 −5.52 >−4.00 >−4.00
    HCT-15 >−4.00 >−4.00 >−4.00
    HT29 >−4.00 >−4.00 >−4.00
    KM12 >−4.00 >−4.00 >−4.00
    SW-620 >−4.00 >−4.00 >−4.00
    CNS Cancer
    SF-268 >−4.00 >−4.00 >−4.00
    SF-295 >−4.00 >−4.00 >−4.00
    SF-539 >−4.00 >−4.00 >−4.00
    SNB-19 >−4.00 >−4.00 >−4.00
    SNB-75 −4.17 >−4.00 >−4.00
    U251 −6.23 >−4.00 >−4.00
    Melanoma
    LOC IMVI >−4.00 >−4.00 >−4.00
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 >−4.00 >−4.00 >−4.00
    SK-MEL-2 >−4.00 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 >−4.00 >−4.00 >−4.00
    UACC-257 −6.56 −6.17 −5.16
    UACC-62 −6.58 >−4.00 >−4.00
    Ovarian Cancer
    IGROVI −5.83 >−4.00 >−4.00
    OVCAR-3 −6.14 >−4.00 >−4.00
    OVCAR-4 >−4.00
    OVCAR-5 −6.88 −6.35 −5.45
    OVCAR-8 −4.48 >−4.00 >−4.00
    SK-OV-3 >−4.00 >−4.00 >−4.00
    Renal Cancer
    786-0 >−4.00 >−4.00 >−4.00
    ACHN >−4.00 >−4.00 >−4.00
    CAKI-1 −6.13 −4.82 >−4.00
    RXF-393 >−4.00 >−4.00 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 −6.42 −5.79 >−4.00
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 >−4.00 >−4.00 >−4.00
    DU-145 >−4.00 >−4.00 >−4.00
    Breast Cancer
    MCF7 −6.17 >−4.00 >−4.00
    MCF7/ADR-RES −4.10 >−4.00 >−4.00
    MDA-MB-231/ATCC >−4.00 >−4.00 >−4.00
    HS 578T >−4.00 >−4.00 >−4.00
    MDA-MB-435 >−4.00 >−4.00 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 −4.20 >−4.00 >−4.00
    T-47D >−4.00 >−4.00
    MG_MID
    Delta −4.58 −4.17 −4.07
    Range 2.31 2.18 1.54
    2.89 2.35 1.61
    NSC: 675245
    Figure US20050203077A1-20050915-C00072
    Leukemia
    CCRF-CEM >−4.00 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −5.17 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC >−4.00 >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 −4.29 >−4.00 >−4.00
    HOP-92 >−4.00 >−4.00 >−4.00
    NCI-H226 −6.01 −5.44 >−4.00
    NCI-H23 >−4.00 >−4.00 >−4.00
    NCI-H322M >−4.00 >−4.00 >−4.00
    NCI-H460 −5.75 >−4.00 >−4.00
    NCI-H522 −4.66 −4.34 −4.02
    Colon Cancer
    COLO 205 >−4.00 >−4.00 >−4.00
    HCC-2998 −5.32 >−4.00 >−4.00
    HCT-116 −6.16 >−4.00 >−4.00
    HCT-15 >−4.00 >−4.00 >−4.00
    HT29 >−4.00 >−4.00 >−4.00
    KM12 >−4.00 >−4.00 >−4.00
    SW-620 −5.38 >−4.00 >−4.00
    CNS Cancer
    SF-268 >−4.00 >−4.00 >−4.00
    SF-295 −4.31 >−4.00 >−4.00
    SF-539 >−4.00 >−4.00 >−4.00
    SNB-19 −4.53 >−4.00 >−4.00
    SNB-75 −5.00 >−4.00 >−4.00
    U251 −5.98 >−4.00 >−4.00
    Melanoma
    LOX IMVI >−4.00 >−4.00 >−4.00
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 >−4.00 >−4.00 >−4.00
    SK-MEL-2 >−4.00 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 −4.14 −4.00 >−4.00
    UACC-257 −6.49 −6.04 >−4.00
    UACC-62 −7.19 >−4.00 >−4.00
    Ovarian Cancer
    IGROV1 −4.17 −4.59 −4.13
