[go: up one dir, main page]

WO2004066994A1 - Anticancer agent - Google Patents

Anticancer agent Download PDF

Info

Publication number
WO2004066994A1
WO2004066994A1 PCT/JP2004/000680 JP2004000680W WO2004066994A1 WO 2004066994 A1 WO2004066994 A1 WO 2004066994A1 JP 2004000680 W JP2004000680 W JP 2004000680W WO 2004066994 A1 WO2004066994 A1 WO 2004066994A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cancer
optionally substituted
compound
anticancer agent
Prior art date
Application number
PCT/JP2004/000680
Other languages
French (fr)
Japanese (ja)
Inventor
Shinya Kimura
Taira Maekawa
Original Assignee
Kansai Technology Licensing Organization Co. Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kansai Technology Licensing Organization Co. Ltd. filed Critical Kansai Technology Licensing Organization Co. Ltd.
Priority to JP2005504703A priority Critical patent/JPWO2004066994A1/en
Publication of WO2004066994A1 publication Critical patent/WO2004066994A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to an anticancer agent and a method for treating cancer.
  • P-glycoprotein P-glycoprotein
  • cells become multidrug-resistant
  • the administration of an anticancer drug to such a patient, who often acquires MDR has a problem in that the drug concentration in the cancer cells is reduced and an effective effect cannot be obtained.
  • An object of the present invention is to provide an anticancer agent having a mechanism different from the conventional one, effective for multidrug-resistant cancer cells, and having a large safety margin.
  • Another object of the present invention is to provide a method for treating cancer.
  • FIG. 1 shows the structures of 15 compounds obtained from Calophyllmn Brasiliensis.
  • FIG. 2 shows the results of examining the inhibitory effect of 15 compounds shown in FIG. 1 on the growth of BV173 cells at concentrations of ⁇ , M, 5 ⁇ , ⁇ , and 30 ⁇ ! .
  • FIG. 3 shows the results of measuring the cytostatic effect of compound 7 (GUT-70) on five types of leukemia cell lines (BV173, SEM, NALM6, HL60, and K562).
  • indicates 0 ⁇ M
  • (translation) is 0.5 ⁇
  • (mouth) is 1.0 ⁇
  • (5.0) is 5.0 ⁇
  • (X) is the measurement result at 10 10. .
  • FIG. 4 shows the results of morphological observation of apoptosis by compound 7.
  • FIG. 4 ⁇ shows the observation results before treatment with compound 7, and
  • FIG. 4 ⁇ shows the observation results after treatment with compound 7.
  • FIG. 5 shows the results of measuring the relationship between cell cycle and apoptosis using TUNEL Atssei.
  • Fig. 5 ⁇ shows the results for the induction of apoptosis when GUT-70 was not treated and Fig. 5 ⁇ shows the results when GUT-70 was treated for 5 ⁇ for 24 hours.
  • Figure 6 shows the leukemia cell line NALM6 cells and BV173 cells treated with compound 7, was examined the temporal change in the P21 WAF./CIP. ⁇ P27 K IPL, p53 and p57 results.
  • FIG. 7A shows the results of comparing the resistance of daunorubicin (DNR) to the P-glycoprotein high-expressing cell K562 / D1-9 and the parent strain 562 cells that do not express P-glycoprotein.
  • FIG. 7B shows the results of comparing the resistance of compound 7 to the P-glycoprotein overexpressing cell K562 / D1-9 and the parent strain K562 cell not overexpressing P-glycoprotein.
  • DNR daunorubicin
  • FIG. 8 shows the results of examining the toxicity of compound 7 on normal cells using normal hematopoietic progenitor cells CFU-GM and BFU-E.
  • FIG. 9 shows the results of examining the effect of compound 7 (GUT-70) on human small cell lung cancer.
  • the inventor of the present invention has made intensive studies with the main object of solving the above problems. As a result, a compound having a specific structure has a remarkable inhibitory effect on the growth of cancer cells. They also found that they are effective against drug-resistant cancer cells, and that they have a large safety margin. The present inventors have made further studies and completed the present invention.
  • the present invention relates to the following anticancer agents and methods for treating cancer.
  • R 3 and R 4 each independently represent a hydrogen atom, a halogen atom, a cyano group, a hydroxy group, an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a propyloxyl group, an alkoxy group, A carboxyl group, an amino group, a monoalkylamino group, a dialkylamino group or a quinoline: a diamino group;
  • R 5 , R 8 and R 9 each independently represent a hydrogen atom, a halogen atom, a cyano group, a hydroxy group, an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a carboxyl group , Alkoxycarbonyl group, optionally substituted aryloxy group, thiol group, alkylthio group, optionally substituted arylthio group, alkylsulfonyl group, optionally substituted arylsulfur group, alkylaminocarbonyl Group, an optionally substituted arylaminosulfonyl group, an alkylaminosulfoyl group, an optionally substituted arylaminosulfonyl group, an alkoxycarbonyl group, an optionally substituted aryloxycarbonyl Group, amino group, monoalkylamino group, dialquinoleamino group, acylamino
  • R 6 represents an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a carboxyl group, an alkoxycarbol group, or a compound represented by the following general formula (2)
  • R 7 represents 0, NH or NR 13 ;
  • R 1 Represents a hydrogen atom, an alkyl group, a cycloalkyl group or an aryl group which may be substituted;
  • R u and R 12 each independently represent a hydrogen atom, an alkyl group, a cycloalkyl group, or a substituted or unsubstituted aryl group;
  • R 13 represents an alkyl group, a cycloalkyl group or an aryl group which may be substituted) or a pharmaceutically acceptable salt thereof as an active ingredient.
  • anticancer agent according to claim 1 which is R 5 force S methoxy group.
  • R 9 is an alkyl group or an optionally substituted aryl group.
  • R 6 is the following general formula (2)
  • R 1G , R 11 and R 12 are as defined above.
  • a method for treating cancer which comprises administering an effective amount of the compound according to any one of Items 1 to 7.
  • the cancer is acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, soft tissue tumor, small cell lung cancer, chronic myelogenous leukemia, medullary thyroid carcinoma, osteosarcoma, head and neck Item 9.
  • cancer selected from the group consisting of acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, chronic myeloid leukemia, and small cell lung cancer.
  • the present invention relates to an anticancer agent preferably containing a compound represented by the general formula (3) as an active ingredient, and a method for treating cancer by administering the compound to a cancer patient.
  • the present invention relates to an anticancer agent containing a compound represented by the formula (3) as an active ingredient. Further, the present invention preferably relates to a method for treating cancer, which comprises administering an anticancer effective amount of a compound represented by the general formula (3).
  • acute antimyelogenous leukemia acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, characterized in that an anticancer effective amount of the compound represented by the general formula (3) is administered.
  • Soft tissue tumor small cell lung cancer, chronic myeloid leukemia, medullary thyroid cancer, osteosarcoma, head and neck cancer, esophageal cancer, non-small cell lung cancer, colorectal cancer, gastric cancer, biliary tract cancer, brain tumor, malignant melanoma, kidney
  • the present invention relates to a method for treating a disease selected from the group consisting of cancer, victory cancer, and liver cancer.
  • the active ingredient of the anticancer agent of the present invention has a structure represented by the following general formula (1).
  • the alkyl group includes a substituted or unsubstituted linear or branched one, and usually has 1 to 18, preferably 1 to 8, and more preferably 1 to 3, for example, Examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutynole, tert-butynole, n-pentyl, isopentyl, hexyl, heptyl, octyl, nonyl and decyl.
  • the cycloalkyl group may be substituted or unsubstituted, and the number of carbon atoms is not particularly limited, but is usually 3 to 10, preferably 3 to 7. Specific examples of the cycloalkyl group include cyclopropanol, cyclobutyl, cyclopentyl, cyclohexynole, and cycloheptyl.
  • the term "aryl group” means a monocyclic or polycyclic group composed of a 5- or 6-membered aromatic hydrocarbon ring, and specific examples include phenyl, naphthyl, fluorenyl, and anthrenyl. , Biphenyl, tetrahydronaphthyl, chromanyl, 2,3-dihydro-1,4-dioxanaphthalenyl, indanyl and phenanthryl.
  • the number of substituents of the aryl group having a substituent is 1 to 3, preferably 1 or 2, and the substituent includes methyl, ethyl, methoxy, ethoxy, amino, methylamino, dimethinoleamino, cyano, nitro, Examples include fluorine, chlorine, bromine, trifluoromethyl, hydroxy, carboxyl, methoxycarbonyl, ethoxycarbol, propoxylproponyl, butoxycarbonyl, CONH2, acetyl.
  • substituted aryl group examples include phenyl, 2-, 3- or 4-fluorophenyl, 2-, 3- or 4-chlorophenol, 2_, 3- or 4-bromophenyl, 2,4- Difluorophenyl, 2, 3_, 2, 4-, 3, 4-, 3, 5- or 2,6-dichlorophenyl, 3-chloro-1-phenylphenyl, 4-isopropylphenyl, 2,6- Dimethynophenyl, 2-, 3- or 4-trifluoromethylphenyl, 4-methoxyphenyl, 4-trifluoromethoxyphenyl, 2-, 3- or 4-dimethylaminophenyl, 2_ , 3- or 4-1-trophenyl, and 4-sulfamoylphenyl.
  • alkoxy group examples include groups represented by ⁇ R ′ (R ′ is the above-mentioned alkyl group).
  • R ′ is the above-mentioned alkyl group.
  • acyl group examples include a group represented by one COR ′ ′ (R ′′ is the above-mentioned alkyl group or the above-mentioned optionally substituted aryl group), and examples thereof include acetyl, propionyl, and benzoyl. Is done.
  • halogen atom examples include fluorine, chlorine, bromine, and iodine.
  • Examples of the asinoleamino group include a group represented by one NHCOR ′ ′ (R ′′ is the above-mentioned alkyl group or the above-mentioned optionally substituted aryl group).
  • R ′′ is the above-mentioned alkyl group or the above-mentioned optionally substituted aryl group.
  • alkoxycarbonyl group examples include a group represented by one CO ⁇ R ′ (R ′ is the aforementioned alkyl group), for example, methoxycarbyl, ethoxycarbonyl, n-propoxycarbinole, isopropoxycarbonyl, Examples are n-butoxycarbonyl, isobutoxycarbonyl, sec-butoxycarbonyl and t-butoxycarbonyl. You may be replaced!
  • Examples of the / aryloxy group include a group represented by -0- (substituted or substituted aryl group), and examples include phenyloxy and naphthyloxy.
  • alkylthio group examples include a group represented by -S- (alkyl group), and examples thereof include methylthio, ethylthio, n -propynolethio, isopropylthio, n-butylthio, isobutylthio, and tert-butylthio.
  • Examples of the / and arylthio groups include groups represented by -S- (substituted or substituted aryl groups), such as phenylthio and naphthylthio.
  • Examples of the monoalkylamino group include an amino group mono-substituted with the above alkyl, such as methylaminocarbonyl, ethylaminocarbonyl, n-propylaminocarbonyl, isopropylaminocarbonyl, n-butylaminocarbonyl, and isobutylamino.
  • an amino group mono-substituted with the above alkyl such as methylaminocarbonyl, ethylaminocarbonyl, n-propylaminocarbonyl, isopropylaminocarbonyl, n-butylaminocarbonyl, and isobutylamino.
  • Examples of the dianolequinole amino group include amino groups di-substituted with the above alkyl, such as dimethylaminocarbonyl, getylaminocarbonyl, di-n-propylaminocarbonyl, diisopropylaminocarbonyl, di-n-butylaminoamino, Examples thereof include diisobutylaminocarbonyl, ditert-butylaminocarbonyl, di-n-pentylaminocarbonyl, diisopentylaminocarbonyl, and dihexylaminocarbonyl.
  • the alkylsulfonyl group, - S0 2 - include groups represented by (alkyl), for example, methylcarbamoyl Roh less Honoré Honi Honoré, E Chino less Honoré Honi Honoré, n- pro Pinot less Honoré Honi Honoré, isopropylidene Roh-less And norfoninole, n-butylsulfonyl, isoptylsulfonyl, and tert-butylsulfonyl.
  • the optionally substituted ⁇ reel sulfonyl group, _S0 2 - include groups represented by (optionally substituted ⁇ aryl group), for example Fuenirusuruho - Le include naphthylsulfonyl El.
  • alkylaminocarbo group examples include an aminocarbonyl group mono- or di-substituted with the alkyl.
  • the amino group mono-substituted with the alkyl include methylaminocanoleponinole, ethylaminocarbonyl, and the like. n-propylaminocarbonyl, isopropylaminocarboel, n-butylaminocarbonyl, isobutylaminocarbonyl, tert-butylaminocarbonyl, n-pentylaminocarbonyl, isopentylaminocarbonyl, and hexinoleaminocarbonyl.
  • amino-disubstituted di-substituted alkyl Dimethylaminopropyl, diethylaminocarbonyl, di-n-propylaminocarbonyl, diisopropylaminocarbonyl, di-n-butylaminocarbonyl, diisobutylaminocarbonyl, di-tert-butylaminocarbonyl, di-n-pentyla Examples include minocarbonyl, diisopentylaminocarbonyl, and dihexylaminocarbonyl.
  • optionally substituted arylaminocarbonyl group examples include a group represented by -CONH- (optionally substituted arylylcarbonyl), such as phenylaminoaminocarbonyl and naphthylaminocarbonyl. No.
  • alkylaminosulfonyl group examples include an aminosulfonyl group mono- or di-substituted with the alkyl.
  • amino group mono-substituted with the alkyl examples include methylaminosulfonyl, ethylaminosulfonyl, and n-propylamino.
  • Sulfonyl isopropylamino sulfoel, n-butylaminosulfonyl, isobutylaminosulfonyl, tert-butynoleaminosulfonyl, n-pentylaminosulfonyl, isopentylaminosulfonyl, hexylaminosulfonyl, and the aminosulfonyl di-substituted with the alkyl.
  • methyl group examples include dimethylaminosulfonyl, getylaminosulfonyl, di-n-propylaminosulfonyl, diisopropylaminosulfonyl, di-n-butylaminosulfonyl, Two Honoré, di tert- heptyl aminosulfonyl, di n- pentyl aminosulfonyl, Jiisopenchirua Minosuruhoniru include carboxymethyl Honoré aminosulfonyl to di.
  • substituents on these ⁇ reel aminosulfonyl group, - S0 2 NH- include groups represented by (substituted Moyoi Ariru group), like for example phenylalanine amino sulfonyl, naphthyl aminosulfonyl Can be
  • the substituted or unsubstituted aryloxycarbonyl group includes a group represented by -COO- (substituted or unsubstituted aryl group), for example, phenylcarboxy group. And naphthyloxycarbonyl.
  • Alkylsulfonyl ⁇ amino group, - NHS0 2 - et include groups represented by (alkyl) is, for example, methylsulfonyl ⁇ Mino, E chill sulfonyl ⁇ amino, n- propylsulfonyl amino, isopropylsulfonyl - Ruamino, n-butylsulfonylamino-containing isobutylsulfonylamino, tert-butylsnolephonylamino, n-pentylsulfonylamino, isopentylsulfonynonamino, hexylsulfonylamino, Substituted by Les, be good Le, as a ⁇ reel sulfonyl ⁇ amino group, - NHS0 2 - include groups (! Substituted / ⁇ also good I Ariru group) represented by, e.g.
  • R 7 is preferably an oxygen atom ( ⁇ ).
  • R 5 is preferably a methoxy group (_OCH 3 ).
  • R 9 is preferably an alkyl group or an aryl group, particularly preferably an n-propyl group.
  • R 6 is a general formula (2)
  • R 1 (3 , R 11 and R 12 are as defined above)
  • the compound of the general formula (3) has a particularly excellent cancer cell growth inhibitory action.
