WO2009110416A1 - Concomitant drug - Google Patents
Concomitant drug Download PDFInfo
- Publication number
- WO2009110416A1 WO2009110416A1 PCT/JP2009/053833 JP2009053833W WO2009110416A1 WO 2009110416 A1 WO2009110416 A1 WO 2009110416A1 JP 2009053833 W JP2009053833 W JP 2009053833W WO 2009110416 A1 WO2009110416 A1 WO 2009110416A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkyl group
- hydrogen atom
- cancer
- alkyl
- compound
- Prior art date
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
Definitions
- the present invention is (1) a formula having HER2 inhibitory action:
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- the present invention also relates to a thymidine synthase production inhibitor comprising the compound represented by the formula (I) or a salt thereof or a prodrug thereof.
- Patent Document 1 describes a HER2 inhibitor having a pyrrolopyrimidine skeleton or a pyrazolopyrimidine skeleton.
- Patent Document 2 discloses a formula that is a HER2 inhibitor:
- the present invention is useful as an agent for preventing or treating cancer (1):
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- N- [2- (4- ⁇ [3-chloro-4- (3-chlorophenoxy) phenyl] amino ⁇ -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) (hereinafter sometimes abbreviated as compound (I)) or a salt or prodrug thereof and (2) a hormonal therapeutic agent or an anticancer agent
- a thymidine synthase production inhibitor hereinafter, referred to as a concomitant agent of the present invention
- a compound represented by the formula (I) or a salt or prodrug thereof It is an object of the present invention to provide a thymidine synthase production inhibitor of the present invention.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- R 1 represents a hydrogen atom
- R 2 is —NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl
- n is an integer of 1 to 4
- R 6 is a hydrogen atom or a C 1-4 alkyl group
- a C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl
- R 3 represents a hydrogen atom or a C 1-6 alkyl group
- R 4 represents a halogen atom or a C 1-6 alkyl group
- R 5 represents a halogen atom or a C 1-6 alkyl group
- X represents a hydrogen atom or a halogen atom.
- the hormonal therapeutic agent may increase the HER family molecule and enhance the stimulation of the HER family molecule, thereby reducing the effect.
- the compound (I) has an action of blocking the HER signal. Therefore, in particular, a pharmaceutical comprising a combination of (1) compound (I), which is a HER2 inhibitor, and (2) a hormonal therapeutic agent, prevents the increase in HER family molecules compared to the case of using a hormonal therapeutic agent alone. Drugs with different mechanisms can be used in combination. Furthermore, the compound represented by the formula (I) or a salt thereof or a prodrug thereof has a thymidine synthase production inhibitory action, so that it is useful as a single agent active ingredient for the safe prevention or treatment of cancer. is there.
- the horizontal axis indicates the concentration of each drug, and the vertical axis indicates the number of cells (%) when the group to which no drug is added is defined as 100%.
- ⁇ ; Compound A alone ( ⁇ M), ⁇ ; AZD6474 alone ( ⁇ M), ⁇ ; Compound A + AZD6474 It is a figure which shows that SK-BR-3 sputum cell proliferation is inhibited more strongly by combined use of compound A and BEZ235 compared with each time alone.
- Cell growth inhibition assay was performed using CellTiter-Glo Luminescent Cell Viability assay kit.
- the horizontal axis indicates the concentration of each drug, and the vertical axis indicates the number of cells (%) when the group to which no drug is added is defined as 100%.
- halogen atom examples include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
- C 1-6 alkyl means straight or branched alkyl having 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, 1-ethylpropyl, hexyl, isohexyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-dimethylbutyl, 2-ethylbutyl, etc. It is done.
- C 1-4 alkyl means straight or branched alkyl having 1 to 4 carbon atoms such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and sec-butyl and tert-butyl.
- R 2 is preferably a C 1-6 alkyl group (particularly an ethyl group) substituted with a group represented by the formula “—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl”.
- R 6a is a hydrogen atom or a methyl group
- R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group.
- R 7 and R 8 are preferably a methyl group.
- R 3 is preferably a hydrogen atom.
- the “halogen atom” represented by R 4 is preferably a chlorine atom.
- the “C 1-6 alkyl group” represented by R 4 is preferably a methyl group.
- R 4 is preferably a chlorine atom or a methyl group.
- the “halogen atom” represented by R 5 is preferably a fluorine atom or a chlorine atom.
- the “C 1-6 alkyl group” represented by R 5 is preferably a methyl group.
- R 5 is preferably a fluorine atom, a chlorine atom or a methyl group.
- halogen atom represented by X is preferably a fluorine atom.
- X is preferably a hydrogen atom or a fluorine atom, more preferably a hydrogen atom.
- R 1 is a hydrogen atom
- R 2 is —NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (wherein R 6a is a hydrogen atom or a methyl group, R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group)
- a C 1-6 alkyl group (particularly an ethyl group) substituted with a group represented by R 3 is a hydrogen atom
- R 4 is a chlorine atom or a methyl group
- R 5 is a fluorine atom, a chlorine atom or a methyl group
- X is a hydrogen atom or a fluorine atom (preferably a hydrogen atom)
- R 1 is a hydrogen atom
- R 2 is Substituted with a group represented by —NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (wherein R 6a represents a hydrogen atom or a methyl group, and R 7 and R 8 represent a methyl group)
- a C 1-6 alkyl group (especially an ethyl group)
- R 3 is a hydrogen atom
- R 4 is a chlorine atom or a methyl group
- R 5 is a fluorine atom, a chlorine atom or a methyl group
- X is a hydrogen atom or a fluorine atom (preferably a hydrogen atom), Compounds.
- Compound (I) and a salt thereof can be produced according to the method described in International Publication No. 2007/064045.
- compound (I) has an isomer such as an optical isomer, a stereoisomer, a positional isomer, a rotational isomer, etc.
- any one of the isomers and a mixture are encompassed in compound (I).
- compound (I) has an optical isomer
- the optical isomer resolved from the racemate is also encompassed in compound (I).
- Each of these isomers can be obtained as a single product by a known synthesis method or separation method (concentration, solvent extraction, column chromatography, recrystallization, etc.).
- Compound (I) may be a crystal, and it is included in compound (I) regardless of whether the crystal form is a single crystal form or a crystal form mixture.
- the crystal can be produced by crystallization by applying a crystallization method known per se.
- Compound (I) may be a solvate (such as a hydrate) or a non-solvate, and both are encompassed in compound (I).
- Compounds labeled with isotopes eg, 2 H, 3 H, 14 C, 35 S, 125 I, etc. are also encompassed in compound (I).
- Examples of the salt of compound (I) include metal salts, ammonium salts, salts with organic bases, salts with inorganic acids, salts with organic acids, salts with basic or acidic amino acids, and the like.
- the metal salt include alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as calcium salt, magnesium salt and barium salt; aluminum salt and the like.
- the salt with organic base include trimethylamine, triethylamine, pyridine, picoline, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, tromethane [tris (hydroxymethyl) methylamine], t-butylamine.
- Salts with cyclohexylamine, dicyclohexylamine, N, N′-dibenzylethylenediamine and the like Preferable examples of the salt with inorganic acid include salts with hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid and the like.
- Preferable examples of the salt with organic acid include formic acid, acetic acid, trifluoroacetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, and benzenesulfone.
- salts with acid p-toluenesulfonic acid and the like.
- salts with basic amino acids include salts with arginine, lysine, ornithine and the like
- salts with acidic amino acids include salts with aspartic acid and glutamic acid, for example. It is done.
- salts with inorganic acids such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, or acetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid
- salts with organic acids such as citric acid, succinic acid, methanesulfonic acid and p-toluenesulfonic acid.
- a salt with p-toluenesulfonic acid is preferred.
- a prodrug of compound (I) or a salt thereof is a compound that is converted into compound (I) or a salt thereof by a reaction with an enzyme, gastric acid or the like under physiological conditions in vivo, that is, enzymatically oxidized, reduced, hydrolyzed, etc. It refers to a compound that changes to compound (I) or a salt thereof, or a compound that undergoes hydrolysis or the like by gastric acid or the like and changes to compound (I) or a salt thereof.
- a compound in which the amino of the compound (I) or a salt thereof is acylated, alkylated or phosphorylated eg, the amino of the compound (I) or a salt thereof is eicosanoylated, Alanylation, pentylaminocarbonylation, (5-methyl-2-oxo-1,3-dioxolen-4-yl) methoxycarbonylation, tetrahydrofuranylation, pyrrolidylmethylation, pivaloyloxymethylation, tert-butyl And the like are used. These compounds can be produced from compound (I) or a salt thereof by a method known per se.
- the prodrug of compound (I) or a salt thereof is compound (I) or a compound thereof under physiological conditions as described in Hirokawa Shoten, 1990, “Pharmaceutical Development”, Volume 7, Molecular Design, pages 163 to 198. It may change into a salt.
- Compound (I) or a salt thereof or a prodrug thereof is abbreviated as Compound (I).
- examples of the ⁇ hormone therapeutic agent '' include phosfestol, diethylstilbestrol, chlorotrianicene, medroxyprogesterone acetate, megestrol acetate, chlormadinone acetate, cyproterone acetate, danazol, dienogest, azoprisnil, Allylestrenol, gestrinone, nomegestrol, tadenane, mepaltricine, raloxifene, oloxifene, levormeloxifene, antiestrogens (eg, tamoxifen citrate, toremifene citrate), ER down regulators (eg fulvestrant ( Faslodex (trademark), etc.), human menopausal gonadotropin, follicle stimulating hormone, pill preparation, mepithiostan, testrolactone, aminoglutethimide, LH-RH agonist ( , Gose
- examples of the “anticancer agent” include a chemotherapeutic agent, an immunotherapeutic agent, a cell growth factor, and a drug that inhibits the action of its receptor.
- chemotherapeutic agent examples include alkylating agents, antimetabolites, anticancer antibiotics, plant-derived anticancer agents and the like.
- alkylating agent examples include nitrogen mustard, nitrogen mustard hydrochloride-N-oxide, chlorambutyl, cyclophosphamide, ifosfamide, thiotepa, carbocon, improsulfan tosylate, busulfan, nimustine hydrochloride, mitoblonitol, Faran, dacarbazine, ranimustine, estramustine phosphate sodium, triethylenemelamine, carmustine, lomustine, streptozocin, piprobroman, etoglucid, carboplatin, cisplatin, miboplatin, nedaplatin, oxaliplatin, altretamine, ambermuthine, dibrospine hydrochloride, fotemustine hydrochloride Predonimustine, pumitep
- antimetabolite examples include mercaptopurine, 6-mercaptopurine riboside, thioinosine, methotrexate, enocitabine, cytarabine, cytarabine okphosphat, ancitabine hydrochloride, 5-FU drugs (eg, fluorouracil, tegafur, UFT, Doxyfluridine, carmofur, galocitabine, emiteful, etc.), aminopterin, leucovorin calcium, tabloid, butosine, folinate calcium, levofolinate calcium, cladribine, emiteful, fludarabine, gemcitabine, hydroxycarbamide, pentostatin, pyritorexime, idoxyuridine, mitoxifridin Ambamustine, pemetrexed disodium salt (Alimta (trademark)) and the like are used.
- 5-FU drugs eg, fluorouracil, tegafur, UFT, Doxyfluridine, car
- anticancer antibiotic examples include actinomycin D, actinomycin C, mitomycin C, chromomycin A3, bleomycin hydrochloride, bleomycin sulfate, pepromycin sulfate, daunorubicin hydrochloride, doxorubicin hydrochloride (Adriacin (trademark)), acrarubicin hydrochloride , Pirarubicin hydrochloride, epirubicin hydrochloride, neocalcinostatin, misramycin, sarcomycin, carcinophylline, mitotane, zorubicin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride and the like.
- plant-derived anticancer agent examples include etoposide, etoposide phosphate, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel (Taxol TM), docetaxel, vinorelbine and the like.
- BRM immunotherapy agent
- examples of the “immunotherapy agent (BRM)” include picibanil, krestin, schizophyllan, lentinan, ubenimex, interferon, interleukin, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, lymphotoxin, BCG vaccine, coryne Bacterium parvum, levamisole, polysaccharide K, procodazole and the like are used.
- the “cell growth factor” in the “drug that inhibits the action of the cell growth factor and its receptor” may be any substance that promotes cell growth, and usually has a molecular weight of 20,000 or less.
- a factor that exerts an action at a low concentration by binding to a receptor is used.
- EGF epidermal growth factor
- Insulin or a substance having substantially the same activity eg, insulin, IGF (insulin-like growth factor) -1, IGF-2, etc.
- FGF fibroblast growth factor
- FGF-10 a substance having substantially the same activity
- CSF erythropoietin
- EPO erythropoietin
- IL-2 interleukin-2
- NGF non grow growth factor
- PDGF platelet-derived growth factor
- TGF ⁇ transforming growth factor ⁇
- HGF hepatocyte growth factor
- the “cell growth factor receptor” may be any receptor that has the ability to bind to the cell growth factor, and specifically includes an EGF receptor, a haregulin receptor (HER2). Insulin receptor, IGF receptor, FGF receptor-1 or FGF receptor-2, and the like.
- Examples of the “drug that inhibits the action of cell growth factor” include HER2 antibody (such as trastuzumab (Herceptin (trademark))), imatinib mesylate, ZD1839 or EGFR antibody (cetuximab (Erbitux (trademark), etc.)) , Antibodies against VEGF (eg, bevacizumab (Avastin TM)), VEGFR antibody, VEGFR inhibitor, EGFR inhibitor (erlotinib (Tarceva TM), gefitinib (Iressa TM), etc.) Is mentioned.
- HER2 antibody such as trastuzumab (Herceptin (trademark))
- imatinib mesylate ZD1839 or EGFR antibody
- cetuximab cetuximab (Erbitux (trademark), etc.
- Antibodies against VEGF eg, bevacizumab (Avastin TM)
- VEGFR antibody e.g,
- mTOR inhibitors temsirolimus, rapamycin, AZD8055, RAD001, CCI-779, Ap23573 / MK8669, etc.
- Akt inhibitors PI3 kinase inhibitors
- hormone therapy agents or anticancer agents include ER down regulators (for example, fulvestrant (Faslodex TM)), HER2 antibodies (trastuzumab (Herceptin TM)) ), EGFR antibody (cetuximab (Erbitux (trademark) etc.), EGFR inhibitor (erlotinib (Tarceva (trademark), gefitinib (Iressa (trademark)) etc.), VEGFR inhibitor or chemotherapeutic agent (Paclitaxel (Taxol (trademark) etc.) is preferable.
- ER down regulators for example, fulvestrant (Faslodex TM)
- HER2 antibodies to stauzumab (Herceptin TM)
- EGFR antibody cetuximab (Erbitux (trademark) etc.
- EGFR inhibitor erlotinib (Tarceva (trademark), gefitinib (Iressa (trademark)) etc.
- fulvestrant Faslodex TM
- trastuzumab Herceptin TM
- cetuximab cetuximab
- erlotinib Tarceva TM
- gefitinib Iressa TM
- Paclitaxel Taxol TM
- preferable examples include doxorubicin hydrochloride (Adriacin (trademark)), irinotecan hydrochloride (topotecin (trademark), campto (trademark)), 5FU, docetaxel or methotrexate.
- the combination agent of the present invention is not limited to the combination of two agents, and three or more agents may be used in combination.
- the timing of administration of Compound (I) and the concomitant drug is not limited, and Compound (I) and the concomitant drug may be administered simultaneously to the administration subject. Alternatively, administration may be performed with a time difference.
- the dose of the concomitant drug may be determined according to the dose used clinically, and can be appropriately selected depending on the administration subject, administration route, disease, combination and the like.
- the administration mode of compound (I) and the concomitant drug is not particularly limited, as long as compound (I) and the concomitant drug are combined at the time of administration.
- Examples of such administration forms include (1) administration of a single preparation obtained by simultaneously formulating compound (I) and a concomitant drug, and (2) separate preparation of compound (I) and concomitant drug. Simultaneous administration of the two preparations obtained by the same route by the same route of administration, (3) with a time difference in the same route of administration of the two formulations obtained by separately formulating the compound (I) and the concomitant drug (4) Simultaneous administration of two kinds of preparations obtained by separately formulating Compound (I) and a concomitant drug by different administration routes, (5) Preparation of Compound (I) and a concomitant drug separately Administration of the two types of preparations obtained by the preparation at different time intervals in different administration routes (for example, administration in the order of Compound (I) ⁇ concomitant drug or administration in the reverse order) and the like are used.
- the combination agent of the present invention is a therapeutic agent that suppresses the growth of cancers expressing HER2 and / or EGFR kinase, and prevents the transition of hormone-dependent and hormone-dependent cancers to hormone-independent cancers It is also useful as a preventive agent. In addition, it has low toxicity (eg acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.), high water solubility, stability, pharmacokinetics (absorbability, distribution) , Metabolism, excretion, etc.), and is also useful as a medicine because of its excellent medicinal effects.
- toxicity eg acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.
- high water solubility eg acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.
- high water solubility eg acute toxicity,
- the concomitant drug of the present invention can be used for various cancers (among others, breast cancer (for example, invasive ductal cancer, non-invasive ductal cancer, inflammatory breast cancer, etc.), prostate cancer (for example, hormone-dependent prostate cancer, hormone).
- breast cancer for example, invasive ductal cancer, non-invasive ductal cancer, inflammatory breast cancer, etc.
- prostate cancer for example, hormone-dependent prostate cancer, hormone.
- pancreatic cancer eg, pancreatic duct cancer, etc.
- stomach cancer eg, papillary adenocarcinoma, mucinous adenocarcinoma, adenosquamous carcinoma, etc.
- lung cancer eg, non-small cell lung cancer, small cell lung cancer, malignant) Mesothelioma
- colon cancer eg, gastrointestinal stromal tumor
- rectal cancer eg, gastrointestinal stromal tumor
- colorectal cancer eg, familial colorectal cancer, hereditary nonpolyposis colorectal cancer, digestion
- Ductal stromal tumors small intestine cancer (eg, non-Hodgkin lymphoma, gastrointestinal stromal tumors), esophageal cancer, duodenal cancer, tongue cancer, pharyngeal cancer (eg, nasopharyngeal cancer, oropharyngeal cancer, hypopharyngeal cancer, etc.) ),
- Tyrosine kinase dependent diseases further include cardiovascular diseases associated with abnormal tyrosine kinase enzyme activity. Therefore, the combination agent of the present invention can also be used as a preventive or therapeutic agent for cardiovascular diseases such as restenosis.
- the combination agent of the present invention is useful as an anticancer agent for prevention or treatment of cancer, particularly breast cancer, ovarian cancer, prostate cancer, lung cancer, pancreatic cancer, kidney cancer, colon cancer, small intestine cancer, esophageal cancer and gastric cancer.
- the concomitant drug of the present invention has low toxicity and is used as a pharmaceutical as it is or mixed with a pharmaceutically acceptable carrier known per se, for example, to mammals (eg, human, horse, cow, dog, cat, rat, mouse, Rabbit, pig, monkey, etc.) can be used as a pharmaceutical composition.
- the concomitant drug of the present invention is prepared by mixing a compound (I) or (and) the above concomitant drug with a pharmacologically acceptable carrier according to a method known per se, for example, a pharmaceutical composition such as a tablet (sugar-coated tablet, film coating).
- the injection can be administered intravenously, intramuscularly, subcutaneously, or into an organ, or directly to the lesion.
- the pharmacologically acceptable carrier that may be used in the production of the concomitant drug of the present invention include various organic or inorganic carrier substances that are commonly used as pharmaceutical materials, such as excipients and lubricants in solid preparations.
- Binders and disintegrants Binders and disintegrants, solvents in liquid preparations, solubilizers, suspending agents, tonicity agents, buffers and soothing agents. If necessary, additives such as conventional preservatives, antioxidants, colorants, sweeteners, adsorbents, wetting agents and the like can be used in appropriate amounts.
- excipient examples include lactose, sucrose, D-mannitol, starch, corn starch, crystalline cellulose, light anhydrous silicic acid and the like.
- lubricant examples include magnesium stearate, calcium stearate, talc, colloidal silica and the like.
- binder examples include crystalline cellulose, sucrose, D-mannitol, dextrin, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, starch, sucrose, gelatin, methylcellulose, sodium carboxymethylcellulose and the like.
- Examples of the disintegrant include starch, carboxymethyl cellulose, carboxymethyl cellulose calcium, carboxymethyl starch sodium, L-hydroxypropyl cellulose, and the like.
- Examples of the solvent include water for injection, alcohol, propylene glycol, macrogol, sesame oil, corn oil, olive oil and the like.
- Examples of the solubilizer include polyethylene glycol, propylene glycol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate and the like.
- suspending agent examples include surfactants such as stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glyceryl monostearate; for example, polyvinyl alcohol, polyvinylpyrrolidone, carboxy
- examples include hydrophilic polymers such as sodium methylcellulose, methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, and hydroxypropylcellulose.
- isotonic agent include glucose, D-sorbitol, sodium chloride, glycerin, D-mannitol and the like.
- buffer examples include buffer solutions of phosphate, acetate, carbonate, citrate and the like.
- soothing agents include benzyl alcohol.
- preservative examples include p-hydroxybenzoates, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid and the like.
- antioxidant examples include sulfite, ascorbic acid, ⁇ -tocopherol and the like.
- the compounding ratio of the compound (I) and the concomitant drug in the combination drug of the present invention can be appropriately selected depending on the administration subject, administration route, disease and the like.
- the content of compound (I) in the combination agent of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50% by weight, based on the whole preparation. More preferably, it is about 0.5 to 20% by weight.
- the content of the concomitant drug in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50% by weight, more preferably about the whole preparation About 0.5 to 20% by weight.
- the content of additives such as carriers in the combination agent of the present invention varies depending on the form of the preparation, but is usually about 1 to 99.99% by weight, preferably about 10 to 90% by weight, based on the whole preparation. .
- the same content may be used when compound (I) and the concomitant drug are formulated separately.
- the compound (I) or the concomitant drug is a dispersant (eg, Tween 80 (manufactured by Atlas Powder, USA), HCO 60 (manufactured by Nikko Chemicals), polyethylene glycol, carboxymethylcellulose, sodium alginate, hydroxypropylmethylcellulose.
- a dispersant eg, Tween 80 (manufactured by Atlas Powder, USA), HCO 60 (manufactured by Nikko Chemicals), polyethylene glycol, carboxymethylcellulose, sodium alginate, hydroxypropylmethylcellulose.
- solubilizers eg, glycerin, ethanol, etc.
- buffers eg, , Phosphoric acid and its alkali metal salts, citric acid and its alkali metal salts, etc.
- isotonic agents eg, sodium chloride, potassium chloride, mannitol, sorbitol, glucose etc.
- pH regulators eg, hydrochloric acid, hydroxylated) Sodium
- preservatives eg, ethyl paraoxybenzoate, benzoic acid, para Methyl xybenzoate, propyl paraoxybenzoate, benzyl alcohol, etc.
- solubilizers eg, concentrated glycerin, meglumine, etc.
- solubilizers eg, propylene glycol
- compound (I) or a concomitant drug is used, for example, an excipient (eg, lactose, sucrose, starch, corn starch, etc.), a disintegrant (eg, starch, calcium carbonate).
- an excipient eg, lactose, sucrose, starch, corn starch, etc.
- a disintegrant eg, starch, calcium carbonate
- binder eg, starch, gum arabic, carboxymethylcellulose, polyvinylpyrrolidone, hydroxypropylcellulose, gelatin, etc.
- lubricant eg, talc, magnesium stearate, polyethylene glycol 6000, etc.
- the coating agent examples include hydroxypropylmethylcellulose, ethylcellulose, hydroxymethylcellulose, hydroxypropylcellulose, polyoxyethylene glycol, Tween 80, Pluronic F68, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxymethylcellulose acetate succinate, Eudragit (Rohm) (Made in Germany, methacrylic acid / acrylic acid copolymer) and dyes (eg, bengara, titanium dioxide, etc.) are used.
- the sugar coating examples include sucrose, talc, gum arabic, pigments (eg, bengara, titanium dioxide, etc.), polishes (eg, beeswax, etc.), and the like.
- the preparation for oral administration may be either an immediate release preparation or a sustained release preparation.
- the compound (I) or the concomitant drug can be converted into an oily or aqueous solid, semisolid or liquid suppository according to a method known per se.
- the oily base used in the above composition include glycerides of higher fatty acids (eg, cacao butter, witepsols (manufactured by Dynamite Nobel, Germany)), intermediate fatty acids (eg, miglyols (manufactured by Dynamite Nobel, Germany) Etc.], or vegetable oils (eg, sesame oil, soybean oil, cottonseed oil, etc.).
- aqueous base examples include polyethylene glycols and propylene glycol.
- aqueous gel base examples include natural gums, cellulose derivatives, vinyl polymers, and acrylic acid polymers.
- sustained-release preparation examples include sustained-release microcapsules. In order to obtain a sustained-release type microcapsule, a method known per se can be employed.
- Compound (I) is preferably molded into a preparation for oral administration such as a solid preparation (eg, powder, granule, tablet, capsule) or into a preparation for rectal administration such as a suppository. Particularly preferred are preparations for oral administration.
- the concomitant drug can be in the above-mentioned dosage form depending on the type of drug.
- the dose of the concomitant drug of the present invention varies depending on the type of compound (I), age, body weight, symptom, dosage form, administration method, administration period, etc., for example, usually per patient (adult, body weight about 60 kg) per person
- the compound (I) and the concomitant drug are each in the range of about 0.1 mg to about 500 mg, preferably in the range of about 1 mg to about 100 mg for oral administration, and about 0.01 mg to about 100 mg for parenteral administration.
- a range can be selected, more preferably from about 0.1 mg to about 10 mg.
- These dosages can be administered in 1 to 3 divided doses per day.
- the dosage varies depending on various conditions.
- a dosage smaller than the dosage may be sufficient, or the dosage may need to be administered beyond the range.
- the amount of the concomitant drug can be set as long as side effects do not become a problem.
- the daily dose as a concomitant drug varies depending on the degree of symptoms, age of the subject, sex, weight, sensitivity difference, timing of administration, interval, nature of the pharmaceutical preparation, formulation, type, type of active ingredient, etc.
- the amount of the drug is usually in the range of about 0.001 to 2000 mg, preferably about 0.01 to 500 mg, more preferably about 0.1 to 100 mg parenterally per kg body weight of the mammal, for example, by oral administration.
- it can be selected from the range of about 0.0001 mg to about 400 mg, more preferably from the range of about 0.001 mg to about 200 mg.
- These dosages are usually administered in 1 to 3 divided doses per day.
- the concomitant drug of the present invention When administering the concomitant drug of the present invention, it may be administered at the same time, but after administering the concomitant drug first, compound (I) may be administered, or compound (I) is administered first. Thereafter, the concomitant drug may be administered.
- the time difference varies depending on the active ingredient, dosage form, and administration method to be administered. For example, when administering the concomitant drug first, within 1 minute to 3 days after administering the concomitant drug, preferably The method includes administering Compound (I) within 10 minutes to 1 day, more preferably within 15 minutes to 1 hour.
- the concomitant drug is administered within 1 minute to 1 day after administration of compound (I), preferably within 10 minutes to 6 hours, more preferably within 15 minutes to 1 hour.
- a method is mentioned.
- a preferable administration method for example, about 0.001 to 200 mg / kg body weight of a concomitant drug formed into a preparation for parenteral administration was injected intravenously or intramuscularly, and after about 15 minutes, it was formed into a preparation for oral administration.
- Compound (I) is orally administered at a daily dose of about 0.005 to 100 mg / kg body weight.
- the compound (I) of the present invention since the compound (I) of the present invention has a thymidine synthase production inhibitory action, it can be used as a single agent active ingredient, as a therapeutic agent that suppresses the growth of cancer, and for hormone-dependent and hormone-dependent cancers. It is useful as a prophylactic agent that prevents the transition to hormone-independent cancer.
- the compound (I) of the present invention has low toxicity (for example, acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.), high water solubility, stability, It is also useful as a medicine because it is excellent in pharmacokinetics (absorbability, distribution, metabolism, excretion, etc.) and in terms of drug efficacy.
- the compound (I) of the present invention can be used as an active ingredient of a single agent as well as various cancers (especially breast cancer (for example, invasive breast cancer, non-invasive breast cancer, inflammatory breast cancer, etc.), prostate cancer) (Eg, hormone-dependent prostate cancer, hormone-independent prostate cancer, etc.), pancreatic cancer (eg, pancreatic duct cancer, etc.), gastric cancer (eg, papillary adenocarcinoma, mucinous adenocarcinoma, adenosquamous carcinoma, etc.), lung cancer (eg, Non-small cell lung cancer, small cell lung cancer, malignant mesothelioma, etc., colon cancer (eg, gastrointestinal stromal tumor), rectal cancer (eg, gastrointestinal stromal tumor), colon cancer (eg, familial colon) Cancer, hereditary non-polyposis colorectal cancer, gastrointestinal stromal tumor, etc.), small intestine cancer (eg, non-Hodgkin lymph
- the thymidine synthase production inhibitor containing the compound (I) of the present invention has low toxicity, and is used as it is as a medicine or mixed with a pharmaceutically acceptable carrier known per se as a mammal (eg, human, horse, etc.). Bovine, dog, cat, rat, mouse, rabbit, pig, monkey, etc.).
- a pharmaceutically acceptable carrier known per se as a mammal (eg, human, horse, etc.). Bovine, dog, cat, rat, mouse, rabbit, pig, monkey, etc.).
- the thymidine synthase production inhibitor of the present invention is prepared, for example, by mixing compound (I) with a pharmacologically acceptable carrier according to a method known per se, including pharmaceutical compositions such as tablets (including sugar-coated tablets and film-coated tablets).
- Powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained release, etc., orally or parenterally (eg, topical, rectal, intravenous administration, etc.) can do.