    OVCAR-3 −6.41 >−4.00 >−4.00
    OVCAR-4 −5.58 >−4.00 >−4.00
    OVCAR-5 −6.11 >−4.00 >−4.00
    OVCAR-8 >−4.00 >−4.00 >−4.00
    Renal Cancer
    786-0 −4.17 >−4.00 >−4.00
    A498 −6.51 −6.05 −5.17
    ACHN −4.24 >−4.00 >−4.00
    CAKI-1 −6.57 −6.03 >−4.00
    RXF-393 −4.92 >−4.00 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 −6.59 −6.09 >−4.00
    UO-31 >−4.00 >−4.00 >−4.00
    Prostate Cancer
    PC-3 >−4.00 >−4.00 >−4.00
    DU-145 >−4.00 >−4.00 >−4.00
    Breast Cancer
    MCF7 −5.83 >−4.00 >−4.00
    MCF7/ADR-RES >−4.00 >−4.00 >−4.00
    MDA-MB-231/ATCC >−4.00 >−4.00 >−4.00
    HS 578T −4.06 >−4.00 >−4.00
    MDA-MB-435 >−4.00 >−4.00 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 >−4.00 >−4.00 >−4.00
    T-47D −4.80 >−4.00 >−4.00
    MG_MID
    Delta −4.67 −4.18 −4.02
    Range 2.53 1.91 1.15
    3.19 2.09 1.17
    NSC: 675244
    Figure US20050203077A1-20050915-C00073
    Leukemia
    CCRF-CEM −5.67 −5.15 >−4.00
    HL-60(TB) −5.61 >−4.00
    K-562 −5.87 −4.48 >−4.00
    MOLT-4 −5.62 −5.12 >−4.00
    RPMI-8226 −5.53 >−4.00 >−4.00
    −5.39
    Non-Small Cell Lung Cancer
    A549/ATCC −4.87 −4.58 −4.29
    EKVX −4.60 −4.19 >−4.00
    HOP-62 −4.92 −4.53 −4.13
    HOP-92 −4.96 −4.59 −4.20
    NCI-H226 −5.65 −5.29 −4.41
    NCI-H23 −4.87 −4.51 −4.16
    NCI-H322M −4.88 −4.55 −4.22
    NCI-H460 −5.65 −4.79 −4.39
    NCI-H522 −5.28 −4.76 −4.37
    Colon Cancer
    COLO 205 −5.50 −4.92 −4.46
    HCC-2998 −5.60 −4.94 −4.41
    HCT-116 −6.42 −4.86 −4.41
    HCT-15 −5.13 −4.58 −4.10
    HT29 −5.49 −4.88 −4.44
    KM12 −5.20 −4.71 −4.34
    SW-620
    CNS Cancer
    SF-268 −5.35 −4.24 >−4.00
    SF-295 −4.82 −4.55 −4.27
    SF-539 −4.77 −4.48 −4.20
    SNB-19 −4.99 −4.52 −4.06
    SNB-75 −5.70 −5.19 >−4.00
    U251 −6.50 −4.86 −4.32
    Melanoma
    LOXIMVI −5.25 −4.68 −4.11
    MALME-3M −4.90 −4.55 −4.19
    M14 −4.94 −4.54 −4.14
    SK-MEL-2 −4.78 −4.34 >−4.00
    SK-MEL-28 −4.88 −4.57 −4.26
    SK-MEL-5 −5.47 −4.86 −4.43
    UACC-257 −6.51 −6.07 −4.48
    UACC-62 −7.15 −4.97 −4.18
    Ovarian Cancer
    IGROV1 −5.48 −4.81 −4.40
    OVCAR-3 −6.55 −5.10 −4.51
    OVCAR-4 −5.80 −4.83 −4.22
    OVCAR-5 −6.26 −4.88 −4.12
    OVCAR-8 −5.04 −4.52 −4.02
    Renal Cancer
    786-0 −5.22 −4.73 −4.36
    A8498 −5.85 −5.53 −5.20
    ACHN −4.99 −4.66 −4.33
    CAKI-1 −6.69 −6.15 −4.84
    RXF-393 −5.71 −5.09 >−4.00
    SN12C −5.00 −4.65 −4.30
    TK-10 −6.54 −6.05 −4.60
    UO-31 −4.81 −4.54 −4.27
    Prostate Cancer
    PC-3 −4.92 −4.49 −4.06
    DU-145 −4.90 −4.60 −4.29
    Breast Cancer
    MCF7 −6.32 −4.94 −4.36
    MCF7/ADR-RES
    HS 578T −5.14 −4.66 −4.27
    MDA-MB-435 −4.99 −4.48 >−4.00
    MDA-N −5.34 −4.71 −4.19
    BT-549 −4.91 −4.51 −4.10
    T-47D −4.95 −4.63 −4.32
    −5.42 −4.45 >−4.00