  • Pharmaceutically acceptable salts of the compounds represented by the general formula (I) of the present invention may be any one of -R 6 , R 8 , R 9 in which a carboxyl group (COOH) or an amino group, a mono- or di-alkylamino group. If the compound has a phenyl group, a pharmaceutically acceptable salt can be prepared.
  • Examples of pharmaceutically acceptable salts of carboxy groups include alkali metal salts (sodium salt, potassium salt, lithium salt), and pharmaceutically acceptable salts of amino, mono- or dialkylamino groups.
  • Inorganic salts such as sulfate, hydrochloride, hydrobromide, lead nitrate and phosphate, fumarate, maleate, succinate, methanesulfonate, toluenesulfonate And other organic acid salts.
  • Such pharmaceutically acceptable salts can be obtained by reacting a compound of the present invention with an alkali metal carbonate or carbonate, an alkali metal hydroxide, or an inorganic or organic acid.
  • the above compounds can be appropriately prepared by various methods, may be chemically synthesized, or may be separated from an appropriate plant by extraction or the like.
  • R 8 and R 9 are as defined above.
  • R ⁇ R 5 , R 8 and R 9 are as defined above.
  • a compound having a structure wherein R 6 is represented by the general formula (2) is a compound represented by the following general formula (10) as a compound represented by the general formula (9).
  • the Friedel 'Crafts (Friede preparative Crafts) catalysts include, for example, A1C1 3, BF 3, ZnCl 2 , Sn Cl 4.
  • the compound of the present invention thus obtained is usually dissolved, extracted, separated, gradient, filtered, concentrated, thin-layer chromatography, column chromatography, gas chromatography, high-performance liquid chromatography, distillation, sublimation, crystal Purification is carried out as appropriate by using a method such as chemical conversion alone or in combination.
  • various conjugates can be obtained by using an intermediate having a desired substituent or by appropriately introducing an appropriate functional group.
  • R 1 and R 2 are methyl groups, R 3 and R 4 are hydrogen, and R 5 is —OCH 3 group, and in the general formula (6), R 8 is hydrogen
  • R 9 is a —CH 2 CH 2 CH 3 group
  • R 10 a compound in which R 1C) is hydrogen, R ′′ and R 12 is a methyl group
  • the compound serving as an active ingredient of the pharmaceutical composition of the present invention can be obtained by extracting an appropriate plant power.
  • the plant include a plant of the family Cucurbitaceae (Guttiferae).
  • plants of the family Tortaceae those belonging to the genus Calophyllum, such as Calophyllum Brasiliensis or Calophyllum inophyllum, are preferred, and Calophyllum Brasiliensis is particularly preferred.
  • the method for obtaining the compound by extracting the compound from the plant can be appropriately selected from known methods, and is not particularly limited.
  • a method in which the bark of a plant is powder-framed and extracted with an organic solvent is used. Can be.
  • the desired compound is obtained by performing appropriate separation and purification such as chromatography, filtration and extraction.
  • an appropriate organic solvent for example, a lower alcohol such as acetone, hexane, methanol, ethanol, etc.
  • the compound of the general formula (3) can be obtained by separating and purifying a plant extract of Calophyllum Brasiliensis (Guttiferae).
  • composition (pharmaceutical formulation)
  • the compound of the general formula (1) or a salt thereof is mixed with a pharmaceutically acceptable carrier, and solid preparations such as tablets, capsules, granules and powders; liquid preparations such as syrups and injections, and patches It can be appropriately formulated as a transdermal absorbent such as an ointment and a plaster, an inhalant, and a suppository.
  • the pharmaceutical composition of the present invention is orally or parenterally administered, and the above compounds may be used alone or in combination of two or more.
  • various organic or inorganic carrier substances commonly used as pharmaceutical materials can be used as the pharmaceutically acceptable carrier. Specifically, excipients, lubricants, binders, disintegrants in solid preparations, solvents, dissolution aids, suspending agents, isotonic agents, buffers, soothing agents, etc. in liquid preparations Can be blended. If necessary, pharmaceutical additives such as preservatives, antioxidants, coloring agents and sweeteners can also be used.
  • excipient examples include lactose, sucrose, D-mannitol, starch, crystalline cellulose, light caffeic anhydride and the like.
  • Preferred examples of the lubricant include, for example, magnesium stearate, calcium stearate, talc, colloidal silica and the like.
  • Suitable examples of the binder include, for example, crystalline cellulose, sucrose, D-mannitol, dextrin, hydroxypropylsenorellose, hydroxypropylmethylcellulose, polyvinylinolepyrrolidone and the like.
  • Preferred examples of the disintegrant include starch, carboxymethylcellulose, carboxymethylcellulose calcium, croscarmellose sodium, carboxymethyl starch sodium and the like.
  • Preferred examples of the solvent include water for injection, alcohol, propylene glycol, macrogol, sesame oil, corn oil and the like.
  • dissolution aid examples include, for example, polyethylene glycol, propylene dalicol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate and the like.
  • the suspending agent include surfactants such as stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glycerin monostearate; polybutyl alcohol , Polyvinylpyrrolidone, sodium carboxymethinolate cellulose, methinoresenololose, hydroxymethylsenorelose, Examples include hydrophilic polymers such as droxicetyl cellulose and hydroxypropyl cellulose.
  • surfactants such as stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glycerin monostearate
  • polybutyl alcohol Polyvinylpyrrolidone, sodium carboxymethinolate cellulose, methinoresenololose, hydroxymethylsenor
  • the tonicity agent include sodium chloride, glycerin, D-mannitol and the like.
  • buffers such as phosphate, acetate, carbonate, and citrate.
  • Preferable examples of the painless darting agent include, for example, benzyl alcohol.
  • Preferable examples of the preservative include, for example, paraoxybenzoic acid esters, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid and the like.
  • the antioxidant include, for example, sulfite, ascorbic acid and the like.
  • the anticancer agent of the present invention can be applied to the treatment of various cancers.
  • the type of cancer is not particularly limited, for example, acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, soft tissue tumor, small cell lung cancer, chronic myeloid leukemia, medullary thyroid cancer Osteosarcoma, head and neck cancer, esophageal cancer, non-small cell lung cancer, colorectal cancer, stomach cancer, biliary tract cancer, brain tumor, malignant melanoma, kidney cancer, knee cancer, and liver cancer.
  • the anticancer agent of the present invention is particularly effective for acute myeloid leukemia, acute lymphocytic leukemia, and small cell lung cancer. Among them, it is suitably used for acute myeloid leukemia, acute lymphocytic leukemia, and small cell lung cancer.
  • the administration conditions and administration method of the anticancer agent of the present invention can be appropriately set according to the type of the disease, the condition of the patient, the application site, and the like.
  • the dose is appropriately adjusted according to the type, size (or weight), symptoms, etc. of the animal.
  • the administration method can also be adjusted appropriately to obtain the optimal therapeutic effect.For example, it can be administered in several divided doses daily, and if a dangerous condition occurs during treatment, the dose may be adjusted according to the condition. And the number of administrations can be reduced as appropriate.
  • the anticancer agent of the present invention can be administered alone to obtain a desired effect. It can also be used in appropriate combination with an anticancer agent, a chemotherapeutic agent, an anti-inflammatory agent or an immunotherapeutic agent.
  • anticancer agents used in combination with the anticancer agent of the present invention include, for example, antimetabolites, alkylating agents, platinum-based anticancer agents, topoisomerase inhibitors, anticancer antibiotics, tyrosine kinase inhibitors, human Antibodies.
  • Human leukemia cells BV173, SUPB15, NAL6, HL60 and SEM were obtained from the American Type Culture Collection.
  • human small cell lung cancer cell lines SBC-1, SBC-3, SBC_5, 25N427, N69, LUT_130, H82, NCI345, LUT134B were used in the experiments.
  • Human leukemia cell lines BV173, SUPB15, NALM6, HL60 and SEM, and human small cell lung cancer cell lines SBC-1, SBC-3, SBC-5, N427, N69, LUT-130, H82, NCI345, LUT134B are non- inactivated the 10% FCS (Hyclone) RPMI- 1640 medium supplemented with in (GibcoBRL), and maintained at 37 ° C, in 5% C0 2 moisture culture Yoshita.
  • SEM is an Iscove culture (GibcoBRL) supplemented with inactivated 10% FCS.
  • MDR K562 / D1-9 cells were maintained in RPMI-1640 culture medium supplemented with inactivated 10% FCS and 0.1 ⁇ M daunorubicin (DNR).
  • DNR daunorubicin
  • Cell proliferation was determined using a count of cells using the trypan blue dye exclusion method and a modified MTT assay using a SF reagent (Nacalai Tesque).
  • Leukemia cells were cultured in flat-bottom 96-well plates (HGreiner labortechnik) at 2 ⁇ 10 4 cells / well in 100 ⁇ l medium and cultured for 72 hours with various concentrations of compound. ⁇ ⁇ ⁇
  • GUT-70 digestion compound 7
  • K562 / D1-9 cells were cultured at various concentrations for GUT-70 or DNR for 72 hours under the same conditions. Cultured. The average of each of the six data was used. There was a linear relationship between TT assay and cell number.
  • Protein samples were separated by SDS / PAGE and nitrocellulose membrane (Amersham
  • the membrane was saturated with a solution of 5% non-fat dry milk in TBST (25 mM Tris-HCl pH 7.8, 140 mM NaCl, 0.1% (vol / vol) Tween20) and persimmon polyclonal p21 w AFI / CIP1 , p27Kipl , Incubated overnight with p53 antibody (diluted to 1000 min). Then, the plate was washed thoroughly with TBST and incubated for 1 hour with donkey anti-Peacock IgG conjugated with horseradish peroxidase (Santa Cruz Biotech.). Detection was performed at Amersham Biosciences;
  • Apoptosis induction was examined for untreated NALM6 and NALM6 cells treated with GUT-70 for 12 hours and 24 hours. May-Giemsa staining was used to study the overall morphology, and a terminal deoxynucleotidyl transferase (TUNEL) assay was used to detect apoptosis. After drug treatment, the 5 X 10 4 cells were washed with PBS (P H7.3), and resuspended in the same buffer. The cytospin preparation of the cell suspension was fixed and stained with May-Giemsa stain. Cell morphology was determined by light microscopy.
  • TUNEL assay was performed using an apoptosis detection kit (R & D systems, Wiesbaden, Germany) according to the instructions. Briefly, NAL 6 cells were resuspended in PBS containing 3.7% formaldehyde on ice for 10 minutes and rinsed with PBS. Then fixed fine The cells were incubated in cytonin for 30 minutes at room temperature and washed with labeling buffer. Cells are labeled using a 25 1 labeling mix (dNTPMix, Mn 2+ , TdT, TdT labeling buffer).
  • the cells were rinsed with PBS, counterstained with 2 ⁇ g / ml propidium iodide for 30 minutes, and subjected to flow analysis using flow cytometry (FACScan; Becton Dickinson. NJ).
  • the sensitivity of normal hematopoietic progenitor cells to GUT-70 was examined by a standard methylcellulose culture assay containing GM-CSF, IL_3, and G-SCF.
  • Normal hematopoietic progenitor cells used were bone marrow cells from a healthy volunteer with informed consent. The cells (1 ⁇ 10 5 ) were cultured in methylcellulose for 12 days at 37 ° C. in a 5% CO 2 incubator at a GUT-70 concentration of 0.20 or 50. The number of CFU-GM and BFU-E colonies was counted under a microscope. Three tissues were tested for each gnorape and the average was determined.
  • Compound 7 having a propyl group showed a more remarkable effect (FIG. 2).
  • the compounds ⁇ and The IC50 value (halHnaximal inhibitory concentrations) of the compound 8 was 3 ⁇ M and 9 ⁇ M, respectively.
  • Compound 7 also exhibited concentration-dependent and time-dependent cell growth inhibitory effects on the other five leukemia cell lines (K562, HL60, SEM, NALM6, and SUPB15) (FIG. 3).
  • GUT-70 also steadily inhibited the growth of human small cell lung cancer cell lines SBC-1, SBC-3, LUT134B, and H82 (IC50 value of 5 ⁇ to 8 ⁇ , FIG. 9).
  • compound 7 (GUT- 70) of exposing the c NALM-6 cells examined the relationship between leukemia cytostatic and apoptosis by changing the concentration of the compound 7 24 hours, morphological techniques and flow in the induction of apoptosis Measured by cytometry. After 24 hours of exposure to compound 7 at a concentration of 5 ⁇ , significant cell apoptosis was observed morphologically (FIG. 4).
  • Compound 7 induces TUNEL-positive (i.e., apoptosis) cells significant number, reduced the percentage of cells in G 2 / M phase.
  • Compound 7 was accumulated in Gi-phase cells by TUNEL-Assy using double-stained piumumoxide staining, and apoptotic cells mainly appeared near the G-S phase (FIG. 5 ).
  • Daunorubicin is generally used as an anticancer drug, and is expressed by P-glycoprotein in contrast to K562 / D1-9, which is a cell that expresses P-glycoprotein. It was about 75 times more resistant than the parent strain K562. On the other hand, the sensitivity of compound 7 was not reduced even for K562 / D1_-9, which overexpresses P-glycoprotein, as compared with the parent strain K562, which does not express P-glycoprotein (Fig. 7). ). These findings indicate that compound 7 (GUT-70) is not involved in the P-glycoprotein-related MDR system.
  • Compound 7 (GUT-70) appears to be a good candidate component to overcome the induction of MDR. Patients with relapse have MDR that induces P-glycoprotein, which is an anticancer drug efflux pump, and clinical trials of compound 7 as an anticancer drug that often shows multidrug resistance are initially applied to patients with relapsed or refractory disease It is thought that it is done.
  • the anticancer agent of the present invention has an action mechanism different from the conventional one, and suppresses cancer cell growth in a p53-independent manner. Therefore, the present invention can be expected to have an effective effect on cancers and the like which could not be expected with conventional anticancer agents. In addition, it is possible to treat a wider variety of cancers and reduce side effects by using them in combination with drugs having different mechanisms.
  • the pharmaceutical composition of the present invention maintains an excellent effect on multidrug-resistant cancer cells in which P-glycoprotein has been induced. Therefore, it can be effectively used for patients whose other anticancer drugs have stopped responding or for patients who need multiple drug combination therapy.
  • the present invention is suitable for clinical use with a large safety margin.
  • the present invention has various requirements that have been eagerly sought in the treatment of cancer and provides an excellent means for treating those diseases.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