- the injection can be administered intravenously, intramuscularly, subcutaneously, or into an organ, or directly to the lesion.
- the pharmacologically acceptable carrier that may be used for the production of the thymidine synthase production inhibitor of the present invention include the pharmacologically acceptable carrier that may be used for the production of the combination agent of the present invention. The same thing is mentioned.
- the content of compound (I) in the thymidine synthase production inhibitor of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50%, based on the whole preparation. % By weight, more preferably about 0.5 to 20% by weight.
- compositions can be produced by a method known per se generally used in the preparation process, and examples thereof include the same methods as those described above for the concomitant drug of the present invention.
- Compound (I) is preferably molded into a preparation for oral administration such as a solid preparation (eg, powder, granule, tablet, capsule) or into a preparation for rectal administration such as a suppository. Particularly preferred are preparations for oral administration.
- the dosage of the thymidine synthase production inhibitor of the present invention varies depending on the type, age, weight, symptom, dosage form, administration method, administration period, etc. of compound (I).
- Per person usually as compound (I), in the range of about 0.1 mg to about 500 mg, preferably in the range of about 1 mg to about 100 mg for oral administration, and about 0.01 mg to about 100 mg for parenteral administration.
- a range can be selected, more preferably from about 0.1 mg to about 10 mg.
- These dosages can be administered in 1 to 3 divided doses per day.
- the dosage varies depending on various conditions. Therefore, a dosage smaller than the dosage may be sufficient, or the dosage may need to be administered beyond the range.
- the production method and use method of the present invention will be described in the following examples and test examples, but the present invention is not limited thereto. Other aspects that fall within the spirit and scope of the invention as defined by the claims are also encompassed by the invention.
- the compound A in Examples and Test Examples is N- [2- (4- ⁇ [3-chloro-4- (3-chlorophenoxy) phenyl] amino ⁇ -5H-pyrrolo [3,2-d]. ] Pyrimidin-5-yl) ethyl] -2-methyl-2- (methylsulfonyl) propanamide means p-toluenesulfonate.
- Example 1 (1) Compound A 8.0g (2) Lactose 60.0g (3) Cornstarch 35.0g (4) Gelatin 3.0g (5) Magnesium stearate 2.0 g After granulating a mixture of Compound A (8.0 g), lactose (60.0 g) and corn starch (35.0 g) with a 10% by weight aqueous gelatin solution (30 mL, 3.0 g as gelatin) through a 1 mm mesh sieve. , Dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
- Example 2 After dissolving 50 mg of trastuzumab in 50 mL of JP distilled water for injection, JP distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
- Example 3 After dissolving 50 mg of cetuximab in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
- Example 4 (1) Compound A 8.0g (2) Erlotinib 8.0g (3) Lactose 60.0g (4) Corn starch 35.0g (5) Gelatin 3.0g (6) Magnesium stearate 2.0 g Using a mixture of Compound A (8.0 g), erlotinib (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
- Example 5 (1) Compound A 8.0g (2) Gefitinib 8.0g (3) Lactose 60.0g (4) Corn starch 35.0g (5) Gelatin 3.0g (6) Magnesium stearate 2.0 g Using a mixture of Compound A (8.0 g), gefitinib (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
- Example 6 Dissolve 50 mg of paclitaxel in 50 mL of JP injection distilled water, and then add JP injection distilled water to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
- Example 7 (1) Compound A 8.0g (2) Fulvestrant 8.0g (3) Lactose 60.0g (4) Corn starch 35.0g (5) Gelatin 3.0g (6) Magnesium stearate 2.0 g A mixture of Compound A (8.0 g), fulvestrant (8.0 g), lactose (60.0 g) and corn starch (35.0 g) was used with a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), After granulating through a 1 mm mesh sieve, it is dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
- Example 8 After dissolving 50 mg of doxorubicin hydrochloride in 50 mL of distilled water for injection, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
- Example 9 After dissolving 50 mg of irinotecan hydrochloride in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
- Example 10 (1) Compound A 8.0g (2) 5FU 8.0g (3) Lactose 60.0g (4) Corn starch 35.0g (5) Gelatin 3.0g (6) Magnesium stearate 2.0 g Using a mixture of Compound A (8.0 g), 5FU (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10% by weight gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
- Example 11 After dissolving 50 mg of docetaxel in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
- Example 12 (1) Compound A 8.0g (2) Methotrexate 8.0g (3) Lactose 60.0g (4) Corn starch 35.0g (5) Gelatin 3.0g (6) Magnesium stearate 2.0 g Using a mixture of Compound A (8.0 g), methotrexate (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
- Test example 1 Combined effect of Compound A and cetuximab (in vitro) Human epidermoid carcinoma cell line A431 cells were seeded in a 96-well plate at 2000 cells / 100 ⁇ L / well. The next day, Compound A alone, cetuximab alone or Compound A and cetuximab were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 50 ⁇ L of 25% glutaraldehyde solution (Wako) was added to 200 ⁇ L of the medium, and allowed to stand at room temperature for 15 minutes.
- Wako 25% glutaraldehyde solution
- the plate was washed once with PBS, 200 ⁇ L of PBS was added, 25 ⁇ L of 50% trichloroacetic acid was added, and the mixture was allowed to stand at 4 ° C. for 1 hour or longer.
- the plate was washed 5 times with tap water, and excess water was removed by tapping on a Kim towel.
- 50 ⁇ L of 0.4% (w / v) Sulforhodamine B (SRB, Sigma) -containing 1% (v / v) acetic acid solution was added to each well, and 15 minutes later, 1% acetic acid solution (v / v) was plated 3 times. Was washed.
- Test example 2 Combined effect of compound A and erlotinib (in vitro) Human lung cancer cell line Calu-3 cells were seeded in a 96-well plate at 5000 cells / 50 ⁇ L / well. The next day, compound A alone, erlotinib alone or compound A and erlotinib were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 ⁇ L of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 ⁇ L of the medium and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The luminescence intensity in the control to which no drug was added was calculated as 100% (FIG. 2). By using Compound A and erlotinib in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
- Test example 3 Combined effect of Compound A and trastuzumab (in vitro) Human breast cancer cell line BT-474 cells were seeded in a 96-well plate at 6000 cells / 50 ⁇ L / well. The next day, compound A alone, trastuzumab alone or compound A and trastuzumab were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 ⁇ L of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 ⁇ L of the medium and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). Calculations were made assuming that the control with no drug added was 100% (FIG. 3). The combined use of Compound A and trastuzumab showed a stronger cell growth inhibitory effect than when each compound was used alone.
- Test example 4 Combined effect of Compound A and trastuzumab (in vivo) Trypsinized human breast cancer cell line BT-474 (ATCC (American Type Culture Collection) catalog No.HTB-20, Lasfargues EY, In Vitro 15: 723-729 (1979)), a HER2 highly expressing cell that has been subcultured
- the suspension was treated and suspended in RPMI-1640 medium (GibcoInvitrogen) containing 10% fetal bovine serum (GibcoInvitrogen).
- the cell density of this cell suspension was measured with a cell counter Sysmex CDA500, and the cell density was adjusted to 1 ⁇ 10 8 cells / mL using the above-mentioned medium.
- nude mice 5 ⁇ 10 7 cells / mL.
- Nude mice for transplantation were BALB / cAJcl-nu / nu, female, 5-week-old mice purchased from CLEA Japan. After arrival, the cells were acclimatized for 1 week, and the cells were transplanted at a concentration of 1 ⁇ 10 7 cells / 200 ⁇ L into the abdomen subcutaneously of a 6-week-old mouse.
- estradiol propionate (Ova hormone Depot 5mg Asuka Pharmaceutical) was injected intramuscularly at a dose of 50 ⁇ L in the right hind paw. Furthermore, one week after transplantation, estradiol propionate was similarly injected intramuscularly into the left hind paw at a dose of 50 ⁇ L. Tumor volume was measured over time from 2 weeks after transplantation, and an individual with a tumor volume of 200 to 300 mm 3 was used.
- Compound A was prepared in 0.5% aqueous methylcellulose solution to a concentration of 13.06 mg / mL, and the above-mentioned Compound A suspension was orally administered at a liquid volume of 10 mL / kg.
- Compound A was administered twice a day in the morning and afternoon (the administration interval was set to be 7 hours or longer), and was administered continuously for 14 days.
- Trastuzumab (Genentech, Inc. Catalog No. NDC 50242-134-68) was prepared by diluting a stock solution of 21 mg / mL to 1 mg / mL with physiological saline and intraperitoneally at a volume of 10 mL / kg. Administered.
- Trastuzumab was administered four times, twice a week. The tumor volume was measured over time every 2 to 3 days, and the final tumor volume was measured on the day after the end of the 14-day administration to determine the combined effect.
- Compound A showed a synergistic effect with trastuzumab on HER2-highly expressing BT-474 breast cancer tumors (FIG. 4).
- Test Example 5 Measurement of TS (thymidine) expression level by addition of compound A Human breast cancer cell line BT-474 (ATCC (American Type Culture Collection) catalog No. HTB-20, Lasfargues EY, In Vitro 15: 723-729 (1979)) was trypsinized and suspended in RPMI-1640 medium (GibcoInvitrogen) containing 10% fetal bovine serum (GibcoInvitrogen). The cell density of this cell suspension was measured with a cell counter Sysmex CDA500, and the cell density was adjusted to 5 ⁇ 10 4 cells / mL using the above-mentioned medium.
- RPMI-1640 medium GibcoInvitrogen
- fetal bovine serum GibcoInvitrogen
- Test Example 6 Combined effect of Compound A and AZD6474 (A431 in vitro) Human epidermoid carcinoma cell line A431 cells were seeded in a 96-well plate at 2000 cells / 50 ⁇ L / well. The next day, Compound A alone, AZD6474 alone or Compound A and AZD6474 were mixed and added in serial dilution. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 ⁇ L of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 ⁇ L of medium and mixed on a plate shaker for about 5 minutes at room temperature.
- CellTiter-Glo Luminescent Cell Viability assay reagent Promega, G7571
- Test Example 7 Combined effect of Compound A and BEZ235 (SK-BR-3 in vitro) Human breast cancer cell line SK-BR-3 cells were seeded in a 96-well plate at 2000 cells / 50 ⁇ L / well. Two days later, Compound A alone, BEZ235 alone or Compound A and BEZ235 were mixed and serially diluted. After the compound was added, it was left in a CO 2 incubator for 4 days. To 100 ⁇ L of the medium, 100 ⁇ L of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO-light (Perkin Elmer). The luminescence intensity in the control with no drug added was calculated as 100% (FIG. 7). By using Compound A and BEZ235 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
- Test Example 8 Combined effect of Compound A and PD0325901 (Calu-3 in vitro) Human lung cancer cell line Calu-3 cells were seeded in a 96-well plate at 5000 cells / 50 ⁇ L / well. The next day, Compound A alone, PD0325901 alone or Compound A and PD0325901 were mixed and serially diluted. After adding the compound, it was left in a CO 2 incubator for 5 days. To 100 ⁇ L of the medium, 100 ⁇ L of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The luminescence intensity in the control to which no drug was added was calculated as 100% (FIG. 8). By using Compound A and PD0325901 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
- Test Example 9 Combined effect of Compound A and rapamycin (BT-474 in vitro) Human breast cancer cell line BT-474 cells were seeded in a 96-well plate at 6000 cells / 50 ⁇ L / well. The next day, compound A alone, rapamycin alone or compound A and rapamycin were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. To 100 ⁇ L of the medium, 100 ⁇ L of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The amount of wells containing only the medium was calculated as the background, and the control with no drug added was calculated as 100% (FIG. 9). The combined use of Compound A and rapamycin showed a stronger cell growth inhibitory effect than when each compound was used alone.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical & Material Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Organic Chemistry (AREA)
- Epidemiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Disclosed is a pharmaceutical agent that is obtained by combining an HER2 inhibitor and either a hormonal therapeutic agent or an anti-cancer agent. Specifically, the pharmaceutical agent consists of a combination of (1) an HER2 inhibitor represented by formula (I) (in formula I, R1 represents a hydrogen atom; R2 represents a C1-6 alkyl group substituted with a group indicated by a C1-4 alkyl that may be -NR6-CO-(CH2)n-SO2-halogenated (therein n is an integer from 1 to 4, R6 is a hydrogen atom or an C1-4 alkyl group, and -(CH2)n- can be substituted with a C1-4 alkyl); R3 represents a hydrogen atom or a C1-6 alkyl group; R4 represents a halogen atom or a C1-6 alkyl group; R5 represents a halogen atom or a C1-6 alkyl group; and X represents a hydrogen atom or a halogen atom; however, N-[2-(4-{[3-chloro-4-(3-chlorophenoxy)phenyl]amino}-5H-pyrrolo[3,2-d]pyrimidine-5-yl)ethyl] -2-(methylsulfonyl)acetoamide, is excluded) or a salt or prodrug thereof, and (2) a hormonal therapeutic agent or an anti-cancer agent.
Description
本発明は(1)HER2阻害作用を有する式:
The present invention is (1) a formula having HER2 inhibitory action:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせてなる医薬に関する。
また、本発明は、前記式(I)で表される化合物またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤に関する。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof and (2) a hormonal therapeutic agent or an anticancer agent.
The present invention also relates to a thymidine synthase production inhibitor comprising the compound represented by the formula (I) or a salt thereof or a prodrug thereof.
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせてなる医薬に関する。
また、本発明は、前記式(I)で表される化合物またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤に関する。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof and (2) a hormonal therapeutic agent or an anticancer agent.
The present invention also relates to a thymidine synthase production inhibitor comprising the compound represented by the formula (I) or a salt thereof or a prodrug thereof.
(発明の背景)
特許文献1にピロロピリミジン骨格またはピラゾロピリミジン骨格を有するHER2阻害剤が記載されている。
また、特許文献2には、HER2阻害剤である式: (Background of the Invention)
Patent Document 1 describes a HER2 inhibitor having a pyrrolopyrimidine skeleton or a pyrazolopyrimidine skeleton.
Patent Document 2 discloses a formula that is a HER2 inhibitor:
特許文献1にピロロピリミジン骨格またはピラゾロピリミジン骨格を有するHER2阻害剤が記載されている。
また、特許文献2には、HER2阻害剤である式: (Background of the Invention)
Patent Document 2 discloses a formula that is a HER2 inhibitor:
で表される化合物とトラスツズマブ等との併用による乳癌治療用途が記載されている。
国際公開第2005/010451号パンフレット
国際公開第2005/120512号パンフレット
The use of a compound represented by the above and trastuzumab in combination for breast cancer treatment is described.
International Publication No. 2005/010451 Pamphlet International Publication No. 2005/120512 Pamphlet
本発明は、癌の予防または治療剤として有用な(1)式:
The present invention is useful as an agent for preventing or treating cancer (1):
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)(以下、化合物(I)と略記することがある)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせてなる医薬(以下、本発明の併用剤と略記することがある)、及び前記式(I)で表される化合物またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤(以下、本発明のサイミジン合成酵素産生阻害剤と略記することがある)を提供することを目的とする。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) (hereinafter sometimes abbreviated as compound (I)) or a salt or prodrug thereof and (2) a hormonal therapeutic agent or an anticancer agent A thymidine synthase production inhibitor (hereinafter, referred to as a concomitant agent of the present invention) and a compound represented by the formula (I) or a salt or prodrug thereof It is an object of the present invention to provide a thymidine synthase production inhibitor of the present invention.
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)(以下、化合物(I)と略記することがある)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせてなる医薬(以下、本発明の併用剤と略記することがある)、及び前記式(I)で表される化合物またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤(以下、本発明のサイミジン合成酵素産生阻害剤と略記することがある)を提供することを目的とする。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) (hereinafter sometimes abbreviated as compound (I)) or a salt or prodrug thereof and (2) a hormonal therapeutic agent or an anticancer agent A thymidine synthase production inhibitor (hereinafter, referred to as a concomitant agent of the present invention) and a compound represented by the formula (I) or a salt or prodrug thereof It is an object of the present invention to provide a thymidine synthase production inhibitor of the present invention.
本発明者らは、鋭意検討した結果、(1)式:
As a result of intensive studies, the present inventors have found that the formula (1):
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせて使用することにより、単剤で用いる場合および他の併用医薬よりも有意な抗癌作用を示すこと、並びに、前記式(I)で表される化合物がサイミジン合成酵素産生阻害作用を示すことを見出し、さらに検討した結果、本発明を完成するに至った。すなわち、本発明は、
〔1〕(1)式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt thereof or a prodrug thereof, and (2) a hormonal therapeutic agent or an anticancer agent in combination for use as a single agent or in other combinations As a result of further investigation, it was found that the compound represented by the above formula (I) exhibits a significant anticancer activity than a pharmaceutical agent, and exhibits a thymidine synthase production inhibitory activity. . That is, the present invention
[1] Formula (1):
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせて使用することにより、単剤で用いる場合および他の併用医薬よりも有意な抗癌作用を示すこと、並びに、前記式(I)で表される化合物がサイミジン合成酵素産生阻害作用を示すことを見出し、さらに検討した結果、本発明を完成するに至った。すなわち、本発明は、
〔1〕(1)式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt thereof or a prodrug thereof, and (2) a hormonal therapeutic agent or an anticancer agent in combination for use as a single agent or in other combinations As a result of further investigation, it was found that the compound represented by the above formula (I) exhibits a significant anticancer activity than a pharmaceutical agent, and exhibits a thymidine synthase production inhibitory activity. . That is, the present invention
[1] Formula (1):
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる医薬;
〔2〕Xが水素原子である上記〔1〕記載の医薬;
〔3〕R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(R6aは水素原子またはメチル基、R7およびR8は、同一または異なって、それぞれ水素原子またはメチル基を表す)で表される基で置換されたC1-6アルキル基、
R3が水素原子、
R4が塩素原子またはメチル基、および
R5がフッ素原子、塩素原子またはメチル基である上記〔2〕記載の医薬;
〔4〕R7およびR8がメチル基である上記〔3〕記載の医薬;
〔5〕(1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔6〕(1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(エチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔7〕(1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-N,2-ジメチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔8〕(1)N-[2-(4-{[3-クロロ-4-(3-メチルフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔9〕(1)N-[2-(4-{[3-クロロ-4-(3-フルオロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔10〕(1)N-[2-(4-{[4-(3-クロロフェノキシ)-3-メチルフェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔11〕抗癌剤がHER2抗体、EGFR抗体、EGFR阻害剤、VEGFR阻害剤または化学療法剤である上記〔1〕記載の医薬;
〔12〕抗癌剤がトラスツズマブ、セツキシマブ、エルロチニブ、ゲフィチニブまたはパクリタキセルである上記〔1〕記載の医薬;
〔13〕ホルモン療法剤がERダウンレギュレーターである上記〔1〕記載の医薬;
〔14〕ホルモン療法剤がフルベストラントである上記〔1〕記載の医薬;
〔15〕癌の予防または治療剤である上記〔1〕記載の医薬;
〔16〕癌が乳癌、卵巣癌、前立腺癌、肺癌、膵癌、腎臓癌、大腸癌、小腸癌、食道癌または胃癌である上記〔15〕記載の医薬;
〔17〕哺乳動物に対して、(1)式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof;
(2) a pharmaceutical comprising a combination of a hormone therapy agent or an anticancer agent;
[2] The medicament according to [1] above, wherein X is a hydrogen atom;
[3] R 1 is a hydrogen atom,
R 2 is
—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (R 6a is a hydrogen atom or a methyl group, R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group) A C 1-6 alkyl group substituted with a group represented by:
R 3 is a hydrogen atom,
The medicament according to the above [2], wherein R 4 is a chlorine atom or a methyl group, and R 5 is a fluorine atom, a chlorine atom or a methyl group;
[4] The medicament according to [3] above, wherein R 7 and R 8 are methyl groups;
[5] (1) N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl 2-methyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[6] (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl ] -2- (ethylsulfonyl) acetamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[7] (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl -N, 2-dimethyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[8] (1) N- [2- (4-{[3-Chloro-4- (3-methylphenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl ] -2- (methylsulfonyl) acetamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[9] (1) N- [2- (4-{[3-Chloro-4- (3-fluorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl ] -2- (methylsulfonyl) acetamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[10] (1) N- [2- (4-{[4- (3-Chlorophenoxy) -3-methylphenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl 2-methyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[11] The medicament according to [1] above, wherein the anticancer agent is a HER2 antibody, EGFR antibody, EGFR inhibitor, VEGFR inhibitor or chemotherapeutic agent;
[12] The medicament according to [1] above, wherein the anticancer agent is trastuzumab, cetuximab, erlotinib, gefitinib or paclitaxel;
[13] The medicament according to [1] above, wherein the hormone therapy agent is an ER down regulator;
[14] The medicament according to [1] above, wherein the hormone therapy agent is fulvestrant;
[15] The medicament according to [1] above, which is a preventive or therapeutic agent for cancer;
[16] The medicament according to [15] above, wherein the cancer is breast cancer, ovarian cancer, prostate cancer, lung cancer, pancreatic cancer, kidney cancer, colon cancer, small intestine cancer, esophageal cancer or stomach cancer;
[17] For mammals, formula (1):
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる医薬;
〔2〕Xが水素原子である上記〔1〕記載の医薬;
〔3〕R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(R6aは水素原子またはメチル基、R7およびR8は、同一または異なって、それぞれ水素原子またはメチル基を表す)で表される基で置換されたC1-6アルキル基、
R3が水素原子、
R4が塩素原子またはメチル基、および
R5がフッ素原子、塩素原子またはメチル基である上記〔2〕記載の医薬;
〔4〕R7およびR8がメチル基である上記〔3〕記載の医薬;
〔5〕(1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔6〕(1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(エチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔7〕(1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-N,2-ジメチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔8〕(1)N-[2-(4-{[3-クロロ-4-(3-メチルフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔9〕(1)N-[2-(4-{[3-クロロ-4-(3-フルオロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔10〕(1)N-[2-(4-{[4-(3-クロロフェノキシ)-3-メチルフェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる上記〔1〕記載の医薬;
〔11〕抗癌剤がHER2抗体、EGFR抗体、EGFR阻害剤、VEGFR阻害剤または化学療法剤である上記〔1〕記載の医薬;
〔12〕抗癌剤がトラスツズマブ、セツキシマブ、エルロチニブ、ゲフィチニブまたはパクリタキセルである上記〔1〕記載の医薬;
〔13〕ホルモン療法剤がERダウンレギュレーターである上記〔1〕記載の医薬;
〔14〕ホルモン療法剤がフルベストラントである上記〔1〕記載の医薬;
〔15〕癌の予防または治療剤である上記〔1〕記載の医薬;
〔16〕癌が乳癌、卵巣癌、前立腺癌、肺癌、膵癌、腎臓癌、大腸癌、小腸癌、食道癌または胃癌である上記〔15〕記載の医薬;
〔17〕哺乳動物に対して、(1)式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof;
(2) a pharmaceutical comprising a combination of a hormone therapy agent or an anticancer agent;
[2] The medicament according to [1] above, wherein X is a hydrogen atom;
[3] R 1 is a hydrogen atom,
R 2 is
—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (R 6a is a hydrogen atom or a methyl group, R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group) A C 1-6 alkyl group substituted with a group represented by:
R 3 is a hydrogen atom,
The medicament according to the above [2], wherein R 4 is a chlorine atom or a methyl group, and R 5 is a fluorine atom, a chlorine atom or a methyl group;
[4] The medicament according to [3] above, wherein R 7 and R 8 are methyl groups;
[5] (1) N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl 2-methyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[6] (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl ] -2- (ethylsulfonyl) acetamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[7] (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl -N, 2-dimethyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[8] (1) N- [2- (4-{[3-Chloro-4- (3-methylphenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl ] -2- (methylsulfonyl) acetamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[9] (1) N- [2- (4-{[3-Chloro-4- (3-fluorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl ] -2- (methylsulfonyl) acetamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[10] (1) N- [2- (4-{[4- (3-Chlorophenoxy) -3-methylphenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl 2-methyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to [1] above, which is combined with a hormone therapy agent or an anticancer agent;
[11] The medicament according to [1] above, wherein the anticancer agent is a HER2 antibody, EGFR antibody, EGFR inhibitor, VEGFR inhibitor or chemotherapeutic agent;
[12] The medicament according to [1] above, wherein the anticancer agent is trastuzumab, cetuximab, erlotinib, gefitinib or paclitaxel;
[13] The medicament according to [1] above, wherein the hormone therapy agent is an ER down regulator;
[14] The medicament according to [1] above, wherein the hormone therapy agent is fulvestrant;
[15] The medicament according to [1] above, which is a preventive or therapeutic agent for cancer;
[16] The medicament according to [15] above, wherein the cancer is breast cancer, ovarian cancer, prostate cancer, lung cancer, pancreatic cancer, kidney cancer, colon cancer, small intestine cancer, esophageal cancer or stomach cancer;
[17] For mammals, formula (1):
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの有効量と、(2)ホルモン療法剤または抗癌剤の有効量とを投与することを特徴とする癌の予防または治療方法;
〔18〕癌の予防または治療剤を製造するための、(1)式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Ile) ethyl] -2- (methylsulfonyl) acetamide)) or a salt thereof or a prodrug thereof, and (2) an effective amount of a hormonal therapeutic agent or an anticancer agent, Or treatment method;
[18] Formula (1) for producing a preventive or therapeutic agent for cancer:
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの有効量と、(2)ホルモン療法剤または抗癌剤の有効量とを投与することを特徴とする癌の予防または治療方法;
〔18〕癌の予防または治療剤を製造するための、(1)式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Ile) ethyl] -2- (methylsulfonyl) acetamide)) or a salt thereof or a prodrug thereof, and (2) an effective amount of a hormonal therapeutic agent or an anticancer agent, Or treatment method;
[18] Formula (1) for producing a preventive or therapeutic agent for cancer:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤の使用;
〔19〕式 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof; and (2) use of a hormonal therapeutic agent or an anticancer agent;
[19] Formula
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤の使用;
〔19〕式 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof; and (2) use of a hormonal therapeutic agent or an anticancer agent;
[19] Formula
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤;
〔20〕哺乳動物に対して、式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof, a thymidine synthase production inhibitor;
[20] For mammals, the formula:
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤;
〔20〕哺乳動物に対して、式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof, a thymidine synthase production inhibitor;
[20] For mammals, the formula:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの有効量を投与することを特徴とするサイミジン合成酵素産生阻害方法;
〔21〕サイミジン合成酵素産生阻害剤を製造するための、式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt thereof or a prodrug thereof, and a method for inhibiting thymidine synthase production comprising administering an effective amount thereof;
[21] A formula for producing a thymidine synthase production inhibitor:
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの有効量を投与することを特徴とするサイミジン合成酵素産生阻害方法;
〔21〕サイミジン合成酵素産生阻害剤を製造するための、式: [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt thereof or a prodrug thereof, and a method for inhibiting thymidine synthase production comprising administering an effective amount thereof;
[21] A formula for producing a thymidine synthase production inhibitor:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの使用;等に関する。
以下に本発明について詳細に説明する。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof;
The present invention is described in detail below.
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの使用;等に関する。
以下に本発明について詳細に説明する。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof;
The present invention is described in detail below.
(1)式:
(1) Formula:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせてなる医薬は、単剤で用いる場合および他の併用医薬よりも有意な効果が認められ、癌の安全な予防または治療剤として有用である。
また、ホルモン療法剤にはHERファミリー分子を増加させ、HERファミリー分子の刺激を亢進することにより効果が減縮する恐れがある一方で、化合物(I)は、HERのシグナルを遮断する作用を有する。従って、特に(1)HER2阻害剤である化合物(I)と、(2)ホルモン療法剤とを組み合わせてなる医薬は、ホルモン療法剤単剤で用いる場合と比較してHERファミリー分子の増加を防ぎ、異なるメカニズムの薬剤を併用できる。
さらに、前記式(I)で表される化合物またはその塩もしくはそのプロドラッグは、サイミジン合成酵素産生阻害作用を有することにより、単剤の有効成分としても、癌の安全な予防または治療に有用である。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Ill) ethyl] -2- (methylsulfonyl) acetamide)) or a salt thereof or a prodrug thereof, and (2) a hormonal therapeutic agent or an anticancer agent are used in combination as a single agent or other combination drugs More significant effects are recognized, and it is useful as a safe preventive or therapeutic agent for cancer.
In addition, the hormonal therapeutic agent may increase the HER family molecule and enhance the stimulation of the HER family molecule, thereby reducing the effect. On the other hand, the compound (I) has an action of blocking the HER signal. Therefore, in particular, a pharmaceutical comprising a combination of (1) compound (I), which is a HER2 inhibitor, and (2) a hormonal therapeutic agent, prevents the increase in HER family molecules compared to the case of using a hormonal therapeutic agent alone. Drugs with different mechanisms can be used in combination.
Furthermore, the compound represented by the formula (I) or a salt thereof or a prodrug thereof has a thymidine synthase production inhibitory action, so that it is useful as a single agent active ingredient for the safe prevention or treatment of cancer. is there.
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤とを組み合わせてなる医薬は、単剤で用いる場合および他の併用医薬よりも有意な効果が認められ、癌の安全な予防または治療剤として有用である。
また、ホルモン療法剤にはHERファミリー分子を増加させ、HERファミリー分子の刺激を亢進することにより効果が減縮する恐れがある一方で、化合物(I)は、HERのシグナルを遮断する作用を有する。従って、特に(1)HER2阻害剤である化合物(I)と、(2)ホルモン療法剤とを組み合わせてなる医薬は、ホルモン療法剤単剤で用いる場合と比較してHERファミリー分子の増加を防ぎ、異なるメカニズムの薬剤を併用できる。
さらに、前記式(I)で表される化合物またはその塩もしくはそのプロドラッグは、サイミジン合成酵素産生阻害作用を有することにより、単剤の有効成分としても、癌の安全な予防または治療に有用である。 [Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Ill) ethyl] -2- (methylsulfonyl) acetamide)) or a salt thereof or a prodrug thereof, and (2) a hormonal therapeutic agent or an anticancer agent are used in combination as a single agent or other combination drugs More significant effects are recognized, and it is useful as a safe preventive or therapeutic agent for cancer.