    MG_MID
    Delta −5.43 −4.78 −4.25
    Range 1.72 1.37 0.96
    2.55 2.15 1.20
    NSC: 675346
    Figure US20050203077A1-20050915-C00074
    Leukemia
    CCRF-CEM >−4.00 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −5.64 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC −4.26 >−4.00 >−4.00
    EKVX −4.11 >−4.00 >−4.00
    HOP-62 −4.19 >−4.00 >−4.00
    HOP-92 −4.73 −4.15 >−4.00
    NCI-H226 −7.76 −6.92 −5.98
    NCI-H23 −4.84 −4.23 >−4.00
    NCI-H322M −4.89 >−4.00 >−4.00
    NCI-H460 −6.60 >−4.00 >−4.00
    NCI-H522 −4.48 >−4.00 >−4.00
    Colon Cancer
    COLO 205 −5.32 >−4.00 >−4.00
    HCC-2998 −6.63 6.15 −4.68
    HCT-116 −6.94 >−4.00 >−4.00
    HCT-15 −4.73 >−4.00 >−4.00
    HT29 >−4.00 >−4.00 >−4.00
    KM12 −4.49 >−4.00 >−4.00
    SW-620 >−4.00 >−4.00
    >−4.00 >−4.00
    CNS Cancer
    SF-268 −4.72 >−4.00 >−4.00
    SF-295 −4.67 −4.16 >−4.00
    SF-539 −4.29 >−4.00 >−4.00
    SNB-19 −4.66 −4.02 >−4.00
    SNB-75 −4.86 −4.24 >−4.00
    U251 −7.24 −4.59 >−4.00
    Melanoma
    LOX IMVI −4.71 >−4.00 >−4.00
    MALME-3M −4.46 >−4.00 >−4.00
    M14 −4.55 >−4.00 >−4.00
    SK-MEL-2 −4.61 >−4.00 >−4.00
    SK-MEL-28 −4.35 >−4.00 >−4.00
    SK-MEL-5 −4.33 >−4.00 >−4.00
    UACC-257 −7.58 −7.08 −6.51
    UACC-62 <−8.00 −7.55 −4.07
    Ovarian Cancer
    IGROV1 −6.79 −6.25 >−4.00
    OVCAR-3 −7.72 −4.67 >−4.00
    OVCAR-4 −6.92 −4.57 >−4.00
    OVCAR-5 −7.35 −6.24 >−4.00
    OVCAR-8 −5.35 >−4.00 >−4.00
    SK-OV-3 −4.90 >−4.00 >−4.00
    Renal Cancer
    786-0 −4.87 >−4.00 >−4.00
    A498 −6.84 −6.53 −6.03
    ACHN −4.64 >−4.00 >−4.00
    RXF-393 −4.79 −4.37 >−4.00
    SN12C >−4.00 >−4.00 >−4.00
    TK-10 −7.20 −6.38 −4.27
    UO-31 −4.45 >−4.00 >−4.00
    Prostate Cancer
    PC-3 −4.21 >−4.00 >−4.00
    DU-145 −4.54 >−4.00 >−4.00
    Breast Cancer
    MCF7 −6.09 >−4.00 >−4.00
    MCF7/ADR-RES −4.88 −4.22 >−4.00
    HS 578T −4.63 >−4.00 >−4.00
    MDA-MB-435 −4.55 −4.06 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 −4.00 >−4.00 >−4.00
    T-47D −4.74 −4.39 −4.04
    −5.77 >−4.00 >−4.00
    MG_MID
    Delta −5.17 −4.42 −4.13
    Range 2.83 3.13 2.38
    4.00 3.55 2.51
    NSC: 675345
    Figure US20050203077A1-20050915-C00075
    Leukemia
    CCRF-CEM >−4.00 >−4.00 >−4.00
    HL-60(TB) >−4.00 >−4.00 >−4.00
    K-562 −4.51 >−4.00 >−4.00
    MOLT-4 >−4.00 >−4.00 >−4.00
    RPMI-8226 >−4.00 >−4.00 >−4.00
    SR >−4.00 >−4.00 >−4.00
    Non-Small Cell Lung Cancer
    A549/ATCC >−4.00 >−4.00 >−4.00
    EKVX >−4.00 >−4.00 >−4.00
    HOP-62 −4.32 >−4.00 >−4.00
    HOP-92 −4.84 −4.26 >−4.00
    NCI-H226 −6.24 −5.61 −5.07
    NCI-H23 −4.66 >−4.00 >−4.00
    NCI-H322M >−4.00 >−4.00 >−4.00
    NCI-H460 −6.09 >−4.00 >−4.00
    NCI-H522 −4.07 >−4.00 >−4.00
    Colon Cancer
    COLO 205 −4.47 >−4.00 >−4.00
