WO2006112330A1 - Inhibitory agent for estrone-3-sulfate transporter activity - Google Patents

Inhibitory agent for estrone-3-sulfate transporter activity Download PDF

Info

Publication number
WO2006112330A1
WO2006112330A1 PCT/JP2006/307751 JP2006307751W WO2006112330A1 WO 2006112330 A1 WO2006112330 A1 WO 2006112330A1 JP 2006307751 W JP2006307751 W JP 2006307751W WO 2006112330 A1 WO2006112330 A1 WO 2006112330A1
Authority
WO
WIPO (PCT)
Prior art keywords
estrone
theaflavin
cells
breast cancer
transporter
Prior art date
Application number
PCT/JP2006/307751
Other languages
French (fr)
Japanese (ja)
Inventor
Ikumi Tamai
Hikaru Yabuuchi
Original Assignee
Genomembrane 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 Genomembrane Co., Ltd. filed Critical Genomembrane Co., Ltd.
Publication of WO2006112330A1 publication Critical patent/WO2006112330A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/121Ketones acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to an inhibitor of transporter activity of an estrone-3-sulfate (estrone-3-sulfate) transporter, a breast cancer cell growth inhibitor, and a breast cancer therapeutic agent.
  • Transporters Membrane proteins that transport substances inside and outside cells are called transporters. When specific molecules bind to transporters embedded in lipid bilayers, the conformation changes and the substances are taken up. Or they are known to be discharged and transported. In recent years, powerful transporters such as organic transporters such as OAT1 that transport organic organic substances, organic cationic transporters such as OCT1 that transport organic cationic substances, PEPT1 that transport peptide substances, etc. Genes such as peptide transporters have been isolated and identified one after another. Some of these transporter genes are ubiquitous in normal tissues 'organs throughout the body, but others are known to be localized in specific tissues' organs such as kidney, liver and brain.
  • estrogen is a so-called female hormone that causes estrus in female animals.
  • estrogen has a steroid structure.
  • Estrone 0 . The biological action of g is 1 IU (international unit), which is used as an estrogen unit.
  • the secretory source is mainly the ovarian follicle and corpus luteum, but it is also secreted by the fetal placenta, adrenal gland, and testis during pregnancy.
  • the secretion of estrogens, such as ovarian force is governed by gonadotropins secreted from the anterior pituitary gland (descending sexual regulation), but conversely, estrogen also has a feedback effect on the pituitary system (ascending sex). Regulation), and it is said that the sexual cycle is established by the mutual relationship between the two.
  • estrogen acts through its receptor (receptor), and the action of estrogen Is the target tissue, not only the anterior pituitary gland and the mammary gland but also the whole body.
  • the main physiological functions are endometrial proliferation, myometrial development, expression of secondary sexual characteristics, establishment of menstrual cycle. Mediation, induction of maternal changes during pregnancy, and promotion of mammary duct growth and secretion.
  • Estrogens are metabolized mainly in the liver and become conjugated estrogens such as estrone trisulfate and are excreted in the urine. Clinically, it is used for treatment of amenorrhea and menstrual abnormalities, artificial movement of menstruation, menopause, hormone therapy for prostate cancer and breast cancer, and osteoporosis.
  • Breast cancer has an abnormal operation of the estrogen receptor system from the time of carcinogenesis, and continues to estrogen-dependent growth at an early stage, but gradually grows out of this control. This suggests that estrogen and its receptor are deeply involved in the development and progression of breast cancer, and its mechanism is still unclear.
  • Breast cancer is a malignant tumor that arises from the peripheral ducts of the epithelial tissue of the mammary gland and the acinar epidermis. Also in Japan, the death rate from breast cancer is on the rise due to the westernization of dietary habits and changes in lifestyle. The most common age is in the 40's.
  • Risk factors include history of breast cancer's family history, unmarried, elderly first birth, early menarche and late menopause, obesity, radiation exposure, high fat diet, and history of benign breast disease. Can do.
  • Hematogenous metastases are common in bone, lung, and liver.
  • the main symptom is a breast mass, surface irregularity, hardness, unclear borders, and low mobility.
  • Non-patent Document 1 2Z3 of breast cancer is estrogen-dependent (Non-patent Document 1), that is, cell proliferation is controlled by estrogen.
  • the biologically active form of estrogen is estradiol, which is synthesized via two main pathways (Non-patent document 2, Non-patent document 3).
  • One is the aromatase pathway where aromatase converts androgen to estrogen, and the other is the sulfatase pathway where steroid sulfatase converts estrone 3 sulfate to estrone.
  • estrone 3 sulfate-derived biological activity estrogen (Non-patent document 3, Non-patent document 3). Reference 4).
  • the circulating plasma concentration of estrone 3 sulfate is about 10 to 20 times that of unconjugated estrogens, and the half life of estrone 3 sulfate is longer than that of estradiol (Non-patent Documents 5 to 7) .
  • estrone 3 sulfate is thought to play an important role in the progression of breast cancer as a reservoir of active estrogens, even though estrone 3 sulfate itself does not exhibit much biological activity.
  • estrone trisulfate Stimulation of breast cancer cell proliferation by estrone trisulfate involves the following series of processes: uptake of estrone trisulfate into cells, desulfurization oxidation by estrogen sulfatase, type I 17 j8-hydroxysteroid Conversion of estrone to estradiol by dehydrogenase, binding to nuclear estrogen receptor, and regulation of gene transcription (Non-Patent Documents 8 to 11).
  • Well-studied forces of enzymes and receptors involved in breast cancer cell responses to estrone trisulfate The transport mechanism through which ligands such as estrone trisulfate cross the cell membrane is unknown.
  • Organic-on transporter (OAT, SLC22A) (Non-patent document 16) family and organic-on transport peptide (OATP, SLC21A, SLCO) (Non-patent document 17, Non-patent document 18)
  • OAT, SLC22A Non-patent document 16
  • OATP, SLC21A, SLCO organic-on transport peptide
  • Non-patent document 18 Some have already been known to transport estrone trisulfate. They are mainly expressed in the liver, kidney, brain, intestine, and other sites. Expression in breast cancer cells has not been reported. Their Km values for estrone 3 sulfate are in the range of 0.05 ⁇ to 59 ⁇ . In women, plasma levels of estrone 3 sulfate range from 1 to 10 ⁇ (19), so these or other transporters are the first step in hormone-dependent growth. It appears to play a role in the uptake of cancer into cancer cells.
  • Non-Patent Document 1 N Engl J Med 302: 78-90, 1980
  • Non-Patent Document 2 J Clin Endocrinol Metab 57: 1125-1128, 1983
  • Non-Patent Document 3 Breast Cancer Res Treat 7: 35-44, 1986
  • Non-Patent Document 4 Ann N Y Acad Sci 464: 126-137, 1986
  • Non-Patent Document 5 J Clin Endocrinol Metab 81: 1460-1464, 1996
  • Non-Patent Document 6 J Biol Chem 236: 1043-1050, 1961
  • Non-Patent Document 7 J Clin Invest 51: 1020-1033, 1972
  • Non-Patent Document 8 Endocrinology 138: 863-870, 1997
  • Non-Patent Document 9 Mol Endocrinol 11: 77-86, 1997
  • Non-Patent Document 10 Endocrinology 123: 1281-1287, 1988
  • Non-Patent Document 11 Breast Cancer 9: 296-302, 2002
  • Non-Patent Document 12 J Clin Endocrinol Metab 59: 1128-1132, 1984
  • Non-Patent Document 13 Endocrinology 106: 1079-1086, 1980
  • Non-Patent Document 14 J Clin Endocrinol Metab 81: 1460-1464, 1996
  • Non-Patent Document 15 J Steroid Biochem Mol Biol 73: 225-235, 2000
  • Non-Patent Document 16 Pflugers Arch 440: 337-350, 2000
  • Non-Patent Document 17 Biochim Biophys Acta 1609: 1-18, 2003
  • Non-Patent Document 18 Pflugers Arch 447: 653-665, 2003
  • Non-Patent Document 19 Hormone Res 27: 61-68, 1987
  • Non-Patent Document 20 J Clin Endocrinol Metab 88: 3902-3912, 2003
  • Non-Patent Document 21 J Clin Endocrinol Metab 88: 3902-3912, 2003
  • Non-Patent Document 22 Pharm Res 18: 1262-1269, 2001
  • Fig. 1 the following methods (blocks 1 to 4) are used to inhibit the growth of breast cancer cells that block upstream the signal transduction due to binding of estrogen and its receptor in breast cancer cells. It ’s known!
  • Block 1 Inhibition of estrogen synthesis from androgen
  • Fadrozole After menopause, it selectively inhibits aromatase, which acts as a rate-limiting enzyme for estrogen synthesis, and also prevents androgenic activity from converting to estrogen, thereby reducing the in vivo estrogen concentration and suppressing breast cancer growth.
  • aromatase acts as a rate-limiting enzyme for estrogen synthesis
  • Block 2 (competitive inhibition at estrogen receptor)
  • Estrogen receptors such as breast cancer tissues are competitively inhibited with estrogen and exert anti-breastogenic activity by showing anti-estrogenic activity
  • Block 3 Inhibition of estrogen synthesis from estrone sulfate
  • Conjugated estrogens are activated by estrogen sulfatase in the cell and promote cancer cell growth. Inhibits this deconjugating enzyme.
  • Block 4 (Cytotoxic effect using antibody to membrane protein)
  • Herceptin Some breast cancer cells highly express HER2 protein. Using this binding of HER2 and antibody, it exerts an antitumor effect due to antibody-dependent cytotoxicity.
  • an antitumor treatment is required unless an aromatase inhibitor, an estrogen sulfatase inhibitor, or an estrogen competitive inhibitor such as tamoxifen or toremifene is incorporated into breast cancer cells.
  • an aromatase inhibitor an estrogen sulfatase inhibitor, or an estrogen competitive inhibitor such as tamoxifen or toremifene is incorporated into breast cancer cells.
  • an aromatase inhibitor an estrogen sulfatase inhibitor
  • an estrogen competitive inhibitor such as tamoxifen or toremifene
  • the present inventors have intensively studied to solve the above problems, and examined the following block 5 (see Fig. 1) as a method for inhibiting the growth of breast cancer cells.
  • Block 5 inhibition of transporter activity
  • Anti-tumor effects appear in estrogen-sensitive breast cancer cells by inhibiting the uptake of estrone trisulfate, an estrogen source.
  • estrone 3 sulfate expressed on the cell surface It was found that the incorporation of estrone trisulfate into cells via the transporter was inhibited, and the proliferation of MCF-7 cells was suppressed, and the present invention was completed.
  • the present invention relates to (1) Genistein, Quercetin, Ginkgolide C, Theaflavin, Theaflavin 3-0-gallate (Th eaflavin 3-O-gallate), Noretin (Rutin), Canolecon (Chalcone), Daidzein (Daidzein), Daidine (Daidzin), Flavanone (Flavanone), Flavonol, Geraldol (Geraldol), Hesperetin (Hesperidin), flavone (Hesperidin), prione , Luteolin-7-O-darcoside (Luteolin-7-0-Glucoside), methoxycanolone (Methoxychalcone), 4, 1-methinole-1-7-methoxyhiso-isoflavone, 5— Morin (5— Morin), Myricetin (Naringenin), Naringenin (7-glycoside), Naringin (Naringin), Choose from Ohesperidin Dihydrochalcone, Nomilin,
  • FIG. 1 is a schematic diagram showing a molecular target for breast cancer.
  • FIG. 2 Results of inhibition of uptake of estrone trisulfate into breast cancer cells by various test substances It is a figure.