In addition, the hormonal therapeutic agent may increase the HER family molecule and enhance the stimulation of the HER family molecule, thereby reducing the effect. On the other hand, the compound (I) has an action of blocking the HER signal. Therefore, in particular, a pharmaceutical comprising a combination of (1) compound (I), which is a HER2 inhibitor, and (2) a hormonal therapeutic agent, prevents the increase in HER family molecules compared to the case of using a hormonal therapeutic agent alone. Drugs with different mechanisms can be used in combination.
Furthermore, the compound represented by the formula (I) or a salt thereof or a prodrug thereof has a thymidine synthase production inhibitory action, so that it is useful as a single agent active ingredient for the safe prevention or treatment of cancer. is there.
(発明の詳細な説明)
以下に、化合物(I)で用いられる各用語について説明する。 (Detailed description of the invention)
Below, each term used by compound (I) is demonstrated.
以下に、化合物(I)で用いられる各用語について説明する。 (Detailed description of the invention)
Below, each term used by compound (I) is demonstrated.
本明細書中、特に記載の無い限り、「ハロゲン原子」(および置換基中の「ハロゲン」)としては、フッ素原子、塩素原子、臭素原子およびヨウ素原子が挙げられる。
Unless otherwise specified, in this specification, examples of the “halogen atom” (and “halogen” in a substituent) include a fluorine atom, a chlorine atom, a bromine atom, and an iodine atom.
本明細書中、特に記載の無い限り、「C1-6アルキル」としては、炭素数1ないし6の直鎖もしくは分枝状のアルキル、例えば、メチル、エチル、プロピル、イソプロピル、ブチル、イソブチル、sec-ブチル、tert-ブチル、ペンチル、イソペンチル、ネオペンチル、1-エチルプロピル、ヘキシル、イソヘキシル、1,1-ジメチルブチル、2,2-ジメチルブチル、3,3-ジメチルブチル、2-エチルブチル等が挙げられる。
In the present specification, unless otherwise specified, the term “C 1-6 alkyl” means straight or branched alkyl having 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, 1-ethylpropyl, hexyl, isohexyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-dimethylbutyl, 2-ethylbutyl, etc. It is done.
本明細書中、特に記載の無い限り、「C1-4アルキル」としては、炭素数1ないし4の直鎖もしくは分枝状のアルキル、例えば、メチル、エチル、プロピル、イソプロピル、ブチル、イソブチル、sec-ブチル、tert-ブチル等が挙げられる。
In the present specification, unless otherwise specified, “C 1-4 alkyl” means straight or branched alkyl having 1 to 4 carbon atoms such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and sec-butyl and tert-butyl.
以下、化合物(I)の各置換基の好ましい基を詳説する。
Hereinafter, preferred groups of each substituent of the compound (I) will be described in detail.
R2としては、式「-NR6a-CO-CR7R8-SO2-C1-4アルキル」で表される基で置換されたC1-6アルキル基(特に、エチル基)が好ましい。
R 2 is preferably a C 1-6 alkyl group (particularly an ethyl group) substituted with a group represented by the formula “—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl”. .
式中、R6aは水素原子またはメチル基、R7およびR8は、同一または異なって、それぞれ水素原子またはメチル基を表す。R7およびR8としては、メチル基が好ましい。
In the formula, R 6a is a hydrogen atom or a methyl group, and R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group. R 7 and R 8 are preferably a methyl group.
R3としては、水素原子が好ましい。
R 3 is preferably a hydrogen atom.
R4で表される「ハロゲン原子」としては、塩素原子が好ましい。また、R4で表される「C1-6アルキル基」としては、メチル基が好ましい。R4としては、塩素原子またはメチル基が好ましい。
The “halogen atom” represented by R 4 is preferably a chlorine atom. The “C 1-6 alkyl group” represented by R 4 is preferably a methyl group. R 4 is preferably a chlorine atom or a methyl group.
R5で表される「ハロゲン原子」としては、フッ素原子および塩素原子が好ましい。また、R5で表される「C1-6アルキル基」としては、メチル基が好ましい。R5としては、フッ素原子、塩素原子またはメチル基が好ましい。
The “halogen atom” represented by R 5 is preferably a fluorine atom or a chlorine atom. Further, the “C 1-6 alkyl group” represented by R 5 is preferably a methyl group. R 5 is preferably a fluorine atom, a chlorine atom or a methyl group.
Xで表される「ハロゲン原子」としては、フッ素原子が好ましい。Xとしては、水素原子またはフッ素原子が好ましく、水素原子がより好ましい。
The “halogen atom” represented by X is preferably a fluorine atom. X is preferably a hydrogen atom or a fluorine atom, more preferably a hydrogen atom.
化合物(I)の好適な態様としては、
R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(式中、R6aは水素原子またはメチル基、R7およびR8は、同一または異なって、それぞれ水素原子またはメチル基を示す)で表される基で置換されたC1-6アルキル基(特に、エチル基)、
R3が水素原子、
R4が塩素原子またはメチル基、
R5がフッ素原子、塩素原子またはメチル基、
Xが水素原子またはフッ素原子(好ましくは、水素原子)である、
化合物が挙げられる。 As a preferred embodiment of compound (I),
R 1 is a hydrogen atom,
R 2 is
—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (wherein R 6a is a hydrogen atom or a methyl group, R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group) A C 1-6 alkyl group (particularly an ethyl group) substituted with a group represented by
R 3 is a hydrogen atom,
R 4 is a chlorine atom or a methyl group,
R 5 is a fluorine atom, a chlorine atom or a methyl group,
X is a hydrogen atom or a fluorine atom (preferably a hydrogen atom),
Compounds.
R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(式中、R6aは水素原子またはメチル基、R7およびR8は、同一または異なって、それぞれ水素原子またはメチル基を示す)で表される基で置換されたC1-6アルキル基(特に、エチル基)、
R3が水素原子、
R4が塩素原子またはメチル基、
R5がフッ素原子、塩素原子またはメチル基、
Xが水素原子またはフッ素原子(好ましくは、水素原子)である、
化合物が挙げられる。 As a preferred embodiment of compound (I),
R 1 is a hydrogen atom,
R 2 is
—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (wherein R 6a is a hydrogen atom or a methyl group, R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group) A C 1-6 alkyl group (particularly an ethyl group) substituted with a group represented by
R 3 is a hydrogen atom,
R 4 is a chlorine atom or a methyl group,
R 5 is a fluorine atom, a chlorine atom or a methyl group,
X is a hydrogen atom or a fluorine atom (preferably a hydrogen atom),
Compounds.
化合物(I)のさらに好適な態様としては、
R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(式中、R6aは水素原子またはメチル基、R7およびR8はメチル基を示す)で表される基で置換されたC1-6アルキル基(特に、エチル基)、
R3が水素原子、
R4が塩素原子またはメチル基、
R5がフッ素原子、塩素原子またはメチル基、
Xが水素原子またはフッ素原子(好ましくは、水素原子)である、
化合物が挙げられる。 As a more preferred embodiment of the compound (I),
R 1 is a hydrogen atom,
R 2 is
Substituted with a group represented by —NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (wherein R 6a represents a hydrogen atom or a methyl group, and R 7 and R 8 represent a methyl group) A C 1-6 alkyl group (especially an ethyl group),
R 3 is a hydrogen atom,
R 4 is a chlorine atom or a methyl group,
R 5 is a fluorine atom, a chlorine atom or a methyl group,
X is a hydrogen atom or a fluorine atom (preferably a hydrogen atom),
Compounds.
R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(式中、R6aは水素原子またはメチル基、R7およびR8はメチル基を示す)で表される基で置換されたC1-6アルキル基(特に、エチル基)、
R3が水素原子、
R4が塩素原子またはメチル基、
R5がフッ素原子、塩素原子またはメチル基、
Xが水素原子またはフッ素原子(好ましくは、水素原子)である、
化合物が挙げられる。 As a more preferred embodiment of the compound (I),
R 1 is a hydrogen atom,
R 2 is
Substituted with a group represented by —NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (wherein R 6a represents a hydrogen atom or a methyl group, and R 7 and R 8 represent a methyl group) A C 1-6 alkyl group (especially an ethyl group),
R 3 is a hydrogen atom,
R 4 is a chlorine atom or a methyl group,
R 5 is a fluorine atom, a chlorine atom or a methyl group,
X is a hydrogen atom or a fluorine atom (preferably a hydrogen atom),
Compounds.
化合物(I)として、特に好ましくは、
N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミド、
N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(エチルスルホニル)アセトアミド、
N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-N,2-ジメチル-2-(メチルスルホニル)プロパンアミド、
N-[2-(4-{[3-クロロ-4-(3-メチルフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミド、
N-[2-(4-{[3-クロロ-4-(3-フルオロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミド、および
N-[2-(4-{[4-(3-クロロフェノキシ)-3-メチルフェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミド、
が挙げられる。 As compound (I), particularly preferably,
N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2-methyl- 2- (methylsulfonyl) propanamide,
N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2- (ethyl Sulfonyl) acetamide,
N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -N, 2- Dimethyl-2- (methylsulfonyl) propanamide,
N- [2- (4-{[3-Chloro-4- (3-methylphenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2- (methyl Sulfonyl) acetamide,
N- [2- (4-{[3-Chloro-4- (3-fluorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2- (methyl Sulfonyl) acetamide, and N- [2- (4-{[4- (3-chlorophenoxy) -3-methylphenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2-methyl-2- (methylsulfonyl) propanamide,
Is mentioned.
N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミド、
N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(エチルスルホニル)アセトアミド、
N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-N,2-ジメチル-2-(メチルスルホニル)プロパンアミド、
N-[2-(4-{[3-クロロ-4-(3-メチルフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミド、
N-[2-(4-{[3-クロロ-4-(3-フルオロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミド、および
N-[2-(4-{[4-(3-クロロフェノキシ)-3-メチルフェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミド、
が挙げられる。 As compound (I), particularly preferably,
N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2-methyl- 2- (methylsulfonyl) propanamide,
N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2- (ethyl Sulfonyl) acetamide,
N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -N, 2- Dimethyl-2- (methylsulfonyl) propanamide,
N- [2- (4-{[3-Chloro-4- (3-methylphenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2- (methyl Sulfonyl) acetamide,
N- [2- (4-{[3-Chloro-4- (3-fluorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2- (methyl Sulfonyl) acetamide, and N- [2- (4-{[4- (3-chlorophenoxy) -3-methylphenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2-methyl-2- (methylsulfonyl) propanamide,
Is mentioned.
化合物(I)及びその塩は、国際公開第2007/064045号パンフレット記載の方法に従い製造することができる。
Compound (I) and a salt thereof can be produced according to the method described in International Publication No. 2007/064045.
化合物(I)が、光学異性体、立体異性体、位置異性体、回転異性体等の異性体を有する場合には、いずれか一方の異性体も混合物も化合物(I)に包含される。例えば、化合物(I)に光学異性体が存在する場合には、ラセミ体から分割された光学異性体も化合物(I)に包含される。これらの異性体は、自体公知の合成手法、分離手法(濃縮、溶媒抽出、カラムクロマトグラフィー、再結晶など)によりそれぞれを単品として得ることができる。
化合物(I)は、結晶であってもよく、結晶形が単一であっても結晶形混合物であっても化合物(I)に包含される。結晶は、自体公知の結晶化法を適用して、結晶化することによって製造することができる。
化合物(I)は、溶媒和物(例えば、水和物等)であっても、無溶媒和物であってもよく、いずれも化合物(I)に包含される。
同位元素(例、2H,3H,14C,35S,125Iなど)などで標識された化合物も、化合物(I)に包含される。 When compound (I) has an isomer such as an optical isomer, a stereoisomer, a positional isomer, a rotational isomer, etc., any one of the isomers and a mixture are encompassed in compound (I). For example, when compound (I) has an optical isomer, the optical isomer resolved from the racemate is also encompassed in compound (I). Each of these isomers can be obtained as a single product by a known synthesis method or separation method (concentration, solvent extraction, column chromatography, recrystallization, etc.).
Compound (I) may be a crystal, and it is included in compound (I) regardless of whether the crystal form is a single crystal form or a crystal form mixture. The crystal can be produced by crystallization by applying a crystallization method known per se.
Compound (I) may be a solvate (such as a hydrate) or a non-solvate, and both are encompassed in compound (I).
Compounds labeled with isotopes (eg, 2 H, 3 H, 14 C, 35 S, 125 I, etc.) are also encompassed in compound (I).
化合物(I)は、結晶であってもよく、結晶形が単一であっても結晶形混合物であっても化合物(I)に包含される。結晶は、自体公知の結晶化法を適用して、結晶化することによって製造することができる。
化合物(I)は、溶媒和物(例えば、水和物等)であっても、無溶媒和物であってもよく、いずれも化合物(I)に包含される。
同位元素(例、2H,3H,14C,35S,125Iなど)などで標識された化合物も、化合物(I)に包含される。 When compound (I) has an isomer such as an optical isomer, a stereoisomer, a positional isomer, a rotational isomer, etc., any one of the isomers and a mixture are encompassed in compound (I). For example, when compound (I) has an optical isomer, the optical isomer resolved from the racemate is also encompassed in compound (I). Each of these isomers can be obtained as a single product by a known synthesis method or separation method (concentration, solvent extraction, column chromatography, recrystallization, etc.).
Compound (I) may be a crystal, and it is included in compound (I) regardless of whether the crystal form is a single crystal form or a crystal form mixture. The crystal can be produced by crystallization by applying a crystallization method known per se.
Compound (I) may be a solvate (such as a hydrate) or a non-solvate, and both are encompassed in compound (I).
Compounds labeled with isotopes (eg, 2 H, 3 H, 14 C, 35 S, 125 I, etc.) are also encompassed in compound (I).
化合物(I)の塩としては、例えば金属塩、アンモニウム塩、有機塩基との塩、無機酸との塩、有機酸との塩、塩基性または酸性アミノ酸との塩等が挙げられる。金属塩の好適な例としては、例えばナトリウム塩、カリウム塩等のアルカリ金属塩;カルシウム塩、マグネシウム塩、バリウム塩等のアルカリ土類金属塩;アルミニウム塩等が挙げられる。有機塩基との塩の好適な例としては、例えばトリメチルアミン、トリエチルアミン、ピリジン、ピコリン、2,6-ルチジン、エタノールアミン、ジエタノールアミン、トリエタノールアミン、トロメタン[トリス(ヒドロキシメチル)メチルアミン]、t-ブチルアミン、シクロヘキシルアミン、ジシクロヘキシルアミン、N,N’-ジベンジルエチレンジアミン等との塩が挙げられる。無機酸との塩の好適な例としては、例えば塩酸、臭化水素酸、硝酸、硫酸、リン酸等との塩が挙げられる。有機酸との塩の好適な例としては、例えばギ酸、酢酸、トリフルオロ酢酸、フタル酸、フマル酸、シュウ酸、酒石酸、マレイン酸、クエン酸、コハク酸、リンゴ酸、メタンスルホン酸、ベンゼンスルホン酸、p-トルエンスルホン酸等との塩が挙げられる。塩基性アミノ酸との塩の好適な例としては、例えばアルギニン、リジン、オルニチン等との塩が挙げられ、酸性アミノ酸との塩の好適な例としては、例えばアスパラギン酸、グルタミン酸等との塩が挙げられる。
Examples of the salt of compound (I) include metal salts, ammonium salts, salts with organic bases, salts with inorganic acids, salts with organic acids, salts with basic or acidic amino acids, and the like. Preferable examples of the metal salt include alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as calcium salt, magnesium salt and barium salt; aluminum salt and the like. Preferable examples of the salt with organic base include trimethylamine, triethylamine, pyridine, picoline, 2,6-lutidine, ethanolamine, diethanolamine, triethanolamine, tromethane [tris (hydroxymethyl) methylamine], t-butylamine. , Salts with cyclohexylamine, dicyclohexylamine, N, N′-dibenzylethylenediamine and the like. Preferable examples of the salt with inorganic acid include salts with hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid and the like. Preferable examples of the salt with organic acid include formic acid, acetic acid, trifluoroacetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, and benzenesulfone. And salts with acid, p-toluenesulfonic acid and the like. Preferable examples of salts with basic amino acids include salts with arginine, lysine, ornithine and the like, and preferable examples of salts with acidic amino acids include salts with aspartic acid and glutamic acid, for example. It is done.
このうち、薬学的に許容し得る塩が好ましく、例えば塩酸、臭化水素酸、硝酸、硫酸、リン酸等無機酸との塩、または酢酸、フタル酸、フマル酸、シュウ酸、酒石酸、マレイン酸、クエン酸、コハク酸、メタンスルホン酸、p-トルエンスルホン酸等の有機酸との塩が挙げられる。特に、p-トルエンスルホン酸との塩が好ましい。
化合物(I)またはその塩のプロドラッグは、生体内における生理条件下で酵素や胃酸等による反応により化合物(I)またはその塩に変換する化合物、即ち酵素的に酸化、還元、加水分解等を起こして化合物(I)またはその塩に変化する化合物、胃酸等により加水分解等を起こして化合物(I)またはその塩に変化する化合物をいう。化合物(I)またはその塩のプロドラッグとしては、化合物(I)またはその塩のアミノがアシル化、アルキル化、りん酸化された化合物(例、化合物(I)またはその塩のアミノがエイコサノイル化、アラニル化、ペンチルアミノカルボニル化、(5-メチル-2-オキソ-1,3-ジオキソレン-4-イル)メトキシカルボニル化、テトラヒドロフラニル化、ピロリジルメチル化、ピバロイルオキシメチル化、tert-ブチル化された化合物等)等が用いられる。これらの化合物は自体公知の方法によって化合物(I)またはその塩から製造することができる。
また、化合物(I)またはその塩のプロドラッグは、広川書店1990年刊「医薬品の開発」第7巻分子設計163頁から198頁に記載されているような生理的条件で化合物(I)またはその塩に変化するものであってもよい。
以下、化合物(I)またはその塩もしくはそのプロドラッグを化合物(I)と略記する。 Of these, pharmaceutically acceptable salts are preferred, for example, salts with inorganic acids such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, or acetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid And salts with organic acids such as citric acid, succinic acid, methanesulfonic acid and p-toluenesulfonic acid. In particular, a salt with p-toluenesulfonic acid is preferred.
A prodrug of compound (I) or a salt thereof is a compound that is converted into compound (I) or a salt thereof by a reaction with an enzyme, gastric acid or the like under physiological conditions in vivo, that is, enzymatically oxidized, reduced, hydrolyzed, etc. It refers to a compound that changes to compound (I) or a salt thereof, or a compound that undergoes hydrolysis or the like by gastric acid or the like and changes to compound (I) or a salt thereof. As a prodrug of the compound (I) or a salt thereof, a compound in which the amino of the compound (I) or a salt thereof is acylated, alkylated or phosphorylated (eg, the amino of the compound (I) or a salt thereof is eicosanoylated, Alanylation, pentylaminocarbonylation, (5-methyl-2-oxo-1,3-dioxolen-4-yl) methoxycarbonylation, tetrahydrofuranylation, pyrrolidylmethylation, pivaloyloxymethylation, tert-butyl And the like are used. These compounds can be produced from compound (I) or a salt thereof by a method known per se.
The prodrug of compound (I) or a salt thereof is compound (I) or a compound thereof under physiological conditions as described in Hirokawa Shoten, 1990, “Pharmaceutical Development”, Volume 7, Molecular Design, pages 163 to 198. It may change into a salt.
Hereinafter, Compound (I) or a salt thereof or a prodrug thereof is abbreviated as Compound (I).
化合物(I)またはその塩のプロドラッグは、生体内における生理条件下で酵素や胃酸等による反応により化合物(I)またはその塩に変換する化合物、即ち酵素的に酸化、還元、加水分解等を起こして化合物(I)またはその塩に変化する化合物、胃酸等により加水分解等を起こして化合物(I)またはその塩に変化する化合物をいう。化合物(I)またはその塩のプロドラッグとしては、化合物(I)またはその塩のアミノがアシル化、アルキル化、りん酸化された化合物(例、化合物(I)またはその塩のアミノがエイコサノイル化、アラニル化、ペンチルアミノカルボニル化、(5-メチル-2-オキソ-1,3-ジオキソレン-4-イル)メトキシカルボニル化、テトラヒドロフラニル化、ピロリジルメチル化、ピバロイルオキシメチル化、tert-ブチル化された化合物等)等が用いられる。これらの化合物は自体公知の方法によって化合物(I)またはその塩から製造することができる。
また、化合物(I)またはその塩のプロドラッグは、広川書店1990年刊「医薬品の開発」第7巻分子設計163頁から198頁に記載されているような生理的条件で化合物(I)またはその塩に変化するものであってもよい。
以下、化合物(I)またはその塩もしくはそのプロドラッグを化合物(I)と略記する。 Of these, pharmaceutically acceptable salts are preferred, for example, salts with inorganic acids such as hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, or acetic acid, phthalic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid And salts with organic acids such as citric acid, succinic acid, methanesulfonic acid and p-toluenesulfonic acid. In particular, a salt with p-toluenesulfonic acid is preferred.
A prodrug of compound (I) or a salt thereof is a compound that is converted into compound (I) or a salt thereof by a reaction with an enzyme, gastric acid or the like under physiological conditions in vivo, that is, enzymatically oxidized, reduced, hydrolyzed, etc. It refers to a compound that changes to compound (I) or a salt thereof, or a compound that undergoes hydrolysis or the like by gastric acid or the like and changes to compound (I) or a salt thereof. As a prodrug of the compound (I) or a salt thereof, a compound in which the amino of the compound (I) or a salt thereof is acylated, alkylated or phosphorylated (eg, the amino of the compound (I) or a salt thereof is eicosanoylated, Alanylation, pentylaminocarbonylation, (5-methyl-2-oxo-1,3-dioxolen-4-yl) methoxycarbonylation, tetrahydrofuranylation, pyrrolidylmethylation, pivaloyloxymethylation, tert-butyl And the like are used. These compounds can be produced from compound (I) or a salt thereof by a method known per se.
The prodrug of compound (I) or a salt thereof is compound (I) or a compound thereof under physiological conditions as described in Hirokawa Shoten, 1990, “Pharmaceutical Development”, Volume 7, Molecular Design, pages 163 to 198. It may change into a salt.
Hereinafter, Compound (I) or a salt thereof or a prodrug thereof is abbreviated as Compound (I).
本明細書中、「ホルモン療法剤」としては、例えば、ホスフェストロール、ジエチルスチルベストロール、クロロトリアニセン、酢酸メドロキシプロゲステロン、酢酸メゲストロール、酢酸クロルマジノン、酢酸シプロテロン、ダナゾール、ジエノゲスト、アソプリスニル、アリルエストレノール、ゲストリノン、ノメゲストロール、タデナン、メパルトリシン、ラロキシフェン、オルメロキシフェン、レボルメロキシフェン、抗エストロゲン(例、クエン酸タモキシフェン、クエン酸トレミフェン等)、ERダウンレギュレーター(例えばフルベストラント(ファスロデックス(商標))等)、ヒト閉経ゴナドトロピン、卵胞刺激ホルモン、ピル製剤、メピチオスタン、テストロラクトン、アミノグルテチイミド、LH-RHアゴニスト(例、酢酸ゴセレリン、ブセレリン、リュープロレリン等)、ドロロキシフェン、エピチオスタノール、スルホン酸エチニルエストラジオール、アロマターゼ阻害薬(例、塩酸ファドロゾール、アナストロゾール、レトロゾール、エキセメスタン、ボロゾール、フォルメスタン等)、抗アンドロゲン(例、フルタミド、ビカルタミド、ニルタミド等)、5α-レダクターゼ阻害薬(例、フィナステリド、デュタステリド、エプリステリド等)、副腎皮質ホルモン系薬剤(例、デキサメタゾン、プレドニゾロン、ベタメタゾン、トリアムシノロン等)、アンドロゲン合成阻害薬(例、アビラテロン等)、レチノイドおよびレチノイドの代謝を遅らせる薬剤(例、リアロゾール等)等が用いられ、なかでもLH-RHアゴニスト(例、酢酸ゴセレリン、ブセレリン、リュープロレリン等)、ERダウンレギュレーター(例えばフルベストラント(ファスロデックス(商標))等)が好ましい。
In the present specification, examples of the `` hormone therapeutic agent '' include phosfestol, diethylstilbestrol, chlorotrianicene, medroxyprogesterone acetate, megestrol acetate, chlormadinone acetate, cyproterone acetate, danazol, dienogest, azoprisnil, Allylestrenol, gestrinone, nomegestrol, tadenane, mepaltricine, raloxifene, oloxifene, levormeloxifene, antiestrogens (eg, tamoxifen citrate, toremifene citrate), ER down regulators (eg fulvestrant ( Faslodex (trademark), etc.), human menopausal gonadotropin, follicle stimulating hormone, pill preparation, mepithiostan, testrolactone, aminoglutethimide, LH-RH agonist ( , Goserelin acetate, buserelin, leuprorelin, etc.), droloxifene, epithiostanol, ethinyl estradiol sulfonate, aromatase inhibitor (eg, fadrozole hydrochloride, anastrozole, letrozole, exemestane, borozole, formestane, etc.), Antiandrogens (eg, flutamide, bicalutamide, nilutamide, etc.), 5α-reductase inhibitors (eg, finasteride, dutasteride, epristeride, etc.), corticosteroids (eg, dexamethasone, prednisolone, betamethasone, triamcinolone, etc.), androgen synthesis inhibition Drugs (eg, abiraterone, etc.), retinoids and drugs that delay the metabolism of retinoids (eg, riarozole, etc.) are used, among them LH-RH agonists (eg, goserelin acetate) Buserelin, leuprorelin etc.), ER down regulator (for example fulvestrant (Faslodex (TM)) and the like) are preferable.
本明細書中、「抗癌剤」としては、例えば、化学療法剤、免疫療法剤、細胞増殖因子ならびにその受容体の作用を阻害する薬剤等があげられる。
In the present specification, examples of the “anticancer agent” include a chemotherapeutic agent, an immunotherapeutic agent, a cell growth factor, and a drug that inhibits the action of its receptor.
該「化学療法剤」としては、例えばアルキル化剤、代謝拮抗剤、抗癌性抗生物質、植物由来抗癌剤等が用いられる。
「アルキル化剤」としては、例えば、ナイトロジェンマスタード、塩酸ナイトロジェンマスタード-N-オキシド、クロラムブチル、シクロフォスファミド、イホスファミド、チオテパ、カルボコン、トシル酸インプロスルファン、ブスルファン、塩酸ニムスチン、ミトブロニトール、メルファラン、ダカルバジン、ラニムスチン、リン酸エストラムスチンナトリウム、トリエチレンメラミン、カルムスチン、ロムスチン、ストレプトゾシン、ピポブロマン、エトグルシド、カルボプラチン、シスプラチン、ミボプラチン、ネダプラチン、オキサリプラチン、アルトレタミン、アンバムスチン、塩酸ジブロスピジウム、フォテムスチン、プレドニムスチン、プミテパ、リボムスチン、テモゾロミド、トレオスルファン、トロフォスファミド、ジノスタチンスチマラマー、アドゼレシン、システムスチン、ビゼレシン等が用いられる。
「代謝拮抗剤」としては、例えば、メルカプトプリン、6-メルカプトプリンリボシド、チオイノシン、メトトレキサート、エノシタビン、シタラビン、シタラビンオクフォスファート、塩酸アンシタビン、5-FU系薬剤(例、フルオロウラシル、テガフール、UFT、ドキシフルリジン、カルモフール、ガロシタビン、エミテフール等)、アミノプテリン、ロイコボリンカルシウム、タブロイド、ブトシン、フォリネイトカルシウム、レボフォリネイトカルシウム、クラドリビン、エミテフール、フルダラビン、ゲムシタビン、ヒドロキシカルバミド、ペントスタチン、ピリトレキシム、イドキシウリジン、ミトグアゾン、チアゾフリン、アンバムスチン、ペメトレキセド二ナトリウム塩(アリムタ(商標))等が用いられる。
「抗癌性抗生物質」としては、例えば、アクチノマイシンD、アクチノマイシンC、マイトマイシンC、クロモマイシンA3、塩酸ブレオマイシン、硫酸ブレオマイシン、硫酸ペプロマイシン、塩酸ダウノルビシン、塩酸ドキソルビシン(アドリアシン(商標))、塩酸アクラルビシン、塩酸ピラルビシン、塩酸エピルビシン、ネオカルチノスタチン、ミスラマイシン、ザルコマイシン、カルチノフィリン、ミトタン、塩酸ゾルビシン、塩酸ミトキサントロン、塩酸イダルビシン等が用いられる。
「植物由来抗癌剤」としては、例えば、エトポシド、リン酸エトポシド、硫酸ビンブラスチン、硫酸ビンクリスチン、硫酸ビンデシン、テニポシド、パクリタキセル(タキソール(商標))、ドセタキセル、ビノレルビン等が用いられる。 Examples of the “chemotherapeutic agent” include alkylating agents, antimetabolites, anticancer antibiotics, plant-derived anticancer agents and the like.
Examples of the “alkylating agent” include nitrogen mustard, nitrogen mustard hydrochloride-N-oxide, chlorambutyl, cyclophosphamide, ifosfamide, thiotepa, carbocon, improsulfan tosylate, busulfan, nimustine hydrochloride, mitoblonitol, Faran, dacarbazine, ranimustine, estramustine phosphate sodium, triethylenemelamine, carmustine, lomustine, streptozocin, piprobroman, etoglucid, carboplatin, cisplatin, miboplatin, nedaplatin, oxaliplatin, altretamine, ambermuthine, dibrospine hydrochloride, fotemustine hydrochloride Predonimustine, pumitepa, ribomustine, temozolomide, treosulphane, trophosphamide, Roh statins scan Chima Lamar, adozelesin, system scan Chin, Bizereshin or the like is used.