    HCC-2998 −5.63 −5.16 −4.13
    HCT-116 −5.39 >−4.00 >−4.00
    HCT-15 >−4.00 >−4.00 >−4.00
    HT29 >−4.00 >−4.00 >−4.00
    KM12 >−4.00 >−4.00 >−4.00
    SW-620 −4.42 >−4.00 >−4.00
    CNS Cancer
    SF-268 >−4.00 >−4.00 >−4.00
    SF-295 −4.41 >−4.00 >−4.00
    SF-539 >−4.00 >−4.00 >−4.00
    SNB-19 −4.50 >−4.00 >−4.00
    SNB-75 −4.28 >−4.00 >−4.00
    U251 −5.64 −4.67 −4.05
    Melanoma
    LOX IMVI >−4.00 >−4.00 >−4.00
    MALME-3M >−4.00 >−4.00 >−4.00
    M14 >−4.00 >−4.00 >−4.00
    SK-MEL-2 −4.11 >−4.00 >−4.00
    SK-MEL-28 >−4.00 >−4.00 >−4.00
    SK-MEL-5 >−4.00 >−4.00 >−4.00
    UACC-257 −5.86 >−4.00 −5.24
    UACC-62 −6.46 −5.55 >−4.00
    Ovarian Cancer
    IGROVI −5.36 −4.64 >−4.00
    OVCAR-3 −5.66 >−4.00 −5.39
    OVCAR-4 >−4.00 >−4.00
    OVCAR-5 −5.81 −5.27 >−4.00
    OVCAR-8 >−4.00 >−4.00 >−4.00
    SK-OV-3 −4.26 >−4.00 >−4.00
    Renal Cancer
    786-0 −4.96 >−4.00 >−4.00
    A498 −6.32 −5.81 −5.39
    ACHN −4.11 >−4.00 >−4.00
    CAKI-1 −4.49 >−4.00 >−4.00
    RXF-393 >−4.00 >−4.00 >−4.00
    SN12C −6.59 −5.78 >−4.00
    TK-10 >−4.00 >−4.00 >−4.00
    UO-31
    Prostate Cancer
    PC-3 >−4.00 >−4.00 >−4.00
    DU-145 >−4.00 >−4.00 >−4.00
    Breast Cancer
    MCF7 −6.14 −5.25 >−4.00
    MCF7/ADR-RES >−4.00 >−4.00 >−4.00
    MDA-MB-231/ATC −4.74 >−4.00 >−4.00
    HS 578T −4.17 >−4.00 >−4.00
    MDA-MB-435 −4.23 >−4.00 >−4.00
    MDA-N >−4.00 >−4.00 >−4.00
    BT-549 >−4.00 >−4.00 >−4.00
    T-47D −4.10 >−4.00 >−4.00
    MG_MID
    Delta −4.75 −4.21 −4.07
    Range 2.03 1.61 1.32
    2.59 1.81 1.39
  • EXAMPLE 54
  • Inhibition of Protein Kinase C
  • The Protein Kinase C (PKC) screening assay utilized in the following experiments is similar to standard PKC assays used by many investigators. Its primary features are that 1) the assay utilizes a 50:50 mixture of recombinant mouse PKC∝ and mouse PKCβ2; 2) employs histone as phosphate-accepting substrate; and 3) the PKC enzymatic activity is activated with phosphatidylserine, TPA and low concentration of calcium, so that both calcium and TPA are somewhat limiting for the extent of activation. In this manner the assay is sensitive to inhibitors of PKC activation. A more detailed description of the assay is provided in the following paragraphs.
  • The recombinant PKC formulation is a mixture (equal parts by activity) of mouse PKC∝ and mouse PKCβ2. The enzymes are expressed in Sf9 insect cells from recombinant baculovirus and partially purified on DEAE-cellulose and Sephacryl 200 gel filtration. Sufficient PKC is added to each reaction to provide approximately 4 pmols phosphate transferred in 30 minute (per total reaction.) The reaction is linear over the time when 4 pmols of phosphate is transferred and the reaction remains linear well beyond this time frame.