  • FIG. 3 is a diagram showing the results of suppression of cell growth promotion effect by estrone 3 sulfate by genistein, quercetin, ginkgolide C, theaflavin, and theaflavin 3-O-gallate.
  • FIG. 4 is a graph showing the results of inhibition of the incorporation of estrone trisulfate into breast cancer cells by various test substances.
  • Inhibitors of transporter activity of the estrone trisulfate transporter of the present invention, breast cancer cell growth inhibitors and breast cancer therapeutic agents include Genistein, Quercetin, Ginkgolide C, Theaflavin (Theaflavin), Theaflavin 3-O-gallate, Rutin, Chalcone, Daidzein, Daidzin, Flavanone, Flavonol, Di Geradolone (Geraldol), Hesperetin, Hesperidin, Ipriflavone, Luteolin-7-O-Dalcoside, Methoxychalcone, 4 , 1-Methyl-7-methoxy-isoflavone (4 '— mety ⁇ 7-methoxy-isoflavone), 5-Morin (5— Morin), Myricetin (Myricetin) Naringenin, Naringenin 7-glycoside, Naringin, Neohesperidin Dihydrochalcone, Nomilin, Primuletin, Ponc
  • genistein, quercetin, ginkgolide C, theaflavin, theaflavin 3-0-gallate are preferable examples. can do. These may be used alone or in combination of two or more. Furthermore, a compound obtained by chemically modifying these can also be used.
  • the inhibitor of transporter activity of the estrone 3 sulfate transporter of the present invention (hereinafter sometimes referred to as “the present inhibitor”) is important in identifying the estrone 3 sulfate transporter.
  • Candidate transporters for estrone 3 sulfate transporters include OAT 1, OAT2, OAT3, OAT4, OATP—A, OATP—B, OATP—C, OATP—D, O ATP-E, OATP-F, OATP-8, NTCP, MRPs, BCRP, etc. You can.
  • cells expressing the candidate estrone 3 sulfate transporter on the cell surface are cultured, and the uptake of the candidate estrone 3 sulfate into the cultured cells is measured in the presence of the inhibitor. Measures the degree of inhibition of the transporter activity of estrone 3 sulfate into cells by the method of 'evaluation' or cell force to express the candidate estrone 3 sulfate transporter on the cell surface.
  • a cell membrane fraction is isolated from the cells to be produced, and vesicles are produced from the cell membrane fraction to form isolated cell membrane vesicles.
  • Contacting the inhibitor, if estrone 3 uptake of sulfuric acid into vesicles by the present inhibitor Ku can be mentioned a method for measuring 'assess the degree of inhibition of uptake.
  • Cultivation of cells in the presence of the above estrone 3 sulfate and the present inhibitor uses a growth medium for the cells used, and at least cell proliferation (in the absence of the present inhibitor) under normal cell culture conditions.
  • Contact with the cell membrane fraction of the above estrone 3 sulfate and the inhibitor, or contact with the isolated cell membrane vesicle of the above estrone 3 sulfate and the inhibitor, which is desired to be carried out until significantly recognized. Can be performed by incubation in a growth medium for the cells used or an appropriate buffer.
  • the above-mentioned inhibitor measures the degree of inhibition of the transporter activity of estrone 3 sulfate into cells by 'measurement of the level of intracellular estrone 3 sulfate' and measures the degree of cell proliferation. 'You can list how to evaluate.
  • cultured human breast cancer cell lines such as MCF-7 cells, KPL-1, MKL-F, or cell lines derived from these cell lines, which are described later.
  • the method of measuring and evaluating the degree of cell proliferation can be used, but when using transformed cells expressing the candidate estrone trisulfate transporter, the method of measuring and evaluating the inhibitor concentration in the cell is used. Can be used.
  • the candidate estrone 3 sulfate transporter gene is expressed in Xenopus laevis oocytes, etc., and changes in membrane potential caused by substrate transport via the transporter are detected using two microelectrodes (current Can also be measured and evaluated by electrophysiological methods.
  • Cell force that expresses estrone trisulfate transporter on the cell surface As a method of isolating the cell membrane fraction, F. Pietri-Rouxel et al. (Eur. J.
  • a transformed cell expressing a candidate estrone 3 sulfate transporter can be used.
  • the origins of genes encoding NTCP, MRPs, BCRP are not particularly limited. Examples include humans, inu, ushi, horses, goats, hidges, monkeys, pigs, usagis, rats, mice, etc. The power that can be cited Human preference is preferred.
  • the method for expressing a gene or cDNA encoding a transporter as described above in a cell is not particularly limited, but Davis et al. (BASIC METHODS IN MOLECULAR BIOL OGY, 1986) and Sambrook et al. (MOLECULAR CLONING: A LABORATORY MANU AL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), for example, as described in many standard laboratory manuals such as calcium phosphate transfer, DEAE-dextran. Mediated transfusion, transvection, microinjection, cationic lipid mediated transfection, electoporation, open-cell introduction, scrape loading, ballistic introduction, infection, etc.
  • Examples of the above expression vectors include adenoviral vectors (Science, 252, 431-434, 1991) used for transient expression in all cells including non-dividing cells (other than blood cells), division, and the like. Retroviral vectors used for long-term expression in cells (Microbiology and Immunology, 158, 1-23, 1992) and adeno-associated viral vectors that can be introduced into non-pathogenic, non-dividing cells and used for long-term expression ( Curr. Top. Microbiol. Immunol, 158, 9 7-129, 1992), Papova Winores vector such as SV40, virus vector such as vaccinia Winores vector etc., ability to specifically mention ribosome etc. It is not limited to these.
  • an adenovirus vector capable of gene expression in cells with high efficiency is particularly preferable.
  • a transporter gene can be introduced into these expression vectors by a conventional method.
  • an expression vector can be constructed by inserting a transporter gene or the like downstream of an appropriate promoter in these expression vectors. it can.
  • the expression vector regulates the expression of the enhancer, terminator, etc. in addition to IRES (ribosome-binding site in the mRNA) to regulate the expression level of the transporter per cell. Including control array.
  • Host cells that express the transporter include insect cells such as Drosophila S2 and Spodoptera Sf9, Vero cells, HeLa cells, CHO cells, WI-38 cells, BHK cells, COS 7 cells, MDCK cells, C127 Cells, HKG cells, human kidney cell lines, CV-1 cells, LLC—MK2 cells, MDBK cells, MRC-5 cells, Caco-2 cells, HT29 cells, human lymphoblasts, Xenopus laevis, etc. And dhfr-deficient strains, HGPRT-deficient strains, and wabain resistant strains.
  • insect cells such as Drosophila S2 and Spodoptera Sf9, Vero cells, HeLa cells, CHO cells, WI-38 cells, BHK cells, COS 7 cells, MDCK cells, C127 Cells, HKG cells, human kidney cell lines, CV-1 cells, LLC—MK2 cells, MDBK cells, MRC-5 cells, Caco-2 cells, HT29 cells, human lymphoblasts,
  • CHO—K1 Choinese hamster ovary cells: ATCC CCL61
  • BHK Nomster kidney cells: ATCC CCL 10
  • COS-7 CV- Origin, SV—40 cells: ATCC CRL1651
  • Vero Cells African green monkey kidney cells: ATCC CCL81
  • human lymphoblasts IM-9, ATCC CCL159
  • Specific antibodies that can be specifically recognized include monoclonal antibodies, polyclonal antibodies, single chain antibodies, humanized antibodies, chimeric antibodies, bifunctional antibodies that can simultaneously recognize two epitopes, and the like. These antibodies are produced by administering cells expressing the estrone trisulfate transporter or the membrane fraction thereof to animals (preferably non-humans) using conventional protocols.
  • monoclonal antibodies The preparations include antibodies produced by continuous cell line cultures, the Hypridoma method (Nature 256, 495-497, 1975), the Trioma method, the human B cell Hypridoma method (Immunol ogy Today 4, 72 1983) and EBV-hybridoma method (MONOCLONAL ANTIBODIE S AND CANCER THERAPY, 77-96, Alan R. Liss, Inc., 1985).
  • the breast cancer cell growth inhibitor of the present invention can be used for identification of an estrone trisulfate transporter and also as a breast cancer therapeutic agent because it can suppress the growth of breast cancer cells.
  • an identification method a cell expressing a candidate estrone trisulfate transporter on the cell surface is cultured in the presence of the breast cancer cell growth inhibitor of the present invention, and the method for measuring and evaluating the degree of cell proliferation is exemplified. be able to.
  • the therapeutic agent for breast cancer of the present invention can also be used as a prophylactic agent for breast cancer.
  • a normal pharmaceutically acceptable carrier binder, stabilizer, enhancer.
  • Various preparation co-components such as a dosage form, a diluent, a pH buffer, a disintegrant, a solubilizer, a solubilizer, and an isotonic agent can be added.
  • These therapeutic agents can be administered orally or parenterally, such as powders, granules, tablets, capsules, syrups, suspensions, etc.
  • a dosage form such as a solution, emulsion, suspension or the like can be administered parenterally by injection.
  • compositions for oral administration When administered parenterally, it can be administered topically.
  • various conventional organic or inorganic carrier substances are used as pharmacologically acceptable carriers. Excipients such as pumps, lubricants such as talc and magnesium stearate, binders such as hydroxypropylcellulose and polybulurpyrrolidone, disintegrants such as carboxymethylcellulose, etc.
  • Solvents such as physiological saline alcohol, solubilizing agents such as polyethylene glycol and propylene glycol, suspending agents such as stearyltriethanolamine, sodium lauryl sulfate and lecithin, isotonic agents such as glycerin and D-manntol Agents, phosphates, acetates, buffers such as kenates, etc. can be blended. If necessary, formulation additives such as preservatives, antioxidants, colorants, sweeteners and the like can be combined.
  • water-soluble solvents such as distilled water and physiological saline
  • solubilizing agents such as sodium salicylate
  • isotonic agents such as sodium chloride sodium, glycerin, D-mannitol
  • Stabilizers such as human serum albumin
  • preservatives such as methylparaben
  • narcotics such as benzyl alcohol
  • the dosage of the therapeutic agent can be appropriately selected depending on the patient's weight and age, dosage form, symptoms, etc.
  • the active ingredient is usually about 0.001 to 500 mg as a single dose, It is preferable to administer 1 to 50 mg once to 3 times a day.
  • MCF-7 cells In the presence of excess estrone 3 sulfate, the inhibitory effects of various test substances on [ 3 H] estrone 3 sulfate uptake by MCF-7 cells (ATCC-HTB22) were examined. MCF-7 cells containing 125 mM NaCl, 4.8 mM KC1, 5.6 mM D-glucose, 1.2 mM CaCl, 1.2 mM KH PO, 1.2 mM MgSO and 25 mM Hepes, p
  • estrone 3 sulfate was examined using genistein, quercetin, ginkgolide C, theaflavin, and theaflavin 3-O-gallate. We examined whether this could be achieved by suppressing intracellular accumulation of estrone 3 sulfate.
  • MCF-7 cells 8 ⁇ 10 3 ZO.
  • estrone 3 sulfate (E 3S; Sigma Chemi cals) was present at a concentration of ⁇
  • BSP bromosulfophthalein
  • 30 ⁇ each dissolved in 0.3% dimethyl sulfoxide (DMSO).
  • DMSO dimethyl sulfoxide
  • estrogen competitive inhibitors such as aromatase inhibitors, estrogen sulfatase inhibitors, tamoxifen and toremifene