Examples of the “antimetabolite” include mercaptopurine, 6-mercaptopurine riboside, thioinosine, methotrexate, enocitabine, cytarabine, cytarabine okphosphat, ancitabine hydrochloride, 5-FU drugs (eg, fluorouracil, tegafur, UFT, Doxyfluridine, carmofur, galocitabine, emiteful, etc.), aminopterin, leucovorin calcium, tabloid, butosine, folinate calcium, levofolinate calcium, cladribine, emiteful, fludarabine, gemcitabine, hydroxycarbamide, pentostatin, pyritorexime, idoxyuridine, mitoxifridin Ambamustine, pemetrexed disodium salt (Alimta (trademark)) and the like are used.
Examples of the “anticancer antibiotic” include actinomycin D, actinomycin C, mitomycin C, chromomycin A3, bleomycin hydrochloride, bleomycin sulfate, pepromycin sulfate, daunorubicin hydrochloride, doxorubicin hydrochloride (Adriacin (trademark)), acrarubicin hydrochloride , Pirarubicin hydrochloride, epirubicin hydrochloride, neocalcinostatin, misramycin, sarcomycin, carcinophylline, mitotane, zorubicin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride and the like.
Examples of the “plant-derived anticancer agent” include etoposide, etoposide phosphate, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel (Taxol ™), docetaxel, vinorelbine and the like.
「アルキル化剤」としては、例えば、ナイトロジェンマスタード、塩酸ナイトロジェンマスタード-N-オキシド、クロラムブチル、シクロフォスファミド、イホスファミド、チオテパ、カルボコン、トシル酸インプロスルファン、ブスルファン、塩酸ニムスチン、ミトブロニトール、メルファラン、ダカルバジン、ラニムスチン、リン酸エストラムスチンナトリウム、トリエチレンメラミン、カルムスチン、ロムスチン、ストレプトゾシン、ピポブロマン、エトグルシド、カルボプラチン、シスプラチン、ミボプラチン、ネダプラチン、オキサリプラチン、アルトレタミン、アンバムスチン、塩酸ジブロスピジウム、フォテムスチン、プレドニムスチン、プミテパ、リボムスチン、テモゾロミド、トレオスルファン、トロフォスファミド、ジノスタチンスチマラマー、アドゼレシン、システムスチン、ビゼレシン等が用いられる。
「代謝拮抗剤」としては、例えば、メルカプトプリン、6-メルカプトプリンリボシド、チオイノシン、メトトレキサート、エノシタビン、シタラビン、シタラビンオクフォスファート、塩酸アンシタビン、5-FU系薬剤(例、フルオロウラシル、テガフール、UFT、ドキシフルリジン、カルモフール、ガロシタビン、エミテフール等)、アミノプテリン、ロイコボリンカルシウム、タブロイド、ブトシン、フォリネイトカルシウム、レボフォリネイトカルシウム、クラドリビン、エミテフール、フルダラビン、ゲムシタビン、ヒドロキシカルバミド、ペントスタチン、ピリトレキシム、イドキシウリジン、ミトグアゾン、チアゾフリン、アンバムスチン、ペメトレキセド二ナトリウム塩(アリムタ(商標))等が用いられる。
「抗癌性抗生物質」としては、例えば、アクチノマイシンD、アクチノマイシンC、マイトマイシンC、クロモマイシンA3、塩酸ブレオマイシン、硫酸ブレオマイシン、硫酸ペプロマイシン、塩酸ダウノルビシン、塩酸ドキソルビシン(アドリアシン(商標))、塩酸アクラルビシン、塩酸ピラルビシン、塩酸エピルビシン、ネオカルチノスタチン、ミスラマイシン、ザルコマイシン、カルチノフィリン、ミトタン、塩酸ゾルビシン、塩酸ミトキサントロン、塩酸イダルビシン等が用いられる。
「植物由来抗癌剤」としては、例えば、エトポシド、リン酸エトポシド、硫酸ビンブラスチン、硫酸ビンクリスチン、硫酸ビンデシン、テニポシド、パクリタキセル(タキソール(商標))、ドセタキセル、ビノレルビン等が用いられる。 Examples of the “chemotherapeutic agent” include alkylating agents, antimetabolites, anticancer antibiotics, plant-derived anticancer agents and the like.
Examples of the “alkylating agent” include nitrogen mustard, nitrogen mustard hydrochloride-N-oxide, chlorambutyl, cyclophosphamide, ifosfamide, thiotepa, carbocon, improsulfan tosylate, busulfan, nimustine hydrochloride, mitoblonitol, Faran, dacarbazine, ranimustine, estramustine phosphate sodium, triethylenemelamine, carmustine, lomustine, streptozocin, piprobroman, etoglucid, carboplatin, cisplatin, miboplatin, nedaplatin, oxaliplatin, altretamine, ambermuthine, dibrospine hydrochloride, fotemustine hydrochloride Predonimustine, pumitepa, ribomustine, temozolomide, treosulphane, trophosphamide, Roh statins scan Chima Lamar, adozelesin, system scan Chin, Bizereshin or the like is used.
Examples of the “antimetabolite” include mercaptopurine, 6-mercaptopurine riboside, thioinosine, methotrexate, enocitabine, cytarabine, cytarabine okphosphat, ancitabine hydrochloride, 5-FU drugs (eg, fluorouracil, tegafur, UFT, Doxyfluridine, carmofur, galocitabine, emiteful, etc.), aminopterin, leucovorin calcium, tabloid, butosine, folinate calcium, levofolinate calcium, cladribine, emiteful, fludarabine, gemcitabine, hydroxycarbamide, pentostatin, pyritorexime, idoxyuridine, mitoxifridin Ambamustine, pemetrexed disodium salt (Alimta (trademark)) and the like are used.
Examples of the “anticancer antibiotic” include actinomycin D, actinomycin C, mitomycin C, chromomycin A3, bleomycin hydrochloride, bleomycin sulfate, pepromycin sulfate, daunorubicin hydrochloride, doxorubicin hydrochloride (Adriacin (trademark)), acrarubicin hydrochloride , Pirarubicin hydrochloride, epirubicin hydrochloride, neocalcinostatin, misramycin, sarcomycin, carcinophylline, mitotane, zorubicin hydrochloride, mitoxantrone hydrochloride, idarubicin hydrochloride and the like.
Examples of the “plant-derived anticancer agent” include etoposide, etoposide phosphate, vinblastine sulfate, vincristine sulfate, vindesine sulfate, teniposide, paclitaxel (Taxol ™), docetaxel, vinorelbine and the like.
該「免疫療法剤(BRM)」としては、例えば、ピシバニール、クレスチン、シゾフィラン、レンチナン、ウベニメクス、インターフェロン、インターロイキン、マクロファージコロニー刺激因子、顆粒球コロニー刺激因子、エリスロポイエチン、リンホトキシン、BCGワクチン、コリネバクテリウムパルブム、レバミゾール、ポリサッカライドK、プロコダゾール等が用いられる。
Examples of the “immunotherapy agent (BRM)” include picibanil, krestin, schizophyllan, lentinan, ubenimex, interferon, interleukin, macrophage colony stimulating factor, granulocyte colony stimulating factor, erythropoietin, lymphotoxin, BCG vaccine, coryne Bacterium parvum, levamisole, polysaccharide K, procodazole and the like are used.
該「細胞増殖因子ならびにその受容体の作用を阻害する薬剤」における「細胞増殖因子」としては、細胞の増殖を促進する物質であればどのようなものでもよく、通常、分子量が20,000以下のペプチドで、受容体との結合により低濃度で作用が発揮される因子が用いられ、具体的には、(1)EGF(epidermal growth factor)またはそれと実質的に同一の活性を有する物質〔例、EGF、ハレグリン等〕、(2)インシュリンまたはそれと実質的に同一の活性を有する物質〔例、インシュリン、IGF(insulin-like growth factor)-1、IGF-2等〕、(3)FGF(fibroblast growth factor)またはそれと実質的に同一の活性を有する物質〔例、酸性FGF、塩基性FGF、KGF(keratinocyte growth factor)、FGF-10等〕、(4)その他の細胞増殖因子〔例、CSF(colony stimulating factor)、EPO(erythropoietin)、IL-2(interleukin-2)、NGF(nerve growth factor)、PDGF(platelet-derived growth factor)、TGFβ(transforming growth factor β)、HGF(hepatocyte growth factor)、VEGF(vascular endothelial growth factor)等〕等が挙げられる。
該「細胞増殖因子の受容体」としては、前記の細胞増殖因子と結合能を有する受容体であればいかなるものであってもよく、具体的には、EGF受容体、ハレグリン受容体(HER2)、インシュリン受容体、IGF受容体、FGF受容体-1またはFGF受容体-2等が挙げられる。
該「細胞増殖因子の作用を阻害する薬剤」としては、HER2抗体(トラスツズマブ(ハーセプチン(商標))等)、メシル酸イマチニブ、ZD1839またはEGFR抗体(セツキシマブ(エルビタックス(Erbitux)(商標))等)、VEGFに対する抗体(例、ベバシツマブ(アバスチン(Avastin)(商標)))、VEGFR抗体、VEGFR阻害剤、EGFR阻害剤(エルロチニブ(タルセバ(Tarceva)(商標))、ゲフィチニブ(イレッサ(商標))等)が挙げられる。 The “cell growth factor” in the “drug that inhibits the action of the cell growth factor and its receptor” may be any substance that promotes cell growth, and usually has a molecular weight of 20,000 or less. In this peptide, a factor that exerts an action at a low concentration by binding to a receptor is used. Specifically, (1) EGF (epidermal growth factor) or a substance having substantially the same activity as that of [e.g. (2) Insulin or a substance having substantially the same activity (eg, insulin, IGF (insulin-like growth factor) -1, IGF-2, etc.), (3) FGF (fibroblast) growth factor) or a substance having substantially the same activity (eg, acid) FGF, basic FGF, KGF (keratinocyte growth factor), FGF-10, etc.], (4) other cell growth factors [eg, CSF (erythropoietin), EPO (erythropoietin), IL-2 (interleukin-2) , NGF (never grow growth factor), PDGF (platelet-derived growth factor), TGFβ (transforming growth factor β), HGF (hepatocyte growth factor), etc.
The “cell growth factor receptor” may be any receptor that has the ability to bind to the cell growth factor, and specifically includes an EGF receptor, a haregulin receptor (HER2). Insulin receptor, IGF receptor, FGF receptor-1 or FGF receptor-2, and the like.
Examples of the “drug that inhibits the action of cell growth factor” include HER2 antibody (such as trastuzumab (Herceptin (trademark))), imatinib mesylate, ZD1839 or EGFR antibody (cetuximab (Erbitux (trademark), etc.)) , Antibodies against VEGF (eg, bevacizumab (Avastin ™)), VEGFR antibody, VEGFR inhibitor, EGFR inhibitor (erlotinib (Tarceva ™), gefitinib (Iressa ™), etc.) Is mentioned.
該「細胞増殖因子の受容体」としては、前記の細胞増殖因子と結合能を有する受容体であればいかなるものであってもよく、具体的には、EGF受容体、ハレグリン受容体(HER2)、インシュリン受容体、IGF受容体、FGF受容体-1またはFGF受容体-2等が挙げられる。
該「細胞増殖因子の作用を阻害する薬剤」としては、HER2抗体(トラスツズマブ(ハーセプチン(商標))等)、メシル酸イマチニブ、ZD1839またはEGFR抗体(セツキシマブ(エルビタックス(Erbitux)(商標))等)、VEGFに対する抗体(例、ベバシツマブ(アバスチン(Avastin)(商標)))、VEGFR抗体、VEGFR阻害剤、EGFR阻害剤(エルロチニブ(タルセバ(Tarceva)(商標))、ゲフィチニブ(イレッサ(商標))等)が挙げられる。 The “cell growth factor” in the “drug that inhibits the action of the cell growth factor and its receptor” may be any substance that promotes cell growth, and usually has a molecular weight of 20,000 or less. In this peptide, a factor that exerts an action at a low concentration by binding to a receptor is used. Specifically, (1) EGF (epidermal growth factor) or a substance having substantially the same activity as that of [e.g. (2) Insulin or a substance having substantially the same activity (eg, insulin, IGF (insulin-like growth factor) -1, IGF-2, etc.), (3) FGF (fibroblast) growth factor) or a substance having substantially the same activity (eg, acid) FGF, basic FGF, KGF (keratinocyte growth factor), FGF-10, etc.], (4) other cell growth factors [eg, CSF (erythropoietin), EPO (erythropoietin), IL-2 (interleukin-2) , NGF (never grow growth factor), PDGF (platelet-derived growth factor), TGFβ (transforming growth factor β), HGF (hepatocyte growth factor), etc.
The “cell growth factor receptor” may be any receptor that has the ability to bind to the cell growth factor, and specifically includes an EGF receptor, a haregulin receptor (HER2). Insulin receptor, IGF receptor, FGF receptor-1 or FGF receptor-2, and the like.
Examples of the “drug that inhibits the action of cell growth factor” include HER2 antibody (such as trastuzumab (Herceptin (trademark))), imatinib mesylate, ZD1839 or EGFR antibody (cetuximab (Erbitux (trademark), etc.)) , Antibodies against VEGF (eg, bevacizumab (Avastin ™)), VEGFR antibody, VEGFR inhibitor, EGFR inhibitor (erlotinib (Tarceva ™), gefitinib (Iressa ™), etc.) Is mentioned.
前記の薬剤の他に、mTOR阻害剤(テムシロリムス、ラパマイシン、AZD8055、RAD001、CCI-779、Ap23573/MK8669等)、Akt阻害剤、PI3キナーゼ阻害剤(PI-103:
In addition to the above drugs, mTOR inhibitors (temsirolimus, rapamycin, AZD8055, RAD001, CCI-779, Ap23573 / MK8669, etc.), Akt inhibitors, PI3 kinase inhibitors (PI-103:
、国際公開WO2005/011686記載の化合物、SF-1126、XL-147、BGT226、GDC-0941等)、PI3キナーゼ/mTOR阻害剤(BEZ235、XL-765等)、c-Met阻害薬(PF-2341066:
, Compounds described in International Publication WO2005 / 011686, SF-1126, XL-147, BGT226, GDC-0941, etc.), PI3 kinase / mTOR inhibitors (BEZ235, XL-765, etc.), c-Met inhibitors (PF-2341066) :
、米国特許出願公開明細書US2004/0198750記載の化合物、米国特許出願公開明細書US2007/0197537記載の化合物、ARQ-197、JNJ-38877605、SGX-523等)、VEGFR/c-Met阻害剤(XL-880、XL-187、MGCD-265等)、VEGR(RET)/EGFR阻害剤(AZD6474等)、MEK阻害剤(PD0325901等)、L-アスパラギナーゼ、アセグラトン、塩酸プロカルバジン、プロトポルフィリン・コバルト錯塩、水銀ヘマトポルフィリン・ナトリウム、トポイソメラーゼI阻害薬(例、塩酸イリノテカン(トポテシン(商標)、カンプト(商標))、トポテカン等)、トポイソメラーゼII阻害薬(例えば、ソブゾキサン等)、分化誘導剤(例、レチノイド、ビタミンD類等)、血管新生阻害薬(例、サリドマイド、SU11248等)、α-ブロッカー(例、塩酸タムスロシン、ナフトピジル、ウラピジル、アルフゾシン、テラゾシン、プラゾシン、シロドシン等)、セリン・スレオニンキナーゼ阻害薬、エンドセリン受容体拮抗薬(例、アトラセンタン等)、プロテアゾーム阻害薬(例、ボルテゾミブ等)、Hsp90阻害薬(例、17-AAG等)、スピロノラクトン、ミノキシジル、11α-ヒドロキシプロゲステロン、骨吸収阻害・転移抑制薬(例、ゾレドロン酸、アレンドロン酸、パミドロン酸、エチドロン酸、イバンドロン酸、クロドロン酸)等も用いることができる。
, Compounds described in US Patent Application Publication US2004 / 0198750, compounds described in US Patent Application Publication US2007 / 0197537, ARQ-197, JNJ-38877605, SGX-523, etc.), VEGFR / c-Met inhibitor (XL -880, XL-187, MGCD-265, etc.), VEGR (RET) / EGFR inhibitor (AZD6474, etc.), MEK inhibitor (PD0325901, etc.), L-asparaginase, acegraton, procarbazine hydrochloride, protoporphyrin / cobalt complex, mercury Hematoporphyrin sodium, topoisomerase I inhibitor (eg, irinotecan hydrochloride (topotecin (trademark), campto (trademark)), topotecan, etc.), topoisomerase II inhibitor (eg, sobuzoxane, etc.), differentiation inducer (eg, retinoid, vitamin) D), angiogenesis inhibitors (eg, thalidomide, SU11248, etc.), α-blockers (eg, tamsulosin hydrochloride, naphthopidyl, urapidil, alfuzosin, terra) Syn, prazosin, silodosin, etc.), serine / threonine kinase inhibitors, endothelin receptor antagonists (eg, atrasentan, etc.), proteasome inhibitors (eg, bortezomib, etc.), Hsp90 inhibitors (eg, 17-AAG, etc.) Spironolactone, minoxidil, 11α-hydroxyprogesterone, bone resorption inhibitor / metastasis inhibitor (eg, zoledronic acid, alendronic acid, pamidronic acid, etidronic acid, ibandronic acid, clodronic acid) and the like can also be used.
前記した中でも、ホルモン療法剤または抗癌剤(以下、併用薬物と略記する)としては、ERダウンレギュレーター(例えばフルベストラント(ファスロデックス(商標))等)、HER2抗体(トラスツズマブ(ハーセプチン(商標))等)、EGFR抗体(セツキシマブ(エルビタックス(Erbitux)(商標)等)、EGFR阻害剤(エルロチニブ(タルセバ(Tarceva)(商標)、ゲフィチニブ(イレッサ(商標))等)、VEGFR阻害剤または化学療法剤(パクリタキセル(タキソール(商標)等)が好ましい。
特に、フルベストラント(ファスロデックス(商標))、トラスツズマブ(ハーセプチン(商標))、セツキシマブ(エルビタックス(Erbitux)(商標))、エルロチニブ(タルセバ(Tarceva)(商標))、ゲフィチニブ(イレッサ(商標))、パクリタキセル(タキソール(商標))等が好ましい。
また、塩酸ドキソルビシン(アドリアシン(商標))、塩酸イリノテカン(トポテシン(商標)、カンプト(商標))、5FU、ドセタキセルまたはメトトレキサートも好ましい例としてあげられる。 Among the above, hormone therapy agents or anticancer agents (hereinafter abbreviated as concomitant drugs) include ER down regulators (for example, fulvestrant (Faslodex ™)), HER2 antibodies (trastuzumab (Herceptin ™)) ), EGFR antibody (cetuximab (Erbitux (trademark) etc.), EGFR inhibitor (erlotinib (Tarceva (trademark), gefitinib (Iressa (trademark)) etc.), VEGFR inhibitor or chemotherapeutic agent (Paclitaxel (Taxol (trademark) etc.) is preferable.
In particular, fulvestrant (Faslodex ™), trastuzumab (Herceptin ™), cetuximab (Erbitux ™), erlotinib (Tarceva ™), gefitinib (Iressa ™) )), Paclitaxel (Taxol ™) and the like are preferred.
Also, preferable examples include doxorubicin hydrochloride (Adriacin (trademark)), irinotecan hydrochloride (topotecin (trademark), campto (trademark)), 5FU, docetaxel or methotrexate.
特に、フルベストラント(ファスロデックス(商標))、トラスツズマブ(ハーセプチン(商標))、セツキシマブ(エルビタックス(Erbitux)(商標))、エルロチニブ(タルセバ(Tarceva)(商標))、ゲフィチニブ(イレッサ(商標))、パクリタキセル(タキソール(商標))等が好ましい。
また、塩酸ドキソルビシン(アドリアシン(商標))、塩酸イリノテカン(トポテシン(商標)、カンプト(商標))、5FU、ドセタキセルまたはメトトレキサートも好ましい例としてあげられる。 Among the above, hormone therapy agents or anticancer agents (hereinafter abbreviated as concomitant drugs) include ER down regulators (for example, fulvestrant (Faslodex ™)), HER2 antibodies (trastuzumab (Herceptin ™)) ), EGFR antibody (cetuximab (Erbitux (trademark) etc.), EGFR inhibitor (erlotinib (Tarceva (trademark), gefitinib (Iressa (trademark)) etc.), VEGFR inhibitor or chemotherapeutic agent (Paclitaxel (Taxol (trademark) etc.) is preferable.
In particular, fulvestrant (Faslodex ™), trastuzumab (Herceptin ™), cetuximab (Erbitux ™), erlotinib (Tarceva ™), gefitinib (Iressa ™) )), Paclitaxel (Taxol ™) and the like are preferred.
Also, preferable examples include doxorubicin hydrochloride (Adriacin (trademark)), irinotecan hydrochloride (topotecin (trademark), campto (trademark)), 5FU, docetaxel or methotrexate.
本発明の併用剤は、2剤の併用に限定されず、3剤以上を併用してもよい。
The combination agent of the present invention is not limited to the combination of two agents, and three or more agents may be used in combination.
化合物(I)と併用薬物との併用に際しては、化合物(I)と併用薬物の投与時期は限定されず、化合物(I)と併用薬物とを、投与対象に対し、同時に投与してもよいし、時間差をおいて投与してもよい。併用薬物の投与量は、臨床上用いられている投与量に準ずればよく、投与対象、投与ルート、疾患、組み合わせ等により適宜選択することができる。
化合物(I)と併用薬物の投与形態は、特に限定されず、投与時に、化合物(I)と併用薬物とが組み合わされていればよい。このような投与形態としては、例えば、(1)化合物(I)と併用薬物とを同時に製剤化して得られる単一の製剤の投与、(2)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の同一投与経路での同時投与、(3)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の同一投与経路での時間差をおいての投与、(4)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の異なる投与経路での同時投与、(5)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の異なる投与経路での時間差をおいての投与(例えば、化合物(I)→併用薬物の順序での投与、あるいは逆の順序での投与)等が用いられる。 In the combined use of Compound (I) and a concomitant drug, the timing of administration of Compound (I) and the concomitant drug is not limited, and Compound (I) and the concomitant drug may be administered simultaneously to the administration subject. Alternatively, administration may be performed with a time difference. The dose of the concomitant drug may be determined according to the dose used clinically, and can be appropriately selected depending on the administration subject, administration route, disease, combination and the like.
The administration mode of compound (I) and the concomitant drug is not particularly limited, as long as compound (I) and the concomitant drug are combined at the time of administration. Examples of such administration forms include (1) administration of a single preparation obtained by simultaneously formulating compound (I) and a concomitant drug, and (2) separate preparation of compound (I) and concomitant drug. Simultaneous administration of the two preparations obtained by the same route by the same route of administration, (3) with a time difference in the same route of administration of the two formulations obtained by separately formulating the compound (I) and the concomitant drug (4) Simultaneous administration of two kinds of preparations obtained by separately formulating Compound (I) and a concomitant drug by different administration routes, (5) Preparation of Compound (I) and a concomitant drug separately Administration of the two types of preparations obtained by the preparation at different time intervals in different administration routes (for example, administration in the order of Compound (I) → concomitant drug or administration in the reverse order) and the like are used.
化合物(I)と併用薬物の投与形態は、特に限定されず、投与時に、化合物(I)と併用薬物とが組み合わされていればよい。このような投与形態としては、例えば、(1)化合物(I)と併用薬物とを同時に製剤化して得られる単一の製剤の投与、(2)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の同一投与経路での同時投与、(3)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の同一投与経路での時間差をおいての投与、(4)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の異なる投与経路での同時投与、(5)化合物(I)と併用薬物とを別々に製剤化して得られる2種の製剤の異なる投与経路での時間差をおいての投与(例えば、化合物(I)→併用薬物の順序での投与、あるいは逆の順序での投与)等が用いられる。 In the combined use of Compound (I) and a concomitant drug, the timing of administration of Compound (I) and the concomitant drug is not limited, and Compound (I) and the concomitant drug may be administered simultaneously to the administration subject. Alternatively, administration may be performed with a time difference. The dose of the concomitant drug may be determined according to the dose used clinically, and can be appropriately selected depending on the administration subject, administration route, disease, combination and the like.
The administration mode of compound (I) and the concomitant drug is not particularly limited, as long as compound (I) and the concomitant drug are combined at the time of administration. Examples of such administration forms include (1) administration of a single preparation obtained by simultaneously formulating compound (I) and a concomitant drug, and (2) separate preparation of compound (I) and concomitant drug. Simultaneous administration of the two preparations obtained by the same route by the same route of administration, (3) with a time difference in the same route of administration of the two formulations obtained by separately formulating the compound (I) and the concomitant drug (4) Simultaneous administration of two kinds of preparations obtained by separately formulating Compound (I) and a concomitant drug by different administration routes, (5) Preparation of Compound (I) and a concomitant drug separately Administration of the two types of preparations obtained by the preparation at different time intervals in different administration routes (for example, administration in the order of Compound (I) → concomitant drug or administration in the reverse order) and the like are used.
本発明の併用剤は、HER2および/またはEGFRキナーゼを発現している癌の増殖を抑制する治療剤として、また、ホルモン依存性癌およびホルモン依存性癌のホルモン非依存性癌への移行を防ぐ予防剤としても有用である。また、毒性(例えば、急性毒性、慢性毒性、遺伝毒性、生殖毒性、心毒性、薬物相互作用、癌原性など)が低く、さらに、水溶性が高く、安定性、体内動態(吸収性、分布、代謝、排泄など)、薬効発現の面でも優れているので、医薬として有用である。
即ち、本発明の併用剤は、種々の癌(なかでも乳癌(例えば、浸潤性乳管癌、非浸潤性乳管癌、炎症性乳癌など)、前立腺癌(例えば、ホルモン依存性前立腺癌、ホルモン非依存性前立腺癌など)、膵癌(例えば、膵管癌など)、胃癌(例えば、乳頭腺癌、粘液性腺癌、腺扁平上皮癌など)、肺癌(例えば、非小細胞肺癌、小細胞肺癌、悪性中皮腫など)、結腸癌(例えば、消化管間質腫瘍など)、直腸癌(例えば、消化管間質腫瘍など)、大腸癌(例えば、家族性大腸癌、遺伝性非ポリポーシス大腸癌、消化管間質腫瘍など)、小腸癌(例えば、非ホジキンリンパ腫、消化管間質腫瘍など)、食道癌、十二指腸癌、舌癌、咽頭癌(例えば、上咽頭癌、中咽頭癌、下咽頭癌など)、唾液腺癌、脳腫瘍(例えば、松果体星細胞腫瘍、毛様細胞性星細胞腫、びまん性星細胞腫、退形成性星細胞腫など)、神経鞘腫、肝臓癌(例えば、原発性肝癌、肝外胆管癌など)、腎臓癌(例えば、腎細胞癌、腎盂と尿管の移行上皮がんなど)、胆管癌、子宮内膜癌、子宮頸癌、卵巣癌(例えば、上皮性卵巣癌、性腺外胚細胞腫瘍、卵巣性胚細胞腫瘍、卵巣低悪性度腫瘍など)、膀胱癌、尿道がん、皮膚癌(例えば、眼内(眼)黒色腫、メルケル細胞がんなど)、血管腫、悪性リンパ腫、悪性黒色腫、甲状腺癌(例えば、甲状腺髄様癌など)、副甲状腺がん、鼻腔がん、副鼻腔がん、骨腫瘍(例えば、骨肉腫、ユーイング腫瘍、子宮肉腫、軟部組織肉腫など)、血管線維腫、網膜肉腫、陰茎癌、精巣腫瘍、小児固形癌(例えば、ウィルムス腫瘍、小児腎腫瘍など)、カポジ肉腫、AIDSに起因するカポジ肉腫、上顎洞腫瘍、線維性組織球腫、平滑筋肉腫、横紋筋肉腫、白血病(例えば、急性骨髄性白血病、急性リンパ芽球性白血病など)等)、アテローム性動脈硬化症、血管新生(例、固形癌および肉腫の成長にともなう血管新生、腫瘍の転移にともなう血管新生、および糖尿病性網膜症にともなう血管新生等)、ウイルス性疾患(HIV感染等)等の異常な細胞増殖による疾患に対する安全な予防または治療剤として用いることができる。
チロシンキナーゼ依存性疾患にはさらに、異常なチロシンキナーゼ酵素活性に関連する心臓血管疾患が含まれる。従って本発明の併用剤は、再狭窄のような心臓血管疾患に対する予防または治療剤として用いることもできる。
本発明の併用剤は、癌、特に乳癌、卵巣癌、前立腺癌、肺癌、膵癌、腎臓癌、大腸癌、小腸癌、食道癌および胃癌等の予防または治療のための抗癌剤として有用である。
本発明の併用剤は、毒性が低く、そのまま医薬として、または自体公知の薬学的に許容しうる担体等と混合して哺乳動物(例、ヒト、ウマ、ウシ、犬、猫、ラット、マウス、ウサギ、ブタ、サル等)に対して医薬組成物として用いることができる。
本発明の併用剤は、例えば、化合物(I)または(および)上記併用薬物を自体公知の方法に従って、薬理学的に許容される担体と混合して医薬組成物、例えば錠剤(糖衣錠、フィルムコーティング錠を含む)、散剤、顆粒剤、カプセル剤(ソフトカプセルを含む)、液剤、注射剤、坐剤、徐放剤等として、経口的又は非経口的(例、局所、直腸、静脈投与等)に安全に投与することができる。注射剤は、静脈内、筋肉内、皮下または臓器内投与あるいは直接病巣に投与することができる。
本発明の併用剤の製造に用いられてもよい薬理学的に許容される担体としては、製剤素材として慣用の各種有機あるいは無機担体物質が挙げられ、例えば固形製剤における賦形剤、滑沢剤、結合剤及び崩壊剤、あるいは液状製剤における溶剤、溶解補助剤、懸濁化剤、等張化剤、緩衝剤及び無痛化剤等が挙げられる。更に必要に応じ、通常の防腐剤、抗酸化剤、着色剤、甘味剤、吸着剤、湿潤剤等の添加物を適宜、適量用いることもできる。 The combination agent of the present invention is a therapeutic agent that suppresses the growth of cancers expressing HER2 and / or EGFR kinase, and prevents the transition of hormone-dependent and hormone-dependent cancers to hormone-independent cancers It is also useful as a preventive agent. In addition, it has low toxicity (eg acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.), high water solubility, stability, pharmacokinetics (absorbability, distribution) , Metabolism, excretion, etc.), and is also useful as a medicine because of its excellent medicinal effects.