  • The PKC screening assay is performed in 96 well polystyrene U bottom micro titer plates, in a total reaction volume of 50 ul. Solution manipulations are performed during the assay utilize a Rainin, motorized EDP-plus M8 eight-channel micropipettor.
  • Samples were typically assayed at three dilutions, however some highly active pure compounds were assayed at six dilutions. Assay samples are dissolved in DMSO at a concentration of 10 mg/ml or less for samples suspected of being more potent. In some cases 50% DMSO:water, water, or methanol is substituted (if essential) for the solvent. At least 25 μl of the highest concentration sample to be assayed is transferred to a well in a 96 well U-bottom polystyrene assay plate. Serial 5-fold or 10-fold dilutions (depending on the dose-range desired) are made using the EDP-plus M8 eight-channel pipettor in dilute mode and mixing by repipetting. Using the 8-channel pipettor, 2 μl of each dilution is transferred to the appropriate wells of the plate(s) to be used for each assay. Duplicate assays are performed for each dose, with each assay, allowing six wells (half the row) for three-dose assays, or 12 wells (the whole row) for six-dose assays. In general, extracts and fractions are assayed at three doses: 400, 40 an 4 ug/ml, while pure compounds are tested at six doses: 400, 80, 16, 3.2, 0.64, 0.128 (8-fold series). The results of these experiments are shown in Table 2.
    TABLE 2
    Inhibition of Protein Kinase C
    Compounds, NSC # Structure IC50 (μg/ml)
    675347
    Figure US20050203077A1-20050915-C00076
    1 × 100
    675346
    Figure US20050203077A1-20050915-C00077
    5 × 10−1
    675345
    Figure US20050203077A1-20050915-C00078
    9 × 10−1
    675344
    Figure US20050203077A1-20050915-C00079
    5 × 101
    675343
    Figure US20050203077A1-20050915-C00080
    3 × 101
    676628
    Figure US20050203077A1-20050915-C00081
    1 × 100
    676629
    Figure US20050203077A1-20050915-C00082
    2 × 10−1
    676630
    Figure US20050203077A1-20050915-C00083
    9 × 10−1
    676631
    Figure US20050203077A1-20050915-C00084
    2 × 100
    676632
    Figure US20050203077A1-20050915-C00085
    5 × 101
    676633
    Figure US20050203077A1-20050915-C00086
    6 × 100
    676634
    Figure US20050203077A1-20050915-C00087
    8 × 10−1
    676635
    Figure US20050203077A1-20050915-C00088
    8 × 100
    674973
    Figure US20050203077A1-20050915-C00089
    7 × 100
    675244
    Figure US20050203077A1-20050915-C00090
    1 × 100
    675245
    Figure US20050203077A1-20050915-C00091
    1 × 100
    675246
    Figure US20050203077A1-20050915-C00092
    3 × 100
    675247
    Figure US20050203077A1-20050915-C00093
    1 × 101