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Saccharide Compounds (AREA)
  • Pyrane Compounds (AREA)

Abstract

Disclosed is an agent for inhibiting the transporter activity of an estrone-3-sulfate transporter, an agent for inhibiting the growth of breast cancer cells, or an agent for the treatment of breast cancer, which does not need to be incorporated in cells, has excellent drug delivery property and has few side effects. The agent comprises at least one component selected from genistein, quercetin, ginkgolide C, theaflavin, theaflavin 3-o-gallate, chalcone, rutin, daidzein, daidzin, flavanone, flavonol, geraldol, hesperetin, hespereridine, ipriflavone, luteolin-7-o-glucoside, methoxychalcone, 4'-methyl-7-methoxy-isoflavone, 5-morin, myricetin, naringenin, naringenin-7-glycoside, naringin, neohesperidin dihydrochalcone, nomilin, primuletin, poncirin, scutellarein, catechin hydrate, and chemically modified products thereof.

Description

明 細 書  Specification
エストロン 3硫酸トランスポーター活性阻害剤  Estrone 3-sulfate transporter activity inhibitor
技術分野  Technical field
[0001] 本発明は、エストロン 3硫酸(エストロンー3—サルフェート; estrone- 3- sulfate)トラン スポーターのトランスポーター活性の阻害剤や乳癌細胞増殖抑制剤や乳癌治療剤 に関する。  The present invention relates to an inhibitor of transporter activity of an estrone-3-sulfate (estrone-3-sulfate) transporter, a breast cancer cell growth inhibitor, and a breast cancer therapeutic agent.
背景技術  Background art
[0002] 細胞内外の物質輸送を行う膜タンパク質等はトランスポーターと呼ばれ、脂質二重 膜中に埋め込まれているトランスポーターに特定の分子が結合すると、コンフオメ一 シヨンが変化して物質が取込みあるいは排出輸送されることが知られて 、る。近年、 力かるトランスポーター、例えば有機ァ-オン性物質を輸送する OAT1等の有機ァ ユオントランスポーター,有機カチオン性物質を輸送する OCT1等の有機カチオント ランスポーター,ペプチド性物質を輸送する PEPT1等のペプチドトランスポーターな どの遺伝子が相次いで単離 '同定されている。これらトランスポーター遺伝子は、全 身の正常組織'臓器に偏在しているものもあるが、腎臓,肝臓,脳などといった特定 の組織'臓器に局在するものも知られている。  [0002] Membrane proteins that transport substances inside and outside cells are called transporters. When specific molecules bind to transporters embedded in lipid bilayers, the conformation changes and the substances are taken up. Or they are known to be discharged and transported. In recent years, powerful transporters such as organic transporters such as OAT1 that transport organic organic substances, organic cationic transporters such as OCT1 that transport organic cationic substances, PEPT1 that transport peptide substances, etc. Genes such as peptide transporters have been isolated and identified one after another. Some of these transporter genes are ubiquitous in normal tissues 'organs throughout the body, but others are known to be localized in specific tissues' organs such as kidney, liver and brain.
[0003] 一方、エストロゲンは雌性動物に発情現象を起こす、いわゆる女性ホルモンであり、 天然に存在するエストロン(E ) ,エストラジオール(E ) ,エストリオール(E ) ,エステ  [0003] On the other hand, estrogen is a so-called female hormone that causes estrus in female animals. Naturally occurring estrone (E), estradiol (E), estriol (E), esthetic
1 2 3 トロール )と、それらと同様の生物活性を有する合成エストロゲンに分類される。ス  1 2 3 tralol) and synthetic estrogens with similar biological activity. The
4  Four
チルべステロールなどの合成型の一部を除 、て、エストロゲンはステロイド構造を有 する。エストロン 0.: gのもつ生物学的作用を 1IU (国際単位)とし、これがエストロ ゲンの単位として用いられて 、る。分泌源は主として卵巣の卵胞及び黄体であるが、 妊娠時の胎児胎盤系、副腎、精巣などからも分泌される。卵巣力ゝらのエストロゲンの 分泌は、下垂体前葉より分泌される性腺刺激ホルモンにより支配されるが(下行性調 節)、逆にエストロゲンによる間脳下垂体系へのフィードバック作用も認められ (上行 性調節)、両者の相互関係により性周期が成立するといわれている。  With the exception of some synthetic forms such as tilbestrol, estrogen has a steroid structure. Estrone 0 .: The biological action of g is 1 IU (international unit), which is used as an estrogen unit. The secretory source is mainly the ovarian follicle and corpus luteum, but it is also secreted by the fetal placenta, adrenal gland, and testis during pregnancy. The secretion of estrogens, such as ovarian force, is governed by gonadotropins secreted from the anterior pituitary gland (descending sexual regulation), but conversely, estrogen also has a feedback effect on the pituitary system (ascending sex). Regulation), and it is said that the sexual cycle is established by the mutual relationship between the two.
[0004] さらに、エストロゲンはその受容体(レセプター)を介して作用し、エストロゲンの作用 は標的組織である間脳 下垂体前葉 性器及び乳腺のみならず全身に及び、主な 生理作用は、子宮内膜の増殖,子宮筋の発育,第二次性徴の発現,月経周期の成 立の媒介,妊娠時の母体変化の惹起,乳腺管の増殖分泌促進などである。エストロ ゲンは主として肝臓において代謝をうけ、エストロン 3硫酸等の抱合型エストロゲンと なり尿中に排泄される。臨床的には、無月経や月経異常の治療,月経の人為的移動 ,更年期障害,前立腺癌や乳癌に対するホルモン療法,骨粗鬆症などに対して用い られている。 [0004] Furthermore, estrogen acts through its receptor (receptor), and the action of estrogen Is the target tissue, not only the anterior pituitary gland and the mammary gland but also the whole body.The main physiological functions are endometrial proliferation, myometrial development, expression of secondary sexual characteristics, establishment of menstrual cycle. Mediation, induction of maternal changes during pregnancy, and promotion of mammary duct growth and secretion. Estrogens are metabolized mainly in the liver and become conjugated estrogens such as estrone trisulfate and are excreted in the urine. Clinically, it is used for treatment of amenorrhea and menstrual abnormalities, artificial movement of menstruation, menopause, hormone therapy for prostate cancer and breast cancer, and osteoporosis.
[0005] 乳癌は、発癌時点からエストロゲン受容体システムの異常作動があり、初期はェスト ロゲン依存性の増殖を続けるが、次第にこの制御を離れた増殖となる。このことから、 乳癌の発生と進展にエストロゲンとその受容体が深く関与して 、ると考えられて 、る 力 その機序は未だ明らかではない。乳癌は乳腺の上皮組織の末梢乳管や腺房上 皮から発生する悪性腫瘍で、欧米では女性の癌死亡の第一位である。また、日本に おいても、近年の食生活の欧米化、生活様式の変化などに伴い乳がんによる死亡率 は上昇傾向にある。好発年齢は 40歳代で、リスク因子として、乳癌の既往'家族歴, 未婚,高齢初産,早期初潮と晩期閉経,肥満,放射線被爆,高脂肪食,良性乳腺疾 患の既往などを挙げることができる。リンパ節転移は腋窩,鎖骨下,胸骨傍に頻度が 高ぐ転移個数は予後と相関する。血行性転移は骨,肺,肝に多い。主症状は乳房 腫瘤であり、表面不整,硬,境界不明瞭で可動性が少ない。  [0005] Breast cancer has an abnormal operation of the estrogen receptor system from the time of carcinogenesis, and continues to estrogen-dependent growth at an early stage, but gradually grows out of this control. This suggests that estrogen and its receptor are deeply involved in the development and progression of breast cancer, and its mechanism is still unclear. Breast cancer is a malignant tumor that arises from the peripheral ducts of the epithelial tissue of the mammary gland and the acinar epidermis. Also in Japan, the death rate from breast cancer is on the rise due to the westernization of dietary habits and changes in lifestyle. The most common age is in the 40's. Risk factors include history of breast cancer's family history, unmarried, elderly first birth, early menarche and late menopause, obesity, radiation exposure, high fat diet, and history of benign breast disease. Can do. The number of metastases of lymph node metastasis, which frequently occur in the axilla, under the clavicle, and parasternally, correlates with prognosis. Hematogenous metastases are common in bone, lung, and liver. The main symptom is a breast mass, surface irregularity, hardness, unclear borders, and low mobility.
[0006] 乳癌の 2Z3は、エストロゲン依存性である(非特許文献 1)、すなわち、細胞増殖が エストロゲンによって制御されて 、る。エストロゲンの生物活性型がエストラジオール であり、 2つの主要経路を経て合成される(非特許文献 2、非特許文献 3)。 1つは、ァ ロマターゼがアンドロゲンをエストロゲンに変換するァロマターゼ経路であり、もう 1つ は、ステロイドスルファターゼがエストロン 3硫酸をエストロンに変換するスルファタ一 ゼ経路である。患者乳癌組織では、スルファターゼの活性はァロマターゼの活性と比 ベて 50〜200倍であるため、スルファターゼ経路は、エストロン 3硫酸由来生物活性 エストロゲンの主要な供給源である (非特許文献 3、非特許文献 4)。また、エストロン 3硫酸の循環血漿濃度は、非抱合エストロゲンの場合と比べて約 10〜20倍であり、 エストロン 3硫酸の半減期はエストラジオールの半減期よりも長い(非特許文献 5〜7) 。したがって、エストロン 3硫酸は、エストロン 3硫酸自体があまり生物活性を示さなくて も、活性エストロゲンの貯蔵庫として、乳癌の進行に重要な役割を果たすと考えられ ている。 [0006] 2Z3 of breast cancer is estrogen-dependent (Non-patent Document 1), that is, cell proliferation is controlled by estrogen. The biologically active form of estrogen is estradiol, which is synthesized via two main pathways (Non-patent document 2, Non-patent document 3). One is the aromatase pathway where aromatase converts androgen to estrogen, and the other is the sulfatase pathway where steroid sulfatase converts estrone 3 sulfate to estrone. In patient breast cancer tissues, the activity of sulfatase is 50-200 times that of aromatase, so the sulfatase pathway is a major source of estrone 3 sulfate-derived biological activity estrogen (Non-patent document 3, Non-patent document 3). Reference 4). The circulating plasma concentration of estrone 3 sulfate is about 10 to 20 times that of unconjugated estrogens, and the half life of estrone 3 sulfate is longer than that of estradiol (Non-patent Documents 5 to 7) . Thus, estrone 3 sulfate is thought to play an important role in the progression of breast cancer as a reservoir of active estrogens, even though estrone 3 sulfate itself does not exhibit much biological activity.
[0007] エストロン 3硫酸による乳癌細胞増殖の刺激には、下記の一連のプロセスが関わつ て 、る:エストロン 3硫酸の細胞への取り込み、エストロゲンスルファターゼによる脱硫 酸化、 I型の 17 j8—ヒドロキシステロイドデヒドロゲナーゼによるエストロンのエストラジ オールへの変換、核エストロゲン受容体への結合、及び遺伝子転写の制御(非特許 文献 8〜11)。エストロン 3硫酸に対する乳癌細胞の応答に関与している酵素や受容 体にっ 、てよく調べてみた力 エストロン 3硫酸などのリガンドが細胞膜を通過する輸 送メカニズムはわかっていない。エストロン 3硫酸の logP値は 1. 4であると報告されて おり、その値はエストロンの値(4. 4)やエストラジオールの値(3. 9)より著しく低いた め、エストロン及びエストラジオールが拡散性であるにも関わらず、拡散によって細胞 膜を通過することは容易ではないであろう。(非特許文献 12)。エストロン 3硫酸ととも にインキュベーションした後の MCF— 7細胞の細胞質画分中で、エストロン 3硫酸は インタタトな形で検出されており(非特許文献 13)、エストロゲン作用を示していた (非 特許文献 4、非特許文献 14、非特許文献 15)。したがって、ホルモン依存性増殖を 示す乳癌細胞へのエストロン 3硫酸の供給には、特異的トランスポーターが関与して いると思われる。  [0007] Stimulation of breast cancer cell proliferation by estrone trisulfate involves the following series of processes: uptake of estrone trisulfate into cells, desulfurization oxidation by estrogen sulfatase, type I 17 j8-hydroxysteroid Conversion of estrone to estradiol by dehydrogenase, binding to nuclear estrogen receptor, and regulation of gene transcription (Non-Patent Documents 8 to 11). Well-studied forces of enzymes and receptors involved in breast cancer cell responses to estrone trisulfate The transport mechanism through which ligands such as estrone trisulfate cross the cell membrane is unknown. The logP value for estrone 3 sulfate is reported to be 1.4, which is significantly lower than the values for estrone (4.4) and estradiol (3.9), so estrone and estradiol are diffusible. Nevertheless, it will not be easy to cross the cell membrane by diffusion. (Non-patent document 12). Estrone 3 sulfate was detected in an intact form in the cytoplasmic fraction of MCF-7 cells after incubation with estrone 3 sulfate (Non-patent document 13), and showed estrogenic action (Non-patent document) 4, Non-patent document 14, Non-patent document 15). Thus, a specific transporter appears to be involved in the supply of estrone trisulfate to breast cancer cells that exhibit hormone-dependent growth.