That is, the concomitant drug of the present invention can be used for various cancers (among others, breast cancer (for example, invasive ductal cancer, non-invasive ductal cancer, inflammatory breast cancer, etc.), prostate cancer (for example, hormone-dependent prostate cancer, hormone). Independent prostate cancer, etc.), pancreatic cancer (eg, pancreatic duct cancer, etc.), stomach cancer (eg, papillary adenocarcinoma, mucinous adenocarcinoma, adenosquamous carcinoma, etc.), lung cancer (eg, non-small cell lung cancer, small cell lung cancer, malignant) Mesothelioma), colon cancer (eg, gastrointestinal stromal tumor), rectal cancer (eg, gastrointestinal stromal tumor), colorectal cancer (eg, familial colorectal cancer, hereditary nonpolyposis colorectal cancer, digestion) Ductal stromal tumors), small intestine cancer (eg, non-Hodgkin lymphoma, gastrointestinal stromal tumors), esophageal cancer, duodenal cancer, tongue cancer, pharyngeal cancer (eg, nasopharyngeal cancer, oropharyngeal cancer, hypopharyngeal cancer, etc.) ), Salivary gland cancer, brain tumor (eg pineal astrocyte tumor, ciliary cell) Cell tumor, diffuse astrocytoma, anaplastic astrocytoma), schwannoma, liver cancer (eg, primary liver cancer, extrahepatic cholangiocarcinoma), kidney cancer (eg, renal cell carcinoma, renal pelvis and urine) Transitional cell carcinoma of the duct), bile duct cancer, endometrial cancer, cervical cancer, ovarian cancer (eg epithelial ovarian cancer, extragonadal germ cell tumor, ovarian germ cell tumor, ovarian low grade tumor, etc.) , Bladder cancer, urethral cancer, skin cancer (eg, intraocular (eye) melanoma, Merkel cell carcinoma), hemangioma, malignant lymphoma, malignant melanoma, thyroid cancer (eg, medullary thyroid cancer), Parathyroid cancer, nasal cavity cancer, sinus cancer, bone tumor (eg, osteosarcoma, Ewing tumor, uterine sarcoma, soft tissue sarcoma, etc.), angiofibroma, retinal sarcoma, penile cancer, testicular cancer, childhood solid cancer (For example, Wilms tumor, childhood kidney tumor, etc.), Kaposi's sarcoma, Disarcoma, maxillary sinus tumor, fibrous histiocytoma, leiomyosarcoma, rhabdomyosarcoma, leukemia (eg, acute myeloid leukemia, acute lymphoblastic leukemia, etc.), atherosclerosis, angiogenesis Diseases caused by abnormal cell proliferation, such as angiogenesis associated with growth of solid tumors and sarcomas, angiogenesis associated with tumor metastasis, and angiogenesis associated with diabetic retinopathy, etc., viral diseases (such as HIV infection) It can be used as a safe preventive or therapeutic agent for.
Tyrosine kinase dependent diseases further include cardiovascular diseases associated with abnormal tyrosine kinase enzyme activity. Therefore, the combination agent of the present invention can also be used as a preventive or therapeutic agent for cardiovascular diseases such as restenosis.
The combination agent of the present invention is useful as an anticancer agent for prevention or treatment of cancer, particularly breast cancer, ovarian cancer, prostate cancer, lung cancer, pancreatic cancer, kidney cancer, colon cancer, small intestine cancer, esophageal cancer and gastric cancer.
The concomitant drug of the present invention has low toxicity and is used as a pharmaceutical as it is or mixed with a pharmaceutically acceptable carrier known per se, for example, to mammals (eg, human, horse, cow, dog, cat, rat, mouse, Rabbit, pig, monkey, etc.) can be used as a pharmaceutical composition.
The concomitant drug of the present invention is prepared by mixing a compound (I) or (and) the above concomitant drug with a pharmacologically acceptable carrier according to a method known per se, for example, a pharmaceutical composition such as a tablet (sugar-coated tablet, film coating). Including tablets), powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained-release agents, etc., orally or parenterally (eg, topical, rectal, intravenous administration, etc.) It can be safely administered. The injection can be administered intravenously, intramuscularly, subcutaneously, or into an organ, or directly to the lesion.
Examples of the pharmacologically acceptable carrier that may be used in the production of the concomitant drug of the present invention include various organic or inorganic carrier substances that are commonly used as pharmaceutical materials, such as excipients and lubricants in solid preparations. , Binders and disintegrants, solvents in liquid preparations, solubilizers, suspending agents, tonicity agents, buffers and soothing agents. If necessary, additives such as conventional preservatives, antioxidants, colorants, sweeteners, adsorbents, wetting agents and the like can be used in appropriate amounts.
即ち、本発明の併用剤は、種々の癌(なかでも乳癌(例えば、浸潤性乳管癌、非浸潤性乳管癌、炎症性乳癌など)、前立腺癌(例えば、ホルモン依存性前立腺癌、ホルモン非依存性前立腺癌など)、膵癌(例えば、膵管癌など)、胃癌(例えば、乳頭腺癌、粘液性腺癌、腺扁平上皮癌など)、肺癌(例えば、非小細胞肺癌、小細胞肺癌、悪性中皮腫など)、結腸癌(例えば、消化管間質腫瘍など)、直腸癌(例えば、消化管間質腫瘍など)、大腸癌(例えば、家族性大腸癌、遺伝性非ポリポーシス大腸癌、消化管間質腫瘍など)、小腸癌(例えば、非ホジキンリンパ腫、消化管間質腫瘍など)、食道癌、十二指腸癌、舌癌、咽頭癌(例えば、上咽頭癌、中咽頭癌、下咽頭癌など)、唾液腺癌、脳腫瘍(例えば、松果体星細胞腫瘍、毛様細胞性星細胞腫、びまん性星細胞腫、退形成性星細胞腫など)、神経鞘腫、肝臓癌(例えば、原発性肝癌、肝外胆管癌など)、腎臓癌(例えば、腎細胞癌、腎盂と尿管の移行上皮がんなど)、胆管癌、子宮内膜癌、子宮頸癌、卵巣癌(例えば、上皮性卵巣癌、性腺外胚細胞腫瘍、卵巣性胚細胞腫瘍、卵巣低悪性度腫瘍など)、膀胱癌、尿道がん、皮膚癌(例えば、眼内(眼)黒色腫、メルケル細胞がんなど)、血管腫、悪性リンパ腫、悪性黒色腫、甲状腺癌(例えば、甲状腺髄様癌など)、副甲状腺がん、鼻腔がん、副鼻腔がん、骨腫瘍(例えば、骨肉腫、ユーイング腫瘍、子宮肉腫、軟部組織肉腫など)、血管線維腫、網膜肉腫、陰茎癌、精巣腫瘍、小児固形癌(例えば、ウィルムス腫瘍、小児腎腫瘍など)、カポジ肉腫、AIDSに起因するカポジ肉腫、上顎洞腫瘍、線維性組織球腫、平滑筋肉腫、横紋筋肉腫、白血病(例えば、急性骨髄性白血病、急性リンパ芽球性白血病など)等)、アテローム性動脈硬化症、血管新生(例、固形癌および肉腫の成長にともなう血管新生、腫瘍の転移にともなう血管新生、および糖尿病性網膜症にともなう血管新生等)、ウイルス性疾患(HIV感染等)等の異常な細胞増殖による疾患に対する安全な予防または治療剤として用いることができる。
チロシンキナーゼ依存性疾患にはさらに、異常なチロシンキナーゼ酵素活性に関連する心臓血管疾患が含まれる。従って本発明の併用剤は、再狭窄のような心臓血管疾患に対する予防または治療剤として用いることもできる。
本発明の併用剤は、癌、特に乳癌、卵巣癌、前立腺癌、肺癌、膵癌、腎臓癌、大腸癌、小腸癌、食道癌および胃癌等の予防または治療のための抗癌剤として有用である。
本発明の併用剤は、毒性が低く、そのまま医薬として、または自体公知の薬学的に許容しうる担体等と混合して哺乳動物(例、ヒト、ウマ、ウシ、犬、猫、ラット、マウス、ウサギ、ブタ、サル等)に対して医薬組成物として用いることができる。
本発明の併用剤は、例えば、化合物(I)または(および)上記併用薬物を自体公知の方法に従って、薬理学的に許容される担体と混合して医薬組成物、例えば錠剤(糖衣錠、フィルムコーティング錠を含む)、散剤、顆粒剤、カプセル剤(ソフトカプセルを含む)、液剤、注射剤、坐剤、徐放剤等として、経口的又は非経口的(例、局所、直腸、静脈投与等)に安全に投与することができる。注射剤は、静脈内、筋肉内、皮下または臓器内投与あるいは直接病巣に投与することができる。
本発明の併用剤の製造に用いられてもよい薬理学的に許容される担体としては、製剤素材として慣用の各種有機あるいは無機担体物質が挙げられ、例えば固形製剤における賦形剤、滑沢剤、結合剤及び崩壊剤、あるいは液状製剤における溶剤、溶解補助剤、懸濁化剤、等張化剤、緩衝剤及び無痛化剤等が挙げられる。更に必要に応じ、通常の防腐剤、抗酸化剤、着色剤、甘味剤、吸着剤、湿潤剤等の添加物を適宜、適量用いることもできる。 The combination agent of the present invention is a therapeutic agent that suppresses the growth of cancers expressing HER2 and / or EGFR kinase, and prevents the transition of hormone-dependent and hormone-dependent cancers to hormone-independent cancers It is also useful as a preventive agent. In addition, it has low toxicity (eg acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.), high water solubility, stability, pharmacokinetics (absorbability, distribution) , Metabolism, excretion, etc.), and is also useful as a medicine because of its excellent medicinal effects.
That is, the concomitant drug of the present invention can be used for various cancers (among others, breast cancer (for example, invasive ductal cancer, non-invasive ductal cancer, inflammatory breast cancer, etc.), prostate cancer (for example, hormone-dependent prostate cancer, hormone). Independent prostate cancer, etc.), pancreatic cancer (eg, pancreatic duct cancer, etc.), stomach cancer (eg, papillary adenocarcinoma, mucinous adenocarcinoma, adenosquamous carcinoma, etc.), lung cancer (eg, non-small cell lung cancer, small cell lung cancer, malignant) Mesothelioma), colon cancer (eg, gastrointestinal stromal tumor), rectal cancer (eg, gastrointestinal stromal tumor), colorectal cancer (eg, familial colorectal cancer, hereditary nonpolyposis colorectal cancer, digestion) Ductal stromal tumors), small intestine cancer (eg, non-Hodgkin lymphoma, gastrointestinal stromal tumors), esophageal cancer, duodenal cancer, tongue cancer, pharyngeal cancer (eg, nasopharyngeal cancer, oropharyngeal cancer, hypopharyngeal cancer, etc.) ), Salivary gland cancer, brain tumor (eg pineal astrocyte tumor, ciliary cell) Cell tumor, diffuse astrocytoma, anaplastic astrocytoma), schwannoma, liver cancer (eg, primary liver cancer, extrahepatic cholangiocarcinoma), kidney cancer (eg, renal cell carcinoma, renal pelvis and urine) Transitional cell carcinoma of the duct), bile duct cancer, endometrial cancer, cervical cancer, ovarian cancer (eg epithelial ovarian cancer, extragonadal germ cell tumor, ovarian germ cell tumor, ovarian low grade tumor, etc.) , Bladder cancer, urethral cancer, skin cancer (eg, intraocular (eye) melanoma, Merkel cell carcinoma), hemangioma, malignant lymphoma, malignant melanoma, thyroid cancer (eg, medullary thyroid cancer), Parathyroid cancer, nasal cavity cancer, sinus cancer, bone tumor (eg, osteosarcoma, Ewing tumor, uterine sarcoma, soft tissue sarcoma, etc.), angiofibroma, retinal sarcoma, penile cancer, testicular cancer, childhood solid cancer (For example, Wilms tumor, childhood kidney tumor, etc.), Kaposi's sarcoma, Disarcoma, maxillary sinus tumor, fibrous histiocytoma, leiomyosarcoma, rhabdomyosarcoma, leukemia (eg, acute myeloid leukemia, acute lymphoblastic leukemia, etc.), atherosclerosis, angiogenesis Diseases caused by abnormal cell proliferation, such as angiogenesis associated with growth of solid tumors and sarcomas, angiogenesis associated with tumor metastasis, and angiogenesis associated with diabetic retinopathy, etc., viral diseases (such as HIV infection) It can be used as a safe preventive or therapeutic agent for.
Tyrosine kinase dependent diseases further include cardiovascular diseases associated with abnormal tyrosine kinase enzyme activity. Therefore, the combination agent of the present invention can also be used as a preventive or therapeutic agent for cardiovascular diseases such as restenosis.
The combination agent of the present invention is useful as an anticancer agent for prevention or treatment of cancer, particularly breast cancer, ovarian cancer, prostate cancer, lung cancer, pancreatic cancer, kidney cancer, colon cancer, small intestine cancer, esophageal cancer and gastric cancer.
The concomitant drug of the present invention has low toxicity and is used as a pharmaceutical as it is or mixed with a pharmaceutically acceptable carrier known per se, for example, to mammals (eg, human, horse, cow, dog, cat, rat, mouse, Rabbit, pig, monkey, etc.) can be used as a pharmaceutical composition.
The concomitant drug of the present invention is prepared by mixing a compound (I) or (and) the above concomitant drug with a pharmacologically acceptable carrier according to a method known per se, for example, a pharmaceutical composition such as a tablet (sugar-coated tablet, film coating). Including tablets), powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained-release agents, etc., orally or parenterally (eg, topical, rectal, intravenous administration, etc.) It can be safely administered. The injection can be administered intravenously, intramuscularly, subcutaneously, or into an organ, or directly to the lesion.
Examples of the pharmacologically acceptable carrier that may be used in the production of the concomitant drug of the present invention include various organic or inorganic carrier substances that are commonly used as pharmaceutical materials, such as excipients and lubricants in solid preparations. , Binders and disintegrants, solvents in liquid preparations, solubilizers, suspending agents, tonicity agents, buffers and soothing agents. If necessary, additives such as conventional preservatives, antioxidants, colorants, sweeteners, adsorbents, wetting agents and the like can be used in appropriate amounts.
賦形剤としては、例えば乳糖、白糖、D-マンニトール、デンプン、コーンスターチ、結晶セルロース、軽質無水ケイ酸等が挙げられる。
滑沢剤としては、例えばステアリン酸マグネシウム、ステアリン酸カルシウム、タルク、コロイドシリカ等が挙げられる。
結合剤としては、例えば結晶セルロース、白糖、D-マンニトール、デキストリン、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、ポリビニルピロリドン、デンプン、ショ糖、ゼラチン、メチルセルロース、カルボキシメチルセルロースナトリウム等が挙げられる。
崩壊剤としては、例えばデンプン、カルボキシメチルセルロース、カルボキシメチルセルロースカルシウム、カルボキシメチルスターチナトリウム、L-ヒドロキシプロピルセルロース等が挙げられる。
溶剤としては、例えば注射用水、アルコール、プロピレングリコール、マクロゴール、ゴマ油、トウモロコシ油、オリーブ油等が挙げられる。
溶解補助剤としては、例えばポリエチレングリコール、プロピレングリコール、D-マンニトール、安息香酸ベンジル、エタノール、トリスアミノメタン、コレステロール、トリエタノールアミン、炭酸ナトリウム、クエン酸ナトリウム等が挙げられる。
懸濁化剤としては、例えばステアリルトリエタノールアミン、ラウリル硫酸ナトリウム、ラウリルアミノプロピオン酸、レシチン、塩化ベンザルコニウム、塩化ベンゼトニウム、モノステアリン酸グリセリン等の界面活性剤;例えばポリビニルアルコール、ポリビニルピロリドン、カルボキシメチルセルロースナトリウム、メチルセルロース、ヒドロキシメチルセルロース、ヒドロキシエチルセルロース、ヒドロキシプロピルセルロース等の親水性高分子等が挙げられる。
等張化剤としては、例えばブドウ糖、D-ソルビトール、塩化ナトリウム、グリセリン、D-マンニトール等が挙げられる。
緩衝剤としては、例えばリン酸塩、酢酸塩、炭酸塩、クエン酸塩等の緩衝液等が挙げられる。
無痛化剤としては、例えばベンジルアルコール等が挙げられる。
防腐剤としては、例えばパラオキシ安息香酸エステル類、クロロブタノール、ベンジルアルコール、フェネチルアルコール、デヒドロ酢酸、ソルビン酸等が挙げられる。
抗酸化剤としては、例えば亜硫酸塩、アスコルビン酸、α-トコフェロール等が挙げられる。 Examples of the excipient include lactose, sucrose, D-mannitol, starch, corn starch, crystalline cellulose, light anhydrous silicic acid and the like.
Examples of the lubricant include magnesium stearate, calcium stearate, talc, colloidal silica and the like.
Examples of the binder include crystalline cellulose, sucrose, D-mannitol, dextrin, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, starch, sucrose, gelatin, methylcellulose, sodium carboxymethylcellulose and the like.
Examples of the disintegrant include starch, carboxymethyl cellulose, carboxymethyl cellulose calcium, carboxymethyl starch sodium, L-hydroxypropyl cellulose, and the like.
Examples of the solvent include water for injection, alcohol, propylene glycol, macrogol, sesame oil, corn oil, olive oil and the like.
Examples of the solubilizer include polyethylene glycol, propylene glycol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate and the like.
Examples of the suspending agent include surfactants such as stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glyceryl monostearate; for example, polyvinyl alcohol, polyvinylpyrrolidone, carboxy Examples include hydrophilic polymers such as sodium methylcellulose, methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, and hydroxypropylcellulose.
Examples of the isotonic agent include glucose, D-sorbitol, sodium chloride, glycerin, D-mannitol and the like.
Examples of the buffer include buffer solutions of phosphate, acetate, carbonate, citrate and the like.
Examples of soothing agents include benzyl alcohol.
Examples of the preservative include p-hydroxybenzoates, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid and the like.
Examples of the antioxidant include sulfite, ascorbic acid, α-tocopherol and the like.
滑沢剤としては、例えばステアリン酸マグネシウム、ステアリン酸カルシウム、タルク、コロイドシリカ等が挙げられる。
結合剤としては、例えば結晶セルロース、白糖、D-マンニトール、デキストリン、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、ポリビニルピロリドン、デンプン、ショ糖、ゼラチン、メチルセルロース、カルボキシメチルセルロースナトリウム等が挙げられる。
崩壊剤としては、例えばデンプン、カルボキシメチルセルロース、カルボキシメチルセルロースカルシウム、カルボキシメチルスターチナトリウム、L-ヒドロキシプロピルセルロース等が挙げられる。
溶剤としては、例えば注射用水、アルコール、プロピレングリコール、マクロゴール、ゴマ油、トウモロコシ油、オリーブ油等が挙げられる。
溶解補助剤としては、例えばポリエチレングリコール、プロピレングリコール、D-マンニトール、安息香酸ベンジル、エタノール、トリスアミノメタン、コレステロール、トリエタノールアミン、炭酸ナトリウム、クエン酸ナトリウム等が挙げられる。
懸濁化剤としては、例えばステアリルトリエタノールアミン、ラウリル硫酸ナトリウム、ラウリルアミノプロピオン酸、レシチン、塩化ベンザルコニウム、塩化ベンゼトニウム、モノステアリン酸グリセリン等の界面活性剤;例えばポリビニルアルコール、ポリビニルピロリドン、カルボキシメチルセルロースナトリウム、メチルセルロース、ヒドロキシメチルセルロース、ヒドロキシエチルセルロース、ヒドロキシプロピルセルロース等の親水性高分子等が挙げられる。
等張化剤としては、例えばブドウ糖、D-ソルビトール、塩化ナトリウム、グリセリン、D-マンニトール等が挙げられる。
緩衝剤としては、例えばリン酸塩、酢酸塩、炭酸塩、クエン酸塩等の緩衝液等が挙げられる。
無痛化剤としては、例えばベンジルアルコール等が挙げられる。
防腐剤としては、例えばパラオキシ安息香酸エステル類、クロロブタノール、ベンジルアルコール、フェネチルアルコール、デヒドロ酢酸、ソルビン酸等が挙げられる。
抗酸化剤としては、例えば亜硫酸塩、アスコルビン酸、α-トコフェロール等が挙げられる。 Examples of the excipient include lactose, sucrose, D-mannitol, starch, corn starch, crystalline cellulose, light anhydrous silicic acid and the like.
Examples of the lubricant include magnesium stearate, calcium stearate, talc, colloidal silica and the like.
Examples of the binder include crystalline cellulose, sucrose, D-mannitol, dextrin, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, starch, sucrose, gelatin, methylcellulose, sodium carboxymethylcellulose and the like.
Examples of the disintegrant include starch, carboxymethyl cellulose, carboxymethyl cellulose calcium, carboxymethyl starch sodium, L-hydroxypropyl cellulose, and the like.
Examples of the solvent include water for injection, alcohol, propylene glycol, macrogol, sesame oil, corn oil, olive oil and the like.
Examples of the solubilizer include polyethylene glycol, propylene glycol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, sodium citrate and the like.
Examples of the suspending agent include surfactants such as stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, benzethonium chloride, and glyceryl monostearate; for example, polyvinyl alcohol, polyvinylpyrrolidone, carboxy Examples include hydrophilic polymers such as sodium methylcellulose, methylcellulose, hydroxymethylcellulose, hydroxyethylcellulose, and hydroxypropylcellulose.
Examples of the isotonic agent include glucose, D-sorbitol, sodium chloride, glycerin, D-mannitol and the like.
Examples of the buffer include buffer solutions of phosphate, acetate, carbonate, citrate and the like.
Examples of soothing agents include benzyl alcohol.
Examples of the preservative include p-hydroxybenzoates, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid and the like.
Examples of the antioxidant include sulfite, ascorbic acid, α-tocopherol and the like.
本発明の併用剤における化合物(I)と併用薬物との配合比は、投与対象、投与ルート、疾患等により適宜選択することができる。
例えば、本発明の併用剤における化合物(I)の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01ないし100重量%、好ましくは約0.1ないし50重量%、さらに好ましくは約0.5ないし20重量%程度である。
本発明の併用剤における併用薬物の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01ないし100重量%、好ましくは約0.1ないし50重量%、さらに好ましくは約0.5ないし20重量%程度である。
本発明の併用剤における担体等の添加剤の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約1ないし99.99重量%、好ましくは約10ないし90重量%程度である。
また、化合物(I)および併用薬物をそれぞれ別々に製剤化する場合も同様の含有量でよい。 The compounding ratio of the compound (I) and the concomitant drug in the combination drug of the present invention can be appropriately selected depending on the administration subject, administration route, disease and the like.
For example, the content of compound (I) in the combination agent of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50% by weight, based on the whole preparation. More preferably, it is about 0.5 to 20% by weight.
The content of the concomitant drug in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50% by weight, more preferably about the whole preparation About 0.5 to 20% by weight.
The content of additives such as carriers in the combination agent of the present invention varies depending on the form of the preparation, but is usually about 1 to 99.99% by weight, preferably about 10 to 90% by weight, based on the whole preparation. .
The same content may be used when compound (I) and the concomitant drug are formulated separately.
例えば、本発明の併用剤における化合物(I)の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01ないし100重量%、好ましくは約0.1ないし50重量%、さらに好ましくは約0.5ないし20重量%程度である。
本発明の併用剤における併用薬物の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01ないし100重量%、好ましくは約0.1ないし50重量%、さらに好ましくは約0.5ないし20重量%程度である。
本発明の併用剤における担体等の添加剤の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約1ないし99.99重量%、好ましくは約10ないし90重量%程度である。
また、化合物(I)および併用薬物をそれぞれ別々に製剤化する場合も同様の含有量でよい。 The compounding ratio of the compound (I) and the concomitant drug in the combination drug of the present invention can be appropriately selected depending on the administration subject, administration route, disease and the like.
For example, the content of compound (I) in the combination agent of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50% by weight, based on the whole preparation. More preferably, it is about 0.5 to 20% by weight.
The content of the concomitant drug in the concomitant drug of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50% by weight, more preferably about the whole preparation About 0.5 to 20% by weight.
The content of additives such as carriers in the combination agent of the present invention varies depending on the form of the preparation, but is usually about 1 to 99.99% by weight, preferably about 10 to 90% by weight, based on the whole preparation. .
The same content may be used when compound (I) and the concomitant drug are formulated separately.
これらの製剤は、製剤工程において通常一般に用いられる自体公知の方法により製造することができる。
例えば、化合物(I)または併用薬物は、分散剤(例、ツイーン(Tween)80(アトラスパウダー社製、米国)、HCO 60(日光ケミカルズ製)、ポリエチレングリコール、カルボキシメチルセルロース、アルギン酸ナトリウム、ヒドロキシプロピルメチルセルロース、デキストリンなど)、安定化剤(例、アスコルビン酸、ピロ亜硫酸ナトリウム等)、界面活性剤(例、ポリソルベート80、マクロゴール等)、可溶化剤(例、グリセリン、エタノール等)、緩衝剤(例、リン酸及びそのアルカリ金属塩、クエン酸及びそのアルカリ金属塩等)、等張化剤(例、塩化ナトリウム、塩化カリウム、マンニトール、ソルビトール、ブドウ糖等)、pH調節剤(例、塩酸、水酸化ナトリウム等)、保存剤(例、パラオキシ安息香酸エチル、安息香酸、パラオキシ安息香酸メチル、パラオキシ安息香酸プロピル、ベンジルアルコール等)、溶解剤(例、濃グリセリン、メグルミン等)、溶解補助剤(例、プロピレングリコール、白糖等)、無痛化剤(例、ブドウ糖、ベンジルアルコール等)などと共に水性注射剤に、あるいはオリーブ油、ゴマ油、綿実油、コーン油などの植物油、プロピレングリコールなどの溶解補助剤に溶解、懸濁あるいは乳化して油性注射剤に成形し、注射剤とすることができる。 These preparations can be produced by a method known per se generally used in the preparation process.
For example, the compound (I) or the concomitant drug is a dispersant (eg, Tween 80 (manufactured by Atlas Powder, USA), HCO 60 (manufactured by Nikko Chemicals), polyethylene glycol, carboxymethylcellulose, sodium alginate, hydroxypropylmethylcellulose. , Dextrin, etc.), stabilizers (eg, ascorbic acid, sodium pyrosulfite, etc.), surfactants (eg, polysorbate 80, macrogol, etc.), solubilizers (eg, glycerin, ethanol, etc.), buffers (eg, , Phosphoric acid and its alkali metal salts, citric acid and its alkali metal salts, etc.), isotonic agents (eg, sodium chloride, potassium chloride, mannitol, sorbitol, glucose etc.), pH regulators (eg, hydrochloric acid, hydroxylated) Sodium), preservatives (eg, ethyl paraoxybenzoate, benzoic acid, para Methyl xybenzoate, propyl paraoxybenzoate, benzyl alcohol, etc.), solubilizers (eg, concentrated glycerin, meglumine, etc.), solubilizers (eg, propylene glycol, sucrose, etc.), soothing agents (eg, glucose, benzyl alcohol) Etc.) in aqueous injections, or in olive oil, sesame oil, cottonseed oil, corn oil and other vegetable oils, or in solubilizing agents such as propylene glycol, and then molded into oily injections to make injections Can do.
例えば、化合物(I)または併用薬物は、分散剤(例、ツイーン(Tween)80(アトラスパウダー社製、米国)、HCO 60(日光ケミカルズ製)、ポリエチレングリコール、カルボキシメチルセルロース、アルギン酸ナトリウム、ヒドロキシプロピルメチルセルロース、デキストリンなど)、安定化剤(例、アスコルビン酸、ピロ亜硫酸ナトリウム等)、界面活性剤(例、ポリソルベート80、マクロゴール等)、可溶化剤(例、グリセリン、エタノール等)、緩衝剤(例、リン酸及びそのアルカリ金属塩、クエン酸及びそのアルカリ金属塩等)、等張化剤(例、塩化ナトリウム、塩化カリウム、マンニトール、ソルビトール、ブドウ糖等)、pH調節剤(例、塩酸、水酸化ナトリウム等)、保存剤(例、パラオキシ安息香酸エチル、安息香酸、パラオキシ安息香酸メチル、パラオキシ安息香酸プロピル、ベンジルアルコール等)、溶解剤(例、濃グリセリン、メグルミン等)、溶解補助剤(例、プロピレングリコール、白糖等)、無痛化剤(例、ブドウ糖、ベンジルアルコール等)などと共に水性注射剤に、あるいはオリーブ油、ゴマ油、綿実油、コーン油などの植物油、プロピレングリコールなどの溶解補助剤に溶解、懸濁あるいは乳化して油性注射剤に成形し、注射剤とすることができる。 These preparations can be produced by a method known per se generally used in the preparation process.