Claims (22)

1. A method for treating a patient having a tumor, said method comprising the step of administering to the patient an effective amount of a compound of the formula
Figure US20050203077A1-20050915-C00094
wherein
R1 and R2 are independently selected from the group consisting of
Figure US20050203077A1-20050915-C00095
H, CHO, CH2OH, and CH2NH2; and
X and Y are independently selected from the group consisting of Se, S, O, NR, and NH, where R is H or C1-C7 alkyl;
R3, R4, R5, and R6 are each independently selected from the group consisting of H, CHO, CH2OH, and CH2NH2; and when any one of R1, R2, R3, R4, R5, and R6 is CH2NH2, the pharmaceutically acceptable salts thereof,
with the proviso that R1 and R2 are not both
Figure US20050203077A1-20050915-C00096
2. The method of claim 1 wherein the administering step includes the compound where R1 and R2 are each H.
3. The method of claim 1 wherein the administering step includes the compound where R3 and R6 are each independently selected from the group consisting of CHO, CH2OH, and CH2NH2.
4. The method of claim 3 wherein the administering step includes the compound where R1 and R2 are each H.
5. The method of claim 1 wherein the administering step includes the compound where at least one of R1 and R2 is selected from the group consisting of CHO, CH2OH, and CH2NH2.
6. The method of claim 1 wherein the administering step includes the compound where at least one of R1, R2, R3, R4, R5, and R6 is selected from the group consisting of CHO, CH2OH, and CH2NH2.
7. The method of claim 1 wherein the administering step includes the compound where one of R1 and R2 is selected from the group consisting of
Figure US20050203077A1-20050915-C00097
8. The method of claim 7 wherein the administering step includes the compound where at least one of R3, R4, R5, and R6 is selected from the group consisting of CHO, CH2OH, and CH2NH2.
9. The method of claim 7 wherein the administering step includes the compound where each of R3, R4, and R6 is H.
10. The method of claim 7 wherein the administering step includes the compound where the other of R1 and R2 is selected from the group consisting of CHO, CH2OH, and CH2NH2.
11. The method of claim 7 wherein the administering step includes the compound where X is Se.
12. The method of claim 1 wherein the administering step includes the compound where one of R1 and R2 is
Figure US20050203077A1-20050915-C00098
13. The method of claim 12 wherein the administering step includes the compound where the other of R1 and R2 is selected from the group consisting of CHO, CH2OH, and CH2NH2.
14. The method of claim 12 wherein the administering step includes the compound where the R5 is selected from the group consisting of CHO, CH2OH, and CH2NH2.
15. The method of claim 12 wherein the administering step includes the compound where X is Se.
16. The method of claim 1 wherein the administering step further comprises a pharmaceutically acceptable carrier therefor.
17. The method of claim 1 wherein the administering step includes the compound as a complex with a cyclodextrin.
18. A process for preparing an intermediate compound of the formula
Figure US20050203077A1-20050915-C00099
the process comprising the step of reacting a compound of the formula
Figure US20050203077A1-20050915-C00100
with a compound of the formula
Figure US20050203077A1-20050915-C00101
in the presence of sodium cyanide and dimethylformamide;
wherein
X and Y are independently selected from the group consisting of Se, S, O, NR, and NH, where R is H or Cl-C7 alkyl; and
R1, R2, R3, R4, and R6 are each independently selected from the group consisting of H, CHO, CH2OH, and CH2NH2.
19. A process for preparing a compound of the formula
Figure US20050203077A1-20050915-C00102
the process comprising the step of reacting a compound of the formula
Figure US20050203077A1-20050915-C00103
with a compound of the formula
Figure US20050203077A1-20050915-C00104
in the presence of sodium cyanide and dimethylformamide to form an intermediate of the formula
Figure US20050203077A1-20050915-C00105
wherein
X, Y, and Z are each independently selected from the group consisting of Se, S, O, NR, and NH, where R is H or C1-C7 alkyl; and
R1, R2, R3, R4, and R6 are each independently selected from the group consisting of H, CHO, CH2OH, and CH2NH2.
20. The process of claim 19 further comprising the step of reacting the intermediate of the formula
Figure US20050203077A1-20050915-C00106
with RNH2Cl in the presence of NaOAc, when Z is NR; with (CH3CO)2O in the presence of HCl, when Z is O; with [(C6H11)3Sn]2S in the presence of BCl3, when Z is S; or with [(C6H11)3Sn]2Se in the presence of BCl3, when Z is Se.
21. The process of claim 20 wherein R1, R2, R3, R4, and R6 are each H.
22. The process of claim 21 further comprising the step of reacting the compound of the formula
Figure US20050203077A1-20050915-C00107
with (a) base, and (b) dimethylformamide, to form a compound of the formula
Figure US20050203077A1-20050915-C00108
wherein R1 is selected from the group consisting of H and CHO.
US11/103,991 1996-06-03 2005-04-12 Selenophene anti-tumor agents Abandoned US20050203077A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/103,991 US20050203077A1 (en) 1996-06-03 2005-04-12 Selenophene anti-tumor agents
US11/593,371 US20070197493A1 (en) 1996-06-03 2006-11-06 Selenophene anti-tumor agents