[0008] 有機ァ-オントランスポーター(OAT、 SLC22A) (非特許文献 16)ファミリー及び 有機ァ-オン輸送ペプチド (OATP、 SLC21A、 SLCO) (非特許文献 17、非特許 文献 18)ファミリーのメンバーのなかには、エストロン 3硫酸を輸送するものがあること が既に知られている。それらは主として肝臓、腎臓、脳、腸、その他の部位で発現す る力 乳癌細胞中での発現は報告されていない。エストロン 3硫酸に対するそれらの Km値は 0. 05 μ Μ〜59 μ Μの範囲内である。女性では、エストロン 3硫酸の血漿中 レベルは 1〜10ηΜの範囲で変動するため(非特許文献 19)、これらの又はその他の トランスポーターは、ホルモン依存性増殖の第一歩としての、エストロン 3硫酸の癌細 胞への取り込みにおいて、ある役割を果たしていると思われる。  [0008] Organic-on transporter (OAT, SLC22A) (Non-patent document 16) family and organic-on transport peptide (OATP, SLC21A, SLCO) (Non-patent document 17, Non-patent document 18) Some have already been known to transport estrone trisulfate. They are mainly expressed in the liver, kidney, brain, intestine, and other sites. Expression in breast cancer cells has not been reported. Their Km values for estrone 3 sulfate are in the range of 0.05 μΜ to 59 μΜ. In women, plasma levels of estrone 3 sulfate range from 1 to 10 ηΜ (19), so these or other transporters are the first step in hormone-dependent growth. It appears to play a role in the uptake of cancer into cancer cells.
[0009] 最近、 Pizzagalli et al.が、ヒトの乳腺におけるステロイドサルフェートトランスポーター である OATP— Bの発現を示唆している(非特許文献 20)。しかし、エストロゲン依存 性 MCF— 7細胞及び T 47D細胞中で O ATP— B mRN Aが検出されなかつたの に対し、 OATP— D&t )ATP—E mRNAはこれらの細胞中で発見されている( 非特許文献 21)。 OATP - D¾tJ^O ATP - E その遺伝子を HEK293細胞中で 一過性に発現させた場合にエストロン 3硫酸に対する輸送活性を示すため(非特許 文献 22)、癌細胞におけるエストロン 3硫酸の輸送に関与していると思われる。しかし 、エストロゲン依存性細胞においてトランスポーターが媒介するエストロン 3硫酸の取 り込みは、これまで調べられていな力 た力 肝臓、胎盤及び脳等のほかの組織に おける取り込みは報告されて ヽる。乳癌細胞の増殖メカニズムを充分に理解するた めには、かかる細胞へのエストロゲンの供給メカニズムを解明しなくてはならない。 非特許文献 1 : N Engl J Med 302: 78-90, 1980 [0009] Recently, Pizzagalli et al. Reported that steroid sulfate transporters in human mammary glands. This suggests the expression of OATP-B (Non-patent Document 20). However, OATP-B mRNA was not detected in estrogen-dependent MCF-7 and T47D cells, whereas OATP-D & t) ATP-E mRNA was found in these cells (non- Patent Document 21). OATP-D¾tJ ^ O ATP-E When the gene is transiently expressed in HEK293 cells, it exhibits transport activity against estrone trisulfate (Non-Patent Document 22) and is involved in the transport of estrone trisulfate in cancer cells It seems to have done. However, the transporter-mediated uptake of estrone trisulfate in estrogen-dependent cells has been reported to be uptake in other tissues such as the liver, placenta and brain, which have not been investigated so far. In order to fully understand the growth mechanism of breast cancer cells, the mechanism of estrogen supply to such cells must be elucidated. Non-Patent Document 1: N Engl J Med 302: 78-90, 1980
非特許文献 2 : J Clin Endocrinol Metab 57: 1125-1128, 1983 Non-Patent Document 2: J Clin Endocrinol Metab 57: 1125-1128, 1983
非特許文献 3 : Breast Cancer Res Treat 7: 35-44, 1986 Non-Patent Document 3: Breast Cancer Res Treat 7: 35-44, 1986
非特許文献 4 : Ann N Y Acad Sci 464: 126-137, 1986 Non-Patent Document 4: Ann N Y Acad Sci 464: 126-137, 1986
非特許文献 5 : J Clin Endocrinol Metab 81: 1460-1464, 1996 Non-Patent Document 5: J Clin Endocrinol Metab 81: 1460-1464, 1996
非特許文献 6 : J Biol Chem 236: 1043-1050, 1961 Non-Patent Document 6: J Biol Chem 236: 1043-1050, 1961
非特許文献 7 : J Clin Invest 51: 1020-1033, 1972 Non-Patent Document 7: J Clin Invest 51: 1020-1033, 1972
非特許文献 8 : Endocrinology 138: 863-870, 1997 Non-Patent Document 8: Endocrinology 138: 863-870, 1997
非特許文献 9 : Mol Endocrinol 11: 77-86, 1997 Non-Patent Document 9: Mol Endocrinol 11: 77-86, 1997
非特許文献 10 : Endocrinology 123: 1281-1287, 1988 Non-Patent Document 10: Endocrinology 123: 1281-1287, 1988
非特許文献 11 : Breast Cancer 9: 296-302, 2002 Non-Patent Document 11: Breast Cancer 9: 296-302, 2002
非特許文献 12 : J Clin Endocrinol Metab 59: 1128-1132, 1984 Non-Patent Document 12: J Clin Endocrinol Metab 59: 1128-1132, 1984
非特許文献 13 : Endocrinology 106: 1079-1086, 1980 Non-Patent Document 13: Endocrinology 106: 1079-1086, 1980
非特許文献 14 : J Clin Endocrinol Metab 81: 1460-1464, 1996 Non-Patent Document 14: J Clin Endocrinol Metab 81: 1460-1464, 1996
非特許文献 15 : J Steroid Biochem Mol Biol 73: 225-235, 2000 Non-Patent Document 15: J Steroid Biochem Mol Biol 73: 225-235, 2000
非特許文献 16 : Pflugers Arch 440: 337-350, 2000 Non-Patent Document 16: Pflugers Arch 440: 337-350, 2000
非特許文献 17 : Biochim Biophys Acta 1609: 1-18, 2003 Non-Patent Document 17: Biochim Biophys Acta 1609: 1-18, 2003
非特許文献 18 : Pflugers Arch 447: 653-665, 2003 非特許文献 19 : Hormone Res 27: 61-68, 1987 Non-Patent Document 18: Pflugers Arch 447: 653-665, 2003 Non-Patent Document 19: Hormone Res 27: 61-68, 1987
非特許文献 20 : J Clin Endocrinol Metab 88: 3902-3912, 2003  Non-Patent Document 20: J Clin Endocrinol Metab 88: 3902-3912, 2003
非特許文献 21 : J Clin Endocrinol Metab 88: 3902-3912, 2003  Non-Patent Document 21: J Clin Endocrinol Metab 88: 3902-3912, 2003
非特許文献 22 : Pharm Res 18: 1262-1269, 2001  Non-Patent Document 22: Pharm Res 18: 1262-1269, 2001
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0011] 乳癌細胞におけるエストロゲンとその受容体との結合によるシグナル伝達をアップ ストリームにブロックする乳癌細胞の増殖阻害については、図 1に示されるように、以 下の方法(ブロック 1〜4)が知られて!/、る。 [0011] As shown in Fig. 1, the following methods (blocks 1 to 4) are used to inhibit the growth of breast cancer cells that block upstream the signal transduction due to binding of estrogen and its receptor in breast cancer cells. It ’s known!
[0012] ブロック 1 (アンドロゲンからのエストロゲンの合成阻害) [0012] Block 1 (Inhibition of estrogen synthesis from androgen)
フアドロゾール:閉経後、エストロゲン合成の律速酵素として働くァロマターゼを選択 的に阻害して、アンドロゲン力もエストロゲンへの変換を妨げることにより、生体内のェ ストロゲン濃度を低下させ、乳癌の増殖を抑制する。  Fadrozole: After menopause, it selectively inhibits aromatase, which acts as a rate-limiting enzyme for estrogen synthesis, and also prevents androgenic activity from converting to estrogen, thereby reducing the in vivo estrogen concentration and suppressing breast cancer growth.
[0013] ブロック 2 (エストロゲンレセプターでの競合阻害) [0013] Block 2 (competitive inhibition at estrogen receptor)
タモキシフェン、トレミフェン:乳癌組織などのエストロゲン受容体に対し、エストロゲ ンと競合的に阻害し、抗エストロゲン作用を示すことによって抗乳癌作用を発揮する  Tamoxifen and toremifene: Estrogen receptors such as breast cancer tissues are competitively inhibited with estrogen and exert anti-breastogenic activity by showing anti-estrogenic activity
[0014] ブロック 3 (エストロン硫酸からのエストロゲン合成阻害) [0014] Block 3 (Inhibition of estrogen synthesis from estrone sulfate)
抱合型エストロゲンは細胞内でエストロゲンスルファターゼによって活性型となり、 癌細胞増殖を促進する。この脱抱合酵素を阻害する。  Conjugated estrogens are activated by estrogen sulfatase in the cell and promote cancer cell growth. Inhibits this deconjugating enzyme.
[0015] ブロック 4 (膜タンパク質に対する抗体を利用した細胞障害作用) [0015] Block 4 (Cytotoxic effect using antibody to membrane protein)
ハーセプチン:乳癌細胞には HER2タンパクが高発現するものがある。この HER2 と抗体の結合を利用し、抗体依存性細胞障害作用による抗腫瘍効果を発揮する。  Herceptin: Some breast cancer cells highly express HER2 protein. Using this binding of HER2 and antibody, it exerts an antitumor effect due to antibody-dependent cytotoxicity.
[0016] 乳癌細胞の増殖阻害における、上記ブロック 1〜4の方法では、ァロマターゼ阻害 剤やエストロゲンスルファターゼ阻害剤、あるいは、タモキシフェンゃトレミフェン等の エストロゲン競合阻害剤が乳癌細胞内に取り込まれないと抗腫瘍効果が発揮されな いこと力 ドラッグデリバリーの面で、また、細胞内に取り込まれると副作用の面で、そ れぞれ問題なしとはいえない。本発明の課題は、細胞内に取り込まれる必要がなぐ ドラッグデリバリー性に優れ、副作用がきわめて少ないエストロン 3硫酸トランスポータ 一のトランスポーター活性の阻害剤や乳癌細胞増殖抑制剤や乳癌治療剤を提供す ることにめる。 [0016] In the method of blocks 1 to 4 above for inhibiting the growth of breast cancer cells, an antitumor treatment is required unless an aromatase inhibitor, an estrogen sulfatase inhibitor, or an estrogen competitive inhibitor such as tamoxifen or toremifene is incorporated into breast cancer cells. Ineffectiveness In terms of drug delivery, and when incorporated into cells, there is no problem in terms of side effects. The problem of the present invention is that it is not necessary to be taken up into cells. Estrone trisulfate transporter with excellent drug delivery properties and extremely few side effects The aim is to provide a single transporter activity inhibitor, breast cancer cell growth inhibitor, and breast cancer therapeutic agent.
課題を解決するための手段  Means for solving the problem
[0017] 本発明者らは、上記課題を解決するために鋭意研究し、乳癌細胞の増殖阻害の方 法として、以下のブロック 5 (図 1参照)について検討した。 [0017] The present inventors have intensively studied to solve the above problems, and examined the following block 5 (see Fig. 1) as a method for inhibiting the growth of breast cancer cells.
ブロック 5 (トランスポーター活性の阻害)  Block 5 (inhibition of transporter activity)
エストロゲン感受性の乳癌細胞に対し、エストロゲンの供給源であるエストロン 3硫 酸の細胞内取込みを阻害することで抗腫瘍効果が現れる。  Anti-tumor effects appear in estrogen-sensitive breast cancer cells by inhibiting the uptake of estrone trisulfate, an estrogen source.
[0018] そして、ヒト乳癌培養細胞 MCF— 7細胞株をエストロン 3硫酸と、被検物質としての フラボノイド等の多くの天然有機化合物の存在下に培養したとき、細胞表面に発現す るエストロン 3硫酸トランスポーターを介してのエストロン 3硫酸の細胞内への取込み が阻害され、 MCF— 7細胞の増殖が抑制されることを見い出し、本発明を完成する に至った。 [0018] When the human breast cancer cell line MCF-7 cell line is cultured in the presence of estrone 3 sulfate and many natural organic compounds such as flavonoids as test substances, estrone 3 sulfate expressed on the cell surface It was found that the incorporation of estrone trisulfate into cells via the transporter was inhibited, and the proliferation of MCF-7 cells was suppressed, and the present invention was completed.