For example, the compound (I) or the concomitant drug is a dispersant (eg, Tween 80 (manufactured by Atlas Powder, USA), HCO 60 (manufactured by Nikko Chemicals), polyethylene glycol, carboxymethylcellulose, sodium alginate, hydroxypropylmethylcellulose. , Dextrin, etc.), stabilizers (eg, ascorbic acid, sodium pyrosulfite, etc.), surfactants (eg, polysorbate 80, macrogol, etc.), solubilizers (eg, glycerin, ethanol, etc.), buffers (eg, , Phosphoric acid and its alkali metal salts, citric acid and its alkali metal salts, etc.), isotonic agents (eg, sodium chloride, potassium chloride, mannitol, sorbitol, glucose etc.), pH regulators (eg, hydrochloric acid, hydroxylated) Sodium), preservatives (eg, ethyl paraoxybenzoate, benzoic acid, para Methyl xybenzoate, propyl paraoxybenzoate, benzyl alcohol, etc.), solubilizers (eg, concentrated glycerin, meglumine, etc.), solubilizers (eg, propylene glycol, sucrose, etc.), soothing agents (eg, glucose, benzyl alcohol) Etc.) in aqueous injections, or in olive oil, sesame oil, cottonseed oil, corn oil and other vegetable oils, or in solubilizing agents such as propylene glycol, and then molded into oily injections to make injections Can do.
経口投与用製剤とするには、自体公知の方法に従い、化合物(I)または併用薬物を例えば、賦形剤(例、乳糖、白糖、デンプン、コーンスターチなど)、崩壊剤(例、デンプン、炭酸カルシウムなど)、結合剤(例、デンプン、アラビアゴム、カルボキシメチルセルロース、ポリビニルピロリドン、ヒドロキシプロピルセルロース、ゼラチンなど)又は滑沢剤(例、タルク、ステアリン酸マグネシウム、ポリエチレングリコール 6000など)などを添加して圧縮成形し、次いで必要により、味のマスキング、腸溶性あるいは持続性の目的のため自体公知の方法でコーティングすることにより経口投与用製剤とすることができる。そのコーティング剤としては、例えば、ヒドロキシプロピルメチルセルロース、エチルセルロース、ヒドロキシメチルセルロース、ヒドロキシプロピルセルロース、ポリオキシエチレングリコール、ツイーン 80、プルロニック F68、セルロースアセテートフタレート、ヒドロキシプロピルメチルセルロースフタレート、ヒドロキシメチルセルロースアセテートサクシネート、オイドラギット(ローム社製、ドイツ、メタアクリル酸・アクリル酸共重合)および色素(例、ベンガラ、二酸化チタン等)などが用いられる。糖衣コーティングとしては、例えば、蔗糖、タルク、アラビアゴム、色素(例、ベンガラ、二酸化チタン等)、艶出し剤(例、ミツロウ等)、などが用いられる。経口投与用製剤は速放性製剤、徐放性製剤のいずれであってもよい。
例えば、坐剤とするには、自体公知の方法に従い、化合物(I)または併用薬物を油性又は水性の固体状、半固体状あるいは液状の坐剤とすることができる。上記組成物に用いる油性基剤としては、例えば、高級脂肪酸のグリセリド〔例、カカオ脂、ウイテプゾル類(ダイナマイトノーベル社製、ドイツ)など〕、中級脂肪酸〔例、ミグリオール類(ダイナマイトノーベル社製、ドイツ)など〕、あるいは植物油(例、ゴマ油、大豆油、綿実油など)などが挙げられる。また、水性基剤としては、例えばポリエチレングリコール類、プロピレングリコール、水性ゲル基剤としては、例えば天然ガム類、セルロース誘導体、ビニル重合体、アクリル酸重合体などが挙げられる。
上記徐放性製剤としては、徐放性マイクロカプセル剤などが挙げられる。
徐放型マイクロカプセルとするには、自体公知の方法を採用できる。
化合物(I)は、固形製剤(例、散剤、顆粒剤、錠剤、カプセル剤)などの経口投与用製剤に成型するか、坐剤などの直腸投与用製剤に成型するのが好ましい。特に経口投与用製剤が好ましい。
併用薬物は、薬物の種類に応じて上記した剤形とすることができる。 In order to obtain a preparation for oral administration, according to a method known per se, compound (I) or a concomitant drug is used, for example, an excipient (eg, lactose, sucrose, starch, corn starch, etc.), a disintegrant (eg, starch, calcium carbonate). Etc.), binder (eg, starch, gum arabic, carboxymethylcellulose, polyvinylpyrrolidone, hydroxypropylcellulose, gelatin, etc.) or lubricant (eg, talc, magnesium stearate, polyethylene glycol 6000, etc.) Then, if necessary, it can be coated by a method known per se for the purpose of taste masking, enteric property or persistence, to obtain a preparation for oral administration. Examples of the coating agent include hydroxypropylmethylcellulose, ethylcellulose, hydroxymethylcellulose, hydroxypropylcellulose, polyoxyethylene glycol, Tween 80, Pluronic F68, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxymethylcellulose acetate succinate, Eudragit (Rohm) (Made in Germany, methacrylic acid / acrylic acid copolymer) and dyes (eg, bengara, titanium dioxide, etc.) are used. Examples of the sugar coating include sucrose, talc, gum arabic, pigments (eg, bengara, titanium dioxide, etc.), polishes (eg, beeswax, etc.), and the like. The preparation for oral administration may be either an immediate release preparation or a sustained release preparation.
For example, to make a suppository, the compound (I) or the concomitant drug can be converted into an oily or aqueous solid, semisolid or liquid suppository according to a method known per se. Examples of the oily base used in the above composition include glycerides of higher fatty acids (eg, cacao butter, witepsols (manufactured by Dynamite Nobel, Germany)), intermediate fatty acids (eg, miglyols (manufactured by Dynamite Nobel, Germany) Etc.], or vegetable oils (eg, sesame oil, soybean oil, cottonseed oil, etc.). Examples of the aqueous base include polyethylene glycols and propylene glycol. Examples of the aqueous gel base include natural gums, cellulose derivatives, vinyl polymers, and acrylic acid polymers.
Examples of the sustained-release preparation include sustained-release microcapsules.
In order to obtain a sustained-release type microcapsule, a method known per se can be employed.
Compound (I) is preferably molded into a preparation for oral administration such as a solid preparation (eg, powder, granule, tablet, capsule) or into a preparation for rectal administration such as a suppository. Particularly preferred are preparations for oral administration.
The concomitant drug can be in the above-mentioned dosage form depending on the type of drug.
例えば、坐剤とするには、自体公知の方法に従い、化合物(I)または併用薬物を油性又は水性の固体状、半固体状あるいは液状の坐剤とすることができる。上記組成物に用いる油性基剤としては、例えば、高級脂肪酸のグリセリド〔例、カカオ脂、ウイテプゾル類(ダイナマイトノーベル社製、ドイツ)など〕、中級脂肪酸〔例、ミグリオール類(ダイナマイトノーベル社製、ドイツ)など〕、あるいは植物油(例、ゴマ油、大豆油、綿実油など)などが挙げられる。また、水性基剤としては、例えばポリエチレングリコール類、プロピレングリコール、水性ゲル基剤としては、例えば天然ガム類、セルロース誘導体、ビニル重合体、アクリル酸重合体などが挙げられる。
上記徐放性製剤としては、徐放性マイクロカプセル剤などが挙げられる。
徐放型マイクロカプセルとするには、自体公知の方法を採用できる。
化合物(I)は、固形製剤(例、散剤、顆粒剤、錠剤、カプセル剤)などの経口投与用製剤に成型するか、坐剤などの直腸投与用製剤に成型するのが好ましい。特に経口投与用製剤が好ましい。
併用薬物は、薬物の種類に応じて上記した剤形とすることができる。 In order to obtain a preparation for oral administration, according to a method known per se, compound (I) or a concomitant drug is used, for example, an excipient (eg, lactose, sucrose, starch, corn starch, etc.), a disintegrant (eg, starch, calcium carbonate). Etc.), binder (eg, starch, gum arabic, carboxymethylcellulose, polyvinylpyrrolidone, hydroxypropylcellulose, gelatin, etc.) or lubricant (eg, talc, magnesium stearate, polyethylene glycol 6000, etc.) Then, if necessary, it can be coated by a method known per se for the purpose of taste masking, enteric property or persistence, to obtain a preparation for oral administration. Examples of the coating agent include hydroxypropylmethylcellulose, ethylcellulose, hydroxymethylcellulose, hydroxypropylcellulose, polyoxyethylene glycol, Tween 80, Pluronic F68, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxymethylcellulose acetate succinate, Eudragit (Rohm) (Made in Germany, methacrylic acid / acrylic acid copolymer) and dyes (eg, bengara, titanium dioxide, etc.) are used. Examples of the sugar coating include sucrose, talc, gum arabic, pigments (eg, bengara, titanium dioxide, etc.), polishes (eg, beeswax, etc.), and the like. The preparation for oral administration may be either an immediate release preparation or a sustained release preparation.
For example, to make a suppository, the compound (I) or the concomitant drug can be converted into an oily or aqueous solid, semisolid or liquid suppository according to a method known per se. Examples of the oily base used in the above composition include glycerides of higher fatty acids (eg, cacao butter, witepsols (manufactured by Dynamite Nobel, Germany)), intermediate fatty acids (eg, miglyols (manufactured by Dynamite Nobel, Germany) Etc.], or vegetable oils (eg, sesame oil, soybean oil, cottonseed oil, etc.). Examples of the aqueous base include polyethylene glycols and propylene glycol. Examples of the aqueous gel base include natural gums, cellulose derivatives, vinyl polymers, and acrylic acid polymers.
Examples of the sustained-release preparation include sustained-release microcapsules.
In order to obtain a sustained-release type microcapsule, a method known per se can be employed.
Compound (I) is preferably molded into a preparation for oral administration such as a solid preparation (eg, powder, granule, tablet, capsule) or into a preparation for rectal administration such as a suppository. Particularly preferred are preparations for oral administration.
The concomitant drug can be in the above-mentioned dosage form depending on the type of drug.
本発明の併用剤の投与量は、化合物(I)の種類、年齢、体重、症状、剤形、投与方法、投与期間などにより異なるが、例えば、患者(成人、体重約60kg)一人あたり、通常、化合物(I)および併用薬物として、それぞれ経口投与の場合、約0.1mg~約500mgの範囲、好ましくは約1mg~約100mgの範囲、非経口投与の場合、約0.01mg~約100mgの範囲、さらに好ましくは約0.1mg~約10mgの範囲から選択できる。これらの投薬量は、1日1~3回に分けて投与できる。もちろん、前記したように投与量は種々の条件で変動するので、前記投与量より少ない量で十分な場合もあり、また範囲を超えて投与する必要のある場合もある。
併用薬物は、副作用が問題とならない範囲でどのような量を設定することも可能である。併用薬物としての一日投与量は、症状の程度、投与対象の年齢、性別、体重、感受性差、投与の時期、間隔、医薬製剤の性質、調剤、種類、有効成分の種類などによって異なり、特に限定されないが、薬物の量として通常、たとえば経口投与で哺乳動物1kg体重あたり約0.001~2000mg、好ましくは約0.01~500mg、さらに好ましくは、約0.1~100mgの範囲、非経口投与の場合、約0.0001mg~約400mgの範囲、さらに好ましくは約0.001mg~約200mgの範囲から選択できる。これらの投薬量は、これを通常1日1~3回に分けて投与する。 The dose of the concomitant drug of the present invention varies depending on the type of compound (I), age, body weight, symptom, dosage form, administration method, administration period, etc., for example, usually per patient (adult, body weight about 60 kg) per person The compound (I) and the concomitant drug are each in the range of about 0.1 mg to about 500 mg, preferably in the range of about 1 mg to about 100 mg for oral administration, and about 0.01 mg to about 100 mg for parenteral administration. A range can be selected, more preferably from about 0.1 mg to about 10 mg. These dosages can be administered in 1 to 3 divided doses per day. Of course, as described above, the dosage varies depending on various conditions. Therefore, a dosage smaller than the dosage may be sufficient, or the dosage may need to be administered beyond the range.
The amount of the concomitant drug can be set as long as side effects do not become a problem. The daily dose as a concomitant drug varies depending on the degree of symptoms, age of the subject, sex, weight, sensitivity difference, timing of administration, interval, nature of the pharmaceutical preparation, formulation, type, type of active ingredient, etc. Although not limited, the amount of the drug is usually in the range of about 0.001 to 2000 mg, preferably about 0.01 to 500 mg, more preferably about 0.1 to 100 mg parenterally per kg body weight of the mammal, for example, by oral administration. For administration, it can be selected from the range of about 0.0001 mg to about 400 mg, more preferably from the range of about 0.001 mg to about 200 mg. These dosages are usually administered in 1 to 3 divided doses per day.
併用薬物は、副作用が問題とならない範囲でどのような量を設定することも可能である。併用薬物としての一日投与量は、症状の程度、投与対象の年齢、性別、体重、感受性差、投与の時期、間隔、医薬製剤の性質、調剤、種類、有効成分の種類などによって異なり、特に限定されないが、薬物の量として通常、たとえば経口投与で哺乳動物1kg体重あたり約0.001~2000mg、好ましくは約0.01~500mg、さらに好ましくは、約0.1~100mgの範囲、非経口投与の場合、約0.0001mg~約400mgの範囲、さらに好ましくは約0.001mg~約200mgの範囲から選択できる。これらの投薬量は、これを通常1日1~3回に分けて投与する。 The dose of the concomitant drug of the present invention varies depending on the type of compound (I), age, body weight, symptom, dosage form, administration method, administration period, etc., for example, usually per patient (adult, body weight about 60 kg) per person The compound (I) and the concomitant drug are each in the range of about 0.1 mg to about 500 mg, preferably in the range of about 1 mg to about 100 mg for oral administration, and about 0.01 mg to about 100 mg for parenteral administration. A range can be selected, more preferably from about 0.1 mg to about 10 mg. These dosages can be administered in 1 to 3 divided doses per day. Of course, as described above, the dosage varies depending on various conditions. Therefore, a dosage smaller than the dosage may be sufficient, or the dosage may need to be administered beyond the range.
The amount of the concomitant drug can be set as long as side effects do not become a problem. The daily dose as a concomitant drug varies depending on the degree of symptoms, age of the subject, sex, weight, sensitivity difference, timing of administration, interval, nature of the pharmaceutical preparation, formulation, type, type of active ingredient, etc. Although not limited, the amount of the drug is usually in the range of about 0.001 to 2000 mg, preferably about 0.01 to 500 mg, more preferably about 0.1 to 100 mg parenterally per kg body weight of the mammal, for example, by oral administration. For administration, it can be selected from the range of about 0.0001 mg to about 400 mg, more preferably from the range of about 0.001 mg to about 200 mg. These dosages are usually administered in 1 to 3 divided doses per day.
本発明の併用剤を投与するに際しては、同時期に投与してもよいが、併用薬物を先に投与した後、化合物(I)を投与してもよいし、化合物(I)を先に投与し、その後で併用薬物を投与してもよい。時間差をおいて投与する場合、時間差は投与する有効成分、剤形、投与方法により異なるが、例えば、併用薬物を先に投与する場合、併用薬物を投与した後1分~3日以内、好ましくは10分~1日以内、より好ましくは15分~1時間以内に化合物(I)を投与する方法が挙げられる。化合物(I)を先に投与する場合、化合物(I)を投与した後、1分~1日以内、好ましくは10分~6時間以内、より好ましくは15分から1時間以内に併用薬物を投与する方法が挙げられる。
好ましい投与方法としては、例えば、非経口投与用製剤に製形された併用薬物約0.001~200mg/kg体重を静脈注射または筋肉注射し、約15分後に経口投与用製剤に製形された化合物(I)約0.005~100mg/kg体重を1日量として経口投与する。 When administering the concomitant drug of the present invention, it may be administered at the same time, but after administering the concomitant drug first, compound (I) may be administered, or compound (I) is administered first. Thereafter, the concomitant drug may be administered. When administered at a time difference, the time difference varies depending on the active ingredient, dosage form, and administration method to be administered. For example, when administering the concomitant drug first, within 1 minute to 3 days after administering the concomitant drug, preferably The method includes administering Compound (I) within 10 minutes to 1 day, more preferably within 15 minutes to 1 hour. When compound (I) is administered first, the concomitant drug is administered within 1 minute to 1 day after administration of compound (I), preferably within 10 minutes to 6 hours, more preferably within 15 minutes to 1 hour. A method is mentioned.
As a preferable administration method, for example, about 0.001 to 200 mg / kg body weight of a concomitant drug formed into a preparation for parenteral administration was injected intravenously or intramuscularly, and after about 15 minutes, it was formed into a preparation for oral administration. Compound (I) is orally administered at a daily dose of about 0.005 to 100 mg / kg body weight.
好ましい投与方法としては、例えば、非経口投与用製剤に製形された併用薬物約0.001~200mg/kg体重を静脈注射または筋肉注射し、約15分後に経口投与用製剤に製形された化合物(I)約0.005~100mg/kg体重を1日量として経口投与する。 When administering the concomitant drug of the present invention, it may be administered at the same time, but after administering the concomitant drug first, compound (I) may be administered, or compound (I) is administered first. Thereafter, the concomitant drug may be administered. When administered at a time difference, the time difference varies depending on the active ingredient, dosage form, and administration method to be administered. For example, when administering the concomitant drug first, within 1 minute to 3 days after administering the concomitant drug, preferably The method includes administering Compound (I) within 10 minutes to 1 day, more preferably within 15 minutes to 1 hour. When compound (I) is administered first, the concomitant drug is administered within 1 minute to 1 day after administration of compound (I), preferably within 10 minutes to 6 hours, more preferably within 15 minutes to 1 hour. A method is mentioned.
As a preferable administration method, for example, about 0.001 to 200 mg / kg body weight of a concomitant drug formed into a preparation for parenteral administration was injected intravenously or intramuscularly, and after about 15 minutes, it was formed into a preparation for oral administration. Compound (I) is orally administered at a daily dose of about 0.005 to 100 mg / kg body weight.
また、本発明の化合物(I)は、サイミジン合成酵素産生阻害作用を有するため、単剤の有効成分としても、癌の増殖を抑制する治療剤として、また、ホルモン依存性癌およびホルモン依存性癌のホルモン非依存性癌への移行を防ぐ予防剤として有用に用いられる。本発明の化合物(I)は、毒性(例えば、急性毒性、慢性毒性、遺伝毒性、生殖毒性、心毒性、薬物相互作用、癌原性など)が低く、さらに、水溶性が高く、安定性、体内動態(吸収性、分布、代謝、排泄など)、薬効発現の面でも優れているので、医薬として有用である。
即ち、本発明の化合物(I)は、単剤の有効成分としても、種々の癌(なかでも乳癌(例えば、浸潤性乳管癌、非浸潤性乳管癌、炎症性乳癌など)、前立腺癌(例えば、ホルモン依存性前立腺癌、ホルモン非依存性前立腺癌など)、膵癌(例えば、膵管癌など)、胃癌(例えば、乳頭腺癌、粘液性腺癌、腺扁平上皮癌など)、肺癌(例えば、非小細胞肺癌、小細胞肺癌、悪性中皮腫など)、結腸癌(例えば、消化管間質腫瘍など)、直腸癌(例えば、消化管間質腫瘍など)、大腸癌(例えば、家族性大腸癌、遺伝性非ポリポーシス大腸癌、消化管間質腫瘍など)、小腸癌(例えば、非ホジキンリンパ腫、消化管間質腫瘍など)、食道癌、十二指腸癌、舌癌、咽頭癌(例えば、上咽頭癌、中咽頭癌、下咽頭癌など)、唾液腺癌、脳腫瘍(例えば、松果体星細胞腫瘍、毛様細胞性星細胞腫、びまん性星細胞腫、退形成性星細胞腫など)、神経鞘腫、肝臓癌(例えば、原発性肝癌、肝外胆管癌など)、腎臓癌(例えば、腎細胞癌、腎盂と尿管の移行上皮がんなど)、胆管癌、子宮内膜癌、子宮頸癌、卵巣癌(例えば、上皮性卵巣癌、性腺外胚細胞腫瘍、卵巣性胚細胞腫瘍、卵巣低悪性度腫瘍など)、膀胱癌、尿道がん、皮膚癌(例えば、眼内(眼)黒色腫、メルケル細胞がんなど)、血管腫、悪性リンパ腫、悪性黒色腫、甲状腺癌(例えば、甲状腺髄様癌など)、副甲状腺がん、鼻腔がん、副鼻腔がん、骨腫瘍(例えば、骨肉腫、ユーイング腫瘍、子宮肉腫、軟部組織肉腫など)、血管線維腫、網膜肉腫、陰茎癌、精巣腫瘍、小児固形癌(例えば、ウィルムス腫瘍、小児腎腫瘍など)、カポジ肉腫、AIDSに起因するカポジ肉腫、上顎洞腫瘍、線維性組織球腫、平滑筋肉腫、横紋筋肉腫、白血病(例えば、急性骨髄性白血病、急性リンパ芽球性白血病など)等)、アテローム性動脈硬化症、血管新生(例、固形癌および肉腫の成長にともなう血管新生、腫瘍の転移にともなう血管新生、および糖尿病性網膜症にともなう血管新生等)、ウイルス性疾患(HIV感染等)等の異常な細胞増殖による疾患に対する安全な予防または治療剤として用いることができる。 In addition, since the compound (I) of the present invention has a thymidine synthase production inhibitory action, it can be used as a single agent active ingredient, as a therapeutic agent that suppresses the growth of cancer, and for hormone-dependent and hormone-dependent cancers. It is useful as a prophylactic agent that prevents the transition to hormone-independent cancer. The compound (I) of the present invention has low toxicity (for example, acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.), high water solubility, stability, It is also useful as a medicine because it is excellent in pharmacokinetics (absorbability, distribution, metabolism, excretion, etc.) and in terms of drug efficacy.
That is, the compound (I) of the present invention can be used as an active ingredient of a single agent as well as various cancers (especially breast cancer (for example, invasive breast cancer, non-invasive breast cancer, inflammatory breast cancer, etc.), prostate cancer) (Eg, hormone-dependent prostate cancer, hormone-independent prostate cancer, etc.), pancreatic cancer (eg, pancreatic duct cancer, etc.), gastric cancer (eg, papillary adenocarcinoma, mucinous adenocarcinoma, adenosquamous carcinoma, etc.), lung cancer (eg, Non-small cell lung cancer, small cell lung cancer, malignant mesothelioma, etc., colon cancer (eg, gastrointestinal stromal tumor), rectal cancer (eg, gastrointestinal stromal tumor), colon cancer (eg, familial colon) Cancer, hereditary non-polyposis colorectal cancer, gastrointestinal stromal tumor, etc.), small intestine cancer (eg, non-Hodgkin lymphoma, gastrointestinal stromal tumor, etc.), esophageal cancer, duodenal cancer, tongue cancer, pharyngeal cancer (eg, nasopharynx) Cancer, oropharyngeal cancer, hypopharyngeal cancer, etc.), salivary gland cancer, brain tumor (eg Pineal astrocytoma, ciliary astrocytoma, diffuse astrocytoma, anaplastic astrocytoma), schwannomas, liver cancer (eg primary liver cancer, extrahepatic cholangiocarcinoma, etc.), Kidney cancer (eg renal cell carcinoma, transitional cell carcinoma of the renal pelvis and ureter), bile duct cancer, endometrial cancer, cervical cancer, ovarian cancer (eg epithelial ovarian cancer, extragonadal germ cell tumor, ovary Germ cell tumor, ovarian low grade tumor, etc.), bladder cancer, urethral cancer, skin cancer (eg, intraocular (eye) melanoma, Merkel cell carcinoma, etc.), hemangioma, malignant lymphoma, malignant melanoma, Thyroid cancer (eg, medullary thyroid cancer), parathyroid cancer, nasal cavity cancer, sinus cancer, bone tumor (eg, osteosarcoma, Ewing tumor, uterine sarcoma, soft tissue sarcoma, etc.), angiofibroma, Retinal sarcoma, penile cancer, testicular tumor, pediatric solid cancer (eg Wilms tumor, pediatric kidney tumor), Disarcoma, Kaposi sarcoma caused by AIDS, maxillary sinus tumor, fibrous histiocytoma, leiomyosarcoma, rhabdomyosarcoma, leukemia (eg, acute myeloid leukemia, acute lymphoblastic leukemia, etc.), atheroma Atherosclerosis, angiogenesis (eg, angiogenesis with growth of solid tumors and sarcomas, angiogenesis with tumor metastasis, angiogenesis with diabetic retinopathy, etc.), viral diseases (HIV infection, etc.), etc. It can be used as a safe preventive or therapeutic agent for diseases caused by abnormal cell proliferation.
即ち、本発明の化合物(I)は、単剤の有効成分としても、種々の癌(なかでも乳癌(例えば、浸潤性乳管癌、非浸潤性乳管癌、炎症性乳癌など)、前立腺癌(例えば、ホルモン依存性前立腺癌、ホルモン非依存性前立腺癌など)、膵癌(例えば、膵管癌など)、胃癌(例えば、乳頭腺癌、粘液性腺癌、腺扁平上皮癌など)、肺癌(例えば、非小細胞肺癌、小細胞肺癌、悪性中皮腫など)、結腸癌(例えば、消化管間質腫瘍など)、直腸癌(例えば、消化管間質腫瘍など)、大腸癌(例えば、家族性大腸癌、遺伝性非ポリポーシス大腸癌、消化管間質腫瘍など)、小腸癌(例えば、非ホジキンリンパ腫、消化管間質腫瘍など)、食道癌、十二指腸癌、舌癌、咽頭癌(例えば、上咽頭癌、中咽頭癌、下咽頭癌など)、唾液腺癌、脳腫瘍(例えば、松果体星細胞腫瘍、毛様細胞性星細胞腫、びまん性星細胞腫、退形成性星細胞腫など)、神経鞘腫、肝臓癌(例えば、原発性肝癌、肝外胆管癌など)、腎臓癌(例えば、腎細胞癌、腎盂と尿管の移行上皮がんなど)、胆管癌、子宮内膜癌、子宮頸癌、卵巣癌(例えば、上皮性卵巣癌、性腺外胚細胞腫瘍、卵巣性胚細胞腫瘍、卵巣低悪性度腫瘍など)、膀胱癌、尿道がん、皮膚癌(例えば、眼内(眼)黒色腫、メルケル細胞がんなど)、血管腫、悪性リンパ腫、悪性黒色腫、甲状腺癌(例えば、甲状腺髄様癌など)、副甲状腺がん、鼻腔がん、副鼻腔がん、骨腫瘍(例えば、骨肉腫、ユーイング腫瘍、子宮肉腫、軟部組織肉腫など)、血管線維腫、網膜肉腫、陰茎癌、精巣腫瘍、小児固形癌(例えば、ウィルムス腫瘍、小児腎腫瘍など)、カポジ肉腫、AIDSに起因するカポジ肉腫、上顎洞腫瘍、線維性組織球腫、平滑筋肉腫、横紋筋肉腫、白血病(例えば、急性骨髄性白血病、急性リンパ芽球性白血病など)等)、アテローム性動脈硬化症、血管新生(例、固形癌および肉腫の成長にともなう血管新生、腫瘍の転移にともなう血管新生、および糖尿病性網膜症にともなう血管新生等)、ウイルス性疾患(HIV感染等)等の異常な細胞増殖による疾患に対する安全な予防または治療剤として用いることができる。 In addition, since the compound (I) of the present invention has a thymidine synthase production inhibitory action, it can be used as a single agent active ingredient, as a therapeutic agent that suppresses the growth of cancer, and for hormone-dependent and hormone-dependent cancers. It is useful as a prophylactic agent that prevents the transition to hormone-independent cancer. The compound (I) of the present invention has low toxicity (for example, acute toxicity, chronic toxicity, genotoxicity, reproductive toxicity, cardiotoxicity, drug interaction, carcinogenicity, etc.), high water solubility, stability, It is also useful as a medicine because it is excellent in pharmacokinetics (absorbability, distribution, metabolism, excretion, etc.) and in terms of drug efficacy.
That is, the compound (I) of the present invention can be used as an active ingredient of a single agent as well as various cancers (especially breast cancer (for example, invasive breast cancer, non-invasive breast cancer, inflammatory breast cancer, etc.), prostate cancer) (Eg, hormone-dependent prostate cancer, hormone-independent prostate cancer, etc.), pancreatic cancer (eg, pancreatic duct cancer, etc.), gastric cancer (eg, papillary adenocarcinoma, mucinous adenocarcinoma, adenosquamous carcinoma, etc.), lung cancer (eg, Non-small cell lung cancer, small cell lung cancer, malignant mesothelioma, etc., colon cancer (eg, gastrointestinal stromal tumor), rectal cancer (eg, gastrointestinal stromal tumor), colon cancer (eg, familial colon) Cancer, hereditary non-polyposis colorectal cancer, gastrointestinal stromal tumor, etc.), small intestine cancer (eg, non-Hodgkin lymphoma, gastrointestinal stromal tumor, etc.), esophageal cancer, duodenal cancer, tongue cancer, pharyngeal cancer (eg, nasopharynx) Cancer, oropharyngeal cancer, hypopharyngeal cancer, etc.), salivary gland cancer, brain tumor (eg Pineal astrocytoma, ciliary astrocytoma, diffuse astrocytoma, anaplastic astrocytoma), schwannomas, liver cancer (eg primary liver cancer, extrahepatic cholangiocarcinoma, etc.), Kidney cancer (eg renal cell carcinoma, transitional cell carcinoma of the renal pelvis and ureter), bile duct cancer, endometrial cancer, cervical cancer, ovarian cancer (eg epithelial ovarian cancer, extragonadal germ cell tumor, ovary Germ cell tumor, ovarian low grade tumor, etc.), bladder cancer, urethral cancer, skin cancer (eg, intraocular (eye) melanoma, Merkel cell carcinoma, etc.), hemangioma, malignant lymphoma, malignant melanoma, Thyroid cancer (eg, medullary thyroid cancer), parathyroid cancer, nasal cavity cancer, sinus cancer, bone tumor (eg, osteosarcoma, Ewing tumor, uterine sarcoma, soft tissue sarcoma, etc.), angiofibroma, Retinal sarcoma, penile cancer, testicular tumor, pediatric solid cancer (eg Wilms tumor, pediatric kidney tumor), Disarcoma, Kaposi sarcoma caused by AIDS, maxillary sinus tumor, fibrous histiocytoma, leiomyosarcoma, rhabdomyosarcoma, leukemia (eg, acute myeloid leukemia, acute lymphoblastic leukemia, etc.), atheroma Atherosclerosis, angiogenesis (eg, angiogenesis with growth of solid tumors and sarcomas, angiogenesis with tumor metastasis, angiogenesis with diabetic retinopathy, etc.), viral diseases (HIV infection, etc.), etc. It can be used as a safe preventive or therapeutic agent for diseases caused by abnormal cell proliferation.