An anticancer agent which contains as an active ingredient a compound represented by the general formula (1): (1) (wherein R1 to R9 are the same as defined in the description) or a pharmaceutically acceptable salt thereof.

Description

明細書  Specification
抗癌剤  Anticancer drug
技術分野  Technical field
本発明は、抗癌剤並びに癌の治療方法に関する。  The present invention relates to an anticancer agent and a method for treating cancer.
背景技術  Background art
従来から、植物より抽出した物質に、抗癌作用や発癌予防作用を有するものがあること が知られており、植物の抽出物力 得られる成分に関して、多くの研究が行われてきた。 例えば、 St. John' s wortという植物から得られる物質が抗腫瘍効果を示すことが報告さ れている(Christoph et al., Oncogene, 2002, 21, 1242-1250)。また、キヤロフイラム属から 得られる物質の中に、癌予防に有効な物質があること (Ito et al, Journal of Natural Products, 2002, 65(3), 267 - 272)、細胞障害性を有する物質があること(David et al., Phytochemistry, 2001, 58, 571- 575)、エイズに対する抑制効果を有する物質があること (Claude et al., Bioorganic & Medicinal Chemistry Letters, 1998, 8, 3475-3478)が報告 されている。これらの文献で報告されてレ、る物質の構造は様々で、機能又は作用との関 連は明確になっていない。  It has been known that some substances extracted from plants have an anti-cancer effect and a carcinogenesis-preventing effect, and many studies have been made on components that can be obtained from plant extracts. For example, it has been reported that a substance obtained from a plant called St. John's wort exhibits an antitumor effect (Christoph et al., Oncogene, 2002, 21, 1242-1250). In addition, among substances obtained from the genus Carophilium, there are substances that are effective in preventing cancer (Ito et al, Journal of Natural Products, 2002, 65 (3), 267-272), and substances that have cytotoxicity (David et al., Phytochemistry, 2001, 58, 571-575) and the presence of substances with an inhibitory effect on AIDS (Claude et al., Bioorganic & Medicinal Chemistry Letters, 1998, 8, 3475-3478). It has been reported. The structures of the substances reported in these documents vary, and their relation to function or action is not clear.
一方、化学合成の分野においても、抗癌剤として有用な様々な薬剤の開発が行われ てきた。これまでにも、高い細胞増殖抑制作用を示す抗癌剤などが種々開発されている。 しかし、これらの中には、正常細胞にも毒性を示すものがあり、患者への実際の投与に十 分な適性を有していないものも少なくなかった。また、癌においては、発症メカニズムと p53との関連が指摘され、 p53を誘導することにより効果を発揮する抗癌剤が多く開発き れてきたが、望まれてレ、る程度の効果を有さなレ、か、又は望まれている程度には異なるタ イブの癌に幅広い効果を示さなレ、ものが少なくなかった。更に、相乗的な作用や副作用 の軽減を目的として、複数の抗癌剤を併用する多剤療法も行われてレ、るが、 P糖蛋白(P glycoprotein)が誘導されて、細胞が多剤耐性 (MDR)を獲得することが多ぐそのような患 者に、抗癌剤を投与しても、癌細胞内の薬物濃度が低下し、有効な効果が得られないと いった問題が生じていた。  On the other hand, in the field of chemical synthesis, various drugs useful as anticancer agents have been developed. Until now, various anticancer agents and the like exhibiting a high cell growth inhibitory action have been developed. However, some of these were also toxic to normal cells, and many did not have sufficient suitability for actual administration to patients. In cancer, the relationship between the onset mechanism and p53 has been pointed out, and many anticancer drugs that exert their effects by inducing p53 have been developed, but they have not been expected to have any degree of effect. Many did not show a broad effect on cancer of different types, or to the extent desired. Furthermore, in order to reduce synergistic effects and side effects, multidrug therapy using a combination of multiple anticancer drugs has been performed. However, P-glycoprotein (P-glycoprotein) is induced, and cells become multidrug-resistant ( The administration of an anticancer drug to such a patient, who often acquires MDR), has a problem in that the drug concentration in the cancer cells is reduced and an effective effect cannot be obtained.
これらのことから、従来とは異なるメカニズムを有し、多剤耐性癌細胞にも有効であって、 し力も安全域の大きレ、、新たな抗癌剤の開発が期待されてレ、た。 本発明は、従来と異なるメカニズムを有し、多剤耐性癌細胞にも有効で、しかも安全域 の大きい抗癌剤を提供することを目的とする。 Based on these facts, it has a mechanism different from conventional ones, is effective for multidrug-resistant cancer cells, has a large safety margin, and is expected to develop new anticancer agents. An object of the present invention is to provide an anticancer agent having a mechanism different from the conventional one, effective for multidrug-resistant cancer cells, and having a large safety margin.
また、本発明は、癌の治療方法を提供することを目的とする。 図面の簡単な説明  Another object of the present invention is to provide a method for treating cancer. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 Calophyllmn Brasiliensisより得られた 15の化合物の構造を示す。  FIG. 1 shows the structures of 15 compounds obtained from Calophyllmn Brasiliensis.
図 2は、図 1に示した 15の化合物について、 BV173細胞の増殖抑制作用を、濃度 Ο μ Μ, M, 5 μ Μ, ΙΟ μ Μ及び 30 Μにお!/ヽて調べた結果を示す。  FIG. 2 shows the results of examining the inhibitory effect of 15 compounds shown in FIG. 1 on the growth of BV173 cells at concentrations of ΟμΜ, M, 5μΜ, ΙΟμΜ, and 30Μ! .
図 3は、化合物 7(GUT- 70)の細胞増殖抑制作用を、 5種類の白血病細胞株 (BV173、 SEM、 NALM6、 HL60及び K562)につ!/、て測定した結果を示す。また(♦)は 0 μ M、 (翻)は 0·5 μ Μ、(口)は 1.0 μ Μ、(◊)は 5.0 μ Μ、(X )は 10 Μの濃度での測定結果 をそれぞれ示す。  FIG. 3 shows the results of measuring the cytostatic effect of compound 7 (GUT-70) on five types of leukemia cell lines (BV173, SEM, NALM6, HL60, and K562). (♦) indicates 0 μM, (translation) is 0.5 μΜ, (mouth) is 1.0 μΜ, (5.0) is 5.0 μΜ, and (X) is the measurement result at 10 10. .
図 4は、化合物 7によるアポトーシスを形態学的に観察レた結果を示す。図 4Αは化合物 7で処理する前、図 4Βは化合物 7で処理後の観察結果をそれぞれ示す。  FIG. 4 shows the results of morphological observation of apoptosis by compound 7. FIG. 4Α shows the observation results before treatment with compound 7, and FIG. 4Β shows the observation results after treatment with compound 7.
図 5は、 TUNELアツセィにより、細胞周期とアポトーシスの関係を測定した結果を示す。 図 5Αは GUT-70無処理時、図 5Βは GUT-70 5 μ Μ、 24時間処理時のアポトーシス誘 導に関する結果をそれぞれ示す。  FIG. 5 shows the results of measuring the relationship between cell cycle and apoptosis using TUNEL Atssei. Fig. 5Α shows the results for the induction of apoptosis when GUT-70 was not treated and Fig. 5Β shows the results when GUT-70 was treated for 5 μΜ for 24 hours.
図 6は、化合物 7で処理した白血病細胞株 NALM6細胞と BV173細胞における、 P21WAF./CIP.^ P27KIPLp53及び p57の時間的変化を調べた結果を示す。 Figure 6 shows the leukemia cell line NALM6 cells and BV173 cells treated with compound 7, was examined the temporal change in the P21 WAF./CIP.^ P27 K IPL, p53 and p57 results.
図 7Aは、 P糖蛋白高発現細胞 K562/D1 - 9と P糖蛋白が発現していない親株 562 細胞に対するダウノルビシン (DNR)の耐性を比較した結果を示す。図 7Bは、 P糖蛋白高 発現細胞 K562/D1 - 9と P糖蛋白が高発現していない親株 K562細胞に対する化 物 7 の耐性を比較した結果を示す。  FIG. 7A shows the results of comparing the resistance of daunorubicin (DNR) to the P-glycoprotein high-expressing cell K562 / D1-9 and the parent strain 562 cells that do not express P-glycoprotein. FIG. 7B shows the results of comparing the resistance of compound 7 to the P-glycoprotein overexpressing cell K562 / D1-9 and the parent strain K562 cell not overexpressing P-glycoprotein.
図 8は、正常造血前駆細胞 CFU- GMと BFU-Eを用いて、正常細胞に対する化合物 7の毒性を調べた結果を示す。  FIG. 8 shows the results of examining the toxicity of compound 7 on normal cells using normal hematopoietic progenitor cells CFU-GM and BFU-E.
図 9は、ヒト小細胞肺ガンに対する化合物 7 (GUT- 70)の効果を調べた結果を示す。  FIG. 9 shows the results of examining the effect of compound 7 (GUT-70) on human small cell lung cancer.
発明の開示  Disclosure of the invention
本発明者は、前記課題を解決することを主な目的として、鋭意検討を重ねた。その結 果、特定の構造を有する化合物 顕著なガン細胞の増殖抑制作用を奏すること、多 剤耐性癌細胞に対しても有効であること、また安全域が大きいことも見出し、更に銳意検 討を重ねて、本発明を完成するに至った。 The inventor of the present invention has made intensive studies with the main object of solving the above problems. As a result, a compound having a specific structure has a remarkable inhibitory effect on the growth of cancer cells. They also found that they are effective against drug-resistant cancer cells, and that they have a large safety margin. The present inventors have made further studies and completed the present invention.
すなわち、本発明は、以下の抗癌剤及び癌の治療方法に関するものである。  That is, the present invention relates to the following anticancer agents and methods for treating cancer.
1.下記一般式 (1)  1.The following general formula (1)
Figure imgf000005_0001
Figure imgf000005_0001
(式中、 R R2は、それぞれ独立に水素原子、アルキル基、シクロアルキル基又は置換 されていてもよいァリール基を表し、或いは、 CR 2は、 C =〇を表し; (Wherein RR 2 independently represents a hydrogen atom, an alkyl group, a cycloalkyl group or an aryl group which may be substituted, or CR 2 represents C = 〇;
R3、 R4は、それぞれ独立に水素原子、ハロゲン原子、シァノ基、ヒドロキシ基、アルキ ル基、シクロアルキル基、置換されていてもよいァリール基、アルコキシ基、ァシル基、力 ルポキシル基、アルコキシカルボ-ル基、アミノ基、モノアルキルアミノ基、ジアルキルアミ ノ基又〖ま了シノレ: Γミノ基を表し; R 3 and R 4 each independently represent a hydrogen atom, a halogen atom, a cyano group, a hydroxy group, an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a propyloxyl group, an alkoxy group, A carboxyl group, an amino group, a monoalkylamino group, a dialkylamino group or a quinoline: a diamino group;
R5、 R8、 R9は、それぞれ独立に水素原子、ハロゲン原子、シァノ基、ヒドロキシ基、アル キル基、シクロアルキル基、置換されていてもよいァリール基、アルコキシ基、ァシル基、 カルボキシル基、アルコキシカルボニル基、置換されていてもよいァリールォキシ基、チ オール基、アルキルチオ基、置換されていてもよいァリールチオ基、アルキルスルホニル 基、置換されていてもよいァリールスルホュル基、アルキルアミノカルボニル基、置換され ていてもよいァリールァミノカルボニル基、アルキルアミノスルホエル基、置換されていて もよぃァリールアミノスルホニル基、アルコキシカルボニル基、置換されていてもよいァリ ールォキシカルボニル基、アミノ基、モノアルキルアミノ基、ジァルキノレアミノ基、ァシルァ ミノ基、アルキルスルホニルァミノ基又は置換されてレ、てもよ!/、ァリーノレスノレホニノレァミノ 基を表し; R 5 , R 8 and R 9 each independently represent a hydrogen atom, a halogen atom, a cyano group, a hydroxy group, an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a carboxyl group , Alkoxycarbonyl group, optionally substituted aryloxy group, thiol group, alkylthio group, optionally substituted arylthio group, alkylsulfonyl group, optionally substituted arylsulfur group, alkylaminocarbonyl Group, an optionally substituted arylaminosulfonyl group, an alkylaminosulfoyl group, an optionally substituted arylaminosulfonyl group, an alkoxycarbonyl group, an optionally substituted aryloxycarbonyl Group, amino group, monoalkylamino group, dialquinoleamino group, acylamino , Alkylsulfonyl § amino group or a substituted been, even! /, Arino Resnore Honinoreamino Represents a group;
R6は、アルキル基、シクロアルキル基、置換されていてもよいァリール基、アルコキシ基、 ァシル基、カルボキシル基、アルコキシカルボ-ル基又は下記一般式(2) R 6 represents an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a carboxyl group, an alkoxycarbol group, or a compound represented by the following general formula (2)
Figure imgf000006_0001
Figure imgf000006_0001
で表される構造を表し; Represents a structure represented by;
R7は、 0、 NH又は NR13を表し; R 7 represents 0, NH or NR 13 ;
R1。は、水素原子、アルキル基、シクロアルキル基又は置換されていてもよいァリール基 を表し; R 1. Represents a hydrogen atom, an alkyl group, a cycloalkyl group or an aryl group which may be substituted;
Ru、 R12は、それぞれ独立に水素原子、アルキル基、シクロアルキル基、置換されていて もよぃァリール基を表し; R u and R 12 each independently represent a hydrogen atom, an alkyl group, a cycloalkyl group, or a substituted or unsubstituted aryl group;
R13は、アルキル基、シクロアルキル基または置換されていてもよいァリール基を表す) で表される化合物またはその薬学的に許容される塩を有効成分とする抗癌剤。 R 13 represents an alkyl group, a cycloalkyl group or an aryl group which may be substituted) or a pharmaceutically acceptable salt thereof as an active ingredient.
2. R7が酸素原子である項 1に記載の抗癌剤。 2. The anticancer agent according to item 1, wherein R 7 is an oxygen atom.
3. R5力 Sメトキシ基である項 1に記載の抗癌剤。 3. anticancer agent according to claim 1 which is R 5 force S methoxy group.
4. R9がアルキル基又は置換されていてもよいァリール基である項 1に記載の抗癌剤4. The anticancer agent according to item 1, wherein R 9 is an alkyl group or an optionally substituted aryl group.
5. R9が n—プロピル基である項 1に記載の抗癌剤。 5. The anticancer agent according to item 1, wherein R 9 is an n-propyl group.
6. R6が、下記一般式 (2) 6. R 6 is the following general formula (2)
Figure imgf000006_0002
Figure imgf000006_0002
(式中、 R1G、 R11および R12は、前記に定義されるとおりである) で表される基である項 1に記載の抗癌剤 c Wherein R 1G , R 11 and R 12 are as defined above. The anticancer agent c according to item 1, which is a group represented by
7. 下記一般式 (3)  7. The following general formula (3)
Figure imgf000007_0001
Figure imgf000007_0001
で表される項 1に記載の抗癌剤。 Item 4. The anticancer agent according to item 1, represented by the formula:
8. 項 1〜7のいずれかに記載の化合物の有効量を投与することを特徴とする癌の 治療方法。  8. A method for treating cancer, which comprises administering an effective amount of the compound according to any one of Items 1 to 7.
9. 癌が、急性骨髄性白血病、急性リンパ性白血病、悪性リンパ腫、絨毛癌、多発 性骨髄腫、軟部腫瘍、小細胞肺ガン、慢性骨髄性白血病、甲状腺髄様癌、骨肉腫、頭 頸部癌、食道癌、非小細胞肺ガン、大腸癌、胃癌、胆道癌、脳腫瘍、悪性黒色腫、腎臓 癌、膝臓癌、肝臓癌からなる群より選択される項 8に記載の方法。  9. The cancer is acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, soft tissue tumor, small cell lung cancer, chronic myelogenous leukemia, medullary thyroid carcinoma, osteosarcoma, head and neck Item 9. The method according to Item 8, which is selected from the group consisting of cancer, esophageal cancer, non-small cell lung cancer, colon cancer, stomach cancer, biliary tract cancer, brain tumor, malignant melanoma, kidney cancer, knee cancer, and liver cancer.
10. 癌が、急性骨髄性白血病、急性リンパ性白血病、悪性リンパ腫、慢性骨髄性 白血病、小細胞肺ガン力 なる群より選択される項 9に記載の方法。  10. The method according to item 9, wherein the cancer is selected from the group consisting of acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, chronic myeloid leukemia, and small cell lung cancer.