[0019] すなわち本発明は、(1)ゲニスティン(Genistein)、ケルセチン(Quercetin)、ギンコ ライド C (Ginkgolide C)、テアフラビン(Theaflavin)、テアフラビン 3— 0—ガレート(Th eaflavin 3— O— gallate)、ノレチン(Rutin)、カノレコン(Chalcone)、ダイゼイン(Daidzein)、 ダイジン(Daidzin)、フラバノン(Flavanone)、フラボノール(Flavonol)、ジェラルドール (Geraldol)、ヘスペレチン(Hesperetin)、ヘスペルジン(Hesperidin)、ィプリフラボン( Ipriflavone)、ルテオリン一 7— O—ダルコシド(Luteolin- 7- 0- Glucoside)、メトキシカ ノレコン(Methoxychalcone)、 4,一メチノレ一 7—メトキシ一イソフラボン(4'- mety卜 7- met hoxy— isoflavone)、 5—モリン (5— Morin)、ミリセチン (Myricetin)ゝナリンゲ-ン (Naring enin)、ナリンゲ-ン一 7—グノレコシド (Naringenin- 7- glycoside)、ナリンジン (Naringin) 、ネオヘスペリジンジノヽイドロカノレコン (Neohesperidin Dihydrochalcone)、ノミリン (No milin)、プリムレチン(Primuletin)、ポンシリン(Poncirin)、スクテラレイン(Scutellarein) 、力テキン水和物(Catechin Hydrate)、及びこれらを化学修飾した化合物から選ば れる 1種以上の成分を有効成分とするエストロン 3硫酸トランスポーターのトランスポ 一ター活性の阻害剤や、(2)ゲニスティン(Genistein)、ケルセチン(Quercetin)、ギ ンコライド C (Ginkgolide C)、テアフラビン(Theaflavin)、テアフラビン 3— O ガレート (Theaflavin3- 0- gallate)、ルチン(Rutin)、カルコン(Chalcone)、ダイゼイン(Daidzein )、ダイジン(Daidzin)、フラバノン(Flavanone)、フラボノール(Flavonol)、ジェラルドー ル(Geraldol)、ヘスペレチン(Hesperetin)、ヘスペルジン(Hesperidin)、ィプリフラボ ン(Ipriflavone)、ルテオリン一 7— O—ダルコシド(Luteolin- 7- 0- Glucoside)、メトキシ カルコン(Methoxychalcone)、 4,一メチル一 7—メトキシ一イソフラボン(4'- mety卜 7- m ethoxy— isoflavone)、 5—モリン (5— Morin)、ミリセチン (Myricetin)ゝナリンゲ-ン (Nari ngenin)、ナリンゲ-ン一 7—グノレコシド (Naringenin— 7— glycoside)、ナリンジン (Naringi n)、ネオヘスペリジンジノヽイドロカノレコン (Neohesperidin Dihydrochalcone)、ノミリン ( Nomilin)、プリムレチン(Primuletin)、ポンシリン(Poncirin)、スクテラレイン(Scutellarei n)、力テキン水和物(Catechin Hydrate)、及びこれらを化学修飾した化合物から選 ばれる 1種以上の成分を有効成分とする乳癌細胞増殖抑制剤や、(3)ゲニスティン( Genistein)、ケルセチン(Quercetin)、ギンコライド C (Ginkgolide C)、テアフラビン(Th eaflavin)、テアフラビン 3— O ガレート(Theaflavin 3-0- gallate)、ルチン(Rutin)、力 ノレコン (Chalcone)、ダイゼイン (Daidzein)、ダイジン (Daidzin)、フラノくノン (Flavanone )、フラボノール (Flavonol)、ジェラルドール (Geraldol)、ヘスペレチン(Hesperetin)、 ヘスペルジン(Hesperidin)、ィプリフラボン(Ipriflavone)、ルテオリン 7— 0—ダルコ シド(Luteolin- 7-0- Glucoside)、メトキシカノレコン(Methoxychalcone)、 4,一メチノレ一 7—メトキシ一イソフラボン(4'- metyl- 7- methoxy- isoflavone)、 5—モリン(5- Morin)、 ミリセチン(Myricetin)、ナリンゲ-ン(Naringenin)、ナリンゲ-ン一 7—ダルコシド(Nar ingenin- 7- glycoside)、ナリンジン (Naringin)、ネオヘスペリジンジノヽイドロカノレコン (N eohespendin Dihydrocnaicone)、ノ リン (Nomilin)、プリムレテン (Primuletinノ、ポンン リン(Poncirin)、スクテラレイン(Scutellarein)、及び力テキン水和物(Catechin Hydrat e)、及びこれらを化学修飾した化合物から選ばれる 1種以上の成分を有効成分とす る乳癌治療剤に関する。 That is, the present invention relates to (1) Genistein, Quercetin, Ginkgolide C, Theaflavin, Theaflavin 3-0-gallate (Th eaflavin 3-O-gallate), Noretin (Rutin), Canolecon (Chalcone), Daidzein (Daidzein), Daidine (Daidzin), Flavanone (Flavanone), Flavonol, Geraldol (Geraldol), Hesperetin (Hesperidin), flavone (Hesperidin), prione , Luteolin-7-O-darcoside (Luteolin-7-0-Glucoside), methoxycanolone (Methoxychalcone), 4, 1-methinole-1-7-methoxyhiso-isoflavone, 5— Morin (5— Morin), Myricetin (Naringenin), Naringenin (7-glycoside), Naringin (Naringin), Choose from Ohesperidin Dihydrochalcone, Nomilin, Primuletin, Poncirin, Scutellarein, Catechin Hydrate, and chemically modified compounds Inhibitors of the transporter activity of estrone trisulfate transporter with one or more active ingredients as active ingredients, (2) Genistein, Quercetin, Ninkolide C (Ginkgolide C), Theaflavin, Theaflavin 3-O gallate (Theaflavin3- 0-gallate), Rutin, Chalcone, Daidzein, Daidin, Flavanone, Flavonol, Geraldol, Hesperetin, Hesperidin, Ipriflavone, Luteolin-7-O-Dalcoside, Methoxychalcone, 4, 1-methyl 7-methoxy-isoflavone, 5- Morin, Myricetin, Naringen, Naringen 7—Gnorecoside (Naringenin—7-glycoside), Naringin, Neohesperidin Dihydrochalcone, Nomilin, Prep Breast cancer cell growth inhibition containing one or more ingredients selected from muruletin (Primuletin), poncilin (Poncirin), scutellarein (Scutellarei n), force techin hydrate (Catechin Hydrate), and compounds chemically modified from these. (3) Genistein, Quercetin, Ginkgolide C, Theaflavin, Theaflavin 3-O-gallate, Rutin, Power Norrecon (Chalcone), daidzein, daidzin, furanonone, flavonol, geraldol, hesperetin, hesperidin, ipriflavone, luteolin 7— 0—Dalcoside (Luteolin-7-0- Glucoside), Methoxychalcone, 4, 1-Methylolone 7-Methoxy-Isoflavone (4 '-metyl- 7-methoxy-isoflavone), 5- Morin, Myricetin, Naringenin, Naringen-7-glycoside, Naringin ( Naringin, Neohespendin Dihydrocnaicone, Nomilin, Primuletin, Poncirin, Scutellarein, and Catechin Hydrat e, and these The present invention relates to a therapeutic agent for breast cancer comprising one or more components selected from compounds chemically modified as active ingredients.
図面の簡単な説明 Brief Description of Drawings
[図 1]乳癌の分子ターゲットを示す模式図である。 FIG. 1 is a schematic diagram showing a molecular target for breast cancer.
[図 2]各種被験物質によるエストロン 3硫酸の乳癌細胞への取込み阻害の結果を示 す図である。 [Fig. 2] Results of inhibition of uptake of estrone trisulfate into breast cancer cells by various test substances It is a figure.
[図 3]エストロン 3硫酸による細胞増殖促進効果のゲニスティン、ケルセチン、ギンコラ イド C、テアフラビン、テアフラビン 3— O—ガレートによる抑制の結果を示す図である  FIG. 3 is a diagram showing the results of suppression of cell growth promotion effect by estrone 3 sulfate by genistein, quercetin, ginkgolide C, theaflavin, and theaflavin 3-O-gallate.
[図 4]各種被験物質によるエストロン 3硫酸の乳癌細胞への取込み阻害の結果を示 す図である。 FIG. 4 is a graph showing the results of inhibition of the incorporation of estrone trisulfate into breast cancer cells by various test substances.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0021] 本発明のエストロン 3硫酸トランスポーターのトランスポーター活性の阻害剤や乳癌 細胞増殖抑制剤や乳癌治療剤としては、ゲニスティン (Genistein)、ケルセチン (Que rcetin)、ギンコライド C (Ginkgolide C)、テアフラビン(Theaflavin)、テアフラビン 3— O ーガレート(Theaflavin 3- 0- gallate)、ルチン(Rutin)、カルコン(Chalcone)、ダイゼィ ン(Daidzein)、ダイジン(Daidzin)、フラバノン(Flavanone)、フラボノール (Flavonol)、 ジエラノレドーノレ (Geraldol)、ヘスペレチン (Hesperetin)、ヘスぺノレジン (Hesperidin)、 ィプリフラボン(Ipriflavone)、ルテオリン一 7— O—ダルコシド(Luteolin- 7- 0- Glucosid e)、メトキシカルコン(Methoxychalcone)、 4,一メチルー 7—メトキシ一イソフラボン(4' — mety卜 7— methoxy— isoflavone)、 5—モリン (5— Morin)、ミリセチン (Myricetin)ゝナリン ゲ-ン (Naringenin)、ナリンゲ-ン一 7—グノレコシド (Naringenin- 7- glycoside)、ナリン ジン (Naringin)、ネオヘスペリジンジノヽイドロカノレコン (Neohesperidin Dihydrochalcon e)、ノミリン(Nomilin)、プリムレチン(Primuletin)、ポンシリン(Poncirin)、スクテラレイ ン(Scutellarein)、力テキン水和物(Catechin Hydrate)を挙げることができるが、中で もゲニスティン、ケルセチン、ギンコライド C、テアフラビン、テアフラビン 3— 0—ガレ ートを好適に例示することができる。また、これらは 1種単独又は 2種以上を併用する ことができる。さらに、これらをィ匕学修飾したィ匕合物も用いることができる。  [0021] Inhibitors of transporter activity of the estrone trisulfate transporter of the present invention, breast cancer cell growth inhibitors and breast cancer therapeutic agents include Genistein, Quercetin, Ginkgolide C, Theaflavin (Theaflavin), Theaflavin 3-O-gallate, Rutin, Chalcone, Daidzein, Daidzin, Flavanone, Flavonol, Di Geradolone (Geraldol), Hesperetin, Hesperidin, Ipriflavone, Luteolin-7-O-Dalcoside, Methoxychalcone, 4 , 1-Methyl-7-methoxy-isoflavone (4 '— mety 卜 7-methoxy-isoflavone), 5-Morin (5— Morin), Myricetin (Myricetin) Naringenin, Naringenin 7-glycoside, Naringin, Neohesperidin Dihydrochalcone, Nomilin, Primuletin, Poncillin (Poncirin), scutellarein, and catechin hydrate can be mentioned. Among them, genistein, quercetin, ginkgolide C, theaflavin, theaflavin 3-0-gallate are preferable examples. can do. These may be used alone or in combination of two or more. Furthermore, a compound obtained by chemically modifying these can also be used.
[0022] 本発明のエストロン 3硫酸トランスポーターのトランスポーター活性の阻害剤(以下「 本件阻害剤」という場合がある)は、エストロン 3硫酸トランスポーターを同定する上で 重要である。エストロン 3硫酸トランスポーターの候補トランスポーターとしては、 OAT 1, OAT2, OAT3, OAT4, OATP—A, OATP— B, OATP— C, OATP—D, O ATP-E, OATP-F, OATP- 8, NTCP, MRPs, BCRPなどを好適に挙げるこ とができる。同定する方法としては、候補エストロン 3硫酸トランスポーターを細胞表面 に発現する細胞を培養し、培養した細胞内への候補エストロン 3硫酸の取り込みを本 件阻害剤の存在下において測定し、本件阻害剤による細胞内へのエストロン 3硫酸 のトランスポーター活性の阻害の程度を測定'評価する方法や、候補エストロン 3硫 酸トランスポーターを細胞表面に発現する細胞力 単離した細胞膜画分に、エストロ ン 3硫酸と本件阻害剤とを接触させ、本件阻害剤によるエストロン 3硫酸の細胞膜画 分への特異的結合の阻害の程度を測定'評価する方法や、候補エストロン 3硫酸トラ ンスポーターを細胞表面に発現する細胞から細胞膜画分を単離し、該細胞膜画分か ら小胞を作製して単離細胞膜小胞とし、該単離細胞膜小胞にエストロン 3硫酸と本件 阻害剤とを接触させ、本件阻害剤による小胞内へのエストロン 3硫酸の取込み量もし くは取込み量の阻害の程度を測定'評価する方法を挙げることができる。上記ェスト ロン 3硫酸と本件阻害剤との共存下の細胞の培養は、使用細胞の増殖用培地を用い 、通常の細胞培養条件下、(本件阻害剤非存在下の場合に)少なくとも細胞増殖が 有意に認められるまで行うことが望ましぐ上記エストロン 3硫酸と本件阻害剤との細 胞膜画分との接触や、上記エストロン 3硫酸と本件阻害剤との単離細胞膜小胞との接 触は、使用細胞の増殖用培地や適当な緩衝液中でインキュベーションすることにより 行うことができる。 The inhibitor of transporter activity of the estrone 3 sulfate transporter of the present invention (hereinafter sometimes referred to as “the present inhibitor”) is important in identifying the estrone 3 sulfate transporter. Candidate transporters for estrone 3 sulfate transporters include OAT 1, OAT2, OAT3, OAT4, OATP—A, OATP—B, OATP—C, OATP—D, O ATP-E, OATP-F, OATP-8, NTCP, MRPs, BCRP, etc. You can. As an identification method, cells expressing the candidate estrone 3 sulfate transporter on the cell surface are cultured, and the uptake of the candidate estrone 3 sulfate into the cultured cells is measured in the presence of the inhibitor. Measures the degree of inhibition of the transporter activity of estrone 3 sulfate into cells by the method of 'evaluation' or cell force to express the candidate estrone 3 sulfate transporter on the cell surface. Contact the sulfuric acid with the inhibitor and measure the degree of inhibition of the specific binding of estrone 3 sulfate to the cell membrane fraction by the inhibitor, or express the candidate estrone 3 sulfate transporter on the cell surface A cell membrane fraction is isolated from the cells to be produced, and vesicles are produced from the cell membrane fraction to form isolated cell membrane vesicles. Contacting the inhibitor, if estrone 3 uptake of sulfuric acid into vesicles by the present inhibitor Ku can be mentioned a method for measuring 'assess the degree of inhibition of uptake. Cultivation of cells in the presence of the above estrone 3 sulfate and the present inhibitor uses a growth medium for the cells used, and at least cell proliferation (in the absence of the present inhibitor) under normal cell culture conditions. Contact with the cell membrane fraction of the above estrone 3 sulfate and the inhibitor, or contact with the isolated cell membrane vesicle of the above estrone 3 sulfate and the inhibitor, which is desired to be carried out until significantly recognized. Can be performed by incubation in a growth medium for the cells used or an appropriate buffer.