本発明の化合物(I)を含有するサイミジン合成酵素産生阻害剤は、毒性が低く、そのまま医薬として、または自体公知の薬学的に許容しうる担体等と混合して哺乳動物(例、ヒト、ウマ、ウシ、犬、猫、ラット、マウス、ウサギ、ブタ、サル等)に対して医薬組成物として用いることができる。
本発明のサイミジン合成酵素産生阻害剤は、例えば、化合物(I)を自体公知の方法に従って、薬理学的に許容される担体と混合して医薬組成物、例えば錠剤(糖衣錠、フィルムコーティング錠を含む)、散剤、顆粒剤、カプセル剤(ソフトカプセルを含む)、液剤、注射剤、坐剤、徐放剤等として、経口的又は非経口的(例、局所、直腸、静脈投与等)に安全に投与することができる。注射剤は、静脈内、筋肉内、皮下または臓器内投与あるいは直接病巣に投与することができる。
本発明のサイミジン合成酵素産生阻害剤の製造に用いられてもよい薬理学的に許容される担体としては、前記本発明の併用剤の製造に用いられてもよい薬理学的に許容される担体と同様のものが挙げられる。 The thymidine synthase production inhibitor containing the compound (I) of the present invention has low toxicity, and is used as it is as a medicine or mixed with a pharmaceutically acceptable carrier known per se as a mammal (eg, human, horse, etc.). Bovine, dog, cat, rat, mouse, rabbit, pig, monkey, etc.).
The thymidine synthase production inhibitor of the present invention is prepared, for example, by mixing compound (I) with a pharmacologically acceptable carrier according to a method known per se, including pharmaceutical compositions such as tablets (including sugar-coated tablets and film-coated tablets). ), Powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained release, etc., orally or parenterally (eg, topical, rectal, intravenous administration, etc.) can do. The injection can be administered intravenously, intramuscularly, subcutaneously, or into an organ, or directly to the lesion.
Examples of the pharmacologically acceptable carrier that may be used for the production of the thymidine synthase production inhibitor of the present invention include the pharmacologically acceptable carrier that may be used for the production of the combination agent of the present invention. The same thing is mentioned.
本発明のサイミジン合成酵素産生阻害剤は、例えば、化合物(I)を自体公知の方法に従って、薬理学的に許容される担体と混合して医薬組成物、例えば錠剤(糖衣錠、フィルムコーティング錠を含む)、散剤、顆粒剤、カプセル剤(ソフトカプセルを含む)、液剤、注射剤、坐剤、徐放剤等として、経口的又は非経口的(例、局所、直腸、静脈投与等)に安全に投与することができる。注射剤は、静脈内、筋肉内、皮下または臓器内投与あるいは直接病巣に投与することができる。
本発明のサイミジン合成酵素産生阻害剤の製造に用いられてもよい薬理学的に許容される担体としては、前記本発明の併用剤の製造に用いられてもよい薬理学的に許容される担体と同様のものが挙げられる。 The thymidine synthase production inhibitor containing the compound (I) of the present invention has low toxicity, and is used as it is as a medicine or mixed with a pharmaceutically acceptable carrier known per se as a mammal (eg, human, horse, etc.). Bovine, dog, cat, rat, mouse, rabbit, pig, monkey, etc.).
The thymidine synthase production inhibitor of the present invention is prepared, for example, by mixing compound (I) with a pharmacologically acceptable carrier according to a method known per se, including pharmaceutical compositions such as tablets (including sugar-coated tablets and film-coated tablets). ), Powders, granules, capsules (including soft capsules), liquids, injections, suppositories, sustained release, etc., orally or parenterally (eg, topical, rectal, intravenous administration, etc.) can do. The injection can be administered intravenously, intramuscularly, subcutaneously, or into an organ, or directly to the lesion.
Examples of the pharmacologically acceptable carrier that may be used for the production of the thymidine synthase production inhibitor of the present invention include the pharmacologically acceptable carrier that may be used for the production of the combination agent of the present invention. The same thing is mentioned.
本発明のサイミジン合成酵素産生阻害剤における化合物(I)の含有量は、製剤の形態によって相違するが、通常製剤全体に対して約0.01ないし100重量%、好ましくは約0.1ないし50重量%、さらに好ましくは約0.5ないし20重量%程度である。
The content of compound (I) in the thymidine synthase production inhibitor of the present invention varies depending on the form of the preparation, but is usually about 0.01 to 100% by weight, preferably about 0.1 to 50%, based on the whole preparation. % By weight, more preferably about 0.5 to 20% by weight.
これらの製剤は、製剤工程において通常一般に用いられる自体公知の方法により製造することができ、例えば、本発明の併用剤について前記したものと同様の方法が挙げられる。
化合物(I)は、固形製剤(例、散剤、顆粒剤、錠剤、カプセル剤)などの経口投与用製剤に成型するか、坐剤などの直腸投与用製剤に成型するのが好ましい。特に経口投与用製剤が好ましい。 These preparations can be produced by a method known per se generally used in the preparation process, and examples thereof include the same methods as those described above for the concomitant drug of the present invention.
Compound (I) is preferably molded into a preparation for oral administration such as a solid preparation (eg, powder, granule, tablet, capsule) or into a preparation for rectal administration such as a suppository. Particularly preferred are preparations for oral administration.
化合物(I)は、固形製剤(例、散剤、顆粒剤、錠剤、カプセル剤)などの経口投与用製剤に成型するか、坐剤などの直腸投与用製剤に成型するのが好ましい。特に経口投与用製剤が好ましい。 These preparations can be produced by a method known per se generally used in the preparation process, and examples thereof include the same methods as those described above for the concomitant drug of the present invention.
Compound (I) is preferably molded into a preparation for oral administration such as a solid preparation (eg, powder, granule, tablet, capsule) or into a preparation for rectal administration such as a suppository. Particularly preferred are preparations for oral administration.
本発明のサイミジン合成酵素産生阻害剤の投与量は、化合物(I)の種類、年齢、体重、症状、剤形、投与方法、投与期間などにより異なるが、例えば、患者(成人、体重約60kg)一人あたり、通常、化合物(I)として、経口投与の場合、約0.1mg~約500mgの範囲、好ましくは約1mg~約100mgの範囲、非経口投与の場合、約0.01mg~約100mgの範囲、さらに好ましくは約0.1mg~約10mgの範囲から選択できる。これらの投薬量は、1日1~3回に分けて投与できる。もちろん、前記したように投与量は種々の条件で変動するので、前記投与量より少ない量で十分な場合もあり、また範囲を超えて投与する必要のある場合もある。
The dosage of the thymidine synthase production inhibitor of the present invention varies depending on the type, age, weight, symptom, dosage form, administration method, administration period, etc. of compound (I). Per person, usually as compound (I), in the range of about 0.1 mg to about 500 mg, preferably in the range of about 1 mg to about 100 mg for oral administration, and about 0.01 mg to about 100 mg for parenteral administration. A range can be selected, more preferably from about 0.1 mg to about 10 mg. These dosages can be administered in 1 to 3 divided doses per day. Of course, as described above, the dosage varies depending on various conditions. Therefore, a dosage smaller than the dosage may be sufficient, or the dosage may need to be administered beyond the range.
以下の実施例および試験例で本発明の製造法および使用法を説明するが、本発明はこれらに限定されるものではない。特許請求の範囲で定義される発明の思想および範囲内に入る他の態様も本発明に含まれる。なお、実施例および試験例中の化合物Aとは、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミド p-トルエンスルホン酸塩を意味する。
The production method and use method of the present invention will be described in the following examples and test examples, but the present invention is not limited thereto. Other aspects that fall within the spirit and scope of the invention as defined by the claims are also encompassed by the invention. The compound A in Examples and Test Examples is N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d]. ] Pyrimidin-5-yl) ethyl] -2-methyl-2- (methylsulfonyl) propanamide means p-toluenesulfonate.
実施例1
(1)化合物A 8.0g
(2)乳糖 60.0g
(3)コーンスターチ 35.0g
(4)ゼラチン 3.0g
(5)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 1
(1) Compound A 8.0g
(2) Lactose 60.0g
(3) Cornstarch 35.0g
(4) Gelatin 3.0g
(5) Magnesium stearate 2.0 g
After granulating a mixture of Compound A (8.0 g), lactose (60.0 g) and corn starch (35.0 g) with a 10% by weight aqueous gelatin solution (30 mL, 3.0 g as gelatin) through a 1 mm mesh sieve. , Dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
(1)化合物A 8.0g
(2)乳糖 60.0g
(3)コーンスターチ 35.0g
(4)ゼラチン 3.0g
(5)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 1
(1) Compound A 8.0g
(2) Lactose 60.0g
(3) Cornstarch 35.0g
(4) Gelatin 3.0g
(5) Magnesium stearate 2.0 g
After granulating a mixture of Compound A (8.0 g), lactose (60.0 g) and corn starch (35.0 g) with a 10% by weight aqueous gelatin solution (30 mL, 3.0 g as gelatin) through a 1 mm mesh sieve. , Dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
実施例2
日局注射用蒸留水50mLにトラスツズマブ50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 2
After dissolving 50 mg of trastuzumab in 50 mL of JP distilled water for injection, JP distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
日局注射用蒸留水50mLにトラスツズマブ50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 2
After dissolving 50 mg of trastuzumab in 50 mL of JP distilled water for injection, JP distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
実施例3
日局注射用蒸留水50mLにセツキシマブ50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 3
After dissolving 50 mg of cetuximab in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
日局注射用蒸留水50mLにセツキシマブ50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 3
After dissolving 50 mg of cetuximab in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
実施例4
(1)化合物A 8.0g
(2)エルロチニブ 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、エルロチニブ(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 4
(1) Compound A 8.0g
(2) Erlotinib 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), erlotinib (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
(1)化合物A 8.0g
(2)エルロチニブ 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、エルロチニブ(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 4
(1) Compound A 8.0g
(2) Erlotinib 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), erlotinib (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
実施例5
(1)化合物A 8.0g
(2)ゲフィチニブ 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、ゲフィチニブ(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 5
(1) Compound A 8.0g
(2) Gefitinib 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), gefitinib (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
(1)化合物A 8.0g
(2)ゲフィチニブ 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、ゲフィチニブ(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 5
(1) Compound A 8.0g
(2) Gefitinib 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), gefitinib (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
実施例6
日局注射用蒸留水50mLにパクリタキセル50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 6
Dissolve 50 mg of paclitaxel in 50 mL of JP injection distilled water, and then add JP injection distilled water to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
日局注射用蒸留水50mLにパクリタキセル50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 6
Dissolve 50 mg of paclitaxel in 50 mL of JP injection distilled water, and then add JP injection distilled water to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
実施例7
(1)化合物A 8.0g
(2)フルベストラント 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、フルベストラント(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 7
(1) Compound A 8.0g
(2) Fulvestrant 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
A mixture of Compound A (8.0 g), fulvestrant (8.0 g), lactose (60.0 g) and corn starch (35.0 g) was used with a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), After granulating through a 1 mm mesh sieve, it is dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
(1)化合物A 8.0g
(2)フルベストラント 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、フルベストラント(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 7
(1) Compound A 8.0g
(2) Fulvestrant 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
A mixture of Compound A (8.0 g), fulvestrant (8.0 g), lactose (60.0 g) and corn starch (35.0 g) was used with a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), After granulating through a 1 mm mesh sieve, it is dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
実施例8
日局注射用蒸留水50mLに塩酸ドキソルビシン50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 8
After dissolving 50 mg of doxorubicin hydrochloride in 50 mL of distilled water for injection, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
日局注射用蒸留水50mLに塩酸ドキソルビシン50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 8
After dissolving 50 mg of doxorubicin hydrochloride in 50 mL of distilled water for injection, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
実施例9
日局注射用蒸留水50mLに塩酸イリノテカン50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 9
After dissolving 50 mg of irinotecan hydrochloride in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
日局注射用蒸留水50mLに塩酸イリノテカン50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 9
After dissolving 50 mg of irinotecan hydrochloride in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
実施例10
(1)化合物A 8.0g
(2)5FU 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、5FU(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 10
(1) Compound A 8.0g
(2) 5FU 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), 5FU (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10% by weight gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
(1)化合物A 8.0g
(2)5FU 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、5FU(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 10
(1) Compound A 8.0g
(2) 5FU 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), 5FU (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10% by weight gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
実施例11
日局注射用蒸留水50mLにドセタキセル50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 11
After dissolving 50 mg of docetaxel in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
日局注射用蒸留水50mLにドセタキセル50mgを溶解した後、日局注射蒸留水を加えて100mLとする。この溶液を滅菌条件下でろ過し、次にこの溶液1mLずつを取り、滅菌条件下、注射用バイアルに充填し、凍結乾燥して密閉する。 Example 11
After dissolving 50 mg of docetaxel in 50 mL of JP injection distilled water, JP injection distilled water is added to make 100 mL. The solution is filtered under sterile conditions, then 1 mL of this solution is taken and filled into injection vials under sterile conditions, lyophilized and sealed.
実施例12
(1)化合物A 8.0g
(2)メトトレキサート 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、メトトレキサート(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 12
(1) Compound A 8.0g
(2) Methotrexate 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), methotrexate (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
(1)化合物A 8.0g
(2)メトトレキサート 8.0g
(3)乳糖 60.0g
(4)コーンスターチ 35.0g
(5)ゼラチン 3.0g
(6)ステアリン酸マグネシウム 2.0g
化合物A(8.0g)、メトトレキサート(8.0g)、乳糖(60.0g)及びコーンスターチ(35.0g)の混合物を10重量%ゼラチン水溶液(30mL、ゼラチンとして3.0g)を用い、1mmメッシュの篩を通して顆粒化した後、40℃で乾燥し再び篩過する。得られる顆粒をステアリン酸マグネシウム(2.0g)と混合し、圧縮する。得られる中心錠を、蔗糖、二酸化チタン、タルク及びアラビアゴムの水懸濁液による糖衣でコーティングする。コーティングが施された錠剤をミツロウで艶出して1000錠のコート錠を得る。 Example 12
(1) Compound A 8.0g
(2) Methotrexate 8.0g
(3) Lactose 60.0g
(4) Corn starch 35.0g
(5) Gelatin 3.0g
(6) Magnesium stearate 2.0 g
Using a mixture of Compound A (8.0 g), methotrexate (8.0 g), lactose (60.0 g) and corn starch (35.0 g) in a 10 wt% gelatin aqueous solution (30 mL, 3.0 g as gelatin), 1 mm mesh And then dried at 40 ° C. and sieved again. The resulting granules are mixed with magnesium stearate (2.0 g) and compressed. The resulting center tablet is coated with a sugar coating with an aqueous suspension of sucrose, titanium dioxide, talc and gum arabic. The coated tablets are polished with beeswax to obtain 1000 coated tablets.
試験例1
化合物Aとセツキシマブとの併用効果 (in vitro)
ヒト類表皮がん細胞株A431細胞を96穴プレートに2000細胞/100μL/wellで播種した。翌日、化合物A単独、セツキシマブ単独または化合物Aとセツキシマブを混合して段階希釈して加えた。化合物を添加してからCO2インキュベーター内に5日間放置した。200μLの培地に対して50μLの25%グルタルアルデヒド溶液(Wako)を加えて、室温で15分間静置した。その後、PBSで1回プレートを洗浄し、200μLのPBSを加えたところに25μLの50%トリクロロ酢酸を加えて4℃で1時間以上静置した。水道水で5回プレートを洗浄し、余分な水分をキムタオルに叩きつけて除去した。50μLの0.4%(w/v) Sulforhodamine B (SRB、Sigma)含有1%(v/v)酢酸溶液を各ウェルに加えてから、15分後に1%酢酸溶液(v/v)で3回プレートを洗浄した。プレートをよく乾燥させてから、10mM Tris溶液を加えて、プレートシェーカーにてよく攪拌し、550 nmでの吸光度を測定した(CORONA ELRCTRIC、MTP-450)。薬物を加えていないコントロールを100%として算出した(図1)。化合物Aとセツキシマブを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test example 1
Combined effect of Compound A and cetuximab (in vitro)
Human epidermoid carcinoma cell line A431 cells were seeded in a 96-well plate at 2000 cells / 100 μL / well. The next day, Compound A alone, cetuximab alone or Compound A and cetuximab were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 50 μL of 25% glutaraldehyde solution (Wako) was added to 200 μL of the medium, and allowed to stand at room temperature for 15 minutes. Thereafter, the plate was washed once with PBS, 200 μL of PBS was added, 25 μL of 50% trichloroacetic acid was added, and the mixture was allowed to stand at 4 ° C. for 1 hour or longer. The plate was washed 5 times with tap water, and excess water was removed by tapping on a Kim towel. 50 μL of 0.4% (w / v) Sulforhodamine B (SRB, Sigma) -containing 1% (v / v) acetic acid solution was added to each well, and 15 minutes later, 1% acetic acid solution (v / v) was plated 3 times. Was washed. After the plate was thoroughly dried, 10 mM Tris solution was added, and the mixture was well stirred with a plate shaker, and the absorbance at 550 nm was measured (CORONA ELRCTRIC, MTP-450). Calculations were made assuming that the control with no drug added was 100% (FIG. 1). The combined use of Compound A and cetuximab showed a strong cell growth inhibitory effect compared to the case of each alone.
化合物Aとセツキシマブとの併用効果 (in vitro)
ヒト類表皮がん細胞株A431細胞を96穴プレートに2000細胞/100μL/wellで播種した。翌日、化合物A単独、セツキシマブ単独または化合物Aとセツキシマブを混合して段階希釈して加えた。化合物を添加してからCO2インキュベーター内に5日間放置した。200μLの培地に対して50μLの25%グルタルアルデヒド溶液(Wako)を加えて、室温で15分間静置した。その後、PBSで1回プレートを洗浄し、200μLのPBSを加えたところに25μLの50%トリクロロ酢酸を加えて4℃で1時間以上静置した。水道水で5回プレートを洗浄し、余分な水分をキムタオルに叩きつけて除去した。50μLの0.4%(w/v) Sulforhodamine B (SRB、Sigma)含有1%(v/v)酢酸溶液を各ウェルに加えてから、15分後に1%酢酸溶液(v/v)で3回プレートを洗浄した。プレートをよく乾燥させてから、10mM Tris溶液を加えて、プレートシェーカーにてよく攪拌し、550 nmでの吸光度を測定した(CORONA ELRCTRIC、MTP-450)。薬物を加えていないコントロールを100%として算出した(図1)。化合物Aとセツキシマブを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test example 1
Combined effect of Compound A and cetuximab (in vitro)
Human epidermoid carcinoma cell line A431 cells were seeded in a 96-well plate at 2000 cells / 100 μL / well. The next day, Compound A alone, cetuximab alone or Compound A and cetuximab were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 50 μL of 25% glutaraldehyde solution (Wako) was added to 200 μL of the medium, and allowed to stand at room temperature for 15 minutes. Thereafter, the plate was washed once with PBS, 200 μL of PBS was added, 25 μL of 50% trichloroacetic acid was added, and the mixture was allowed to stand at 4 ° C. for 1 hour or longer. The plate was washed 5 times with tap water, and excess water was removed by tapping on a Kim towel. 50 μL of 0.4% (w / v) Sulforhodamine B (SRB, Sigma) -containing 1% (v / v) acetic acid solution was added to each well, and 15 minutes later, 1% acetic acid solution (v / v) was plated 3 times. Was washed. After the plate was thoroughly dried, 10 mM Tris solution was added, and the mixture was well stirred with a plate shaker, and the absorbance at 550 nm was measured (CORONA ELRCTRIC, MTP-450). Calculations were made assuming that the control with no drug added was 100% (FIG. 1). The combined use of Compound A and cetuximab showed a strong cell growth inhibitory effect compared to the case of each alone.
試験例2
化合物Aとエルロチニブとの併用効果 (in vitro)
ヒト肺がん細胞株Calu-3細胞を96穴プレートに5000細胞/50μL/wellで播種した。翌日、化合物A単独、エルロチニブ単独または化合物Aとエルロチニブを混合して段階希釈して加えた。化合物を添加してからCO2インキュベーター内に5日間放置した。100μLの培地に対して100μLのCellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571)を加えてよく攪拌した。発光強度をARVO light (Perkin Elmer)にて測定した。薬物を加えていないコントロールでの発光強度を100%として算出した(図2)。化合物Aとエルロチニブを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test example 2
Combined effect of compound A and erlotinib (in vitro)
Human lung cancer cell line Calu-3 cells were seeded in a 96-well plate at 5000 cells / 50 μL / well. The next day, compound A alone, erlotinib alone or compound A and erlotinib were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 μL of the medium and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The luminescence intensity in the control to which no drug was added was calculated as 100% (FIG. 2). By using Compound A and erlotinib in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
化合物Aとエルロチニブとの併用効果 (in vitro)
ヒト肺がん細胞株Calu-3細胞を96穴プレートに5000細胞/50μL/wellで播種した。翌日、化合物A単独、エルロチニブ単独または化合物Aとエルロチニブを混合して段階希釈して加えた。化合物を添加してからCO2インキュベーター内に5日間放置した。100μLの培地に対して100μLのCellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571)を加えてよく攪拌した。発光強度をARVO light (Perkin Elmer)にて測定した。薬物を加えていないコントロールでの発光強度を100%として算出した(図2)。化合物Aとエルロチニブを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test example 2
Combined effect of compound A and erlotinib (in vitro)
Human lung cancer cell line Calu-3 cells were seeded in a 96-well plate at 5000 cells / 50 μL / well. The next day, compound A alone, erlotinib alone or compound A and erlotinib were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 μL of the medium and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The luminescence intensity in the control to which no drug was added was calculated as 100% (FIG. 2). By using Compound A and erlotinib in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
試験例3
化合物Aとトラスツズマブとの併用効果 (in vitro)
ヒト乳がん細胞株BT-474細胞を96穴プレートに6000細胞/50μL/wellで播種した。翌日、化合物A単独、トラスツズマブ単独または化合物Aとトラスツズマブを混合して段階希釈して加えた。化合物を添加してからCO2インキュベーター内に5日間放置した。100μLの培地に対して100μLのCellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571)を加えてよく攪拌した。発光強度をARVO light (Perkin Elmer)にて測定した。薬物を加えていないコントロールを100%として算出した(図3)。化合物Aとトラスツズマブを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test example 3
Combined effect of Compound A and trastuzumab (in vitro)
Human breast cancer cell line BT-474 cells were seeded in a 96-well plate at 6000 cells / 50 μL / well. The next day, compound A alone, trastuzumab alone or compound A and trastuzumab were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 μL of the medium and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). Calculations were made assuming that the control with no drug added was 100% (FIG. 3). The combined use of Compound A and trastuzumab showed a stronger cell growth inhibitory effect than when each compound was used alone.
化合物Aとトラスツズマブとの併用効果 (in vitro)
ヒト乳がん細胞株BT-474細胞を96穴プレートに6000細胞/50μL/wellで播種した。翌日、化合物A単独、トラスツズマブ単独または化合物Aとトラスツズマブを混合して段階希釈して加えた。化合物を添加してからCO2インキュベーター内に5日間放置した。100μLの培地に対して100μLのCellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571)を加えてよく攪拌した。発光強度をARVO light (Perkin Elmer)にて測定した。薬物を加えていないコントロールを100%として算出した(図3)。化合物Aとトラスツズマブを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test example 3
Combined effect of Compound A and trastuzumab (in vitro)
Human breast cancer cell line BT-474 cells were seeded in a 96-well plate at 6000 cells / 50 μL / well. The next day, compound A alone, trastuzumab alone or compound A and trastuzumab were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 μL of the medium and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). Calculations were made assuming that the control with no drug added was 100% (FIG. 3). The combined use of Compound A and trastuzumab showed a stronger cell growth inhibitory effect than when each compound was used alone.
試験例4
化合物Aとトラスツズマブとの併用効果 (in vivo)
継代培養しているHER2高発現細胞であるヒト乳癌細胞株BT-474(ATCC (American Type Culture Collection)カタログNo.HTB-20、Lasfargues EY, In Vitro 15:723-729(1979))をトリプシン処理し、10%牛胎児血清 (GibcoInvitrogen)を含むRPMI-1640培地 (GibcoInvitrogen)に懸濁した。この細胞懸濁液の細胞密度を細胞計数装置シスメックスCDA500で測定し、上述の培地を用いて細胞密度を1 x 108細胞/mLに調製した。さらにこれを同量のMatrigel (Basement Membrane Matrix, phenol-red free, Cat.No.40234C, Becton Dickinson)にて希釈しヌードマウス移植用細胞懸濁液(5 x 107 細胞/ mL)とした。移植用ヌードマウスはBALB/cAJcl-nu/nu、雌性、5週齢のマウスを日本クレア株式会社から購入した。入荷後1週間順化を行い、6週齢時のマウスの腹部皮下に上記細胞懸濁液を1 x 107細胞/200μLの濃度にて細胞を移植した。移植時に、プロピオン酸エストラジオール(オバホルモン・デポー 5mg あすか製薬)を右後足に50μLの用量で筋肉内注射した。さらに、移植1週間後、同様にプロピオン酸エストラジオールを左後足に50μLの用量で筋肉内注射した。移植2週間後から腫瘍体積を経時的に測定し、腫瘍体積が200から300mm3になった個体を使用した。
化合物Aを0.5%メチルセルロース水溶液にて13.06mg / mLの濃度になるよう調製し、上記化合物A懸濁液を10 mL / kgの液量で経口投与した。化合物Aは1日2回午前と午後の投与を行い(投与間隔は7時間以上あけることとした)、14日間連続投与した。トラスツズマブ(Genentech,Inc. カタログ No. NDC 50242-134-68)を、原液21mg / mLの溶液を生理食塩水にて1mg / mLとなるよう希釈調製し、10 mL / kgの液量で腹腔内投与した。トラスツズマブは1週間に2回、計4回の投与を行った。2日から3日毎に経時的に腫瘍体積を測定し、14日間投薬終了翌日に最終腫瘍体積を測定し、併用効果の判定を行った。
HER2高発現BT-474乳癌腫瘍に対して、化合物Aはトラスツズマブと相乗効果を示した(図4)。 Test example 4
Combined effect of Compound A and trastuzumab (in vivo)
Trypsinized human breast cancer cell line BT-474 (ATCC (American Type Culture Collection) catalog No.HTB-20, Lasfargues EY, In Vitro 15: 723-729 (1979)), a HER2 highly expressing cell that has been subcultured The suspension was treated and suspended in RPMI-1640 medium (GibcoInvitrogen) containing 10% fetal bovine serum (GibcoInvitrogen). The cell density of this cell suspension was measured with a cell counter Sysmex CDA500, and the cell density was adjusted to 1 × 10 8 cells / mL using the above-mentioned medium. This was further diluted with the same amount of Matrigel (Basement Membrane Matrix, phenol-red free, Cat. No. 40234C, Becton Dickinson) to obtain a cell suspension for transplantation of nude mice (5 × 10 7 cells / mL). Nude mice for transplantation were BALB / cAJcl-nu / nu, female, 5-week-old mice purchased from CLEA Japan. After arrival, the cells were acclimatized for 1 week, and the cells were transplanted at a concentration of 1 × 10 7 cells / 200 μL into the abdomen subcutaneously of a 6-week-old mouse. At the time of transplantation, estradiol propionate (Ova hormone Depot 5mg Asuka Pharmaceutical) was injected intramuscularly at a dose of 50 μL in the right hind paw. Furthermore, one week after transplantation, estradiol propionate was similarly injected intramuscularly into the left hind paw at a dose of 50 μL. Tumor volume was measured over time from 2 weeks after transplantation, and an individual with a tumor volume of 200 to 300 mm 3 was used.
Compound A was prepared in 0.5% aqueous methylcellulose solution to a concentration of 13.06 mg / mL, and the above-mentioned Compound A suspension was orally administered at a liquid volume of 10 mL / kg. Compound A was administered twice a day in the morning and afternoon (the administration interval was set to be 7 hours or longer), and was administered continuously for 14 days. Trastuzumab (Genentech, Inc. Catalog No. NDC 50242-134-68) was prepared by diluting a stock solution of 21 mg / mL to 1 mg / mL with physiological saline and intraperitoneally at a volume of 10 mL / kg. Administered. Trastuzumab was administered four times, twice a week. The tumor volume was measured over time every 2 to 3 days, and the final tumor volume was measured on the day after the end of the 14-day administration to determine the combined effect.
Compound A showed a synergistic effect with trastuzumab on HER2-highly expressing BT-474 breast cancer tumors (FIG. 4).