11. 癌が多剤耐性 ( DR)の癌である項 8に記載の方法。  11. The method according to item 8, wherein the cancer is a multidrug resistant (DR) cancer.
本発明は、好ましくは一般式(3)で表される化合物を有効成分として含有する抗癌剤、 並びに該化合物を癌患者に投与する癌の治療方法に係るものである。  The present invention relates to an anticancer agent preferably containing a compound represented by the general formula (3) as an active ingredient, and a method for treating cancer by administering the compound to a cancer patient.
更に好ましくは、急性骨髄性白血病、急性リンパ性白血病、悪性リンパ腫、絨毛癌、多 発性骨髄腫、軟部腫瘍、小細胞肺ガン、慢性骨髄性白血病、甲状腺髄様癌、骨肉腫、 頭頸部癌、食道癌、非小細胞肺ガン、大腸癌、胃癌、胆道癌、脳腫瘍、悪性黒色腫、腎 S蔵癌、膝臓癌、肝臓癌力 なる群より選択される疾患を治療するための、一般式 (3)で表 される化合物を有効成分として含有する抗癌剤に係るものである。 また、本発明は、好ましくは、一般式 (3)で表される化合物の抗癌有効量を投与するこ とを特徴とする癌の治療方法に係るものである。 More preferably, acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, soft tissue tumor, small cell lung cancer, chronic myelogenous leukemia, medullary thyroid carcinoma, osteosarcoma, head and neck cancer Esophageal cancer, non-small cell lung cancer, colorectal cancer, gastric cancer, biliary tract cancer, brain tumor, malignant melanoma, renal S tumor, knee cancer, liver cancer. The present invention relates to an anticancer agent containing a compound represented by the formula (3) as an active ingredient. Further, the present invention preferably relates to a method for treating cancer, which comprises administering an anticancer effective amount of a compound represented by the general formula (3).
更に好ましくは、一般式 (3)で表される化合物の抗癌有効量を投与することを特徴とす る、急性骨髄性白血病、急性リンパ性白血病、悪性リンパ腫、絨毛癌、多発性骨髄腫、 軟部腫瘍、小細胞肺ガン、慢性骨髄性白血病、甲状腺髄様癌、骨肉腫、頭頸部癌、食 道癌、非小細胞肺ガン、大腸癌、胃癌、胆道癌、脳腫瘍、悪性黒色腫、腎臓癌、勝臓癌、 肝臓癌からなる群より選択される疾患の治療方法に係るものである。  More preferably, acute antimyelogenous leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, characterized in that an anticancer effective amount of the compound represented by the general formula (3) is administered. Soft tissue tumor, small cell lung cancer, chronic myeloid leukemia, medullary thyroid cancer, osteosarcoma, head and neck cancer, esophageal cancer, non-small cell lung cancer, colorectal cancer, gastric cancer, biliary tract cancer, brain tumor, malignant melanoma, kidney The present invention relates to a method for treating a disease selected from the group consisting of cancer, victory cancer, and liver cancer.
以下、本発明を詳細に説明する。  Hereinafter, the present invention will be described in detail.
抗癌化合物の構造  Structure of anticancer compound
本発明の抗癌剤の有効成分は、下記 般式(1)で表される構造を有する。  The active ingredient of the anticancer agent of the present invention has a structure represented by the following general formula (1).
-般式 (1)  -General formula (1)
Figure imgf000008_0001
Figure imgf000008_0001
(式中、 R1〜 は、前記に定義されるとおりである。 ) (In the formula, R 1 to are as defined above.)
3において、アルキル基は、置換又は非置換の直鎖状或いは分岐状のものを 含み、炭素数は通常は 1〜18、好ましくは 1〜8、更に好ましくは 1〜3であり、例えば、メ チル、ェチル、 n—プロピル、イソプロピル、 n—ブチル、イソブチノレ、 tert—ブチノレ、 n— ペンチル、イソペンチル、へキシル、ヘプチル、ォクチル、ノニル及ぴデシルが挙げられ る。 In to 3 , the alkyl group includes a substituted or unsubstituted linear or branched one, and usually has 1 to 18, preferably 1 to 8, and more preferably 1 to 3, for example, Examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutynole, tert-butynole, n-pentyl, isopentyl, hexyl, heptyl, octyl, nonyl and decyl.
シクロアルキル基は、置換又は非置換のものを含み、炭素数も特に限定されないが、 通常は 3〜: 10、好ましくは 3〜7である。シクロアルキル基の具体例としては、シクロプロピ ノレ、シクロブチル、シクロペンチル、シクロへキシノレ及びシクロへプチルが挙げられる。 ァリール基としては、「ァリール基」とは、 5又は 6員の芳香族炭化水素環からなる単環 又は多環系の基を意味し、具体例としては、フエ-ル、ナフチル、フルォレニル、アントリ ル、ビフエ二リル、テトラヒドロナフチル、クロマニル、 2, 3—ジヒドロー 1, 4ージォキサナフ タレニル、インダニル及ぴフエナントリルが挙げられる。 The cycloalkyl group may be substituted or unsubstituted, and the number of carbon atoms is not particularly limited, but is usually 3 to 10, preferably 3 to 7. Specific examples of the cycloalkyl group include cyclopropanol, cyclobutyl, cyclopentyl, cyclohexynole, and cycloheptyl. As the aryl group, the term "aryl group" means a monocyclic or polycyclic group composed of a 5- or 6-membered aromatic hydrocarbon ring, and specific examples include phenyl, naphthyl, fluorenyl, and anthrenyl. , Biphenyl, tetrahydronaphthyl, chromanyl, 2,3-dihydro-1,4-dioxanaphthalenyl, indanyl and phenanthryl.
置換基を有するァリール基の置換基の数は 1〜3個、好ましくは 1または 2個であり、該 置換基としては、メチル、ェチル、メトキシ、エトキシ、ァミノ、メチルァミノ、ジメチノレアミノ、 シァノ、ニトロ、フッ素、塩素、臭素、トリフルォロメチル、ヒドロキシ、カルボキシル、メトキシ カルボニル、エトキシカルボ-ル、プロポキシ力ルポニル、ブトキシカルポニル、 CONH2, ァセチルが例示される。置換ァリール基の例示としては、フエニル、 2—、 3—若しくは 4一 フルオロフェニル、 2—、 3—若しくは 4一クロ口フエ-ル、 2_、 3—若しくは 4一ブロモフエ 二ノレ、 2, 4ージフルオロフェニル、 2, 3_、 2, 4—、 3, 4—、 3, 5—若しくは 2, 6—ジクロ 口フエニル、 3—クロ口一 4ーフゾレオ口フエニル、 4—イソプロピルフエニル、 2, 6—ジメチ ノレフエ二ノレ、 2—、 3—若しくは 4—トリフルォロメチルフエニル、 4—メトキシフエ二ル、 4— トリフルォロメトキシフエニル、 2—、 3 _若しくは 4—ジメチルァミノフエエル、 2_、 3—若し くは 4一-トロフエニル、 4—スルファモイルフヱニルが例示される。  The number of substituents of the aryl group having a substituent is 1 to 3, preferably 1 or 2, and the substituent includes methyl, ethyl, methoxy, ethoxy, amino, methylamino, dimethinoleamino, cyano, nitro, Examples include fluorine, chlorine, bromine, trifluoromethyl, hydroxy, carboxyl, methoxycarbonyl, ethoxycarbol, propoxylproponyl, butoxycarbonyl, CONH2, acetyl. Examples of the substituted aryl group include phenyl, 2-, 3- or 4-fluorophenyl, 2-, 3- or 4-chlorophenol, 2_, 3- or 4-bromophenyl, 2,4- Difluorophenyl, 2, 3_, 2, 4-, 3, 4-, 3, 5- or 2,6-dichlorophenyl, 3-chloro-1-phenylphenyl, 4-isopropylphenyl, 2,6- Dimethynophenyl, 2-, 3- or 4-trifluoromethylphenyl, 4-methoxyphenyl, 4-trifluoromethoxyphenyl, 2-, 3- or 4-dimethylaminophenyl, 2_ , 3- or 4-1-trophenyl, and 4-sulfamoylphenyl.
アルコキシ基としては、一〇R' (R'は前記アルキル基である)で表される基が挙げられ、 例えば、メトキシ、エトキシ、 n—プロポキシ、イソプロポキシ、 n -ブトキシ、イソブトキシ、 sec-ブトキシ、 t -ブトキシが例示される。  Examples of the alkoxy group include groups represented by 〇R ′ (R ′ is the above-mentioned alkyl group). For example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy , T-butoxy.
ァシル基としては、一 COR' ' ( R"は前記アルキル基または前記置換されていてもよ ぃァリール基である)で表される基が挙げられ、例えば、ァセチル、プロピオニル、ベンゾ ィルが例示される。  Examples of the acyl group include a group represented by one COR ′ ′ (R ″ is the above-mentioned alkyl group or the above-mentioned optionally substituted aryl group), and examples thereof include acetyl, propionyl, and benzoyl. Is done.
ハロゲン原子としては、フッ素、塩素、臭素、ヨウ素が挙げられる。  Examples of the halogen atom include fluorine, chlorine, bromine, and iodine.
アシノレアミノ基としては、一 NHCOR' ' ( R"は前記アルキル基または前記置換されて いてもよいァリール基である)で表される基が挙げられ、例えば、ァセチルァミノ、プロピオ ニノレアミノ、ブチリルァミノ、ベンゾィルァミノが挙げられる。  Examples of the asinoleamino group include a group represented by one NHCOR ′ ′ (R ″ is the above-mentioned alkyl group or the above-mentioned optionally substituted aryl group). Can be
アルコキシカルボニル基としては、一 CO〇R' (R'は前記アルキル基である)で表され る基が挙げられ、例えば、メトキシカルボエル、エトキシカルボニル、 n—プロポキシカルボ 二ノレ、イソプロポキシカルボニル、 n -ブトキシカルボニル、イソブトキシカルボニル、 sec -ブ トキシカルボニル、 t-ブトキシカルボニルが例示される。 置換されてレヽてもよ!/ヽァリールォキシ基としては、 -0- (置換されてレ、てもよレヽァリール 基)で表される基が挙げられ、例えばフエニルォキシ、ナフチルォキシが挙げられる。 Examples of the alkoxycarbonyl group include a group represented by one CO〇R ′ (R ′ is the aforementioned alkyl group), for example, methoxycarbyl, ethoxycarbonyl, n-propoxycarbinole, isopropoxycarbonyl, Examples are n-butoxycarbonyl, isobutoxycarbonyl, sec-butoxycarbonyl and t-butoxycarbonyl. You may be replaced! Examples of the / aryloxy group include a group represented by -0- (substituted or substituted aryl group), and examples include phenyloxy and naphthyloxy.
アルキルチオ基としては、 -S- (アルキル基)で表される基が拳げられ、例えばメチルチ ォ、ェチルチオ、 n—プロピノレチォ、イソプロピルチオ、 n—プチルチオ、イソプチルチオ、 tert—ブチルチオが挙げられる。 Examples of the alkylthio group include a group represented by -S- (alkyl group), and examples thereof include methylthio, ethylthio, n -propynolethio, isopropylthio, n-butylthio, isobutylthio, and tert-butylthio.
置換されてレ、てもよ!/、ァリールチオ基としては、 - S- (置換されてレ、てもよレ、ァリール基) で表される基が挙げられ、例えばフエ二ルチオ、ナフチルチオが挙げられる。  You can be replaced! Examples of the / and arylthio groups include groups represented by -S- (substituted or substituted aryl groups), such as phenylthio and naphthylthio.
モノアルキルアミノ基としては、前記アルキルでモノ置換されたァミノ基、例えばメチル ァミノカルボニル、ェチルァミノカルボニル、 n—プロピルアミノカルボニル、イソプロピル ァミノカルボニル、 n—プチルァミノカルボニル、イソブチルァミノカルボニル、 tert—プチ ルァミノカルボニル、 n—ペンチルァミノ力ルポ-ル、イソペンチルァミノカルボニル、へキ シルァミノカルボニルが挙げられる。  Examples of the monoalkylamino group include an amino group mono-substituted with the above alkyl, such as methylaminocarbonyl, ethylaminocarbonyl, n-propylaminocarbonyl, isopropylaminocarbonyl, n-butylaminocarbonyl, and isobutylamino. Carbonyl, tert-butylaminocarbonyl, n-pentylaminophenol, isopentylaminocarbonyl and hexylaminocarbonyl.
ジァノレキノレアミノ基としては、前記アルキルでジ置換されたアミノ基、例えばジメチルァ ミノカルボニル、ジェチルァミノカルボニル、ジ n—プロピルアミノカルボニル、ジイソプロピ ルァミノカルボニル、ジ n—ブチルァミノ力ルポニル、ジイソブチルァミノカルボニル、ジ tert—ブチルァミノ力ルポニル、ジ n—ペンチルァミノカルボニル、ジイソペンチルァミノ力 ルポ二ノレ、ジへキシルァミノカルボニルが挙げられる。  Examples of the dianolequinole amino group include amino groups di-substituted with the above alkyl, such as dimethylaminocarbonyl, getylaminocarbonyl, di-n-propylaminocarbonyl, diisopropylaminocarbonyl, di-n-butylaminoamino, Examples thereof include diisobutylaminocarbonyl, ditert-butylaminocarbonyl, di-n-pentylaminocarbonyl, diisopentylaminocarbonyl, and dihexylaminocarbonyl.
アルキルスルホニル基としては、 - S02 - (アルキル基)で表される基が挙げられ、例えば メチノレスノレホニノレ、ェチノレスノレホニノレ、 n—プロピノレスノレホニノレ、イソプロピノレスノレホニノレ、 n—ブチルスルホニル、イソプチルスルホニル、 tert—プチルスルホニルが挙げられる。 置換されていてもよいァリールスルホニル基としては、 _S02 -(置換されていてもよいァ リール基)で表される基が挙げられ、例えばフエニルスルホ -ル、ナフチルスルホエルが 挙げられる。 The alkylsulfonyl group, - S0 2 - include groups represented by (alkyl), for example, methylcarbamoyl Roh less Honoré Honi Honoré, E Chino less Honoré Honi Honoré, n- pro Pinot less Honoré Honi Honoré, isopropylidene Roh-less And norfoninole, n-butylsulfonyl, isoptylsulfonyl, and tert-butylsulfonyl. The optionally substituted § reel sulfonyl group, _S0 2 - include groups represented by (optionally substituted § aryl group), for example Fuenirusuruho - Le include naphthylsulfonyl El.
アルキルアミノカルボ-ル基としては、前記アルキルでモノ置換またはジ置換されたァ ミノカルボニル基が挙げられ、前記アルキルでモノ置換されたァミノ基としては、メチルァ ミノカノレポ二ノレ、ェチルァミノカルボニル、 n—プロピルアミノカルボニル、イソプロピルアミ ノカルボエル、 n—ブチルァミノカルボニル、イソプチルァミノカルボニル、 tert—ブチルァ ミノカルボニル、 n—ペンチルァミノカルボニル、イソペンチルァミノカルボニル、へキシノレ ァミノカルボニルが挙げられ、前記アルキルでジ置換されたァミノカルボニル基としては、 ジメチルァミノ力ルポニル、ジェチルァミノカルボニル、ジ n—プロピルアミノカルボニル、 ジイソプロピルアミノカルボニル、ジ n—プチルァミノカルボニル、ジイソプチルァミノカル ボニル、ジ tert—ブチルァミノカルボニル、ジ n—ペンチルァミノカルボニル、ジイソペン チルァミノカルボニル、ジへキシルァミノカルボニルが挙げられる。 Examples of the alkylaminocarbo group include an aminocarbonyl group mono- or di-substituted with the alkyl. Examples of the amino group mono-substituted with the alkyl include methylaminocanoleponinole, ethylaminocarbonyl, and the like. n-propylaminocarbonyl, isopropylaminocarboel, n-butylaminocarbonyl, isobutylaminocarbonyl, tert-butylaminocarbonyl, n-pentylaminocarbonyl, isopentylaminocarbonyl, and hexinoleaminocarbonyl. As the amino-disubstituted di-substituted alkyl, Dimethylaminopropyl, diethylaminocarbonyl, di-n-propylaminocarbonyl, diisopropylaminocarbonyl, di-n-butylaminocarbonyl, diisobutylaminocarbonyl, di-tert-butylaminocarbonyl, di-n-pentyla Examples include minocarbonyl, diisopentylaminocarbonyl, and dihexylaminocarbonyl.
置換されていてもよいァリールァミノカルボニル基としては、 - CONH -(置換されていて もよぃァリール基)で表される基が挙げられ、例えばフエニルァミノカルボニル、ナフチル ァミノカルボニルが挙げられる。  Examples of the optionally substituted arylaminocarbonyl group include a group represented by -CONH- (optionally substituted arylylcarbonyl), such as phenylaminoaminocarbonyl and naphthylaminocarbonyl. No.
アルキルアミノスルホニル基としては、前記アルキルでモノ置換またはジ置換されたアミ ノスルホニル基が挙げられ、前記アルキルでモノ置換されたァミノ基としては、メチルアミ ノスルホニル、ェチルアミノスルホニル、 n—プロピルアミノスルホニル、イソプロピルアミノ スルホエル、 n—ブチルアミノスルホニル、イソブチルアミノスルホニル、 tert—ブチノレアミ ノスルホニル、 n—ペンチルアミノスルホニル、イソペンチルアミノスルホニル、へキシルァ ミノスルホニルが挙げられ、前記アルキルでジ置換されたアミノスルホ -ル基としては、ジ メチルアミノスルホニル、ジェチルアミノスルホニル、ジ n—プロピルアミノスルホニル、ジィ ソプロピルアミノスルホニル、ジ n -ブチルアミノスルホニル、ジイソブチルアミノスルホニ ノレ、ジ tert—プチルアミノスルホニル、ジ n—ペンチルアミノスルホニル、ジイソペンチルァ ミノスルホニル、ジへキシノレアミノスルホニルが挙げられる。  Examples of the alkylaminosulfonyl group include an aminosulfonyl group mono- or di-substituted with the alkyl. Examples of the amino group mono-substituted with the alkyl include methylaminosulfonyl, ethylaminosulfonyl, and n-propylamino. Sulfonyl, isopropylamino sulfoel, n-butylaminosulfonyl, isobutylaminosulfonyl, tert-butynoleaminosulfonyl, n-pentylaminosulfonyl, isopentylaminosulfonyl, hexylaminosulfonyl, and the aminosulfonyl di-substituted with the alkyl. Examples of the methyl group include dimethylaminosulfonyl, getylaminosulfonyl, di-n-propylaminosulfonyl, diisopropylaminosulfonyl, di-n-butylaminosulfonyl, Two Honoré, di tert- heptyl aminosulfonyl, di n- pentyl aminosulfonyl, Jiisopenchirua Minosuruhoniru include carboxymethyl Honoré aminosulfonyl to di.
置換されていてもよいァリールアミノスルホニル基としては、- S02NH- (置換されていて もよぃァリール基)で表される基が挙げられ、例えばフエニルアミノスルホニル、ナフチル アミノスルホニルが挙げられる。 The substituents on these § reel aminosulfonyl group, - S0 2 NH- include groups represented by (substituted Moyoi Ariru group), like for example phenylalanine amino sulfonyl, naphthyl aminosulfonyl Can be