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US1909596P 1996-06-03 1996-06-03
PCT/US1997/009717 WO1997046225A1 (en) 1996-06-03 1997-06-03 Selenophene anti-tumor agents
US18051498A 1998-11-11 1998-11-11
US10/061,480 US6620804B2 (en) 1996-06-03 2002-02-01 Selenophene anti-tumor agents
US10/658,175 US6894039B2 (en) 1996-06-03 2003-09-09 Selenophene anti-tumor agents
US11/103,991 US20050203077A1 (en) 1996-06-03 2005-04-12 Selenophene anti-tumor agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/658,175 Division US6894039B2 (en) 1996-06-03 2003-09-09 Selenophene anti-tumor agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/593,371 Continuation US20070197493A1 (en) 1996-06-03 2006-11-06 Selenophene anti-tumor agents

Publications (1)

Publication Number Publication Date
US20050203077A1 true US20050203077A1 (en) 2005-09-15

Family

ID=26691840

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/061,480 Expired - Fee Related US6620804B2 (en) 1996-06-03 2002-02-01 Selenophene anti-tumor agents
US10/658,175 Expired - Fee Related US6894039B2 (en) 1996-06-03 2003-09-09 Selenophene anti-tumor agents
US11/103,991 Abandoned US20050203077A1 (en) 1996-06-03 2005-04-12 Selenophene anti-tumor agents
US11/593,371 Abandoned US20070197493A1 (en) 1996-06-03 2006-11-06 Selenophene anti-tumor agents

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/061,480 Expired - Fee Related US6620804B2 (en) 1996-06-03 2002-02-01 Selenophene anti-tumor agents
US10/658,175 Expired - Fee Related US6894039B2 (en) 1996-06-03 2003-09-09 Selenophene anti-tumor agents

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/593,371 Abandoned US20070197493A1 (en) 1996-06-03 2006-11-06 Selenophene anti-tumor agents

Country Status (1)

Country Link
US (4) US6620804B2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090163494A1 (en) * 2007-12-19 2009-06-25 Pao-Chiung Hong Protein kinase inhibitors
WO2009085040A1 (en) * 2007-12-27 2009-07-09 Dcb-Usa Llc Protein kinase inhibitors

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060040909A1 (en) * 2004-08-23 2006-02-23 Development Center For Biotechnology Selenophene compounds
US7803799B2 (en) * 2006-07-07 2010-09-28 National Health Research Institutes Selenophene compounds
TWI408136B (en) * 2006-10-02 2013-09-11 Nat Health Research Institutes Thiophene compounds and pharmaceutical composition using the same
TWI339205B (en) * 2006-10-02 2011-03-21 Nat Health Research Institutes Pyrazole compounds and pharmaceutical composition
EP2468747B1 (en) * 2006-12-07 2014-09-03 China Medical University 2-Selenophene-4-quinolones as anticancer agents
TWI434686B (en) * 2008-11-03 2014-04-21 Nat Health Research Institutes Imidazol-4-one and imidazole-4-thione compounds
CA2759519A1 (en) * 2009-04-27 2010-11-04 Kasina Laila Innova Pharmaceuticals Private Limited Anti-cancer drugs, and uses relating for malignant melanoma and other cancers
ES2652363T3 (en) 2010-08-18 2018-02-01 Samumed, Llc Dicetonas and hydroxyketones as activators of the catenin signaling pathway
CN103221048A (en) 2010-11-18 2013-07-24 卡斯纳莱拉创新药物私人有限公司 Substituted 4-(selenophen-2(or 3)-ylamino)pyrimidine compounds and methods of use thereof
US9090633B2 (en) 2010-11-18 2015-07-28 Kasina Laila Innova Pharmaceuticals Private Limited Substituted 4-(arylamino) selenophenopyrimidine compounds and methods of use thereof
RU2680716C2 (en) * 2013-02-22 2019-02-26 СЭМЬЮМЕД, ЭлЭлСи Gamma-diketones as wnt/beta-catenin signaling pathway activators
CN104130252B (en) * 2014-07-15 2016-09-14 南开大学 Organic photoelectric compound and its preparation method and application
EP3206686B1 (en) 2014-08-20 2019-10-09 Samumed, LLC Gamma-diketones for treatment and prevention of aging skin and wrinkles