化合物Aとトラスツズマブとの併用効果 (in vivo)
継代培養しているHER2高発現細胞であるヒト乳癌細胞株BT-474(ATCC (American Type Culture Collection)カタログNo.HTB-20、Lasfargues EY, In Vitro 15:723-729(1979))をトリプシン処理し、10%牛胎児血清 (GibcoInvitrogen)を含むRPMI-1640培地 (GibcoInvitrogen)に懸濁した。この細胞懸濁液の細胞密度を細胞計数装置シスメックスCDA500で測定し、上述の培地を用いて細胞密度を1 x 108細胞/mLに調製した。さらにこれを同量のMatrigel (Basement Membrane Matrix, phenol-red free, Cat.No.40234C, Becton Dickinson)にて希釈しヌードマウス移植用細胞懸濁液(5 x 107 細胞/ mL)とした。移植用ヌードマウスはBALB/cAJcl-nu/nu、雌性、5週齢のマウスを日本クレア株式会社から購入した。入荷後1週間順化を行い、6週齢時のマウスの腹部皮下に上記細胞懸濁液を1 x 107細胞/200μLの濃度にて細胞を移植した。移植時に、プロピオン酸エストラジオール(オバホルモン・デポー 5mg あすか製薬)を右後足に50μLの用量で筋肉内注射した。さらに、移植1週間後、同様にプロピオン酸エストラジオールを左後足に50μLの用量で筋肉内注射した。移植2週間後から腫瘍体積を経時的に測定し、腫瘍体積が200から300mm3になった個体を使用した。
化合物Aを0.5%メチルセルロース水溶液にて13.06mg / mLの濃度になるよう調製し、上記化合物A懸濁液を10 mL / kgの液量で経口投与した。化合物Aは1日2回午前と午後の投与を行い(投与間隔は7時間以上あけることとした)、14日間連続投与した。トラスツズマブ(Genentech,Inc. カタログ No. NDC 50242-134-68)を、原液21mg / mLの溶液を生理食塩水にて1mg / mLとなるよう希釈調製し、10 mL / kgの液量で腹腔内投与した。トラスツズマブは1週間に2回、計4回の投与を行った。2日から3日毎に経時的に腫瘍体積を測定し、14日間投薬終了翌日に最終腫瘍体積を測定し、併用効果の判定を行った。
HER2高発現BT-474乳癌腫瘍に対して、化合物Aはトラスツズマブと相乗効果を示した(図4)。 Test example 4
Combined effect of Compound A and trastuzumab (in vivo)
Trypsinized human breast cancer cell line BT-474 (ATCC (American Type Culture Collection) catalog No.HTB-20, Lasfargues EY, In Vitro 15: 723-729 (1979)), a HER2 highly expressing cell that has been subcultured The suspension was treated and suspended in RPMI-1640 medium (GibcoInvitrogen) containing 10% fetal bovine serum (GibcoInvitrogen). The cell density of this cell suspension was measured with a cell counter Sysmex CDA500, and the cell density was adjusted to 1 × 10 8 cells / mL using the above-mentioned medium. This was further diluted with the same amount of Matrigel (Basement Membrane Matrix, phenol-red free, Cat. No. 40234C, Becton Dickinson) to obtain a cell suspension for transplantation of nude mice (5 × 10 7 cells / mL). Nude mice for transplantation were BALB / cAJcl-nu / nu, female, 5-week-old mice purchased from CLEA Japan. After arrival, the cells were acclimatized for 1 week, and the cells were transplanted at a concentration of 1 × 10 7 cells / 200 μL into the abdomen subcutaneously of a 6-week-old mouse. At the time of transplantation, estradiol propionate (Ova hormone Depot 5mg Asuka Pharmaceutical) was injected intramuscularly at a dose of 50 μL in the right hind paw. Furthermore, one week after transplantation, estradiol propionate was similarly injected intramuscularly into the left hind paw at a dose of 50 μL. Tumor volume was measured over time from 2 weeks after transplantation, and an individual with a tumor volume of 200 to 300 mm 3 was used.
Compound A was prepared in 0.5% aqueous methylcellulose solution to a concentration of 13.06 mg / mL, and the above-mentioned Compound A suspension was orally administered at a liquid volume of 10 mL / kg. Compound A was administered twice a day in the morning and afternoon (the administration interval was set to be 7 hours or longer), and was administered continuously for 14 days. Trastuzumab (Genentech, Inc. Catalog No. NDC 50242-134-68) was prepared by diluting a stock solution of 21 mg / mL to 1 mg / mL with physiological saline and intraperitoneally at a volume of 10 mL / kg. Administered. Trastuzumab was administered four times, twice a week. The tumor volume was measured over time every 2 to 3 days, and the final tumor volume was measured on the day after the end of the 14-day administration to determine the combined effect.
Compound A showed a synergistic effect with trastuzumab on HER2-highly expressing BT-474 breast cancer tumors (FIG. 4).
試験例5
化合物Aの添加によるTS(サイミジン)発現量の測定
継代培養しているHER2高発現細胞であるヒト乳癌細胞株BT-474 (ATCC (American Type Culture Collection)カタログNo.HTB-20、Lasfargues EY, In Vitro 15:723-729(1979))をトリプシン処理し、10%牛胎児血清 (GibcoInvitrogen)を含むRPMI-1640培地(GibcoInvitrogen)に懸濁した。この細胞懸濁液の細胞密度を細胞計数装置シスメックスCDA500で測定し、上述の培地を用いて細胞密度を5 x 104 細胞/mLに調製した。これを6 well マルチウエル培養プレート(Corning)の各ウエルに2 mLずつ分注し、37℃、5% CO2下で一夜培養した。その培養プレートに化合物Aを100nmol/Lとなるように添加した。化合物添加2日後にRNA easy Mini kit(QIAGEN)により各々のtotal RNAを抽出し、TaqMan Reverse Transcription Reagent (Applied Biosystems) を用いてcDNAに変換した。このcDNAを鋳型としTS発現量をTaqMan Gene Expression Assays (Applied Biosystems)を用いて調べた。GeneAmp7700 (Applied Biosystems)により検出し、GAPDHを内部標準として
(Applied Biosystems社参照)を用いて解析した(図5)。化合物Aを100 nmol/L添加することにより、TS発現量が顕著に低下した。 Test Example 5
Measurement of TS (thymidine) expression level by addition of compound A Human breast cancer cell line BT-474 (ATCC (American Type Culture Collection) catalog No. HTB-20, Lasfargues EY, In Vitro 15: 723-729 (1979)) was trypsinized and suspended in RPMI-1640 medium (GibcoInvitrogen) containing 10% fetal bovine serum (GibcoInvitrogen). The cell density of this cell suspension was measured with a cell counter Sysmex CDA500, and the cell density was adjusted to 5 × 10 4 cells / mL using the above-mentioned medium. 2 mL of this was dispensed into each well of a 6-well multi-well culture plate (Corning) and cultured overnight at 37 ° C. and 5% CO 2 . Compound A was added to the culture plate at 100 nmol / L. Two days after compound addition, each total RNA was extracted with RNA easy Mini kit (QIAGEN) and converted into cDNA using TaqMan Reverse Transcription Reagent (Applied Biosystems). Using this cDNA as a template, the TS expression level was examined using TaqMan Gene Expression Assays (Applied Biosystems). Detected with GeneAmp7700 (Applied Biosystems) and GAPDH as internal standard
(See Applied Biosystems) (FIG. 5). By adding 100 nmol / L of compound A, the TS expression level was significantly reduced.
化合物Aの添加によるTS(サイミジン)発現量の測定
継代培養しているHER2高発現細胞であるヒト乳癌細胞株BT-474 (ATCC (American Type Culture Collection)カタログNo.HTB-20、Lasfargues EY, In Vitro 15:723-729(1979))をトリプシン処理し、10%牛胎児血清 (GibcoInvitrogen)を含むRPMI-1640培地(GibcoInvitrogen)に懸濁した。この細胞懸濁液の細胞密度を細胞計数装置シスメックスCDA500で測定し、上述の培地を用いて細胞密度を5 x 104 細胞/mLに調製した。これを6 well マルチウエル培養プレート(Corning)の各ウエルに2 mLずつ分注し、37℃、5% CO2下で一夜培養した。その培養プレートに化合物Aを100nmol/Lとなるように添加した。化合物添加2日後にRNA easy Mini kit(QIAGEN)により各々のtotal RNAを抽出し、TaqMan Reverse Transcription Reagent (Applied Biosystems) を用いてcDNAに変換した。このcDNAを鋳型としTS発現量をTaqMan Gene Expression Assays (Applied Biosystems)を用いて調べた。GeneAmp7700 (Applied Biosystems)により検出し、GAPDHを内部標準として
(Applied Biosystems社参照)を用いて解析した(図5)。化合物Aを100 nmol/L添加することにより、TS発現量が顕著に低下した。 Test Example 5
Measurement of TS (thymidine) expression level by addition of compound A Human breast cancer cell line BT-474 (ATCC (American Type Culture Collection) catalog No. HTB-20, Lasfargues EY, In Vitro 15: 723-729 (1979)) was trypsinized and suspended in RPMI-1640 medium (GibcoInvitrogen) containing 10% fetal bovine serum (GibcoInvitrogen). The cell density of this cell suspension was measured with a cell counter Sysmex CDA500, and the cell density was adjusted to 5 × 10 4 cells / mL using the above-mentioned medium. 2 mL of this was dispensed into each well of a 6-well multi-well culture plate (Corning) and cultured overnight at 37 ° C. and 5% CO 2 . Compound A was added to the culture plate at 100 nmol / L. Two days after compound addition, each total RNA was extracted with RNA easy Mini kit (QIAGEN) and converted into cDNA using TaqMan Reverse Transcription Reagent (Applied Biosystems). Using this cDNA as a template, the TS expression level was examined using TaqMan Gene Expression Assays (Applied Biosystems). Detected with GeneAmp7700 (Applied Biosystems) and GAPDH as internal standard
(See Applied Biosystems) (FIG. 5). By adding 100 nmol / L of compound A, the TS expression level was significantly reduced.
試験例6
化合物AとAZD6474との併用効果 (A431 in vitro)
ヒト類表皮がん細胞株 A431 細胞を 96 穴プレートに2000 cells/ 50 μL/well で播種した。翌日に化合物A単独、AZD6474単独または化合物AとAZD6474を混合して段階希釈して加えた。化合物を添加してから CO2 インキュベーター内に 5 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) を加えて、室温で 5 分程度プレートシェーカーにて混和した。その後10分程度室温にプレートを放置し、発光をARVO-Lightにて測定した (Perkin Elmer)。培地のみが入ったwellの発行量をバックグラウンドとし、薬物を加えていないコントロールを 100% として算出した(図6)。化合物Aと AZD6474を併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 6
Combined effect of Compound A and AZD6474 (A431 in vitro)
Human epidermoid carcinoma cell line A431 cells were seeded in a 96-well plate at 2000 cells / 50 μL / well. The next day, Compound A alone, AZD6474 alone or Compound A and AZD6474 were mixed and added in serial dilution. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 μL of medium and mixed on a plate shaker for about 5 minutes at room temperature. Thereafter, the plate was left at room temperature for about 10 minutes, and luminescence was measured with ARVO-Light (Perkin Elmer). The amount of wells containing only the medium was calculated as the background, and the control with no drug added was calculated as 100% (FIG. 6). By using Compound A and AZD6474 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
化合物AとAZD6474との併用効果 (A431 in vitro)
ヒト類表皮がん細胞株 A431 細胞を 96 穴プレートに2000 cells/ 50 μL/well で播種した。翌日に化合物A単独、AZD6474単独または化合物AとAZD6474を混合して段階希釈して加えた。化合物を添加してから CO2 インキュベーター内に 5 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) を加えて、室温で 5 分程度プレートシェーカーにて混和した。その後10分程度室温にプレートを放置し、発光をARVO-Lightにて測定した (Perkin Elmer)。培地のみが入ったwellの発行量をバックグラウンドとし、薬物を加えていないコントロールを 100% として算出した(図6)。化合物Aと AZD6474を併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 6
Combined effect of Compound A and AZD6474 (A431 in vitro)
Human epidermoid carcinoma cell line A431 cells were seeded in a 96-well plate at 2000 cells / 50 μL / well. The next day, Compound A alone, AZD6474 alone or Compound A and AZD6474 were mixed and added in serial dilution. After adding the compound, it was left in a CO 2 incubator for 5 days. 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added to 100 μL of medium and mixed on a plate shaker for about 5 minutes at room temperature. Thereafter, the plate was left at room temperature for about 10 minutes, and luminescence was measured with ARVO-Light (Perkin Elmer). The amount of wells containing only the medium was calculated as the background, and the control with no drug added was calculated as 100% (FIG. 6). By using Compound A and AZD6474 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
試験例7
化合物AとBEZ235との併用効果 (SK-BR-3 in vitro)
ヒト乳がん細胞株 SK-BR-3 細胞を 96 穴プレートに2000 cells/ 50 μL/well で播種した。2日後、化合物A単独、BEZ235単独または化合物AとBEZ235を混合して段階希釈して加えた。化合物を添加してから CO2インキュベーター内に 4 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega、G7571) を加えてよく攪拌した。発光強度を ARVO-light (Perkin Elmer) にて測定した。薬物を加えていないコントロールでの発光強度を 100% として算出した(図7)。化合物AとBEZ235を併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 7
Combined effect of Compound A and BEZ235 (SK-BR-3 in vitro)
Human breast cancer cell line SK-BR-3 cells were seeded in a 96-well plate at 2000 cells / 50 μL / well. Two days later, Compound A alone, BEZ235 alone or Compound A and BEZ235 were mixed and serially diluted. After the compound was added, it was left in a CO 2 incubator for 4 days. To 100 μL of the medium, 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO-light (Perkin Elmer). The luminescence intensity in the control with no drug added was calculated as 100% (FIG. 7). By using Compound A and BEZ235 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
化合物AとBEZ235との併用効果 (SK-BR-3 in vitro)
ヒト乳がん細胞株 SK-BR-3 細胞を 96 穴プレートに2000 cells/ 50 μL/well で播種した。2日後、化合物A単独、BEZ235単独または化合物AとBEZ235を混合して段階希釈して加えた。化合物を添加してから CO2インキュベーター内に 4 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega、G7571) を加えてよく攪拌した。発光強度を ARVO-light (Perkin Elmer) にて測定した。薬物を加えていないコントロールでの発光強度を 100% として算出した(図7)。化合物AとBEZ235を併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 7
Combined effect of Compound A and BEZ235 (SK-BR-3 in vitro)
Human breast cancer cell line SK-BR-3 cells were seeded in a 96-well plate at 2000 cells / 50 μL / well. Two days later, Compound A alone, BEZ235 alone or Compound A and BEZ235 were mixed and serially diluted. After the compound was added, it was left in a CO 2 incubator for 4 days. To 100 μL of the medium, 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO-light (Perkin Elmer). The luminescence intensity in the control with no drug added was calculated as 100% (FIG. 7). By using Compound A and BEZ235 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
試験例8
化合物AとPD0325901との併用効果 (Calu-3 in vitro)
ヒト肺がん細胞株 Calu-3 細胞を 96 穴プレートに5000 cells/ 50 μL/well で播種した。翌日、化合物A単独、PD0325901 単独または化合物AとPD0325901を混合して段階希釈して加えた。化合物を添加してから CO2 インキュベーター内に 5 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega、G7571) を加えてよく攪拌した。発光強度を ARVO light (Perkin Elmer) にて測定した。薬物を加えていないコントロールでの発光強度を 100% として算出した(図8)。化合物AとPD0325901を併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 8
Combined effect of Compound A and PD0325901 (Calu-3 in vitro)
Human lung cancer cell line Calu-3 cells were seeded in a 96-well plate at 5000 cells / 50 μL / well. The next day, Compound A alone, PD0325901 alone or Compound A and PD0325901 were mixed and serially diluted. After adding the compound, it was left in a CO 2 incubator for 5 days. To 100 μL of the medium, 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The luminescence intensity in the control to which no drug was added was calculated as 100% (FIG. 8). By using Compound A and PD0325901 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
化合物AとPD0325901との併用効果 (Calu-3 in vitro)
ヒト肺がん細胞株 Calu-3 細胞を 96 穴プレートに5000 cells/ 50 μL/well で播種した。翌日、化合物A単独、PD0325901 単独または化合物AとPD0325901を混合して段階希釈して加えた。化合物を添加してから CO2 インキュベーター内に 5 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega、G7571) を加えてよく攪拌した。発光強度を ARVO light (Perkin Elmer) にて測定した。薬物を加えていないコントロールでの発光強度を 100% として算出した(図8)。化合物AとPD0325901を併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 8
Combined effect of Compound A and PD0325901 (Calu-3 in vitro)
Human lung cancer cell line Calu-3 cells were seeded in a 96-well plate at 5000 cells / 50 μL / well. The next day, Compound A alone, PD0325901 alone or Compound A and PD0325901 were mixed and serially diluted. After adding the compound, it was left in a CO 2 incubator for 5 days. To 100 μL of the medium, 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The luminescence intensity in the control to which no drug was added was calculated as 100% (FIG. 8). By using Compound A and PD0325901 in combination, the cell growth inhibitory action was stronger than when each compound was used alone.
試験例9
化合物Aとラパマイシンとの併用効果 (BT-474 in vitro)
ヒト乳がん細胞株 BT-474 細胞を 96 穴プレートに6000 cells/ 50 μL/well で播種した。翌日に化合物A単独、ラパマイシン単独または化合物Aとラパマイシンを混合して段階希釈して加えた。化合物を添加してから CO2 インキュベーター内に 5 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega、G7571) を加えてよく攪拌した。発光強度を ARVO light (Perkin Elmer) にて測定した。培地のみが入ったwellの発行量をバックグラウンドとし、薬物を加えていないコントロールを 100% として算出した(図9)。化合物Aとラパマイシンを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 9
Combined effect of Compound A and rapamycin (BT-474 in vitro)
Human breast cancer cell line BT-474 cells were seeded in a 96-well plate at 6000 cells / 50 μL / well. The next day, compound A alone, rapamycin alone or compound A and rapamycin were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. To 100 μL of the medium, 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The amount of wells containing only the medium was calculated as the background, and the control with no drug added was calculated as 100% (FIG. 9). The combined use of Compound A and rapamycin showed a stronger cell growth inhibitory effect than when each compound was used alone.
化合物Aとラパマイシンとの併用効果 (BT-474 in vitro)
ヒト乳がん細胞株 BT-474 細胞を 96 穴プレートに6000 cells/ 50 μL/well で播種した。翌日に化合物A単独、ラパマイシン単独または化合物Aとラパマイシンを混合して段階希釈して加えた。化合物を添加してから CO2 インキュベーター内に 5 日間放置した。100 μL の培地に対して 100 μL の CellTiter-Glo Luminescent Cell Viability assay reagent (Promega、G7571) を加えてよく攪拌した。発光強度を ARVO light (Perkin Elmer) にて測定した。培地のみが入ったwellの発行量をバックグラウンドとし、薬物を加えていないコントロールを 100% として算出した(図9)。化合物Aとラパマイシンを併用することで、それぞれ各単独時に比べて強い細胞増殖阻害作用を示した。 Test Example 9
Combined effect of Compound A and rapamycin (BT-474 in vitro)
Human breast cancer cell line BT-474 cells were seeded in a 96-well plate at 6000 cells / 50 μL / well. The next day, compound A alone, rapamycin alone or compound A and rapamycin were mixed and added in serial dilutions. After adding the compound, it was left in a CO 2 incubator for 5 days. To 100 μL of the medium, 100 μL of CellTiter-Glo Luminescent Cell Viability assay reagent (Promega, G7571) was added and stirred well. Luminescence intensity was measured with ARVO light (Perkin Elmer). The amount of wells containing only the medium was calculated as the background, and the control with no drug added was calculated as 100% (FIG. 9). The combined use of Compound A and rapamycin showed a stronger cell growth inhibitory effect than when each compound was used alone.
本出願は、日本で出願された特願2008-052715を基礎としており、その内容は本明細書に全て包含されるものである。
This application is based on Japanese Patent Application No. 2008-052715 filed in Japan, the contents of which are incorporated in full herein.
Claims (21)
- (1)式:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる医薬。 (1) Formula:
[Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof;
(2) A pharmaceutical comprising a combination of a hormone therapy agent or an anticancer agent. - Xが水素原子である請求項1記載の医薬。 The medicine according to claim 1, wherein X is a hydrogen atom.
- R1が水素原子、
R2が、
-NR6a-CO-CR7R8-SO2-C1-4アルキル(R6aは水素原子またはメチル基、R7およびR8は、同一または異なって、それぞれ水素原子またはメチル基を表す)で表される基で置換されたC1-6アルキル基、
R3が水素原子、
R4が塩素原子またはメチル基、および
R5がフッ素原子、塩素原子またはメチル基である請求項2記載の医薬。 R 1 is a hydrogen atom,
R 2 is
—NR 6a —CO—CR 7 R 8 —SO 2 —C 1-4 alkyl (R 6a is a hydrogen atom or a methyl group, R 7 and R 8 are the same or different and each represents a hydrogen atom or a methyl group) A C 1-6 alkyl group substituted with a group represented by:
R 3 is a hydrogen atom,
The pharmaceutical according to claim 2, wherein R 4 is a chlorine atom or a methyl group, and R 5 is a fluorine atom, a chlorine atom or a methyl group. - R7およびR8がメチル基である請求項3記載の医薬。 The pharmaceutical according to claim 3, wherein R 7 and R 8 are methyl groups.
- (1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる請求項1記載の医薬。 (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2 -Methyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to claim 1, which is a combination of a hormone therapy agent or an anticancer agent. - (1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(エチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる請求項1記載の医薬。 (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2 -(Ethylsulfonyl) acetamide;
(2) The medicament according to claim 1, which is a combination of a hormone therapy agent or an anticancer agent. - (1)N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-N,2-ジメチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる請求項1記載の医薬。 (1) N- [2- (4-{[3-Chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -N , 2-dimethyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to claim 1, which is a combination of a hormone therapy agent or an anticancer agent. - (1)N-[2-(4-{[3-クロロ-4-(3-メチルフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる請求項1記載の医薬。 (1) N- [2- (4-{[3-Chloro-4- (3-methylphenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2 -(Methylsulfonyl) acetamide;
(2) The medicament according to claim 1, which is a combination of a hormone therapy agent or an anticancer agent. - (1)N-[2-(4-{[3-クロロ-4-(3-フルオロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる請求項1記載の医薬。 (1) N- [2- (4-{[3-Chloro-4- (3-fluorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2 -(Methylsulfonyl) acetamide;
(2) The medicament according to claim 1, which is a combination of a hormone therapy agent or an anticancer agent. - (1)N-[2-(4-{[4-(3-クロロフェノキシ)-3-メチルフェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-メチル-2-(メチルスルホニル)プロパンアミドと、
(2)ホルモン療法剤または抗癌剤
とを組み合わせてなる請求項1記載の医薬。 (1) N- [2- (4-{[4- (3-Chlorophenoxy) -3-methylphenyl] amino} -5H-pyrrolo [3,2-d] pyrimidin-5-yl) ethyl] -2 -Methyl-2- (methylsulfonyl) propanamide;
(2) The medicament according to claim 1, which is a combination of a hormone therapy agent or an anticancer agent. - 抗癌剤がHER2抗体、EGFR抗体、EGFR阻害剤、VEGFR阻害剤または化学療法剤である請求項1記載の医薬。 The medicament according to claim 1, wherein the anticancer agent is a HER2 antibody, an EGFR antibody, an EGFR inhibitor, a VEGFR inhibitor or a chemotherapeutic agent.
- 抗癌剤がトラスツズマブ、セツキシマブ、エルロチニブ、ゲフィチニブまたはパクリタキセルである請求項1記載の医薬。 The medicament according to claim 1, wherein the anticancer agent is trastuzumab, cetuximab, erlotinib, gefitinib or paclitaxel.
- ホルモン療法剤がERダウンレギュレーターである請求項1記載の医薬。 The medicament according to claim 1, wherein the hormone therapy agent is an ER down regulator.
- ホルモン療法剤がフルベストラントである請求項1記載の医薬。 The medicine according to claim 1, wherein the hormone therapy agent is fulvestrant.
- 癌の予防または治療剤である請求項1記載の医薬。 The medicament according to claim 1, which is a preventive or therapeutic agent for cancer.
- 癌が乳癌、卵巣癌、前立腺癌、肺癌、膵癌、腎臓癌、大腸癌、小腸癌、食道癌または胃癌である請求項15記載の医薬。 The medicament according to claim 15, wherein the cancer is breast cancer, ovarian cancer, prostate cancer, lung cancer, pancreatic cancer, kidney cancer, colon cancer, small intestine cancer, esophageal cancer or stomach cancer.
- 哺乳動物に対して、(1)式:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの有効量と、(2)ホルモン療法剤または抗癌剤の有効量とを投与することを特徴とする癌の予防または治療方法。 For mammals, formula (1):
[Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Ile) ethyl] -2- (methylsulfonyl) acetamide)) or a salt thereof or a prodrug thereof, and (2) an effective amount of a hormonal therapeutic agent or an anticancer agent, Or treatment method. - 癌の予防または治療剤を製造するための、(1)式:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグと、(2)ホルモン療法剤または抗癌剤の使用。 Formula (1) for producing a preventive or therapeutic agent for cancer:
[Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt thereof or a prodrug thereof, and (2) use of a hormone therapeutic agent or an anticancer agent. - 式:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグを含有してなるサイミジン合成酵素産生阻害剤。 formula:
[Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide), or a salt or prodrug thereof, a thymidine synthase production inhibitor. - 哺乳動物に対して、式:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの有効量を投与することを特徴とするサイミジン合成酵素産生阻害方法。 For mammals, the formula:
[Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt thereof or a prodrug thereof, and a method for inhibiting thymidine synthase production, which comprises administering an effective amount thereof. - サイミジン合成酵素産生阻害剤を製造するための、式:
[式中、R1は水素原子を、
R2は、
-NR6-CO-(CH2)n-SO2-ハロゲン化されていてもよいC1-4アルキル(nは1から4の整数であり、R6は水素原子またはC1-4アルキル基であり、-(CH2)n-はC1-4アルキルで置換されていてもよい)で表される基で置換されたC1-6アルキル基を、
R3は水素原子またはC1-6アルキル基を、
R4はハロゲン原子またはC1-6アルキル基を、
R5はハロゲン原子またはC1-6アルキル基を、
Xは水素原子またはハロゲン原子を
示す。]で表される化合物(ただし、N-[2-(4-{[3-クロロ-4-(3-クロロフェノキシ)フェニル]アミノ}-5H-ピロロ[3,2-d]ピリミジン-5-イル)エチル]-2-(メチルスルホニル)アセトアミドを除く)またはその塩もしくはそのプロドラッグの使用。 Formula for the production of thymidine synthase production inhibitor:
[Wherein R 1 represents a hydrogen atom,
R 2 is
—NR 6 —CO— (CH 2 ) n —SO 2 —optionally halogenated C 1-4 alkyl (n is an integer of 1 to 4; R 6 is a hydrogen atom or a C 1-4 alkyl group; A C 1-6 alkyl group substituted with a group represented by the formula : — (CH 2 ) n — may be substituted with C 1-4 alkyl),
R 3 represents a hydrogen atom or a C 1-6 alkyl group,
R 4 represents a halogen atom or a C 1-6 alkyl group,
R 5 represents a halogen atom or a C 1-6 alkyl group,
X represents a hydrogen atom or a halogen atom. Embedded image wherein N- [2- (4-{[3-chloro-4- (3-chlorophenoxy) phenyl] amino} -5H-pyrrolo [3,2-d] pyrimidine-5- Yl) ethyl] -2- (methylsulfonyl) acetamide) or a salt or prodrug thereof.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2008-052715 | 2008-03-03 | ||
JP2008052715 | 2008-03-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2009110416A1 true WO2009110416A1 (en) | 2009-09-11 |
Family
ID=41055971
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2009/053833 WO2009110416A1 (en) | 2008-03-03 | 2009-03-02 | Concomitant drug |
Country Status (4)
Country | Link |
---|---|
AR (1) | AR070735A1 (en) |
TW (1) | TW200940546A (en) |
UY (1) | UY31683A (en) |
WO (1) | WO2009110416A1 (en) |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007064045A1 (en) * | 2005-12-02 | 2007-06-07 | Takeda Pharmaceutical Company Limited | Fused heterocyclic compound |
-
2009
- 2009-03-02 TW TW098106628A patent/TW200940546A/en unknown
- 2009-03-02 AR ARP090100731A patent/AR070735A1/en unknown
- 2009-03-02 UY UY0001031683A patent/UY31683A/en not_active Application Discontinuation
- 2009-03-02 WO PCT/JP2009/053833 patent/WO2009110416A1/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007064045A1 (en) * | 2005-12-02 | 2007-06-07 | Takeda Pharmaceutical Company Limited | Fused heterocyclic compound |
Also Published As
Publication number | Publication date |
---|---|
TW200940546A (en) | 2009-10-01 |
UY31683A (en) | 2009-11-10 |
AR070735A1 (en) | 2010-04-28 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
KR102021157B1 (en) | Combinations of akt inhibitor compounds and abiraterone, and methods of use | |
EP3233852B1 (en) | Tetrahydro-pyrido[3,4-b]indoles as estrogen receptor modulators and uses thereof | |
RU2492864C2 (en) | Method of treating cancer carrying egfr mutations | |
JP6754071B2 (en) | Combination medications containing metformin and dihydroquercetin, and use for the treatment of cancer | |
KR20070050478A (en) | Combinations for the treatment of diseases involving cell proliferation | |
TW201639578A (en) | Active metabolites of apilimod and uses thereof | |
CN111837146A (en) | Excellent dosage regimen for G1T38 | |
BR112019027967A2 (en) | isolated crystalline form b, pharmaceutical composition, method for treating a disorder, method of a compound or composition, and, process for producing crystalline form b. | |
JP2010505745A (en) | Concomitant medication | |
WO2009110415A1 (en) | Concomitant drug | |
WO2009110416A1 (en) | Concomitant drug | |
US20240343728A1 (en) | Compounds that inhibit pi3k isoform alpha and methods for treating cancer | |
WO2018186366A1 (en) | Novel cyclin-dependent kinase 8 and/or 19 inhibitor | |
JP2012097032A (en) | Concomitant medicine |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 09717204 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 09717204 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: JP |