置換されてレ、てもよレ、ァリールォキシカルボニル基としては、 -COO- (置換されてレ、て もよぃァリール基)で表される基が挙げられ、例えばフエ-ルォキシカルボュル、ナフチル ォキシカルボニルが挙げられる。  The substituted or unsubstituted aryloxycarbonyl group includes a group represented by -COO- (substituted or unsubstituted aryl group), for example, phenylcarboxy group. And naphthyloxycarbonyl.
アルキルスルホニルァミノ基としては、 - NHS02 -(アルキル基)で表される基が挙げら れ、例えばメチルスルホニルァミノ、ェチルスルホニルァミノ、 n—プロピルスルホニルアミ ノ、イソプロピルスルホ-ルァミノ、 n—ブチルスルホニルアミ入イソブチルスルホニルアミ ノ、 tert一ブチルスノレホニルァミノ、 n—ペンチルスルホニルァミノ、イソペンチルスルホニ ノレアミノ、へキシルスルホニルァミノが挙げられ、 置換されてレ、てもよレ、ァリールスルホニルァミノ基としては、 - NHS02- (置換されて!/ヽ てもよぃァリール基)で表される基が挙げられ、例えばフエニルスルホニルアミ入ナフチ ルスルホニルァミノが挙げられる。 Alkylsulfonyl § amino group, - NHS0 2 - et include groups represented by (alkyl) is, for example, methylsulfonyl § Mino, E chill sulfonyl § amino, n- propylsulfonyl amino, isopropylsulfonyl - Ruamino, n-butylsulfonylamino-containing isobutylsulfonylamino, tert-butylsnolephonylamino, n-pentylsulfonylamino, isopentylsulfonynonamino, hexylsulfonylamino, Substituted by Les, be good Le, as a § reel sulfonyl § amino group, - NHS0 2 - include groups (! Substituted /ヽalso good I Ariru group) represented by, e.g., phenylalanine sulfonyl Examples include naphthylsulfonylamino with amide.
本発明の一般式(1)で表される化合物に関し、 R7は酸素原子 (〇) が好ましい。 R5はメトキシ基(_OCH3)が好ましい。 In the compound represented by the general formula (1) of the present invention, R 7 is preferably an oxygen atom (〇). R 5 is preferably a methoxy group (_OCH 3 ).
R9はアルキル基又はァリール基が好ましく、特に n—プロピル基が好ましい。 R 9 is preferably an alkyl group or an aryl group, particularly preferably an n-propyl group.
また、 R6は、一般式 (2) R 6 is a general formula (2)
Figure imgf000012_0001
Figure imgf000012_0001
(式中、 R1(3、 R11および R12は、前記に定義されるとおりである) Wherein R 1 (3 , R 11 and R 12 are as defined above)
で表される基が好ましい。 The group represented by is preferred.
本発明においては、一般式 (3)  In the present invention, the general formula (3)
Figure imgf000012_0002
Figure imgf000012_0002
で表される化合物(5-メトキシ- 2,2 -ジメチル _6- (2-メチル -トォキソ -2-ブテニル)- 10-プ 口ピル- 2H,8H-ベンゾ [1,2 - b;3,4- b,]ジピラン- 8-オン(5- methoxy- 2,2 - dimethy卜 6-(2 - methtl-l-oxo-2-butenyl)-10-propyl-2H,8H-benzo[l,2-b;3,4-b']dipyran-8-one) )が抗 癌剤として好ましい。 (5-methoxy-2,2-dimethyl_6- (2-methyl-oxo-2-butenyl) -10-prop-pyr-2H, 8H-benzo [1,2-b; 3,4 -b,] dipyran-8-one (5-methoxy-2,2-dimethy 6- (2- methtl-l-oxo-2-butenyl) -10-propyl-2H, 8H-benzo [l, 2-b; 3,4-b '] dipyran-8-one)) is preferred as an anticancer agent.
この一般式 (3)の化合物は、特に優れた癌細胞増殖抑制作用を有する。  The compound of the general formula (3) has a particularly excellent cancer cell growth inhibitory action.
本発明の一般式ひ)で表される化合物の薬学的に許容される塩は、 〜 R6、 R8, R9 のいずれかがカルボキシル基(COOH)あるいはアミノ基、モノ一またはジ一アルキルアミ ノ基を有する場合、薬学的に許容される塩を調製できる。薬学的に許容されるカルボキシ ル基の塩としては、アルカリ金属塩 (ナトリウム塩、カリウム塩、リチウム塩)が例示され、薬 学的に許容されるァミノ基、モノーまたはジ一アルキルアミノ基の塩としては、硫酸塩、塩 酸塩、臭化水素酸塩、硝酸鉛、リン酸塩などの無機酸塩、フマル酸塩、マレイン酸塩、コ ハク酸塩、メタンスルホン酸塩、トルエンスルホン酸塩などの有機酸塩が挙げられる。この ような薬学的に許容される塩は、本発明の化合物とアルカリ金属の炭酸塩または炭酸水 素塩、アルカリ金属水酸化物、あるいは、無機酸または有機酸を作用させることにより得 ることがでさる。 Pharmaceutically acceptable salts of the compounds represented by the general formula (I) of the present invention may be any one of -R 6 , R 8 , R 9 in which a carboxyl group (COOH) or an amino group, a mono- or di-alkylamino group. If the compound has a phenyl group, a pharmaceutically acceptable salt can be prepared. Examples of pharmaceutically acceptable salts of carboxy groups include alkali metal salts (sodium salt, potassium salt, lithium salt), and pharmaceutically acceptable salts of amino, mono- or dialkylamino groups. Inorganic salts such as sulfate, hydrochloride, hydrobromide, lead nitrate and phosphate, fumarate, maleate, succinate, methanesulfonate, toluenesulfonate And other organic acid salts. Such pharmaceutically acceptable salts can be obtained by reacting a compound of the present invention with an alkali metal carbonate or carbonate, an alkali metal hydroxide, or an inorganic or organic acid. Monkey
一般式 (1)の化合物の製造方法  Method for producing compound of general formula (1)
上記化合物は、種々の方法で適宜調製することができ、化学的に合成してもよぐまた、 適当な植物から抽出などにより分離してもよい。  The above compounds can be appropriately prepared by various methods, may be chemically synthesized, or may be separated from an appropriate plant by extraction or the like.
(i)化学的合成方法 (i) Chemical synthesis method
化学的に合成する方法としては、適宜公知の方法を用/、ることができる。  As a method of chemically synthesizing, a known method can be appropriately used.
例えば、以下のような方法が挙げられる。  For example, the following method can be used.
下記一般式 (4)  The following general formula (4)
Figure imgf000013_0001
Figure imgf000013_0001
(式中、 〜 は、前記に定義されるとおりである)  (Wherein is as defined above)
で表される化合物、又は、下記一般式(5)
Figure imgf000014_0001
Or a compound represented by the following general formula (5)
Figure imgf000014_0001
(式中、 〜1 5は、前記に定義されるとおりである。 Zは水素原子または R13(R13は前記 定義されるとおりである)を示す。 ) (Wherein, to 1 5, wherein is as defined. Z represents a hydrogen atom or R 13 (R 13 is as defined above).)
で表される化合物と、下記一般式 (6) And a compound represented by the following general formula (6)
Figure imgf000014_0002
Figure imgf000014_0002
(式中、 R8及び R9は、前記に定義されるとおりである) Wherein R 8 and R 9 are as defined above.
で表される化合物とを、濃硫酸を用いて加熱反応させて、 The compound represented by is heated and reacted using concentrated sulfuric acid,
それぞれ、下記一般式 (7) Each of the following general formula (7)
Figure imgf000014_0003
Figure imgf000014_0003
(式中、 R^R5, R8及び R9は、前記に定義されるとおりである) Wherein R ^ R 5 , R 8 and R 9 are as defined above.
で表される化合物、又は、下記一般式 (8)
Figure imgf000015_0001
Or a compound represented by the following general formula (8)
Figure imgf000015_0001
(式中、 〜 、 R8、 R9及ぴ Zは、前記に定義されるとおりである) (Wherein, R 8 , R 9 and Z are as defined above)
で表される化合物を得る。 Is obtained.
次いで、一般式 (7)又は (8)で表される化合物に、例えば、下記一般式 (9) R6— C1 (9) Next, the compound represented by the general formula (7) or (8) is added to, for example, the following general formula (9) R 6 — C1 (9)
で表される化合物を、フリーデル ·クラフツ (Friede卜 Crafts)触媒を用いて反応させること によって、下記一般式 (1) Is reacted with a Friedel Crafts catalyst to obtain a compound represented by the following general formula (1)
Figure imgf000015_0002
Figure imgf000015_0002
(式中、 〜 は、前記に定義されるとおりである)  (Wherein is as defined above)
で表される化合物を得る。 Is obtained.
R6が一般式 (2)で表される構造の化合物は、一般式 (9)で表される化合物として、下 記一般式(10)
Figure imgf000016_0001
A compound having a structure wherein R 6 is represented by the general formula (2) is a compound represented by the following general formula (10) as a compound represented by the general formula (9).
Figure imgf000016_0001
(式中、 R1Q〜R12は、前記に定義されるとおりである) (Wherein, R 1Q to R 12 are as defined above)
で表される化合物を、フリーデル 'クラフツ (Friedel - Crafts)触媒を用いてァシル化反応さ せて得ることができる。 Can be obtained by an acylation reaction using a Friedel-Crafts catalyst.
フリーデル 'クラフツ(Friedeト Crafts)触媒としては、例えば、 A1C13、 BF3、 ZnCl2、 Sn Cl4などが挙げられる。 The Friedel 'Crafts (Friede preparative Crafts) catalysts include, for example, A1C1 3, BF 3, ZnCl 2 , Sn Cl 4.
一般式 (4)〜(10)で表される化合物は、いずれも、汎用の方法により得ることができ、 市販品がある場合には、それをそのまま用いればよい。  All of the compounds represented by the general formulas (4) to (10) can be obtained by a general-purpose method, and when there is a commercially available product, it may be used as it is.
斯くして得られる本発明の化合物は、通常、溶解、抽出、分液、傾斜、濾過、濃縮、薄 層クロマトグラフィー、カラムクロマトグラフィー、ガスクロマトグラフィー、高速液体クロマト グラフィ一、蒸留、昇華、結晶化などの方法を単独又は組み合わせて、適宜精製を行う。 上記と同様の方法において、所望の置換基を有する中間物を用い、又は適当な官能 基を適宜導入することによって、各種ィ匕合物を得ることができる。  The compound of the present invention thus obtained is usually dissolved, extracted, separated, gradient, filtered, concentrated, thin-layer chromatography, column chromatography, gas chromatography, high-performance liquid chromatography, distillation, sublimation, crystal Purification is carried out as appropriate by using a method such as chemical conversion alone or in combination. In the same manner as described above, various conjugates can be obtained by using an intermediate having a desired substituent or by appropriately introducing an appropriate functional group.
例えば、一般式 (4)において、 R1, R2がメチル基、 R3、 R4が水素、 R5がー OCH3基で ある化合物を用い、一般式(6)において、 R8が水素、 R9がー CH2CH2CH3基である化合 物を用い、一般式(10)において、 R1C)が水素、 R"、 R12がメチル基である化合物を用いる 場合には、 For example, in the general formula (4), a compound in which R 1 and R 2 are methyl groups, R 3 and R 4 are hydrogen, and R 5 is —OCH 3 group, and in the general formula (6), R 8 is hydrogen When a compound in which R 9 is a —CH 2 CH 2 CH 3 group is used, and in the general formula (10), a compound in which R 1C) is hydrogen, R ″ and R 12 is a methyl group,
下記一般式 (3) The following general formula (3)
Figure imgf000017_0001
Figure imgf000017_0001
で表される化合物を得ることができる。 Can be obtained.
(ii)植物からの抽出方法  (ii) Extraction method from plants
本願発明の医薬組成物の有効成分となる化合物は、適当な植物力 抽出して得ること もできる。植物としては、オトギリ草科(Guttiferae)の植物が例示できる。オトギリ草科の植 物では、キヤロフィラム(Calophyllum)属のもの、例えば、 Calophyllum Brasiliensis又は Calophyllum inophyllumなどが好適であり、特に、 Calophyllum Brasiliensisが好適に用い られる。  The compound serving as an active ingredient of the pharmaceutical composition of the present invention can be obtained by extracting an appropriate plant power. Examples of the plant include a plant of the family Cucurbitaceae (Guttiferae). For plants of the family Tortaceae, those belonging to the genus Calophyllum, such as Calophyllum Brasiliensis or Calophyllum inophyllum, are preferred, and Calophyllum Brasiliensis is particularly preferred.
植物から該化合物を抽出して得る方法は、適宜公知の方法を用レ、ることができ、特に 限定されないが、例えば、植物の樹皮などを粉枠し、有機溶媒で抽出する方法などを用 いることができる。  The method for obtaining the compound by extracting the compound from the plant can be appropriately selected from known methods, and is not particularly limited. For example, a method in which the bark of a plant is powder-framed and extracted with an organic solvent is used. Can be.
具体的には、次のように行うことができる。 Calophyllum Brasiliensis又は Calophyllum inophyllumの乾燥した樹皮の粉砕物に、適当な有機溶媒、例えば、アセトン、へキサン、 メタノール、エタノール等の低級アルコールなどを適宜組み合わせて用いて、室温で抽 出した後、低圧下溶媒を蒸発する。更にクロマトグラフィーやろ過、抽出など、適宜分離 精製を行って、所望の化合物を得る。  Specifically, it can be performed as follows. After extracting the dried bark of Calophyllum Brasiliensis or Calophyllum inophyllum with an appropriate organic solvent, for example, a lower alcohol such as acetone, hexane, methanol, ethanol, etc., at appropriate temperature, extract at room temperature, and then extract under low pressure. Evaporate the solvent. Further, the desired compound is obtained by performing appropriate separation and purification such as chromatography, filtration and extraction.
例えば、一般式(3)の化合物は、 Calophyllum Brasiliensis (Guttiferae)の植物抽出物 を分離精製して得ることができる。  For example, the compound of the general formula (3) can be obtained by separating and purifying a plant extract of Calophyllum Brasiliensis (Guttiferae).
医薬組成物(医薬製剤)の製法 一般式 (1)の化合物またはその塩は、薬学的に許容される担体と配合し、錠剤、カブ セル剤、顆粒剤、散剤などの固形製剤;シロップ剤、注射剤などの液状製剤、貼付剤、軟 膏剤、硬膏剤などの経皮吸収剤、吸入剤、坐剤として、適宜製剤化することができる。 本発明の医薬組成物は、経口または非経口投与され、上記化合物を 1種単独で用い てもよく、又は 2種以上を併用して用いてもよい。 Manufacturing method of pharmaceutical composition (pharmaceutical formulation) The compound of the general formula (1) or a salt thereof is mixed with a pharmaceutically acceptable carrier, and solid preparations such as tablets, capsules, granules and powders; liquid preparations such as syrups and injections, and patches It can be appropriately formulated as a transdermal absorbent such as an ointment and a plaster, an inhalant, and a suppository. The pharmaceutical composition of the present invention is orally or parenterally administered, and the above compounds may be used alone or in combination of two or more.
薬学的に許容される担体としては、製剤素材として慣用されている各種有機あるいは 無機担体物質を用いることができる。具体的には、固形製剤における賦形剤、滑沢剤、 結合剤、崩壌剤、液状製剤における溶剤、溶解補助剤、懸濁化剤、等張化剤、緩衝剤、 無痛化剤などを配合することができる。また必要に応じて、防腐剤、抗酸化剤、着色剤、 甘味剤などの製剤添加物を用いることもできる。  As the pharmaceutically acceptable carrier, various organic or inorganic carrier substances commonly used as pharmaceutical materials can be used. Specifically, excipients, lubricants, binders, disintegrants in solid preparations, solvents, dissolution aids, suspending agents, isotonic agents, buffers, soothing agents, etc. in liquid preparations Can be blended. If necessary, pharmaceutical additives such as preservatives, antioxidants, coloring agents and sweeteners can also be used.
賦形剤の好適な例としては、例えば乳糖、白糖、 D—マンニトール、デンプン、結晶セ ルロース、軽質無水ケィ酸などが挙げられる。  Preferred examples of the excipient include lactose, sucrose, D-mannitol, starch, crystalline cellulose, light caffeic anhydride and the like.
滑沢剤の好適な例としては、例えばステアリン酸マグネシウム、ステアリン酸カルシウム、 タルク、コロイドシリカなどが挙げられる。  Preferred examples of the lubricant include, for example, magnesium stearate, calcium stearate, talc, colloidal silica and the like.
結合剤の好適な例としては、例えば結晶セルロース、白糖、 D—マンニトール、デキス トリン、ヒドロキシプロピルセノレロース、ヒドロキシプロピルメチルセルロース、ポリビニノレピロ リドンなどが挙げられる。  Suitable examples of the binder include, for example, crystalline cellulose, sucrose, D-mannitol, dextrin, hydroxypropylsenorellose, hydroxypropylmethylcellulose, polyvinylinolepyrrolidone and the like.