上記本件阻害剤による細胞内へのエストロン 3硫酸のトランスポーター活性の阻害 の程度を測定'評価する方法としては、細胞内のエストロン 3硫酸濃度を測定'評価 する方法や、細胞増殖の程度を測定'評価する方法を挙げることができる。後述する MCF— 7細胞、 KPL— 1, MKL— F又はそれら細胞株由来の細胞株等のヒト乳癌 培養細胞株を用いる場合は、細胞内の本件阻害剤濃度を測定'評価する方法と、細 胞増殖の程度を測定'評価する方法とを利用することができるが、候補エストロン 3硫 酸トランスポーターを発現する形質転換細胞を用いる場合は、細胞内の本件阻害剤 濃度を測定'評価する方法を利用することができる。また、候補エストロン 3硫酸トラン スポーター遺伝子をアフリカッメガエル卵母細胞等に発現させ、 2本の微小電極を用 いて、トランスポーターを介した基質輸送に伴って生じる膜電位の変化を検出(電流 を測定)する電気生理学的方法により測定'評価することもできる。 [0024] エストロン 3硫酸トランスポーターを細胞表面に発現する細胞力 細胞膜画分を単 離する方法としては、 F. Pietri— Rouxelら(Eur. J. Biochem., 247, 1174-1179, 1997)の 方法など、従来公知の方法を用いることができ、また、細胞膜画分から小胞を作製し て単離細胞膜小胞とする方法としては、 J.E. Levele d. Biol. Chem., 252, 1990-199 7, 1977)の方法など、従来公知の方法を用いることができる。そしてまた、本件阻害 剤によるエストロン 3硫酸の細胞膜画分への特異的結合の阻害の程度を測定する方 法としては、放射性標識エストロン 3硫酸を用いた定常状態結合阻害カゝら平衡解離 定数 Kdを測定する方法を例示することができ、本件阻害剤による単離細胞膜小胞 内へのエストロン 3硫酸の取込み量もしくは取込み量の阻害の程度を測定する方法と しては、 I. Tamaiら(Biochim. Biophys. Acta, 1512, 273-284, 2001)記載の方法など、 従来公知の手法を細胞内エストロン 3硫酸の測定方法として用いることができる。 The above-mentioned inhibitor measures the degree of inhibition of the transporter activity of estrone 3 sulfate into cells by 'measurement of the level of intracellular estrone 3 sulfate' and measures the degree of cell proliferation. 'You can list how to evaluate. When using cultured human breast cancer cell lines such as MCF-7 cells, KPL-1, MKL-F, or cell lines derived from these cell lines, which are described later, The method of measuring and evaluating the degree of cell proliferation can be used, but when using transformed cells expressing the candidate estrone trisulfate transporter, the method of measuring and evaluating the inhibitor concentration in the cell is used. Can be used. In addition, the candidate estrone 3 sulfate transporter gene is expressed in Xenopus laevis oocytes, etc., and changes in membrane potential caused by substrate transport via the transporter are detected using two microelectrodes (current Can also be measured and evaluated by electrophysiological methods. [0024] Cell force that expresses estrone trisulfate transporter on the cell surface As a method of isolating the cell membrane fraction, F. Pietri-Rouxel et al. (Eur. J. Biochem., 247, 1174-1179, 1997) Conventionally known methods such as the method can be used, and as a method for producing vesicles from the cell membrane fraction to obtain isolated cell membrane vesicles, JE Leveled Biol. Chem., 252, 1990-199 7 , 1977) and the like can be used. In addition, as a method for measuring the degree of inhibition of the specific binding of estrone 3 sulfate to the cell membrane fraction by the inhibitor, steady-state binding inhibition using radiolabeled estrone 3 sulfate was used as an equilibrium dissociation constant Kd. As a method for measuring the amount of estrone 3 sulfate incorporated into isolated cell membrane vesicles by this inhibitor or the degree of inhibition of the amount incorporated, I. Tamai et al. ( Biochim. Biophys. Acta, 1512, 273-284, 2001) can be used as a method for measuring intracellular estrone trisulfate, such as a method described in the related art.
[0025] 上記エストロン 3硫酸トランスポーターを細胞表面に発現する細胞としては、候補ェ ストロン 3硫酸トランスポーターを発現する形質転換細胞を使用することができる。力 力る形質転換細胞の作製に使用される前記 OAT1, OAT2, OAT3, OAT4, OA TP -A, OATP-B, OATP-C, OATP— D, OATP— E, OATP— F, OATP -8, NTCP, MRPs, BCRPをコードする遺伝子の由来としては特に制限されるも のではなぐ例えばヒト,ィヌ,ゥシ,ゥマ,ャギ,ヒッジ,サル,ブタ,ゥサギ,ラット,マ ウス等を挙げることができる力 ヒト由来が好まし 、。  [0025] As the cell expressing the estrone 3 sulfate transporter on the cell surface, a transformed cell expressing a candidate estrone 3 sulfate transporter can be used. OAT1, OAT2, OAT3, OAT4, OATP-A, OATP-B, OATP-C, OATP—D, OATP—E, OATP—F, OATP-8, The origins of genes encoding NTCP, MRPs, BCRP are not particularly limited. Examples include humans, inu, ushi, horses, goats, hidges, monkeys, pigs, usagis, rats, mice, etc. The power that can be cited Human preference is preferred.
[0026] 上記のようなトランスポーターをコードする遺伝子や cDNAを細胞に発現させる方 法としては特に限定されないが、 Davisら(BASIC METHODS IN MOLECULAR BIOL OGY, 1986)及び Sambrookら(MOLECULAR CLONING: A LABORATORY MANU AL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989 )などの多くの標準的な実験室マニュアルに記載される方法、例えば、リン酸カルシゥ ムトランスフエクシヨン, DEAE—デキストラン媒介トランスフエクシヨン, トランスべクシ ヨン(transvection) ,マイクロインジェクション,カチオン性脂質媒介トランスフエクショ ン,エレクト口ポレーシヨン,开質導入,スクレープローデイング(scrape loading) ,弾丸 導入 (ballisticintroduction) ,感染等の遺伝子導入法により、トランスポーター遺伝子 を宿主細胞へ導入することにより発現させることができる力 トランスポーター遺伝子 を含む発現ベクターを上記宿主細胞に導入することが好ましぐ中でもトランスポータ 一遺伝子を含む発現ベクターを宿主細胞に感染させることにより、トランスポーターを 細胞に発現させることがより好ましい。 [0026] The method for expressing a gene or cDNA encoding a transporter as described above in a cell is not particularly limited, but Davis et al. (BASIC METHODS IN MOLECULAR BIOL OGY, 1986) and Sambrook et al. (MOLECULAR CLONING: A LABORATORY MANU AL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), for example, as described in many standard laboratory manuals such as calcium phosphate transfer, DEAE-dextran. Mediated transfusion, transvection, microinjection, cationic lipid mediated transfection, electoporation, open-cell introduction, scrape loading, ballistic introduction, infection, etc. Expression by introducing a transporter gene into a host cell by gene transfer method The ability to transport transporter genes Among them, it is more preferable to express the transporter in the cell by infecting the host cell with an expression vector containing the transporter gene, even though it is preferable to introduce the expression vector containing the above into the host cell.
[0027] 上記発現ベクターとしては、例えば、非分裂細胞を含む全ての細胞 (血球系以外) での一過性発現に用いられるアデノウイルスベクター(Science, 252, 431-434, 1991) や、分裂細胞での長期発現に用いられるレトロウイルスベクター(Microbiology and I mmunology, 158, 1-23, 1992)や、非病原性、非分裂細胞にも導入可能で、長期発 現に用いられるアデノ随伴ウィルスベクター(Curr. Top. Microbiol. Immunol, 158, 9 7-129, 1992)、 SV40のようなパポバウイノレスベクター、ワクシニアウイノレスベクタ一等 のウィルスベクターの他、リボソーム等を具体的に挙げることができる力 これらに限 定されるものではない。これらの中でも、高い効率で細胞における遺伝子発現が可 能なアデノウイルスベクターが特に好ましい。また、これらの発現ベクターへのトランス ポーター遺伝子の導入は常法によって行うことができ、例えばこれら発現ベクター中 の適当なプロモーターの下流にトランスポーター遺伝子等を挿入することにより発現 ベクターを構築することができる。また、発現ベクターはトランスポーター遺伝子及び マーカー遺伝子に加え、トランスポーターの細胞当たりの発現量を規格ィ匕するための IRES (mRNA内部のリボソーム結合サイト)の他、ェンハンサー,ターミネータ一等 発現を調節する制御配列を含んで 、てもよ 、。  [0027] Examples of the above expression vectors include adenoviral vectors (Science, 252, 431-434, 1991) used for transient expression in all cells including non-dividing cells (other than blood cells), division, and the like. Retroviral vectors used for long-term expression in cells (Microbiology and Immunology, 158, 1-23, 1992) and adeno-associated viral vectors that can be introduced into non-pathogenic, non-dividing cells and used for long-term expression ( Curr. Top. Microbiol. Immunol, 158, 9 7-129, 1992), Papova Winores vector such as SV40, virus vector such as vaccinia Winores vector etc., ability to specifically mention ribosome etc. It is not limited to these. Among these, an adenovirus vector capable of gene expression in cells with high efficiency is particularly preferable. In addition, a transporter gene can be introduced into these expression vectors by a conventional method. For example, an expression vector can be constructed by inserting a transporter gene or the like downstream of an appropriate promoter in these expression vectors. it can. In addition to the transporter gene and marker gene, the expression vector regulates the expression of the enhancer, terminator, etc. in addition to IRES (ribosome-binding site in the mRNA) to regulate the expression level of the transporter per cell. Including control array.
[0028] トランスポーターを発現する宿主細胞としては、ドロソフイラ S 2,スポドプテラ Sf 9等 の昆虫細胞, Vero細胞, HeLa細胞, CHO細胞, WI— 38細胞, BHK細胞, COS 7細胞, MDCK細胞, C127細胞, HKG細胞,ヒト腎細胞株, CV— 1細胞, LLC — MK2細胞, MDBK細胞, MRC— 5細胞, Caco— 2細胞, HT29細胞,ヒトリンノ 芽球細胞,アフリカッメガエル等の卵母細胞や、これらの dhfr欠損株, HGPRT欠損 株,ゥァバイン耐性株等を挙げることができる。具体的には、 CHO— K1 (チヤィニー ズハムスター卵巣細胞: ATCC CCL61) , BHK (ノヽムスター腎細胞: ATCC CCL 10) , COS - 7 (CV- l Origin, SV—40細胞: ATCC CRL1651) , Vero細胞( アフリカミドリザル腎細胞: ATCC CCL81) ,ヒトリンパ芽球細胞(IM— 9, ATCC CCL159)を f列示することができる。 [0029] トランスポーターは細胞膜表面に発現する膜タンパク質であるため、エストロン 3硫 酸トランスポーターに対する中和抗体は細胞外力 選択的に標的を阻害する可能性 があり、このことから本件阻害剤によりエストロン 3硫酸トランスポーターを同定すること ができると、エストロン 3硫酸トランスポーターに対する中和抗体を作製することができ る。特異的に認識する抗体としては、モノクローナル抗体,ポリクローナル抗体,一本 鎖抗体,ヒト化抗体,キメラ抗体, 2つのェピトープを同時に認識することができる二 機能性抗体等を例示することができる。これら抗体は、慣用のプロトコールを用いて、 動物 (好ましくはヒト以外)にエストロン 3硫酸トランスポーターを膜表面に発現してい る細胞やその細胞膜画分を投与することにより産生され、例えばモノクローナル抗体 の調製には、連続細胞系の培養物により産生される抗体をもたらす、ハイプリドーマ 法(Nature 256, 495-497, 1975) ,トリオ一マ法,ヒト B細胞ハイプリドーマ法(Immunol ogy Today 4, 72, 1983)及び EBV—ハイブリドーマ法(MONOCLONAL ANTIBODIE S AND CANCER THERAPY, 77-96, Alan R.Liss, Inc., 1985)など任意の方法を用い ることがでさる。 [0028] Host cells that express the transporter include insect cells such as Drosophila S2 and Spodoptera Sf9, Vero cells, HeLa cells, CHO cells, WI-38 cells, BHK cells, COS 7 cells, MDCK cells, C127 Cells, HKG cells, human kidney cell lines, CV-1 cells, LLC—MK2 cells, MDBK cells, MRC-5 cells, Caco-2 cells, HT29 cells, human lymphoblasts, Xenopus laevis, etc. And dhfr-deficient strains, HGPRT-deficient strains, and wabain resistant strains. Specifically, CHO—K1 (Chinese hamster ovary cells: ATCC CCL61), BHK (Nomster kidney cells: ATCC CCL 10), COS-7 (CV- Origin, SV—40 cells: ATCC CRL1651), Vero Cells (African green monkey kidney cells: ATCC CCL81) and human lymphoblasts (IM-9, ATCC CCL159) can be shown in column f. [0029] Since the transporter is a membrane protein expressed on the cell membrane surface, neutralizing antibodies against the estrone trisulphate transporter may selectively inhibit the target against extracellular force. Once the trisulfate transporter can be identified, neutralizing antibodies against the estrone trisulfate transporter can be generated. Specific antibodies that can be specifically recognized include monoclonal antibodies, polyclonal antibodies, single chain antibodies, humanized antibodies, chimeric antibodies, bifunctional antibodies that can simultaneously recognize two epitopes, and the like. These antibodies are produced by administering cells expressing the estrone trisulfate transporter or the membrane fraction thereof to animals (preferably non-humans) using conventional protocols. For example, monoclonal antibodies The preparations include antibodies produced by continuous cell line cultures, the Hypridoma method (Nature 256, 495-497, 1975), the Trioma method, the human B cell Hypridoma method (Immunol ogy Today 4, 72 1983) and EBV-hybridoma method (MONOCLONAL ANTIBODIE S AND CANCER THERAPY, 77-96, Alan R. Liss, Inc., 1985).