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5578636A (en) * 1994-04-08 1996-11-26 Purdue Research Foundation Polythiophene anti-tumor agents

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5578636A (en) * 1994-04-08 1996-11-26 Purdue Research Foundation Polythiophene anti-tumor agents

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090163494A1 (en) * 2007-12-19 2009-06-25 Pao-Chiung Hong Protein kinase inhibitors
WO2009085040A1 (en) * 2007-12-27 2009-07-09 Dcb-Usa Llc Protein kinase inhibitors

Also Published As

Publication number Publication date
US20070197493A1 (en) 2007-08-23
US20030028015A1 (en) 2003-02-06
US6620804B2 (en) 2003-09-16
US6894039B2 (en) 2005-05-17
US20040063662A1 (en) 2004-04-01

Similar Documents

Publication Publication Date Title
US20050203077A1 (en) Selenophene anti-tumor agents
Liang et al. Exploration and synthesis of curcumin analogues with improved structural stability both in vitro and in vivo as cytotoxic agents
Tozkoparan et al. 6-benzylidenethiazolo [3, 2-b]-1, 2, 4-triazole-5 (6h)-onessubstituted with ibuprofen: Synthesis, characterizationand evaluation of anti-inflammatory activity
Rostom et al. Synthesis of some pyrazolines and pyrimidines derived from polymethoxy chalcones as anticancer and antimicrobial agents
EP0845991B1 (en) Use of phenol substituted diphosphonates as antineoplastic agents
Cocco et al. Synthesis and antitumour activity of 4-hydroxy-2-pyridone derivatives
Andreani et al. Potential antitumor agents. Part 291: synthesis and potential coanthracyclinic activity of imidazo [2, 1-b] thiazole guanylhydrazones
US6337346B1 (en) Naphtho- and dihydrobenzo-thiophene derivatives as cytotoxic antitumor agents
EP1021178B1 (en) Selenophene anti-tumor agents
Chen et al. Synthesis and Cytotoxicity Evaluation of Certain α‐Methylidene‐γ‐butyrolactones Bearing Coumarin, Flavone, Xanthone, Carbazole, and Dibenzofuran Moieties
Tzeng et al. Synthesis and Cytotoxic evaluation of a series of γ-substituted γ-aryloxymethyl-α-methylene-γ-butyrolactones against cancer cells
Tantawy et al. Structure-based design, synthesis, and biological evaluation of novel piperine–resveratrol hybrids as antiproliferative agents targeting SIRT-2
US5994394A (en) Polyheterocyclic compounds
US20060040909A1 (en) Selenophene compounds
US5741811A (en) Polythiophene anti-tumor agents
JPS61158961A (en) Polycyclic vermicidal compounds, manufacture and drug containing them
US6046229A (en) Polyaryl antitumor agents
KR20070095016A (en) Pyridine derivatives, a method for preparation thereof and a composition containing the same for treating and preventing cancer disease
CN103848833B (en) Pyrrolo-triazine analog derivative, its preparation method and in application pharmaceutically
US7750181B2 (en) Antiproliferative agents
PL188075B1 (en) Novel water-soluble c-ring analogues of 20 (s)- camptotecin
JP2001506624A (en) Polythiophene antitumor agent
JPH1180152A (en) Polyheterocyclic compound
US4866070A (en) Acenaphtho[1,2b]quinolinylmethylamino derivatives and pharmaceutical compositions containing them
KR101263263B1 (en) 2,4-Diaminoquinazoline compounds having anticancer activity

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:PURDUE UNIVERSITY;REEL/FRAME:021734/0301

Effective date: 20060630