崩壌剤の好適な例としては、例えばデンプン、カルボキシメチルセルロース、カルボキ シメチルセルロースカルシウム、クロスカルメロースナトリウム、カルボキシメチルスターチ ナトリウムなどが挙げられる。  Preferred examples of the disintegrant include starch, carboxymethylcellulose, carboxymethylcellulose calcium, croscarmellose sodium, carboxymethyl starch sodium and the like.
溶剤の好適な例としては、例えば注射用水、アルコール、プロピレングリコール、マクロ ゴール、ゴマ油、トウモロコシ油などが挙げられる。  Preferred examples of the solvent include water for injection, alcohol, propylene glycol, macrogol, sesame oil, corn oil and the like.
溶解補助剤の好適な例としては、例えばポリエチレングリコール、プロピレンダリコール、 D—マンニトール、安息香酸ベンジル、エタノール、トリスァミノメタン、コレステロール、トリ エタノールァミン、炭酸ナトリウム、クェン酸ナトリウムなどが挙げられる。  Preferred examples of the dissolution aid include, for example, polyethylene glycol, propylene dalicol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate and the like. .
懸濁化剤の好適な例としては、例えばステアリルトリエタノールァミン、ラウリル硫酸ナト リウム、ラウリルアミノプロピオン酸、レシチン、塩化ベンザルコニゥム、塩化べンゼトニゥム、 モノステアリン酸グリセリンなどの界面活性剤;ポリビュルアルコール、ポリビニルピロリドン、 カルボキシメチノレセルロースナトリウム、メチノレセノレロース、ヒドロキシメチルセノレロース、ヒ ドロキシェチルセルロース、ヒドロキシプロピルセルロースなどの親水性高分子などが挙 げられる。 Preferable examples of the suspending agent include surfactants such as stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glycerin monostearate; polybutyl alcohol , Polyvinylpyrrolidone, sodium carboxymethinolate cellulose, methinoresenololose, hydroxymethylsenorelose, Examples include hydrophilic polymers such as droxicetyl cellulose and hydroxypropyl cellulose.
等張化剤の好適な例としては、例えば塩化ナトリウム、グリセリン、 D—マンニトールな どが挙げられる。  Preferable examples of the tonicity agent include sodium chloride, glycerin, D-mannitol and the like.
緩衝剤の好適な例としては、例えばリン酸塩、酢酸塩、炭酸塩、クェン酸塩などの緩衝 液などが挙げられる。  Preferred examples of the buffer include buffers such as phosphate, acetate, carbonate, and citrate.
無痛ィ匕剤の好適な例としては、例えばべンジルアルコールなどが挙げられる。防腐剤 の好適な例としては、例えば、パラォキシ安息香酸エステル類、クロロブタノール、ベンジ ルアルコール、フエネチルアルコール、デヒドロ酢酸、ソルビン酸などが挙げられる。  Preferable examples of the painless darting agent include, for example, benzyl alcohol. Preferable examples of the preservative include, for example, paraoxybenzoic acid esters, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid and the like.
抗酸化剤の好適な例としては、例えば亜硫酸塩、ァスコルビン酸などが挙げられる。 本発明の抗癌剤は、各種の癌の治療に適用することができる。  Preferable examples of the antioxidant include, for example, sulfite, ascorbic acid and the like. The anticancer agent of the present invention can be applied to the treatment of various cancers.
癌の種類は、特に限定されないが、例えば、急性骨髄性白血病、急性リンパ性白血病、 悪性リンパ腫、絨毛癌、多発性骨髄腫、軟部腫瘍、小細胞肺ガン、慢性骨髄性白血病、 甲状腺髄様癌、骨肉腫、頭頸部癌、食道癌、非小細胞肺ガン、大腸癌、胃癌、胆道癌、 脳腫瘍、悪性黒色腫、腎臓癌、膝臓癌、肝臓癌が挙げられる。  The type of cancer is not particularly limited, for example, acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, soft tissue tumor, small cell lung cancer, chronic myeloid leukemia, medullary thyroid cancer Osteosarcoma, head and neck cancer, esophageal cancer, non-small cell lung cancer, colorectal cancer, stomach cancer, biliary tract cancer, brain tumor, malignant melanoma, kidney cancer, knee cancer, and liver cancer.
本発明の抗癌剤は、特に、急性骨髄性白血病、急性リンパ性白血病、小細胞肺ガン、 に有効である。中でも急性骨髄性白血病、急性リンパ性白血病、小細胞肺ガンに好適に 用いられる。  The anticancer agent of the present invention is particularly effective for acute myeloid leukemia, acute lymphocytic leukemia, and small cell lung cancer. Among them, it is suitably used for acute myeloid leukemia, acute lymphocytic leukemia, and small cell lung cancer.
本発明の抗癌剤の投与条件及び投与方法は、疾患の種類や患者の状態又は適用部 位などに応じて、適宜設定することができる。  The administration conditions and administration method of the anticancer agent of the present invention can be appropriately set according to the type of the disease, the condition of the patient, the application site, and the like.
具体的には、投与量は、動物の種類や大きさ (または体重)、症状等に応じて適宜調 節する。ヒト又はヒト以外の動物に投与する場合には、約 0. 1— 100mg/kg (体重)程度、 好ましくは約 1. 0_ 10mg/kg (体重)程度、更に好ましくは約 1. 0 - 3. Omg/kg (体重)程 度の量で投与する。また投与方法も、最適の治療効果が得られるように適宜調節でき、 例えば、毎日数回に分けて投与でき、治療中に危険な状態が生じた場合には、その状 態に応じて投与量や投与回数を適宜減らすこともできる。  Specifically, the dose is appropriately adjusted according to the type, size (or weight), symptoms, etc. of the animal. When administered to humans or non-human animals, about 0.1 to 100 mg / kg (body weight), preferably about 1.0 to 10 mg / kg (body weight), more preferably about 1.0 to 3. Administer Omg / kg (body weight). The administration method can also be adjusted appropriately to obtain the optimal therapeutic effect.For example, it can be administered in several divided doses daily, and if a dangerous condition occurs during treatment, the dose may be adjusted according to the condition. And the number of administrations can be reduced as appropriate.
本願発明の抗癌剤は、所望の効果を得るために、単独で投与することもでき、また他の 抗癌剤、化学療法剤、抗炎症剤または免疫療法剤などと適宜組み合わせて用いることも でさる。 The anticancer agent of the present invention can be administered alone to obtain a desired effect. It can also be used in appropriate combination with an anticancer agent, a chemotherapeutic agent, an anti-inflammatory agent or an immunotherapeutic agent.
本発明の抗癌剤と組み合わせて用レ、る他の抗癌剤としては、例えば、代謝拮抗剤、ァ ルキル化薬、白金系抗癌剤、トポイソメラーゼ阻害剤、抗癌性抗生物質、チロシンキナー 5 ゼ阻害剤、ヒト化抗体が挙げられる。  Other anticancer agents used in combination with the anticancer agent of the present invention include, for example, antimetabolites, alkylating agents, platinum-based anticancer agents, topoisomerase inhibitors, anticancer antibiotics, tyrosine kinase inhibitors, human Antibodies.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
以下、実施例、試験例等を挙げて、本発明をより一層具体的に説明するが、本発明は これらの実施例に限定されることはない。  Hereinafter, the present invention will be described more specifically with reference to Examples and Test Examples, but the present invention is not limited to these Examples.
' 化合物の調製  '' Preparation of compounds
10 ブラジノレで採取されたキヤロフィラムブラジリエンシス (Calophyllum Brasiliensis)を用 いて、次のような手順で複数の化合物を得た。まず、 Calophyllum Brasiliensis乾燥樹皮 (630g)を粉砕した後、アセトン (3L)を用いて 3回冷浸抽出を行った。得られたアセトン抽 出液を減圧下濃縮し、残渣をアセトン抽出物(19.26g)とした。このアセトン抽出物につい て、溶出液としてへキサン一アセトンを用いて、 Siゲルカラムクロマトグラフィーを行ったと 10 Using Calophyllum Brasiliensis collected from Brassinole, several compounds were obtained by the following procedure. First, the dry bark (630 g) of Calophyllum Brasiliensis was pulverized and then subjected to cold immersion extraction three times with acetone (3 L). The obtained acetone extract was concentrated under reduced pressure, and the residue was used as an acetone extract (19.26 g). This acetone extract was subjected to Si gel column chromatography using hexane-acetone as eluent.
15 ころ、 85:15溶出部より、化合物 7 (GUT - 70) (137.8mg)が得られた。溶出液として、へキ サン一アセトン、アセトン、 CH2C12 - MeOH、及ぴメタノールを用いて、同様の処理を行つ た結果、化合物 7を含む 15種類の化合物が得られた。得られた 15の化合物の構造を 図 1に示す。 At around 15: Compound 7 (GUT-70) (137.8 mg) was obtained from the eluate at 85:15. The same treatment was carried out using hexane-acetone, acetone, CH 2 C 12 -MeOH, and methanol as eluents. As a result, 15 kinds of compounds including compound 7 were obtained. Figure 1 shows the structures of the obtained 15 compounds.
なお、化合物 7(GUT - 70)は、 Palmer, C.J.; Josephs, J.L. Tetrahedron Lett. 1994, 35, 20 5363-5366, and references thereinに基づいて同定した。また、他の化合物の構造につ いても、公知文献に基づいて同定した。  Compound 7 (GUT-70) was identified based on Palmer, C.J .; Josephs, J.L. Tetrahedron Lett. 1994, 35, 20 5363-5366, and references therein. The structures of other compounds were also identified based on known literature.
細胞の調製  Cell preparation
ヒト白血病細胞 BV173、 SUPB15、 NAL 6, HL60及ぴ SEMを American Type Culture Collectionから得た。また、ヒト小細胞肺癌細胞株 SBC- 1、 SBC- 3、 SBC_5、 25 N427、 N69、 LUT_130、 H82、 NCI345, LUT134Bを実験に使用した。ヒト白血病細胞株 BV173, SUPB15、 NALM6, HL60及ぴ SEM、並びにヒト小細胞肺癌細胞株 SBC- 1、 SBC- 3、 SBC- 5、 N427、 N69、 LUT- 130、 H82、 NCI345, LUT134Bは、非働化した 10% FCS (Hyclone)を添加した RPMI-1640培養液(GibcoBRL)中、 37°C、 5%C02の湿気中培 養下で維持した。 SEMは、非働化した 10%FCSを添加した Iscove培養液(GibcoBRL) 中で培養した。 P糖蛋白高発現多剤耐性 (MDR) K562/D1- 9細胞を非働化した 10% FCSと 0.1 μ Mダウノルビシン(DNR)を添加した RPMI - 1640培養液で維持した。 Human leukemia cells BV173, SUPB15, NAL6, HL60 and SEM were obtained from the American Type Culture Collection. In addition, human small cell lung cancer cell lines SBC-1, SBC-3, SBC_5, 25N427, N69, LUT_130, H82, NCI345, LUT134B were used in the experiments. Human leukemia cell lines BV173, SUPB15, NALM6, HL60 and SEM, and human small cell lung cancer cell lines SBC-1, SBC-3, SBC-5, N427, N69, LUT-130, H82, NCI345, LUT134B are non- inactivated the 10% FCS (Hyclone) RPMI- 1640 medium supplemented with in (GibcoBRL), and maintained at 37 ° C, in 5% C0 2 moisture culture Yoshita. SEM is an Iscove culture (GibcoBRL) supplemented with inactivated 10% FCS. Cultured in P-glycoprotein high expression multidrug resistance (MDR) K562 / D1-9 cells were maintained in RPMI-1640 culture medium supplemented with inactivated 10% FCS and 0.1 μM daunorubicin (DNR).
In vitro細胞增殖活性の測定  In vitro cell proliferation activity measurement
細胞増殖は、トリパンブルー染色排除方法 (trypan blue dye exclusion method)を使用 した細胞数のカウントと SF試薬 (Nacalai Tesque)を用いた改変 MTTアツセィ(modified MTT assay)を使用して決定した。白血病細胞は、平底 96ゥェルプレー HGreiner labortechnik)で 100 μ 1媒体中 1ゥエルあたり 2 X 104細胞として培養し、種々の濃度の化 合物とともに 72時間培養した。 Ρ糖蛋白高発現細胞における GUT - 70 (ィ匕合物 7)の効 果を調べるために、 K562/D1- 9細胞を、 GUT-70又は DNRについて種々の濃度で 72 時間、同様の条件で培養した。それぞれ 6つのデータの平均値を使用した。 TTアツセ ィと細胞数の間には直線関係が見られた。 Cell proliferation was determined using a count of cells using the trypan blue dye exclusion method and a modified MTT assay using a SF reagent (Nacalai Tesque). Leukemia cells were cultured in flat-bottom 96-well plates (HGreiner labortechnik) at 2 × 10 4 cells / well in 100 μl medium and cultured for 72 hours with various concentrations of compound.調 べ る In order to examine the effect of GUT-70 (digestion compound 7) on cells expressing high levels of glycoprotein, K562 / D1-9 cells were cultured at various concentrations for GUT-70 or DNR for 72 hours under the same conditions. Cultured. The average of each of the six data was used. There was a linear relationship between TT assay and cell number.
ウェスタンプロット分析  Western plot analysis
蛋白質試料を SDS/PAGEによって分離し、ニトロセルロース膜 (Amersham  Protein samples were separated by SDS / PAGE and nitrocellulose membrane (Amersham
Bioscience)上でエレクトロブロットした。膜を、 5%ノンフアット乾燥ミルクを含有する TBST (25mM Tris-HCl pH7.8, 140mM NaCl,0. l%(vol/vol)Tween20)溶液で飽和させ、ゥサギ ポリクローナル p21wAFI/CIP1p27Kiplp53抗体(1000分の に希釈)で一晩インキュベート した。次いで TBSTで完全に洗浄し、西洋ヮサビペルォキシダーゼ(Santa Cruz Biotech.)と複合体ィ匕したロバ抗ゥサギ IgGで 1時間インキュベートした。検出は、増幅ィ匕 学 ¾光キッ (Amersham Biosciences;で実施し 7こ。 Bioscience). The membrane was saturated with a solution of 5% non-fat dry milk in TBST (25 mM Tris-HCl pH 7.8, 140 mM NaCl, 0.1% (vol / vol) Tween20) and persimmon polyclonal p21 w AFI / CIP1 , p27Kipl , Incubated overnight with p53 antibody (diluted to 1000 min). Then, the plate was washed thoroughly with TBST and incubated for 1 hour with donkey anti-Peacock IgG conjugated with horseradish peroxidase (Santa Cruz Biotech.). Detection was performed at Amersham Biosciences;
アポトーシス誘導分析  Apoptosis induction analysis
未処理の NALM6及び GUT- 70で 12時間及ぴ 24時間処理した NALM6細胞につ いて、アポトーシス誘導を調べた。全体の形態を研究するために May- Giemsa染色を用 レ、、またアポトーシスを検出するためにターミナルデォキシヌクレオチジルトランスフェラ ーゼアツセィ (terminal deoxynucleotidyl transferase (TUNEL) assay)を用いた。薬物処 理した後、 5 X 104細胞を PBS (PH7.3)で洗浄し、同じバッファーで再懸濁した。細胞懸濁 液のサイトスピン調製液を固定し、 May- Giemsa染色液で染色した。細胞形態は光学顕 微鏡で決定した。 TUNELアツセィはアポトーシス検出キット(R&D systems, Wiesbaden, Germany)を用いて指示書に従って行った。簡単に説明すると、 NAL 6細胞を、 3.7%ホ ルムアルデヒド含有 PBS中で氷上 10分間再懸濁し、 PBSで濯いだ。次いで固定した細 胞を cytonin中 30分間、室温でインキュベートし、標識バッファーで洗浄した。細胞は、 ラベリングミックス(dNTPMix、 Mn 2+、 TdT、 TdTラベリングバッファー)を 25 1用いて Apoptosis induction was examined for untreated NALM6 and NALM6 cells treated with GUT-70 for 12 hours and 24 hours. May-Giemsa staining was used to study the overall morphology, and a terminal deoxynucleotidyl transferase (TUNEL) assay was used to detect apoptosis. After drug treatment, the 5 X 10 4 cells were washed with PBS (P H7.3), and resuspended in the same buffer. The cytospin preparation of the cell suspension was fixed and stained with May-Giemsa stain. Cell morphology was determined by light microscopy. TUNEL assay was performed using an apoptosis detection kit (R & D systems, Wiesbaden, Germany) according to the instructions. Briefly, NAL 6 cells were resuspended in PBS containing 3.7% formaldehyde on ice for 10 minutes and rinsed with PBS. Then fixed fine The cells were incubated in cytonin for 30 minutes at room temperature and washed with labeling buffer. Cells are labeled using a 25 1 labeling mix (dNTPMix, Mn 2+ , TdT, TdT labeling buffer).
37°Cで 1時間再懸濁し、停止バッファーで反応を停止した。次いで、ストレプトアビジン (streptavidine)を含む溶液 25 μ 1を用いて暗室中室温で 10分間再懸濁した。細胞を After resuspending at 37 ° C for 1 hour, the reaction was stopped with a stop buffer. Next, the cells were resuspended in a dark room at room temperature for 10 minutes using 25 μl of a solution containing streptavidin. Cells
PBSで濯ぎ、 2 μ g/mlプロビジゥムヨウ化物(propidium iodide)で 30分間対比染色し、フ ローサイトメトリー(FACScan ; Becton Dickinson.NJ)で角军析を行った。 The cells were rinsed with PBS, counterstained with 2 μg / ml propidium iodide for 30 minutes, and subjected to flow analysis using flow cytometry (FACScan; Becton Dickinson. NJ).