[0030] 本発明の乳癌細胞増殖抑制剤は、エストロン 3硫酸トランスポーターの同定用として 、また乳癌細胞の増殖を抑制しうることから乳癌治療剤として用いることができる。同 定する方法としては、候補エストロン 3硫酸トランスポーターを細胞表面に発現する細 胞を、本発明の乳癌細胞増殖抑制剤の存在下において培養し、細胞増殖の程度を 測定'評価する方法を挙げることができる。  [0030] The breast cancer cell growth inhibitor of the present invention can be used for identification of an estrone trisulfate transporter and also as a breast cancer therapeutic agent because it can suppress the growth of breast cancer cells. As an identification method, a cell expressing a candidate estrone trisulfate transporter on the cell surface is cultured in the presence of the breast cancer cell growth inhibitor of the present invention, and the method for measuring and evaluating the degree of cell proliferation is exemplified. be able to.
[0031] 本発明の乳癌治療剤は、乳癌の予防薬としても用いることができ、医薬用の治療剤 として用いる場合は、薬学的に許容される通常の担体、結合剤、安定化剤、賦形剤、 希釈剤、 pH緩衝剤、崩壊剤、可溶化剤、溶解補助剤、等張剤などの各種調剤用配 合成分を添加することができる。これら治療剤は、経口的又は非経口的に投与するこ とができ、例えば粉末、顆粒、錠剤、カプセル剤、シロップ剤、懸濁液等の剤型で経 口的に投与することができ、あるいは、例えば溶液、乳剤、懸濁液等の剤型にしたも のを注射の型で非経口投与することができる。非経口投与する場合、局所に直接投 与することができる。経口的に投与する製剤の場合、薬理学的に許容される担体とし ては、慣用の各種有機あるいは無機担体物質が用いられ、例えば錠剤には乳糖、デ ンプン等の賦形剤、タルク、ステアリン酸マグネシウム等の滑沢剤、ヒドロキシプロピル セルロース、ポリビュルピロリドン等の結合剤、カルボキシメチルセルロース等の崩壊 剤等を配合することができ、懸濁液製剤には生理的食塩水アルコール等の溶剤、ポ リエチレングリコール、プロピレングリコール等の溶解補助剤、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、レシチン等の懸濁化剤、グリセリン、 D—マン-トー ル等の等張化剤、リン酸塩、酢酸塩、クェン酸塩等の緩衝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配 合することもできる。非経口的に投与する製剤の場合、蒸留水、生理的食塩水等の 水溶性溶剤、サリチル酸ナトリウム等の溶解補助剤、塩ィ匕ナトリウム、グリセリン、 D— マン-トール等の等張化剤、ヒト血清アルブミン等の安定化剤、メチルパラベン等の 保存剤、ベンジルアルコール等の局麻剤を配合することができる。治療薬の投与量 は、患者の体重や年齢、投与形態、症状等により適宜選定することができるが、例え ば成人に投与する場合、有効成分を通常 1回量として約 0. 001〜500mg、好ましく は l〜50mgを 1日 1回〜 3回投与するのが望ましい。 [0031] The therapeutic agent for breast cancer of the present invention can also be used as a prophylactic agent for breast cancer. When used as a therapeutic agent for pharmaceuticals, a normal pharmaceutically acceptable carrier, binder, stabilizer, enhancer. Various preparation co-components such as a dosage form, a diluent, a pH buffer, a disintegrant, a solubilizer, a solubilizer, and an isotonic agent can be added. These therapeutic agents can be administered orally or parenterally, such as powders, granules, tablets, capsules, syrups, suspensions, etc. Alternatively, for example, a dosage form such as a solution, emulsion, suspension or the like can be administered parenterally by injection. When administered parenterally, it can be administered topically. In the case of preparations for oral administration, various conventional organic or inorganic carrier substances are used as pharmacologically acceptable carriers. Excipients such as pumps, lubricants such as talc and magnesium stearate, binders such as hydroxypropylcellulose and polybulurpyrrolidone, disintegrants such as carboxymethylcellulose, etc. Solvents such as physiological saline alcohol, solubilizing agents such as polyethylene glycol and propylene glycol, suspending agents such as stearyltriethanolamine, sodium lauryl sulfate and lecithin, isotonic agents such as glycerin and D-manntol Agents, phosphates, acetates, buffers such as kenates, etc. can be blended. If necessary, formulation additives such as preservatives, antioxidants, colorants, sweeteners and the like can be combined. In the case of preparations administered parenterally, water-soluble solvents such as distilled water and physiological saline, solubilizing agents such as sodium salicylate, isotonic agents such as sodium chloride sodium, glycerin, D-mannitol, Stabilizers such as human serum albumin, preservatives such as methylparaben, and narcotics such as benzyl alcohol can be added. The dosage of the therapeutic agent can be appropriately selected depending on the patient's weight and age, dosage form, symptoms, etc. For example, when administered to an adult, the active ingredient is usually about 0.001 to 500 mg as a single dose, It is preferable to administer 1 to 50 mg once to 3 times a day.
[0032] 以下、実施例により本発明をより具体的に説明するが、本発明の技術的範囲はこ れらの例示に限定されるものではない。 [0032] Hereinafter, the present invention will be described more specifically with reference to examples, but the technical scope of the present invention is not limited to these examples.
実施例 1  Example 1
[0033] (エストロン 3硫酸の取り込み活性の阻害)  [0033] (Inhibition of uptake activity of estrone 3 sulfate)
過剰のエストロン 3硫酸の存在下、 MCF—7細胞(ATCC—HTB22)による [3H] エストロン 3硫酸の取り込みに対する、種々の被験物質の阻害作用を調べた。 MCF — 7細胞を 125mMの NaCl、 4. 8mMの KC1、 5. 6mMの D—グルコース、 1. 2mM の CaCl、 1. 2mMの KH PO、 1. 2mMの MgSO及び 25mMの Hepesを含み、 pIn the presence of excess estrone 3 sulfate, the inhibitory effects of various test substances on [ 3 H] estrone 3 sulfate uptake by MCF-7 cells (ATCC-HTB22) were examined. MCF-7 cells containing 125 mM NaCl, 4.8 mM KC1, 5.6 mM D-glucose, 1.2 mM CaCl, 1.2 mM KH PO, 1.2 mM MgSO and 25 mM Hepes, p
2 2 4 4 2 2 4 4
H7. 4に調製された輸送培地に懸濁した。輸送培地中の細胞懸濁液並びに非標識 ImMエストロン 3硫酸、 3. 6nMの [3H]エストロン 3硫酸及びそれぞれ 30 Μの被 験化合物を含む溶液を、 10分間 37°Cで個別にインキュベーションして輸送を開始さ せた。適時、混合液を 150 μ Lずつ取り除き、シリコーンオイル(SH550、 Toray Dow Corning Co.社製)混合液の層、及び密度 1. 03の流動パラフィン (Wako Pure Chem ical Industries社製)を通しての遠心濾過によって、輸送培地から細胞を分離した。各 細胞ペレットを 3Nの KOH中で可溶化し、 HC1で中和した。その後、液体シンチレ一 シヨン流体としてクリアゾルー 1 (Nacalai tesque社製)を用いた液体シンチレーシヨン力 ゥンターで、関連する放射能を測定した。結果を図 2に示す。その結果、ゲニスティン 、ケルセチン、ギンコライド C、テアフラビン、テアフラビン 3— O—ガレートを用いたと き、エストロン 3硫酸の取り込み活性が強く阻害されることがわ力つた。 It was suspended in the transport medium prepared in H7.4. Cell suspension in transport medium and a solution containing unlabeled ImM estrone trisulfate, 3.6 nM [ 3 H] estrone trisulfate and 30 μL of each test compound individually for 10 minutes at 37 ° C. And started transportation. When appropriate, remove 150 μL of the mixture and centrifuge through a layer of silicone oil (SH550, Toray Dow Corning Co.) and liquid paraffin of density 1.03 (Wako Pure Chemical Industries). The cells were separated from the transport medium by each Cell pellets were solubilized in 3N KOH and neutralized with HC1. Thereafter, the associated radioactivity was measured with a liquid scintillation force counter using Clearsol 1 (manufactured by Nacalai Tesque) as the liquid scintillation fluid. The result is shown in figure 2. As a result, when genistein, quercetin, ginkgolide C, theaflavin, and theaflavin 3-O-gallate were used, it was found that the uptake activity of estrone 3 sulfate was strongly inhibited.
実施例 2  Example 2
[0034] (エストロン 3硫酸による細胞増殖促進効果の抑制)  [0034] (Inhibition of cell growth promoting effect by estrone trisulfate)
MCF— 7細胞 (ATCC— HTB22)のエストロン 3硫酸による細胞増殖促進効果の 抑制を、ゲニスティン、ケルセチン、ギンコライド C、テアフラビン、テアフラビン 3— O ーガレートを用いて調べた。エストロン 3硫酸の細胞内蓄積を抑えることで達成される か否かを検討した。 MCF— 7細胞を 8 X 103個 ZO. 32cm2の濃度で 96ゥエル プレ ートに播種し 1日培養した後、 ΙΟΟηΜの濃度のエストロン 3硫酸(E 3S ; Sigma Chemi cals社製)の存在下又は不存在下に、ポジティブコントロールとしての 30 μ Μのブロ モスルフオフタレイン(BSP : Sigma Chemicals社製)、及び、 0. 3%ジメチルスルフォ キシド(DMSO)に溶解したそれぞれ 30 μ Μのゲニスティン、ケルセチン、ギンコライ ドじ、テアフラビン、テアフラビン 3— Ο—ガレートを含む培地をそれぞれ添カ卩し、さら に 3日間培養した。その後、細胞量を測定した。結果を図 3に示す。図 3から明らかな ように、コントロールであるエストロン 3硫酸単独存在下及び 0. 3%DMSOに溶解し たエストロン 3硫酸存在下においては、細胞増殖促進効果が観察された。一方、 100 nMの濃度のエストロン 3硫酸(E 3S)の存在下、 0. 3%DMSOに溶解したそれぞれ 30 μ Μのゲ-スティン、ケルセチン、ギンコライド C、テアフラビン、テアフラビン 3— O ーガレートを添カ卩した場合、 BSPを添カ卩した場合と同様に、コントロールに比べて細 胞増殖が阻害された。 Inhibition of the cell growth promoting effect of estrone 3 sulfate on MCF-7 cells (ATCC—HTB22) was examined using genistein, quercetin, ginkgolide C, theaflavin, and theaflavin 3-O-gallate. We examined whether this could be achieved by suppressing intracellular accumulation of estrone 3 sulfate. MCF-7 cells 8 × 10 3 ZO. Seed in 96-well plate at a concentration of 32 cm 2 and cultured for 1 day, then estrone 3 sulfate (E 3S; Sigma Chemi cals) was present at a concentration of ΙΟΟηΜ In the absence or absence of 30 μΜ bromosulfophthalein (BSP: Sigma Chemicals) as a positive control and 30 μΜ each dissolved in 0.3% dimethyl sulfoxide (DMSO). Medium containing genistin, quercetin, ginkgolide, theaflavin, and theaflavin 3- 3-gallate were added, and cultured for another 3 days. Thereafter, the amount of cells was measured. The results are shown in Figure 3. As is clear from FIG. 3, cell growth promoting effects were observed in the presence of estrone 3 sulfate alone as a control and in the presence of estrone 3 sulfate dissolved in 0.3% DMSO. On the other hand, 30 μΜ of gestin, quercetin, ginkgolide C, theaflavin, and theaflavin 3-O-gallate dissolved in 0.3% DMSO in the presence of 100 nM estrone 3 sulfate (E 3S) were added. In the same manner as in the case of supplementing with BSP, cell growth was inhibited compared to the control.
実施例 3  Example 3
[0035] (エストロン 3硫酸の取り込み活性の阻害)  [0035] (Inhibition of estrone trisulfate uptake activity)
実施例 1と同様にして、各種被験物質によるエストロン 3硫酸の取り込み活性の阻 害作用を調べた。結果を図 4に示す。その結果、カルコン、ダイゼイン、ダイジン、フラ バノン、フラボノール、ジヱラルドール、ヘスペレチン、ヘスペルジン、ィプリフラボン、 ルテオリン一 7— O—ダルコシド、メトキシカルコン、 4'—メチル一 7—メトキシ一イソフ ラボン、 5—モリン(5- Morin)、ミリセチン、ナリンゲ-ン、ナリンゲ-ンー 7—ダルコシド 、ナリンジン、ネオヘスペリジンジハイド口カルコン、ノミリン、プリムレチン、ポンシリン、 スクテラレイン、及び力テキン水和物に、強いエストロン 3硫酸の取り込み活性阻害作 用が認められた。 In the same manner as in Example 1, the inhibitory effect of various test substances on the uptake activity of estrone 3 sulfate was examined. The results are shown in Fig. 4. As a result, chalcone, daidzein, daidzin, flavanone, flavonol, dizalraldol, hesperetin, hesperzin, ypriflavone, Luteolin 7-O-Dalcoside, Methoxychalcone, 4'-Methyl-1-7-methoxy-Isoflavone, 5-Morine, Myricetin, Naringen, Naringen 7-Dalcoside, Naringin, Neohesperidin Hyde mouth chalcone, nomiline, primuletin, poncillin, scutellarein, and force tekin hydrate were found to have a strong inhibitory action on estrone trisulfate uptake.
産業状の利用可能性 Industrial applicability
本発明によると、ァロマターゼ阻害剤やエストロゲンスルファターゼ阻害剤、タモキ シフェンゃトレミフェン等のエストロゲン競合阻害剤と異なり、細胞内に取り込まれる必 要がなぐドラッグデリバリー性に優れ、副作用がきわめて少ないエストロン 3硫酸トラ ンスポーターの阻害剤や乳癌細胞増殖抑制剤や乳癌治療剤を提供することができる  According to the present invention, unlike estrogen competitive inhibitors such as aromatase inhibitors, estrogen sulfatase inhibitors, tamoxifen and toremifene, it has excellent drug delivery properties that do not need to be taken into cells and has very few side effects. Can provide a transporter inhibitor, a breast cancer cell growth inhibitor, and a breast cancer therapeutic agent.