正常造血前駆細胞に対する GUT- 70の毒性評価  Toxicity evaluation of GUT-70 on normal hematopoietic progenitor cells
正常造血前駆細胞の GUT- 70に対する感受性を、 GM- CSF、 IL_3、 G-SCFを含む標 準メチノレセノレロース培養アツセィ (standard methylcellulose culture assay)によって調べ た。正常造血前駆細胞は、インフォームドコンセントで同意を得た健常なポランティアから の骨髄細胞を使用した。 GUT- 70の濃度を 0,20又は 50 として、該細胞(1 X 105)を、 5%C02の培養器内で、 37°Cで 12日間、メチルセルロース中で培養した。 CFU- GM及び BFU-Eのコロニーの数は顕微鏡で数えた。各グノレープの試験に対し 3つのティッシュを 用い、その平均を求めた。 The sensitivity of normal hematopoietic progenitor cells to GUT-70 was examined by a standard methylcellulose culture assay containing GM-CSF, IL_3, and G-SCF. Normal hematopoietic progenitor cells used were bone marrow cells from a healthy volunteer with informed consent. The cells (1 × 10 5 ) were cultured in methylcellulose for 12 days at 37 ° C. in a 5% CO 2 incubator at a GUT-70 concentration of 0.20 or 50. The number of CFU-GM and BFU-E colonies was counted under a microscope. Three tissues were tested for each gnorape and the average was determined.
統計分析  Statistical analysis
統計は、 Student' s t testを使用した。 0.05より小さい P値は統計的に有意と見なし、 two-sided testsから引き出した。  Statistics used Student's st test. P values less than 0.05 were considered statistically significant and were drawn from two-sided tests.
細胞増殖抑制評価  Cell growth inhibition evaluation
C. Brasiliensisからの 15の天然抽出物、すなわち 6つのキサントン (Xanthones) (1-6) 及ぴ 9つの 4-フエユルクマリン類(4- phenylcoumarins) (これは、 2つの三環型クマリンィ匕 合物(7,8)、 6つの四環型クマリン(9-14)及ぴ 1つのジメチルシクロプロピル(15)を含む) が得られた。化合物 8は、以前に Calophyllolide (Spino)又は Oblongulide(Dharmaratan) として同定されてレ、る化合物である。これらにつレ、て MTTアツセィを用いて 5つの白血 病細胞の抗白血病活性を調べた。 BV173細胞の増殖は三環形クマリン構造を含む 4 -フ ェニルクマリンィヒ合物 (ィ匕合物 7 (GUT - 70)及ぴ 8)によって抑制された。この増殖抑制作 用は、 Mから 30 μ Mの範囲の濃度で濃度依存的であった。一方、他の 13の化合物 は増殖抑制作用を示さなかった。  Fifteen natural extracts from C. Brasiliensis, namely six Xanthones (1-6) and nine 4-phenylcoumarins (which are two tricyclic coumarinides) 7,8), six tetracyclic coumarins (9-14) and one dimethylcyclopropyl (15) were obtained. Compound 8 is a compound previously identified as Calophyllolide (Spino) or Oblongulide (Dharmaratan). In addition, the anti-leukemia activity of five leukemia cells was examined using MTT Atsey. The proliferation of BV173 cells was suppressed by a 4-phenylcoumarin conjugate containing a tricyclic coumarin structure (i-Dai-conjugate 7 (GUT-70) and 8). This growth inhibitory effect was concentration dependent at concentrations ranging from M to 30 μM. On the other hand, the other 13 compounds did not show a growth inhibitory effect.
化合物 7と化合物 8のどちらも BV173細胞の増殖抑制作用を示したが、プロピル基 を有する化合物 7 (GUT_70)の方が、より顕著な作用を示した(図 2)。また、化合物 Ίと ィ匕合物 8の IC50値 (halHnaximal inhibitory concentrations)はそれぞれ 3 μ Mと 9 μ M であった。更に、化合物 7は、その他 5種類の白血病細胞株 (K562、 HL60、 SEM、 NALM6, SUPB15)についても、濃度依存的及び時間依存的な細胞増殖抑制作用を示し た(図 3)。又 GUT- 70は、ヒト小細胞肺癌細胞株 SBC- 1、 SBC- 3、 LUT134B, H82の増 殖を着実に抑制した (IC50値 5 μ Μ〜8 μ Μ、図 9)。 While both Compound 7 and Compound 8 showed an inhibitory effect on the growth of BV173 cells, Compound 7 having a propyl group (GUT_70) showed a more remarkable effect (FIG. 2). The compounds Ί and The IC50 value (halHnaximal inhibitory concentrations) of the compound 8 was 3 μM and 9 μM, respectively. Furthermore, Compound 7 also exhibited concentration-dependent and time-dependent cell growth inhibitory effects on the other five leukemia cell lines (K562, HL60, SEM, NALM6, and SUPB15) (FIG. 3). GUT-70 also steadily inhibited the growth of human small cell lung cancer cell lines SBC-1, SBC-3, LUT134B, and H82 (IC50 value of 5 μΜ to 8 μΜ, FIG. 9).
- 上記の結果から明らかなように、ブラジノレで採取された Calophyllum Brasiliensisの茎 より抽出された三環形クマリン構造を有する化合物力 白血病細胞及び小細胞肺癌細胞 において、顕著な細胞増殖抑制作用を有することが明らかとなった。 -As is evident from the above results, a compound with a tricyclic coumarin structure extracted from the stem of Calophyllum Brasiliensis collected from Brassinole has a significant cytostatic effect on leukemia cells and small cell lung cancer cells. It became clear.
化合物 7及び 8を含む三環形クマリン化合物 (I)は濃度依存的に細胞の増殖を抑制し た。一方、他の構造の 13の化合物は増殖を抑制しな力 た。このうち、四環形クマリンィ匕 合物は、発癌予防作用を示すものも一部に報告されているが、増殖抑制作用は示さなか つた。また化合物 7の作用は、化合物 8よりも顕著であることが分力、つた。  Tricyclic coumarin compounds (I), including compounds 7 and 8, suppressed cell growth in a concentration-dependent manner. On the other hand, 13 compounds with other structures did not inhibit growth. Among them, some tetracyclic coumarindi conjugates have been reported to have a carcinogenesis-preventing action, but have no growth-suppressing action. Also, the effect of Compound 7 was more remarkable than Compound 8.
これらの知見は、三環形クマリン構造が増殖抑制作用に必須の構造であることを示す ものであり、また側鎖のプロピル基が抗白血病作用を増加することを示すものである。プ 口ピル基を側鎖に有する化合物 7がより効果的な作用を示すのは、膜に対するより強い 親和性を有するためと推根 I]される。この側鎖の重要性について得られた知見は、三環系 クマリンを含む 4-フエニルクマリン化合物の作用の改善につな力 Sることが予測される。  These findings indicate that the tricyclic coumarin structure is an essential structure for the growth inhibitory effect, and that the propyl group in the side chain increases the anti-leukemia effect. The reason that compound 7 having a propyl group in the side chain exhibits a more effective action is because of its stronger affinity for the membrane [I]. The knowledge gained about the importance of this side chain is expected to improve the action of 4-phenylcoumarin compounds, including tricyclic coumarins.
アポトーシス誘導による細胞増殖抑制  Cell proliferation suppression by inducing apoptosis
次に化合物 7 (GUT- 70)の白血病細胞増殖抑制作用とアポトーシスとの関連を調べた c NALM-6細胞を化合物 7の濃度を変えて 24時間さらし、アポトーシスの誘導を形態学 的手法とフローサイトメトリーで測定した。 5 μ Μの濃度の化合物 7に 24時間さらした後、 顕著な細胞のアポトーシスが形態学的に観察された(図 4)。化合物 7は顕著な数の TUNEL陽性 (即ちアポトーシス)細胞を誘導し、 G2/M期の細胞の割合を減少させた。プ 口ピウムョード 2重染色を用いた TUNELアツセィにより、化合物 7が Gi期の細胞に蓄積 され、アポトーシス細胞は主に Gノ S期付近で現れた(図5)。 Then compound 7 (GUT- 70) of exposing the c NALM-6 cells examined the relationship between leukemia cytostatic and apoptosis by changing the concentration of the compound 7 24 hours, morphological techniques and flow in the induction of apoptosis Measured by cytometry. After 24 hours of exposure to compound 7 at a concentration of 5 μΜ, significant cell apoptosis was observed morphologically (FIG. 4). Compound 7 induces TUNEL-positive (i.e., apoptosis) cells significant number, reduced the percentage of cells in G 2 / M phase. Compound 7 was accumulated in Gi-phase cells by TUNEL-Assy using double-stained piumumoxide staining, and apoptotic cells mainly appeared near the G-S phase (FIG. 5 ).
上記のように、 in vitroで 5つの異なるヒト白血病細胞に対し増殖抑制作用を示した化 合物 7 の作用について、幾つかの異なる方法を用いて、細胞のアポトーシスを評価した。 その結果、化合物 7で処理した白血病細胞は、 DNAの開裂を弓 Iき起こし、アポトーシス を誘導すること、また、主に Gi/S境界でアポトーシスを誘導してレ、ることが明ら力となった。 細胞周期関連蛋白に対する化合物 7 (GUT- 70)の影響 As described above, cell apoptosis was evaluated using several different methods for the effect of compound 7, which showed an antiproliferative effect on five different human leukemia cells in vitro. As a result, it is clear that leukemia cells treated with compound 7 can cause DNA cleavage and induce apoptosis, and induce apoptosis mainly at the Gi / S boundary. became. Effect of compound 7 (GUT-70) on cell cycle related proteins
NALM6細胞と BV173細胞において、 p21WAFI/CIP1、 p27Kipl、 p53及び p57のタイムコー スを調べた。化合物 7は、 NALM6細胞において、癌抑制遺伝子産物 p53及ぴサイクリ ン依存性キナーゼインヒビターである p27及び p57を誘導した力 p21は誘導しなかった。 また BV173細胞において、 p27及び p57を誘導したが、 P21及ぴ p53は誘導しなかった (図 6)。また、前述したように、化合物 7は、野生型 p53を欠損するヒト白血病細胞株 HL60にも増殖抑制作用を示した。これらのことから、化合物 7は、 p53非依存的であるこ と、即ち、化合物 7の細胞増殖抑制作用は、 p53誘導を介した p21の発現による作用で ないことがわかった。 The time course of p21 WAFI / CIP1 , p27 Kipl , p53 and p57 was examined in NALM6 cells and BV173 cells. Compound 7 did not induce the p21 which induced the tumor suppressor gene product p53 and the cyclin-dependent kinase inhibitors p27 and p57 in NALM6 cells. In BV173 cells, induced a p27 and p57, P 21及Pi p53 did not induce (Figure 6). In addition, as described above, Compound 7 also showed a growth inhibitory effect on human leukemia cell line HL60 lacking wild-type p53. From these results, it was found that Compound 7 was p53-independent, that is, the cell growth inhibitory effect of Compound 7 was not an effect of p21 expression through p53 induction.
また、全ての細胞において、 p27Kiplの発現が増加した力 一方、 p53の発現が増加した のは、 NALM6のみであった。それゆえ、化合物 Ίの処理後の主なシグナル伝達経路は p53-p21経路ではなぐ G1/S移行をプロックする p27の経路であることがわかった。 Moreover, in all cells, the expression of p27 Kipl increased, whereas the expression of p53 increased only in NALM6. Therefore, it was found that the main signaling pathway after the treatment of Compound で is the p27 pathway that blocks G1 / S translocation rather than the p53-p21 pathway.
P糖蛋白髙発現細胞 K562/D1-9に対する化合物 Ί (GUT- 70)の作用  Effect of compound Ί (GUT-70) on P-glycoprotein 髙 -expressing cells K562 / D1-9
抗癌剤として一般に用レ、られてレ、るダウノルビシン (DNR)は、 P糖蛋白が髙発現して V、る細胞である K562/D1- 9に対し、 P糖蛋白が発現してレヽなレ、親株 K562の約 75倍の 耐性を示した。これに対し、化合物 7の感受性は、 P糖蛋白が高発現している K562/D1_ - 9に対しても、 P糖蛋白が発現していない親株 K562に比べて低下しな力 た(図 7)。 これらの知見は、化合物 7 (GUT-70) 、 P糖蛋白関連 MDRシステムに関与しなレ、こ とを示すものである。化合物 7 (GUT-70)は、 MDRの誘導を克服する好適な候補成分と なるものと思われる。再発患者は、抗癌剤の排出ポンプとなる P糖蛋白を誘導する MDR を有し、多剤耐性を示すことが多ぐ抗癌剤としての化合物 7の臨床試験は、当初再発又 は難治性の患者に適用されるものと考えられる。  Daunorubicin (DNR) is generally used as an anticancer drug, and is expressed by P-glycoprotein in contrast to K562 / D1-9, which is a cell that expresses P-glycoprotein. It was about 75 times more resistant than the parent strain K562. On the other hand, the sensitivity of compound 7 was not reduced even for K562 / D1_-9, which overexpresses P-glycoprotein, as compared with the parent strain K562, which does not express P-glycoprotein (Fig. 7). ). These findings indicate that compound 7 (GUT-70) is not involved in the P-glycoprotein-related MDR system. Compound 7 (GUT-70) appears to be a good candidate component to overcome the induction of MDR. Patients with relapse have MDR that induces P-glycoprotein, which is an anticancer drug efflux pump, and clinical trials of compound 7 as an anticancer drug that often shows multidrug resistance are initially applied to patients with relapsed or refractory disease It is thought that it is done.
正常造血前駆細胞に対する化合物 7の安全性評価  Safety evaluation of compound 7 on normal hematopoietic progenitor cells
正常造血前駆細胞を 20 μ Mと 50 μ Mの化合物 7 (GUT-70)で処理した結果、 CFU - G コロニーの数は、それぞれコントロールの 75% ± 10%、 5% ±2%であることが明らか になった。また BFU- Εコロニーの数は、それぞれコントロールの 80% ± 10%、 3% ± 1% であることが明らかになった(図 8)。これらの結果から、濃度 20 μ Μまで、化合物 7 (GUT - 70)の正常造血前駆細胞に対する増殖抑制作用が極めて低いことが明らかとなつ た。このことから、化合物 7 (GUT- 70)は、安全域が大きい化合物であることがわかった。 上記の結果に示されるように、本願発明の医薬組成物は、顕著な癌増殖抑制作用を 有し、癌治療、特に白血病及び小細胞肺癌の治療に対し、優れた効果を奏し得るもので ある。 Normal hematopoietic progenitor cells treated with 20 μM and 50 μM Compound 7 (GUT-70) should result in 75% ± 10% and 5% ± 2% CFU-G colonies, respectively, of controls Became evident. The number of BFU-II colonies was found to be 80% ± 10% and 3% ± 1% of the control, respectively (Fig. 8). These results revealed that compound 7 (GUT-70) had a very low inhibitory effect on proliferation of normal hematopoietic progenitor cells up to a concentration of 20 μM. From this, it was found that compound 7 (GUT-70) was a compound having a large safety margin. As shown in the above results, the pharmaceutical composition of the present invention has a remarkable inhibitory effect on cancer growth, and can exert an excellent effect on cancer treatment, particularly on leukemia and small cell lung cancer. .
本願発明の抗癌剤は、従来とは異なる作用メカニズムを有し、 p53非依存的に癌細胞 増殖を抑制する。従って、本願発明は、従来の抗癌剤などでは効果が期待できなかった 癌などに対しても有効な効果が期待できる。また、異なるメカニズムを有する薬剤と併用 することによって、より広範な種類の癌に対する治療や副作用の軽減などを行うことが可 能となる。  The anticancer agent of the present invention has an action mechanism different from the conventional one, and suppresses cancer cell growth in a p53-independent manner. Therefore, the present invention can be expected to have an effective effect on cancers and the like which could not be expected with conventional anticancer agents. In addition, it is possible to treat a wider variety of cancers and reduce side effects by using them in combination with drugs having different mechanisms.
更に、本願発明の医薬組成物は、 P糖蛋白が誘導された多剤耐性癌細胞に対しても 優れた効果を維持する。従って、他の抗癌剤が効かなくなった患者や多剤併用療法を必 要とする患者に対しても有効に用いることができる。  Furthermore, the pharmaceutical composition of the present invention maintains an excellent effect on multidrug-resistant cancer cells in which P-glycoprotein has been induced. Therefore, it can be effectively used for patients whose other anticancer drugs have stopped responding or for patients who need multiple drug combination therapy.
し力も、本願発明は、安全域が大きぐ臨床での使用に適している。  Also, the present invention is suitable for clinical use with a large safety margin.
このように、本願発明は、癌治療において切望されてレ、る種々の要件を備えており、そ れらの疾患の治療に優れた手段を提供するものである。  As described above, the present invention has various requirements that have been eagerly sought in the treatment of cancer and provides an excellent means for treating those diseases.