Claims

請求の範囲 The scope of the claims
[1] ゲ-スティン(Genistein)、ケルセチン(Quercetin)、ギンコライド C (GinkgolideC)、 テアフラビン(Theaflavin)、テアフラビン 3— O—ガレート(Theaflavin 3-O-gallate)、 ルチン(Rutin)、カルコン(Chalcone)、ダイゼイン(Daidzein)、ダイジン(Daidzin)、フ ラノ ノン (Flavanone)、フラボノーノレ (Flavonol)、ジエラノレドーノレ (Geraldol)、ヘスペレ チン(Hesperetin)、ヘスペルジン(Hesperidin)、ィプリフラボン(Ipriflavone)、ルテオリ ン一 7— O—ダルコシド(Luteolin- 7- 0- Glucoside)、メトキシカルコン(Methoxychalco ne)、 4,一メチノレ一 7—メトキシ一イソフラボン(4'— mety卜 7— methoxy— isoflavone)、 5— モリン(5- Morin)、ミリセチン(Myricetin)、ナリンゲ-ン(Naringenin)、ナリンゲ-ン一 7—グノレコシド (Naringenin- 7- glycoside)、ナリンジン (Naringin)、ネオヘスペリジンジ ノヽイドロカノレコン (Neohesperidin Dihydrochalcone)、ノミリン (Nomilin)、プリムレチン ( Primuletin)、ポンシリン(Poncirin)、スクテラレイン(Scutellarein)、力テキン水和物(Ca techin Hydrate)、及びこれらを化学修飾した化合物から選ばれる 1種以上の成分を 有効成分とするエストロン 3硫酸トランスポーターのトランスポーター活性の阻害剤。  [1] Genistein, Quercetin, Ginkgolide C, Theaflavin, Theaflavin 3-O-gallate, Rutin, Chalcone , Daidzein, Daidzin, Flavanone, Flavonol, Geraldol, Hesperetin, Hesperidin, Ipriflavone, Luteolin 1—O-Dalcoside (Luteolin-7-0-Glucoside), 4, Methoxychalcone, 4, 1-Methylolone 7-Methoxy-isoflavone (4'—mety 卜 7-methoxy-isoflavone), 5-—Molin ( 5- Morin), Myricetin, Naringenin, Naringenin 7-glycoside, Naringin, Neohesperidin dinoid One or more selected from canorecon (Neohesperidin Dihydrochalcone), nomilin, primuletin, poncilin, scutellarein, force techin hydrate (Ca techin Hydrate), and compounds chemically modified from these An inhibitor of the transporter activity of the estrone trisulfate transporter, comprising as an active ingredient.
[2] ゲ-スティン(Genistein)、ケルセチン(Quercetin)、ギンコライド C (GinkgolideC)、 テアフラビン(Theaflavin)、テアフラビン 3— O—ガレート(Theaflavin 3-O-gallate)、 ルチン(Rutin)、カルコン(Chalcone)、ダイゼイン(Daidzein)、ダイジン(Daidzin)、フ ラノ ノン (Flavanone)、フラボノーノレ (Flavonol)、ジエラノレドーノレ (Geraldol)、ヘスペレ チン(Hesperetin)、ヘスペルジン(Hesperidin)、ィプリフラボン(Ipriflavone)、ルテオリ ン一 7— O—ダルコシド(Luteolin- 7-0- Glucoside)、メトキシカルコン(Methoxychalco ne)、 4,一メチノレ一 7—メトキシ一イソフラボン(4'— mety卜 7— methoxy— isoflavone)、 5— モリン(5- Morin)、ミリセチン(Myricetin)、ナリンゲ-ン(Naringenin)、ナリンゲ-ン一 7—グノレコシド (Naringenin- 7- glycoside)、ナリンジン (Naringin)、ネオヘスペリジンジ ノヽイドロカノレコン (Neohesperidin Dihydrochalcone)、ノミリン (Nomilin)、プリムレチン ( Primuletin)、ポンシリン(Poncirin)、スクテラレイン(Scutellarein)、力テキン水和物(Ca techin Hydrate)、及びこれらを化学修飾した化合物から選ばれる 1種以上の成分を 有効成分とする乳癌細胞増殖抑制剤。  [2] Genistein, Quercetin, Ginkgolide C, Theaflavin, Theaflavin 3-O-gallate, Rutin, Chalcone , Daidzein, Daidzin, Flavanone, Flavonol, Geraldol, Hesperetin, Hesperidin, Ipriflavone, Luteolin 1—O-Dalcoside (Luteolin-7-0- Glucoside), Methoxychalcone, 4, 1-Methylolone 7-Methoxy-1-isoflavone, 5—Molin ( 5- Morin), Myricetin, Naringenin, Naringenin 7-glycoside, Naringin, Neohesperidin dinoiduro One or more selected from canorecon (Neohesperidin Dihydrochalcone), nomilin, primuletin, poncilin, scutellarein, force techin hydrate (Ca techin Hydrate), and compounds chemically modified from these A breast cancer cell growth inhibitor comprising the above ingredients as an active ingredient.
[3] ゲ-スティン(Genistein)、ケルセチン(Quercetin)、ギンコライド C (GinkgolideC)、 テアフラビン(Theaflavin)、テアフラビン 3— O—ガレート(Theaflavin 3-O-gallate)、 ルチン(Rutin)、カルコン(Chalcone)、ダイゼイン(Daidzein)、ダイジン(Daidzin)、フ ラノ ノン (Flavanone)、フラボノーノレ (Flavonol)、ジエラノレドーノレ (Geraldol)、ヘスペレ チン(Hesperetin)、ヘスペルジン(Hesperidin)、ィプリフラボン(Ipriflavone)、ルテオリ ン一 7— O—ダルコシド(Luteolin- 7- 0- Glucoside)、メトキシカルコン(Methoxychalco ne)、 4,一メチノレ一 7—メトキシ一イソフラボン(4'— mety卜 7— methoxy— isoflavone)、 5— モリン(5- Morin)、ミリセチン(Myricetin)、ナリンゲ-ン(Naringenin)、ナリンゲ-ン一 7—グノレコシド (Naringenin- 7- glycoside)、ナリンジン (Naringin)、ネオヘスペリジンジ ノヽイドロカノレコン (Neohesperidin Dihydrochalcone)、ノミリン (Nomilin)、プリムレチン ( Primuletin)、ポンシリン(Poncirin)、スクテラレイン(Scutellarein)、及び力テキン水和 物(Catechin Hydrate)、及びこれらを化学修飾した化合物力 選ばれる 1種以上の 成分を有効成分とする乳癌治療剤。 [3] Genistein, Quercetin, Ginkgolide C, Theaflavin, Theaflavin 3-O-gallate, Theaflavin 3-O-gallate, Rutin, Chalcone, Daidzein, Daidzin, Flavanone, Flavonol ), Geraldol, Hesperetin, Hesperidin, Ipriflavone, Luteolin-7-O-Dalcoside, Methoxychalcone ), 4, 1-methylol 7-methoxy-isoflavone (4'-methy 卜 7-methoxy-isoflavone), 5- Morin (5- Morin), myricetin (Myricetin), Naringenin, Naringen I 7—Gnorecoside (Naringenin-7-glycoside), Naringin (Naringin), Neohesperidin Dihydrochalcone (Neohesperidin Dihydrochalcone), Nomilin (Primilet) in), Poncirin, Scutellarein, Catechin Hydrate, and a compound force obtained by chemically modifying these. A therapeutic agent for breast cancer comprising one or more selected components as active ingredients.
PCT/JP2006/307751 2005-04-15 2006-04-12 Inhibitory agent for estrone-3-sulfate transporter activity WO2006112330A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2005-119098 2005-04-15
JP2005119098A JP2006298781A (en) 2005-04-15 2005-04-15 Estrone-3-sulfate transporter activity inhibitor

Publications (1)

Publication Number Publication Date
WO2006112330A1 true WO2006112330A1 (en) 2006-10-26

Family

ID=37115054

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/307751 WO2006112330A1 (en) 2005-04-15 2006-04-12 Inhibitory agent for estrone-3-sulfate transporter activity

Country Status (2)

Country Link
JP (1) JP2006298781A (en)
WO (1) WO2006112330A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016062281A1 (en) * 2014-10-24 2016-04-28 朗齐生物医学股份有限公司 Applications of cardiovascular disease medicaments in preparing cancer-inhibiting pharmaceutical composition
EP3333153A1 (en) * 2011-06-15 2018-06-13 The U.S.A. as represented by the Secretary, Department of Health and Human Services Nuclear receptor modulators and their use for the treatment and prevention of cancer
CN111171042A (en) * 2020-01-15 2020-05-19 中国科学院西北高原生物研究所 Separation preparation process and application of natural free radical scavenger in saxifrage

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6082737B2 (en) * 2011-07-07 2017-02-15 リサーチ キャンサー インスティテュート オブ アメリカResearch Cancer Institute Of America Systems, methods, and formulations for treating cancer
US8933078B2 (en) 2011-07-14 2015-01-13 Research Cancer Institute Of America Method of treating cancer with combinations of histone deacetylase inhibitors (HDAC1) substances
WO2018157081A1 (en) 2017-02-27 2018-08-30 Research Cancer Institute Of America Compositions, methods, systems and/or kits for preventing and/or treating neoplasms
WO2018170457A1 (en) 2017-03-17 2018-09-20 Research Cancer Institute Of America Compositions, methods, systems and/or kits for preventing and/or treating neoplasms

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS60178815A (en) * 1984-02-24 1985-09-12 Rikagaku Kenkyusho Carcinostatic agent
JP2001252045A (en) * 2000-03-13 2001-09-18 Nicchu Igaku Kenkyusho:Kk Anti-dioxin health food with genistein as main material extracted from fermented soybean
JP2001335484A (en) * 2000-05-25 2001-12-04 Protein Technol Internatl Inc Composition or method for prophylaxis or treatment of mammary cancer
JP2002293779A (en) * 2001-03-30 2002-10-09 Pokka Corp Flavonoid compound and method for producing the same
JP2003002830A (en) * 2001-06-20 2003-01-08 Sangaku Renkei Kiko Kyushu:Kk Method for inhibiting expression of estrogen activity using flavonoid and composition for inhibiting expression of estrogen activity
JP2003510240A (en) * 1997-09-26 2003-03-18 ケイジーケイ シナーギーズ インコーポレイテッド Use of citrus limonoids, flavonoids and tocotrienols as cancer treatment
WO2003089423A2 (en) * 2002-04-18 2003-10-30 Sri International Novel flavanoids and chalcones as chemotherapeutic, chemopreventive, and antiangiogenic agents
JP2005029778A (en) * 2003-06-20 2005-02-03 Pokka Corp Antioxidant, antibacterial, antineoplastic, and food and drink

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS60178815A (en) * 1984-02-24 1985-09-12 Rikagaku Kenkyusho Carcinostatic agent
JP2003510240A (en) * 1997-09-26 2003-03-18 ケイジーケイ シナーギーズ インコーポレイテッド Use of citrus limonoids, flavonoids and tocotrienols as cancer treatment
JP2001252045A (en) * 2000-03-13 2001-09-18 Nicchu Igaku Kenkyusho:Kk Anti-dioxin health food with genistein as main material extracted from fermented soybean
JP2001335484A (en) * 2000-05-25 2001-12-04 Protein Technol Internatl Inc Composition or method for prophylaxis or treatment of mammary cancer
JP2002293779A (en) * 2001-03-30 2002-10-09 Pokka Corp Flavonoid compound and method for producing the same
JP2003002830A (en) * 2001-06-20 2003-01-08 Sangaku Renkei Kiko Kyushu:Kk Method for inhibiting expression of estrogen activity using flavonoid and composition for inhibiting expression of estrogen activity
WO2003089423A2 (en) * 2002-04-18 2003-10-30 Sri International Novel flavanoids and chalcones as chemotherapeutic, chemopreventive, and antiangiogenic agents
JP2005029778A (en) * 2003-06-20 2005-02-03 Pokka Corp Antioxidant, antibacterial, antineoplastic, and food and drink

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
NOZAWA T. ET AL.: "Involvement of estrone-3-sulfate transporters in proliferation of hormone-dependent breast cancer cells", J. PHARMACOL. EXP. THER. NLM, vol. 311, no. 3, December 2004 (2004-12-01), pages 1032 - 1037, XP003001230 *
SATOH H. ET AL.: "Citrus juices inhibit the function of human organic anion-transporting polypeptide OATP-B", DRUG METABOLISM AND DISPOSITION, vol. 33, no. 4, 2005, pages 518 - 523, XP003001228 *
SOODVILAI S. ET AL.: "Involvement of tyrosine kinase and PI3K in the regulation of OAT3-mediated estrone sulfate transport in isolated rabbit renal proximal tubules", AMERICAN JOURNAL OF PHYSIOLOGY, vol. 289, no. 5, PT. 2, 2005, pages F1057 - F1064, XP003001229 *
SOODVILAI S. ET AL.: "Protein tyrosine kinase regulates OAT3-mediated organic anion transport in intact renal proximal tubules", FASEB JOURNAL, vol. 19, no. 4, SUPPL. S, PART 1, 4 March 2005 (2005-03-04), pages A149, XP003001227 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3333153A1 (en) * 2011-06-15 2018-06-13 The U.S.A. as represented by the Secretary, Department of Health and Human Services Nuclear receptor modulators and their use for the treatment and prevention of cancer
US10071945B2 (en) 2011-06-15 2018-09-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nuclear receptor modulators and their use for the treatment and prevention of cancer
AU2017248517B2 (en) * 2011-06-15 2019-06-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nuclear receptor modulators and their use for the treatment and prevention of cancer
US10737995B2 (en) 2011-06-15 2020-08-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Nuclear receptor modulators and their use for the treatment and prevention of cancer
WO2016062281A1 (en) * 2014-10-24 2016-04-28 朗齐生物医学股份有限公司 Applications of cardiovascular disease medicaments in preparing cancer-inhibiting pharmaceutical composition
CN111171042A (en) * 2020-01-15 2020-05-19 中国科学院西北高原生物研究所 Separation preparation process and application of natural free radical scavenger in saxifrage

Also Published As

Publication number Publication date
JP2006298781A (en) 2006-11-02

Similar Documents

Publication Publication Date Title
Brodie et al. Aromatase and COX-2 expression in human breast cancers
Gershanovich et al. Letrozole, a new oral aromatase inhibitor: randomised trial comparing 2.5 mg daily, 0.5 mg daily and aminoglutethimide in postmenopausal women with advanced breast cancer
WO2006112330A1 (en) Inhibitory agent for estrone-3-sulfate transporter activity
Goss et al. Aromatase inhibitors in the treatment and prevention of breast cancer
Riggins et al. Antiestrogens, aromatase inhibitors, and apoptosis in breast cancer
Liao et al. Promotion of estrogen-induced mammary gland carcinogenesis by androgen in the male Noble rat: probable mediation by steroid receptors.
Spoerri et al. Endocrine control of canine mammary neoplasms: serum reproductive hormone levels and tissue expression of steroid hormone, prolactin and growth hormone receptors
JP2001515038A (en) Methods for preventing or treating estrogen-dependent diseases and disorders
Howell et al. Tamoxifen as an agonist for metastatic breast cancer
AU785439B2 (en) Hormone receptor modulation
JPWO2004035089A1 (en) Treatment for hormone-dependent cancer
JPWO2009008461A1 (en) Pharmaceutical composition for diseases caused by sex hormone-sensitive cell proliferation
Bennett et al. Pharmacodynamic and pharmacokinetic properties of AFPep, a novel peptide for the treatment of breast cancer
Mazzucchelli et al. Somatostatin receptor subtypes in hormone-refractory (castration-resistant) prostatic carcinoma
Goss et al. Chemoprevention with aromatase inhibitors—trial strategies
US20070148703A1 (en) Method of screening remedy for breast cancer
JP2006176427A (en) Estrone 3-sulfate transporter activity inhibitor
Babaei et al. 17β-Estradiol inhibits calcium-dependent and-independent contractions in isolated human saphenous vein
Zhou et al. In Vivo Study of the Effects of ER on Apoptosis and Proliferation of Hormone-Independent Prostate Cancer Cell Lines PC-3M
Vergote et al. The oestrogen receptor and its selective modulators in gynaecological and breast cancer
US20090036507A1 (en) Bis (cyanophenyl) methyl-triazole for use in prevention of breast cancer
Kittayaruksakul et al. Activation of constitutive androstane receptor inhibits intestinal CFTR-mediated chloride transport
US20110178052A1 (en) 9-alpha estratriene derivatives as er-beta selective ligands for the prevention and treatment of intestinal cancer
Broulik et al. Raloxifen prevents bone loss in castrated male mice
US8729053B2 (en) Nuclear factor kappa B pathway inhibitor composition and use of same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06731687

Country of ref document: EP

Kind code of ref document: A1