Claims

請求の範囲 The scope of the claims
1.下記一般式(1)  1. The following general formula (1)
Figure imgf000026_0001
Figure imgf000026_0001
(式中、 R R2は、それぞれ独立に水素原子、アルキル基、シクロアルキル基又は置換 されていてもよいァリール基を表し、或いは、 CR 2は、 C==0を表し; (Wherein RR 2 independently represents a hydrogen atom, an alkyl group, a cycloalkyl group or an aryl group which may be substituted, or CR 2 represents C == 0;
R R4は、それぞれ独立に水素原子、ハロゲン原子、シァノ基、ヒドロキシ基、アルキ ル基、シクロアルキル基、置換されていてもよいァリール基、アルコキシ基、ァシル基、力 ルポキシル基、アルコキシカルボエル基、アミノ基、モノアルキルアミノ基、ジアルキルアミ ノ基又はァシルアミノ基を表し; RR 4 independently represents a hydrogen atom, a halogen atom, a cyano group, a hydroxy group, an alkyl group, a cycloalkyl group, an aryl group which may be substituted, an alkoxy group, an acyl group, a propyloxyl group, an alkoxycarboyl group , An amino group, a monoalkylamino group, a dialkylamino group or an acylamino group;
R5、 R8、 R9は、それぞれ独立に水素原子、ハロゲン原子、シァノ基、ヒドロキシ基、アル キル基、シクロアルキル基、置換されていてもよいァリール基、アルコキシ基、ァシル基、 カルボキシル基、アルコキシカルボニル基、置換されていてもよいァリールォキシ基、チ オール基、アルキルチオ基、置換されていてもよいァリールチオ基、アルキルスルホニル 基、置換されていてもよいァリールスルホニル基、アルキルアミノカルボニル基、置換され ていてもよいァリールァミノカルボニル基、アルキルアミノスルホ -ル基、置換されていて もよぃァリールアミノスルホニル基、アルコキシカルボニル基、置換されていてもよいァリ ールォキシカルボニル基、アミノ基、モノアルキルアミノ基、ジアルキルアミノ基、ァシルァ ミノ基、アルキノレスルホニルァミノ基又は置換されていてもよいァリールスルホニルァミノ 基を表し; R6は、アルキル基、シクロアルキル基、置換されていてもよいァリール基、アルコキシ基、 ァシル基、カルボキシル基、アルコキシカルボ-ル基又は下記一般式(2) R 5 , R 8 and R 9 each independently represent a hydrogen atom, a halogen atom, a cyano group, a hydroxy group, an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a carboxyl group An alkoxycarbonyl group, an optionally substituted aryloxy group, a thiol group, an alkylthio group, an optionally substituted arylthio group, an alkylsulfonyl group, an optionally substituted arylarylsulfonyl group, an alkylaminocarbonyl group, An optionally substituted arylaminosulfonyl group, an alkylaminosulfonyl group, an optionally substituted arylaminosulfonyl group, an alkoxycarbonyl group, an optionally substituted aryloxycarbonyl group , Amino group, monoalkylamino group, dialkylamino group, acylamino group It represents alkylene Honoré sulfonyl § amino group or an optionally substituted § reel sulfonyl § amino group; R 6 represents an alkyl group, a cycloalkyl group, an optionally substituted aryl group, an alkoxy group, an acyl group, a carboxyl group, an alkoxycarbol group, or a compound represented by the following general formula (2)
Figure imgf000027_0001
Figure imgf000027_0001
で表される構造を表し; Represents a structure represented by;
R7は、〇、 NH又は NR13を表し; R 7 represents 〇, NH or NR 13 ;
R1。は、水素原子、アルキル基、シクロアルキル基又は置換されていてもよいァリール基 を表し; R 1. Represents a hydrogen atom, an alkyl group, a cycloalkyl group or an aryl group which may be substituted;
Rn、 R12は、それぞれ独立に水素原子、アルキル基、シクロアルキル基、置換されていて もよぃァリール基を表し; R n and R 12 each independently represent a hydrogen atom, an alkyl group, a cycloalkyl group, or a substituted or unsubstituted aryl group;
R13は、アルキル基、シクロアルキル基または置換されていてもよいァリール基を表す) で表される化合物またはその薬学的に許容される塩を有効成分とする抗癌剤。 R 13 represents an alkyl group, a cycloalkyl group or an aryl group which may be substituted) or a pharmaceutically acceptable salt thereof as an active ingredient.
2. R7が酸素原子である請求項 1に記載の抗癌剤。 2. The anticancer agent according to claim 1, wherein R 7 is an oxygen atom.
3. R5がメトキシ基である請求項 1に記載の抗癌剤。 3. anticancer agent according to claim 1 R 5 is a methoxy group.
4. R9がアルキル基又は置換されていてもよいァリール基である請求項 1に記載の抗 癌剤。 4. The anticancer agent according to claim 1, wherein R 9 is an alkyl group or an optionally substituted aryl group.
5. R9が n—プロピル基である請求項 1に記載の抗癌剤。 5. The anticancer agent according to claim 1, wherein R 9 is an n-propyl group.
6. R6が、下記一般式 (2) 6. R 6 is the following general formula (2)
Figure imgf000027_0002
Figure imgf000027_0002
(式中、 R1D、 R11および R12は、前記に定義されるとおりである) で表される基である請求項 1に記載の抗癌剤。 Wherein R 1D , R 11 and R 12 are as defined above. 2. The anticancer agent according to claim 1, which is a group represented by the formula:
7. 下記一般式 (3)  7. The following general formula (3)
Figure imgf000028_0001
Figure imgf000028_0001
で表される請求項 1に記載の抗癌剤。 The anticancer agent according to claim 1, which is represented by:
8. 請求項 1〜7のいずれかに記載の化合物の有効量を投与することを特徴とする 癌の治療方法。  8. A method for treating cancer, which comprises administering an effective amount of the compound according to any one of claims 1 to 7.
9. 癌が、急性骨髄性白血病、急性リンパ性白血病、悪性リンパ腫、絨毛癌、多発 性骨髄腫、軟部腫瘍、小細胞肺ガン、慢性骨髄性白血病、甲状腺髄様癌、骨肉腫、頭 頸部癌、食道癌、非小細胞肺ガン、大腸癌、胃癌、胆道癌、脳腫瘍、悪性黒色腫、腎臓 癌、膝臓癌、肝臓癌からなる群より選択される請求項 8に記載の方法。  9. The cancer is acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, choriocarcinoma, multiple myeloma, soft tissue tumor, small cell lung cancer, chronic myelogenous leukemia, medullary thyroid carcinoma, osteosarcoma, head and neck 9. The method according to claim 8, wherein the method is selected from the group consisting of cancer, esophageal cancer, non-small cell lung cancer, colorectal cancer, gastric cancer, biliary tract cancer, brain tumor, malignant melanoma, kidney cancer, knee cancer, and liver cancer.
10. 癌が、急性骨髄性白血病、急性リンパ性白血病、悪性リンパ腫、慢性骨髄性 白血病、小細胞肺ガンからなる群より選択される請求項 9に記載の方法。  10. The method according to claim 9, wherein the cancer is selected from the group consisting of acute myeloid leukemia, acute lymphocytic leukemia, malignant lymphoma, chronic myelogenous leukemia, and small cell lung cancer.
11. 癌が多剤耐性 (MDR)の癌である請求項 8に記載の方法。  11. The method according to claim 8, wherein the cancer is a multidrug resistant (MDR) cancer.
PCT/JP2004/000680 2003-01-28 2004-01-27 Anticancer agent WO2004066994A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2005504703A JPWO2004066994A1 (en) 2003-01-28 2004-01-27 Anticancer drug

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2003-018321 2003-01-28
JP2003018321 2003-01-28

Publications (1)

Publication Number Publication Date
WO2004066994A1 true WO2004066994A1 (en) 2004-08-12

Family

ID=32820580

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/000680 WO2004066994A1 (en) 2003-01-28 2004-01-27 Anticancer agent

Country Status (2)

Country Link
JP (1) JPWO2004066994A1 (en)
WO (1) WO2004066994A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009060835A1 (en) * 2007-11-05 2009-05-14 Kyoto University Novel ubiquilin-binding small molecule
JP5622856B2 (en) * 2010-09-16 2014-11-12 株式会社エコリソース研究所 Xanthone derivatives and uses thereof
CN112409368A (en) * 2020-11-23 2021-02-26 昆明医科大学 A class of C-4 substituted coumarin compounds and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000056737A2 (en) * 1999-03-19 2000-09-28 Parker Hughes Institute Calanolides for inhibiting btk
JP2002193969A (en) * 2000-12-23 2002-07-10 Japan Science & Technology Corp Asymmetric synthesis of chromanone ring compounds and synthesis of carophyllam coumarin compounds
WO2002066475A2 (en) * 2001-02-23 2002-08-29 Ligand Pharmaceuticals Incorporated Tricyclic androgen receptor modulator compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000056737A2 (en) * 1999-03-19 2000-09-28 Parker Hughes Institute Calanolides for inhibiting btk
JP2002193969A (en) * 2000-12-23 2002-07-10 Japan Science & Technology Corp Asymmetric synthesis of chromanone ring compounds and synthesis of carophyllam coumarin compounds
WO2002066475A2 (en) * 2001-02-23 2002-08-29 Ligand Pharmaceuticals Incorporated Tricyclic androgen receptor modulator compounds

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
COMPTES RENDUS DES SEANCES DE L'ACADEMIE DES SCIENCES, SERIE C: SCIENCES CHIMIQUES, vol. 275, no. 19, 1972, pages 1105 - 1107 *
DATABASE CAPLUS [online] 1973, CAVE A. ET AL: "Structure of a new 4-alkylcoumarin isolated from Calophyllum inophyllum", XP002979346, Database accession no. 1973:58264 *
FAN W. ET AL: "Apocynins A-D: New Phenylpropanoid-substituted Flavan-3-ols isolated from Leaves of Apocynum venetum (Luobuma-Ye)", CHEM. PHARM. BULL., vol. 47, no. 7, 1999, pages 1049 - 1050, XP002979344 *
ITO H. ET AL: "Polyphenols from Eriobotrya japonica and Their Cytotoxicity against Human Oral Tumor Cell Lines", CHEM. PHARM. BULL., vol. 48, no. 5, 2000, pages 687 - 693, XP002979343 *
ITOIGAWA M. ET AL: "Cancer chemopreventive agents, 4-phenylcoumarins from Calophyllum inophyllum", CANCER LETTERS, vol. 169, no. 1, 2001, pages 15 - 19, XP002979339 *
JU Y. ET AL: "Cytotoxic Coumarins and Lignans from Extracts of the Northern Prickly Ash (Zanthoxylum americanum)", PHYTOTHERAPY RESEARCH, vol. 15, no. 5, 2001, pages 441 - 443, XP002979340 *
KAWAII S. ET AL: "Antiproliferative effect of isopentenylated coumarins on several cancer cell lines", ANTICANCER RESEARCH, vol. 21, no. 3B, 2001, pages 1905 - 1911, XP002979341 *
PALMER CJ ET AL: "Synthesis of the Calophyllum coumarins. Part 2", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 1: ORGANIC AND BIO-ORGANIC CHEMISTRY, no. 24, 1995, pages 3135 - 3152, XP002045573 *
PALMER CJ ET AL: "Sythesis of the Calophyllum Coumarins", TETRAHEDRON LETTERS, vol. 35, no. 30, 1994, pages 5363 - 5366, XP002148382 *
SATOH M. ET AL: "Cytotoxic Constituents from Erythroxylum catuaba Isolation and Cytotoxic Activities of Cinchonain", NATURAL MEDICINES, vol. 54, no. 2, 2000, pages 97 - 100, XP002979342 *
TANAKA T. ET AL: "Enantioselective total synthesis of anti HIV-1 active (+)-calanolide A through a quinine-catalyzed asymmetric intramolecular oxo-Michael addition", TETRAHEDRON LETTERS, vol. 41, no. 52, 2000, pages 10229 - 10232, XP004225164 *
XIONG Q. ET AL: "Hepatoprotective Effect of Apocynum venetum and its Active Constituents", PLANTA MEDICA, vol. 66, no. 2, 2000, pages 127 - 133, XP002979345 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009060835A1 (en) * 2007-11-05 2009-05-14 Kyoto University Novel ubiquilin-binding small molecule
JP5622856B2 (en) * 2010-09-16 2014-11-12 株式会社エコリソース研究所 Xanthone derivatives and uses thereof
CN112409368A (en) * 2020-11-23 2021-02-26 昆明医科大学 A class of C-4 substituted coumarin compounds and preparation method and application thereof

Also Published As

Publication number Publication date
JPWO2004066994A1 (en) 2006-05-18

Similar Documents

Publication Publication Date Title
US9549911B2 (en) Ginger metabolites and uses thereof
Baskar et al. Chemopreventive potential of β-sitosterol in experimental colon cancer model-an in vitro and in vivo study
Bądziul et al. Combined treatment with quercetin and imperatorin as a potent strategy for killing HeLa and Hep-2 cells
Qiu et al. Synthesis and evaluation of asymmetric curcuminoid analogs as potential anticancer agents that downregulate NF-κB activation and enhance the sensitivity of gastric cancer cell lines to irinotecan chemotherapy
JP2001501590A (en) Methods for treating malignant tumors with thyroxine analogs without significant hormonal activity
KR20080031266A (en) PAP Modulators and Cancer Treatment
Gaur et al. In vitro and in vivo synergistic interaction of substituted chalcone derivatives with norfloxacin against methicillin resistant Staphylococcus aureus
FR2921927A1 (en) IMIDAZO [1,2-A] QUINOXALINS AND DERIVATIVES FOR THE TREATMENT OF CANCER
WO2012136910A1 (en) Derivatives of 4-arylcoumarin and 4-arylquinoline, therapeutic uses thereof and method for synthesising same
CN112423743A (en) Activator of unfolded protein response
JP2005516938A (en) Isoindigo, derivatives of indigo and indirubin, and use in cancer treatment
Lin et al. N-Benzoyl-12-nitrodehydroabietylamine-7-one, a novel dehydroabietylamine derivative, induces apoptosis and inhibits proliferation in HepG2 cells
EP2694059A2 (en) Small molecule inhibitors of xbp1 splicing
Xia et al. W346 inhibits cell growth, invasion, induces cycle arrest and potentiates apoptosis in human gastric cancer cells in vitro through the NF-κB signaling pathway
Cui et al. Natural products targeting cancer stem cells: a revisit
CN115715201A (en) Glucose triptolide conjugate and application thereof
Nyein et al. Synthesis and anti-glioblastoma effects of artemisinin-isothiocyanate derivatives
US10947249B2 (en) Nimbolide analogs as anti-cancer agents and preparation thereof
JP2014517813A (en) Sulindac derivatives, their use and their preparation
WO2004066994A1 (en) Anticancer agent
US11098052B2 (en) 4-azapodophylotoxins compounds
US20100279964A1 (en) Angular Pyranocoumarins, Process for Preparation and Uses Thereof
Liu et al. Synthesis of a celastrol derivative as a cancer stem cell inhibitor through regulation of the STAT3 pathway for treatment of ovarian cancer
CA2409664C (en) Use of substituted acryloyl distamycin derivatives in the treatment of tumors associated with high levels of glutathione
WO2014048313A1 (en) Condensation product of theanine derivative and carboxylic acid coumarin derivative, intermediate of the condensation product, method for preparing same, and use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005504703

Country of ref document: JP

122 Ep: pct application non-entry in european phase